1
|
El Emrani S, Jansen EJS, Goeman JJ, Termote JUM, Lopriore E, Schalij-Delfos NE, van der Meeren LE. Comprehensive assessment of placental inflammation: Novel approach in predicting retinopathy of prematurity. Early Hum Dev 2025; 204:106239. [PMID: 40086020 DOI: 10.1016/j.earlhumdev.2025.106239] [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: 01/15/2025] [Revised: 03/03/2025] [Accepted: 03/06/2025] [Indexed: 03/16/2025]
Abstract
The purpose of this study was to determine the independent association between placental inflammation and the development of retinopathy of prematurity (ROP). This retrospective cohort study included 591 neonates born with a gestational age (GA) ≤32 weeks and/or a birthweight (BW) ≤1500 g. Clinical data were retrospectively collected, and placentas were reexamined for acute (e.g. chorioamnionitis and funisitis) and chronic placental inflammation (e.g. chorioamnionitis, villitis of unknown etiology and chronic deciduitis). Severe acute chorioamnionitis was defined as the presence of confluent polymorphonuclear leukocytes or subchorionic microabscesses. Outcomes explored were GA, BW, small for gestational age (SGA), mechanical ventilation duration, postnatal corticosteroids, sepsis, necrotizing enterocolitis, and ROP. Acute histological chorioamnionitis and funisitis were associated with lower GA, lower SGA rates, increased duration of mechanical ventilation and increased ROP rates, while chronic chorioamnionitis and villitis were associated with higher GA and increased SGA rates. BW was significantly lower in neonates with chronic deciduitis. Subanalysis of placentas without maternal and fetal vascular malperfusion also showed increased rates of severe acute chorioamnionitis (42 % vs. 21 %), funisitis (61 % vs. 35 %) in neonates with ROP. Multivariable regression analysis revealed two placental inflammatory factors to be independently associated with ROP: severe acute chorioamnionitis (OR 2.1; 95 % CI 1.1-3.8) and funisitis (OR 1.7; 95 % CI 1.0-2.9). Structured placental evaluation of the presence of severe acute chorioamnionitis and funisitis is valuable in predicting the development of ROP. This increased risk of ROP development could be integrated into the neonatal treatment approach of high-risk neonates in a very early stage in order to reduce ROP.
Collapse
Affiliation(s)
- Salma El Emrani
- Department of Ophthalmology, Leiden University Medical Center, Leiden, the Netherlands; Division of Neonatology, Willem-Alexander Children's Hospital, Department of Pediatrics, Leiden University Medical Center, Leiden, the Netherlands.
| | - Esther J S Jansen
- Division of Neonatology, Wilhelmina Children's Hospital, Department of Women and Neonate, 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
| | - Jacqueline U M Termote
- Division of Neonatology, Wilhelmina Children's Hospital, Department of Women and Neonate, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Enrico Lopriore
- Division of Neonatology, Willem-Alexander Children's Hospital, Department of Pediatrics, Leiden University Medical Center, Leiden, 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
|
2
|
El Emrani S, van der Meeren LE, Jansen EJ, Goeman JJ, Termote JU, Lopriore E, Schalij-Delfos NE. Early-Onset Sepsis as an Early Predictor for Retinopathy of Prematurity: A Meta-analysis. Am J Perinatol 2025; 42:387-394. [PMID: 39029916 PMCID: PMC11793952 DOI: 10.1055/a-2369-6690] [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: 04/09/2024] [Accepted: 07/16/2024] [Indexed: 07/21/2024]
Abstract
OBJECTIVE Neonatal sepsis has been established as a risk factor for retinopathy of prematurity (ROP) but previous meta-analyses have predominately focused on late-onset sepsis (LOS). This meta-analysis aims to explore the association between early-onset sepsis (EOS) and the risk of ROP. STUDY DESIGN Observational studies reporting (unadjusted) data on proven EOS in neonates with ROP were included. PubMed, Embase, and Cochrane Library were searched. Proven EOS was defined as a positive blood or cerebrospinal fluid culture. Effect sizes were calculated by using logistic random-effects models and meta-regression analyses. Primary outcomes were any stage ROP and severe ROP (≥stage 3, type I, aggressive [posterior] ROP, plus disease or requiring treatment). Potential confounders explored were gestational age at birth, birth weight, small for gestational age, maternal steroid use, necrotizing enterocolitis, LOS, and mechanical ventilation duration. RESULTS Seventeen studies reporting the incidence of proven EOS in neonates with ROP were included. Proven EOS showed no significant association with any stage ROP (odds ratio [OR] = 1.90; 95% confidence interval [CI]: 0.96-3.79, p = 0.067) but heterogeneity between studies was significantly high. Neonates with proven EOS had an increased risk for severe ROP (OR = 2.21; 95% CI: 1.68-2.90), and no significant confounders influencing this effect size were found in the meta-regression analysis. CONCLUSION Neonates with proven EOS are at increased risk of severe ROP. Neonatologists need to be aware that EOS is an early predictor of ROP and should adapt their policy and treatment decisions where possible to reduce ROP. KEY POINTS · This meta-analysis reveals a 2.2-fold increased risk of severe ROP in neonates with proven EOS.. · Future studies should distinguish between EOS and LOS when investigating risk factors of ROP.. · Treatment decisions should be adapted where possible in neonates with EOS before ROP screening begins..
Collapse
Affiliation(s)
- Salma El Emrani
- Department of Ophthalmology, Leiden University Medical Center, Leiden, The Netherlands
- Neonatology, Department of Pediatrics, Willem-Alexander Children's Hospital, Leiden University Medical Center, Leiden, 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
| | - Esther J.S. Jansen
- Neonatology, Department of Women and Neonate, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Jelle J. Goeman
- Medical Statistics, Department of Biomedical Data Science, Leiden University Medical Center, Leiden, The Netherlands
| | - Jacqueline U.M. Termote
- Neonatology, Department of Women and Neonate, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Enrico Lopriore
- Neonatology, Department of Pediatrics, Willem-Alexander Children's Hospital, Leiden University Medical Center, Leiden, The Netherlands
| | | |
Collapse
|
3
|
Zgutka K, Tkacz M, Grabowska M, Mikołajek-Bedner W, Tarnowski M. Sirtuins and Their Implications in the Physiopathology of Gestational Diabetes Mellitus. Pharmaceuticals (Basel) 2025; 18:41. [PMID: 39861104 PMCID: PMC11768332 DOI: 10.3390/ph18010041] [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: 12/04/2024] [Revised: 12/23/2024] [Accepted: 12/27/2024] [Indexed: 01/27/2025] Open
Abstract
Gestational diabetes mellitus (GDM) imposes serious short- and long-term health problems for the mother and her child. An effective therapeutic that can reduce the incidence of GDM and improve long-term outcomes is a major research priority and is very important for public health. Unfortunately, despite numerous studies, the molecular mechanisms underlying GDM are not fully defined and require further study. Chronic low-grade inflammation, oxidative stress, and insulin resistance are central features of pregnancies complicated by GDM. There is evidence of the involvement of sirtuins, which are NAD+-dependent histone deacetylases, in energy metabolism and inflammation. Taking these facts into consideration, the role of sirtuins in the pathomechanism of GDM will be discussed.
Collapse
Affiliation(s)
- Katarzyna Zgutka
- Department of Physiology in Health Sciences, Faculty of Health Sciences, Pomeranian Medical University, 70-210 Szczecin, Poland
| | - Marta Tkacz
- Department of Physiology in Health Sciences, Faculty of Health Sciences, Pomeranian Medical University, 70-210 Szczecin, Poland
| | - Marta Grabowska
- Department of Histology and Developmental Biology, Faculty of Health Sciences, Pomeranian Medical University, 71-210 Szczecin, Poland
| | - Wioletta Mikołajek-Bedner
- Department of Obstetrics and Gynecology, Pomeranian Medical University in Szczecin, 70-204 Szczecin, Poland
| | - Maciej Tarnowski
- Department of Physiology in Health Sciences, Faculty of Health Sciences, Pomeranian Medical University, 70-210 Szczecin, Poland
| |
Collapse
|
4
|
Lawrence SM, Wynn JL, Gordon SM. Neonatal bacteremia and sepsis. REMINGTON AND KLEIN'S INFECTIOUS DISEASES OF THE FETUS AND NEWBORN INFANT 2025:183-232.e25. [DOI: 10.1016/b978-0-323-79525-8.00015-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
5
|
Collins JM, Keane JM, Deady C, Khashan AS, McCarthy FP, O'Keeffe GW, Clarke G, Cryan JF, Caputi V, O'Mahony SM. Prenatal stress impacts foetal neurodevelopment: Temporal windows of gestational vulnerability. Neurosci Biobehav Rev 2024; 164:105793. [PMID: 38971516 DOI: 10.1016/j.neubiorev.2024.105793] [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: 04/09/2024] [Revised: 06/27/2024] [Accepted: 07/01/2024] [Indexed: 07/08/2024]
Abstract
Prenatal maternal stressors ranging in severity from everyday occurrences/hassles to the experience of traumatic events negatively impact neurodevelopment, increasing the risk for the onset of psychopathology in the offspring. Notably, the timing of prenatal stress exposure plays a critical role in determining the nature and severity of subsequent neurodevelopmental outcomes. In this review, we evaluate the empirical evidence regarding temporal windows of heightened vulnerability to prenatal stress with respect to motor, cognitive, language, and behavioural development in both human and animal studies. We also explore potential temporal windows whereby several mechanisms may mediate prenatal stress-induced neurodevelopmental effects, namely, excessive hypothalamic-pituitary-adrenal axis activity, altered serotonin signalling and sympathetic-adrenal-medullary system, changes in placental function, immune system dysregulation, and alterations of the gut microbiota. While broadly defined developmental windows are apparent for specific psychopathological outcomes, inconsistencies arise when more complex cognitive and behavioural outcomes are considered. Novel approaches to track molecular markers reflective of the underlying aetiologies throughout gestation to identify tractable biomolecular signatures corresponding to critical vulnerability periods are urgently required.
Collapse
Affiliation(s)
- James M Collins
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland.
| | - James M Keane
- APC Microbiome Ireland, University College Cork, Cork, Ireland.
| | - Clara Deady
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland.
| | - Ali S Khashan
- School of Public Health, University College Cork, Cork, Ireland; The Irish Centre for Maternal and Child Health Research (INFANT), Cork University Maternity Hospital, Cork, Ireland.
| | - Fergus P McCarthy
- The Irish Centre for Maternal and Child Health Research (INFANT), Cork University Maternity Hospital, Cork, Ireland; Department of Obstetrics and Gynaecology, University College Cork, Cork, Ireland.
| | - Gerard W O'Keeffe
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; The Irish Centre for Maternal and Child Health Research (INFANT), Cork University Maternity Hospital, Cork, Ireland.
| | - Gerard Clarke
- APC Microbiome Ireland, University College Cork, Cork, Ireland; The Irish Centre for Maternal and Child Health Research (INFANT), Cork University Maternity Hospital, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland.
| | - John F Cryan
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland.
| | - Valentina Caputi
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland.
| | | |
Collapse
|
6
|
Sheng JA, Tobet SA. Maternal immune activation with toll-like receptor 7 agonist during mid-gestation alters juvenile and adult developmental milestones and behavior. J Neuroendocrinol 2024; 36:e13417. [PMID: 38822791 PMCID: PMC11296912 DOI: 10.1111/jne.13417] [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: 11/08/2023] [Revised: 05/07/2024] [Accepted: 05/15/2024] [Indexed: 06/03/2024]
Abstract
Infections during pregnancy are associated with increased risk for adult neuropsychiatric disease, such as major depressive disorder, schizophrenia, and autism spectrum disorder. In mouse models of maternal immune activation (MIA), different toll-like receptors (TLRs) are stimulated to initiate inflammatory responses in mother and fetus. The goal of this study was to determine sex-dependent aspects of MIA using a TLR7/8 agonist, Resiquimod (RQ), on neurodevelopment. RQ was administered to timed-pregnant mice on embryonic day (E) 12.5. At E15, maternal/fetal plasma cytokines were measured by enzyme-linked immunosorbent assay (ELISA). Maternal cytokines interleukin (IL)-6 and IL-10 were higher while tumor necrosis factor (TNF)-α and IL-17 were lower in pregnant dams exposed to RQ. Fetal cytokines (E15) were altered at the same timepoint with fetal plasma IL-6 and IL-17 greater after RQ compared to vehicle, while IL-10 and TNF-α were higher in male fetuses but not female. Other timed-pregnant dams were allowed to give birth. MIA with RQ did not alter the female to male ratio of offspring born per litter. Body weights were reduced significantly in both sexes at birth, and over the next 5 weeks. Offspring from RQ-injected mothers opened their eyes 5 days later than controls. Similarly, female offspring from RQ-injected mothers exhibited pubertal delay based on vaginal opening 2-3 days later than control females. On the behavioral side, juvenile and adult male and female MIA offspring exhibited less social-like behavior in a social interaction test. Anhedonia-like behavior was greater in MIA adult female mice. This study provides support for sex-dependent influences of fetal antecedents for altered brain development and behavioral outputs that could be indicative of increased susceptibility for adult disorders through immune mechanisms. Future studies are needed to determine neural cellular and molecular mechanisms for such programming effects.
Collapse
Affiliation(s)
| | - Stuart A. Tobet
- Biomedical Sciences, Colorado State University, Fort Collins, CO
- Department of Psychiatry, Mass General Hospital, Harvard Medical School, Boston, MA
- School of Biomedical Engineering, Colorado State University, Fort Collins, CO
- Innovation Center on Sex Differences in Medicine, Mass General Hospital
| |
Collapse
|
7
|
Shen Q, Dong S, Shah NK, Liang Y, Wang J, Shan YH, He J. Peripartum outcomes and immune responses after SARS-CoV-2 infection in the third trimester of pregnancy. BMC Pregnancy Childbirth 2024; 24:498. [PMID: 39048938 PMCID: PMC11267945 DOI: 10.1186/s12884-024-06707-5] [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: 04/30/2024] [Accepted: 07/18/2024] [Indexed: 07/27/2024] Open
Abstract
BACKGROUND SARS-CoV-2 infection in pregnant women during the third trimester resulted in overall adverse pregnancy outcomes compared to non-infected controls and a unique humoral and cellular response at delivery. In this study we aimed to assess the impact of SARS-CoV-2 infection on maternal/neonatal peripartum outcomes andimmunological profiles. METHOD In this study, we recruited 304 SARS-CoV-2 infected pregnant women and 910 SARS-CoV-2 non-infected pregnant women who were admitted for delivery. Peripartum and neonates' outcomes response to SARS-CoV-2 infection were analyzed. Furthermore, we characterized the antibody and cytokines profile in SARS-CoV-2 infected maternal blood (MB) and cord blood (CB). We also assessed routine laboratory tests and liver function tests in MB before labor. Unpaired T test, Mann-Whitney test and Spearman test were used to analyze the data. RESULTS SARS-CoV-2 infected pregnant women were significantly associated with increased risk of adverse pregnancy outcomes, including preterm labor (13.8% vs. 9.5%, p = 0.033) and meconium-stained amniotic fluid (8.9% vs. 5.5%, p = 0.039). The risk of low birth weight (< 2500 g) (10.5% vs. 6.5%, p = 0.021) and Apgar score < 8 at 1-minute (9.2% vs. 5.8%, p = 0.049) significantly increased in newborns from COVID-19 positive mothers than their counterparts. Our results showed that antibodies were increased in adverse-outcome SARS-CoV-2 infected mothers and their neonates, and abnormal proportion of immune cells were detected in SARS-CoV-2 infected mothers. While the immune response showed no difference between adverse-outcome infected pregnant women and normal-outcome infected pregnant women. Thus, SARS-CoV-2 infection during the third trimester of pregnancy induced a unique humoral and cellular response at delivery. CONCLUSION SARS-CoV-2 infection closer to delivery could incline to adverse pregnancy outcomes. Therefore, the utmost care is required for SARS-CoV-2 infected pregnant women and their newborns. TRIAL REGISTRATION The study protocol was approved by the Institutional Review Board of the First Hospital of Jilin University with the approval code number 23K170-001, and informed consent was obtained from all enrolled patients prior to sample collection.
