1
|
Kathuria S, Gupta A, Tracy AR, Luna Ramirez RI, Thulasingam SK, Zaghloul N, Ahmed M, Limesand SW. Systemic inflammatory responses to repeated and increasing endotoxin challenges in fetal sheep. Physiol Rep 2025; 13:e70316. [PMID: 40268878 PMCID: PMC12018166 DOI: 10.14814/phy2.70316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Revised: 03/28/2025] [Accepted: 03/28/2025] [Indexed: 04/25/2025] Open
Abstract
Repeated low-dose administration of lipopolysaccharide (LPS) attenuates subsequent fetal responses, which makes it challenging to investigate interventions to prolonged exposure. Our aim was to develop a fetal inflammatory response syndrome (FIRS) model that consistently and effectively elicits a marked physiological response to increasing LPS doses. Four intravenous LPS boluses (0.3, 1.5, 3, and 15 μg) were administered to fetal sheep over 5 days. Physiological responses were measured via blood gases, pH, lactate, and cortisol concentrations. Fetal peripheral blood mononuclear cells (PBMCs) were analyzed for transcriptomic changes and tissue cytokine expression postmortem. All LPS challenges increased lactate, cortisol, and pCO2 concentrations and decreased pH and pO2 levels at 3 and 5 hours. No interaction was found between day (increasing LPS doses) and hour (LPS response to each dose). PBMC numbers increase with LPS challenges. Transcriptional analysis on PBMCs identified several enriched gene clusters indicating upregulation of inflammatory gene signatures along with complement activation and NFκB signaling pathways. Expression of pro-inflammatory cytokines (TNFα, IL-6, or IL-1β) was measured in lung, heart, liver, placenta, and spleen. Physiological indices show both respiratory and metabolic acidosis with successive and increasing LPS challenges that demonstrate a robust systemic response despite tachyphylaxis to LPS in fetal sheep.
Collapse
Affiliation(s)
- Sanya Kathuria
- School of Animal and Comparative Biomedical SciencesUniversity of ArizonaTucsonArizonaUSA
| | - Akash Gupta
- Division of Neonatology, Department of Pediatrics, College of MedicineUniversity of ArizonaTucsonArizonaUSA
| | - Ayna R. Tracy
- School of Animal and Comparative Biomedical SciencesUniversity of ArizonaTucsonArizonaUSA
| | - Rosa I. Luna Ramirez
- School of Animal and Comparative Biomedical SciencesUniversity of ArizonaTucsonArizonaUSA
| | - Senthil Kumar Thulasingam
- Division of Neonatology, Department of Pediatrics, College of MedicineUniversity of ArizonaTucsonArizonaUSA
| | - Nahla Zaghloul
- Division of Neonatology, Department of Pediatrics, College of MedicineUniversity of ArizonaTucsonArizonaUSA
- Division of Neonatology, Department of Pediatrics, College of MedicineUniversity of FloridaGainesvilleFloridaUSA
| | - Mohamed Ahmed
- Division of Neonatology, Department of Pediatrics, College of MedicineUniversity of ArizonaTucsonArizonaUSA
- Division of Neonatology, Department of Pediatrics, College of MedicineUniversity of FloridaGainesvilleFloridaUSA
| | - Sean W. Limesand
- School of Animal and Comparative Biomedical SciencesUniversity of ArizonaTucsonArizonaUSA
| |
Collapse
|
2
|
Domingues RR, Wiltbank MC, Hernandez LL, Adcock SJJ. Prenatal treatment with the antidepressant fluoxetine on maternal and neonatal behavior in sheep. Pediatr Res 2025:10.1038/s41390-025-03799-3. [PMID: 39809853 DOI: 10.1038/s41390-025-03799-3] [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: 05/14/2024] [Revised: 11/18/2024] [Accepted: 12/15/2024] [Indexed: 01/16/2025]
Abstract
BACKGROUND Fluoxetine is commonly prescribed to treat depression during pregnancy. We aimed to evaluate the effects of prenatal fluoxetine exposure on maternal-offspring behavior in a non-depressed sheep model. METHODS On day 119 ± 1 of a 151-day expected gestation, Hampshire ewes were randomly assigned to receive intravenous fluoxetine (10 mg/kg for the first 2 days and 5 mg/kg daily thereafter until parturition) or a control vehicle. Video was recorded of 8 fluoxetine-treated ewes and 10 control ewes for 2 h before and after parturition. RESULTS Fluoxetine did not alter dam behavior during the peripartum period, including time spent lying before the first birth, lying bout duration, probability of needing birth assistance, duration of birth assistance when provided, and time spent touching her lambs. However, in utero exposure impaired neonatal vigor as lambs spent less time standing and tended to spend less time nursing compared to unexposed lambs. CONCLUSION Neonatal behavioral impairments are consistent with those associated with fluoxetine exposure during human gestation. This effect appears to be independent of maternal behavior, which was unaffected by antidepressant use. IMPACT Lambs exposed to SSRI in utero spent less time standing and tended to spend less time nursing than control lambs, consistent with neonatal behavioral outcomes encountered in clinical practice. The reduced neonatal vigor was likely unrelated to maternal behavior, which was not altered by SSRI treatment. Non-depressed sheep models can help to elucidate the behavioral effects of antidepressant use during pregnancy to enhance health outcomes and patient care.
Collapse
Affiliation(s)
- Rafael R Domingues
- Department of Animal and Dairy Sciences, University of Wisconsin, Madison, WI, USA
- Endocrinology and Reproductive Physiology Program, University of Wisconsin, Madison, WI, USA
- Department of Animal Sciences, The Ohio State University, Columbus, OH, USA
| | - Milo C Wiltbank
- Department of Animal and Dairy Sciences, University of Wisconsin, Madison, WI, USA
- Endocrinology and Reproductive Physiology Program, University of Wisconsin, Madison, WI, USA
| | - Laura L Hernandez
- Department of Animal and Dairy Sciences, University of Wisconsin, Madison, WI, USA
- Endocrinology and Reproductive Physiology Program, University of Wisconsin, Madison, WI, USA
| | - Sarah J J Adcock
- Department of Animal and Dairy Sciences, University of Wisconsin, Madison, WI, USA.
| |
Collapse
|
3
|
Prasad J, Van Steenwinckel J, Gunn AJ, Bennet L, Korzeniewski SJ, Gressens P, Dean JM. Chronic Inflammation Offers Hints About Viable Therapeutic Targets for Preeclampsia and Potentially Related Offspring Sequelae. Int J Mol Sci 2024; 25:12999. [PMID: 39684715 PMCID: PMC11640791 DOI: 10.3390/ijms252312999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 11/22/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024] Open
Abstract
The combination of hypertension with systemic inflammation during pregnancy is a hallmark of preeclampsia, but both processes also convey dynamic information about its antecedents and correlates (e.g., fetal growth restriction) and potentially related offspring sequelae. Causal inferences are further complicated by the increasingly frequent overlap of preeclampsia, fetal growth restriction, and multiple indicators of acute and chronic inflammation, with decreased gestational length and its correlates (e.g., social vulnerability). This complexity prompted our group to summarize information from mechanistic studies, integrated with key clinical evidence, to discuss the possibility that sustained or intermittent systemic inflammation-related phenomena offer hints about viable therapeutic targets, not only for the prevention of preeclampsia, but also the neurobehavioral and other developmental deficits that appear to be overrepresented in surviving offspring. Importantly, we feel that carefully designed hypothesis-driven observational studies are necessary if we are to translate the mechanistic evidence into child health benefits, namely because multiple pregnancy disorders might contribute to heightened risks of neuroinflammation, arrested brain development, or dysconnectivity in survivors who exhibit developmental problems later in life.
Collapse
Affiliation(s)
- Jaya Prasad
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1142, New Zealand; (J.P.); (A.J.G.); (L.B.); (J.M.D.)
| | | | - Alistair J. Gunn
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1142, New Zealand; (J.P.); (A.J.G.); (L.B.); (J.M.D.)
| | - Laura Bennet
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1142, New Zealand; (J.P.); (A.J.G.); (L.B.); (J.M.D.)
| | - Steven J. Korzeniewski
- C.S. Mott Center for Human Growth and Development, Department of Emergency Medicine, Wayne State University School of Medicine, Detroit, MI 48202, USA
| | - Pierre Gressens
- Inserm, Neurodiderot, Université de Paris, 75019 Paris, France;
- Centre for the Developing Brain, Division of Imaging Sciences and Department of Biomedical Engineering, King’s College London, King’s Health Partners, St. Thomas’ Hospital, London SE1 7EH, UK
| | - Justin M. Dean
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1142, New Zealand; (J.P.); (A.J.G.); (L.B.); (J.M.D.)
| |
Collapse
|
4
|
Alayli Y, Brown LS, Tolentino-Plata K, Leon R, Heyne R, Chalak LF, Mir IN. Neurodevelopmental outcomes in extremely preterm infants with placental pathologic evidence of fetal inflammatroy response. Pediatr Res 2024:10.1038/s41390-024-03391-1. [PMID: 39112787 DOI: 10.1038/s41390-024-03391-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 06/13/2024] [Accepted: 06/23/2024] [Indexed: 11/28/2024]
Abstract
OBJECTIVES Neonates born with fetal inflammatory response (FIR) are at increased risk for adverse neonatal outcomes. Our objective was to determine whether FIR and its severity is associated with neurodevelopmental impairment (NDI) at 2 years of age or death among preterm infants. METHODS A retrospective cohort study of prospectively collected data of all infants born <29 weeks gestational age (GA). FIR and its severity were diagnosed according to the Amsterdam Placental Workshop Group Consensus Statement. Neurodevelopmental outcomes among all participants were quantified according to Bayley III. RESULTS Mothers of infants with FIR were significantly younger (P = 0.04) and had a greater prevalence of antenatal steroid use (P < 0.01), infection during pregnancy (P = 0.01), PPROM (P < 0.01), and clinical chorioamnionitis (P < 0.01). Infants with FIR had longer duration of hospitalization (P < 0.01), days on oxygen (P < 0.01), congenital pneumonia (P = 0.03), moderate/severe bronchopulmonary dysplasia (BPD; P < 0.01). Notably, infants with FIR were not at increased risk of NDI or death (primary outcome). Those with moderate to severe FIR (≥ stage 2 FIR) were at increased risk of developing motor & language impairment or death (P < 0.01). CONCLUSION This is the first report demonstrating an association between the severity of FIR and subsequent NDI in preterm infants born. IMPACT STATEMENT Fetal Inflammatory Response (FIR) is not associated Neurodevelopmental Impairement (NDI) or Death in preterm infants However, there is significant relationship between moderate to severe FIR and NDI at 2 years of age in preterm infants. This is the first study demonstrating the impact of progression and severity of FIR on NDI or Death in preterm infants. These observations provide additional insight into understanding the impact of intrauterine exposure to inflammation on the NDI or death in preterm infants.
