1
|
Boyle AK, Tetorou K, Suff N, Beecroft L, Mazzaschi M, Karda R, Hristova M, Waddington SN, Peebles D. Ascending Vaginal Infection in Mice Induces Preterm Birth and Neonatal Morbidity. THE AMERICAN JOURNAL OF PATHOLOGY 2025; 195:891-906. [PMID: 39892780 DOI: 10.1016/j.ajpath.2025.01.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 12/06/2024] [Accepted: 01/10/2025] [Indexed: 02/04/2025]
Abstract
Preterm birth (PTB; delivery before 37 weeks), the main cause of neonatal death worldwide, can lead to adverse neurodevelopmental outcomes, as well as lung and gut pathology. PTB can be associated with ascending vaginal infection. Ascending Escherichia coli infection in pregnant mice induces PTB and reduces pup survival. The current study demonstrated that this model recapitulates the pathology observed in human preterm neonates (namely, neuroinflammation, lung injury, and gut inflammation). In neonatal brains, there is widespread cell death, microglial activation, astrogliosis, and reduced neuronal density. The utility of this model was validated by assessing the efficacy of maternal cervical gene therapy with an adeno-associated viral vector containing human β defensin 3. This improved pup survival and reduced tumor necrosis factor alpha mRNA expression in perinatal pup brains exposed to E. coli. This model provides a unique opportunity to evaluate the therapeutic benefit of preterm labor interventions on perinatal pathology.
Collapse
Affiliation(s)
- Ashley K Boyle
- Elizabeth Garrett Anderson Institute for Women's Health, University College London, London, United Kingdom.
| | - Konstantina Tetorou
- Elizabeth Garrett Anderson Institute for Women's Health, University College London, London, United Kingdom
| | - Natalie Suff
- Department of Women and Children's Health, St Thomas' Hospital, King's College London, London, United Kingdom
| | - Laura Beecroft
- Elizabeth Garrett Anderson Institute for Women's Health, University College London, London, United Kingdom
| | - Margherita Mazzaschi
- Elizabeth Garrett Anderson Institute for Women's Health, University College London, London, United Kingdom
| | - Rajvinder Karda
- Elizabeth Garrett Anderson Institute for Women's Health, University College London, London, United Kingdom
| | - Mariya Hristova
- Elizabeth Garrett Anderson Institute for Women's Health, University College London, London, United Kingdom
| | - Simon N Waddington
- Elizabeth Garrett Anderson Institute for Women's Health, University College London, London, United Kingdom; Faculty of Health Sciences, Wits/South African Medical Research Council Antiviral Gene Therapy Research Unit, Johannesburg, South Africa
| | - Donald Peebles
- Elizabeth Garrett Anderson Institute for Women's Health, University College London, London, United Kingdom
| |
Collapse
|
2
|
El-Raghi AA, Essawi WM, Hassan MAE, Hashem NM, Abdelnour SA. Interactions among factors affecting stillbirths in Egyptian buffaloes (Bubalus bubalis). Trop Anim Health Prod 2025; 57:183. [PMID: 40272636 DOI: 10.1007/s11250-025-04402-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 03/21/2025] [Indexed: 04/27/2025]
Abstract
In buffaloes, stillbirth (SB) is a major source of reproductive and economic losses. Hence, the objectives of this study were: 1) investigating the relationship between potential risk factors (body condition score [BCS], gestation period, calving season, calf sex, and dam parity) and SB occurrence in Egyptian buffaloes; and 2) identifying blood metabolites, the redox status, and immune-inflammatory attributes in calves that may be related to SB. The incidence of SB was 6.64%. Among the evaluated risk factors, BCS was a significant risk factor for SB. There was a 73.7% lower odds (lower odds odd ratio, OR = 0.246) of SB for dams with a gestation length ≥ 305 days, compared to those with a gestation length < 305 days. The risk of SB decreased steadily with increasing dam parity. The odds of SB were 2.48 times higher in male calves compared to female calves. In comparison to the spring season, the probability of SB doubled during the summer season. Calf blood serum analysis showed that SB-born calves had higher blood biochemical and cytokines alterations than normal-born calves. On the other hand, immunoglobulins and glutathione peroxidase were significantly lower in SB-born calves. Our results indicated that factors related to the induction of inflammation and/or disrupted immune system responses, such as obesity, high temperature, and oxidative stress, are the main evoking factors for SB in buffaloes; therefore, protective measures against SB in Egyptian buffaloes should be based on controlling these factors, either by nutritional interventions or management practices.
Collapse
Affiliation(s)
- Ali Ali El-Raghi
- Department of Animal, Poultry, and Fish Production, Faculty of Agriculture, Damietta University, Damietta, 34517, Egypt
| | - Walaa M Essawi
- Department of Theriogenology, Faculty of Veterinary Medicine, Aswan University, Aswan, 81528, Egypt.
| | - Mahmoud A E Hassan
- Agriculture Research Center, Animal Production Research Institute (APRI), Ministry of Agriculture, Dokki, 12619, Giza, Egypt
| | - Nesrein M Hashem
- Department of Animal and Fish Production, Faculty of Agriculture, Alexandria University, Alexandria, 21545, Egypt
| | - Sameh A Abdelnour
- Animal Production Department, Faculty of Agriculture, Zagazig University, Zagazig, 44511, Egypt
| |
Collapse
|
3
|
Auriti C, De Rose DU, Maddaloni C, Ravà L, Martini L, Di Tommaso E, Bernaschi P, Paionni E, Porzio O, Piersigilli F, Iannetta M, Dotta A, Ronchetti MP. The accuracy of presepsin in diagnosing neonatal late-onset sepsis in critically ill neonates: a prospective study. Clin Chem Lab Med 2025:cclm-2025-0128. [PMID: 40249949 DOI: 10.1515/cclm-2025-0128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Accepted: 04/10/2025] [Indexed: 04/20/2025]
Abstract
OBJECTIVES The diagnostic accuracy of presepsin (P-SEP) in the newborn is still under evaluation. METHODS In a multicenter study, we studied the accuracy of P-SEP as a diagnostic marker of late-onset sepsis (LOS) in critical newborns with underlying disorders, to define the most accurate cut-off to distinguish infected from uninfected patients. RESULTS Sixty-nine/351 newborns without infections at admission developed LOS. The median P-SEP value at T0 (admission) was 518.0 ng/L (IQR 313.0-789.0), without significant differences related to underlying diseases (p=0.52). In neonates who developed LOS, P-SEP increased at the onset of infection (T1) (median: 816.0 ng/L) and after 24-48 h (median: 901.0 ng/L) compared with their value at admission (median: 560.0 ng/L) (p<0.01 and p=0.03, respectively). The area under the ROC curve at T1 was 0.71 (95 % CI 0.65-0.78) when all cases of sepsis were included in the analysis and increased to 0.74 (95 % CI 0.66-0.81) considering only confirmed sepsis. Approximately two-thirds of patients were correctly classified, setting the cut-off at 713 ng/L, with a negative predictive value of 89.0 %. CONCLUSIONS At a cut-off of 713 ng/L, P-SEP has good accuracy in diagnosing LOS in critically ill newborns. In uninfected newborns, the median value of P-SEP is not influenced by any underlying pathology.
Collapse
Affiliation(s)
- Cinzia Auriti
- Saint Camillus International University of Health and Medical Sciences, Rome, Italy
- Casa di Cura Villa Margherita, Rome, Italy
| | - Domenico Umberto De Rose
- Neonatal Intensive Care Unit - "Bambino Gesù" Children's Hospital IRCCS, Rome, Italy
- Faculty of Medicine and Surgery, PhD Course in Microbiology, Immunology, Infectious Diseases, and Transplants (MIMIT), "Tor Vergata" University of Rome, Rome, Italy
| | - Chiara Maddaloni
- Neonatal Intensive Care Unit - "Bambino Gesù" Children's Hospital IRCCS, Rome, Italy
| | - Lucilla Ravà
- Epidemiology Unit -"Bambino Gesù" Children's Hospital IRCCS, Rome, Italy
| | - Ludovica Martini
- Neonatal Intensive Care Unit - "Bambino Gesù" Children's Hospital IRCCS, Rome, Italy
| | - Eleonora Di Tommaso
- Neonatal Intensive Care Unit - Mediterranean Pediatric Cardiology Center, San Vincenzo Hospital, Taormina, Italy
| | - Paola Bernaschi
- Microbiology Unit - "Bambino Gesù" Children's Hospital IRCCS, Rome, Italy
| | - Emanuel Paionni
- Clinical Chemistry Laboratory Unit - "Bambino Gesù" Children's Hospital IRCCS, Rome, Italy
| | - Ottavia Porzio
- Clinical Chemistry Laboratory Unit - "Bambino Gesù" Children's Hospital IRCCS, Rome, Italy
- Department of Experimental Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Fiammetta Piersigilli
- Neonatal Intensive Care Unit, Cliniques Universitaires Saint Luc, Université Catholique de Louvain, Bruxelles, Belgium
| | - Marco Iannetta
- Tor Vergata University Hospital, Infectious Disease Clinic, Department of Systems Medicine, Tor Vergata University, Rome, Italy
| | - Andrea Dotta
- Neonatal Intensive Care Unit - "Bambino Gesù" Children's Hospital IRCCS, Rome, Italy
| | - Maria Paola Ronchetti
- Neonatal Intensive Care Unit - "Bambino Gesù" Children's Hospital IRCCS, Rome, Italy
| |
Collapse
|
4
|
Fox Á, Doyle E, Reynolds A, Geary M, Cuzzilla R, Hayes B. Placental histology for infants with hypoxic ischaemic encephalopathy compared with healthy controls: a case-control study. BMC Pediatr 2025; 25:294. [PMID: 40229702 PMCID: PMC11995492 DOI: 10.1186/s12887-025-05652-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 03/31/2025] [Indexed: 04/16/2025] Open
Abstract
BACKGROUND The role of the placenta in the development of hypoxic ischaemic encephalopathy (HIE) remains undefined. There is limited research comparing placental histology for infants with HIE and healthy controls. This is limiting our ability to understand its role in HIE. This study hypothesised that placental pathology is more common in infants with HIE compared with healthy infants and aimed to report the differences in placental histology between infants with HIE and healthy controls. METHODS A case-control study of infants with moderate andsevere HIE and healthy controls at a single tertiary Neonatal Intensive Care Unit. Placental histology was reviewed by one perinatal histopathologist using consensus guidelines. RESULTS Seventy-four cases and 98 controls were included. Cases had a higher incidence of pathology, including fetal vascular malperfusion, histological chorioamnionitis and delayed villous maturation. CONCLUSION This study demonstrates a higher incidence of placental pathology for infants born with HIE suggesting that the placenta is an important factor in the pathogenesis of HIE. Further research is required to delineate this relationship.
Collapse
Affiliation(s)
- Áine Fox
- Royal College of Surgeons (RCSI), Dublin, Ireland.
- Department of Neonatology, Rotunda Hospital, Dublin, Ireland.
| | - Emma Doyle
- Department of Pathology, Rotunda Hospital, Dublin, Ireland
| | | | - Michael Geary
- Royal College of Surgeons (RCSI), Dublin, Ireland
- Department of Obstetrics, Rotunda Hospital, Dublin, Ireland
| | - Rocco Cuzzilla
- Royal College of Surgeons (RCSI), Dublin, Ireland
- Newborn Research and Neonatal Services, The Royal Women's Hospital, Melbourne, Australia
- Department of Obstetrics, Gynaecology and Newborn Health, The University of Melbourne, Melbourne, Australia
| | - Breda Hayes
- Royal College of Surgeons (RCSI), Dublin, Ireland
- Department of Neonatology, Rotunda Hospital, Dublin, Ireland
| |
Collapse
|
5
|
Bitsadze V, Lazarchuk A, Vorobev A, Khizroeva J, Tretyakova M, Makatsariya N, Gashimova N, Grigoreva K, Tatarintseva A, Karpova A, Mostovoi A, Zainulina M, Kapanadze D, Blbulyan A, Kuneshko N, Gris JC, Elalamy I, Gerotziafas G, Makatsariya A. Systemic Inflammatory Response Syndrome, Thromboinflammation, and Septic Shock in Fetuses and Neonates. Int J Mol Sci 2025; 26:3259. [PMID: 40244141 PMCID: PMC11989690 DOI: 10.3390/ijms26073259] [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: 01/24/2025] [Revised: 03/24/2025] [Accepted: 03/28/2025] [Indexed: 04/18/2025] Open
Abstract
This article explores systemic inflammatory response syndrome (SIRS), thromboinflammation, and septic shock in fetuses and neonates, offering a comprehensive examination of their pathophysiology, diagnostic criteria, and clinical implications. It identifies SIRS as an exaggerated response to external stress, disrupting the balance between inflammation and adaptive mechanisms, driven by cytokines such as TNF-α and IL-1. The fetal inflammatory response syndrome (FIRS), a subset of SIRS, is noted for its role in adverse neonatal outcomes, including organ damage, inflammation, and long-term developmental disorders. The article discusses the extensive effects of FIRS on critical systems, including the blood, lungs, central nervous system, and kidneys. It highlights the challenges in diagnosing and managing septic shock in neonates, focusing on the relationship between inflammation and the hemostatic system. Additionally, the paper points out recent advancements, such as the convergent model of coagulation and emerging biomarkers like microRNAs for early detection. Despite this progress, gaps remain in understanding the molecular mechanisms underlying these conditions and in developing effective therapeutic strategies. This highlights the necessity for targeted research to mitigate the morbidity and mortality associated with septic shock in neonates.
Collapse
Affiliation(s)
- Victoria Bitsadze
- Department of Obstetrics, Gynecology and Perinatal Medicine, The I.M. Sechenov First Moscow State Medical University (Sechenov University), Trubetskaya Str 8-2, Moscow 119435, Russia; (V.B.); (A.L.); (A.V.); (M.T.); (N.M.); (N.G.); (K.G.); (A.T.); (J.-C.G.); (I.E.); (G.G.); (A.M.)
| | - Arina Lazarchuk
- Department of Obstetrics, Gynecology and Perinatal Medicine, The I.M. Sechenov First Moscow State Medical University (Sechenov University), Trubetskaya Str 8-2, Moscow 119435, Russia; (V.B.); (A.L.); (A.V.); (M.T.); (N.M.); (N.G.); (K.G.); (A.T.); (J.-C.G.); (I.E.); (G.G.); (A.M.)
| | - Alexander Vorobev
- Department of Obstetrics, Gynecology and Perinatal Medicine, The I.M. Sechenov First Moscow State Medical University (Sechenov University), Trubetskaya Str 8-2, Moscow 119435, Russia; (V.B.); (A.L.); (A.V.); (M.T.); (N.M.); (N.G.); (K.G.); (A.T.); (J.-C.G.); (I.E.); (G.G.); (A.M.)
| | - Jamilya Khizroeva
- Department of Obstetrics, Gynecology and Perinatal Medicine, The I.M. Sechenov First Moscow State Medical University (Sechenov University), Trubetskaya Str 8-2, Moscow 119435, Russia; (V.B.); (A.L.); (A.V.); (M.T.); (N.M.); (N.G.); (K.G.); (A.T.); (J.-C.G.); (I.E.); (G.G.); (A.M.)
| | - Maria Tretyakova
- Department of Obstetrics, Gynecology and Perinatal Medicine, The I.M. Sechenov First Moscow State Medical University (Sechenov University), Trubetskaya Str 8-2, Moscow 119435, Russia; (V.B.); (A.L.); (A.V.); (M.T.); (N.M.); (N.G.); (K.G.); (A.T.); (J.-C.G.); (I.E.); (G.G.); (A.M.)
| | - Natalia Makatsariya
- Department of Obstetrics, Gynecology and Perinatal Medicine, The I.M. Sechenov First Moscow State Medical University (Sechenov University), Trubetskaya Str 8-2, Moscow 119435, Russia; (V.B.); (A.L.); (A.V.); (M.T.); (N.M.); (N.G.); (K.G.); (A.T.); (J.-C.G.); (I.E.); (G.G.); (A.M.)
| | - Nilufar Gashimova
- Department of Obstetrics, Gynecology and Perinatal Medicine, The I.M. Sechenov First Moscow State Medical University (Sechenov University), Trubetskaya Str 8-2, Moscow 119435, Russia; (V.B.); (A.L.); (A.V.); (M.T.); (N.M.); (N.G.); (K.G.); (A.T.); (J.-C.G.); (I.E.); (G.G.); (A.M.)
| | - Kristina Grigoreva
- Department of Obstetrics, Gynecology and Perinatal Medicine, The I.M. Sechenov First Moscow State Medical University (Sechenov University), Trubetskaya Str 8-2, Moscow 119435, Russia; (V.B.); (A.L.); (A.V.); (M.T.); (N.M.); (N.G.); (K.G.); (A.T.); (J.-C.G.); (I.E.); (G.G.); (A.M.)
| | - Alena Tatarintseva
- Department of Obstetrics, Gynecology and Perinatal Medicine, The I.M. Sechenov First Moscow State Medical University (Sechenov University), Trubetskaya Str 8-2, Moscow 119435, Russia; (V.B.); (A.L.); (A.V.); (M.T.); (N.M.); (N.G.); (K.G.); (A.T.); (J.-C.G.); (I.E.); (G.G.); (A.M.)
| | - Anna Karpova
- Vorokhobov City Clinical Hospital No 67, Moscow Healthcare Department, 2/44 Salyama Adilya Str., Moscow 123423, Russia; (A.K.); (A.M.)