Collapse
Affiliation(s)
- Qi Shen
- Department of obstetrics, Obstetrics and Gynaecology Center, The First Hospital of Jilin University, Changchun, 130061, China
| | - Shuai Dong
- Department of obstetrics, Obstetrics and Gynaecology Center, The First Hospital of Jilin University, Changchun, 130061, China
| | - Neelam Kumari Shah
- Department of obstetrics, Obstetrics and Gynaecology Center, The First Hospital of Jilin University, Changchun, 130061, China
| | - Yuan Liang
- Department of obstetrics, Obstetrics and Gynaecology Center, The First Hospital of Jilin University, Changchun, 130061, China
| | - Jie Wang
- Department of obstetrics, Obstetrics and Gynaecology Center, The First Hospital of Jilin University, Changchun, 130061, China
| | - Yan-Hong Shan
- Department of obstetrics, Obstetrics and Gynaecology Center, The First Hospital of Jilin University, Changchun, 130061, China.
| | - Jin He
- Department of obstetrics, Obstetrics and Gynaecology Center, The First Hospital of Jilin University, Changchun, 130061, China.
| |
Collapse
|
8
|
McLellan J, Croen LA, Iosif AM, Ashwood P, Yoshida C, Berger K, Van de Water J. Differences in mid-gestational and early postnatal neonatal cytokines and chemokines are associated with patterns of maternal autoantibodies in the context of autism. Cereb Cortex 2024; 34:50-62. [PMID: 38696596 PMCID: PMC11065110 DOI: 10.1093/cercor/bhae082] [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: 11/01/2023] [Revised: 01/25/2024] [Accepted: 01/06/2024] [Indexed: 05/04/2024] Open
Abstract
Associations between maternal immune dysregulation (including autoimmunity and skewed cytokine/chemokine profiles) and offspring neurodevelopmental disorders such as autism have been reported. In maternal autoantibody-related autism, specific maternally derived autoantibodies can access the fetal compartment to target eight proteins critical for neurodevelopment. We examined the relationship between maternal autoantibodies to the eight maternal autoantibody-related autism proteins and cytokine/chemokine profiles in the second trimester of pregnancy in mothers of children later diagnosed with autism and their neonates' cytokine/chemokine profiles. Using banked maternal serum samples from 15 to 19 weeks of gestation from the Early Markers for Autism Study and corresponding banked newborn bloodspots, we identified three maternal/offspring groups based on maternal autoantibody status: (1) mothers with autoantibodies to one or more of the eight maternal autoantibody-related autismassociated proteins but not a maternal autoantibody-related autism-specific pattern, (2) mothers with a known maternal autoantibody-related autism pattern, and (3) mothers without autoantibodies to any of the eight maternal autoantibody-related autism proteins. Using a multiplex platform, we measured maternal second trimester and neonatal cytokine/chemokine levels. This combined analysis aimed to determine potential associations between maternal autoantibodies and the maternal and neonatal cytokine/chemokine profiles, each of which has been shown to have implications on offspring neurodevelopment independently.
Collapse
Affiliation(s)
- Janna McLellan
- Department of Internal Medicine, Division of Rheumatology, Allergy, and Clinical Immunology, University of California Davis, 451 Health Sciences Drive, Suite 6505C, Davis, CA 95616, United States
| | - Lisa A Croen
- Division of Research, Kaiser Permanente Northern California, 2000 Broadway, Oakland, CA 94612, United States
| | - Ana-Maria Iosif
- Department of Public Health Sciences, Division of Biostatistics, University of California Davis, Medical Sciences 1C, Davis, CA, 95616, United States
| | - Paul Ashwood
- MIND Institute, University of California Davis, 2805 Wet Lab Building, Sacramento, CA 95817, United States
- Department of Medical Microbiology and Immunology, University of California Davis, 3146 One Shields Avenue, Tupper Hall, Davis, CA 95616, United States
| | - Cathleen Yoshida
- Division of Research, Kaiser Permanente Northern California, 2000 Broadway, Oakland, CA 94612, United States
| | - Kimberly Berger
- Sequoia Foundation, 741 Addison Suite B, Berkeley, CA 94710, United States
| | - Judy Van de Water
- Department of Internal Medicine, Division of Rheumatology, Allergy, and Clinical Immunology, University of California Davis, 451 Health Sciences Drive, Suite 6505C, Davis, CA 95616, United States
- MIND Institute, University of California Davis, 2805 Wet Lab Building, Sacramento, CA 95817, United States
| |
Collapse
|
9
|
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
|
10
|
Perrone S, Carloni S, Dell'Orto VG, Filonzi L, Beretta V, Petrolini C, Lembo C, Buonocore G, Esposito S, Nonnis Marzano F. Hypoxic ischemic brain injury: animal models reveal new mechanisms of melatonin-mediated neuroprotection. Rev Neurosci 2024; 35:331-339. [PMID: 38153803 DOI: 10.1515/revneuro-2023-0126] [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: 10/13/2023] [Accepted: 11/24/2023] [Indexed: 12/30/2023]
Abstract
Oxidative stress (OS) and inflammation play a key role in the development of hypoxic-ischemic (H-I) induced brain damage. Following H-I, rapid neuronal death occurs during the acute phase of inflammation, and activation of the oxidant-antioxidant system contributes to the brain damage by activated microglia. So far, in an animal model of perinatal H-I, it was showed that neuroprostanes are present in all brain damaged areas, including the cerebral cortex, hippocampus and striatum. Based on the interplay between inflammation and OS, it was demonstrated in the same model that inflammation reduced brain sirtuin-1 expression and affected the expression of specific miRNAs. Moreover, through proteomic approach, an increased expression of genes and proteins in cerebral cortex synaptosomes has been revealed after induction of neonatal H-I. Administration of melatonin in the experimental treatment of brain damage and neurodegenerative diseases has produced promising therapeutic results. Melatonin protects against OS, contributes to reduce the generation of pro-inflammatory factors and promotes tissue regeneration and repair. Starting from the above cited aspects, this educational review aims to discuss the inflammatory and OS main pathways in H-I brain injury, focusing on the role of melatonin as neuroprotectant and providing current and emerging evidence.
Collapse
Affiliation(s)
- Serafina Perrone
- Neonatology Unit, Department of Medicine and Surgery, University of Parma, Pietro Barilla Children's Hospital, Via Gramsci 14, 43126 Parma, Italy
| | - Silvia Carloni
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Via Aurelio Saffi 2, 61029 Urbino, Italy
| | - Valentina Giovanna Dell'Orto
- Neonatology Unit, Department of Medicine and Surgery, University of Parma, Pietro Barilla Children's Hospital, Via Gramsci 14, 43126 Parma, Italy
| | - Laura Filonzi
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parco Area delle Scienze 11/A, 43124 Parma, Italy
| | - Virginia Beretta
- Neonatology Unit, Department of Medicine and Surgery, University of Parma, Pietro Barilla Children's Hospital, Via Gramsci 14, 43126 Parma, Italy
| | - Chiara Petrolini
- Neonatology Unit, Department of Medicine and Surgery, University of Parma, Pietro Barilla Children's Hospital, Via Gramsci 14, 43126 Parma, Italy
| | - Chiara Lembo
- Department of Neonatology, APHP, Necker-Enfants, Malades Hospital, 149 Rue de Sèvres, 75015 Paris, France
| | - Giuseppe Buonocore
- Department of Molecular and Developmental Medicine, University of Siena, Via Banchi di Sotto 55, 53100 Siena, Italy
| | - Susanna Esposito
- Pediatric Clinic, Department of Medicine and Surgery, University of Parma, Pietro Barilla Children's Hospital, Via Gramsci 14, 43126 Parma, Italy
| | - Francesco Nonnis Marzano
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parco Area delle Scienze 11/A, 43124 Parma, Italy
| |
Collapse
|
11
|
Pergialiotis V, Sapantzoglou I, Rodolaki K, Varthaliti A, Theodora M, Antsaklis P, Thomakos N, Stavros S, Daskalakis G, Papapanagiotou A. Maternal and neonatal outcomes following magnesium sulfate in the setting of chorioamnionitis: a meta-analysis. Arch Gynecol Obstet 2024; 309:917-927. [PMID: 37768342 PMCID: PMC10866770 DOI: 10.1007/s00404-023-07221-3] [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: 06/15/2023] [Accepted: 09/01/2023] [Indexed: 09/29/2023]
Abstract
PURPOSE Magnesium sulfate (MgSO4) has been widely used in obstetrics as a mean to help decrease maternal and neonatal morbidity in various antenatal pathology. As a factor, it seems to regulate immunity and can, thus, predispose to infectious morbidity. To date, it remains unknown if its administration can increase the risk of chorioamnionitis. In the present meta-analysis, we sought to accumulate the available evidence. METHODS We systematically searched Medline, Scopus, Clinicaltrials.gov, EMBASE, Cochrane Central Register of Controlled Trials CENTRAL, and Google Scholar databases in our primary search along with the reference lists of electronically retrieved full-text papers. RESULTS Eight studies were included that investigated the incidence of chorioamnionitis among parturient that received MgSO4 and control patients. Magnesium sulfate was administered in 3229 women and 3330 women served as controls as they did not receive MgSO4. The meta-analysis of data revealed that there was no association between the administration of magnesium sulfate and the incidence of chorioamnionitis (OR 0.98, 95% CI 0.73, 1.32). Rucker's analysis revealed that small studies did not significantly influence the statistical significance of this finding (OR 1.12, 95% CI 0.82, 1.53). Trial sequential analysis revealed that the required number to safely interpret the primary outcome was not reached. Two studies evaluated the impact of MgSO4 in neonates delivered in the setting of chorioamnionitis. Neither of these indicated the presence of a beneficial effect in neonatal morbidity, including the risk of cerebral palsy, intraventricular hemorrhage, necrotizing enterocolitis, bronchopulmonary dysplasia, sepsis, stillbirth, or neonatal death. CONCLUSION Current evidence indicates that magnesium sulfate is not associated with an increased risk of maternal chorioamnionitis. However, it should be noted that its effect on neonatal outcomes of offspring born in the setting of chorioamnionitis might be subtle if any, although the available evidence is very limited.
Collapse
Affiliation(s)
- Vasilios Pergialiotis
- First Department of Obstetrics and Gynecology, "Alexandra" General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Ioakim Sapantzoglou
- First Department of Obstetrics and Gynecology, "Alexandra" General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Kalliopi Rodolaki
- First Department of Obstetrics and Gynecology, "Alexandra" General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Antonia Varthaliti
- First Department of Obstetrics and Gynecology, "Alexandra" General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Marianna Theodora
- First Department of Obstetrics and Gynecology, "Alexandra" General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Panagiotis Antsaklis
- First Department of Obstetrics and Gynecology, "Alexandra" General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Nikolaos Thomakos
- First Department of Obstetrics and Gynecology, "Alexandra" General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Sofoklis Stavros
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Georgios Daskalakis
- First Department of Obstetrics and Gynecology, "Alexandra" General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Aggeliki Papapanagiotou
- Third Department of Obstetrics and Gynecology, Attikon General Hospital, National and Kapodistrian University of Athens, 2, Lourou Str., 11523, Athens, Greece.
| |
Collapse
|
12
|
Kontovazainitis CG, Gialamprinou D, Theodoridis T, Mitsiakos G. Hemostasis in Pre-Eclamptic Women and Their Offspring: Current Knowledge and Hemostasis Assessment with Viscoelastic Tests. Diagnostics (Basel) 2024; 14:347. [PMID: 38337863 PMCID: PMC10855316 DOI: 10.3390/diagnostics14030347] [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: 12/15/2023] [Revised: 01/22/2024] [Accepted: 01/30/2024] [Indexed: 02/12/2024] Open
Abstract
Pre-eclampsia (PE) is a placenta-mediated disease and remains a major cause of maternal and neonatal mortality and morbidity. As PE develops, normal pregnancy's hypercoagulable balance is disrupted, leading to platelet hyperactivation, excessive pathological hypercoagulability, and perturbed fibrinolysis. This narrative review aims to summarize the current knowledge regarding hemostasis in PE compared with healthy gestation and the potential effects of maternal PE on neonatal hemostasis. Finally, it aims to discuss hemostasis assessments for normal pregnancies and PE, emphasizing the role of viscoelastic tests, namely, thromboelastography (TEG) and thromboelastometry (ROTEM), for monitoring PE-associated hemostatic alterations. The use of TEG/ROTEM for assessing the hemostatic profile of PE women has been little considered, even though conventional coagulation tests (CCTs) have not helped to monitor hemostasis in this population. Compared with normal pregnancy, TEG/ROTEM in PE reveals an excessive hypercoagulability analogous with the severity of the disease, characterized by higher-stability fibrin clots. The TEG/ROTEM parameters can reflect PE severity and may be used for monitoring and as predictive markers for the disease.