Collapse
Affiliation(s)
- Yasmeen Alayli
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | | | - Kristine Tolentino-Plata
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Rachel Leon
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Roy Heyne
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Lina F Chalak
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Imran N Mir
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
5
|
White TA, Miller SL, Sutherland AE, Allison BJ, Camm EJ. Perinatal compromise affects development, form, and function of the hippocampus part one; clinical studies. Pediatr Res 2024; 95:1698-1708. [PMID: 38519794 PMCID: PMC11245394 DOI: 10.1038/s41390-024-03105-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 01/30/2024] [Accepted: 02/05/2024] [Indexed: 03/25/2024]
Abstract
The hippocampus is a neuron-rich specialised brain structure that plays a central role in the regulation of emotions, learning and memory, cognition, spatial navigation, and motivational processes. In human fetal development, hippocampal neurogenesis is principally complete by mid-gestation, with subsequent maturation comprising dendritogenesis and synaptogenesis in the third trimester of pregnancy and infancy. Dendritogenesis and synaptogenesis underpin connectivity. Hippocampal development is exquisitely sensitive to perturbations during pregnancy and at birth. Clinical investigations demonstrate that preterm birth, fetal growth restriction (FGR), and acute hypoxic-ischaemic encephalopathy (HIE) are common perinatal complications that alter hippocampal development. In turn, deficits in hippocampal development and structure mediate a range of neurodevelopmental disorders, including cognitive and learning problems, autism, and Attention-Deficit/Hyperactivity Disorder (ADHD). In this review, we summarise the developmental profile of the hippocampus during fetal and neonatal life and examine the hippocampal deficits observed following common human pregnancy complications. IMPACT: The review provides a comprehensive summary of the developmental profile of the hippocampus in normal fetal and neonatal life. We address a significant knowledge gap in paediatric research by providing a comprehensive summary of the relationship between pregnancy complications and subsequent hippocampal damage, shedding new light on this critical aspect of early neurodevelopment.
Collapse
Affiliation(s)
- Tegan A White
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia.
- Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC, Australia.
| | - Suzanne L Miller
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC, Australia
| | - Amy E Sutherland
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC, Australia
| | - Beth J Allison
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC, Australia
| | - Emily J Camm
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia.
- Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC, Australia.
| |
Collapse
|
6
|
White TA, Miller SL, Sutherland AE, Allison BJ, Camm EJ. Perinatal compromise affects development, form, and function of the hippocampus part two; preclinical studies. Pediatr Res 2024; 95:1709-1719. [PMID: 38519795 PMCID: PMC11245392 DOI: 10.1038/s41390-024-03144-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 02/15/2024] [Accepted: 03/03/2024] [Indexed: 03/25/2024]
Abstract
The hippocampus is a vital brain structure deep in the medial temporal lobe that mediates a range of functions encompassing emotional regulation, learning, memory, and cognition. Hippocampal development is exquisitely sensitive to perturbations and adverse conditions during pregnancy and at birth, including preterm birth, fetal growth restriction (FGR), acute hypoxic-ischaemic encephalopathy (HIE), and intrauterine inflammation. Disruptions to hippocampal development due to these conditions can have long-lasting functional impacts. Here, we discuss a range of preclinical models of prematurity and FGR and conditions that induce hypoxia and inflammation, which have been critical in elucidating the underlying mechanisms and cellular and subcellular structures implicated in hippocampal dysfunction. Finally, we discuss potential therapeutic targets to reduce the burden of these perinatal insults on the developing hippocampus. IMPACT: The review explores the preclinical literature examining the association between pregnancy and birth complications, and hippocampal form and function. The developmental processes and cellular mechanisms that are disrupted within the hippocampus following perinatal compromise are described, and potential therapeutic targets are discussed.
Collapse
Affiliation(s)
- Tegan A White
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia.
- Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC, Australia.
| | - Suzanne L Miller
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC, Australia
| | - Amy E Sutherland
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC, Australia
| | - Beth J Allison
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC, Australia
| | - Emily J Camm
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia.
- Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC, Australia.
| |
Collapse
|
7
|
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
|
8
|
Lipopolysaccharide-Induced Immunological Tolerance in Monocyte-Derived Dendritic Cells. IMMUNO 2022. [DOI: 10.3390/immuno2030030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Bacterial lipopolysaccharides (LPS), also referred to as endotoxins, are major outer surface membrane components present on almost all Gram-negative bacteria and are major determinants of sepsis-related clinical complications including septic shock. LPS acts as a strong stimulator of innate or natural immunity in a wide variety of eukaryotic species ranging from insects to humans including specific effects on the adaptive immune system. However, following immune stimulation, lipopolysaccharide can induce tolerance which is an essential immune-homeostatic response that prevents overactivation of the inflammatory response. The tolerance induced by LPS is a state of reduced immune responsiveness due to persistent and repeated challenges, resulting in decreased expression of pro-inflammatory modulators and up-regulation of antimicrobials and other mediators that promote a reduction of inflammation. The presence of environmental-derived LPS may play a key role in decreasing autoimmune diseases and gut tolerance to the plethora of ingested antigens. The use of LPS may be an important immune adjuvant as demonstrated by the promotion of IDO1 increase when present in the fusion protein complex of CTB-INS (a chimera of the cholera toxin B subunit linked to proinsulin) that inhibits human monocyte-derived DC (moDC) activation, which may act through an IDO1-dependent pathway. The resultant state of DC tolerance can be further enhanced by the presence of residual E. coli lipopolysaccharide (LPS) which is almost always present in partially purified CTB-INS preparations. The approach to using an adjuvant with an autoantigen in immunotherapy promises effective treatment for devastating tissue-specific autoimmune diseases like multiple sclerosis (MS) and type 1 diabetes (T1D).
Collapse
|
9
|
Zhou J, Teng Y, Zhang F, Ru X, Li P, Wang J, Yan S, Zhu P, Tao F, Huang K. Sex-specific association between placental inflammatory cytokine mRNA expression and preschoolers' behavioral development: The Ma'anshan birth cohort study. Brain Behav Immun 2022; 104:110-121. [PMID: 35661681 DOI: 10.1016/j.bbi.2022.05.017] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 04/25/2022] [Accepted: 05/29/2022] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Placental inflammation may contribute to brain abnormalities and childhood neuropsychiatric disorders, but limited knowledge is available on the association of placental inflammatory cytokine levels and offspring's behavioral development. This study aimed to examine the sex-specific association between placental inflammatory cytokine mRNA expression and preschoolers' behavioral development. METHODS 3474 pregnant women were recruited as the initial study population in the Ma'anshan birth cohort (MABC) study. Placentas (n = 2519) were collected during childbirth, and the mRNA expression of IL-8, IL-1β, CRP, TNF-α, IL-6, IL-10, and IL-4 was assessed. The Child Behavior Checklist 1.5-5 (CBCL 1.5-5) was used to assess children's behavioral development at 4 years old. A T-score ≥ 60 on summary scales or a score ≥ 65 on syndrome scales was regarded as the borderline clinical range. Multiple linear regression models and binary logistic regression models were applied to explore the sex-specific associations between placental inflammatory cytokines mRNA transcript levels and preschoolers' behavioral development. RESULTS Sex-specific associations between placental inflammatory cytokines mRNA expression and preschoolers' behavioral development were observed. There was a positive association between IL-8 and CBCL scores for boys on anxious/depressed problems, aggressive behaviors, externalizing problems and total problems. Logistic regression models showed that high levels of IL-8 were associated with a higher risk of girls' emotionally reactive problems and sleep problems compared to low/medium levels. High TNF-α was correlated with increased sleep problem scores in boys, and medium TNF-α (vs. low levels) was associated with an increased risk of girls' externalizing problems. Medium levels of CRP, IL-1β, and IL-6 were found to be associated with a decreased risk of girls' behavioral problems compared to low/high levels. For anti-inflammatory cytokines, medium IL-10 and IL-4 (vs. low levels) were observed to be associated with a lower risk of internalizing problems in boys and externalizing problems in girls, respectively. High IL-10 was correlated with decreased attention problem scores in boys. CONCLUSION This study indicates that placental inflammatory cytokine mRNA expression of IL-8, CRP, TNF-α, IL-1β, IL-4 and IL-10 may be associated with preschoolers' behavioral development in a sex-specific manner.
Collapse
Affiliation(s)
- Jixing Zhou
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University; Key Laboratory of Population Health Across Life Cycle (AHMU), MOE, Hefei 230032, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Hefei 230032, China; Anhui Provincial Key Laboratory of Population Health and Aristogenics, Hefei 230032, China
| | - Yuzhu Teng
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University; Key Laboratory of Population Health Across Life Cycle (AHMU), MOE, Hefei 230032, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Hefei 230032, China; Anhui Provincial Key Laboratory of Population Health and Aristogenics, Hefei 230032, China
| | - Fu Zhang
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University; Key Laboratory of Population Health Across Life Cycle (AHMU), MOE, Hefei 230032, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Hefei 230032, China; Anhui Provincial Key Laboratory of Population Health and Aristogenics, Hefei 230032, China
| | - Xue Ru
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University; Key Laboratory of Population Health Across Life Cycle (AHMU), MOE, Hefei 230032, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Hefei 230032, China; Anhui Provincial Key Laboratory of Population Health and Aristogenics, Hefei 230032, China
| | - Peixuan Li
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University; Key Laboratory of Population Health Across Life Cycle (AHMU), MOE, Hefei 230032, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Hefei 230032, China; Anhui Provincial Key Laboratory of Population Health and Aristogenics, Hefei 230032, China
| | - Jianqing Wang
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University
| | - Shuangqin Yan
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University; Maternal and Child Health Care Center of Ma'anshan, No 24 Jiashan Road, Ma'anshan 243011, Anhui, China
| | - Peng Zhu
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University; Key Laboratory of Population Health Across Life Cycle (AHMU), MOE, Hefei 230032, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Hefei 230032, China; Anhui Provincial Key Laboratory of Population Health and Aristogenics, Hefei 230032, China
| | - Fangbiao Tao
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University; Key Laboratory of Population Health Across Life Cycle (AHMU), MOE, Hefei 230032, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Hefei 230032, China; Anhui Provincial Key Laboratory of Population Health and Aristogenics, Hefei 230032, China
| | - Kun Huang
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University; Key Laboratory of Population Health Across Life Cycle (AHMU), MOE, Hefei 230032, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Hefei 230032, China; Anhui Provincial Key Laboratory of Population Health and Aristogenics, Hefei 230032, China; Scientific Research Center in Preventive Medicine, School of Public Health, Anhui Medical University, Anhui Province, China.
| |
Collapse
|
10
|
Murata T, Kyozuka H, Yasuda S, Fukuda T, Tanaka T, Fujimori K. Effects of maternal ritodrine hydrochloride administration on the heart rate of preterm fetal sheep with intraamniotic inflammation. PLoS One 2022; 17:e0265872. [PMID: 35358222 PMCID: PMC8970407 DOI: 10.1371/journal.pone.0265872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 03/04/2022] [Indexed: 11/18/2022] Open
Abstract
Ritodrine hydrochloride is used for pregnancy prolongation and intrauterine fetal resuscitation. However, its clinical significance in intraamniotic inflammation during preterm labor and intrauterine fetal distress is unclear. We investigated the effects of maternal ritodrine hydrochloride administration (MRA; 200 μg/min for 2 h, followed by 800 μg/min for 2 h after 24 h) on fetal physiological parameters. For this purpose, we used chronically instrumented pregnant sheep at 113–119 d (term = 145 d) of gestation without (Group 1, n = 5) and with (Group 2, n = 5) intraamniotic inflammation induced by lipopolysaccharide injection into the amniotic cavity. The changes in fetal heart rate (FHR) and short-term variability (STV) and long-term variability (LTV) in FHR, fetal blood pressure, and fetal arterial blood gas (FABG) values were measured before and at 1 and 2 h after initiating MRA. Before MRA, all parameters were similar between Groups 1 and 2; however, there was significantly higher STV in Group 2 than in Group 1 before MRA at 800 μg/min, significantly higher partial arterial pressure of carbon dioxide in FABG in Group 2 than in Group 1 before MRA at 200 μg/min, and significantly lower blood glucose (BG) in Group 2 than in Group 1 before MRA at 800 μg/min. One hour after MRA, the FHR, STV, and LTV were significantly higher at 800 μg/min than those at the baseline in Group 1, as determined by the Friedman test; however, no significant difference was observed in Group 2. Additionally, the FABG pH significantly decreased 1 h after MRA at 800 μg/min in Group 2, whereas FABG lactate and BG significantly increased 2 h after MRA at 800 μg/min in Groups 1 and 2. Thus, short-term MRA at 800 μg/min increased the FHR, STV, and LTV significantly; these values were further modified under intraamniotic inflammation.