- Russian Medical Academy of Continuous Professional Education, Health Ministry of Russian Federation, 2/1 bldg. 1, Barrikadnaya Str., Moscow 123993, Russia
- Department of Polyclinic Therapy, Clinical Laboratory Diagnostics and Medical Biochemistry of Institute of Postgraduate Education of Yaroslavl State Medical University, Yaroslavl State Medical University, Health Ministry of Russian Federation, 5 Revolutsionnaya Str., Yaroslavl 150000, Russia
| | - Aleksei Mostovoi
- Vorokhobov City Clinical Hospital No 67, Moscow Healthcare Department, 2/44 Salyama Adilya Str., Moscow 123423, Russia; (A.K.); (A.M.)
- Russian Medical Academy of Continuous Professional Education, Health Ministry of Russian Federation, 2/1 bldg. 1, Barrikadnaya Str., Moscow 123993, Russia
- Department of Polyclinic Therapy, Clinical Laboratory Diagnostics and Medical Biochemistry of Institute of Postgraduate Education of Yaroslavl State Medical University, Yaroslavl State Medical University, Health Ministry of Russian Federation, 5 Revolutsionnaya Str., Yaroslavl 150000, Russia
| | - Marina Zainulina
- Snegirev Maternity Hospital No 6, 5 Mayakovskogo Str., Saint Petersburg 192014, Russia;
- Department of Obstetrics, Gynecology and Reproductology of Pavlov First Saint Petersburg State Medical University, Pavlov First Saint Petersburg State Medical University, Health Ministry of Russian Federation, 6/8 Lev Tolstoy Str., Saint Petersburg 197022, Russia
| | - Daredzhan Kapanadze
- Center of Pathology of Pregnancy and Hemostasis «Medlabi», 340112 Tbilisi, Georgia;
| | - Armen Blbulyan
- Research Center of Maternal and Child Health Protection, 22 Mashtots Avenue, Yerevan 0002, Armenia;
| | - Nart Kuneshko
- Moscow’s Region Odintsovo Maternity Hospital, Odintsovo 143003, Russia;
| | - Jean-Christophe Gris
- Department of Obstetrics, Gynecology and Perinatal Medicine, The I.M. Sechenov First Moscow State Medical University (Sechenov University), Trubetskaya Str 8-2, Moscow 119435, Russia; (V.B.); (A.L.); (A.V.); (M.T.); (N.M.); (N.G.); (K.G.); (A.T.); (J.-C.G.); (I.E.); (G.G.); (A.M.)
- Faculty of Pharmaceutical and Biological Sciences, Montpellier University, 34093 Montpellier, France
| | - Ismail Elalamy
- Department of Obstetrics, Gynecology and Perinatal Medicine, The I.M. Sechenov First Moscow State Medical University (Sechenov University), Trubetskaya Str 8-2, Moscow 119435, Russia; (V.B.); (A.L.); (A.V.); (M.T.); (N.M.); (N.G.); (K.G.); (A.T.); (J.-C.G.); (I.E.); (G.G.); (A.M.)
- Faculté Privee de Médecine de Marrakech (FPMM), Route Amizmiz, Marrakech 42312, Morocco
- Hopital Americain de Paris, 55 rue du Château, Neuilly Sur Seine, 92200 Paris, France
| | - Grigoris Gerotziafas
- Department of Obstetrics, Gynecology and Perinatal Medicine, The I.M. Sechenov First Moscow State Medical University (Sechenov University), Trubetskaya Str 8-2, Moscow 119435, Russia; (V.B.); (A.L.); (A.V.); (M.T.); (N.M.); (N.G.); (K.G.); (A.T.); (J.-C.G.); (I.E.); (G.G.); (A.M.)
- INSERM UMR_S_938, Saint-Antoine Research Center (CRSA), Team “Cancer Biology and Therapeutics”, Group “Cancer—Angiogenesis—Thrombosis”, University Institute of Cancerology (UIC), Sorbonne University, 34 Rue du Crozatier, 75012 Paris, France
- Thrombosis Center, Tenon—Saint Antoine University Hospital, Hôpitaux Universitaires Est Parisien, Assitance Publique Hôpitaix de Paris (AP-HP), 4 Rue de la Chine, 75020 Paris, France
| | - Alexander Makatsariya
- Department of Obstetrics, Gynecology and Perinatal Medicine, The I.M. Sechenov First Moscow State Medical University (Sechenov University), Trubetskaya Str 8-2, Moscow 119435, Russia; (V.B.); (A.L.); (A.V.); (M.T.); (N.M.); (N.G.); (K.G.); (A.T.); (J.-C.G.); (I.E.); (G.G.); (A.M.)
| |
Collapse
|
6
|
Kathuria S, Gupta A, Tracy AR, Luna Ramirez RI, Thulasingam SK, Zaghloul N, Ahmed M, Limesand SW. Systemic inflammatory responses to repeated and increasing endotoxin challenges in fetal sheep. Physiol Rep 2025; 13:e70316. [PMID: 40268878 PMCID: PMC12018166 DOI: 10.14814/phy2.70316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Revised: 03/28/2025] [Accepted: 03/28/2025] [Indexed: 04/25/2025] Open
Abstract
Repeated low-dose administration of lipopolysaccharide (LPS) attenuates subsequent fetal responses, which makes it challenging to investigate interventions to prolonged exposure. Our aim was to develop a fetal inflammatory response syndrome (FIRS) model that consistently and effectively elicits a marked physiological response to increasing LPS doses. Four intravenous LPS boluses (0.3, 1.5, 3, and 15 μg) were administered to fetal sheep over 5 days. Physiological responses were measured via blood gases, pH, lactate, and cortisol concentrations. Fetal peripheral blood mononuclear cells (PBMCs) were analyzed for transcriptomic changes and tissue cytokine expression postmortem. All LPS challenges increased lactate, cortisol, and pCO2 concentrations and decreased pH and pO2 levels at 3 and 5 hours. No interaction was found between day (increasing LPS doses) and hour (LPS response to each dose). PBMC numbers increase with LPS challenges. Transcriptional analysis on PBMCs identified several enriched gene clusters indicating upregulation of inflammatory gene signatures along with complement activation and NFκB signaling pathways. Expression of pro-inflammatory cytokines (TNFα, IL-6, or IL-1β) was measured in lung, heart, liver, placenta, and spleen. Physiological indices show both respiratory and metabolic acidosis with successive and increasing LPS challenges that demonstrate a robust systemic response despite tachyphylaxis to LPS in fetal sheep.
Collapse
Affiliation(s)
- Sanya Kathuria
- School of Animal and Comparative Biomedical SciencesUniversity of ArizonaTucsonArizonaUSA
| | - Akash Gupta
- Division of Neonatology, Department of Pediatrics, College of MedicineUniversity of ArizonaTucsonArizonaUSA
| | - Ayna R. Tracy
- School of Animal and Comparative Biomedical SciencesUniversity of ArizonaTucsonArizonaUSA
| | - Rosa I. Luna Ramirez
- School of Animal and Comparative Biomedical SciencesUniversity of ArizonaTucsonArizonaUSA
| | - Senthil Kumar Thulasingam
- Division of Neonatology, Department of Pediatrics, College of MedicineUniversity of ArizonaTucsonArizonaUSA
| | - Nahla Zaghloul
- Division of Neonatology, Department of Pediatrics, College of MedicineUniversity of ArizonaTucsonArizonaUSA
- Division of Neonatology, Department of Pediatrics, College of MedicineUniversity of FloridaGainesvilleFloridaUSA
| | - Mohamed Ahmed
- Division of Neonatology, Department of Pediatrics, College of MedicineUniversity of ArizonaTucsonArizonaUSA
- Division of Neonatology, Department of Pediatrics, College of MedicineUniversity of FloridaGainesvilleFloridaUSA
| | - Sean W. Limesand
- School of Animal and Comparative Biomedical SciencesUniversity of ArizonaTucsonArizonaUSA
| |
Collapse
|
7
|
Zeming KK, Llanora G, Quek K, Goh CR, Ng NZH, Han J, Yeo KT. Whole blood biophysical immune profiling of newborn infants correlates with immune responses. Pediatr Res 2025:10.1038/s41390-025-03952-y. [PMID: 40164874 DOI: 10.1038/s41390-025-03952-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Accepted: 02/10/2025] [Indexed: 04/02/2025]
Abstract
BACKGROUND There is a current, absence of reliable, blood-sparing, diagnostic tools to measure and trend real-time changes in the levels of inflammation and its effects on the immune cells in the infant. METHODS We deployed the BiophysicaL Immune Profiling for Infants (BLIPI) system in the neonatal intensive care unit to describe immune cell biophysical profiles using 50 microliters of blood per sample from term and preterm infants. RESULTS A total of 19 infants (8 term, 11 preterm) were recruited and 24 blood samples were collected in their first month. Based on the profiles of immune cells' size and deformation, there was a clear distinction between term and preterm infants, with 48/50 markers significantly different. A preterm infant with late-onset bacterial sepsis had notable size and deformability differences compared to the rest of the preterm cohort. There was a significant correlation between immune cell biophysical profiles and clinical markers such as C-reactive protein, white blood cell counts, and immature-to-total neutrophil (I:T) ratios, with Pearson correlation coefficients for linear regression models of 0.98, 0.97 and 0.94 respectively. CONCLUSION This study highlights the potential for the biophysical immune cell profiling system to provide an overview of the infant's current immune activation and response. IMPACT We present a novel, minimally invasive diagnostic system that leverages the physical properties of immune cells to provide a rapid and direct assessment of the immune status, requiring 20 times less blood volume than standard tests. This study demonstrates the potential of a compact, deployable system that is capable of performing biophysical profiling to assess immune cell activation in term and preterm infants, by revealing distinct differences in cell size and deformation between groups. The system's sensitive, quantitative measures were correlated with routine clinical biomarkers, highlighting its ability to provide a rapid, minimally invasive, real-time monitoring of neonatal immune status.
Collapse
Affiliation(s)
- Kerwin Kwek Zeming
- Critical Analytics for Manufacturing of Personalized Medicine, Singapore-MIT Alliance for Research and Technology, Singapore, Singapore
| | - Genevieve Llanora
- Department of Neonatology, KK Women's & Children's Hospital, Singapore, Singapore
| | - Kaiyun Quek
- Critical Analytics for Manufacturing of Personalized Medicine, Singapore-MIT Alliance for Research and Technology, Singapore, Singapore
| | - Chin Ren Goh
- Critical Analytics for Manufacturing of Personalized Medicine, Singapore-MIT Alliance for Research and Technology, Singapore, Singapore
| | - Nicholas Zhi Heng Ng
- Critical Analytics for Manufacturing of Personalized Medicine, Singapore-MIT Alliance for Research and Technology, Singapore, Singapore
| | - Jongyoon Han
- Critical Analytics for Manufacturing of Personalized Medicine, Singapore-MIT Alliance for Research and Technology, Singapore, Singapore.
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Electrical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Antimicrobial Resistance, Singapore-MIT Alliance for Research and Technology, Singapore, Singapore.
| | - Kee Thai Yeo
- Department of Neonatology, KK Women's & Children's Hospital, Singapore, Singapore.
- Duke-NUS Medical School, Singapore, Singapore.
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
8
|
Matasariu DR, Condac C, Bîrluțiu V, Lozneanu L, Bujor IE, Boiculese VL, Sava M, Ursache A. Placental Molecular Expression of Different Pathogenic Vaginal Infections. Int J Mol Sci 2025; 26:2863. [PMID: 40243431 PMCID: PMC11988318 DOI: 10.3390/ijms26072863] [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: 02/18/2025] [Revised: 03/19/2025] [Accepted: 03/19/2025] [Indexed: 04/18/2025] Open
Abstract
This study evaluated the differential expression of four placental markers-vitamin D receptor (VDR), Cluster of Differentiation 44 (CD44), osteopontin (OPN), and cyclooxygenase-2 (COX-2)-in response to pathogens, which may contribute to our understanding of pathogen-specific impacts on pregnancy outcomes. We immunohistochemically (IHC) analyzed placental tissues obtained from 70 healthy-term pregnant women in the control group and compared them to tissues obtained from 78 women with pregnancy above 24 weeks of gestation, single-pathogen vaginal infection, and premature rupture of membranes/preterm premature rupture of membranes (PROM/PPROM). We detected high expression of these four molecules in cases of Group B Streptococcus (GBS) and Ureaplasma urealyticum vaginal infections, and moderate expression in cases of Enterobacteriaceae infections, except for Klebsiella; the cases with Klebsiella and Candida species (spp.) vaginitis exhibited a lower expression compared to the healthy control group. VDR, CD44, and OPN had increased placental expression in GBS and Ureaplasma urealyticum vaginal infections; the opportunistic pathogenicity of both Escherichia coli and Candida spp. explains their low IHC positivity, and the tremendous ability of Gram-negative bacteria to elude the host immunity is revealed by the negative IHC staining in cases of Klebsiella vaginitis. These findings suggest that pathogen-specific alterations in the expression of these markers may contribute to the differential risk stratification of pregnancy complications and may mitigate the risks of adverse maternal and fetal outcomes. Interventions aiming to modulate these pathways might improve pregnancy outcomes.
Collapse
Affiliation(s)
- Daniela Roxana Matasariu
- Department of Obstetrics and Gynecology, “Cuza Voda” Hospital, 700038 Iasi, Romania;
- Department of Mother and Child, University of Medicine and Pharmacy “Gr. T. Popa”, 700115 Iasi, Romania; (I.E.B.); (A.U.)
| | - Constantin Condac
- Department of Anesthesia and Intensive Care, “Cuza Voda” Hospital, 700038 Iasi, Romania;
- Department of Infectious Diseases, University of Medicine and Pharmacy “Lucian Blaga”, 550169 Sibiu, Romania;
| | - Victoria Bîrluțiu
- Department of Infectious Diseases, University of Medicine and Pharmacy “Lucian Blaga”, 550169 Sibiu, Romania;
| | - Ludmila Lozneanu
- Department of Morpho-Functional Sciences I—Histology, University of Medicine and Pharmacy “Gr. T. Popa”, 700115 Iasi, Romania
| | - Iuliana Elena Bujor
- Department of Mother and Child, University of Medicine and Pharmacy “Gr. T. Popa”, 700115 Iasi, Romania; (I.E.B.); (A.U.)
| | - Vasile Lucian Boiculese
- Biostatistics, Department of Preventive Medicine and Interdisciplinarity, University of Medicine and Pharmacy “Gr. T. Popa”, 700115 Iasi, Romania;
| | - Mihai Sava
- Department of Anesthesia and Intensive Care, University of Medicine and Pharmacy “Lucian Blaga”, 550169 Sibiu, Romania;
| | - Alexandra Ursache
- Department of Mother and Child, University of Medicine and Pharmacy “Gr. T. Popa”, 700115 Iasi, Romania; (I.E.B.); (A.U.)
| |
Collapse
|
9
|
Kumasaka S, Negishi Y, Morita R, Migita M, Shima Y. Immunological role of zinc in preterm neonates. Immunol Med 2025; 48:78-88. [PMID: 39450996 DOI: 10.1080/25785826.2024.2420426] [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: 04/08/2024] [Accepted: 10/14/2024] [Indexed: 10/26/2024] Open
Abstract
Zinc (Zn), an essential trace element, plays a significant role in fetal development and biological defense during the embryonic and neonatal periods. Therefore, exploring the kinetics of Zn related to immune disturbances in preterm neonates is important. We here performed the measurement of Zn concentration along with immunological analysis of neonates and investigated the role of Zn in the neonatal period. Serum Zn concentrations were measured immediately after birth in neonates (329 cases). Moreover, for 25 cases, the kinetics of various immune cells and cytokines were measured by flow cytometry and electrochemiluminescence. We observed that Zn levels were inversely correlated with gestational weeks. Immune cell and cytokine analysis revealed an inverse correlation between HLA-DR on monocytes and Zn levels and between inflammatory cytokine interleukin-12 and Zn levels. Furthermore, oxidative stress status was inversely correlated with Zn levels. Our results suggested that the Zn dynamics immediately after birth, which show a negative correlation with the gestational week, can provide an anti-inflammatory and anti-oxidative environment for preterm neonates. The increased Zn concentration in the blood of preterm neonates may consequently protect neonates from perinatal stress.
Collapse
Affiliation(s)
- Sakae Kumasaka
- Department of Pediatrics, Japan Red Cross Tokyo Katsushika Perinatal Center, Tokyo, Japan
- Department of Pediatrics, Nippon Medical School, Tokyo, Japan
| | - Yasuyuki Negishi
- Department of Microbiology and Immunology, Nippon Medical School, Tokyo, Japan
- Department of Obstetrics and Gynecology, Nippon Medical School, Tokyo, Japan
| | - Rimpei Morita
- Department of Microbiology and Immunology, Nippon Medical School, Tokyo, Japan
| | - Makoto Migita
- Department of Pediatrics, Nippon Medical School, Tokyo, Japan
- Department of Pediatrics, Nippon Medical School Musashikosugi Hospital, Kanagawa, Japan
| | - Yoshio Shima
- Department of Pediatrics, Nippon Medical School, Tokyo, Japan
- Department of Neonatal Medicine, Nippon Medical School Musashikosugi Hospital, Kanagawa, Japan
| |
Collapse
|
10
|
Yuan J, Wu Y, Zhang Y, Zeng L, Zhou J, Piao M, Tong X, Wei Y, Cui L, Han T. Diagnostic Value of Umbilical Cord Blood Interleukin-6 Level in Premature Infants with Early-Onset Sepsis. CHILDREN (BASEL, SWITZERLAND) 2025; 12:301. [PMID: 40150583 PMCID: PMC11941668 DOI: 10.3390/children12030301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2025] [Revised: 02/25/2025] [Accepted: 02/26/2025] [Indexed: 03/29/2025]
Abstract
OBJECTIVE Early-onset sepsis (EOS) is a serious, grave, and frequently fatal condition in premature infants. This study aimed to assess the diagnostic value of interleukin-6 (IL-6) levels in umbilical cord blood for identifying EOS in preterm infants. METHODS This prospective cohort study was conducted on preterm infants between May 2019 and April 2021. Based on the diagnostic criteria for EOS, the participants were divided into EOS and non-EOS groups. Receiver operating characteristic (ROC) curve analysis was performed to evaluate the diagnostic efficacy of cord blood IL-6 levels for EOS. RESULTS The levels of IL-6 were significantly higher in the EOS group (n = 10) compared to the non-EOS group (n = 178) [617.5 pg/mL (323.3, 1579.8) vs. 49.7 pg/mL (15.8, 142.8), respectively; p = 0.000]. ROC curve analysis demonstrated that a cutoff value of 250.5 pg/mL for cord blood IL-6 yielded a sensitivity of 90%, specificity of 82%, and area under the curve of 0.876, with a confidence interval of 0.753-0.999, indicating its high accuracy as a diagnostic marker for EOS among preterm infants (p < 0.001). CONCLUSIONS The detection of IL-6 in the umbilical cord blood offers convenience and exhibits significant diagnostic potential for EOS in preterm infants, thereby providing valuable support for clinical decision-making.