Collapse
Affiliation(s)
- Christos-Georgios Kontovazainitis
- 2nd Neonatal Department and Neonatal Intensive Care Unit (NICU), “Papageorgiou” University Hospital, Aristotle University of Thessaloniki, 56403 Thessaloniki, Greece; (C.-G.K.); (D.G.)
| | - Dimitra Gialamprinou
- 2nd Neonatal Department and Neonatal Intensive Care Unit (NICU), “Papageorgiou” University Hospital, Aristotle University of Thessaloniki, 56403 Thessaloniki, Greece; (C.-G.K.); (D.G.)
| | - Theodoros Theodoridis
- 1st Department of Obstetrics and Gynecology, “Papageorgiou” University Hospital, Aristotle University of Thessaloniki, 56403 Thessaloniki, Greece;
| | - Georgios Mitsiakos
- 2nd Neonatal Department and Neonatal Intensive Care Unit (NICU), “Papageorgiou” University Hospital, Aristotle University of Thessaloniki, 56403 Thessaloniki, Greece; (C.-G.K.); (D.G.)
| |
Collapse
|
13
|
Dammann O, Stansfield BK. Neonatal sepsis as a cause of retinopathy of prematurity: An etiological explanation. Prog Retin Eye Res 2024; 98:101230. [PMID: 37984792 PMCID: PMC10842718 DOI: 10.1016/j.preteyeres.2023.101230] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 11/12/2023] [Accepted: 11/13/2023] [Indexed: 11/22/2023]
Abstract
Retinopathy of prematurity (ROP) is a complex neonatal disorder with multiple contributing factors. In this paper we have mounted the evidence in support of the proposal that neonatal sepsis meets all requirements for being a cause of ROP (not a condition, mechanism, or even innocent bystander) by means of initiating the early stages of the pathomechanism of ROP occurrence, systemic inflammation. We use the model of etiological explanation, which distinguishes between two overlapping processes in ROP causation. It can be shown that sepsis can initiate the early stages of the pathomechanism via systemic inflammation (causation process) and that systemic inflammation can contribute to growth factor aberrations and the retinal characteristics of ROP (disease process). The combined contribution of these factors with immaturity at birth (as intrinsic risk modifier) and prenatal inflammation (as extrinsic facilitator) seems to provide a cogent functional framework of ROP occurrence. Finally, we apply the Bradford Hill heuristics to the available evidence. Taken together, the above suggests that neonatal sepsis is a causal inducer of ROP.
Collapse
Affiliation(s)
- Olaf Dammann
- Dept. of Public Health & Community Medicine, Tufts University School of Medicine, Boston, USA; Dept. of Gynecology & Obstetrics, Hannover Medical School, Hannover, Germany; Dept. of Neuromedicine & Movement Science, Norwegian University of Science & Technology, Trondheim, Norway; Dept. of Philosophy, University of Johannesburg, Johannesburg, South Africa.
| | | |
Collapse
|
14
|
Gray JM, Major K, Castillo-Ruiz A, Shipley M, Gangappa S, Forger NG. The inflammatory response to birth requires MyD88 and is driven by both mother and offspring. Brain Behav Immun 2024; 115:617-630. [PMID: 37967662 PMCID: PMC11913035 DOI: 10.1016/j.bbi.2023.11.011] [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: 07/19/2023] [Revised: 10/15/2023] [Accepted: 11/11/2023] [Indexed: 11/17/2023] Open
Abstract
Birth is an inflammatory event for the newborn, characterized by elevations in interleukin (IL)-6, IL-10, and tumor necrosis factor (TNF)-α peripherally and/or centrally, as well as changes in brain microglia. However, the mechanism(s) underlying these responses is unknown. Toll-like receptors (TLRs) play crucial roles in innate immunity and initiate inflammatory cascades upon recognition of endogenous or exogenous antigens. Most TLR signaling depends on the adaptor molecule myeloid differentiation primary response 88 (MyD88). We independently varied MyD88 gene status in mouse dams and their offspring to determine whether the inflammatory response to birth depends on MyD88 signaling and, if so, whether that signaling occurs in the offspring, the mother, or both. We find that the perinatal surges in plasma IL-6 and brain expression of TNF-α depend solely on MyD88 gene status of the offspring, whereas postnatal increases in plasma IL-10 and TNF-α depend on MyD88 in both the pup and dam. Interestingly, MyD88 genotype of the dam primarily drives differences in offspring brain microglial density and has robust effects on developmental neuronal cell death. Milk cytokines were evaluated as a possible source of postnatal maternal influence; although we found high levels of CXCL1/GROα and several other cytokines in ingested post-partum milk, their presence did not require MyD88. Thus, the inflammatory response previously described in the late-term fetus and newborn depends on MyD88 (and, by extension, TLRs), with signaling in both the dam and offspring contributing. Unexpectedly, naturally-occuring neuronal cell death in the newborn is modulated primarily by maternal MyD88 gene status.
Collapse
Affiliation(s)
- Jennifer M Gray
- Neuroscience Institute, Georgia State University, Atlanta, GA 30303, USA
| | - Kharli Major
- Neuroscience Institute, Georgia State University, Atlanta, GA 30303, USA
| | | | - Michael Shipley
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, GA 30329, USA
| | - Shivaprakash Gangappa
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, GA 30329, USA
| | - Nancy G Forger
- Neuroscience Institute, Georgia State University, Atlanta, GA 30303, USA.
| |
Collapse
|
15
|
Xie F, Li L, Peng M, Zhang H. Overexpression of miR-199a-5p improves brain injury in newborn rats with intrauterine infection via inhibition of astrocyte activation. Brain Res 2023; 1820:148560. [PMID: 37648092 DOI: 10.1016/j.brainres.2023.148560] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/17/2023] [Accepted: 08/27/2023] [Indexed: 09/01/2023]
Abstract
White matter injury is the most common form of brain injury in preterm infants. In addition to hypoxia ischemia, intrauterine infection is most closely related to brain white matter injury. Our study aimed to explore the mechanism of the miR-199a-5p/HIF-1α axis on astrocyte activation and brain injury in newborn rats caused by intrauterine infection. The animal/cell model was established via escherichia coli infection/lipopolysaccharide induction, followed by the measurement of body weight, brain weight, and the pathological changes in brain tissues of newborn rats, and the pathological changes in placenta and uterus wall of pregnant rats. Also, the levels of GFAP, TNF-α, MDA, GSH, SOD, miR-199a-5p, and HIF-1α were detected though corresponding assays or kits. In vitro, cell viability and apoptosis and the levels of IL-6 and TNF-α were evaluated in astrocytes. Moreover, the targeting relationship between miR-199a-5p and HIF-1α was verified. miR-199a-5p was lowly expressed in the brain tissues of newborn rats with intrauterine infection. Overexpression of miR-199a-5p relieved the injury of placenta and uterus wall in pregnant rats and brain injury in newborn rats, accompanied by decreased HIF-1α, GFAP, TNF-α, and MDA levels and increased GSH and SOD levels. Results from cell models showed that miR-199a-5p overexpression inhibited astrocyte activation, shown by enhanced cell viability, weakened cell apoptosis, and decreased GFAP, IL-6, and TNF-α. Mechanistically, miR-199a-5p targeted HIF-1α to decrease its expression. Collectively, miR-199a-5p inhibited astrocyte activation and alleviated brain injury in newborn rats with intrauterine infection by reducing HIF-1α expression.
Collapse
Affiliation(s)
- Fan Xie
- Obstetrics Department, Maternal and Child Health Hospital of Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, NO.745 Wuluo Road, Hongshan District, Wuhan, Hubei 430070, PR China
| | - Li Li
- Obstetrics Department, Maternal and Child Health Hospital of Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, NO.745 Wuluo Road, Hongshan District, Wuhan, Hubei 430070, PR China
| | - Min Peng
- Obstetrics Department, Maternal and Child Health Hospital of Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, NO.745 Wuluo Road, Hongshan District, Wuhan, Hubei 430070, PR China.
| | - Huan Zhang
- Obstetrics Department, Maternal and Child Health Hospital of Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, NO.745 Wuluo Road, Hongshan District, Wuhan, Hubei 430070, PR China.
| |
Collapse
|
16
|
Menon R, Muglia LJ, Levin LH. Review on new approach methods to gain insight into the feto-maternal interface physiology. Front Med (Lausanne) 2023; 10:1304002. [PMID: 38098843 PMCID: PMC10720461 DOI: 10.3389/fmed.2023.1304002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 11/13/2023] [Indexed: 12/17/2023] Open
Abstract
Non-human animals represent a large and important feature in the history of biomedical research. The validity of their use, in terms of reproducible outcomes and translational confidence to the human situation, as well as ethical concerns surrounding that use, have been and remain controversial topics. Over the last 10 years, the communities developing microphysiological systems (MPS) have produced new approach method (NAMs) such as organoids and organs-on-a-chip. These alternative methodologies have shown indications of greater reliability and translatability than animal use in some areas, represent more humane substitutions for animals in these settings, and - with continued scientific effort - may change the conduct of basic research, clinical studies, safety testing, and drug development. Here, we present an introduction to these more human-relevant methodologies and suggest how a suite of pregnancy associated feto-maternal interface system-oriented NAMs may be integrated as reliable partial-/full animal replacements for investigators, significantly aid animal-/environmental welfare, and improve healthcare outcomes.
Collapse
Affiliation(s)
- Ramkumar Menon
- Department of Obstetrics and Gynecology, Division of Basic Science and Translational Research, The University of Texas Medical Branch at Galveston, Galveston, TX, United States
| | - Louis J. Muglia
- The Burroughs Wellcome Fund, Research Triangle Park, NC, United States
- Cincinnati Children’s Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | | |
Collapse
|
17
|
Kitase Y, Madurai NK, Hamimi S, Hellinger RL, Odukoya OA, Ramachandra S, Muthukumar S, Vasan V, Sevensky R, Kirk SE, Gall A, Heck T, Ozen M, Orsburn BC, Robinson S, Jantzie LL. Chorioamnionitis disrupts erythropoietin and melatonin homeostasis through the placental-fetal-brain axis during critical developmental periods. Front Physiol 2023; 14:1201699. [PMID: 37546540 PMCID: PMC10398572 DOI: 10.3389/fphys.2023.1201699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 07/06/2023] [Indexed: 08/08/2023] Open
Abstract
Introduction: Novel therapeutics are emerging to mitigate damage from perinatal brain injury (PBI). Few newborns with PBI suffer from a singular etiology. Most experience cumulative insults from prenatal inflammation, genetic and epigenetic vulnerability, toxins (opioids, other drug exposures, environmental exposure), hypoxia-ischemia, and postnatal stressors such as sepsis and seizures. Accordingly, tailoring of emerging therapeutic regimens with endogenous repair or neuro-immunomodulatory agents for individuals requires a more precise understanding of ligand, receptor-, and non-receptor-mediated regulation of essential developmental hormones. Given the recent clinical focus on neurorepair for PBI, we hypothesized that there would be injury-induced changes in erythropoietin (EPO), erythropoietin receptor (EPOR), melatonin receptor (MLTR), NAD-dependent deacetylase sirtuin-1 (SIRT1) signaling, and hypoxia inducible factors (HIF1α, HIF2α). Specifically, we predicted that EPO, EPOR, MLTR1, SIRT1, HIF1α and HIF2α alterations after chorioamnionitis (CHORIO) would reflect relative changes observed in human preterm infants. Similarly, we expected unique developmental regulation after injury that would reveal potential clues to mechanisms and timing of inflammatory and oxidative injury after CHORIO that could inform future therapeutic development to treat PBI. Methods: To induce CHORIO, a laparotomy was performed on embryonic day 18 (E18) in rats with transient uterine artery occlusion plus intra-amniotic injection of lipopolysaccharide (LPS). Placentae and fetal brains were collected at 24 h. Brains were also collected on postnatal day 2 (P2), P7, and P21. EPO, EPOR, MLTR1, SIRT1, HIF1α and HIF2α levels were quantified using a clinical electrochemiluminescent biomarker platform, qPCR, and/or RNAscope. MLT levels were quantified with liquid chromatography mass spectrometry. Results: Examination of EPO, EPOR, and MLTR1 at 24 h showed that while placental levels of EPO and MLTR1 mRNA were decreased acutely after CHORIO, cerebral levels of EPO, EPOR and MLTR1 mRNA were increased compared to control. Notably, CHORIO brains at P2 were SIRT1 mRNA deficient with increased HIF1α and HIF2α despite normalized levels of EPO, EPOR and MLTR1, and in the presence of elevated serum EPO levels. Uniquely, brain levels of EPO, EPOR and MLTR1 shifted at P7 and P21, with prominent CHORIO-induced changes in mRNA expression. Reductions at P21 were concomitant with increased serum EPO levels in CHORIO rats compared to controls and variable MLT levels. Discussion: These data reveal that commensurate with robust inflammation through the maternal placental-fetal axis, CHORIO impacts EPO, MLT, SIRT1, and HIF signal transduction defined by dynamic changes in EPO, EPOR, MLTR1, SIRT1, HIF1α and HIF2α mRNA, and EPO protein. Notably, ligand-receptor mismatch, tissue compartment differential regulation, and non-receptor-mediated signaling highlight the importance, complexity and nuance of neural and immune cell development and provide essential clues to mechanisms of injury in PBI. As the placenta, immune cells, and neural cells share many common, developmentally regulated signal transduction pathways, further studies are needed to clarify the perinatal dynamics of EPO and MLT signaling and to capitalize on therapies that target endogenous neurorepair mechanisms.
Collapse
Affiliation(s)
- Yuma Kitase
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Nethra K. Madurai
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Sarah Hamimi
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Ryan L. Hellinger
- Division of Pediatric Neurosurgery, Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - O. Angel Odukoya
- Division of Pediatric Neurosurgery, Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Sindhu Ramachandra
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Sankar Muthukumar
- Division of Pediatric Neurosurgery, Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Vikram Vasan
- Division of Pediatric Neurosurgery, Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Riley Sevensky
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Shannon E. Kirk
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Alexander Gall
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Timothy Heck
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Maide Ozen
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Benjamin C. Orsburn
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Shenandoah Robinson
- Division of Pediatric Neurosurgery, Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Lauren L. Jantzie
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Division of Pediatric Neurosurgery, Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Kennedy Krieger Institute, Baltimore, MD, United States
| |
Collapse
|
18
|
Woods R, Lorusso J, Fletcher J, ElTaher H, McEwan F, Harris I, Kowash H, D'Souza SW, Harte M, Hager R, Glazier JD. Maternal immune activation and role of placenta in the prenatal programming of neurodevelopmental disorders. Neuronal Signal 2023; 7:NS20220064. [PMID: 37332846 PMCID: PMC10273029 DOI: 10.1042/ns20220064] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 05/16/2023] [Accepted: 05/17/2023] [Indexed: 06/20/2023] Open
Abstract
Maternal infection during pregnancy, leading to maternal immune activation (mIA) and cytokine release, increases the offspring risk of developing a variety of neurodevelopmental disorders (NDDs), including schizophrenia. Animal models have provided evidence to support these mechanistic links, with placental inflammatory responses and dysregulation of placental function implicated. This leads to changes in fetal brain cytokine balance and altered epigenetic regulation of key neurodevelopmental pathways. The prenatal timing of such mIA-evoked changes, and the accompanying fetal developmental responses to an altered in utero environment, will determine the scope of the impacts on neurodevelopmental processes. Such dysregulation can impart enduring neuropathological changes, which manifest subsequently in the postnatal period as altered neurodevelopmental behaviours in the offspring. Hence, elucidation of the functional changes that occur at the molecular level in the placenta is vital in improving our understanding of the mechanisms that underlie the pathogenesis of NDDs. This has notable relevance to the recent COVID-19 pandemic, where inflammatory responses in the placenta to SARS-CoV-2 infection during pregnancy and NDDs in early childhood have been reported. This review presents an integrated overview of these collective topics and describes the possible contribution of prenatal programming through placental effects as an underlying mechanism that links to NDD risk, underpinned by altered epigenetic regulation of neurodevelopmental pathways.