Collapse
Affiliation(s)
- Tsuyoshi Murata
- Department of Obstetrics and Gynecology, Fukushima Medical University School of Medicine, Fukushima, Japan
- * E-mail:
| | - Hyo Kyozuka
- Department of Obstetrics and Gynecology, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Shun Yasuda
- Department of Obstetrics and Gynecology, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Toma Fukuda
- Department of Obstetrics and Gynecology, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Teruyoshi Tanaka
- Department of Biomolecular Science, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Keiya Fujimori
- Department of Obstetrics and Gynecology, Fukushima Medical University School of Medicine, Fukushima, Japan
| |
Collapse
|
11
|
Geraniol improved memory impairment and neurotoxicity induced by zinc oxide nanoparticles in male wistar rats through its antioxidant effect. Life Sci 2021; 282:119823. [PMID: 34273375 DOI: 10.1016/j.lfs.2021.119823] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 06/27/2021] [Accepted: 07/06/2021] [Indexed: 11/23/2022]
Abstract
AIMS Zinc oxide nanoparticles (ZnO-NPs) are currently applied in food and pharmaceutical industries whose neurotoxic effect on the central nervous system (CNS) is a major concern. Considering the pharmacological properties (antioxidant, anti-inflammatory) of the geraniol (GE), we aimed to investigate the efficacy of geraniol on ZnO-NPs neurotoxicity. MATERIALS AND METHODS We used 32 male Wistar rats, randomly assigned to four groups (n = 8): Control, GE (daily received 100 mg/kg of GE by gavage), ZnO-NPs (received intraperitoneal injection of 75 mg/kg of ZnO-NPs twice a week), and ZnO-NPs + GE (received both GE and ZnO-NPs at same doses above during 4 weeks). Morris water maze (MWM) and Y-maze tasks were done to evaluate learning and memory function. Biochemical assays were done to measure total antioxidant capacity (TAC), malondialdehyde (MDA), superoxide dismutase (SOD), glutathione peroxidase (GPX) and ZnO-NPs bioaccumulation. Nissl and H&E staining were performed for histological evaluations. KEY FINDINGS The results of behavioral study revealed that GE improved learning and memory impairment induced by ZnO-NPs. Moreover, neuroprotective effect of GE significantly decreased pathological parameters such as necrosis and gliosis, and consequently increased the number of nerve cells in the cortex and different hippocampal areas. Furthermore, biochemical studies demonstrated that GE significantly increased antioxidant indices (namely, TAC, SOD, and GPX) and reduced oxidative stress marker (MDA) and Zn bioaccumulation in ZnO-NPs treated animals. SIGNIFICANCE Our results provide experimental evidence to further investigate the precise mechanisms underlying the geraniol as a promising therapeutic approach for improvement of cognitive function and neurotoxicity induce by ZnO-NPs.
Collapse
|
12
|
Usuda H, Watanabe S, Saito M, Ikeda H, Koshinami S, Sato S, Musk GC, Fee E, Carter S, Kumagai Y, Takahashi T, Takahashi Y, Kawamura S, Hanita T, Kure S, Yaegashi N, Newnham JP, Kemp MW. Successful use of an artificial placenta-based life support system to treat extremely preterm ovine fetuses compromised by intrauterine inflammation. Am J Obstet Gynecol 2020; 223:755.e1-755.e20. [PMID: 32380175 DOI: 10.1016/j.ajog.2020.04.036] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Revised: 04/23/2020] [Accepted: 04/28/2020] [Indexed: 01/03/2023]
Abstract
BACKGROUND Ex vivo uterine environment therapy is an experimental intensive care strategy for extremely preterm infants born between 21 and 24 weeks of gestation. Gas exchange is performed by membranous oxygenators connected by catheters to the umbilical vessels. The fetus is submerged in a bath of synthetic amniotic fluid. The lungs remain fluid filled, and pulmonary respiration does not occur. Intrauterine inflammation is strongly associated with extremely preterm birth and fetal injury. At present, there are no data that we are aware of to show that artificial placenta-based systems can be used to support extremely preterm fetuses compromised by exposure to intrauterine inflammation. OBJECTIVE To evaluate the ability of our ex vivo uterine environment therapy platform to support extremely preterm ovine fetuses (95-day gestational age; approximately equivalent to 24 weeks of human gestation) exposed to intrauterine inflammation for a period of 120 hours, the following primary endpoints were chosen: (1) maintenance of key physiological variables within normal ranges, (2) absence of infection and inflammation, (3) absence of brain injury, and (4) gross fetal growth and cardiovascular function matching that of age-matched in utero controls. STUDY DESIGN Ten ewes with singleton pregnancies were each given a single intraamniotic injection of 10-mg Escherichia coli lipopolysaccharides under ultrasound guidance 48 hours before undergoing surgical delivery for adaptation to ex vivo uterine environment therapy at 95-day gestation (term=150 days). Fetuses were adapted to ex vivo uterine environment therapy and maintained for 120 hours with constant monitoring of key vital parameters (ex vivo uterine environment group) before being killed at 100-day equivalent gestational age. Umbilical artery blood samples were regularly collected to assess blood gas data, differential counts, biochemical parameters, inflammatory markers, and microbial load to exclude infection. Ultrasound was conducted at 48 hours after intraamniotic lipopolysaccharides (before surgery) to confirm fetal viability and at the conclusion of the experiments (before euthanasia) to evaluate cardiac function. Brain injury was evaluated by gross anatomic and histopathologic investigations. Eight singleton pregnant control animals were similarly exposed to intraamniotic lipopolysaccharides at 93-day gestation and were killed at 100-day gestation to allow comparative postmortem analyses (control group). Biobanked samples from age-matched saline-treated animals served as an additional comparison group. Successful instillation of lipopolysaccharides into the amniotic fluid exposure was confirmed by amniotic fluid analysis at the time of administration and by analyzing cytokine levels in fetal plasma and amniotic fluid. Data were tested for mean differences using analysis of variance. RESULTS Six of 8 lipopolysaccharide control group (75%) and 8 of 10 ex vivo uterine environment group fetuses (80%) successfully completed their protocols. Six of 8 ex vivo uterine environment group fetuses required dexamethasone phosphate treatment to manage profound refractory hypotension. Weight and crown-rump length were reduced in ex vivo uterine environment group fetuses at euthanasia than those in lipopolysaccharide control group fetuses (P<.05). There were no biologically significant differences in cardiac ultrasound measurement, differential leukocyte counts (P>.05), plasma tumor necrosis factor α, monocyte chemoattractant protein-1 concentrations (P>.05), or liver function tests between groups. Daily blood cultures were negative for aerobic and anaerobic growth in all ex vivo uterine environment group animals. No cases of intraventricular hemorrhage were observed. White matter injury was identified in 3 of 6 lipopolysaccharide control group fetuses and 3 of 8 vivo uterine environment group fetuses. CONCLUSION We report the use of an artificial placenta-based system to support extremely preterm lambs compromised by exposure to intrauterine inflammation. Our data highlight key challenges (refractory hypotension, growth restriction, and white matter injury) to be overcome in the development and use of artificial placenta technology for extremely preterm infants. As such challenges seem largely absent from studies based on healthy pregnancies, additional experiments of this nature using clinically relevant model systems are essential for further development of this technology and its eventual clinical application.
Collapse
Affiliation(s)
- Haruo Usuda
- Division of Obstetrics and Gynecology, The University of Western Australia, Crawley, Western Australia, Australia; Center for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Miyagi, Japan.
| | - Shimpei Watanabe
- Center for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Miyagi, Japan
| | - Masatoshi Saito
- Division of Obstetrics and Gynecology, The University of Western Australia, Crawley, Western Australia, Australia; Center for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Miyagi, Japan
| | - Hideyuki Ikeda
- Center for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Miyagi, Japan
| | - Shota Koshinami
- Center for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Miyagi, Japan
| | - Shinichi Sato
- Center for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Miyagi, Japan
| | - Gabrielle C Musk
- Division of Obstetrics and Gynecology, The University of Western Australia, Crawley, Western Australia, Australia; Animal Care Services, The University of Western Australia, Crawley, Western Australia, Australia
| | - Erin Fee
- Division of Obstetrics and Gynecology, The University of Western Australia, Crawley, Western Australia, Australia
| | - Sean Carter
- Division of Obstetrics and Gynecology, The University of Western Australia, Crawley, Western Australia, Australia
| | - Yusaku Kumagai
- Center for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Miyagi, Japan
| | - Tsukasa Takahashi
- Division of Obstetrics and Gynecology, The University of Western Australia, Crawley, Western Australia, Australia; Center for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Miyagi, Japan
| | - Yuki Takahashi
- Division of Obstetrics and Gynecology, The University of Western Australia, Crawley, Western Australia, Australia; Center for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Miyagi, Japan
| | | | - Takushi Hanita
- Center for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Miyagi, Japan
| | - Shigeo Kure
- Center for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Miyagi, Japan
| | - Nobuo Yaegashi
- Center for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Miyagi, Japan
| | - John P Newnham
- Division of Obstetrics and Gynecology, The University of Western Australia, Crawley, Western Australia, Australia; School of Veterinary and Life Sciences, Murdoch University, Western Australia, Australia
| | - Matthew W Kemp
- Division of Obstetrics and Gynecology, The University of Western Australia, Crawley, Western Australia, Australia; Center for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Miyagi, Japan; School of Veterinary and Life Sciences, Murdoch University, Western Australia, Australia
| |
Collapse
|
13
|
Zarate MA, Wesolowski SR, Nguyen LM, De Dios RK, Wilkening RB, Rozance PJ, Wright CJ. In utero inflammatory challenge induces an early activation of the hepatic innate immune response in late gestation fetal sheep. Innate Immun 2020; 26:549-564. [PMID: 32538259 PMCID: PMC7556190 DOI: 10.1177/1753425920928388] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 04/20/2020] [Accepted: 04/26/2020] [Indexed: 12/15/2022] Open
Abstract
Chorioamnionitis is associated with inflammatory end-organ damage in the fetus. Tissues in direct contact with amniotic fluid drive a pro-inflammatory response and contribute to this injury. However, due to a lack of direct contact with the amniotic fluid, the liver contribution to this response has not been fully characterized. Given its role as an immunologic organ, we hypothesized that the fetal liver would demonstrate an early innate immune response to an in utero inflammatory challenge. Fetal sheep (131 ± 1 d gestation) demonstrated metabolic acidosis and high cortisol and norepinephrine values within 5 h of exposure to intra-amniotic LPS. Likewise, expression of pro-inflammatory cytokines increased significantly at 1 and 5 h of exposure. This was associated with NF-κB activation, by inhibitory protein IκBα degradation, and nuclear translocation of NF-κB subunits (p65/p50). Corroborating these findings, LPS exposure significantly increased pro-inflammatory innate immune gene expression in fetal sheep hepatic macrophages in vitro. Thus, an in utero inflammatory challenge induces an early hepatic innate immune response with systemic metabolic and stress responses. Within the fetal liver, hepatic macrophages respond robustly to LPS exposure. Our results demonstrate that the fetal hepatic innate immune response must be considered when developing therapeutic approaches to attenuate end-organ injury associated with in utero inflammation.