Collapse
Affiliation(s)
- Jinfang Yuan
- Department of Pediatric, Peking University Third Hospital, Beijing 100191, China; (J.Y.); (Y.Z.); (M.P.); (X.T.)
| | - Yufeng Wu
- Clinical Laboratory, Peking University Third Hospital, Beijing 100191, China; (Y.W.); (J.Z.); (L.C.)
| | - Yahui Zhang
- Department of Pediatric, Peking University Third Hospital, Beijing 100191, China; (J.Y.); (Y.Z.); (M.P.); (X.T.)
| | - Lin Zeng
- Research Center of Clinical Epidemiology, Peking University Third Hospital, Beijing 100191, China;
| | - Jiansuo Zhou
- Clinical Laboratory, Peking University Third Hospital, Beijing 100191, China; (Y.W.); (J.Z.); (L.C.)
| | - Meihua Piao
- Department of Pediatric, Peking University Third Hospital, Beijing 100191, China; (J.Y.); (Y.Z.); (M.P.); (X.T.)
| | - Xiaomei Tong
- Department of Pediatric, Peking University Third Hospital, Beijing 100191, China; (J.Y.); (Y.Z.); (M.P.); (X.T.)
| | - Yuan Wei
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing 100191, China;
| | - Liyan Cui
- Clinical Laboratory, Peking University Third Hospital, Beijing 100191, China; (Y.W.); (J.Z.); (L.C.)
| | - Tongyan Han
- Department of Pediatric, Peking University Third Hospital, Beijing 100191, China; (J.Y.); (Y.Z.); (M.P.); (X.T.)
| |
Collapse
|
11
|
Hu L, Sun Q, Liu Z, Huang H, Zhao E, Chen H, Wu Y, Ge Y, Ouyang D, Tang B. Structural Characterization of APSN from Astragalus membranaceus and Its Potential Therapeutic Effect on Immune Dysregulation and Tissue Damage. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:4042-4054. [PMID: 39918058 DOI: 10.1021/acs.jafc.4c08632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/20/2025]
Abstract
Addressing the global health impact of inflammatory and immune-mediated diseases, this study focused on purifying and characterizing a neutral polysaccharide, APSN, from the Astragalus membranaceus. Employing high-performance gel permeation chromatography (HPGPC), Fourier-transform infrared spectroscopy (FTIR), and nuclear magnetic resonance (NMR) spectroscopy, we elucidated APSN's structural features, revealing a highly branched glucan with a 1,4-α-d-glucopyranosyl main chain and side chains at the O-6 position. Separately, we assessed APSN's biological activity, finding that it significantly modulates immune responses by inhibiting the NF-κB pathway in RAW264.7 macrophages and promotes endothelial cell proliferation and angiogenesis-related gene expression in HUVECs. These results position APSN as a potential therapeutic for diseases characterized by immune dysregulation and tissue damage, warranting further investigation of its mechanisms and clinical efficacy.
Collapse
Affiliation(s)
- Liqiu Hu
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Qili Sun
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Zhanpeng Liu
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Hanwei Huang
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Enze Zhao
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Haotian Chen
- Department of Material Science and Engineering, Columbia University, New York, New York 10027, United States
| | - Yuxun Wu
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Yongmei Ge
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Dongfang Ouyang
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
- Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129, United States
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, Ontario M5S 3G8, Canada
| | - Bin Tang
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
- Guangdong Provincial Key Laboratory of Advanced Biomaterial, Shenzhen, Guangdong 518055, China
| |
Collapse
|
12
|
Bohm EK, Castañeda D, Lu Q, Cameron MD, Aliota MT. Zika virus-induced fetal demise is driven by strain- and dose-specific RLR-driven activation of the interferon response in the decidua, placenta, and fetus in Ifnar1 -/- mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.12.637947. [PMID: 39990459 PMCID: PMC11844458 DOI: 10.1101/2025.02.12.637947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
Congenital Zika syndrome (CZS), the set of fetal and neonatal complications associated with Zika virus (ZIKV) infection in pregnancy, was first noted during the outbreak in the Americas in 2015-16. However, there was an unequal distribution of ZIKV cases and severe outcomes in all areas where ZIKV emerged in the Americas, demonstrating that the risk of CZS varied over space and time. Recently, we demonstrated that phenotypic heterogeneity existed between closely-related ZIKV strains. All ZIKV strains tested infected the placenta but varied in their capacity to cause overt fetal harm. Here, we further characterized the relative contributions of virus genotype and infecting dose of two phenotypically distinct ZIKV strains across multiple timepoints in gestation in pregnant mice that lack type-I interferon receptor function ( Ifnar1 -/- ). To better understand the underlying causes of adverse fetal outcomes, we used RNA sequencing to compare ZIKV-infected and uninfected tissues. We found that ZIKV infection triggers retinoic acid-inducible gene I (RIG-I)-like receptor-mediated activation of the interferon response at the maternal-fetal interface. However, modest chemical inhibition of RIG-I activation in the decidua and placenta did not protect against fetal demise. Instead, the fetal interferon response was significantly associated with fetal demise. Together, these findings suggest that the response to ZIKV at the maternal-fetal interface can vary depending on the infecting ZIKV genotype and dose, and that the fetal immune response is an important mediator of fetal harm. IMPORTANCE Previously, we used a mouse model of ZIKV infection during pregnancy to assess the pathogenic potential to the fetus of a panel of five, low-passage ZIKV strains representing the viral genetic diversity in the Americas. We found that phenotypic heterogeneity existed between these closely-related ZIKV strains. Here, we show that this heterogeneity is driven by retinoic acid-inducible gene I (RIG-I)-like receptor-mediated activation of the interferon response at the maternal-fetal interface. We used chemical inhibition of the RIG-I pathway and measured the transcriptional activity of interferon stimulated genes in fetuses to demonstrate that the fetal immune response may contribute to fetal demise.
Collapse
|
13
|
Lanzarone V, Polkinghorne A, Eslick G, Branley J. Diagnostic tests for the prediction of histological chorioamnionitis and funisitis in pregnant women with preterm premature rupture of membranes: A systematic review. Aust N Z J Obstet Gynaecol 2025; 65:13-24. [PMID: 39132916 PMCID: PMC11924156 DOI: 10.1111/ajo.13864] [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: 05/29/2023] [Accepted: 07/09/2024] [Indexed: 08/13/2024]
Abstract
BACKGROUND Infection of the amniotic cavity is an important driver and/or consequence of preterm prelabour rupture of membranes (PPROM). Prediction of infection is challenging, limiting guidance for interventions during the antenatal period. Infection typically triggers a host inflammatory response, and non-invasive indirect markers of the maternal or fetal inflammatory response have been reported in the context of PPROM and intra-amniotic infection. Some of these markers have also been tested in amniotic fluid (AF) samples. AIMS This study compared markers of the inflammatory response in women with PPROM against the outcome standard of histological chorioamnionitis (HCA) or funisitis (FUS). METHODS Searches were conducted for studies reporting diagnostic test sensitivity and specificity for proven HCA or FUS in pregnant women with PPROM after 20 weeks' gestation. Weighted mean pooled sensitivity (Se), specificity (Sp), positive predictive value, negative predictive value, diagnostic odds ratio and 95% confidence intervals were calculated for each of the selected diagnostic tests. RESULTS Except ultrasonographic detection of fetal thymic involution, almost all index tests analysed showed relatively low sensitivity. Maternal white cell count, interleukin-6 (IL-6) and AF IL-6 had credible specificity. Testing of AF markers, while more consistent than serum markers, showed no clear diagnostic accuracy improvement. CONCLUSIONS There is a clear lack of evidence for the reliability of any individual diagnostic test to assist in the detection of HCA or FUS in women with PPROM. Combining several markers into a predictive model for improved diagnosis may be worth investigating.
Collapse
Affiliation(s)
- Valeria Lanzarone
- Christopher Kohlenberg Department of Perinatal UltrasoundNepean HospitalPenrithNew South WalesAustralia
| | - Adam Polkinghorne
- Department of Microbiology and Infectious DiseasesNew South Wales Health Pathology, Nepean Blue Mountains Pathology ServicePenrithNew South WalesAustralia
- Nepean Clinical School, Faculty of Medicine and HealthUniversity of SydneySydneyNew South WalesAustralia
| | - Guy Eslick
- Clinical Links Using Evidence‐based Data (CLUED) Pty LtdSydneyNew South WalesAustralia
| | - James Branley
- Department of Microbiology and Infectious DiseasesNew South Wales Health Pathology, Nepean Blue Mountains Pathology ServicePenrithNew South WalesAustralia
- Nepean Clinical School, Faculty of Medicine and HealthUniversity of SydneySydneyNew South WalesAustralia
| |
Collapse
|
14
|
Pereira S, Costa P, Oumar M, Garcia-Ruiz I. A pragmatic approach to recognize intrapartum chorioamnionitis. Eur J Obstet Gynecol Reprod Biol 2025; 305:199-203. [PMID: 39705989 DOI: 10.1016/j.ejogrb.2024.12.012] [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: 09/27/2024] [Revised: 11/18/2024] [Accepted: 12/08/2024] [Indexed: 12/23/2024]
Abstract
Chorioamnionitis is a significant contributor to non-hypoxic fetal compromise, increasing the risk of neonatal encephalopathy and cerebral palsy. This paper highlights the limitations of traditional diagnostic methods for chorioamnionitis, which rely on histopathology, microbiology, and clinical signs, and emphasizes the importance of recognizing the fetal inflammatory response using intrapartum cardiotocography (CTG). By understanding the physiological pathways of inflammation, clinicians can identify characteristic CTG patterns. This pragmatic approach allows for earlier detection and intervention, potentially improving perinatal outcomes. The paper calls for a re-evaluation of clinical management practices and advocates for a multifaceted approach that incorporates CTG findings into the diagnosis of chorioamnionitis to prevent adverse neonatal outcomes.
Collapse
Affiliation(s)
- Susana Pereira
- Fetal Medicine Unit, The Royal London Hospital, Barts Health NHS Trust, London, UK; Universitat Autònoma de Barcelona, Barcelona, Spain.
| | - Paulina Costa
- Department of Obstetrics and Gynecology, Unidade Local de Saúde do Médio Ave, Santo Tirso, Portugal
| | - Mohamed Oumar
- Fetal Medicine Unit, The Royal London Hospital, Barts Health NHS Trust, London, UK
| | - Itziar Garcia-Ruiz
- Maternal-Fetal Medicine Unit, Department of Obstetrics, Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| |
Collapse
|
15
|
Molloy EJ. Early immune modulation in cerebral palsy medical management of cerebral palsy series. Pediatr Res 2025:10.1038/s41390-025-03798-4. [PMID: 39863783 DOI: 10.1038/s41390-025-03798-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Revised: 11/20/2024] [Accepted: 11/20/2024] [Indexed: 01/27/2025]
Affiliation(s)
- E J Molloy
- Discipline of Paediatrics, Trinity College, the University of Dublin, Dublin, Ireland.
- Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland.
- Trinity Research in Childhood Centre (TRiCC), Trinity College Dublin, Dublin, Ireland.
- Paediatrics, Coombe Women and Infants University Hospital, Dublin, Ireland.
- Neonatology & Neurodisability, Childrens Health Ireland at Crumlin & Tallaght, Dublin, Ireland.
| |
Collapse
|
16
|
Wu HS, Huang HC, Chen IL. Analysis of Salivary Cytokines in Retinopathy of Prematurity. CHILDREN (BASEL, SWITZERLAND) 2025; 12:80. [PMID: 39857911 PMCID: PMC11764213 DOI: 10.3390/children12010080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 01/04/2025] [Accepted: 01/07/2025] [Indexed: 01/27/2025]
Abstract
BACKGROUND/OBJECTIVES This cohort study aimed to establish a correlation between salivary cytokines and retinopathy of prematurity (ROP) in premature neonates. Additionally, we sought to identify a minimally invasive method for cytokine detection in this population. METHODS We recruited premature neonates born at less than 34 weeks gestational age (GA), with no history of maternal or neonatal infections. Salivary samples were collected on their first (D1) and seventh (D7) days of life, and cytokine levels were measured using the MILLPLEXMAP Human multiplex assay. RESULTS A total of 125 neonates were included in the study, categorized into two groups based on the severity of ROP: None to Mild and Moderate to Severe. The salivary levels of interleukin (IL)-6, IL-8, vascular endothelial growth factor (VEGF), and tumor necrosis factor (TNF)-α on D1 and D7 were significantly higher in the Moderate to Severe ROP group compared to the None to Mild ROP group (p = 0.005, 0.004, 0.026, 0.018, 0.001, 0.007, 0.025, and 0.002, respectively). After adjusting for GA, the levels of IL-6 and VEGF on D7 were significantly elevated in the Moderate to Severe ROP group compared to the None to Mild ROP group (p = 0.024 and 0.016, respectively). CONCLUSIONS This study establishes a novel, non-invasive method for the early prediction of ROP in premature neonates by correlating salivary cytokine levels in early life with the subsequent development of ROP.
Collapse
Affiliation(s)
- Hwa-Shiu Wu
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, College of Medicine, Chang Gung University, Kaohsiung 83301, Taiwan; (H.-S.W.); (H.-C.H.)
| | - Hsin-Chun Huang
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, College of Medicine, Chang Gung University, Kaohsiung 83301, Taiwan; (H.-S.W.); (H.-C.H.)
- School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Linkou 33302, Taiwan
| | - I-Lun Chen
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, College of Medicine, Chang Gung University, Kaohsiung 83301, Taiwan; (H.-S.W.); (H.-C.H.)
- School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Linkou 33302, Taiwan
| |
Collapse
|
17
|
Murillo-Bravo C, Balcells-Solé J, Cobo T, Crispi Brillas F. Cardiac remodeling in patients with intra-amniotic infection and/or inflammation. Am J Obstet Gynecol 2025; 232:e16-e17. [PMID: 39151770 DOI: 10.1016/j.ajog.2024.08.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 08/08/2024] [Indexed: 08/19/2024]
Affiliation(s)
- Clara Murillo-Bravo
- 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, Universitat de Barcelona, Barcelona, Spain
| | - Judith Balcells-Solé
- 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
| | - 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, Universitat de Barcelona, Barcelona, Spain; Center for Biomedical Research on Rare Diseases, Instituto de Salud Carlos III, Madrid, Spain.
| | - Fàtima Crispi Brillas
- 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, Universitat de Barcelona, Barcelona, Spain; Center for Biomedical Research on Rare Diseases, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
18
|
van Otterdijk SD, Binder AM, Michels KB. Placental methylation and pro-inflammatory protein levels in cord blood. Placenta 2024; 158:231-239. [PMID: 39514934 DOI: 10.1016/j.placenta.2024.10.067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 10/28/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024]
Abstract
INTRODUCTION The neonates' inflammatory response may in part be shaped during development by the placental epigenome, but evidence is scarce. We investigated the association between placental DNA methylation and pro-inflammatory proteins in cord blood. METHODS A total of 249 mother-child dyads from the Harvard Epigenetic Birth Cohort were included in this study. Genome-wide methylation in placental DNA was assessed using the Illumina Human Methylation 450 Bead Chip array and verified by pyrosequencing. Cord blood inflammation markers assessed were interleukin-6, interleukin-8 and tumor necrosis factor α, intercellular adhesion molecule 1, serum amyloid A, and C-reactive protein. RESULTS We identified differential placental DNA methylation of three loci in the HIVEP3 gene shore region that were associated with TNFα protein levels in cord blood. TNFα levels were associated with mode of delivery, gestational age and birth order. Three other loci located in the open sea region of the BCL11B gene were associated with SAA protein levels in cord blood. SAA levels were associated with birthweight, gestational age, and infant sex. CONCLUSIONS Our results suggest a potential role for HIVEP3 and BCL11B placental DNA methylation in the acute immune response of the neonate. These immune markers were correlated with several mother and child characteristics.