Collapse
Affiliation(s)
- Rebecca M. Woods
- Division of Evolution, Infection and Genomics, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, U.K
| | - Jarred M. Lorusso
- Division of Evolution, Infection and Genomics, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, U.K
| | - Jennifer Fletcher
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Medicine, Biology and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, U.K
| | - Heidi ElTaher
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Medicine, Biology and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, U.K
- Department of Physiology, Faculty of Medicine, Alexandria University, Egypt
| | - Francesca McEwan
- Division of Evolution, Infection and Genomics, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, U.K
| | - Isabella Harris
- Division of Evolution, Infection and Genomics, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, U.K
| | - Hager M. Kowash
- Division of Developmental Biology and Medicine, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9WL, U.K
| | - Stephen W. D'Souza
- Division of Developmental Biology and Medicine, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9WL, U.K
| | - Michael Harte
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Medicine, Biology and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, U.K
| | - Reinmar Hager
- Division of Evolution, Infection and Genomics, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, U.K
| | - Jocelyn D. Glazier
- Division of Evolution, Infection and Genomics, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, U.K
| |
Collapse
|
19
|
Song WS, Hung TH, Liu SH, Zheng YT, Lin HM, Yang FY. Neuroprotection by Abdominal Ultrasound in Lipopolysaccharide-Induced Systemic Inflammation. Int J Mol Sci 2023; 24:ijms24119329. [PMID: 37298275 DOI: 10.3390/ijms24119329] [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: 03/22/2023] [Revised: 05/10/2023] [Accepted: 05/24/2023] [Indexed: 06/12/2023] Open
Abstract
Systemic inflammation is associated with intestinal inflammation and neuroinflammation by imbalancing the gut-brain axis. Low-intensity pulsed ultrasound (LIPUS) has neuroprotective and anti-inflammatory effects. This study explored LIPUS's neuroprotective effects against lipopolysaccharide (LPS)-induced neuroinflammation through transabdominal stimulation. Male C57BL/6J mice were intraperitoneally injected with LPS (0.75 mg/kg) daily for seven days, and abdominal LIPUS was applied to the abdominal area for 15 min/day during the last six days. One day after the last LIPUS treatment, biological samples were collected for microscopic and immunohistochemical analysis. Histological examination showed that LPS administration leads to tissue damage in the colon and brain. Transabdominal LIPUS stimulation attenuated colonic damage, reducing histological score, colonic muscle thickness, and villi shortening. Furthermore, abdominal LIPUS reduced hippocampal microglial activation (labeled by ionized calcium-binding adaptor molecule-1 [Iba-1]) and neuronal cell loss (labeled by microtubule-associated protein 2 [MAP2]). Moreover, abdominal LIPUS attenuated the number of apoptotic cells in the hippocampus and cortex. Altogether, our results indicate that abdominal LIPUS stimulation attenuates LPS-induced colonic inflammation and neuroinflammation. These findings provide new insights into the treatment strategy for neuroinflammation-related brain disorders and may facilitate method development through the gut-brain axis pathway.
Collapse
Affiliation(s)
- Wen-Shin Song
- Division of Neurosurgery, Cheng Hsin General Hospital, Taipei 112, Taiwan
- Department of Neurological Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan
| | - Tai-Ho Hung
- Department of Obstetrics and Gynecology, Taipei Chang Gung Memorial Hospital, Taipei 105, Taiwan
- Department of Obstetrics and Gynecology, Keelung Chang Gung Memorial Hospital, Keelung 204, Taiwan
| | - Shing-Hwa Liu
- Institute of Toxicology, College of Medicine, National Taiwan University, Taipei 106, Taiwan
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung 404, Taiwan
| | - Yin-Ting Zheng
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Hsin-Mei Lin
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Feng-Yi Yang
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| |
Collapse
|
20
|
Crocker CE, Sharmeen R, Tran TT, Khan AM, Li W, Alcorn JL. Surfactant protein a attenuates generalized and localized neuroinflammation in neonatal mice. Brain Res 2023; 1807:148308. [PMID: 36871846 PMCID: PMC10065943 DOI: 10.1016/j.brainres.2023.148308] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 02/24/2023] [Accepted: 02/28/2023] [Indexed: 03/06/2023]
Abstract
Surfactant protein A (SP-A) has important roles in innate immunity and modulation of pulmonary and extrapulmonary inflammation. Given SP-A has been detected in rat and human brain, we sought to determine if SP-A has a role in modulating inflammation in the neonatal mouse brain. Neonatal wildtype (WT) and SP-A-deficient (SP-A-/-) mice were subjected to three models of brain inflammation: systemic sepsis, intraventricular hemorrhage (IVH) and hypoxic-ischemic encephalopathy (HIE). Following each intervention, RNA was isolated from brain tissue and expression of cytokine and SP-A mRNA was determined by real-time quantitative RT-PCR analysis. In the sepsis model, expression of most cytokine mRNAs was significantly increased in brains of WT and SP-A-/- mice with significantly greater expression of all cytokine mRNA levels in SP-A-/- mice compared to WT. In the IVH model, expression of all cytokine mRNAs was significantly increased in WT and SP-A-/- mice and levels of most cytokine mRNAs were significantly increased in SP-A-/- mice compared to WT. In the HIE model, only TNF-α mRNA levels were significantly increased in WT brain tissue while all pro-inflammtory cytokine mRNAs were significantly increased in SP-A-/- mice, and all pro-inflammatory cytokine mRNA levels were significantly higher in SP-A-/- mice compared to WT. SP-A mRNA was not detectable in brain tissue of adult WT mice nor in WT neonates subjected to these models. These results suggest that SP-A-/- neonatal mice subjected to models of neuroinflammation are more susceptible to both generalized and localized neuroinflammation compared to WT mice, thus supporting the hypothesis that SP-A attenuates inflammation in neonatal mouse brain.
Collapse
Affiliation(s)
- Caroline E Crocker
- Division of Neonatology, Department of Pediatrics, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Romana Sharmeen
- Department of Neurology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Thu T Tran
- Division of Neonatology, Department of Pediatrics, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Amir M Khan
- Division of Neonatology, Department of Pediatrics, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Wen Li
- Division of Clinical and Translational Sciences, Department of Internal Medicine, the University of Texas McGovern Medical School at Houston, Houston, TX 77030, USA; Biostatistics/Epidemiology/Research Design Component, Center for Clinical and Translational Sciences, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Joseph L Alcorn
- Division of Neonatology, Department of Pediatrics, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA; Pediatric Research Center, Department of Pediatrics, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA.
| |
Collapse
|
21
|
Yang Y, Shen Y, Lin J, Dai S, Lu X, Xun G, Li Y, Wu R, Xia K, Luo X, Zhao J, Ou J. Association between history of miscarriage and autism spectrum disorder. Eur Arch Psychiatry Clin Neurosci 2023; 273:687-697. [PMID: 36251093 DOI: 10.1007/s00406-022-01494-6] [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: 02/22/2022] [Accepted: 09/13/2022] [Indexed: 11/03/2022]
Abstract
This case-control study was designed to examine the association between different types of miscarriage history and autism spectrum disorder (ASD), and determine whether the number of miscarriage history affects the risk of ASD. All of 2274 children with ASD and 1086 healthy controls were recruited. Sociodemographic and prenatal, perinatal, and neonatal characteristics were compared between the two groups. Multivariable logistic regression analyses were applied to investigate association between miscarriage history and ASD. Stratified analyses based on sex and types of miscarriages were similarly performed. History of miscarriage was potential risk factors for ASD ([aOR] = 2.919; 95% [CI] = 2.327-3.517). Stratified analyses revealed that induced ([aOR] = 2.763, 95% [CI] = 2.259-3.379) and spontaneous miscarriage history ([aOR] = 3.341, 95% [CI] = 1.939-4.820) were associated with high risk of ASD, respectively. A sex-biased ratio in the risk of ASD was observed between females ([aOR] = 3.049, 95% [CI] = 2.153-4.137) and males ([aOR] = 2.538, 95% [CI] = 1.978-3.251). Stratified analysis of induced miscarriage history revealed that only iatrogenic miscarriage history was associated with an increased risk ASD ([aOR] = 2.843, 95% [CI] = 1.534-4.268). Also, multiple spontaneous miscarriage histories ([aOR] = 1.836, 95% [CI] = 1.252-2.693) were associated with higher autism risk than one spontaneous miscarriages history ([aOR] = 3.016, 95% [CI] = 1.894-4.174). In conclusion, miscarriage history is related to an increased risk for ASD in offspring, which is affected by the types of miscarriage and sex of the fetus.
Collapse
Affiliation(s)
- Ye Yang
- Department of Psychiatry, and National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China
| | - Yidong Shen
- Department of Psychiatry, and National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China
| | - Jingjing Lin
- Department of Psychiatry, and National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China
| | - Si Dai
- Department of Psychiatry, and National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China
| | - Xiaozi Lu
- Qingdao Mental Health Center, Qingdao, 266034, Shandong, China
| | - Guanglei Xun
- Shandong Mental Health Center, 49 East Wenhua Road, Jinan, 250014, Shandong, China
| | - Yamin Li
- Clinical Nursing Teaching and Research Section, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Renrong Wu
- Department of Psychiatry, and National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China
| | - Kun Xia
- Center for Medical Genetics and School of Life Sciences, Central South University, Changsha, 410078, Hunan, China
| | - Xuerong Luo
- Department of Psychiatry, and National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China
| | - Jingping Zhao
- Department of Psychiatry, and National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China
| | - Jianjun Ou
- Department of Psychiatry, and National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China.
| |
Collapse
|
22
|
Gervasi MT, Romero R, Cainelli E, Veronese P, Tran MR, Jung E, Suksai M, Bosco M, Gotsch F. Intra-amniotic inflammation in the mid-trimester of pregnancy is a risk factor for neuropsychological disorders in childhood. J Perinat Med 2023; 51:363-378. [PMID: 36173676 PMCID: PMC10010737 DOI: 10.1515/jpm-2022-0255] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 07/17/2022] [Indexed: 11/15/2022]
Abstract
OBJECTIVES Intra-amniotic inflammation is a subclinical condition frequently caused by either microbial invasion of the amniotic cavity or sterile inflammatory stimuli, e.g., alarmins. An accumulating body of evidence supports a role for maternal immune activation in the genesis of fetal neuroinflammation and the occurrence of neurodevelopmental disorders such as cerebral palsy, schizophrenia, and autism. The objective of this study was to determine whether fetal exposure to mid-trimester intra-amniotic inflammation is associated with neurodevelopmental disorders in children eight to 12 years of age. METHODS This is a retrospective case-control study comprising 20 children with evidence of prenatal exposure to intra-amniotic inflammation in the mid-trimester and 20 controls matched for gestational age at amniocentesis and at delivery. Amniotic fluid samples were tested for concentrations of interleukin-6 and C-X-C motif chemokine ligand 10, for bacteria by culture and molecular microbiologic methods as well as by polymerase chain reaction for eight viruses. Neuropsychological testing of children, performed by two experienced psychologists, assessed cognitive and behavioral domains. Neuropsychological dysfunction was defined as the presence of an abnormal score (<2 standard deviations) on at least two cognitive tasks. RESULTS Neuropsychological dysfunction was present in 45% (9/20) of children exposed to intra-amniotic inflammation but in only 10% (2/20) of those in the control group (p=0.03). The relative risk (RR) of neuropsychological dysfunction conferred by amniotic fluid inflammation remained significant after adjusting for gestational age at delivery [aRR=4.5 (1.07-16.7)]. Of the 11 children diagnosed with neuropsychological dysfunction, nine were delivered at term and eight of them had mothers with intra-amniotic inflammation. Children exposed to intra-amniotic inflammation were found to have abnormalities in neuropsychological tasks evaluating complex skills, e.g., auditory attention, executive functions, and social skills, whereas the domains of reasoning, language, and memory were not affected in the cases and controls. CONCLUSIONS Asymptomatic sterile intra-amniotic inflammation in the mid-trimester of pregnancy, followed by a term birth, can still confer to the offspring a substantial risk for neurodevelopmental disorders in childhood. Early recognition and treatment of maternal immune activation in pregnancy may be a strategy for the prevention of subsequent neurodevelopmental disorders in offspring.
Collapse
Affiliation(s)
- Maria Teresa Gervasi
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD, USA, and Detroit, MI, USA
- Gynaecology and Obstetrics Unit, Department of Women’s and Children’s Health, University Hospital of Padua, Padua, Italy
| | - Roberto Romero
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD, USA, and Detroit, MI, USA
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, USA
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI, USA
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, Michigan, USA
- Detroit Medical Center, Detroit, MI, USA
| | - Elisa Cainelli
- Department of General Psychology, University of Padova, Padova, Italy
| | - Paola Veronese
- Maternal-Fetal Medicine Unit, Department of Women’s and Children’s Health, AOPD, Padua, Italy
| | - Maria Rosa Tran
- Gynaecology and Obstetrics Unit, Department of Women’s and Children’s Health, University Hospital of Padua, Padua, Italy
| | - Eunjung Jung
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD, USA, and Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Manaphat Suksai
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD, USA, and Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Mariachiara Bosco
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD, USA, and Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Francesca Gotsch
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD, USA, and Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| |
Collapse
|
23
|
Intrauterine Inflammation Leads to Select Sex- and Age-Specific Behavior and Molecular Differences in Mice. Int J Mol Sci 2022; 24:ijms24010032. [PMID: 36613475 PMCID: PMC9819857 DOI: 10.3390/ijms24010032] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Revised: 11/30/2022] [Accepted: 12/15/2022] [Indexed: 12/24/2022] Open
Abstract
Sex-specific differences in behavior have been observed in anxiety and learning in children exposed to prenatal inflammation; however, whether these behaviors manifest differently by age is unknown. This study assesses possible behavioral changes due to in utero inflammation as a function of age in neonatal, juvenile, and adult animals and presents potential molecular targets for observed differences. CD-1 timed pregnant dams were injected in utero with lipopolysaccharide (LPS, 50 μg/animal) or saline at embryonic day 15. No differences in stress responses were measured by neonatal ultrasonic vocalizations between LPS- and saline-exposed groups of either sex. By contrast, prenatal inflammation caused a male-specific increase in anxiety in mature but not juvenile animals. Juvenile LPS-exposed females had decreased movement in open field testing that was not present in adult animals. We additionally observed improved memory retrieval after in utero LPS in the juvenile animals of both sexes, which in males may be related to a perseverative phenotype. However, there was an impairment of long-term memory in only adult LPS-exposed females. Finally, gene expression analyses revealed that LPS induced sex-specific changes in genes involved in hippocampal neurogenesis. In conclusion, intrauterine inflammation has age- and sex-specific effects on anxiety and learning that may correlate to sex-specific disruption of gene expression associated with neurogenesis in the hippocampus.