Collapse
Affiliation(s)
- Miguel A Zarate
- Section of Neonatology, Department of Pediatrics, Children’s Hospital Colorado, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Stephanie R Wesolowski
- Section of Neonatology, Department of Pediatrics, Children’s Hospital Colorado, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Leanna M Nguyen
- Section of Neonatology, Department of Pediatrics, Children’s Hospital Colorado, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Robyn K De Dios
- Section of Neonatology, Department of Pediatrics, Children’s Hospital Colorado, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Randall B Wilkening
- Section of Neonatology, Department of Pediatrics, Children’s Hospital Colorado, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Paul J Rozance
- Section of Neonatology, Department of Pediatrics, Children’s Hospital Colorado, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Clyde J Wright
- Section of Neonatology, Department of Pediatrics, Children’s Hospital Colorado, University of Colorado School of Medicine, Aurora, CO 80045, USA
| |
Collapse
|
14
|
Jung E, Romero R, Yeo L, Diaz-Primera R, Marin-Concha J, Para R, Lopez AM, Pacora P, Gomez-Lopez N, Yoon BH, Kim CJ, Berry SM, Hsu CD. The fetal inflammatory response syndrome: the origins of a concept, pathophysiology, diagnosis, and obstetrical implications. Semin Fetal Neonatal Med 2020; 25:101146. [PMID: 33164775 PMCID: PMC10580248 DOI: 10.1016/j.siny.2020.101146] [Citation(s) in RCA: 131] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The fetus can deploy a local or systemic inflammatory response when exposed to microorganisms or, alternatively, to non-infection-related stimuli (e.g., danger signals or alarmins). The term "Fetal Inflammatory Response Syndrome" (FIRS) was coined to describe a condition characterized by evidence of a systemic inflammatory response, frequently a result of the activation of the innate limb of the immune response. FIRS can be diagnosed by an increased concentration of umbilical cord plasma or serum acute phase reactants such as C-reactive protein or cytokines (e.g., interleukin-6). Pathologic evidence of a systemic fetal inflammatory response indicates the presence of funisitis or chorionic vasculitis. FIRS was first described in patients at risk for intraamniotic infection who presented preterm labor with intact membranes or preterm prelabor rupture of the membranes. However, FIRS can also be observed in patients with sterile intra-amniotic inflammation, alloimmunization (e.g., Rh disease), and active autoimmune disorders. Neonates born with FIRS have a higher rate of complications, such as early-onset neonatal sepsis, intraventricular hemorrhage, periventricular leukomalacia, and death, than those born without FIRS. Survivors are at risk for long-term sequelae that may include bronchopulmonary dysplasia, neurodevelopmental disorders, such as cerebral palsy, retinopathy of prematurity, and sensorineuronal hearing loss. Experimental FIRS can be induced by intra-amniotic administration of bacteria, microbial products (such as endotoxin), or inflammatory cytokines (such as interleukin-1), and animal models have provided important insights about the mechanisms responsible for multiple organ involvement and dysfunction. A systemic fetal inflammatory response is thought to be adaptive, but, on occasion, may become dysregulated whereby a fetal cytokine storm ensues and can lead to multiple organ dysfunction and even fetal death if delivery does not occur ("rescued by birth"). Thus, the onset of preterm labor in this context can be considered to have survival value. The evidence so far suggests that FIRS may compound the effects of immaturity and neonatal inflammation, thus increasing the risk of neonatal complications and long-term morbidity. Modulation of a dysregulated fetal inflammatory response by the administration of antimicrobial agents, anti-inflammatory agents, or cell-based therapy holds promise to reduce infant morbidity and mortality.
Collapse
Affiliation(s)
- Eunjung Jung
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD, and Detroit, MI, USA; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Roberto Romero
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD, and Detroit, MI, USA; Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, USA; Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI, USA; Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, USA; Detroit Medical Center, Detroit, MI, USA; Department of Obstetrics and Gynecology, Florida International University, Miami, FL, USA.
| | - Lami Yeo
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD, and Detroit, MI, USA; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Ramiro Diaz-Primera
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD, and Detroit, MI, USA; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Julio Marin-Concha
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD, and Detroit, MI, USA; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Robert Para
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD, and Detroit, MI, USA; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Ashley M Lopez
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD, and Detroit, MI, USA; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Percy Pacora
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD, and Detroit, MI, USA; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Nardhy Gomez-Lopez
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD, and Detroit, MI, USA; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA; Department of Biochemistry, Microbiology and Immunology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Bo Hyun Yoon
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD, and Detroit, MI, USA; Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Chong Jai Kim
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD, and Detroit, MI, USA; Department of Pathology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Stanley M Berry
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD, and Detroit, MI, USA; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Chaur-Dong Hsu
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD, and Detroit, MI, USA; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA; Department of Physiology, Wayne State University School of Medicine, Detroit, MI, USA
| |
Collapse
|
15
|
Gutiérrez-Expósito D, Arteche-Villasol N, Vallejo-García R, Ferreras-Estrada MC, Ferre I, Sánchez-Sánchez R, Ortega-Mora LM, Pérez V, Benavides J. Characterization of Fetal Brain Damage in Early Abortions of Ovine Toxoplasmosis. Vet Pathol 2020; 57:535-544. [PMID: 32406321 DOI: 10.1177/0300985820921539] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
There is an unacknowledged clinical presentation of ovine toxoplasmosis characterized by early abortions and lesions of fetal leukoencephalomalacia. To investigate the pathogenesis of this condition, the extent and distribution of leukomalacia and the variations in the cell populations associated with it were characterized in 32 fetal brains from 2 previously published experimental studies of Toxoplasma gondii infection in pregnant sheep. Immunohistochemical labeling of βAPP allowed for the detection of leukomalacia in 100/110 (91%) studied samples. There was no clear influence of the challenge dose or the area of the brain (frontal lobe, corpus callosum, midbrain, and cerebellum). In tissues with leukomalacia, there was loss of oligodendrocytes and increased number of astrocytes and microglia both in the areas of necrosis but also in the surrounding area. These findings were similar to those described in ovine experimental models (inflammation syndrome and hypoxic models) of periventricular leukomalacia in humans. Thus, a fetal inflammatory syndrome may be involved in the pathogenesis of early abortion in ovine toxoplasmosis. However, further studies are needed to determine the pathogenesis of this clinical presentation because placental thrombosis and resulting hypoxia could also be responsible for the leukomalacia.
Collapse
Affiliation(s)
- Daniel Gutiérrez-Expósito
- Universidad de León, Campus de Vegazana, León, Spain.,Instituto de Ganadería de Montaña (CSIC-Universidad de León), Grulleros, León, Spain
| | - Noive Arteche-Villasol
- Universidad de León, Campus de Vegazana, León, Spain.,Instituto de Ganadería de Montaña (CSIC-Universidad de León), Grulleros, León, Spain
| | - Raquel Vallejo-García
- Universidad de León, Campus de Vegazana, León, Spain.,Instituto de Ganadería de Montaña (CSIC-Universidad de León), Grulleros, León, Spain
| | - María C Ferreras-Estrada
- Universidad de León, Campus de Vegazana, León, Spain.,Instituto de Ganadería de Montaña (CSIC-Universidad de León), Grulleros, León, Spain
| | | | | | | | - Valentín Pérez
- Universidad de León, Campus de Vegazana, León, Spain.,Instituto de Ganadería de Montaña (CSIC-Universidad de León), Grulleros, León, Spain
| | - Julio Benavides
- Instituto de Ganadería de Montaña (CSIC-Universidad de León), Grulleros, León, Spain
| |
Collapse
|
16
|
Galinsky R, Dhillon SK, Dean JM, Davidson JO, Lear CA, Wassink G, Nott F, Kelly SB, Fraser M, Yuill C, Bennet L, Gunn AJ. Tumor necrosis factor inhibition attenuates white matter gliosis after systemic inflammation in preterm fetal sheep. J Neuroinflammation 2020; 17:92. [PMID: 32293473 PMCID: PMC7087378 DOI: 10.1186/s12974-020-01769-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Accepted: 03/09/2020] [Indexed: 12/21/2022] Open
Abstract
Background Increased circulating levels of tumor necrosis factor (TNF) are associated with greater risk of impaired neurodevelopment after preterm birth. In this study, we tested the hypothesis that systemic TNF inhibition, using the soluble TNF receptor Etanercept, would attenuate neuroinflammation in preterm fetal sheep exposed to lipopolysaccharide (LPS). Methods Chronically instrumented preterm fetal sheep at 0.7 of gestation were randomly assigned to receive saline (control; n = 7), LPS infusion (100 ng/kg i.v. over 24 h then 250 ng/kg/24 h for 96 h plus 1 μg LPS boluses at 48, 72, and 96 h, to induce inflammation; n = 8) or LPS plus two i.v. infusions of Etanercept (2 doses, 5 mg/kg infused over 30 min, 48 h apart) started immediately before LPS-exposure (n = 8). Sheep were killed 10 days after starting infusions, for histology. Results LPS boluses were associated with increased circulating TNF, interleukin (IL)-6 and IL-10, electroencephalogram (EEG) suppression, hypotension, tachycardia, and increased carotid artery perfusion (P < 0.05 vs. control). In the periventricular and intragyral white matter, LPS exposure increased gliosis, TNF-positive cells, total oligodendrocytes, and cell proliferation (P < 0.05 vs control), but did not affect myelin expression or numbers of neurons in the cortex and subcortical regions. Etanercept delayed the rise in circulating IL-6, prolonged the increase in IL-10 (P < 0.05 vs. LPS), and attenuated EEG suppression, hypotension, and tachycardia after LPS boluses. Histologically, Etanercept normalized LPS-induced gliosis, and increase in TNF-positive cells, proliferation, and total oligodendrocytes. Conclusion TNF inhibition markedly attenuated white matter gliosis but did not affect mature oligodendrocytes after prolonged systemic inflammation in preterm fetal sheep. Further studies of long-term brain maturation are now needed.