Collapse
Affiliation(s)
- Sanne D van Otterdijk
- Institute for Prevention and Cancer Epidemiology, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany; Obstetrics and Gynecology Epidemiology Center, Department of Obstetrics, Gynecology and Reproductive Biology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Alexandra M Binder
- Department of Epidemiology, Fielding School of Public Health, University of California, Los Angeles, CA, USA; Cancer Epidemiology Program, University of Hawaii Cancer Center, Honolulu, HI, USA
| | - Karin B Michels
- Institute for Prevention and Cancer Epidemiology, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany; Obstetrics and Gynecology Epidemiology Center, Department of Obstetrics, Gynecology and Reproductive Biology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Epidemiology, Harvard School of Public Health, Boston, MA, USA; Department of Epidemiology, Fielding School of Public Health, University of California, Los Angeles, CA, USA.
| |
Collapse
|
19
|
Garcia-Flores V, Liu Z, Romero R, Pique-Regi R, Xu Y, Miller D, Levenson D, Galaz J, Winters AD, Farias-Jofre M, Panzer JJ, Theis KR, Gomez-Lopez N. Homeostatic Macrophages Prevent Preterm Birth and Improve Neonatal Outcomes by Mitigating In Utero Sterile Inflammation in Mice. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:1620-1634. [PMID: 39431882 PMCID: PMC11572957 DOI: 10.4049/jimmunol.2400467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 09/28/2024] [Indexed: 10/22/2024]
Abstract
Preterm birth (PTB), often preceded by preterm labor, is a major cause of neonatal morbidity and mortality worldwide. Most PTB cases involve intra-amniotic inflammation without detectable microorganisms, termed in utero sterile inflammation, for which there is no established treatment. In this study, we propose homeostatic macrophages to prevent PTB and adverse neonatal outcomes caused by in utero sterile inflammation. Single-cell atlases of the maternal-fetal interface revealed that homeostatic maternal macrophages are reduced with human labor. M2 macrophage treatment prevented PTB and reduced adverse neonatal outcomes in mice with in utero sterile inflammation. Specifically, M2 macrophages halted premature labor by suppressing inflammatory responses in the amniotic cavity, including inflammasome activation, and mitigated placental and offspring lung inflammation. Moreover, M2 macrophages boosted gut inflammation in neonates and improved their ability to fight systemic bacterial infections. Our findings show that M2 macrophages are a promising strategy to mitigate PTB and improve neonatal outcomes resulting from in utero sterile inflammation.
Collapse
Affiliation(s)
- Valeria Garcia-Flores
- Pregnancy 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, Detroit, MI and Bethesda, MD
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
- Center for Reproductive Health Sciences, Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, MO
| | - Zhenjie Liu
- Pregnancy 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, Detroit, MI and Bethesda, MD
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
| | - Roberto Romero
- Pregnancy 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, Detroit, MI and Bethesda, MD
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI
| | - Roger Pique-Regi
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI
| | - Yi Xu
- Pregnancy 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, Detroit, MI and Bethesda, MD
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
- Center for Reproductive Health Sciences, Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, MO
| | - Derek Miller
- Pregnancy 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, Detroit, MI and Bethesda, MD
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
- Center for Reproductive Health Sciences, Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, MO
| | - Dustyn Levenson
- Center for Reproductive Health Sciences, Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, MO
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI
| | - Jose Galaz
- Pregnancy 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, Detroit, MI and Bethesda, MD
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
- Division of Obstetrics and Gynecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Andrew D. Winters
- Pregnancy 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, Detroit, MI and Bethesda, MD
- Department of Biochemistry, Microbiology, and Immunology, Wayne State University School of Medicine, Detroit, MI
| | - Marcelo Farias-Jofre
- Pregnancy 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, Detroit, MI and Bethesda, MD
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
- Division of Obstetrics and Gynecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Jonathan J. Panzer
- Department of Biochemistry, Microbiology, and Immunology, Wayne State University School of Medicine, Detroit, MI
| | - Kevin R. Theis
- Pregnancy 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, Detroit, MI and Bethesda, MD
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
- Department of Biochemistry, Microbiology, and Immunology, Wayne State University School of Medicine, Detroit, MI
| | - Nardhy Gomez-Lopez
- Pregnancy 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, Detroit, MI and Bethesda, MD
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
- Center for Reproductive Health Sciences, Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, MO
- Department of Biochemistry, Microbiology, and Immunology, Wayne State University School of Medicine, Detroit, MI
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
20
|
Pongchaikul P, Romero R, Wongsurawat T, Jenjaroenpun P, Kruasuwan W, Mongkolsuk P, Vivithanaporn P, Thaipisuttikul I, Singsaneh A, Khamphakul J, Santanirand P, Kotchompoo K, Bhuwapathanapun M, Warintaksa P, Chaemsaithong P. Molecular evidence that GBS early neonatal sepsis results from ascending infection: comparative hybrid genomics analyses show that microorganisms in the vaginal ecosystem, amniotic fluid, chorioamniotic membranes, and neonatal blood are the same. J Perinat Med 2024; 52:977-990. [PMID: 39405032 PMCID: PMC11560570 DOI: 10.1515/jpm-2024-0310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 08/20/2024] [Indexed: 11/12/2024]
Abstract
OBJECTIVES Streptococcus agalactiae, or Group B Streptococcus (GBS), is a leading cause of neonatal sepsis. Materno-fetal transmission of the microorganisms present in the lower genital tract/perineum is considered to be the most frequent mode for acquisition of infection. It has also been proposed that, in a subset of cases, GBS causes acute chorioamnionitis, intraamniotic infection, and fetal/neonatal sepsis. However, the evidence to support this ascending pathway is derived from microbiologic studies that rely on cultivation methods, which do not have the resolution to determine if the microorganisms causing neonatal sepsis are the same as those found in the amniotic fluid and the vaginal ecosystem. METHODS We used whole genome sequencing of the microorganisms isolated from the vagina, amniotic fluid, chorioamniotic membranes, and neonatal blood (four isolates) in a case of early neonatal sepsis. Using hybrid genome assembly, we characterized the genomic features including virulence factors and antimicrobial resistance in four isolates from the same mother, placenta, and newborn. RESULTS Whole genome sequencing revealed that the microorganisms in the four clinical isolates corresponded to S. agalactiae sequence type 1, clonal complexes 1, and serotype Ib. Comparative genomic analysis illustrated similar DNA sequences of the four genomes. CONCLUSIONS This study presents the first evidence of the genomic similarity of microorganisms in the vaginal ecosystem, the space between the chorioamniotic membranes of the placenta, amniotic fluid, and neonatal blood.
Collapse
Affiliation(s)
- Pisut Pongchaikul
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital Mahidol University, Samut Prakan 10540, Thailand
- Integrative Computational BioScience Center, Mahidol University, Nakhon Pathom 73170, Thailand
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool L7 3EA, United Kingdom
| | - Roberto Romero
- Pregnancy 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, United States Department of Health and Human Services, Bethesda, Maryland 20892, USA
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, Michigan 48109, USA
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, Michigan 48824, USA
| | - Thidathip Wongsurawat
- Division of Medical Bioinformatics, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Piroon Jenjaroenpun
- Division of Medical Bioinformatics, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Worarat Kruasuwan
- Division of Medical Bioinformatics, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Paninee Mongkolsuk
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital Mahidol University, Samut Prakan 10540, Thailand
| | - Pornpun Vivithanaporn
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital Mahidol University, Samut Prakan 10540, Thailand
| | - Iyarit Thaipisuttikul
- Department of Microbiology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Arunee Singsaneh
- Department of Pathology, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand
| | - Jakkrit Khamphakul
- Department of Pathology, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand
| | - Pitak Santanirand
- Department of Pathology, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand
| | - Kanyaphat Kotchompoo
- Department of Obstetrics and Gynecology, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand
| | - Maolee Bhuwapathanapun
- Department of Obstetrics and Gynecology, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand
| | - Puntabut Warintaksa
- Department of Obstetrics and Gynecology, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand
| | - Piya Chaemsaithong
- Department of Obstetrics and Gynecology, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand
| |
Collapse
|
21
|
Obeagu EI, Obeagu GU. Neutrophil phenotypes in prolonged labor: Implications for therapeutic strategies. Medicine (Baltimore) 2024; 103:e40611. [PMID: 39560573 PMCID: PMC11576028 DOI: 10.1097/md.0000000000040611] [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: 12/19/2023] [Accepted: 11/01/2024] [Indexed: 11/20/2024] Open
Abstract
Prolonged labor, defined as labor extending beyond 20 hours for nulliparas and 14 hours for multiparas, poses significant risks to both maternal and neonatal health. The inflammatory response plays a crucial role in the pathophysiology of prolonged labor, with neutrophils being key players in this process. Neutrophils, the most abundant leukocytes, exhibit diverse phenotypes and functions in response to prolonged labor, influencing both the onset and progression of labor through their inflammatory actions. Classical neutrophils (N1) are involved in acute inflammatory responses, aiding in tissue remodeling and labor onset, but their prolonged activation can lead to tissue damage. Regulatory neutrophils (N2), which produce anti-inflammatory cytokines, help resolve inflammation and facilitate labor progression. Low-density granulocytes and aged neutrophils, associated with chronic inflammation and impaired function respectively, contribute to labor complications. The balance among these neutrophil phenotypes is crucial for maintaining a controlled inflammatory response during labor. Therapeutic strategies targeting neutrophil recruitment, NETosis, and cytokine production hold promise for managing prolonged labor. Modulating chemokine pathways, regulating NET formation, and balancing cytokine profiles may reduce inflammation and improve labor outcomes. Further research into the mechanisms of neutrophil regulation and the development of targeted therapies is essential for mitigating the adverse effects of prolonged labor and enhancing maternal and neonatal health.
Collapse
|
22
|
Lear BA, Zhou KQ, Dhillon SK, Lear CA, Bennet L, Gunn AJ. Preventive, rescue and reparative neuroprotective strategies for the fetus and neonate. Semin Fetal Neonatal Med 2024; 29:101542. [PMID: 39472238 DOI: 10.1016/j.siny.2024.101542] [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] [Indexed: 12/13/2024]
Abstract
Neonatal encephalopathy remains a major contributor to death and disability around the world. Acute hypoxia-ischaemia before, during or after birth creates a series of events that can lead to neonatal brain injury. Understanding the evolution of injury underpinned the development of therapeutic hypothermia. This review discusses the determinants of injury, including maturity, the pattern of exposure to HI, impaired placental function, often associated with fetal growth restriction and in the long-term, socio-economic deprivation. Chorioamnionitis has been associated with the presence of NE, but it is important to note that experimentally, inflammation can either sensitize to greater neural injury after HI or alleviate injury, depending on its precise timing. As fetal surveillance tools improve it is likely that improved detection of specific pathways will offer future opportunities for preventive and reparative interventions in utero and after birth.
Collapse
Affiliation(s)
- Benjamin A Lear
- Fetal Physiology and Neuroscience Group, Department of Physiology, The University of Auckland, Auckland, New Zealand
| | - Kelly Q Zhou
- Fetal Physiology and Neuroscience Group, Department of Physiology, The University of Auckland, Auckland, New Zealand
| | - Simerdeep K Dhillon
- Fetal Physiology and Neuroscience Group, Department of Physiology, The University of Auckland, Auckland, New Zealand
| | - Christopher A Lear
- Fetal Physiology and Neuroscience Group, Department of Physiology, The University of Auckland, Auckland, New Zealand
| | - Laura Bennet
- Fetal Physiology and Neuroscience Group, Department of Physiology, The University of Auckland, Auckland, New Zealand
| | - Alistair J Gunn
- Fetal Physiology and Neuroscience Group, Department of Physiology, The University of Auckland, Auckland, New Zealand.
| |
Collapse
|
23
|
Hernández-Rodríguez J, Pérez-Hernández J, Flores-Espinosa P, Olmos-Ortiz A, Velazquez P, Zamora-Escudero R, Islas-López M, Helguera-Repetto AC, Hernández-Bones K, Rodríguez-Flores S, Jiménez-Escutia R, Fortanel-Fonseca A, Flores-Pliego A, Lopez-Vancell R, Zaga-Clavellina V. Galectin-1 Elicits a Tissue-Specific Anti-Inflammatory and Anti-Degradative Effect Upon LPS-Induced Response in an Ex Vivo Model of Human Fetal Membranes Modeling an Intraamniotic Inflammation. Am J Reprod Immunol 2024; 92:e70016. [PMID: 39575516 PMCID: PMC11582940 DOI: 10.1111/aji.70016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 10/16/2024] [Accepted: 10/28/2024] [Indexed: 11/24/2024] Open
Abstract
PROBLEM Intrauterine infection is one of the most jeopardizing conditions associated with adverse outcomes, including preterm birth; however, multiple tolerance mechanisms operate at the maternal-fetal interface to avoid the rejection of the fetus. Among the factors that maintain the uterus as an immunoprivileged site, Galectin-1 (Gal-1), an immunomodulatory glycan-binding protein secreted by the maternal-fetal unit, is pivotal in promoting immune cell homeostasis. This work aimed to evaluate the role of Gal-1 during a lipopolysaccharide (LPS)-induced-inflammatory milieu. METHOD OF STUDY Using an ex vivo culture with two independent compartments, human fetal membranes at term were pretreated with 40 and 80 ng/mL of Gal-1, then to reproduce an intraamniotic inflammation, the fetal side of membranes was stimulated with 500 ng/mL of LPS for 24 h. The concentrations of tumor necrosis factor (TNF)-α, interleukin (IL)-1β, IL-6, monocyte chemoattractant protein (MCP1), macrophage inflammatory protein (MIP1) α, regulated upon activation normal T cell expressed and secreted (RANTES), and matrix metalloproteinase (MMP)-9 were measured in both amnion and choriodecidua compartments. RESULTS In a tissue-specific fashion profile, pretreatment with the physiologic concentration of Gal-1 significantly diminished the LPS-dependent secretion of TNF-α, IL-1β, Il-6, MCP1, MIP1α, RANTES, and MMP-9. CONCLUSION Gal-1 elicits an anti-inflammatory effect on the human fetal membranes stimulated with LPS, which supports the hypothesis that Gal-1 is part of the immunomodulatory mechanisms intended to stop the harmful effect of inflammation of the maternal-fetal interface.
Collapse
Affiliation(s)
- Jazmin Hernández-Rodríguez
- Departamento de Inmunobioquímica, Instituto Nacional de Perinatología, Ciudad de México, Mexico
- Laboratorio de Patología Experimental UME, Unidad de Medicina Experimental, Facultad de Medicina UNAM, Ciudad Universitaria, Ciudad de México, Mexico
| | - Jesús Pérez-Hernández
- Laboratorio de Patología Experimental UME, Unidad de Medicina Experimental, Facultad de Medicina UNAM, Ciudad Universitaria, Ciudad de México, Mexico
| | - Pilar Flores-Espinosa
- Departamento de Inmunobioquímica, Instituto Nacional de Perinatología, Ciudad de México, Mexico
| | - Andrea Olmos-Ortiz
- Departamento de Inmunobioquímica, Instituto Nacional de Perinatología, Ciudad de México, Mexico
| | - Pilar Velazquez
- Departamento de Ginecología y Obstetricia, Hospital Ángeles México, Ciudad de México, Mexico
| | | | - Marcela Islas-López
- Ginecología y Obstetricia, Hospital Ángeles Lomas-UNAM, Huixquilucan, Mexico
| | | | - Karla Hernández-Bones
- Departamento de Inmunobioquímica, Instituto Nacional de Perinatología, Ciudad de México, Mexico
- Posgrado en Ciencias Médicas, Escuela Superior de Medicina, Instituto Politécnico Nacional, Ciudad de México, Mexico
| | - Samara Rodríguez-Flores
- Departamento de Inmunobioquímica, Instituto Nacional de Perinatología, Ciudad de México, Mexico
- Posgrado en Ciencias Médicas, Escuela Superior de Medicina, Instituto Politécnico Nacional, Ciudad de México, Mexico
| | - Rodrigo Jiménez-Escutia
- Departamento de Inmunobioquímica, Instituto Nacional de Perinatología, Ciudad de México, Mexico
- Posgrado en Ciencias Biológicas, Unidad de Posgrado, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | | | - Arturo Flores-Pliego
- Departamento de Inmunobioquímica, Instituto Nacional de Perinatología, Ciudad de México, Mexico
| | - Rosario Lopez-Vancell
- Laboratorio de Patología Experimental UME, Unidad de Medicina Experimental, Facultad de Medicina UNAM, Ciudad Universitaria, Ciudad de México, Mexico
| | | |
Collapse
|
24
|
Jalkanen K, Virtanen A, Aittoniemi J, Flinck H, Ampuja S, Huhtala H, Tihtonen K. Novel Biomarkers: Soluble Urokinase-Type Plasminogen Activator Receptor and Procalcitonin- and Histological Chorioamnionitis after Preterm Premature Rupture of Membranes. Reprod Sci 2024; 31:3175-3182. [PMID: 39227528 PMCID: PMC11438667 DOI: 10.1007/s43032-024-01678-6] [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: 01/09/2024] [Accepted: 08/12/2024] [Indexed: 09/05/2024]
Abstract
Fetal inflammatory response syndrome or infection after preterm premature rupture of membranes (PPROM) increases neonatal morbidity in preterm deliveries. Biochemical markers from the amniotic fluid (AF) have been used to evaluate possible intra-amniotic infection during the asymptomatic phase after PPROM. This study aimed to describe whether soluble urokinase-type plasminogen activator receptor (suPAR) or procalcitonin (PCT) from AF or maternal sera could reveal fetal inflammatory response or infection after PPROM. AF and maternal serum samples were collected weekly after PPROM (23+ 0 - 34+ 6 gestational weeks) until delivery from twenty women and two women with possible chorioamnionitis with intact membranes. Levels of suPAR, PCT, interleukin-6 (IL-6), glucose, lactate dehydrogenase (LDH), and bacterial PCR were determined from AF and suPAR and PCT and IL-6 from maternal sera. Fetal infection or inflammation response were determined by the histology of the placenta after delivery. AF glucose was significantly lower and AF LDH higher in the fetal site histologic chorioamnionitis (HCA) group, while AF suPAR concentrations tended to be higher in this group. AF suPAR correlated significantly with AF glucose and LDH. Based on receiver operating characteristic (ROC) analysis, AF glucose had the best predictability for fetal site histological chorioamnionitis. The findings of AF PCT were insignificant considering HCA. AF glucose had the highest accuracy in predicting fetal site histologic chorioamnionitis. AF suPAR may be a promising marker; however, our findings were limited by a small study population.