Collapse
|
24
|
Kelly RS, Lee-Sarwar K, Chen YC, Laranjo N, Fichorova R, Chu SH, Prince N, Lasky-Su J, Weiss ST, Litonjua AA. Maternal Inflammatory Biomarkers during Pregnancy and Early Life Neurodevelopment in Offspring: Results from the VDAART Study. Int J Mol Sci 2022; 23:15249. [PMID: 36499584 PMCID: PMC9739845 DOI: 10.3390/ijms232315249] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 11/30/2022] [Accepted: 12/01/2022] [Indexed: 12/12/2022] Open
Abstract
Maternal infection and stress during the prenatal period have been associated with adverse neurodevelopmental outcomes in offspring, suggesting that biomarkers of increased inflammation in the mothers may associate with poorer developmental outcomes. In 491 mother-child pairs from the Vitamin D Antenatal Asthma Reduction Trial (VDAART), we investigated the association between maternal levels of two inflammatory biomarkers; interleukin-8 (IL-8) and C-Reactive Protein (CRP) during early (10-18 wks) and late (32-38 wks) pregnancy with offspring scores in the five domains of the Ages and Stages Questionnaire, a validated screening tool for assessing early life development. We identified a robust association between early pregnancy IL-8 levels and decreased fine-motor (β: -0.919, 95%CI: -1.425, -0.414, p = 3.9 × 10-4) and problem-solving skills at age two (β: -1.221, 95%CI: -1.904, -0.414, p = 4.9 × 10-4). Associations between IL-8 with other domains of development and those for CRP did not survive correction for multiple testing. Similarly, while there was some evidence that the detrimental effects of early pregnancy IL-8 were strongest in boys and in those who were not breastfed, these interactions were not robust to correction for multiple testing. However, further research is required to determine if other maternal inflammatory biomarkers associate with offspring neurodevelopment and work should continue to focus on the management of factors leading to increases in IL-8 levels in pregnant women.
Collapse
Affiliation(s)
- Rachel S. Kelly
- Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Kathleen Lee-Sarwar
- Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
- Division of Allergy and Clinical Immunology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02467, USA
| | - Yih-Chieh Chen
- Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
- Division of Allergy and Clinical Immunology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02467, USA
| | - Nancy Laranjo
- Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Raina Fichorova
- Laboratory of Genital Tract Biology, Department of Obstetrics, Gynecology and Reproductive Biology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Su H. Chu
- Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Nicole Prince
- Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Jessica Lasky-Su
- Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Scott T. Weiss
- Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Augusto A. Litonjua
- Division of Pediatric Pulmonary Medicine, Golisano Children’s Hospital at Strong, University of Rochester Medical Center, Rochester, NY 14642, USA
| |
Collapse
|
25
|
Affiliation(s)
- Lauren L Jantzie
- Division of Neonatal-Perinatal Medicine, Departments of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America; Neurosciences Intensive Care Nursery, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America; Division of Pediatric Neurosurgery, Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America; Kennedy Krieger Institute, Baltimore, MD, United States of America
| |
Collapse
|
26
|
Pregestational Exposure to T. gondii Produces Maternal Antibodies That Recognize Fetal Brain Mimotopes and Induces Neurochemical and Behavioral Dysfunction in the Offspring. Cells 2022; 11:cells11233819. [PMID: 36497079 PMCID: PMC9741080 DOI: 10.3390/cells11233819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 11/23/2022] [Accepted: 11/25/2022] [Indexed: 11/30/2022] Open
Abstract
The activation of the maternal immune system by a prenatal infection is considered a risk factor for developing psychiatric disorders in the offspring. Toxoplasma gondii is one of the pathogenic infections associated with schizophrenia. Recent studies have shown an association between high levels of IgG anti-T. gondii from mothers and their neonates, with a higher risk of developing schizophrenia. The absence of the parasite and the levels of IgGs found in the early stages of life suggest a transplacental transfer of the anti-T. gondii IgG antibodies, which could bind fetal brain structures by molecular mimicry and induce alterations in neurodevelopment. This study aimed to determine the maternal pathogenic antibodies formation that led to behavioral impairment on the progeny of rats immunized with T. gondii. Female rats were immunized prior to gestation with T. gondii lysate (3 times/once per week). The anti-T. gondii IgG levels were determined in the serum of pregestational exposed females' previous mating. After this, locomotor activity, cognitive and social tests were performed. Cortical neurotransmitter levels for dopamine and glutamate were evaluated at 60 PND in the progeny of rats immunized before gestation (Pregestational group). The maternal pathogenic antibodies were evidenced by their binding to fetal brain mimotopes in the Pregestational group and the reactivity of the serum containing anti-T. gondii IgG was tested in control fetal brains (non-immunized). These results showed that the Pregestational group presented impairment in short and long-term memory, hypoactivity and alteration in social behavior, which was also associated with a decrease in cortical glutamate and dopamine levels. We also found the IgG antibodies bound to brain mimotopes in fetuses from females immunized with T. gondii, as well as observing a strong reactivity of the serum females immunized for fetal brain structures of fetuses from unimmunized mothers. Our results suggest that the exposure to T. gondii before gestation produced maternal pathogenic antibodies that can recognize fetal brain mimotopes and lead to neurochemical and behavioral alterations in the offspring.
Collapse
|
27
|
Butticci R, Habre C, Hernandez A, Barcos-Munoz F, Pfister R, Hanquinet S, Beuchée A, Baud O. Early arterial pressure monitoring and term-equivalent age MRI findings in very preterm infants. Pediatr Res 2022; 92:822-828. [PMID: 34799666 DOI: 10.1038/s41390-021-01839-2] [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: 05/19/2021] [Revised: 10/21/2021] [Accepted: 10/28/2021] [Indexed: 11/09/2022]
Abstract
INTRODUCTION Variability of arterial blood pressure (ABP) has been associated with intraventricular hemorrhage in very preterm neonates (VPT) and may predict other brain lesions assessed at term-equivalent of age (TEA). METHODS This was a prospective single-center study including VPT with early invasive continuous ABP monitoring and assessed at TEA using brain magnetic resonance imaging (TEA-MRI). The association between early mean ABP (MABP) and TEA-MRI findings was modeled by multivariate logistic regression analysis using covariates selected by the LASSO method. RESULTS Among 99 VPT, the LASSO procedure selected consecutive periods of lowest MABP of 30 min on day 1 (d1) and 10 min on day 2 (d2) as the most relevant durations to predict TEA-MRI findings (OR [95% CI], 1.11 [1.02-1.23], p = 0.03 and 1.13 [1.01-1.27], p = 0.03, respectively). ROC curve analysis showed optimal thresholds at 30.25 mmHg on d1 and 33.25 mmHg on d2. This significant association persisted after adjustment with covariates including birthweight, gestational age, sex, and inotrope exposure. Final models selected by LASSO included the decile of the birthweight and lowest MABP for 30 min on d1 and 10 min on d2, for which the areas under the ROC curve were 74% and 75%, respectively. CONCLUSION Early continuous ABP monitoring may predict brain TEA-MRI findings in VPT. IMPACT Early arterial blood pressure monitoring may contribute to predicting brain damage upon MRI at term-equivalent of age for infants born very preterm. Careful blood pressure continuous monitoring in very preterm infants may identify infants at risk of long-term brain damage. Umbilical artery catheterization provides the best option for continuously monitoring arterial blood pressure in very preterm infants.
Collapse
Affiliation(s)
- Roberta Butticci
- Division of Neonatology and Pediatric Intensive Care, Children's University Hospital of Geneva and University of Geneva, 1205, Geneva, Switzerland
| | - Céline Habre
- Pediatric Radiology Unit, Division of Radiology, Children's University Hospital of Geneva and University of Geneva, 1211, Geneva, Switzerland
| | - Alfredo Hernandez
- University of Rennes, CHU Rennes, Inserm, LTSI-UMR 1099, F-35000, Rennes, France
| | - Francisca Barcos-Munoz
- Division of Neonatology and Pediatric Intensive Care, Children's University Hospital of Geneva and University of Geneva, 1205, Geneva, Switzerland
| | - Riccardo Pfister
- Division of Neonatology and Pediatric Intensive Care, Children's University Hospital of Geneva and University of Geneva, 1205, Geneva, Switzerland
| | - Sylviane Hanquinet
- Pediatric Radiology Unit, Division of Radiology, Children's University Hospital of Geneva and University of Geneva, 1211, Geneva, Switzerland
| | - Alain Beuchée
- University of Rennes, CHU Rennes, Inserm, LTSI-UMR 1099, F-35000, Rennes, France
| | - Olivier Baud
- Division of Neonatology and Pediatric Intensive Care, Children's University Hospital of Geneva and University of Geneva, 1205, Geneva, Switzerland. .,NeuroDiderot, UMR 1141, Inserm, Université de Paris, Paris, France.
| |
Collapse
|
28
|
Menon R. Epithelial to mesenchymal transition (EMT) of feto-maternal reproductive tissues generates inflammation: a detrimental factor for preterm birth. BMB Rep 2022. [PMID: 35880430 PMCID: PMC9442346 DOI: 10.5483/bmbrep.2022.55.8.174] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Human pregnancy is a delicate and complex process where multiorgan interactions between two independent systems, the mother, and her fetus, maintain pregnancy. Intercellular interactions that can define homeostasis at the various cellular level between the two systems allow uninterrupted fetal growth and development until delivery. Interactions are needed for tissue remodeling during pregnancy at both fetal and maternal tissue layers. One of the mechanisms that help tissue remodeling is via cellular transitions where epithelial cells undergo a cyclic transition from epithelial to mesenchymal (EMT) and back from mesenchymal to epithelial (MET). Two major pregnancy-associated tissue systems that use EMT, and MET are the fetal membrane (amniochorion) amnion epithelial layer and cervical epithelial cells and will be reviewed here. EMT is often associated with localized inflammation, and it is a well-balanced process to facilitate tissue remodeling. Cyclic transition processes are important because a terminal state or the static state of EMT can cause accumulation of proinflammatory mesenchymal cells in the matrix regions of these tissues and increase localized inflammation that can cause tissue damage. Interactions that determine homeostasis are often controlled by both endocrine and paracrine mediators. Pregnancy maintenance hormone progesterone and its receptors are critical for maintaining the balance between EMT and MET. Increased intrauterine oxidative stress at term can force a static (terminal) EMT and increase inflammation that are physiologic processes that destabilize homeostasis that maintain pregnancy to promote labor and delivery of the fetus. However, conditions that can produce an untimely increase in EMT and inflammation can be pathologic. These tissue damages are often associated with adverse pregnancy complications such as preterm prelabor rupture of the membranes (pPROM) and spontaneous preterm birth (PTB). Therefore, an understanding of the biomolecular processes that maintain cyclic EMT-MET is critical to reducing the risk of pPROM and PTB. Extracellular vesicles (exosomes of 40-160 nm) that can carry various cargo are involved in cellular transitions as paracrine mediators. Exosomes can carry a variety of biomolecules as cargo. Studies specifically using exosomes from cells undergone EMT can carry a pro-inflammatory cargo and in a paracrine fashion can modify the neighboring tissue environment to cause enhancement of uterine inflammation.
Collapse
Affiliation(s)
- Ramkumar Menon
- Division of Basic and Translational Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston 77555-1062, TX, USA
| |
Collapse
|
29
|
Genome-Wide Knockout Screen Identifies Human Sialomucin CD164 as an Essential Entry Factor for Lymphocytic Choriomeningitis Virus. mBio 2022; 13:e0020522. [PMID: 35502904 PMCID: PMC9239079 DOI: 10.1128/mbio.00205-22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Lymphocytic choriomeningitis virus (LCMV) is a well-studied mammarenavirus that can be fatal in congenital infections. However, our understanding of LCMV and its interactions with human host factors remains incomplete. Here, host determinants affecting LCMV infection were investigated through a genome-wide CRISPR knockout screen in A549 cells, a human lung adenocarcinoma line. We identified and validated a variety of novel host factors that play a functional role in LCMV infection. Among these, knockout of the sialomucin CD164, a heavily glycosylated transmembrane protein, was found to ablate infection with multiple LCMV strains but not other hemorrhagic mammarenaviruses in several cell types. Further characterization revealed a dependency of LCMV entry on the cysteine-rich domain of CD164, including an N-linked glycosylation site at residue 104 in that region. Given the documented role of LCMV with respect to transplacental human infections, CD164 expression was investigated in human placental tissue and placental cell lines. CD164 was found to be highly expressed in the cytotrophoblast cells, an initial contact site for pathogens within the placenta, and LCMV infection in placental cells was effectively blocked using a monoclonal antibody specific to the cysteine-rich domain of CD164. Together, this study identifies novel factors associated with LCMV infection of human tissues and highlights the importance of CD164, a sialomucin that previously had not been associated with viral infection. IMPORTANCE Lymphocytic choriomeningitis virus (LCMV) is a human-pathogenic mammarenavirus that can be fatal in congenital infections. Although frequently used in the study of persistent infections in the field of immunology, aspects of this virus's life cycle remain incomplete. For example, while viral entry has been shown to depend on a cell adhesion molecule, DAG1, genetic knockout of this gene allows for residual viral infection, implying that additional receptors can mediate cell entry. The significance of our study is the identification of host factors important for successful infection, including the sialomucin CD164, which had not been previously associated with viral infection. We demonstrated that CD164 is essential for LCMV entry into human cells and can serve as a possible therapeutic target for treatment of congenital infection.