Collapse
Affiliation(s)
- Robert Galinsky
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, Private bag 92019, Auckland, 1023, New Zealand.,The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia.,Department of Obstetrics and Gynaecology, Monash University, Melbourne, Victoria, Australia
| | - Simerdeep K Dhillon
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, Private bag 92019, Auckland, 1023, New Zealand
| | - Justin M Dean
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, Private bag 92019, Auckland, 1023, New Zealand
| | - Joanne O Davidson
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, Private bag 92019, Auckland, 1023, New Zealand
| | - Christopher A Lear
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, Private bag 92019, Auckland, 1023, New Zealand
| | - Guido Wassink
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, Private bag 92019, Auckland, 1023, New Zealand
| | - Fraser Nott
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia
| | - Sharmony B Kelly
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia
| | - Mhoyra Fraser
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, Private bag 92019, Auckland, 1023, New Zealand
| | - Caroline Yuill
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, Private bag 92019, Auckland, 1023, New Zealand
| | - Laura Bennet
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, Private bag 92019, Auckland, 1023, New Zealand
| | - Alistair Jan Gunn
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, Private bag 92019, Auckland, 1023, New Zealand.
| |
Collapse
|
17
|
Pre- and Neonatal Exposure to Lead (Pb) Induces Neuroinflammation in the Forebrain Cortex, Hippocampus and Cerebellum of Rat Pups. Int J Mol Sci 2020; 21:ijms21031083. [PMID: 32041252 PMCID: PMC7037720 DOI: 10.3390/ijms21031083] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 02/01/2020] [Accepted: 02/03/2020] [Indexed: 12/11/2022] Open
Abstract
Lead (Pb) is a heavy metal with a proven neurotoxic effect. Exposure is particularly dangerous to the developing brain in the pre- and neonatal periods. One postulated mechanism of its neurotoxicity is induction of inflammation. This study analyzed the effect of exposure of rat pups to Pb during periods of brain development on the concentrations of selected cytokines and prostanoids in the forebrain cortex, hippocampus and cerebellum. Methods: Administration of 0.1% lead acetate (PbAc) in drinking water ad libitum, from the first day of gestation to postnatal day 21, resulted in blood Pb in rat pups reaching levels below the threshold considered safe for humans by the Centers for Disease Control and Prevention (10 µg/dL). Enzyme-linked immunosorbent assay (ELISA) method was used to determine the levels of interleukins IL-1β, IL-6, transforming growth factor-β (TGF-β), prostaglandin E2 (PGE2) and thromboxane B2 (TXB2). Western blot and quantitative real-time PCR were used to determine the expression levels of cyclooxygenases COX-1 and COX-2. Finally, Western blot was used to determine the level of nuclear factor kappa B (NF-κB). Results: In all studied brain structures (forebrain cortex, hippocampus and cerebellum), the administration of Pb caused a significant increase in all studied cytokines and prostanoids (IL-1β, IL-6, TGF-β, PGE2 and TXB2). The protein and mRNA expression of COX-1 and COX-2 increased in all studied brain structures, as did NF-κB expression. Conclusions: Chronic pre- and neonatal exposure to Pb induces neuroinflammation in the forebrain cortex, hippocampus and cerebellum of rat pups.
Collapse
|
18
|
Persistent Toll-like receptor 7 stimulation induces behavioral and molecular innate immune tolerance. Brain Behav Immun 2019; 82:338-353. [PMID: 31499172 PMCID: PMC6956569 DOI: 10.1016/j.bbi.2019.09.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 09/04/2019] [Accepted: 09/05/2019] [Indexed: 12/18/2022] Open
Abstract
Toll-like receptors 7 and 8 (TLR7 and TLR8) are endosomal pattern recognition receptors that detect a variety of single-stranded RNA species. While TLR7/8 agonists have robust therapeutic potential, clinical utility of these agents is limited by sickness responses associated with treatment induction. To understand the kinetics and mechanism of these responses, we characterized the acute and chronic effects of TLR7 stimulation. Single-cell RNA-sequencing studies, RNAscope, and radiolabeled in situ hybridization demonstrate that central nervous system gene expression of TLR7 is exclusive to microglia. In vitro studies demonstrate that microglia are highly sensitive to TLR7 stimulation, and respond in a dose-dependent manner to the imidazoquinoline R848. In vivo, both intraperitoneal (IP) and intracerebroventricular (ICV) R848 induce acute sickness responses including hypophagia, weight loss, and decreased voluntary locomotor activity, associated with increased CNS pro-inflammatory gene expression and changes to glial morphology. However, chronic daily IP R848 resulted in rapid tachyphylaxis of behavioral and molecular manifestations of illness. In microglial in vitro assays, pro-inflammatory transcriptional responses rapidly diminished in the context of repeated R848. In addition to TLR7 desensitization, we found that microglia become partially refractory to lipopolysaccharide (LPS) following R848 pretreatment, associated with induction of negative regulators A20 and Irak3. Similarly, mice pre-treated with R848 demonstrate reduced sickness responses, hypothalamic inflammation, and hepatic inflammation in response to LPS. These data combined demonstrate that TLR7 stimulation induces acute behavioral and molecular evidence of sickness responses. Following prolonged dosing, R848 induces a refractory state to both TLR7 and TLR4 activation, consistent with induced immune tolerance.
Collapse
|
19
|
Cavarsan CF, Gorassini MA, Quinlan KA. Animal models of developmental motor disorders: parallels to human motor dysfunction in cerebral palsy. J Neurophysiol 2019; 122:1238-1253. [PMID: 31411933 PMCID: PMC6766736 DOI: 10.1152/jn.00233.2019] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 08/07/2019] [Accepted: 08/08/2019] [Indexed: 12/12/2022] Open
Abstract
Cerebral palsy (CP) is the most common motor disability in children. Much of the previous research on CP has focused on reducing the severity of brain injuries, whereas very few researchers have investigated the cause and amelioration of motor symptoms. This research focus has had an impact on the choice of animal models. Many of the commonly used animal models do not display a prominent CP-like motor phenotype. In general, rodent models show anatomically severe injuries in the central nervous system (CNS) in response to insults associated with CP, including hypoxia, ischemia, and neuroinflammation. Unfortunately, most rodent models do not display a prominent motor phenotype that includes the hallmarks of spasticity (muscle stiffness and hyperreflexia) and weakness. To study motor dysfunction related to developmental injuries, a larger animal model is needed, such as rabbit, pig, or nonhuman primate. In this work, we describe and compare various animal models of CP and their potential for translation to the human condition.
Collapse
Affiliation(s)
- Clarissa F Cavarsan
- George and Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, Rhode Island
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, Rhode Island
| | - Monica A Gorassini
- Department of Biomedical Engineering, University of Alberta, Edmonton, Alberta, Canada
| | - Katharina A Quinlan
- George and Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, Rhode Island
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, Rhode Island
| |
Collapse
|
20
|
Brandenburg JE, Fogarty MJ, Sieck GC. A Critical Evaluation of Current Concepts in Cerebral Palsy. Physiology (Bethesda) 2019; 34:216-229. [PMID: 30968751 PMCID: PMC7938766 DOI: 10.1152/physiol.00054.2018] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 01/11/2019] [Accepted: 01/23/2019] [Indexed: 11/22/2022] Open
Abstract
Spastic cerebral palsy (CP), despite the name, is not consistently identifiable by specific brain lesions. CP animal models focus on risk factors for development of CP, yet few reproduce the diagnostic symptoms. Animal models of CP must advance beyond risk factors to etiologies, including both the brain and spinal cord.
Collapse
Affiliation(s)
- Joline E Brandenburg
- Department of Physical Medicine and Rehabilitation, Mayo Clinic College of Medicine , Rochester, Minnesota
- Department of Pediatric and Adolescent Medicine, Mayo Clinic College of Medicine , Rochester, Minnesota
| | - Matthew J Fogarty
- Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine , Rochester, Minnesota
| | - Gary C Sieck
- Department of Physical Medicine and Rehabilitation, Mayo Clinic College of Medicine , Rochester, Minnesota
- Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine , Rochester, Minnesota
- Department of Anesthesiology, Mayo Clinic College of Medicine , Rochester, Minnesota
| |
Collapse
|
21
|
Feng SYS, Hollis JH, Samarasinghe T, Phillips DJ, Rao S, Yu VYH, Walker AM. Endotoxin-induced cerebral pathophysiology: differences between fetus and newborn. Physiol Rep 2019; 7:e13973. [PMID: 30785235 PMCID: PMC6381816 DOI: 10.14814/phy2.13973] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 11/23/2018] [Accepted: 11/26/2018] [Indexed: 02/07/2023] Open
Abstract
As the comparative pathophysiology of perinatal infection in the fetus and newborn is uncertain, this study contrasted the cerebral effects of endotoxemia in conscious fetal sheep and newborn lambs. Responses to intravenous bacterial endotoxin (lipopolysaccharide, LPS) or normal saline were studied on three consecutive days in fetal sheep (LPS 1 μg/kg, n = 5; normal saline n = 5) and newborn lambs (LPS 2 μg/kg, n = 10; normal saline n = 5). Cerebro-vascular function was assessed by monitoring cerebral blood flow (CBF) and cerebral vascular resistance (CVR) over 12 h each day, and inflammatory responses were assessed by plasma TNF alpha (TNF-α), nitrate and nitrite concentrations. Brain injury was quantified by counting both resting and active macrophages in the caudate nucleus and periventricular white matter (PVWM). An acute cerebral vasoconstriction (within 1 h of LPS injection) occurred in both the fetus (ΔCVR +53%) and newborn (ΔCVR +63%); subsequently prolonged cerebral vasodilatation occurred in the fetus (ΔCVR -33%) in association with double plasma nitrate/nitrite concentrations, but not in the newborn. Abundant infiltration of activated macrophages was observed in both CN and PVWM at each age, with the extent being 2-3 times greater in the fetus (P < 0.001). In conclusion, while the fetus and newborn experience a similar acute disruption of the cerebral circulation after LPS, the fetus suffers a more prolonged circulatory disruption, a greater infiltration of activated macrophages, and an exaggerated susceptibility to brain injury.