Collapse
Affiliation(s)
- Kati Jalkanen
- Department of Obstetric and Gynecology, Tampere University Hospital, Elämänaukio, Kuntokatu 2, Tampere, 33520, Finland.
| | - Anita Virtanen
- Department of Obstetric and Gynecology, Tampere University Hospital, Elämänaukio, Kuntokatu 2, Tampere, 33520, Finland
| | - Janne Aittoniemi
- Department of Clinical Microbiology, Fimlab Laboratories, Tampere, Finland
| | - Heidi Flinck
- Department of Clinical Microbiology, Fimlab Laboratories, Tampere, Finland
| | - Sinikka Ampuja
- Department of Pathology, Tampere University Hospital, Tampere, Finland
| | - Heini Huhtala
- Faculty of Social Sciences, Tampere University, Tampere, Finland
| | - Kati Tihtonen
- Department of Obstetric and Gynecology, Tampere University Hospital, Elämänaukio, Kuntokatu 2, Tampere, 33520, Finland
| |
Collapse
|
25
|
Yu H, Li D, Zhao X, Fu J. Fetal origin of bronchopulmonary dysplasia: contribution of intrauterine inflammation. Mol Med 2024; 30:135. [PMID: 39227783 PMCID: PMC11373297 DOI: 10.1186/s10020-024-00909-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 08/23/2024] [Indexed: 09/05/2024] Open
Abstract
Bronchopulmonary dysplasia (BPD) is a common chronic lung disease in infants and the most frequent adverse outcome of premature birth, despite major efforts to minimize injury. It is thought to result from aberrant repair response triggered by either prenatal or recurrent postnatal injury to the lungs during development. Intrauterine inflammation is an important risk factor for prenatal lung injury, which is also increasingly linked to BPD. However, the specific mechanisms remain unclear. This review summarizes clinical and animal research linking intrauterine inflammation to BPD. We assess how intrauterine inflammation affects lung alveolarization and vascular development. In addition, we discuss prenatal therapeutic strategies targeting intrauterine inflammation to prevent or treat BPD.
Collapse
Affiliation(s)
- Haoting Yu
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, 110004, China
| | - Danni Li
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, 110004, China
| | - Xinyi Zhao
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, 110004, China
| | - Jianhua Fu
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, 110004, China.
| |
Collapse
|
26
|
Ling LJ, Zhou Q, Zhang F, Lei WJ, Li MD, Lu JW, Wang WS, Sun K, Ying H. The dual role of glucocorticoid regeneration in inflammation at parturition. Front Immunol 2024; 15:1459489. [PMID: 39290694 PMCID: PMC11405189 DOI: 10.3389/fimmu.2024.1459489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 08/19/2024] [Indexed: 09/19/2024] Open
Abstract
Introduction Fetal membrane inflammation is an integral event of parturition. However, excessive pro-inflammatory cytokines can impose threats to the fetus. Coincidentally, the fetal membranes express abundant 11β-hydroxysteroid dehydrogenase 1 (11β-HSD1), which generates biologically active cortisol to promote labor through induction of prostaglandin synthesis. Given the well-recognized anti-inflammatory actions of glucocorticoids, we hypothesized that cortisol regenerated in the fetal membranes might be engaged in restraining fetus-hazardous pro-inflammatory cytokine production for the safety of the fetus, while reserving pro-labor effect on prostaglandin synthesis to ensure safe delivery of the fetus. Methods The hypothesis was examined in human amnion tissue and cultured primary human amnion fibroblasts as well as a mouse model. Results 11β-HSD1 was significantly increased in the human amnion in infection-induced preterm birth. Studies in human amnion fibroblasts showed that lipopolysaccharide (LPS) induced 11β-HSD1 expression synergistically with cortisol. Cortisol completely blocked NF-κB-mediated pro-inflammatory cytokine expression by LPS, but STAT3-mediated cyclooxygenase 2 expression, a crucial prostaglandin synthetic enzyme, remained. Further studies in pregnant mice showed that corticosterone did not delay LPS-induced preterm birth, but alleviated LPS-induced fetal organ damages, along with increased 11β-HSD1, cyclooxygenase 2, and decreased pro-inflammatory cytokine in the fetal membranes. Discussion There is a feed-forward cortisol regeneration in the fetal membranes in infection, and cortisol regenerated restrains pro-inflammatory cytokine expression, while reserves pro-labor effect on prostaglandin synthesis. This dual role of cortisol regeneration can prevent excessive pro-inflammatory cytokine production, while ensure in-time delivery for the safety of the fetus.
Collapse
Affiliation(s)
- Li-Jun Ling
- Department of Obstetrics, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai, China
| | - Qiong Zhou
- Department of Obstetrics and Gynecology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fan Zhang
- Center for Reproductive Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Wen-Jia Lei
- Center for Reproductive Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Meng-Die Li
- Center for Reproductive Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Jiang-Wen Lu
- Center for Reproductive Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Wang-Sheng Wang
- Center for Reproductive Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Kang Sun
- Center for Reproductive Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Hao Ying
- Department of Obstetrics, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai, China
| |
Collapse
|
27
|
Tantengco OAG, Diwa MH, Millagrosa PMM, Velayo CL. Epidemiology and placental pathology of intrauterine fetal demise in a tertiary hospital in the Philippines. Eur J Obstet Gynecol Reprod Biol X 2024; 23:100338. [PMID: 39286338 PMCID: PMC11404223 DOI: 10.1016/j.eurox.2024.100338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 08/19/2024] [Accepted: 08/23/2024] [Indexed: 09/19/2024] Open
Abstract
Objectives The Philippines has at least 25,000 stillbirth or intrauterine fetal demise (IUFD) cases every year. Despite its burden, there is scarce information on IUFD epidemiology in the Philippines. Hence, this study reported the epidemiology and placental pathology of IUFD in a tertiary hospital in the Philippines. Study design This cross-sectional study analyzed second- and third-trimester IUFD cases at the Philippine General Hospital from 2012 to 2021. We reviewed maternal sociodemographic and clinical characteristics and evaluated placental pathology. All statistical tests were done with GraphPad Prism software version 8.0. Results We recorded 947 (2.28 %) cases of IUFD out of 41,562 obstetric deliveries from 2012 to 2021. Out of 947 IUFD cases, 532 had placental pathology reports. Second-trimester IUFD cases showed higher rates of no antenatal care (42.86 %) compared to third-trimester cases (10.61 %). Hypertensive disorders were more common in third-trimester IUFD. Infarcts (23.34 %), calcifications (4.12 %), and hemorrhages/hematomas (3.00 %) were the most prevalent placental abnormalities. While these abnormalities were more common in third-trimester IUFD, placental and fetal membrane infections like chorioamnionitis were more frequent in second-trimester IUFD. Conclusion The results highlighted the differences in maternal sociodemographic and clinical characteristics, and placental pathology between second- and third-trimester cases of IUFD. These observations revealed distinct pathological processes and potential etiologies contributing to IUFD in the Philippines.
Collapse
Affiliation(s)
- Ourlad Alzeus G Tantengco
- Department of Physiology, College of Medicine, University of the Philippines Manila, Manila, Philippines
| | - Michele H Diwa
- Department of Pathology, College of Medicine, University of the Philippines Manila, Manila, Philippines
- Department of Laboratories, University of the Philippines - Philippine General Hospital, Taft Avenue, Manila, Philippines
| | | | - Clarissa L Velayo
- Department of Physiology, College of Medicine, University of the Philippines Manila, Manila, Philippines
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, University of the Philippines - Philippine General Hospital, Taft Avenue, Manila, Philippines
| |
Collapse
|
28
|
Soucek O, Kacerovsky M, Musilova IK, Stranik J, Kukla R, Bolehovska R, Hornychova H, Andrys C. Changes in Relative Counts of Different Leukocyte Subpopulations in Peripheral and Umbilical Cord Blood of Women With Preterm Prelabor Rupture of Membranes With Respect to Intraamniotic Inflammation and Fetal Inflammatory Response Syndrome. Am J Reprod Immunol 2024; 92:e13926. [PMID: 39302219 DOI: 10.1111/aji.13926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 08/21/2024] [Accepted: 09/01/2024] [Indexed: 09/22/2024] Open
Abstract
OBJECTIVE The aim of this study was to evaluate changes in the relative counts of different leukocyte subsets in peripheral and umbilical cord blood in pregnancies complicated by preterm prelabor rupture of membranes (PPROM) with respect to the presence of intraamniotic inflammation (IAI) and fetal inflammatory response syndrome (FIRS). METHODS Fifty-two women with singleton pregnancies complicated by PPROM were included in this study. From samples of peripheral and umbilical cord blood, relative counts of these leukocyte subpopulations were determined using multicolor flow cytometry: granulocytes, monocytes, lymphocytes, T cells and their subpopulations, B cells and their subpopulations, and NK cells and their subpopulations. IAI was defined as increased concentrations of interleukin 6 in the amniotic fluid. Amniotic fluid samples were obtained by transabdominal amniocentesis. RESULTS Women with IAI had higher relative counts of monocytes (p = 0.04) in peripheral blood. There was an increased relative number of granulocytes (p = 0.003) and a decreased number of lymphocytes (p = 0.0048), helper CD4+ T cells (p = 0.019), NK cells (p = 0.0001) within leukocytes, NK cells within lymphocytes (p = 0.003) and CD16+ NK cells within NK cells (p = 0.005) in umbilical cord blood samples of women with FIRS. However, after adjusting the results for gestational age at sampling, all differences disappeared. CONCLUSIONS The presence of IAI or FIRS is not accompanied by significant changes in the relative counts of immune cells in peripheral blood or umbilical cord blood in pregnancies complicated by PPROM.
Collapse
Affiliation(s)
- Ondrej Soucek
- Department of Immunology, University Hospital Hradec Kralove, Hradec Kralove, Czech Republic
| | - Marian Kacerovsky
- Department of Obstetrics and Gynecology, Hospital Most, Krajska zdravotni a.s., Most, Czech Republic
- Biomedical Research Center, University Hospital Hradec Kralove, Hradec Kralove, Czech Republic
| | - Ivana Kacerovska Musilova
- Department of Obstetrics and Gynecology, Hospital Most, Krajska zdravotni a.s., Most, Czech Republic
- Biomedical Research Center, University Hospital Hradec Kralove, Hradec Kralove, Czech Republic
| | - Jaroslav Stranik
- Department of Obstetrics and Gynecology, University Hospital Hradec Kralove, Hradec Kralove, Czech Republic
| | - Rudolf Kukla
- Department of Microbiology, University Hospital Hradec Kralove, Hradec Kralove, Czech Republic
| | - Radka Bolehovska
- Department of Microbiology, University Hospital Hradec Kralove, Hradec Kralove, Czech Republic
| | - Helena Hornychova
- Department of Pathology, University Hospital Hradec Kralove, Hradec Kralove, Czech Republic
| | - Ctirad Andrys
- Department of Immunology, University Hospital Hradec Kralove, Hradec Kralove, Czech Republic
| |
Collapse
|
29
|
Feng MT, Ji Q, Liu DD, Xu W. Two cases of Leukemoid reaction in premature infants caused by fetal inflammatory response syndrome. BMC Pediatr 2024; 24:546. [PMID: 39182037 PMCID: PMC11344429 DOI: 10.1186/s12887-024-05006-4] [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: 08/27/2023] [Accepted: 08/12/2024] [Indexed: 08/27/2024] Open
Abstract
BACKGROUND Fetal inflammatory response syndrome (FIRS) is a systemic inflammatory response caused by the activation of the fetal immune system. The serological diagnostic criterion for fetal inflammatory response syndrome is a cord blood interleukin-6 concentration that exceeds 11 pg/mL, while pathologic evidence indicates the presence of funisitis or chorionic vasculitis. It can affect all systems of the fetus. Alterations in patients' hematopoietic system are primarily reflected by changes in peripheral blood leukocyte and neutrophil counts. CASE PRESENTATION We performed placental pathology to identify FIRS and showed two cases of neonatal leukemoid reaction caused by FIRS. These two babies' alterations in hematopoietic system resolves spontaneously with the inflammation relief, without specific interventions. During the 16‑month and14- month follow‑up period, their motor and intellectual development was normal. CONCLUSIONS . Neonatal leukemoid reaction is a reactive disease characterized by abnormal blood parameters similar to those of leukemia, but not leukemia. It is an aberrant hematopoietic response that typically resolves spontaneously with cause relief without requiring specific interventions.
Collapse
Affiliation(s)
- Meng-Ting Feng
- Department of Neonatology, Children's Medical Center, The First Hospital of Jilin University, NO.1 Xinmin Street, Changchun, 130012, China
| | - Qiong Ji
- Department of Neonatology, Children's Medical Center, The First Hospital of Jilin University, NO.1 Xinmin Street, Changchun, 130012, China
| | - Dan-Dan Liu
- Department of Neonatology, Children's Medical Center, The First Hospital of Jilin University, NO.1 Xinmin Street, Changchun, 130012, China
| | - Wei Xu
- Department of Neonatology, Children's Medical Center, The First Hospital of Jilin University, NO.1 Xinmin Street, Changchun, 130012, China.
| |
Collapse
|
30
|
王 燕, 曾 雅, 陈 雪, 黄 智, 杨 传. [Clinical characteristics of Ureaplasma urealyticum infection and colonization in extremely preterm infants]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2024; 26:811-816. [PMID: 39148384 PMCID: PMC11334552 DOI: 10.7499/j.issn.1008-8830.2403002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 06/25/2024] [Indexed: 08/17/2024]
Abstract
OBJECTIVES To investigate the clinical characteristics of Ureaplasma urealyticum (UU) infection and colonization in extremely preterm infants and its impact on the incidence of bronchopulmonary dysplasia (BPD). METHODS A retrospective analysis was conducted on 258 extremely preterm infants who were admitted to the Department of Neonatology, Shenzhen Maternity and Child Healthcare Hospital, from September 2018 to September 2022. According to the results of UU nucleic acid testing and the evaluation criteria for UU infection and colonization, the subjects were divided into three groups: UU-negative group (155 infants), UU infection group (70 infants), and UU colonization group (33 infants). The three groups were compared in terms of general information and primary and secondary clinical outcomes. RESULTS Compared with the UU-negative group, the UU infection group had significant increases in the incidence rate of BPD, total oxygen supply time, and the length of hospital stay (P<0.05), while there were no significant differences in the incidence rates of BPD and moderate/severe BPD between the UU colonization group and the UU-negative group (P>0.05). CONCLUSIONS The impact of UU on the incidence of BPD in extremely preterm infants is associated with the pathogenic state of UU (i.e., infection or colonization), and there are significant increases in the incidence rate of BPD, total oxygen supply time, and the length of hospital stay in extremely preterm infants with UU infection. UU colonization is not associated with the incidence of BPD and moderate/severe BPD in extremely preterm infants.
Collapse
|
31
|
Alayli Y, Brown LS, Tolentino-Plata K, Leon R, Heyne R, Chalak LF, Mir IN. Neurodevelopmental outcomes in extremely preterm infants with placental pathologic evidence of fetal inflammatroy response. Pediatr Res 2024:10.1038/s41390-024-03391-1. [PMID: 39112787 DOI: 10.1038/s41390-024-03391-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 06/13/2024] [Accepted: 06/23/2024] [Indexed: 11/28/2024]
Abstract
OBJECTIVES Neonates born with fetal inflammatory response (FIR) are at increased risk for adverse neonatal outcomes. Our objective was to determine whether FIR and its severity is associated with neurodevelopmental impairment (NDI) at 2 years of age or death among preterm infants. METHODS A retrospective cohort study of prospectively collected data of all infants born <29 weeks gestational age (GA). FIR and its severity were diagnosed according to the Amsterdam Placental Workshop Group Consensus Statement. Neurodevelopmental outcomes among all participants were quantified according to Bayley III. RESULTS Mothers of infants with FIR were significantly younger (P = 0.04) and had a greater prevalence of antenatal steroid use (P < 0.01), infection during pregnancy (P = 0.01), PPROM (P < 0.01), and clinical chorioamnionitis (P < 0.01). Infants with FIR had longer duration of hospitalization (P < 0.01), days on oxygen (P < 0.01), congenital pneumonia (P = 0.03), moderate/severe bronchopulmonary dysplasia (BPD; P < 0.01). Notably, infants with FIR were not at increased risk of NDI or death (primary outcome). Those with moderate to severe FIR (≥ stage 2 FIR) were at increased risk of developing motor & language impairment or death (P < 0.01). CONCLUSION This is the first report demonstrating an association between the severity of FIR and subsequent NDI in preterm infants born. IMPACT STATEMENT Fetal Inflammatory Response (FIR) is not associated Neurodevelopmental Impairement (NDI) or Death in preterm infants However, there is significant relationship between moderate to severe FIR and NDI at 2 years of age in preterm infants. This is the first study demonstrating the impact of progression and severity of FIR on NDI or Death in preterm infants. These observations provide additional insight into understanding the impact of intrauterine exposure to inflammation on the NDI or death in preterm infants.
Collapse
Affiliation(s)
- Yasmeen Alayli
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | | | - Kristine Tolentino-Plata
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Rachel Leon
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Roy Heyne
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Lina F Chalak
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Imran N Mir
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
32
|
Deng ZM, Dai FF, Wang RQ, Deng HB, Yin TL, Cheng YX, Chen GT. Organ-on-a-chip: future of female reproductive pathophysiological models. J Nanobiotechnology 2024; 22:455. [PMID: 39085921 PMCID: PMC11290169 DOI: 10.1186/s12951-024-02651-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 06/18/2024] [Indexed: 08/02/2024] Open
Abstract
The female reproductive system comprises the internal and external genitalia, which communicate through intricate endocrine pathways. Besides secreting hormones that maintain the female secondary sexual characteristics, it also produces follicles and offspring. However, the in vitro systems have been very limited in recapitulating the specific anatomy and pathophysiology of women. Organ-on-a-chip technology, based on microfluidics, can better simulate the cellular microenvironment in vivo, opening a new field for the basic and clinical research of female reproductive system diseases. This technology can not only reconstruct the organ structure but also emulate the organ function as much as possible. The precisely controlled fluidic microenvironment provided by microfluidics vividly mimics the complex endocrine hormone crosstalk among various organs of the female reproductive system, making it a powerful preclinical tool and the future of pathophysiological models of the female reproductive system. Here, we review the research on the application of organ-on-a-chip platforms in the female reproductive systems, focusing on the latest progress in developing models that reproduce the physiological functions or disease features of female reproductive organs and tissues, and highlighting the challenges and future directions in this field.