Collapse
|
30
|
Crawford TM, Andersen CC, Hodyl NA, Robertson SA, Stark MJ. Effect of washed versus unwashed red blood cells on transfusion-related immune responses in preterm newborns. Clin Transl Immunology 2022; 11:e1377. [PMID: 35284073 PMCID: PMC8907378 DOI: 10.1002/cti2.1377] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 01/04/2022] [Accepted: 02/04/2022] [Indexed: 12/22/2022] Open
Abstract
Objectives Transfusion with washed packed red blood cells (PRBCs) may be associated with reduced transfusion‐related pro‐inflammatory cytokine production. This may be because of alterations in recipient immune responses. Methods This randomised trial evaluated the effect of transfusion with washed compared with unwashed PRBCs on pro‐inflammatory cytokines and endothelial activation in 154 preterm newborns born before 29 weeks’ gestation. Changes in plasma cytokines and measures of endothelial activation in recipient blood were analysed after each of the first three transfusions. Results By the third transfusion, infants receiving unwashed blood had an increase in IL‐17A (P = 0.04) and TNF (P = 0.007), whereas infants receiving washed blood had reductions in IL‐17A (P = 0.013), TNF (P = 0.048), IL‐6 (P = 0.001), IL‐8 (P = 0.037), IL‐12 (P = 0.001) and IFN‐γ (P = 0.001). The magnitude of the post‐transfusion increase in cytokines did not change between the first and third transfusions in the unwashed group but decreased in the washed group for IL‐12 (P = 0.001), IL‐17A (P = 0.01) and TNF (P = 0.03), with the difference between the groups reaching significance by the third transfusion (P < 0.001 for each cytokine). Conclusion The pro‐inflammatory immune response to transfusion in preterm infants can be modified when PRBCs are washed prior to transfusion. Further studies are required to determine whether the use of washed PRBCs for neonatal transfusion translates into reduced morbidity and mortality.
Collapse
Affiliation(s)
- Tara M Crawford
- The Women's and Children's Hospital Adelaide SA Australia.,The Robinson Research Institute The University of Adelaide Adelaide SA Australia
| | - Chad C Andersen
- The Women's and Children's Hospital Adelaide SA Australia.,The Robinson Research Institute The University of Adelaide Adelaide SA Australia
| | - Nicolette A Hodyl
- The Robinson Research Institute The University of Adelaide Adelaide SA Australia
| | - Sarah A Robertson
- The Robinson Research Institute The University of Adelaide Adelaide SA Australia
| | - Michael J Stark
- The Women's and Children's Hospital Adelaide SA Australia.,The Robinson Research Institute The University of Adelaide Adelaide SA Australia
| |
Collapse
|
31
|
Menon R. Fetal inflammatory response at the fetomaternal interface: A requirement for labor at term and preterm. Immunol Rev 2022; 308:149-167. [PMID: 35285967 DOI: 10.1111/imr.13075] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 03/04/2022] [Indexed: 12/20/2022]
Abstract
Human parturition at term and preterm is an inflammatory process synchronously executed by both fetomaternal tissues to transition them from a quiescent state t an active state of labor to ensure delivery. The initiators of the inflammatory signaling mechanism can be both maternal and fetal. The placental (fetal)-maternal immune and endocrine mediated homeostatic imbalances and inflammation are well reported. However, the fetal inflammatory response (FIR) theories initiated by the fetal membranes (amniochorion) at the choriodecidual interface are not well established. Although immune cell migration, activation, and production of proparturition cytokines to the fetal membranes are reported, cellular level events that can generate a unique set of inflammation are not well discussed. This review discusses derangements to fetal membrane cells (physiologically and pathologically at term and preterm, respectively) in response to both endogenous and exogenous factors to generate inflammatory signals. In addition, the mechanisms of inflammatory signal propagation (fetal signaling of parturition) and how these signals cause immune imbalances at the choriodecidual interface are discussed. In addition to maternal inflammation, this review projects FIR as an additional mediator of inflammatory overload required to promote parturition.
Collapse
Affiliation(s)
- Ramkumar Menon
- Division of Basic Science and Translational Research, Department of Obstetrics & Gynecology, The University of Texas Medical Branch, Galveston, Texas, USA
| |
Collapse
|
32
|
Abstract
Individuals born extremely preterm (before 28 weeks of gestation) comprise only about 0.7% of births in the United States and an even lower proportion in other high resource countries. However, these individuals account for a disproportionate number of children with cerebral palsy, intellectual deficit, autism spectrum disorder, attention deficit hyperactivity disorder, and epilepsy. This review describes two large multiple center cohorts comprised of individuals born extremely preterm: the EPICURE cohort, recruited 1995 in the United Kingdom and the Republic of Ireland, and the Extremely Low Gestational Age Newborn (ELGAN), recruited 2002-2004 in five states in the United States. The primary focus of these studies has been neurodevelopmental disorders, but also of interest are growth, respiratory illness, and parent- and self-reported global health and well-being. Both of these studies indicate that among individuals born extremely preterm the risks of most neurodevelopmental disorders are increased. Early life factors that contribute to this risk include perinatal brain damage, some of which can be identified using neonatal head ultrasound, bronchopulmonary dysplasia, and neonatal systemic inflammation. Prenatal factors, particularly the family's socioeconomic position, also appear to contribute to risk. For most adverse outcomes, the risk is higher in males. Young adults born extremely preterm who have neurodevelopmental impairment, as compared to those without such impairment, rate their quality of life lower. However, young adults born extremely preterm who do not have neurodevelopmental impairments rate their quality of life as being similar to that of young adults born at term. Finally, we summarize the current state of interventions designed to improve the life course of extremely premature infants, with particular focus on efforts to prevent premature birth and on postnatal efforts to prevent adverse neurodevelopmental outcomes.
Collapse
Affiliation(s)
- Genevieve L Taylor
- Genevieve L Taylor MD: Department of Pediatrics, Division of Neonatal-Perinatal Medicine, University of North Carolina School of Medicine
| | - T Michael O'Shea
- T. Michael O'Shea, MD, MPH: Department of Pediatrics, Division of Neonatal-Perinatal Medicine, University of North Carolina School of Medicine.
| |
Collapse
|
33
|
Brégère C, Schwendele B, Radanovic B, Guzman R. Microglia and Stem-Cell Mediated Neuroprotection after Neonatal Hypoxia-Ischemia. Stem Cell Rev Rep 2022; 18:474-522. [PMID: 34382141 PMCID: PMC8930888 DOI: 10.1007/s12015-021-10213-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/28/2021] [Indexed: 12/14/2022]
Abstract
Neonatal hypoxia-ischemia encephalopathy (HIE) refers to a brain injury in term infants that can lead to death or lifelong neurological deficits such as cerebral palsy (CP). The pathogenesis of this disease involves multiple cellular and molecular events, notably a neuroinflammatory response driven partly by microglia, the brain resident macrophages. Treatment options are currently very limited, but stem cell (SC) therapy holds promise, as beneficial outcomes are reported in animal studies and to a lesser degree in human trials. Among putative mechanisms of action, immunomodulation is considered a major contributor to SC associated benefits. The goal of this review is to examine whether microglia is a cellular target of SC-mediated immunomodulation and whether the recruitment of microglia is linked to brain repair. We will first provide an overview on microglial activation in the rodent model of neonatal HI, and highlight its sensitivity to developmental age. Two complementary questions are then addressed: (i) do immune-related treatments impact microglia and provide neuroprotection, (ii) does stem cell treatment modulates microglia? Finally, the immune-related findings in patients enrolled in SC based clinical trials are discussed. Our review points to an impact of SCs on the microglial phenotype, but heterogeneity in experimental designs and methodological limitations hamper our understanding of a potential contribution of microglia to SC associated benefits. Thorough analyses of the microglial phenotype are warranted to better address the relevance of the neuroimmune crosstalk in brain repair and improve or advance the development of SC protocols in humans.
Collapse
Affiliation(s)
- Catherine Brégère
- Department of Biomedicine and Department of Neurosurgery, Faculty of Medicine, University Hospital Basel, Basel, Switzerland
| | - Bernd Schwendele
- Department of Biomedicine and Department of Neurosurgery, Faculty of Medicine, University Hospital Basel, Basel, Switzerland
| | - Boris Radanovic
- Department of Biomedicine and Department of Neurosurgery, Faculty of Medicine, University Hospital Basel, Basel, Switzerland
| | - Raphael Guzman
- Department of Biomedicine and Department of Neurosurgery, Faculty of Medicine, University Hospital Basel, Basel, Switzerland.
| |
Collapse
|
34
|
Gall AR, Amoah SK, Kitase Y, Jantzie LL. Placental mediated mechanisms of perinatal brain injury: Evolving inflammation and exosomes. Exp Neurol 2022; 347:113914. [PMID: 34752783 PMCID: PMC8712107 DOI: 10.1016/j.expneurol.2021.113914] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Revised: 10/29/2021] [Accepted: 11/01/2021] [Indexed: 01/03/2023]
Abstract
Pregnancy is an inflammatory process that is carefully regulated by the placenta via immunomodulation and cell-to-cell communication of maternal and fetal tissues. Exosomes, types of extracellular vesicles, facilitate the intercellular communication and traffic biologically modifying cargo within the maternal-placental-fetal axis in normal and pathologic pregnancies. Chorioamnionitis is characterized by inflammation of chorioamniotic membranes that produces systemic maternal and fetal inflammatory responses of cytokine dysregulation and has been associated with brain injury and neurodevelopmental disorders. This review focuses on how pathologic placental exosomes propagate acute and chronic inflammation leading to brain injury. The evidence reviewed here highlights the need to investigate exosomes from pathologic pregnancies and those with known brain injury to identify new diagnostics, biomarkers, and potential therapeutic targets.
Collapse
Affiliation(s)
- Alexander R Gall
- Division of Neonatology, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Stephen K Amoah
- Division of Neonatology, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Yuma Kitase
- Division of Neonatology, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Lauren L Jantzie
- Division of Neonatology, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA,Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA,Kennedy Krieger Institute, Baltimore, MD, USA,Corresponding author at: 600 N. Wolfe Street, CMSC Building, 6-104A, Baltimore, MD 21287, USA. (L.L. Jantzie)
| |
Collapse
|
35
|
McLellan J, Kim DHJ, Bruce M, Ramirez-Celis A, Van de Water J. Maternal Immune Dysregulation and Autism-Understanding the Role of Cytokines, Chemokines and Autoantibodies. Front Psychiatry 2022; 13:834910. [PMID: 35722542 PMCID: PMC9201050 DOI: 10.3389/fpsyt.2022.834910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 04/20/2022] [Indexed: 11/29/2022] Open
Abstract
Autism spectrum disorder (ASD) is acknowledged as a highly heterogeneous, behaviorally defined neurodevelopmental disorder with multiple etiologies. In addition to its high heritability, we have come to recognize a role for maternal immune system dysregulation as a prominent risk factor for the development of ASD in the child. Examples of these risk factors include altered cytokine/chemokine activity and the presence of autoantibodies in mothers that are reactive to proteins in the developing brain. In addition to large clinical studies, the development of pre-clinical models enables the ability to evaluate the cellular and molecular underpinnings of immune-related pathology. For example, the novel animal models of maternal autoantibody-related (MAR) ASD described herein will serve as a preclinical platform for the future testing of targeted therapeutics for one 'type' of ASD. Identification of the cellular targets will advance precision medicine efforts toward tailored therapeutics and prevention. This minireview highlights emerging evidence for the role of maternal immune dysregulation as a potential biomarker, as well as a pathologically relevant mechanism for the development of ASD in offspring. Further, we will discuss the current limitations of these models as well as potential avenues for future research.
Collapse
Affiliation(s)
- Janna McLellan
- Division of Rheumatology, Department of Internal Medicine, Allergy, and Clinical Immunology, University of California, Davis, Davis, CA, United States
| | - Danielle H J Kim
- Division of Rheumatology, Department of Internal Medicine, Allergy, and Clinical Immunology, University of California, Davis, Davis, CA, United States
| | - Matthew Bruce
- Division of Rheumatology, Department of Internal Medicine, Allergy, and Clinical Immunology, University of California, Davis, Davis, CA, United States
| | - Alexandra Ramirez-Celis
- Division of Rheumatology, Department of Internal Medicine, Allergy, and Clinical Immunology, University of California, Davis, Davis, CA, United States
| | - Judy Van de Water
- Division of Rheumatology, Department of Internal Medicine, Allergy, and Clinical Immunology, University of California, Davis, Davis, CA, United States.,MIND Institute, University of California, Davis, Davis, CA, United States
| |
Collapse
|
36
|
Ingvaldsen SH, Morken TS, Austeng D, Dammann O. Visuopathy of prematurity: is retinopathy just the tip of the iceberg? Pediatr Res 2022; 91:1043-1048. [PMID: 34168272 PMCID: PMC9122817 DOI: 10.1038/s41390-021-01625-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Revised: 05/27/2021] [Accepted: 05/29/2021] [Indexed: 02/06/2023]
Abstract
Research on retinopathy of prematurity (ROP) focuses mainly on the abnormal vascularization patterns that are directly visible for ophthalmologists. However, recent findings indicate that children born prematurely also exhibit changes in the retinal cellular architecture and along the dorsal visual stream, such as structural changes between and within cortical areas. Moreover, perinatal sustained systemic inflammation (SSI) is associated with an increased risk for ROP and the visual deficits that follow. In this paper, we propose that ROP might just be the tip of an iceberg we call visuopathy of prematurity (VOP). The VOP paradigm comprises abnormal vascularization of the retina, alterations in retinal cellular architecture, choroidal degeneration, and abnormalities in the visual pathway, including cortical areas. Furthermore, VOP itself might influence the developmental trajectories of cerebral structures and functions deemed responsible for visual processing, thereby explaining visual deficits among children born preterm.
Collapse
Affiliation(s)
- Sigrid Hegna Ingvaldsen
- Department of Neuromedicine and Movement Science, Norwegian University of Science and Technology, Trondheim, Norway.
| | - Tora Sund Morken
- grid.5947.f0000 0001 1516 2393Department of Neuromedicine and Movement Science, Norwegian University of Science and Technology, Trondheim, Norway ,grid.52522.320000 0004 0627 3560Department of Ophthalmology, St. Olav Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Dordi Austeng
- grid.5947.f0000 0001 1516 2393Department of Neuromedicine and Movement Science, Norwegian University of Science and Technology, Trondheim, Norway ,grid.52522.320000 0004 0627 3560Department of Ophthalmology, St. Olav Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Olaf Dammann
- grid.5947.f0000 0001 1516 2393Department of Neuromedicine and Movement Science, Norwegian University of Science and Technology, Trondheim, Norway ,grid.67033.310000 0000 8934 4045Department of Public Health and Community Medicine, Tufts University School of Medicine, Boston, MA USA ,grid.10423.340000 0000 9529 9877Department of Gynecology and Obstetrics, Hannover Medical School, Hannover, Germany
| |
Collapse
|
37
|
Blaylock RL, Faria M. New concepts in the development of schizophrenia, autism spectrum disorders, and degenerative brain diseases based on chronic inflammation: A working hypothesis from continued advances in neuroscience research. Surg Neurol Int 2021; 12:556. [PMID: 34877042 PMCID: PMC8645502 DOI: 10.25259/sni_1007_2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 10/05/2021] [Indexed: 12/14/2022] Open
Abstract
This paper was written prompted by a poignant film about adolescent girl with schizophrenia who babysits for a younger girl in an isolated cabin. Schizophrenia is an illness that both authors are fascinated with and that they continue to study and investigate. There is now compelling evidence that schizophrenia is a very complex syndrome that involves numerous neural pathways in the brain, far more than just dopaminergic and serotonergic systems. One of the more popular theories in recent literature is that it represents a hypo glutaminergic deficiency of certain pathways, including thalamic ones. After much review of research and study in this area, we have concluded that most such theories contain a number of shortcomings. Most are based on clinical responses to certain drugs, particularly antipsychotic drugs affecting the dopaminergic neurotransmitters; thus, assuming dopamine release was the central cause of the psychotic symptoms of schizophrenia. The theory was limited in that dopamine excess could only explain the positive symptoms of the disorder. Antipsychotic medications have minimal effectiveness for the negative and cognitive symptoms associated with schizophrenia. It has been estimated that 20–30% of patients show either a partial or no response to antipsychotic medications. In addition, the dopamine hypothesis does not explain the neuroanatomic findings in schizophrenia.