Collapse
Affiliation(s)
- Susan Y. S. Feng
- The Ritchie CentreHudson Institute of Medical ResearchClaytonVictoriaAustralia
- Neonatal DirectorateKing Edward Memorial HospitalPerth Children's HospitalSubiacoWestern AustraliaAustralia
| | - Jacob H. Hollis
- Department of PhysiologyMonash UniversityClaytonVictoriaAustralia
| | | | - David J. Phillips
- Academic & Medical PortfolioEpworth HealthCareRichmondVictoriaAustralia
| | - Shripada Rao
- Neonatal DirectorateKing Edward Memorial HospitalPerth Children's HospitalSubiacoWestern AustraliaAustralia
| | - Victor Y. H. Yu
- The Ritchie CentreHudson Institute of Medical ResearchClaytonVictoriaAustralia
- Monash NewbornMonash Medical CentreClaytonVictoriaAustralia
| | - Adrian M. Walker
- The Ritchie CentreHudson Institute of Medical ResearchClaytonVictoriaAustralia
| |
Collapse
|
22
|
Morrison JL, Berry MJ, Botting KJ, Darby JRT, Frasch MG, Gatford KL, Giussani DA, Gray CL, Harding R, Herrera EA, Kemp MW, Lock MC, McMillen IC, Moss TJ, Musk GC, Oliver MH, Regnault TRH, Roberts CT, Soo JY, Tellam RL. Improving pregnancy outcomes in humans through studies in sheep. Am J Physiol Regul Integr Comp Physiol 2018; 315:R1123-R1153. [PMID: 30325659 DOI: 10.1152/ajpregu.00391.2017] [Citation(s) in RCA: 119] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Experimental studies that are relevant to human pregnancy rely on the selection of appropriate animal models as an important element in experimental design. Consideration of the strengths and weaknesses of any animal model of human disease is fundamental to effective and meaningful translation of preclinical research. Studies in sheep have made significant contributions to our understanding of the normal and abnormal development of the fetus. As a model of human pregnancy, studies in sheep have enabled scientists and clinicians to answer questions about the etiology and treatment of poor maternal, placental, and fetal health and to provide an evidence base for translation of interventions to the clinic. The aim of this review is to highlight the advances in perinatal human medicine that have been achieved following translation of research using the pregnant sheep and fetus.
Collapse
Affiliation(s)
- Janna L Morrison
- Early Origins of Adult Health Research Group, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - Mary J Berry
- Department of Paediatrics and Child Health, University of Otago , Wellington , New Zealand
| | - Kimberley J Botting
- Department of Physiology, Development, and Neuroscience, University of Cambridge , Cambridge , United Kingdom
| | - Jack R T Darby
- Early Origins of Adult Health Research Group, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - Martin G Frasch
- Department of Obstetrics and Gynecology, University of Washington , Seattle, Washington
| | - Kathryn L Gatford
- Robinson Research Institute and Adelaide Medical School, University of Adelaide , Adelaide, South Australia , Australia
| | - Dino A Giussani
- Department of Physiology, Development, and Neuroscience, University of Cambridge , Cambridge , United Kingdom
| | - Clint L Gray
- Department of Paediatrics and Child Health, University of Otago , Wellington , New Zealand
| | - Richard Harding
- Department of Anatomy and Developmental Biology, Monash University , Clayton, Victoria , Australia
| | - Emilio A Herrera
- Pathophysiology Program, Biomedical Sciences Institute (ICBM), Faculty of Medicine, University of Chile , Santiago , Chile
| | - Matthew W Kemp
- Division of Obstetrics and Gynecology, University of Western Australia , Perth, Western Australia , Australia
| | - Mitchell C Lock
- Early Origins of Adult Health Research Group, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - I Caroline McMillen
- Early Origins of Adult Health Research Group, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - Timothy J Moss
- The Ritchie Centre, Hudson Institute of Medical Research, Department of Obstetrics and Gynaecology, Monash University , Clayton, Victoria , Australia
| | - Gabrielle C Musk
- Animal Care Services, University of Western Australia , Perth, Western Australia , Australia
| | - Mark H Oliver
- Liggins Institute, University of Auckland , Auckland , New Zealand
| | - Timothy R H Regnault
- Department of Obstetrics and Gynecology and Department of Physiology and Pharmacology, Western University, and Children's Health Research Institute , London, Ontario , Canada
| | - Claire T Roberts
- Robinson Research Institute and Adelaide Medical School, University of Adelaide , Adelaide, South Australia , Australia
| | - Jia Yin Soo
- Early Origins of Adult Health Research Group, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - Ross L Tellam
- Early Origins of Adult Health Research Group, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, South Australia, Australia
| |
Collapse
|
23
|
Bennet L, Dhillon S, Lear CA, van den Heuij L, King V, Dean JM, Wassink G, Davidson JO, Gunn AJ. Chronic inflammation and impaired development of the preterm brain. J Reprod Immunol 2018; 125:45-55. [DOI: 10.1016/j.jri.2017.11.003] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 11/13/2017] [Accepted: 11/24/2017] [Indexed: 12/17/2022]
|
24
|
Stojanovska V, Miller SL, Hooper SB, Polglase GR. The Consequences of Preterm Birth and Chorioamnionitis on Brainstem Respiratory Centers: Implications for Neurochemical Development and Altered Functions by Inflammation and Prostaglandins. Front Cell Neurosci 2018; 12:26. [PMID: 29449803 PMCID: PMC5799271 DOI: 10.3389/fncel.2018.00026] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 01/17/2018] [Indexed: 11/16/2022] Open
Abstract
Preterm birth is a major cause for neonatal morbidity and mortality, and is frequently associated with adverse neurological outcomes. The transition from intrauterine to extrauterine life at birth is particularly challenging for preterm infants. The main physiological driver for extrauterine transition is the establishment of spontaneous breathing. However, preterm infants have difficulty clearing lung liquid, have insufficient surfactant levels, and underdeveloped lungs. Further, preterm infants have an underdeveloped brainstem, resulting in reduced respiratory drive. These factors facilitate the increased requirement for respiratory support. A principal cause of preterm birth is intrauterine infection/inflammation (chorioamnionitis), and infants with chorioamnionitis have an increased risk and severity of neurological damage, but also demonstrate impaired autoresuscitation capacity and prevalent apnoeic episodes. The brainstem contains vital respiratory centers which provide the neural drive for breathing, but the impact of preterm birth and/or chorioamnionitis on this brain region is not well understood. The aim of this review is to provide an overview of the role and function of the brainstem respiratory centers, and to highlight the proposed mechanisms of how preterm birth and chorioamnionitis may affect central respiratory functions.
Collapse
Affiliation(s)
- Vanesa Stojanovska
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia
| | - Suzanne L Miller
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia.,Department of Obstetrics and Gynaecology, Monash University and Hudson Institute of Medical Research, Melbourne, VIC, Australia
| | - Stuart B Hooper
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia.,Department of Obstetrics and Gynaecology, Monash University and Hudson Institute of Medical Research, Melbourne, VIC, Australia
| | - Graeme R Polglase
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia.,Department of Obstetrics and Gynaecology, Monash University and Hudson Institute of Medical Research, Melbourne, VIC, Australia
| |
Collapse
|
25
|
Vagus nerve stimulation in pregnant rats and effects on inflammatory markers in the brainstem of neonates. Pediatr Res 2018; 83:514-519. [PMID: 29053705 PMCID: PMC5866172 DOI: 10.1038/pr.2017.265] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 09/25/2017] [Indexed: 01/29/2023]
Abstract
BackgroundVagus nerve stimulation (VNS) is an Food and Drug Administration-approved method delivering electrical impulses for treatment of depression and epilepsy in adults. The vagus nerve innervates the majority of visceral organs and cervix, but potential impacts of VNS on the progress of pregnancy and the fetus are not well studied.MethodsWe tested the hypothesis that VNS in pregnant dams does not induce inflammatory changes in the cardio-respiratory control regions of the pups' brainstem, potentially impacting the morbidity and mortality of offspring. Pregnant dams were implanted with stimulators providing intermittent low or high frequency electrical stimulation of the sub-diaphragmatic esophageal segment of the vagus nerve for 6-7 days until delivery. After birth, we collected pup brainstems that included cardio-respiratory control regions and counted the cells labeled for pro-inflammatory cytokines (interleukin (IL)-1β, IL-6, tumor necrosis factor-α) and high mobility group box 1.ResultsNeither pup viability nor number of cells labeled for pro-inflammatory cytokines in nucleus tractus solitarii or hypoglossal motor nucleus was impaired by VNS. We provide evidence suggesting that chronic VNS of pregnant mothers does not impede the progress or outcome of pregnancy.ConclusionVNS does not cause preterm birth, affect well-being of progeny, or impact central inflammatory processes that are critical for normal cardiovascular and respiratory function in newborns.
Collapse
|
26
|
Microglia and Neonatal Brain Injury. Neuroscience 2018; 405:68-76. [PMID: 29352997 DOI: 10.1016/j.neuroscience.2018.01.023] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 12/21/2017] [Accepted: 01/09/2018] [Indexed: 12/16/2022]
Abstract
Microglial cells are now recognized as the "gate-keepers" of healthy brain microenvironment with their disrupted functions adversely affecting neurovascular integrity, neuronal homeostasis, and network connectivity. The perception that these cells are purely toxic under neurodegenerative conditions has been challenged by a continuously increasing understanding of their complexity, the existence of a broad array of microglial phenotypes, and their ability to rapidly change in a context-dependent manner to attenuate or exacerbate injuries of different nature. Recent studies have demonstrated that microglial cells exert crucial physiological functions during embryonic and postnatal brain development, some of these functions being unique to particular stages of development, and extending far beyond sensing dangerous signals and serving as antigen presenting cells. In this focused review we cover the roles of microglial cells in regulating embryonic vasculogenesis, neurogenesis, and establishing network connectivity during postnatal brain development. We further discuss context-dependent microglial contribution to neonatal brain injuries associated with prenatal and postnatal infection and inflammation, in relation to neurodevelopmental disorders, as well as perinatal hypoxia-ischemia and arterial focal stroke. We also emphasize microglial phenotypic diversity, notably at the ultrastructural level, and their sex-dependent influence on the pathophysiology of neurodevelopmental disorders.
Collapse
|
27
|
Stojanovska V, Atik A, Nitsos I, Skiöld B, Barton SK, Zahra VA, Rodgers K, Hooper SB, Polglase GR, Galinsky R. Effects of Intrauterine Inflammation on Cortical Gray Matter of Near-Term Lambs. Front Pediatr 2018; 6:145. [PMID: 29963540 PMCID: PMC6013568 DOI: 10.3389/fped.2018.00145] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 05/01/2018] [Indexed: 12/25/2022] Open
Abstract
Introduction: Ventilation causes cerebral white matter inflammation and injury, which is exacerbated by intrauterine inflammation. However, the effects on cortical gray matter are not well-known. Our aim was to examine the effect of ventilation on the cerebral cortex of near-term lambs exposed to intrauterine inflammation. Method:Pregnant ewes at 119 ± 1 days gestation received an intra-amniotic injection of saline or lipopolysaccharide (LPS; 10 mg). Seven days later, lambs were randomized to either a high tidal volume injurious ventilation strategy (INJSALN = 6, INJLPSN = 5) or a protective ventilation strategy (PROTSALN = 5, PROTLPSN = 6). Respiratory parameters, heart rate and blood gases were monitored during the neonatal period. At post-mortem, the brain was collected and processed for immunohistochemical assessment. Neuronal density (NeuN), apoptotic cell death (caspase 8 and TUNEL), microglial density (Iba-1), astrocytic density (GFAP), and vascular protein extravasation (sheep serum) were assessed within the frontal, parietal, temporal and occipital lobes of the cerebral cortex. Results:A significant reduction in the number of neurons in all cortical layers except 4 was observed in LPS-exposed lambs compared to controls (layer #1: p = 0.041; layers #2 + 3: p = 0.023; layers #5 + 6: p = 0.016). LPS treatment caused a significant increase in gray matter area, indicative of edema. LPS+ventilation did not cause apoptotic cell death in the gray matter. Astrogliosis was not observed following PROT or INJ ventilation, with or without LPS exposure. LPS exposure was associated with vascular protein extravasation. Conclusion:Ventilation had little effect on gray matter inflammation and injury. Intrauterine inflammation reduced neuronal cell density, caused edema of the cortical gray matter, and blood vessel extravasation in the brain of near-term lambs.