Collapse
Affiliation(s)
- Zhi-Min Deng
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, China
| | - Fang-Fang Dai
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, China
| | - Rui-Qi Wang
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, China
| | - Hong-Bing Deng
- Hubei International Scientific and Technological Cooperation Base of Sustainable Resource and Energy, Hubei Key Laboratory of Biomass Resource Chemistry and Environmental Biotechnology, School of Resource and Environmental Science, Wuhan University, Wuhan, Hubei, 430060, China
| | - Tai-Lang Yin
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, China.
| | - Yan-Xiang Cheng
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, China.
| | - Gan-Tao Chen
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, China.
| |
Collapse
|
33
|
Yates EF, Mulkey SB. Viral infections in pregnancy and impact on offspring neurodevelopment: mechanisms and lessons learned. Pediatr Res 2024; 96:64-72. [PMID: 38509227 PMCID: PMC11257821 DOI: 10.1038/s41390-024-03145-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 02/28/2024] [Accepted: 03/04/2024] [Indexed: 03/22/2024]
Abstract
Pregnant individuals with viral illness may experience significant morbidity and have higher rates of pregnancy and neonatal complications. With the growing number of viral infections and new viral pandemics, it is important to examine the effects of infection during pregnancy on both the gestational parent and the offspring. Febrile illness and inflammation during pregnancy are correlated with risk for autism, attention deficit/hyperactivity disorder, and developmental delay in the offspring in human and animal models. Historical viral epidemics had limited follow-up of the offspring of affected pregnancies. Infants exposed to seasonal influenza and the 2009 H1N1 influenza virus experienced increased risks of congenital malformations and neuropsychiatric conditions. Zika virus exposure in utero can lead to a spectrum of abnormalities, ranging from severe microcephaly to neurodevelopmental delays which may appear later in childhood and in the absence of Zika-related birth defects. Vertical infection with severe acute respiratory syndrome coronavirus-2 has occurred rarely, but there appears to be a risk for developmental delays in the infants with antenatal exposure. Determining how illness from infection during pregnancy and specific viral pathogens can affect pregnancy and neurodevelopmental outcomes of offspring can better prepare the community to care for these children as they grow. IMPACT: Viral infections have impacted pregnant people and their offspring throughout history. Antenatal exposure to maternal fever and inflammation may increase risk of developmental and neurobehavioral disorders in infants and children. The recent SARS-CoV-2 pandemic stresses the importance of longitudinal studies to follow pregnancies and offspring neurodevelopment.
Collapse
Affiliation(s)
- Emma F Yates
- Frank H. Netter School of Medicine at Quinnipiac University, North Haven, CT, USA
| | - Sarah B Mulkey
- Children's National Hospital, Washington, DC, USA.
- Department of Neurology, The George Washington University School of Medicine and Health Sciences, Washington, DC, USA.
- Department of Pediatrics, The George Washington University School of Medicine and Health Sciences, Washington, DC, USA.
| |
Collapse
|
34
|
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
|
35
|
Galindo AN, Frey Rubio DA, Hettiaratchi MH. Biomaterial strategies for regulating the neuroinflammatory response. MATERIALS ADVANCES 2024; 5:4025-4054. [PMID: 38774837 PMCID: PMC11103561 DOI: 10.1039/d3ma00736g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 04/07/2024] [Indexed: 05/24/2024]
Abstract
Injury and disease in the central nervous system (CNS) can result in a dysregulated inflammatory environment that inhibits the repair of functional tissue. Biomaterials present a promising approach to tackle this complex inhibitory environment and modulate the mechanisms involved in neuroinflammation to halt the progression of secondary injury and promote the repair of functional tissue. In this review, we will cover recent advances in biomaterial strategies, including nanoparticles, hydrogels, implantable scaffolds, and neural probe coatings, that have been used to modulate the innate immune response to injury and disease within the CNS. The stages of inflammation following CNS injury and the main inflammatory contributors involved in common neurodegenerative diseases will be discussed, as understanding the inflammatory response to injury and disease is critical for identifying therapeutic targets and designing effective biomaterial-based treatment strategies. Biomaterials and novel composites will then be discussed with an emphasis on strategies that deliver immunomodulatory agents or utilize cell-material interactions to modulate inflammation and promote functional tissue repair. We will explore the application of these biomaterial-based strategies in the context of nanoparticle- and hydrogel-mediated delivery of small molecule drugs and therapeutic proteins to inflamed nervous tissue, implantation of hydrogels and scaffolds to modulate immune cell behavior and guide axon elongation, and neural probe coatings to mitigate glial scarring and enhance signaling at the tissue-device interface. Finally, we will present a future outlook on the growing role of biomaterial-based strategies for immunomodulation in regenerative medicine and neuroengineering applications in the CNS.
Collapse
Affiliation(s)
- Alycia N Galindo
- Phil and Penny Knight Campus for Accelerating Scientific Impact, University of Oregon Eugene OR USA
| | - David A Frey Rubio
- Phil and Penny Knight Campus for Accelerating Scientific Impact, University of Oregon Eugene OR USA
| | - Marian H Hettiaratchi
- Phil and Penny Knight Campus for Accelerating Scientific Impact, University of Oregon Eugene OR USA
- Department of Chemistry and Biochemistry, University of Oregon Eugene OR USA
| |
Collapse
|
36
|
Chen JJ, Chen XJ, She QM, Li JX, Luo QH. Clinical risk factors for preterm birth and evaluating maternal psychology in the postpartum period. World J Psychiatry 2024; 14:661-669. [PMID: 38808088 PMCID: PMC11129159 DOI: 10.5498/wjp.v14.i5.661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 03/26/2024] [Accepted: 04/11/2024] [Indexed: 05/16/2024] Open
Abstract
BACKGROUND Although the specific pathogenesis of preterm birth (PTB) has not been thoroughly clarified, it is known to be related to various factors, such as pregnancy complications, maternal socioeconomic factors, lifestyle habits, reproductive history, environmental and psychological factors, prenatal care, and nutritional status. PTB has serious implications for newborns and families and is associated with high mortality and complications. Therefore, the prediction of PTB risk can facilitate early intervention and reduce its resultant adverse consequences. AIM To analyze the risk factors for PTB to establish a PTB risk prediction model and to assess postpartum anxiety and depression in mothers. METHODS A retrospective analysis of 648 consecutive parturients who delivered at Shenzhen Bao'an District Songgang People's Hospital between January 2019 and January 2022 was performed. According to the diagnostic criteria for premature infants, the parturients were divided into a PTB group (n = 60) and a full-term (FT) group (n = 588). Puerperae were assessed by the Self-rating Anxiety Scale (SAS) and Self-rating Depression Scale (SDS), based on which the mothers with anxiety and depression symptoms were screened for further analysis. The factors affecting PTB were analyzed by univariate analysis, and the related risk factors were identified by logistic regression. RESULTS According to univariate analysis, the PTB group was older than the FT group, with a smaller weight change and greater proportions of women who underwent artificial insemination and had gestational diabetes mellitus (P < 0.05). In addition, greater proportions of women with reproductive tract infections and greater white blood cell (WBC) counts (P < 0.05), shorter cervical lengths in the second trimester and lower neutrophil percentages (P < 0.001) were detected in the PTB group than in the FT group. The PTB group exhibited higher postpartum SAS and SDS scores than did the FT group (P < 0.0001), with a higher number of mothers experiencing anxiety and depression (P < 0.001). Multivariate logistic regression analysis revealed that a greater maternal weight change, the presence of gestational diabetes mellitus, a shorter cervical length in the second trimester, a greater WBC count, and the presence of maternal anxiety and depression were risk factors for PTB (P < 0.01). Moreover, the risk score of the FT group was lower than that of the PTB group, and the area under the curve of the risk score for predicting PTB was greater than 0.9. CONCLUSION This study highlights the complex interplay between postpartum anxiety and PTB, where maternal anxiety may be a potential risk factor for PTB, with PTB potentially increasing the incidence of postpartum anxiety in mothers. In addition, a greater maternal weight change, the presence of gestational diabetes mellitus, a shorter cervical length, a greater WBC count, and postpartum anxiety and depression were identified as risk factors for PTB.
Collapse
Affiliation(s)
- Jia-Jun Chen
- Department of Clinical Laboratory, Shenzhen Bao’an District Songgang People’s Hospital, Shenzhen 518000, Guangdong Province, China
| | - Xue-Jin Chen
- Department of Otolaryngology Head and Neck Surgery Outpatient, Shenzhen Children’s Hospital, Shenzhen 518000, Guangdong Province, China
| | - Qiu-Min She
- Department of Clinical Laboratory, Shenzhen Bao’an District Songgang People’s Hospital, Shenzhen 518000, Guangdong Province, China
| | - Jie-Xi Li
- Department of Prevention and Health Care, Shenzhen Bao’an District Songgang People’s Hospital, Shenzhen 518000, Guangdong Province, China
| | - Qiu-Hong Luo
- Department of Obstetrics, Shenzhen Bao’an District Songgang People’s Hospital, Shenzhen 518000, Guangdong Province, China
| |
Collapse
|
37
|
Bebell LM, Ngonzi J, Butler A, Kumbakumba E, Adong J, Loos C, Boatin AA, Bassett IV, Siedner MJ, Williams PL, Mattie H, Hedt-Gauthier B, Correia KFB, Lake E, Alter G. Distinct cytokine profiles in late pregnancy in Ugandan people with HIV. Sci Rep 2024; 14:10980. [PMID: 38744864 PMCID: PMC11093984 DOI: 10.1038/s41598-024-61764-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 05/09/2024] [Indexed: 05/16/2024] Open
Abstract
During pregnancy, multiple immune regulatory mechanisms establish an immune-tolerant environment for the allogeneic fetus, including cellular signals called cytokines that modify immune responses. However, the impact of maternal HIV infection on these responses is incompletely characterized. We analyzed paired maternal and umbilical cord plasma collected during labor from 147 people with HIV taking antiretroviral therapy and 142 HIV-uninfected comparators. Though cytokine concentrations were overall similar between groups, using Partial Least Squares Discriminant Analysis we identified distinct cytokine profiles in each group, driven by higher IL-5 and lower IL-8 and MIP-1α levels in pregnant people with HIV and higher RANTES and E-selectin in HIV-unexposed umbilical cord plasma (P-value < 0.01). Furthermore, maternal RANTES, SDF-α, gro α -KC, IL-6, and IP-10 levels differed significantly by HIV serostatus (P < 0.01). Although global maternal and umbilical cord cytokine profiles differed significantly (P < 0.01), umbilical cord plasma profiles were similar by maternal HIV serostatus. We demonstrate that HIV infection is associated with a distinct maternal plasma cytokine profile which is not transferred across the placenta, indicating a placental role in coordinating local inflammatory response. Furthermore, maternal cytokine profiles in people with HIV suggest an incomplete shift from Th2 to Th1 immune phenotype at the end of pregnancy.
Collapse
Affiliation(s)
- Lisa M Bebell
- Medical Practice Evaluation Center and Center for Global Health, Massachusetts General Hospital Division of Infectious Diseases, GRJ-504, 55 Fruit St, Boston, MA, 02114, USA.
| | - Joseph Ngonzi
- Department of Obstetrics and Gynaecology, Mbarara University of Science and Technology, Mbarara, Uganda
| | - Audrey Butler
- State University of New York Upstate Medical University, Syracuse, NY, USA
| | - Elias Kumbakumba
- Department of Paediatrics and Child Health, Mbarara University of Science and Technology, Mbarara, Uganda
| | - Julian Adong
- Department of Paediatrics and Child Health, Mbarara University of Science and Technology, Mbarara, Uganda
| | - Carolin Loos
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - Adeline A Boatin
- Department of Obstetrics and Gynecology and Center for Global Health, Massachusetts General Hospital, Boston, MA, USA
| | - Ingrid V Bassett
- Division of Infectious Diseases and Medical Practice Evaluation Center, Massachusetts General Hospital, Boston, MA, USA
| | - Mark J Siedner
- Division of Infectious Diseases and Medical Practice Evaluation Center, Massachusetts General Hospital, Boston, MA, USA
| | - Paige L Williams
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Heather Mattie
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Bethany Hedt-Gauthier
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Global Health and Social Medicine, Harvard Medical School, Boston, MA, USA
| | | | - Erin Lake
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Galit Alter
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| |
Collapse
|
38
|
Kovács K, Kovács ŐZ, Bajzát D, Imrei M, Nagy R, Németh D, Kói T, Szabó M, Fintha A, Hegyi P, Garami M, Gasparics Á. The histologic fetal inflammatory response and neonatal outcomes: systematic review and meta-analysis. Am J Obstet Gynecol 2024; 230:493-511.e3. [PMID: 37967697 DOI: 10.1016/j.ajog.2023.11.1223] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 10/30/2023] [Accepted: 11/05/2023] [Indexed: 11/17/2023]
Abstract
OBJECTIVE This study aimed to investigate the prognostic role of concomitant histological fetal inflammatory response with chorioamnionitis on neonatal outcomes through a systematic review and meta-analysis of existing literature. DATA SOURCES The primary search was conducted on October 17, 2021, and it was updated on May 26, 2023, across 4 separate databases (MEDLINE, the Cochrane Central Register of Controlled Trials, Embase, and Scopus) without using any filters. STUDY ELIGIBILITY CRITERIA Observational studies reporting obstetrical and neonatal outcomes of infant-mother dyads with histological chorioamnionitis and histological fetal inflammatory response vs infant-mother dyads with histological chorioamnionitis alone were eligible. Studies that enrolled only preterm neonates, studies on neonates born before 37 weeks of gestation, or studies on neonates with very low birthweight (birthweight <1500 g) were included. The protocol was registered with the International Prospective Register of Systematic Reviews (registration number: CRD42021283448). METHODS The records were selected by title, abstract, and full text, and disagreements were resolved by consensus. Random-effect model-based pooled odds ratios with corresponding 95% confidence intervals were calculated for dichotomous outcomes. RESULTS Overall, 50 studies were identified. A quantitative analysis of 14 outcomes was performed. Subgroup analysis using the mean gestational age of the studies was performed, and a cutoff of 28 weeks of gestation was implemented. Among neonates with lower gestational ages, early-onset sepsis (pooled odds ratio, 2.23; 95% confidence interval, 1.76-2.84) and bronchopulmonary dysplasia (pooled odds ratio, 1.30; 95% confidence interval, 1.02-1.66) were associated with histological fetal inflammatory response. Our analysis showed that preterm neonates with a concomitant histological fetal inflammatory response are more likely to develop intraventricular hemorrhage (pooled odds ratio, 1.54; 95% confidence interval, 1.18-2.02) and retinopathy of prematurity (pooled odds ratio, 1.37; 95% confidence interval, 1.03-1.82). The odds of clinical chorioamnionitis were almost 3-fold higher among infant-mother dyads with histological fetal inflammatory response than among infant-mother dyads with histological chorioamnionitis alone (pooled odds ratio, 2.99; 95% confidence interval, 1.96-4.55). CONCLUSION This study investigated multiple neonatal outcomes and found association in the case of 4 major morbidities: early-onset sepsis, bronchopulmonary dysplasia, intraventricular hemorrhage, and retinopathy of prematurity.