Collapse
Affiliation(s)
| | - Miguel Faria
- Clinical Professor of Surgery (Neurosurgery, ret.) and Adjunct Professor of Medical History (ret.), Mercer University School of Medicine, United States
| |
Collapse
|
38
|
Goudreau AD, Everest C, Nagpal TS, Puranda JL, Bhattacharjee J, Vasanthan T, Adamo KB. Elucidating the interaction between maternal physical activity and circulating myokines throughout gestation: A scoping review. Am J Reprod Immunol 2021; 86:e13488. [PMID: 34331363 DOI: 10.1111/aji.13488] [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: 05/27/2021] [Revised: 07/05/2021] [Accepted: 07/26/2021] [Indexed: 11/29/2022] Open
Abstract
Physical activity (PA) during pregnancy provides both maternal and fetal health benefits. It has been theorized that myokines, peptides secreted by contracting skeletal muscle, may play an important mechanistic role in facilitating the health benefits obtained from prenatal exercise. The objective of this review was to synthesize the current literature on the relationship between maternal PA and myokine response. A search strategy was developed using the terms pregnancy, PA, IL-6, IL-10, IL-13, and TNF-α. A systematic search was completed in July 2020, in Medline, SPORTDiscus, EMBASE, CENTRAL, and in November 2020 for unpublished dissertations (grey literature; Proquest). Both human- and animal-based studies of any design were included, while commentaries and editorial articles were excluded. Data were extracted by two independent reviewers and summarized narratively. Data were thematically summarized based on the myokine and whether findings were from human or animal studies. Ten studies were included in this review. Findings from studies that examined IL-6, IL-10, and TNF-α suggest a trimester-specific interaction between PA and myokine levels; no studies evaluated IL-13. Future research should investigate the PA-myokine relationship throughout all stages of gestation.
Collapse
Affiliation(s)
| | - Catherine Everest
- Faculty of Health Sciences, University of Ottawa, Ottawa, ON, Canada
| | - Taniya S Nagpal
- Faculty of Health Sciences, University of Ottawa, Ottawa, ON, Canada
- Society of Obstetricians and Gynaecologists of Canada, Ottawa, ON, Canada
| | - Jessica L Puranda
- Faculty of Health Sciences, University of Ottawa, Ottawa, ON, Canada
| | - Jayonta Bhattacharjee
- Faculty of Health Sciences, University of Ottawa, Ottawa, ON, Canada
- Department of Surgery and Obstetrics, Bangladesh Agricultural University, Mymensingh, Bangladesh
| | | | - Kristi B Adamo
- Faculty of Health Sciences, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
39
|
Griffin A, Spencer SK, Bowles T, Solis L, Robinson R, Ramarao S, Wallace K. Male HELLP pups experience sensorimotor delays and reduced body weight. Physiol Behav 2021; 241:113567. [PMID: 34474060 DOI: 10.1016/j.physbeh.2021.113567] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 08/26/2021] [Accepted: 08/27/2021] [Indexed: 01/12/2023]
Abstract
Offspring of Preeclampsia (PreE) and HELLP Syndrome are at an increased risk of developing neurodevelopmental disorders. In the current study we sought to determine if offspring from experimental models of PreE and HELLP had evidence of early onset neurodevelopmental delay. Offspring from PreE, HELLP and normal pregnant dams were assessed in a battery of sensorimotor tests beginning on postnatal day (PND) 3. Male HELLP offspring showed altered behavior in the surface righting reflex on PND 3 and cliff avoidance task from PND 3-6 relative to other groups. Results suggest that there are sex differences in offspring born to dams with PreE and HELLP.
Collapse
Affiliation(s)
- Ashley Griffin
- Program in Neuroscience, 2500 North State Street, University of Mississippi Medical Center, Jackson, MS 39216, United States
| | - Shauna-Kay Spencer
- Department of Obstetrics and Gynecology, 2500 North State Street, University of Mississippi Medical Center, Jackson, MS 39216, United States
| | - Teylor Bowles
- Department of Obstetrics and Gynecology, 2500 North State Street, University of Mississippi Medical Center, Jackson, MS 39216, United States
| | - Lucia Solis
- Department of Obstetrics and Gynecology, 2500 North State Street, University of Mississippi Medical Center, Jackson, MS 39216, United States
| | - Reanna Robinson
- Department of Obstetrics and Gynecology, 2500 North State Street, University of Mississippi Medical Center, Jackson, MS 39216, United States
| | - Sumana Ramarao
- Department of Pediatrics, 2500 North State Street, University of Mississippi Medical Center, Jackson, MS 39216, United States
| | - Kedra Wallace
- Department of Obstetrics and Gynecology, 2500 North State Street, University of Mississippi Medical Center, Jackson, MS 39216, United States; Department of Neurobiology and Anatomical Sciences, 2500 North State Street, University of Mississippi Medical Center, Jackson, MS 39216, United States.
| |
Collapse
|
40
|
Schachinger F, Farr S. The Effects of Preterm Birth on Musculoskeletal Health-Related Disorders. J Clin Med 2021; 10:5082. [PMID: 34768599 PMCID: PMC8584797 DOI: 10.3390/jcm10215082] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 10/27/2021] [Accepted: 10/29/2021] [Indexed: 12/21/2022] Open
Abstract
Preterm birth is associated with various diseases and conditions which demand multidisciplinary medical care. Approximately 10% of all neonates are born prematurely with an increasing survival rate in almost all Western countries. This ongoing, yet desirable trend is creating new challenges for sufficient medical treatment regimens, which should be upheld throughout the patients' lives. Orthopedic surgeons are focused on musculoskeletal disorders and the improvement of patients' ability to cope with the challenges of everyday life. The most common conditions associated with preterm birth are cerebral palsy and a dysregulation of the calcium/phosphorus metabolism, which may lead to fractures. These diseases may vary greatly in their organic manifestation and clinical presentation. This demands multidisciplinary cooperation and parental support. Clinical management is aimed on the early enhancement of a patient's physical, as well as neurological condition, and to prevent the development of secondary musculoskeletal disorders. In this article, we give an overview of the current literature on the most common musculoskeletal disorders associated with preterm birth and critically discuss state of the art diagnostic standards and treatment algorithms.
Collapse
Affiliation(s)
| | - Sebastian Farr
- Department of Pediatric Orthopaedics and Foot and Ankle Surgery, Orthopaedic Hospital Speising, Speisingerstrasse 109, A-1130 Vienna, Austria;
| |
Collapse
|
41
|
Chauhan P, Kaur G, Prasad R, Singh H. Pharmacotherapy of schizophrenia: immunological aspects and potential role of immunotherapy. Expert Rev Neurother 2021; 21:1441-1453. [PMID: 34654348 DOI: 10.1080/14737175.2021.1994857] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
INTRODUCTION Schizophrenia is a complex disorder owing to diversity in clinical phenotypes, overlapping symptoms, and heterogeneous clinical presentation. Even after decades of research, the exact causative mechanisms of schizophrenia are not completely known. Recent evidence indicates the role of immune dysfunction in schizophrenia pathogenesis as observed from alteration in immune cells, increased activity of complement cascade, and development of autoantibodies against neurotransmitter receptors. Immunotherapy involving immunosuppressants and cytokine-targeting drugs, have shown promising results in several clinical studies and it demands further research in this area. AREAS COVERED Here, the authors review the immunopathogenesis of schizophrenia, limitations of conventional, and atypical antipsychotic drugs and the potential role and limitations of immunotherapeutic drugs in schizophrenia management. EXPERT OPINION Schizophrenia is a complex disorder and poses a challenge to the currently available treatment approaches. Nearly 30% schizophrenia patients exhibit minimal response toward conventional and atypical antipsychotic drugs. Immune system dysfunction plays an important part of schizophrenia pathophysiology and existing monoclonal antibody (mAb) drugs targeting specific components of the immune system are being repositioned in schizophrenia. The authors call upon public and private funders to facilitate urgent and rigorous research efforts in exploring potential role of immunotherapy in schizophrenia.
Collapse
Affiliation(s)
- Prerna Chauhan
- Multidisciplinary Research Unit, Government Medical College & Hospital, Chandigarh, India
| | - Gurjit Kaur
- Department of Physiology, Government Medical College & Hospital, Chandigarh, India
| | - Rajendra Prasad
- Department of Biochemistry, Maharishi Markandeshwar Institute of Medical Sciences and Research, Ambala, Haryana, India
| | - Harmanjit Singh
- Department of Pharmacology, Government Medical College & Hospital, Chandigarh, India
| |
Collapse
|
42
|
O'Connor TG, Ciesla AA. Maternal Immune Activation Hypotheses for Human Neurodevelopment: Some Outstanding Questions. BIOLOGICAL PSYCHIATRY: COGNITIVE NEUROSCIENCE AND NEUROIMAGING 2021; 7:471-479. [PMID: 34688920 PMCID: PMC9021321 DOI: 10.1016/j.bpsc.2021.10.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 10/05/2021] [Accepted: 10/13/2021] [Indexed: 12/17/2022]
Abstract
The Maternal Immune Activation (MIA) hypothesis is a leading model for understanding prenatal influences on individual differences in, and clinical syndromes of, neurodevelopment. Experimental animal and human research has proliferated in recent years, and there is now a sizable research base. Several meta-analyses demonstrate general support for an association between prenatal immune activation and neurodevelopment in human research. However, questions remain about the nature of the immune activation, the network of underlying mechanisms involved, and the breadth of impact across behavioral phenotypes. Complementing recent reviews of results, the current review places particular emphasis on how advances in understanding mechanisms may be improved with greater attention to addressing the methodological variation and limitations of existing studies, and identifies areas for further clinical research.
Collapse
Affiliation(s)
- Thomas G O'Connor
- Department of Psychiatry, University of Rochester; Department of Psycholog, University of Rochestery; Department of Neuroscience, University of Rochester; Department of Obstetrics and Gynecology, University of Rochester; Wynne Center for Family Research, University of Rochester.
| | | |
Collapse
|
43
|
Kitase Y, Chin EM, Ramachandra S, Burkhardt C, Madurai NK, Lenz C, Hoon AH, Robinson S, Jantzie LL. Sustained peripheral immune hyper-reactivity (SPIHR): an enduring biomarker of altered inflammatory responses in adult rats after perinatal brain injury. J Neuroinflammation 2021; 18:242. [PMID: 34666799 PMCID: PMC8527679 DOI: 10.1186/s12974-021-02291-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 10/07/2021] [Indexed: 01/12/2023] Open
Abstract
Background Chorioamnionitis (CHORIO) is a principal risk factor for preterm birth and is the most common pathological abnormality found in the placentae of preterm infants. CHORIO has a multitude of effects on the maternal–placental–fetal axis including profound inflammation. Cumulatively, these changes trigger injury in the developing immune and central nervous systems, thereby increasing susceptibility to chronic sequelae later in life. Despite this and reports of neural–immune changes in children with cerebral palsy, the extent and chronicity of the peripheral immune and neuroinflammatory changes secondary to CHORIO has not been fully characterized. Methods We examined the persistence and time course of peripheral immune hyper-reactivity in an established and translational model of perinatal brain injury (PBI) secondary to CHORIO. Pregnant Sprague–Dawley rats underwent laparotomy on embryonic day 18 (E18, preterm equivalent). Uterine arteries were occluded for 60 min, followed by intra-amniotic injection of lipopolysaccharide (LPS). Serum and peripheral blood mononuclear cells (PBMCs) were collected at young adult (postnatal day P60) and middle-aged equivalents (P120). Serum and PBMCs secretome chemokines and cytokines were assayed using multiplex electrochemiluminescent immunoassay. Multiparameter flow cytometry was performed to interrogate immune cell populations. Results Serum levels of interleukin-1β (IL-1β), IL-5, IL-6, C–X–C Motif Chemokine Ligand 1 (CXCL1), tumor necrosis factor-α (TNF-α), and C–C motif chemokine ligand 2/monocyte chemoattractant protein-1 (CCL2/MCP-1) were significantly higher in CHORIO animals compared to sham controls at P60. Notably, CHORIO PBMCs were primed. Specifically, they were hyper-reactive and secreted more inflammatory mediators both at baseline and when stimulated in vitro. While serum levels of cytokines normalized by P120, PBMCs remained primed, and hyper-reactive with a robust pro-inflammatory secretome concomitant with a persistent change in multiple T cell populations in CHORIO animals. Conclusions The data indicate that an in utero inflammatory insult leads to neural–immune changes that persist through adulthood, thereby conferring vulnerability to brain and immune system injury throughout the lifespan. This unique molecular and cellular immune signature including sustained peripheral immune hyper-reactivity (SPIHR) and immune cell priming may be a viable biomarker of altered inflammatory responses following in utero insults and advances our understanding of the neuroinflammatory cascade that leads to perinatal brain injury and later neurodevelopmental disorders, including cerebral palsy.