Collapse
Affiliation(s)
- Vanesa Stojanovska
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia
| | - Anzari Atik
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia
| | - Ilias Nitsos
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia
| | - Béatrice Skiöld
- Department of Women's and Children's Health, Karolinska Institute, Stockholm, Sweden
| | - Samantha K Barton
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia.,Centre of Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Valerie A Zahra
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia
| | - Karyn Rodgers
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia
| | - Stuart B Hooper
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia.,Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC, Australia
| | - Graeme R Polglase
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia.,Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC, Australia
| | - Robert Galinsky
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia.,Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC, Australia
| |
Collapse
|
28
|
Girardi G. Complement activation, a threat to pregnancy. Semin Immunopathol 2017; 40:103-111. [PMID: 28900713 DOI: 10.1007/s00281-017-0645-x] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 08/03/2017] [Indexed: 12/11/2022]
Abstract
Pregnancy poses a challenge for the immune systems of placental mammals. As fetal tissues are semi-allogeneic and alloantibodies that commonly develop in the mother, the fetus and the placenta might be subject to complement-mediated immune attack with the potential risk of adverse pregnancy outcomes. Here, I describe how the use of animal models was pivotal in demonstrating that complement inhibition at the fetomaternal interface is essential for a successful pregnancy. Studies in animals also helped the identification of uncontrolled complement activation as a crucial effector in the pathogenesis of recurrent miscarriages, intrauterine growth restriction, preeclampsia, and preterm birth. Clinical studies employing complement biomarkers in plasma and urine showed an association between dysregulation of the complement system and adverse pregnancy outcomes. A better understanding of the role of the complement system in pregnancy complications will allow a rational approach to manipulate its activation as a potential therapeutic strategy with the goal of protecting pregnancies and improving long-term outcomes for mother and child.
Collapse
Affiliation(s)
- Guillermina Girardi
- Pregnancy Laboratory, Department of Women and Children's Health, The Rayne Institute, St Thomas' Hospital, King's College London, London, SE1 7EH, UK.
| |
Collapse
|
29
|
Lee YA. White Matter Injury of Prematurity: Its Mechanisms and Clinical Features. J Pathol Transl Med 2017; 51:449-455. [PMID: 28797157 PMCID: PMC5611534 DOI: 10.4132/jptm.2017.07.25] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 07/19/2017] [Accepted: 07/25/2017] [Indexed: 11/17/2022] Open
Abstract
A developing central nervous system is vulnerable to various insults such as infection and ischemia. While increased understanding of the dynamic nature of brain development allows a deeper insight into the pathophysiology of perinatal brain injury, the precise nature of specific fetal and neonatal brain injuries and their short- and long-term clinical consequences need special attention and further elucidation. The current review will describe the pathophysiological aspects and clinical significance of white matter injury of prematurity, a main form of perinatal brain injury in premature newborns, with a particular emphasis on its potential antenatal components.
Collapse
Affiliation(s)
- Young Ah Lee
- Division of Pediatric Neurology, Department of Pediatrics, Beaumont Hospital, Oakland University School of Medicine, Royal Oak, MI, USA
| |
Collapse
|
30
|
McDougall ARA, Hale N, Rees S, Harding R, De Matteo R, Hooper SB, Tolcos M. Erythropoietin Protects Against Lipopolysaccharide-Induced Microgliosis and Abnormal Granule Cell Development in the Ovine Fetal Cerebellum. Front Cell Neurosci 2017; 11:224. [PMID: 28804448 PMCID: PMC5532439 DOI: 10.3389/fncel.2017.00224] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2017] [Accepted: 07/13/2017] [Indexed: 11/13/2022] Open
Abstract
Erythropoietin (EPO) ameliorates inflammation-induced injury in cerebral white matter (WM). However, effects of inflammation on the cerebellum and neuroprotective effects of EPO are unknown. Our aims were to determine: (i) whether lipopolysaccharide (LPS)-induced intrauterine inflammation causes injury to, and/or impairs development of the cerebellum; and (ii) whether recombinant human EPO (rhEPO) mitigates these changes. At 107 ± 1 days gestational age (DGA; ~0.7 of term), fetal sheep received LPS (~0.9 μg/kg; i.v.) or an equivalent volume of saline, followed 1 h later with 5000 IU/kg rhEPO (i.v.) or an equivalent volume of saline (i.v.). This generated the following experimental groups: control (saline + saline; n = 6), LPS (LPS + saline, n = 8) and LPS + rhEPO (n = 8). At necropsy (116 ± 1 DGA; ~0.8 of term) the brain was perfusion-fixed and stained histologically (H&E) and immunostained to identify granule cells (Neuronal Nuclei, NeuN), granule cell proliferation (Ki67), Bergmann glia (glial fibrillary acidic protein, GFAP), astrogliosis (GFAP) and microgliosis (Iba-1). In comparison to controls, LPS fetuses had an increased density of Iba-1-positive microglia (p < 0.005) in the lobular WM; rhEPO prevented this increase (p < 0.05). The thickness of both the proliferative (Ki67-positive) and post-mitotic zones (Ki67-negative) of the EGL were increased in LPS-exposed fetuses compared to controls (p < 0.05), but were not different between controls and LPS + rhEPO fetuses. LPS also increased (p < 0.001) the density of granule cells (NeuN-positive) in the internal granule layer (IGL); rhEPO prevented the increase (p < 0.01). There was no difference between groups in the areas of the vermis (total cross-section), molecular layer (ML), IGL or WM, the density of NeuN-positive granule cells in the ML, the linear density of Bergmann glial fibers, the areal density or somal area of the Purkinje cells, the areal coverage of GFAP-positive astrocytes in the lobular and deep WM, the density of Iba-1-positive microglia in the deep WM or the density of apopotic cells in the cerebellum. LPS-induced intrauterine inflammation caused microgliosis and abnormal development of granule cells. rhEPO ameliorated these changes, suggesting that it is neuroprotective against LPS-induced inflammatory effects in the cerebellum.
Collapse
Affiliation(s)
- Annie R A McDougall
- The Ritchie Centre, Hudson Institute of Medical ResearchClayton, VIC, Australia.,Department of Obstetrics and Gynaecology, Monash UniversityClayton, VIC, Australia
| | - Nadia Hale
- The Ritchie Centre, Hudson Institute of Medical ResearchClayton, VIC, Australia
| | - Sandra Rees
- Department of Anatomy and Neuroscience, University of MelbourneParkville, VIC, Australia
| | - Richard Harding
- Department of Anatomy and Developmental Biology, Monash UniversityClayton, VIC, Australia
| | - Robert De Matteo
- Department of Anatomy and Developmental Biology, Monash UniversityClayton, VIC, Australia
| | - Stuart B Hooper
- The Ritchie Centre, Hudson Institute of Medical ResearchClayton, VIC, Australia.,Department of Obstetrics and Gynaecology, Monash UniversityClayton, VIC, Australia
| | - Mary Tolcos
- School of Health and Biomedical Sciences, RMIT UniversityMelbourne, VIC, Australia
| |
Collapse
|
31
|
Pascual M, Montesinos J, Montagud-Romero S, Forteza J, Rodríguez-Arias M, Miñarro J, Guerri C. TLR4 response mediates ethanol-induced neurodevelopment alterations in a model of fetal alcohol spectrum disorders. J Neuroinflammation 2017; 14:145. [PMID: 28738878 PMCID: PMC5525270 DOI: 10.1186/s12974-017-0918-2] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 07/12/2017] [Indexed: 01/15/2023] Open
Abstract
Background Inflammation during brain development participates in the pathogenesis of early brain injury and cognitive dysfunctions. Prenatal ethanol exposure affects the developing brain and causes neural impairment, cognitive and behavioral effects, collectively known as fetal alcohol spectrum disorders (FASD). Our previous studies demonstrate that ethanol activates the innate immune response and TLR4 receptor and causes neuroinflammation, brain damage, and cognitive defects in the developmental brain stage of adolescents. We hypothesize that by activating the TLR4 response, maternal alcohol consumption during pregnancy triggers the release of cytokines and chemokines in both the maternal sera and brains of fetuses/offspring, which impairs brain ontogeny and causes cognitive dysfunction. Methods WT and TLR4-KO female mice treated with or without 10% ethanol in the drinking water during gestation and lactation were used. Cytokine/chemokine levels were determined by ELISA in the amniotic fluid, maternal serum, and cerebral cortex, as well as in the offspring cerebral cortex. Microglial and neuronal markers (evaluated by western blotting), myelin proteins (immunohistochemical and western blotting) and synaptic parameters (western blotting and electron microscopy) were assessed in the cortices of the WT and TLR4-KO pups on PND 0, 20, and 66. Behavioral tests (elevated plus maze and passive avoidance) were performed in the WT and TLR4-KO mice on PND 66 exposed or not to ethanol. Results We show that alcohol intake during gestation and lactation increases the levels of several cytokines/chemokines (IL-1β, IL-17, MIP-1α, and fractalkine) in the maternal sera, amniotic fluid, and brains of fetuses and offspring. The upregulation of cytokines/chemokines is associated with an increase in activated microglia markers (CD11b and MHC-II), and with a reduction in some synaptic (synaptotagmin, synapsin IIa) and myelin (MBP, PLP) proteins in the brains of offspring on days 0, 20, and 66 (long-term effects). These changes are associated with long-term behavioral impairments, in the 66-day-old alcohol-exposed pups. TLR4-deficient mice are protected against ethanol-induced cytokine/chemokine production in alcohol-treated dams and offspring, along with synaptic and myelin alterations, and the log-term behavioral dysfunction induced by ethanol in offspring. Conclusions These results suggest that the immune system activation, through the TLR4 response, might play an important role in the neurodevelopmental defects in FASD. Electronic supplementary material The online version of this article (doi:10.1186/s12974-017-0918-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- María Pascual
- Department of Molecular and Cellular Pathology of Alcohol, Principe Felipe Research Center, C/Eduardo Primo Yúfera 3, 46012, Valencia, Spain.,Red Temática de Investigación Cooperativa en Salud (RETICS-Trastornos Adictivos), Instituto de Salud Carlos III, MICINN and FEDER, Madrid, Spain.,Department of Physiology, School of Medicine, Universitat de Valencia, Valencia, Spain
| | - Jorge Montesinos
- Department of Molecular and Cellular Pathology of Alcohol, Principe Felipe Research Center, C/Eduardo Primo Yúfera 3, 46012, Valencia, Spain.,Red Temática de Investigación Cooperativa en Salud (RETICS-Trastornos Adictivos), Instituto de Salud Carlos III, MICINN and FEDER, Madrid, Spain
| | - Sandra Montagud-Romero
- Department of Psychobiology, Facultad de Psicología, Universitat de Valencia, Valencia, Spain
| | - Jerónimo Forteza
- Instituto Valenciano de Patología, Unidad Mixta de Patología Molecular, Principe Felipe Research Center, Valencia, Spain
| | - Marta Rodríguez-Arias
- Department of Psychobiology, Facultad de Psicología, Universitat de Valencia, Valencia, Spain.,Red Temática de Investigación Cooperativa en Salud (RETICS-Trastornos Adictivos), Instituto de Salud Carlos III, MICINN and FEDER, Madrid, Spain
| | - José Miñarro
- Department of Psychobiology, Facultad de Psicología, Universitat de Valencia, Valencia, Spain.,Red Temática de Investigación Cooperativa en Salud (RETICS-Trastornos Adictivos), Instituto de Salud Carlos III, MICINN and FEDER, Madrid, Spain
| | - Consuelo Guerri
- Department of Molecular and Cellular Pathology of Alcohol, Principe Felipe Research Center, C/Eduardo Primo Yúfera 3, 46012, Valencia, Spain. .,Red Temática de Investigación Cooperativa en Salud (RETICS-Trastornos Adictivos), Instituto de Salud Carlos III, MICINN and FEDER, Madrid, Spain.