Collapse
Affiliation(s)
- Kinga Kovács
- Centre for Translational Medicine, Semmelweis University, Budapest, Hungary
| | - Őzike Zsuzsanna Kovács
- Department of Obstetrics and Gynecology, Semmelweis University, Budapest, Hungary; Department of Obstetrics and Gynecology, Semmelweis University, Budapest, Hungary
| | - Dorina Bajzát
- Centre for Translational Medicine, Semmelweis University, Budapest, Hungary; Heim Pál National Pediatric Institute, Budapest, Hungary
| | - Marcell Imrei
- Centre for Translational Medicine, Semmelweis University, Budapest, Hungary; Heim Pál National Pediatric Institute, Budapest, Hungary
| | - Rita Nagy
- Centre for Translational Medicine, Semmelweis University, Budapest, Hungary
| | - Dávid Németh
- Centre for Translational Medicine, Semmelweis University, Budapest, Hungary
| | - Tamás Kói
- Centre for Translational Medicine, Semmelweis University, Budapest, Hungary
| | - Miklós Szabó
- Department of Obstetrics and Gynecology, Semmelweis University, Budapest, Hungary; Division of Neonatology, First Department of Pediatrics, Semmelweis University, Budapest, Hungary
| | - Attila Fintha
- Faculty of Medicine, Department of Pathology and Cancer Research, Semmelweis University, Budapest, Hungary
| | - Péter Hegyi
- Centre for Translational Medicine, Semmelweis University, Budapest, Hungary; Institute for Translational Medicine, Szentágothai Research Centre, Medical School, University of Pécs, Pécs, Hungary; Institute of Pancreatic Diseases, Semmelweis University, Budapest, Hungary
| | - Miklós Garami
- Centre for Translational Medicine, Semmelweis University, Budapest, Hungary; Department of Pediatrics, Semmelweis University, Budapest, Hungary
| | - Ákos Gasparics
- Department of Obstetrics and Gynecology, Semmelweis University, Budapest, Hungary.
| |
Collapse
|
39
|
Duan J, Xu F, Zhu C, Wang J, Zhang X, Xu Y, Li B, Peng X, Zhu J, Wang X, Zhu C. Histological chorioamnionitis and pathological stages on very preterm infant outcomes. Histopathology 2024; 84:1024-1037. [PMID: 38253913 DOI: 10.1111/his.15147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 01/01/2024] [Accepted: 01/09/2024] [Indexed: 01/24/2024]
Abstract
AIMS Histological chorioamnionitis (HCA) is a condition linked to preterm birth and neonatal infection and its relationship with various pathological stages in extremely preterm neonates, and with their associated short- and long-term consequences, remains a subject of research. This study investigated the connection between different pathological stages of HCA and both short-term complications and long-term outcomes in preterm infants born at or before 32 weeks of gestational age. METHODS Preterm infants born at ≤ 32 weeks of gestation who underwent placental pathology evaluation and were followed-up at 18-24 months of corrected age were included. Neonates were classified based on their exposure to HCA and were further subdivided into different groups according to maternal inflammatory responses (MIR) and fetal inflammatory responses (FIR) stages. We compared short-term complications during their hospital stay between the HCA-exposed and -unexposed groups and examined the influence of HCA stages on long-term outcomes. RESULTS The HCA group exhibited distinct characteristics such as higher rates of premature rupture of membranes > 18 h, reduced amniotic fluid, early-onset sepsis, bronchopulmonary dysplasia and intraventricular haemorrhage (IVH) grades III-IV (P < 0.05). The moderate-severe HCA group displayed lower gestational age, lower birth weight and higher incidence of IVH (grades III-IV) and preterm sepsis compared with the mild HCA group (P < 0.05). After adjusting for confounders, the MIR stages 2-3 group showed associations with cognitive impairment and cerebral palsy (P < 0.05), and the FIR stages 2-3 group also showed poor long-term outcomes and cognitive impairment (P < 0.05). CONCLUSIONS Moderate-severe HCA was associated with increased early-onset sepsis, severe IVH and poor long-term outcomes, including cognitive impairment and cerebral palsy. Vigilant prevention strategies are warranted for severe HCA cases in order to mitigate poorer clinical outcomes.
Collapse
Affiliation(s)
- Jiajia Duan
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Institute of Neuroscience and Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Pediatrics, Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Falin Xu
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Institute of Neuroscience and Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Pediatrics, Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Chaoya Zhu
- Department of Pathology, Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ju Wang
- Department of Anesthesiology, Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiaoli Zhang
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Institute of Neuroscience and Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Pediatrics, Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yiran Xu
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Institute of Neuroscience and Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Pediatrics, Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Bingbing Li
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Institute of Neuroscience and Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Pediatrics, Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xirui Peng
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Institute of Neuroscience and Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Pediatrics, Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jinjin Zhu
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Institute of Neuroscience and Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Pediatrics, Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiaoyang Wang
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Institute of Neuroscience and Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Center for Perinatal Medicine and Health, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Changlian Zhu
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Institute of Neuroscience and Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
40
|
Grill A, Goeral K, Leitich H, Farr A, Berger A, Rittenschober-Boehm J. Maternal biomarkers in predicting neonatal sepsis after preterm premature rupture of membranes in preterm infants. Acta Paediatr 2024; 113:962-972. [PMID: 38265123 DOI: 10.1111/apa.17114] [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: 10/30/2023] [Revised: 01/03/2024] [Accepted: 01/10/2024] [Indexed: 01/25/2024]
Abstract
AIM This retrospective cohort study aimed to assess the utility of maternal C-reactive protein (CRP) and leukocyte levels in predicting neonatal sepsis after preterm premature rupture of membranes (pPROM). METHODS We conducted a retrospective cohort study (2009-2021), encompassing preterm infants born ≤29 + 6 weeks of gestation following pPROM. The primary outcome was early-onset neonatal sepsis within the initial 72 h of life. RESULTS We analysed data from 706 patients with a median gestational age at pPROM of 25.1 weeks and a median gestational age at birth of 26.4 weeks. Overall survival rate was 86.1%, with 65.7% survival without severe morbidities. These rates were significantly worse in preterm infants with sepsis. Maternal CRP and leukocyte levels correlated significantly with neonatal infection markers and sepsis. However, their predictive values, correlation coefficients, and area under the curve values were generally low. Using maternal CRP ≥2 mg/dL to predict neonatal sepsis yielded a positive predictive value of 18.5%, negative predictive value of 91.5%, AUC of 0.589, 45.5% sensitivity, and 74.5% specificity. CONCLUSION Maternal CRP and leukocyte levels were ineffective as a tool for predicting early-onset neonatal sepsis following early pPROM. Consequently, these biomarkers lack the reliability required for clinical decision-making in this context.
Collapse
Affiliation(s)
- Agnes Grill
- Division of Neonatology, Intensive Care and Neuropediatrics, Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, Vienna, Austria
| | - Katharina Goeral
- Division of Neonatology, Intensive Care and Neuropediatrics, Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, Vienna, Austria
| | - Harald Leitich
- Division of Obstetrics and Fetomaternal Medicine, Department of Obstetrics and Gynecology, Comprehensive Center for Pediatrics, Medical University of Vienna, Vienna, Austria
| | - Alex Farr
- Division of Obstetrics and Fetomaternal Medicine, Department of Obstetrics and Gynecology, Comprehensive Center for Pediatrics, Medical University of Vienna, Vienna, Austria
| | - Angelika Berger
- Division of Neonatology, Intensive Care and Neuropediatrics, Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, Vienna, Austria
| | - Judith Rittenschober-Boehm
- Division of Neonatology, Intensive Care and Neuropediatrics, Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
41
|
El Emrani S, Jansen EJS, Goeman JJ, Lopriore E, Termote JUM, Schalij-Delfos NE, van der Meeren LE. Histological Chorioamnionitis and Funisitis as New Risk Factors for Retinopathy of Prematurity: A Meta-analysis. Am J Perinatol 2024; 41:e3264-e3273. [PMID: 37989252 PMCID: PMC11150066 DOI: 10.1055/a-2215-0662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2023]
Abstract
OBJECTIVE The role of placental inflammation in neonatal morbidities is underestimated due to lack of placental examination. This meta-analysis aims to assess the association between histological chorioamnionitis (HCA) with and without funisitis (FUN) and risk of retinopathy of prematurity (ROP). STUDY DESIGN Forty-five studies reporting (unadjusted) data on HCA without FUN and HCA with FUN in neonates with ROP were included. Primary outcomes were any stage ROP and severe ROP. Potential confounders explored were gestational age (GA) at birth, birthweight, maternal steroid use, necrotizing enterocolitis, sepsis (suspected/proven) and mechanical ventilation duration. RESULTS Neonates with HCA had increased risk for any stage ROP (odds ratio [OR] 1.8; 95% confidence interval [CI] 1.3-2.4) and severe ROP (OR 1.5; 95% CI 1.2-1.8) compared with neonates without HCA. The rates of any stage ROP (OR 1.8; 95% CI 1.4-2.2) and severe ROP (OR 1.4; 95% CI 1.1-1.6) were higher in neonates with FUN compared with neonates without FUN. Multivariate meta-regression analysis suggests that lower GA increases the effect size between FUN and severe ROP. CONCLUSION This meta-analysis confirms that presence of HCA and FUN are risk factors for any stage ROP and severe ROP. Structured histological placental examination of HCA and FUN may be a tool to further refine the ROP risk profile. KEY POINTS · This systematic review confirms that HCA is a risk factor for ROP.. · This meta-analysis reveals that FUN results in an even higher risk for developing ROP.. · Placental examination of HCA/FUN may be a tool to further refine the ROP risk profile..
Collapse
Affiliation(s)
- Salma El Emrani
- Department of Ophthalmology, Leiden University Medical Center, Leiden, The Netherlands
- Division of Neonatology, Department of Pediatrics, Willem-Alexander Children's Hospital, Leiden University Medical Center, Leiden, The Netherlands
| | - Esther J S Jansen
- Division of Neonatology, Department of Women and Neonate, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Jelle J Goeman
- Division of Medical Statistics, Department of Biomedical Data Science, Leiden University Medical Center, Leiden, The Netherlands
| | - Enrico Lopriore
- Division of Neonatology, Department of Pediatrics, Willem-Alexander Children's Hospital, Leiden University Medical Center, Leiden, The Netherlands
| | - Jacqueline U M Termote
- Division of Neonatology, Department of Women and Neonate, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, The Netherlands
| | | | - Lotte E van der Meeren
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
- Department of Pathology, Erasmus Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
42
|
Bradley T, Tucker M, Sampath V. Triggered - does maternal COVID-19 program an exaggerated immune response in neonates? Pediatr Res 2024; 95:1400-1401. [PMID: 38172211 DOI: 10.1038/s41390-023-03007-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 12/15/2023] [Indexed: 01/05/2024]
Affiliation(s)
- Todd Bradley
- Genomic Medicine Center, Children's Mercy Kansas City, Kansas City, MO, USA
| | - Megan Tucker
- Division of Neonatology, Children's Mercy Kansas City, Kansas City, MO, USA
| | - Venkatesh Sampath
- Division of Neonatology, Children's Mercy Kansas City, Kansas City, MO, USA.
| |
Collapse
|
43
|
Gilley A, Boly TJ, Paden A, Bermick J. Neonatal immune cells have heightened responses following in-utero exposure to chorioamnionitis or COVID-19. Pediatr Res 2024; 95:1483-1492. [PMID: 37949998 PMCID: PMC11082064 DOI: 10.1038/s41390-023-02888-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 09/25/2023] [Accepted: 10/20/2023] [Indexed: 11/12/2023]
Abstract
BACKGROUND Chorioamnionitis alters neonatal immune responses. Gestational COVID-19 infection is associated with adverse pregnancy outcomes, but its impact on neonatal immunity is unclear. We hypothesized that gestational COVID-19 exposure would result in exaggerated neonatal immune responses, similar to chorioamnionitis-exposed neonates. METHODS Term umbilical cord blood mononuclear cells (CBMCs) were isolated from neonates exposed to chorioamnionitis, gestational COVID-19 or unexposed controls. CBMCs were cultured and stimulated with heat-killed Escherichia coli, Streptococcus agalactiae or Staphylococcus epidermidis. A multiplexed protein assay was used to measure cytokine levels in cell culture supernatants and flow cytometry was used to evaluate cellular-level cytokine expression. RESULTS Both chorioamnionitis-exposed and COVID-19 exposed CBMCs demonstrated upregulation of IL-1β and IL-6 compared to unexposed CBMCs, while only COVID-19 exposure resulted in IL-8 upregulation. There were no differences between chorioamnionitis-exposed and COVID-19 exposed CBMCs when these groups were directly compared. Flow cytometry demonstrated immune cell subset specific differences in cytokine expression between the exposure groups. CONCLUSION The fetal/neonatal response to maternal inflammation differed based on immune cell subset and etiology of inflammation, but the global neonatal cytokine responses were similar between exposure groups. This suggests that targeting perinatal inflammation rather than the specific etiology may be a possible therapeutic approach. IMPACT Neonatal immune cells have similar pathogen-associated global cytokine responses, but different cell-level immune responses, following in-utero exposure to chorioamnionitis or COVID-19. This is the first study to directly compare immune responses between neonates exposed to chorioamnionitis and COVID-19. This suggests that the fetal/neonatal cellular response to perinatal inflammation differs based on the etiology and severity of maternal inflammation, but still results in a similar overall inflammatory profile regardless of the cause of perinatal inflammation.
Collapse
Affiliation(s)
- Annemarie Gilley
- Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, University of Iowa, Iowa City, IA, USA.
| | - Timothy J Boly
- Division of Neonatology, Department of Pediatrics, University of Iowa, Iowa City, IA, USA
- Iowa Inflammation Program, University of Iowa, Iowa City, IA, USA
| | - Austin Paden
- Division of Neonatology, Department of Pediatrics, University of Iowa, Iowa City, IA, USA
- Iowa Inflammation Program, University of Iowa, Iowa City, IA, USA
| | - Jennifer Bermick
- Division of Neonatology, Department of Pediatrics, University of Iowa, Iowa City, IA, USA
- Iowa Inflammation Program, University of Iowa, Iowa City, IA, USA
| |
Collapse
|
44
|
McKissic D, Perez FA, Puia-Dumitrescu M, Ryan R, Hendrixson DT, Billimoria Z, DiGeronimo R, Sawyer T. Maternal COVID-19 Infection Associated with Fetal Systemic Inflammatory Complications in COVID-19-Negative Neonates: A Case-Series. Am J Perinatol 2024; 41:e1451-e1458. [PMID: 38134940 DOI: 10.1055/a-2234-8064] [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] [Indexed: 12/24/2023]
Abstract
OBJECTIVE This study aimed to examine fetal and neonatal inflammatory and neurologic complications associated with maternal coronavirus disease 2019 (COVID-19) infection. STUDY DESIGN Case-series using a convenience sample of neonates cared for in a large referral-based children's hospital neonatal intensive care unit between September 2021 and May 2022. RESULTS We identified seven neonates with exposure to maternal severe acute respiratory syndrome related coronavirus 2 (SARS-CoV-2) and a presentation consistent with inflammatory complications. All had some degree of neurologic injury with neuroimaging findings including restricted diffusion indicating injury in the white matter, cortex, deep gray structures, and splenium of the corpus callosum as well as intracranial hemorrhage. In addition, many infants had cytopenia and abnormal coagulation studies. Placental pathology, when available, revealed inflammation, clot with calcifications, and hematomas with associated infarcts. CONCLUSION Neonates born to mothers with SARS-CoV-2, even when negative for the virus themselves, may have complications consistent with a systemic inflammatory syndrome. Placental pathology as well as neurologic imaging in infants with neurologic findings may help to support this diagnosis. KEY POINTS · A systemic inflammatory response may cause illness in babies born to mothers with a history of COVID-19.. · Inflammatory markers and placental pathology are helpful in supporting this diagnosis.. · Consider neuroimaging in infants of mothers with a history of COVID-19 with neurologic findings..
Collapse
Affiliation(s)
- Devin McKissic
- Division of Neonatology, Department of Pediatrics, University of Washington and Seattle Children's Hospital, Seattle, Washington
| | - Francisco A Perez
- Department of Radiology, University of Washington and Seattle Children's Hospital, Seattle, Washington
| | - Mihai Puia-Dumitrescu
- Division of Neonatology, Department of Pediatrics, University of Washington and Seattle Children's Hospital, Seattle, Washington
| | - Ramah Ryan
- Division of Neonatology, Department of Pediatrics, University of Washington and Seattle Children's Hospital, Seattle, Washington
| | - D Taylor Hendrixson
- Division of Neonatology, Department of Pediatrics, University of Washington and Seattle Children's Hospital, Seattle, Washington
- Division of Infectious Disease, Department of Pediatrics, University of Washington, Seattle, Washington
| | - Zeenia Billimoria
- Division of Neonatology, Department of Pediatrics, University of Washington and Seattle Children's Hospital, Seattle, Washington
| | - Robert DiGeronimo
- Division of Neonatology, Department of Pediatrics, University of Washington and Seattle Children's Hospital, Seattle, Washington
| | - Taylor Sawyer
- Division of Neonatology, Department of Pediatrics, University of Washington and Seattle Children's Hospital, Seattle, Washington
| |
Collapse
|
45
|
Halpern MD, Gupta A, Zaghloul N, Thulasingam S, Calton CM, Camp SM, Garcia JGN, Ahmed M. Extracellular Nicotinamide Phosphoribosyltransferase Is a Therapeutic Target in Experimental Necrotizing Enterocolitis. Biomedicines 2024; 12:970. [PMID: 38790933 PMCID: PMC11118767 DOI: 10.3390/biomedicines12050970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 04/21/2024] [Accepted: 04/24/2024] [Indexed: 05/26/2024] Open
Abstract
Necrotizing enterocolitis (NEC) is the most common gastrointestinal emergency of prematurity. Postulated mechanisms leading to inflammatory necrosis of the ileum and colon include activation of the pathogen recognition receptor Toll-like receptor 4 (TLR4) and decreased levels of transforming growth factor beta (TGFβ). Extracellular nicotinamide phosphoribosyltransferase (eNAMPT), a novel damage-associated molecular pattern (DAMP), is a TLR4 ligand and plays a role in a number of inflammatory disease processes. To test the hypothesis that eNAMPT is involved in NEC, an eNAMPT-neutralizing monoclonal antibody, ALT-100, was used in a well-established animal model of NEC. Preterm Sprague-Dawley pups delivered prematurely from timed-pregnant dams were exposed to hypoxia/hypothermia and randomized to control-foster mother dam-fed rats, injected IP with saline (vehicle) 48 h after delivery; control + mAB-foster dam-fed rats, injected IP with 10 µg of ALT-100 at 48 h post-delivery; NEC-orally gavaged, formula-fed rats injected with saline; and NEC + mAb-formula-fed rats, injected IP with 10 µg of ALT-100 at 48 h. The distal ileum was processed 96 h after C-section delivery for histological, biochemical, molecular, and RNA sequencing studies. Saline-treated NEC pups exhibited markedly increased fecal blood and histologic ileal damage compared to controls (q < 0.0001), and findings significantly reduced in ALT-100 mAb-treated NEC pups (q < 0.01). Real-time PCR in ileal tissues revealed increased NAMPT in NEC pups compared to pups that received the ALT-100 mAb (p < 0.01). Elevated serum levels of tumor necrosis factor alpha (TNFα), interleukin 6 (IL-6), interleukin-8 (IL-8), and NAMPT were observed in NEC pups compared to NEC + mAb pups (p < 0.01). Finally, RNA-Seq confirmed dysregulated TGFβ and TLR4 signaling pathways in NEC pups that were attenuated by ALT-100 mAb treatment. These data strongly support the involvement of eNAMPT in NEC pathobiology and eNAMPT neutralization as a strategy to address the unmet need for NEC therapeutics.