Collapse
Affiliation(s)
- Yuma Kitase
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Johns Hopkins University School of Medicine, 600 N. Wolfe Street, CMSC Building, 6-104A, Baltimore, MD, USA
| | - Eric M Chin
- Department of Neurology and Developmental Medicine, Kennedy Krieger Institute, Baltimore, MD, USA
| | - Sindhu Ramachandra
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Johns Hopkins University School of Medicine, 600 N. Wolfe Street, CMSC Building, 6-104A, Baltimore, MD, USA
| | - Christopher Burkhardt
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Johns Hopkins University School of Medicine, 600 N. Wolfe Street, CMSC Building, 6-104A, Baltimore, MD, USA
| | - Nethra K Madurai
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Johns Hopkins University School of Medicine, 600 N. Wolfe Street, CMSC Building, 6-104A, Baltimore, MD, USA
| | - Colleen Lenz
- Department of Neurology and Developmental Medicine, Kennedy Krieger Institute, Baltimore, MD, USA
| | - Alexander H Hoon
- Department of Neurology and Developmental Medicine, Kennedy Krieger Institute, Baltimore, MD, USA
| | - Shenandoah Robinson
- Division of Pediatric Neurosurgery, Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Lauren L Jantzie
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Johns Hopkins University School of Medicine, 600 N. Wolfe Street, CMSC Building, 6-104A, Baltimore, MD, USA. .,Department of Neurology and Developmental Medicine, Kennedy Krieger Institute, Baltimore, MD, USA. .,Division of Pediatric Neurosurgery, Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA. .,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
44
|
Al-Lawama M, Badran E, Ghanim N, Irsheid A, Qtaishat H, Al-Ammouri I, Al-Zyadneh E, Al-Iede M, Daher AH, Bakri FG, Massad G. Perinatal Transmission and Clinical Outcomes of Neonates Born to SARS-CoV-2-Positive Mothers. J Clin Med Res 2021; 13:420-424. [PMID: 34527097 PMCID: PMC8425792 DOI: 10.14740/jocmr4578] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Accepted: 08/20/2021] [Indexed: 12/25/2022] Open
Abstract
Background The aim of the study was to investigate the clinical outcomes and rate of virus detection in neonates born to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-positive mothers. Methods This prospective study included neonates born to SARS-CoV-2-positive mothers, documenting their viral polymerase chain reaction results and clinical outcomes. Results Of the 130 neonates born to 122 SARS-CoV-2-positive mothers, 12% tested positive. Most (62%) neonates were delivered via cesarean section at an average gestational age of 36 weeks, with a birth weight of 2,900 g. Only 38% neonates required admission. SARS-CoV-2-positive infants were born at a significantly lower gestational age; had a significantly lower birth weight; and had significantly higher admission rates, surfactant therapy, and bradycardia than SARS-CoV-2-negative infants. There was no significant difference in mortality rates. Conclusion This study documents perinatal transmission of SARS-CoV-2. It reports for the first time the occurrence of neonatal bradycardia as a complication of maternal SARS-CoV-2 infection. Despite that, neonates born to SARS-CoV-2-positive mothers had relatively good short-term outcomes.
Collapse
Affiliation(s)
- Manar Al-Lawama
- Pediatric Department, School of Medicine, The University of Jordan, Amman, Jordan
| | - Eman Badran
- Pediatric Department, School of Medicine, The University of Jordan, Amman, Jordan
| | - Noor Ghanim
- Pediatric Department, School of Medicine, The University of Jordan, Amman, Jordan
| | - Ayah Irsheid
- Pediatric Department, School of Medicine, The University of Jordan, Amman, Jordan
| | - Hiba Qtaishat
- Pediatric Department, School of Medicine, The University of Jordan, Amman, Jordan
| | - Iyad Al-Ammouri
- Pediatric Department, School of Medicine, The University of Jordan, Amman, Jordan
| | - Enas Al-Zyadneh
- Pediatric Department, School of Medicine, The University of Jordan, Amman, Jordan
| | - Montaha Al-Iede
- Pediatric Department, School of Medicine, The University of Jordan, Amman, Jordan
| | - Amira H Daher
- Pediatric Department, School of Medicine, The University of Jordan, Amman, Jordan
| | - Fares G Bakri
- Division of Infectious Diseases, Department of Medicine, Jordan University Hospital, Amman, Jordan.,Infectious Disease and Vaccine Center, The University of Jordan, Amman, Jordan
| | - Ghada Massad
- Nursing Department, Jordan University Hospital, Amman, Jordan
| |
Collapse
|
45
|
Dammann O, Rivera JC, Chemtob S. The prenatal phase of retinopathy of prematurity. Acta Paediatr 2021; 110:2521-2528. [PMID: 34028096 DOI: 10.1111/apa.15945] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 04/29/2021] [Accepted: 05/18/2021] [Indexed: 12/22/2022]
Abstract
AIM To explore the current literature on prenatal inflammation-associated risk factors for retinopathy of prematurity (ROP). METHODS Subjective summary of selected experimental and epidemiological publications that support the authors' central hypothesis that the aetiology of ROP begins before birth. RESULTS Based on current evidence we suggest that, contrary to current aetiological models, the process of ROP development begins with a prephase in utero. This beginning is likely initiated by inflammatory responses that are associated with intrauterine infection. CONCLUSION We propose a novel aetio-pathogenetic model of ROP and suggest that the effects of postnatal exposure to inflammatory stressors (resulting from infection or hyperoxia or both) as well as those of other pre- and postnatal contributors to the complex pathogenesis of ROP might be modified by the prenatal phase of the disease.
Collapse
Affiliation(s)
- Olaf Dammann
- Deptartments of Public Health & Community Medicine, Pediatrics, and Ophthalmology Tufts University School of Medicine Boston USA
- Department of Gynecology and Obstetrics Hannover Medical School Hannover Germany
- Department of Neuromedicine and Movement Science NTNU Norwegian University of Science and Technology Trondheim Norway
| | - José Carlos Rivera
- Departments of Pediatrics, Ophthalmology, and Pharmacology Hôpital Maisonneuve‐Rosemont Research Center Montreal QC Canada
- CHU Sainte Justine Research Centre Montreal QC Canada
| | - Sylvain Chemtob
- Departments of Pediatrics, Ophthalmology, and Pharmacology Hôpital Maisonneuve‐Rosemont Research Center Montreal QC Canada
- CHU Sainte Justine Research Centre Montreal QC Canada
- Department of Pharmacology and Therapeutics McGill University Montreal QC Canada
| |
Collapse
|
46
|
Placental Macrophages Demonstrate Sex-Specific Response to Intrauterine Inflammation and May Serve as a Marker of Perinatal Neuroinflammation. J Reprod Immunol 2021; 147:103360. [PMID: 34390899 DOI: 10.1016/j.jri.2021.103360] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 07/06/2021] [Accepted: 07/26/2021] [Indexed: 12/23/2022]
Abstract
Preterm birth (PTB) is considered to be one of the most frequent causes of neonatal death. Prompt and effective measures to predict adverse fetal outcome following PTB are urgently needed. Placenta macrophages are a critical immune cell population during pregnancy, phenotypically divided into M1 and M2 subsets. An established mouse model of intrauterine inflammation (IUI) was applied. Placenta (labyrinth) and corresponding fetal brain were harvested within 24 hours post injection (hpi). Flow cytometry, Western blot, real-time qPCR, and regular histology were utilized to examine the cytokines, macrophage polarization, and sex-specificity. Placental exposure to LPS led to significantly reduced labyrinth thickness compared to PBS-exposed controls as early as 3 hpi, accompanied by apoptosis and necrosis. Pro-inflammatory M1 markers, Il-1β, and iNOS, and anti-inflammatory M2 marker Il-10 increased significantly in placentas exposed to IUI. Analysis of flow cytometry revealed that fetal macrophages (Hofbauer cell, HBCs) were mostly M1-like and that maternal inter-labyrinth macrophages (MIM) were M2-like in their features in IUI. Male fetuses displayed significantly decreased M2-like features in HBCs at 3 and 6 hpi, while female fetuses showed significant increase in M2-like features in MIM at 3 and 6 hpi. Furthermore, there was a significant correlation between the frequency of HBCs and corresponding microglial marker expression at 3 and 6 hpi. Placental macrophages demonstrated sex-specific features in response to IUI. Specifically, HBCs may be a potential biomarker for fetal brain injury at preterm birth.
Collapse
|
47
|
Chesnut M, Hartung T, Hogberg H, Pamies D. Human Oligodendrocytes and Myelin In Vitro to Evaluate Developmental Neurotoxicity. Int J Mol Sci 2021; 22:7929. [PMID: 34360696 PMCID: PMC8347131 DOI: 10.3390/ijms22157929] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 07/14/2021] [Accepted: 07/21/2021] [Indexed: 01/01/2023] Open
Abstract
Neurodevelopment is uniquely sensitive to toxic insults and there are concerns that environmental chemicals are contributing to widespread subclinical developmental neurotoxicity (DNT). Increased DNT evaluation is needed due to the lack of such information for most chemicals in common use, but in vivo studies recommended in regulatory guidelines are not practical for the large-scale screening of potential DNT chemicals. It is widely acknowledged that developmental neurotoxicity is a consequence of disruptions to basic processes in neurodevelopment and that testing strategies using human cell-based in vitro systems that mimic these processes could aid in prioritizing chemicals with DNT potential. Myelination is a fundamental process in neurodevelopment that should be included in a DNT testing strategy, but there are very few in vitro models of myelination. Thus, there is a need to establish an in vitro myelination assay for DNT. Here, we summarize the routes of myelin toxicity and the known models to study this particular endpoint.
Collapse
Affiliation(s)
- Megan Chesnut
- Center for Alternatives to Animal Testing (CAAT), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA; (M.C.); (T.H.)
| | - Thomas Hartung
- Center for Alternatives to Animal Testing (CAAT), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA; (M.C.); (T.H.)
- Center for Alternatives to Animal Testing (CAAT-Europe), University of Konstanz, 78464 Konstanz, Germany
| | - Helena Hogberg
- Center for Alternatives to Animal Testing (CAAT), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA; (M.C.); (T.H.)
| | - David Pamies
- Center for Alternatives to Animal Testing (CAAT), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA; (M.C.); (T.H.)
- Department of Physiology, University of Lausanne, 1005 Lausanne, Switzerland
- Swiss Centre for Applied Human Toxicology (SCAHT), 4055 Basel, Switzerland
| |
Collapse
|
48
|
Association between maternal cervicovaginal swab positivity for Ureaplasma spp. or other microorganisms and neonatal respiratory outcome and mortality. J Perinatol 2021; 41:1-11. [PMID: 32908191 DOI: 10.1038/s41372-020-00808-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Revised: 08/04/2020] [Accepted: 08/27/2020] [Indexed: 11/09/2022]
Abstract
OBJECTIVE We investigated the association between maternal cervicovaginal cultures, its antibiotic treatment, and neonatal outcome. STUDY DESIGN This retrospective cohort study enrolled 480 neonates born prior to 32 weeks' gestation. They were divided into groups according to maternal cervicovaginal culture results. Multivariate logistic regression analysis was used to predict neonatal outcome based on maternal culture results, adjusted for perinatal risk factors and neonatal morbidities. RESULT Maternal cervicovaginal Ureaplasma colonization was independently associated with bronchopulmonary dysplasia at 36 weeks (BPD) (OR 8.34; 95% CI 1.21-57.45). In neonates with and without maternal cervicovaginal Ureaplasma colonization BPD occurred in 12.3% and 3.8%, respectively. Maternal colonization with other microorganisms was associated with a higher neonatal mortality (p = 0.002), lower gestational age (p = 0.026), and birth weight (p = 0.036). CONCLUSIONS This study underscores the role of the maternal cervicovaginal microbiome as a predictor of neonatal outcome. Cervicovaginal Ureaplasma colonization seems not to be an innocent bystander in the multifactorial etiology of BPD.
Collapse
|
49
|
Ma X, Shi Y. Whether erythropoietin can be a neuroprotective agent against premature brain injury: cellular mechanisms and clinical efficacy. Curr Neuropharmacol 2021; 20:611-629. [PMID: 34030616 DOI: 10.2174/1570159x19666210524154519] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 04/27/2021] [Accepted: 05/08/2021] [Indexed: 11/22/2022] Open
Abstract
Preterm infants are at high risk of brain injury. With more understanding of the preterm brain injury's pathogenesis, neuroscientists are looking for more effective methods to prevent and treat it, among which erythropoietin (Epo) is considered as a prime candidate. This review tries to clarify the possible mechanisms of Epo in preterm neuroprotection and summarize updated evidence considering Epo as a pharmacological neuroprotective strategy in animal models and clinical trials. To date, various animal models have validated that Epo is an anti-apoptotic, anti-inflammatory, anti-oxidant, anti-excitotoxic, neurogenetic, erythropoietic, angiogenetic, and neurotrophic agent, thus preventing preterm brain injury. However, although the scientific rationale and preclinical data for Epo's neuroprotective effect are promising, when translated to bedside, the results vary in different studies, especially in its long-term efficacy. Based on existing evidence, it is still too early to recommend Epo as the standard treatment for preterm brain injury.
Collapse
Affiliation(s)
- Xueling Ma
- Department of Neonatology, Children's Hospital of Chongqing Medical University; National Clinical Research Center for Child Health and Disorders; Ministry of Education Key Laboratory of Child Development and Disorders; Chongqing 400014, China
| | - Yuan Shi
- Department of Neonatology, Children's Hospital of Chongqing Medical University; National Clinical Research Center for Child Health and Disorders; Ministry of Education Key Laboratory of Child Development and Disorders; Chongqing 400014, China
| |
Collapse
|
50
|
Rasile M, Lauranzano E, Mirabella F, Matteoli M. Neurological consequences of neurovascular unit and brain vasculature damages: potential risks for pregnancy infections and COVID-19-babies. FEBS J 2021; 289:3374-3392. [PMID: 33998773 PMCID: PMC8237015 DOI: 10.1111/febs.16020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 05/11/2021] [Accepted: 05/13/2021] [Indexed: 01/08/2023]
Abstract
Intragravidic and perinatal infections, acting through either direct viral effect or immune-mediated responses, are recognized causes of liability for neurodevelopmental disorders in the progeny. The large amounts of epidemiological data and the wealth of information deriving from animal models of gestational infections have contributed to delineate, in the last years, possible underpinning mechanisms for this phenomenon, including defects in neuronal migration, impaired spine and synaptic development, and altered activation of microglia. Recently, dysfunctions of the neurovascular unit and anomalies of the brain vasculature have unexpectedly emerged as potential causes at the origin of behavioral abnormalities and psychiatric disorders consequent to prenatal and perinatal infections. This review aims to discuss the up-to-date literature evidence pointing to the neurovascular unit and brain vasculature damages as the etiological mechanisms in neurodevelopmental syndromes. We focus on the inflammatory events consequent to intragravidic viral infections as well as on the direct viral effects as the potential primary triggers. These authors hope that a timely review of the literature will help to envision promising research directions, also relevant for the present and future COVID-19 longitudinal studies.
Collapse
Affiliation(s)
- Marco Rasile
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy.,IRCCS Humanitas Clinical and Research Center, Rozzano, Italy
| | | | - Filippo Mirabella
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
| | - Michela Matteoli
- IRCCS Humanitas Clinical and Research Center, Rozzano, Italy.,CNR Institute of Neuroscience, Milano, Italy
| |
Collapse
|