Collapse
Affiliation(s)
- Melissa D. Halpern
- Division of Neonatology, Department of Pediatrics, College of Medicine, University of Arizona, Tucson, AZ 85724, USA
| | - Akash Gupta
- Division of Neonatology, Department of Pediatrics, College of Medicine, University of Arizona, Tucson, AZ 85724, USA
| | - Nahla Zaghloul
- Division of Neonatology, Department of Pediatrics, College of Medicine, University of Arizona, Tucson, AZ 85724, USA
| | - Senthilkumar Thulasingam
- Division of Neonatology, Department of Pediatrics, College of Medicine, University of Arizona, Tucson, AZ 85724, USA
| | - Christine M. Calton
- Division of Neonatology, Department of Pediatrics, College of Medicine, University of Arizona, Tucson, AZ 85724, USA
| | - Sara M. Camp
- Center for Inflammation Science and Systems Medicine, University of Florida Scripps Research Institute, Jupiter, FL 33458, USA (J.G.N.G.)
| | - Joe G. N. Garcia
- Center for Inflammation Science and Systems Medicine, University of Florida Scripps Research Institute, Jupiter, FL 33458, USA (J.G.N.G.)
| | - Mohamed Ahmed
- Division of Neonatology, Department of Pediatrics, College of Medicine, University of Arizona, Tucson, AZ 85724, USA
| |
Collapse
|
46
|
Chen B, Wang L, Xie D, Wang Y. Bioinformatics-based discovery of biomarkers and immunoinflammatory targets in children with cerebral palsy: An observational study. Medicine (Baltimore) 2024; 103:e37828. [PMID: 38640267 PMCID: PMC11029991 DOI: 10.1097/md.0000000000037828] [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: 02/12/2024] [Revised: 03/12/2024] [Accepted: 03/15/2024] [Indexed: 04/21/2024] Open
Abstract
Cerebral palsy (CP) is the most common disabling disease in children, and motor dysfunction is the core symptom of CP. Although relevant risk factors have been found to be closely associated with CP: congenital malformations, multiple gestation, prematurity, intrauterine inflammation and infection, birth asphyxia, thrombophilia, and perinatal stroke. Its important pathophysiological mechanism is amniotic fluid infection and intraamniotic inflammation leading to fetal developing brain damage, which may last for many years. However, the molecular mechanism of CP is still not well explained. This study aimed to use bioinformatics to identify key biomarker-related signaling pathways in CP. The expression profile of children with CP was selected from the Gene Expression Comprehensive Database, and the CP disease gene data set was obtained from GeneCards. A protein-protein interaction network was established and functional enrichment analysis was performed using Gene Ontology and Kyoto Encyclopedia of Genes and Genomes databases. A total of 144 differential key intersection genes and 10 hub genes were identified through molecular biology. Gene Ontology functional enrichment analysis results show that differentially expressed genes are mainly concentrated in biological processes, such as immune response and neurogenesis. The cellular components involved mainly include axons, postsynaptic membranes, etc, and their molecular functions mainly involve proteoglycan binding, collagen binding, etc. Kyoto Encyclopedia of Genes and Genomes analysis shows that the intersection genes are mainly in signaling pathways related to the immune system, inflammatory response, and nervous system, such as Th17 cell differentiation, Toll-like receptor signaling pathway, tumor necrosis factor signaling pathway, NF-κB signaling pathway, axon guidance, PI3K-Akt signaling pathway, HIF-1 signaling pathway, gap junction, etc. Jak-STAT signaling pathway, mTOR signaling pathway, and related hub genes regulate immune cells and inflammatory factors and play an important role in the development and progression of CP.
Collapse
Affiliation(s)
- Bo Chen
- Department of Rehabilitation, The Affiliated Hospital of Southwest Medical University, Southwest Medical University, Luzhou, China
- Department of Rehabilitation Science, Hong Kong Polytechnic University, Hong Kong, China
| | - Ling Wang
- Department of Operating Room, The Affiliated Hospital of Southwest Medical University, Southwest Medical University, Luzhou, China
| | - Dongke Xie
- Pediatric Surgery, The Affiliated Hospital of Southwest Medical University, Southwest Medical University, Luzhou, China
- Sichuan Clinical Research Center for Birth Defects, The Affiliated Hospital of Southwest Medical University, Southwest Medical University, Luzhou, China
| | - Yuanhui Wang
- Pediatric Surgery, The Affiliated Hospital of Southwest Medical University, Southwest Medical University, Luzhou, China
- Sichuan Clinical Research Center for Birth Defects, The Affiliated Hospital of Southwest Medical University, Southwest Medical University, Luzhou, China
| |
Collapse
|
47
|
Fortin O, DeBiasi RL, Mulkey SB. Congenital infectious encephalopathies from the intrapartum period to postnatal life. Semin Fetal Neonatal Med 2024:101526. [PMID: 38677956 DOI: 10.1016/j.siny.2024.101526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/29/2024]
Abstract
Congenital infections are a common but often underrecognized cause of fetal brain abnormalities, as well as fetal-neonatal morbidity and mortality, that should be considered by all healthcare professionals providing neurological care to fetuses and newborns. Maternal infection with various pathogens (cytomegalovirus, Toxoplasmosis, Rubella virus, Parvovirus B19, lymphocytic choriomeningitis virus, syphilis, Zika virus, varicella zoster virus) during pregnancy can be transmitted to the developing fetus, which can cause multisystem dysfunction and destructive or malformative central nervous system lesions. These can be recognized on fetal and neonatal imaging, including ultrasound and MRI. Imaging and clinical features often overlap, but some distinguishing features can help identify specific pathogens and guide subsequent testing strategies. Some pathogens can be specifically treated, and others can be managed with targeted interventions or symptomatic therapy based on expected complications. Neurological and neurodevelopmental complications related to congenital infections vary widely and are likely driven by a combination of pathophysiologic factors, alone or in combination. These include direct invasion of the fetal central nervous system by pathogens, inflammation of the maternal-placental-fetal triad in response to infection, and long-term effects of immunogenic and epigenetic changes in the fetus in response to maternal-fetal infection. Congenital infections and their neurodevelopmental impacts should be seen as an issue of public health policy, given that infection and the associated complications disproportionately affect woman and children from low- and middle-income countries and those with lower socio-economic status in high-income countries. Congenital infections may be preventable and treatable, which can improve long-term neurodevelopmental outcomes in children.
Collapse
Affiliation(s)
- Olivier Fortin
- Zickler Family Prenatal Pediatrics Institute, Children's National Hospital, Washington DC, USA
| | - Roberta L DeBiasi
- Division of Pediatric Infectious Disease, Children's National Hospital, Washington DC, USA; Department of Pediatrics, The George Washington University School of Medicine and Health Sciences, Washington DC, USA; Department of Tropical Medicine, Microbiology and Infectious Diseases, The George Washington University School of Medicine and Health Sciences, Washington DC, USA
| | - Sarah B Mulkey
- Zickler Family Prenatal Pediatrics Institute, Children's National Hospital, Washington DC, USA; Department of Pediatrics, The George Washington University School of Medicine and Health Sciences, Washington DC, USA; Department of Neurology and Rehabilitation Medicine, The George Washington University School of Medicine and Health Sciences, Washington DC, USA.
| |
Collapse
|
48
|
Salmon F, Kayem G, Maisonneuve E, Foix-L'Hélias L, Benhammou V, Kaminski M, Marchand-Martin L, Kana G, Subtil D, Lorthe E, Ancel PY, Letouzey M. Clinical Chorioamnionitis and Neurodevelopment at 5 Years of Age in Children Born Preterm: The EPIPAGE-2 Cohort Study. J Pediatr 2024; 267:113921. [PMID: 38242316 DOI: 10.1016/j.jpeds.2024.113921] [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: 03/23/2023] [Revised: 01/04/2024] [Accepted: 01/10/2024] [Indexed: 01/21/2024]
Abstract
OBJECTIVE To assess the association between clinical chorioamnionitis and neurodevelopmental disorders at 5 years of age in children born preterm. STUDY DESIGN EPIPAGE 2 is a national, population-based cohort study of children born before 35 weeks of gestation in France in 2011. We included infants born alive between 240/7 and 346/7 weeks after preterm labor or preterm premature rupture of membranes. Clinical chorioamnionitis was defined as maternal fever before labor (>37.8°C) with ≥2 of the following criteria: maternal tachycardia, hyperleukocytosis, uterine contractions, purulent amniotic fluid, or fetal tachycardia. The primary outcome was a composite, including cerebral palsy, coordination disorders, cognitive disorders, sensory disorders, or behavioral disorders. We also analyzed each of these disorders separately as secondary outcomes. We performed a multivariable analysis using logistic regression models. We accounted for the nonindependence of twins and missing data by generalized estimating equation models and multiple imputations, respectively. RESULTS Among 2927 children alive at 5 years of age, 124 (3%) were born in a context of clinical chorioamnionitis. Overall, 8.2% and 9.6% of children exposed and unexposed, respectively, to clinical chorioamnionitis had moderate-to-severe neurodevelopmental disorders. After multiple imputations and multivariable analysis, clinical chorioamnionitis was not associated with the occurrence of moderate-to-severe neurodevelopmental disorders (aOR, 0.9; 95% CI, 0.5-1.8). CONCLUSIONS We did not find any association between clinical chorioamnionitis and neurodevelopmental disorders at 5 years of age in children born at <35 weeks of gestation after preterm labor or preterm premature rupture of membrane.
Collapse
Affiliation(s)
- Fanny Salmon
- Université Paris Cité, Sorbonne Paris-Nord, Inserm, INRAE, CRESS, Obstetrical Perinatal and Pediatric Epidemiology Research Team, EPOPé, Paris, France; Department of Gynecology and Obstetrics, Robert Debré Hospital, AP-HP, Université Paris-Cité, Paris, France.
| | - Gilles Kayem
- Université Paris Cité, Sorbonne Paris-Nord, Inserm, INRAE, CRESS, Obstetrical Perinatal and Pediatric Epidemiology Research Team, EPOPé, Paris, France; Department of Gynecology and Obstetrics, Armand Trousseau Hospital, APHP, Paris Sorbonne University, Paris, France
| | - Emeline Maisonneuve
- Institute of Primary Health Care (BIHAM), University of Bern, Brisbane, Australia
| | - Laurence Foix-L'Hélias
- Université Paris Cité, Sorbonne Paris-Nord, Inserm, INRAE, CRESS, Obstetrical Perinatal and Pediatric Epidemiology Research Team, EPOPé, Paris, France; Department of Neonatal Pediatrics, Armand Trousseau Hospital, APHP, Paris Sorbonne University, Paris, France
| | - Valérie Benhammou
- Université Paris Cité, Sorbonne Paris-Nord, Inserm, INRAE, CRESS, Obstetrical Perinatal and Pediatric Epidemiology Research Team, EPOPé, Paris, France
| | - Monique Kaminski
- Université Paris Cité, Sorbonne Paris-Nord, Inserm, INRAE, CRESS, Obstetrical Perinatal and Pediatric Epidemiology Research Team, EPOPé, Paris, France
| | - Laetitia Marchand-Martin
- Université Paris Cité, Sorbonne Paris-Nord, Inserm, INRAE, CRESS, Obstetrical Perinatal and Pediatric Epidemiology Research Team, EPOPé, Paris, France
| | - Gildas Kana
- Université Paris Cité, Sorbonne Paris-Nord, Inserm, INRAE, CRESS, Obstetrical Perinatal and Pediatric Epidemiology Research Team, EPOPé, Paris, France
| | - Damien Subtil
- Department of Obstetrics and Gynecology, CHU Lille, EA 2694 METRICS, University of Lille, Lille, France
| | - Elsa Lorthe
- Université Paris Cité, Sorbonne Paris-Nord, Inserm, INRAE, CRESS, Obstetrical Perinatal and Pediatric Epidemiology Research Team, EPOPé, Paris, France; Department of Primary Care Medicine, Unit of Population Epidemiology, Geneva University Hospitals, Geneva, Switzerland
| | - Pierre-Yves Ancel
- Université Paris Cité, Sorbonne Paris-Nord, Inserm, INRAE, CRESS, Obstetrical Perinatal and Pediatric Epidemiology Research Team, EPOPé, Paris, France; Center for Clinical Investigation P1419, APHP, APHP. Centre- Université Paris Cité, Paris, France
| | - Mathilde Letouzey
- Université Paris Cité, Sorbonne Paris-Nord, Inserm, INRAE, CRESS, Obstetrical Perinatal and Pediatric Epidemiology Research Team, EPOPé, Paris, France; Department of Neonatal Pediatrics, Poissy Saint Germain Hospital, Clamart, France
| |
Collapse
|
49
|
Warintaksa P, Lertrat W, Romero R, Vivithanaporn P, Mongkolsuk P, Kamlungkuea T, Settacomkul R, Pongchaikul P, Chaemsaithong P. Anaphylaxis-induced premature uterine contractions: a case report and literature review. BMC Pregnancy Childbirth 2024; 24:197. [PMID: 38481196 PMCID: PMC10935842 DOI: 10.1186/s12884-024-06297-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 01/28/2024] [Indexed: 03/17/2024] Open
Abstract
BACKGROUND Preterm labor is caused by multiple etiologies, including intra-amniotic infection and/or intra-amniotic inflammation, vascular disorders, cervical disease, decidual senescence, and breakdown of maternal-fetal tolerance. Accumulating evidence in vivo and in vitro has shown that an allergic reaction, including anaphylaxis, can induce preterm uterine contractions. This report describes a case of a pregnant woman who developed anaphylaxis and regular uterine contractions after the ingestion of a strawberry-coated biscuit. We also review the mechanism of allergic reaction (hypersensitivity)-induced preterm labor. Case presentation A 31-year-old woman (gravida 1, para 0) at 30+2 weeks of gestation was admitted to the labor and delivery unit with regular uterine contractions and anaphylactic symptoms after she ingested a strawberry-coated biscuit as a snack. The uterine contractions resolved after the treatment of anaphylaxis by administering antihistamines and epinephrine. The patient subsequently delivered at 39+3 weeks of gestation. The amniotic fluid profile showed no infection or inflammation. A postpartum skin-prick test confirmed a positive type 1 hypersensitivity reaction to the strawberry-coated biscuit. CONCLUSIONS We report a case of anaphylaxis-induced uterine contractility in which uterine contractions subsided after the treatment of anaphylaxis. The absence of intra-amniotic infection and/or intra-amniotic inflammation and the cause of the anaphylaxis were confirmed. Our findings indicate that maternal allergic reactions may be one of the mechanisms of preterm labor.
Collapse
Affiliation(s)
- Puntabut Warintaksa
- Department of Obstetrics and Gynecology, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, 10400, Thailand
| | - Waranyu Lertrat
- Department of Obstetrics and Gynecology, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, 10400, Thailand
| | - Roberto Romero
- Pregnancy 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, 20892, USA.
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, 48109, USA.
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI, 48824, USA.
| | - Pornpun Vivithanaporn
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital Mahidol University, Samut Prakan, 10540, Thailand
| | - Paninee Mongkolsuk
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital Mahidol University, Samut Prakan, 10540, Thailand
| | - Threebhorn Kamlungkuea
- Department of Obstetrics and Gynecology, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, 10400, Thailand
| | - Rapeewan Settacomkul
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital Mahidol University, Samut Prakan, 10540, Thailand
| | - Pisut Pongchaikul
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital Mahidol University, Samut Prakan, 10540, Thailand.
- Integrative Computational BioScience Center, Mahidol University, Nakhon Pathom, 73170, Thailand.
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, L69 3BX, UK.
| | - Piya Chaemsaithong
- Department of Obstetrics and Gynecology, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, 10400, Thailand.
| |
Collapse
|
50
|
Olguín-Ortega A, Figueroa-Damian R, Palafox-Vargas ML, Reyes-Muñoz E. Risk of adverse perinatal outcomes among women with clinical and subclinical histopathological chorioamnionitis. Front Med (Lausanne) 2024; 11:1242962. [PMID: 38510456 PMCID: PMC10953497 DOI: 10.3389/fmed.2024.1242962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 02/19/2024] [Indexed: 03/22/2024] Open
Abstract
Introduction Histologic chorioamnionitis (HCA) is a placental inflammatory condition associated with adverse perinatal outcomes (APOs). This historical cohort study explores the risk of APOs in pregnant women with HCA and compares the impact of clinical chorioamnionitis (CCA) with subclinical chorioamnionitis (SCCA). Methodology Placentas were evaluated by a perinatal pathologist tand all women with HCA were included. Two groups were integrated: (1) women with clinical chorioamnionitis (CCA) and (2) women with subclinical chorioamnionitis (SCCA). Additionally, we conducted a secondary analysis to compare the prevalence of APOs among stage 1, 2 and 3 of HCA and the risk of APOs between grades 1 and 2 of HCA. The APOs analyzed were preterm birth, stillbirth, neonatal weight < 1,500 g, neonatal sepsis. Relative risk with 95% confidence interval was calculated. Results The study included 41 cases of CCA and 270 cases of SCCA. The mean gestational age at diagnosis and birth was 30.2 ± 5.4 weeks and 32.5 ± 5.1 weeks, for group 1 and 2, respectively. The study also found that women with HCA stage 3 and grade 2 had a higher prevalence and risk of adverse perinatal outcomes. Discussion The findings of this study suggest the importance of placental histological study to excluded SCCA, which represents a significant risk to both maternal and neonatal health, contributing to high morbidity and mortality.
Collapse
Affiliation(s)
- Andrea Olguín-Ortega
- Department of Gynecology, National Institute of Perinatology, Mexico City, Mexico
| | | | | | - Enrique Reyes-Muñoz
- Coordination of Gynecological and Perinatal Endocrinology, National Institute of Perinatology, Mexico City, Mexico
| |
Collapse
|