| |
Collapse
|
32
|
Moss TJM, Westover AJ. Inflammation-induced preterm lung maturation: lessons from animal experimentation. Paediatr Respir Rev 2017; 23:72-77. [PMID: 27856214 DOI: 10.1016/j.prrv.2016.10.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2015] [Revised: 07/08/2016] [Accepted: 10/05/2016] [Indexed: 02/05/2023]
Abstract
Intrauterine inflammation, or chorioamnionitis, is a major contributor to preterm birth. Prematurity per se is associated with considerable morbidity and mortality resulting from lung immaturity but exposure to chorioamnionitis reduces the risk of neonatal respiratory distress syndrome (RDS) in preterm infants. Animal experiments have identified that an increase in pulmonary surfactant production by the preterm lungs likely underlies this decreased risk of RDS in infants exposed to chorioamnionitis. Further animal experimentation has shown that infectious or inflammatory agents in amniotic fluid exert their effects on lung development by direct effects within the developing respiratory tract, and probably not by systemic pathways. Differences in the effects of intrauterine inflammation and glucocorticoids demonstrate that canonical glucocorticoid-mediated lung maturation is not responsible for inflammation-induced changes in lung development. Animal experimentation is identifying alternative lung maturational pathways, and transgenic animals and cell culture techniques will allow identification of novel mechanisms of lung maturation that may lead to new treatments for the prevention of RDS.
Collapse
Affiliation(s)
- Timothy J M Moss
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia; Department of Obstetrics and Gynecology, Monash University, Clayton, VIC 3168, Australia.
| | - Alana J Westover
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia
| |
Collapse
|
33
|
Millar LJ, Shi L, Hoerder-Suabedissen A, Molnár Z. Neonatal Hypoxia Ischaemia: Mechanisms, Models, and Therapeutic Challenges. Front Cell Neurosci 2017; 11:78. [PMID: 28533743 PMCID: PMC5420571 DOI: 10.3389/fncel.2017.00078] [Citation(s) in RCA: 231] [Impact Index Per Article: 28.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Accepted: 03/07/2017] [Indexed: 12/11/2022] Open
Abstract
Neonatal hypoxia-ischaemia (HI) is the most common cause of death and disability in human neonates, and is often associated with persistent motor, sensory, and cognitive impairment. Improved intensive care technology has increased survival without preventing neurological disorder, increasing morbidity throughout the adult population. Early preventative or neuroprotective interventions have the potential to rescue brain development in neonates, yet only one therapeutic intervention is currently licensed for use in developed countries. Recent investigations of the transient cortical layer known as subplate, especially regarding subplate's secretory role, opens up a novel set of potential molecular modulators of neonatal HI injury. This review examines the biological mechanisms of human neonatal HI, discusses evidence for the relevance of subplate-secreted molecules to this condition, and evaluates available animal models. Neuroserpin, a neuronally released neuroprotective factor, is discussed as a case study for developing new potential pharmacological interventions for use post-ischaemic injury.
Collapse
Affiliation(s)
- Lancelot J. Millar
- Molnár Group, Department of Physiology, Anatomy and Genetics, University of OxfordOxford, UK
| | - Lei Shi
- Molnár Group, Department of Physiology, Anatomy and Genetics, University of OxfordOxford, UK
- JNU-HKUST Joint Laboratory for Neuroscience and Innovative Drug Research, College of Pharmacy, Jinan UniversityGuangzhou, China
| | | | - Zoltán Molnár
- Molnár Group, Department of Physiology, Anatomy and Genetics, University of OxfordOxford, UK
| |
Collapse
|
34
|
Paton MCB, McDonald CA, Allison BJ, Fahey MC, Jenkin G, Miller SL. Perinatal Brain Injury As a Consequence of Preterm Birth and Intrauterine Inflammation: Designing Targeted Stem Cell Therapies. Front Neurosci 2017; 11:200. [PMID: 28442989 PMCID: PMC5385368 DOI: 10.3389/fnins.2017.00200] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Accepted: 03/24/2017] [Indexed: 12/20/2022] Open
Abstract
Chorioamnionitis is a major cause of preterm birth and brain injury. Bacterial invasion of the chorion and amnion, and/or the placenta, can lead to a fetal inflammatory response, which in turn has significant adverse consequences for the developing fetal brain. Accordingly, there is a strong causal link between chorioamnionitis, preterm brain injury and the pathogenesis of severe postnatal neurological deficits and cerebral palsy. Currently there are no treatments to protect or repair against brain injury in preterm infants born after pregnancy compromised by intrauterine infection. This review describes the injurious cascade of events in the preterm brain in response to a severe fetal inflammatory event. We will highlight specific periods of increased vulnerability, and the potential effects of therapeutic intervention with cell-based therapies. Many clinical trials are underway to investigate the efficacy of stem cells to treat patients with cerebral palsy. Stem cells, obtained from umbilical cord tissue and cord blood, normally discarded after birth, are emerging as a safe and potentially effective therapy. It is not yet known, however, which stem cell type(s) are the most efficacious for administration to preterm infants to treat brain injury-mediated inflammation. Individual stem cell populations found in cord blood and tissue, such as mesenchymal stem cells (MSCs) and endothelial progenitor cells (EPCs), have a number of potential benefits that may specifically target preterm inflammatory-induced brain injury. MSCs have strong immunomodulatory potential, protecting against global and local neuroinflammatory cascades triggered during infection to the fetus. EPCs have angiogenic and vascular reparative qualities that make them ideal for neurovascular repair. A combined therapy using both MSCs and EPCs to target inflammation and promote angiogenesis for re-establishment of vital vessel networks is a treatment concept that warrants further investigation.
Collapse
Affiliation(s)
- Madison C B Paton
- Neurodevelopment and Neuroprotection Research Group, The Ritchie Centre, Hudson Institute of Medical Research, Monash UniversityClayton, VIC, Australia.,Department of Obstetrics and Gynaecology, Monash Medical Centre, Monash UniversityClayton, VIC, Australia
| | - Courtney A McDonald
- Neurodevelopment and Neuroprotection Research Group, The Ritchie Centre, Hudson Institute of Medical Research, Monash UniversityClayton, VIC, Australia
| | - Beth J Allison
- Neurodevelopment and Neuroprotection Research Group, The Ritchie Centre, Hudson Institute of Medical Research, Monash UniversityClayton, VIC, Australia
| | - Michael C Fahey
- Neurodevelopment and Neuroprotection Research Group, The Ritchie Centre, Hudson Institute of Medical Research, Monash UniversityClayton, VIC, Australia.,Department of Paediatrics, Monash UniversityClayton, VIC, Australia
| | - Graham Jenkin
- Neurodevelopment and Neuroprotection Research Group, The Ritchie Centre, Hudson Institute of Medical Research, Monash UniversityClayton, VIC, Australia.,Department of Obstetrics and Gynaecology, Monash Medical Centre, Monash UniversityClayton, VIC, Australia
| | - Suzanne L Miller
- Neurodevelopment and Neuroprotection Research Group, The Ritchie Centre, Hudson Institute of Medical Research, Monash UniversityClayton, VIC, Australia.,Department of Obstetrics and Gynaecology, Monash Medical Centre, Monash UniversityClayton, VIC, Australia
| |
Collapse
|
35
|
Huang L, Zhao F, Qu Y, Zhang L, Wang Y, Mu D. Animal models of hypoxic-ischemic encephalopathy: optimal choices for the best outcomes. Rev Neurosci 2017; 28:31-43. [PMID: 27559689 DOI: 10.1515/revneuro-2016-0022] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Accepted: 07/15/2016] [Indexed: 12/21/2022]
Abstract
AbstractHypoxic-ischemic encephalopathy (HIE), a serious disease leading to neonatal death, is becoming a key area of pediatric neurological research. Despite remarkable advances in the understanding of HIE, the explicit pathogenesis of HIE is unclear, and well-established treatments are absent. Animal models are usually considered as the first step in the exploration of the underlying disease and in evaluating promising therapeutic interventions. Various animal models of HIE have been developed with distinct characteristics, and it is important to choose an appropriate animal model according to the experimental objectives. Generally, small animal models may be more suitable for exploring the mechanisms of HIE, whereas large animal models are better for translational studies. This review focuses on the features of commonly used HIE animal models with respect to their modeling strategies, merits, and shortcomings, and associated neuropathological changes, providing a comprehensive reference for improving existing animal models and developing new animal models.
Collapse
Affiliation(s)
- Lan Huang
- 1Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China
- 2Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects of the Ministry of Education, Sichuan University, Chengdu 610041, China
| | - Fengyan Zhao
- 1Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China
- 2Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects of the Ministry of Education, Sichuan University, Chengdu 610041, China
| | - Yi Qu
- 1Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China
- 2Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects of the Ministry of Education, Sichuan University, Chengdu 610041, China
| | - Li Zhang
- 1Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China
- 2Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects of the Ministry of Education, Sichuan University, Chengdu 610041, China
| | - Yan Wang
- 1Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China
- 2Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects of the Ministry of Education, Sichuan University, Chengdu 610041, China
| | - Dezhi Mu
- 1Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China
- 2Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects of the Ministry of Education, Sichuan University, Chengdu 610041, China
- 3Department of Pediatrics, University of California, San Francisco, CA 94143, USA
| |
Collapse
|
36
|
Alahmari DM, Skiöld B, Barton SK, Nitsos I, McDonald C, Miller SL, Zahra V, Galinsky R, Wu Q, Farrell MJ, Moss TJ, Hooper SB, Pearson JT, Polglase GR. Diffusion Tensor Imaging Colour Mapping Threshold for Identification of Ventilation-Induced Brain Injury after Intrauterine Inflammation in Preterm Lambs. Front Pediatr 2017; 5:70. [PMID: 28424764 PMCID: PMC5380678 DOI: 10.3389/fped.2017.00070] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Accepted: 03/20/2017] [Indexed: 11/23/2022] Open
Abstract
PURPOSE The aim of this study is to examine whether advanced magnetic resonance imaging (MRI) techniques can detect early brain injury caused by intrauterine inflammation and inappropriate initial respiratory support in preterm lambs. HYPOTHESIS Neuropathology caused by intrauterine inflammation is exacerbated by mechanical ventilation at birth and is detectable with advanced MRI techniques. METHODS Pregnant ewes received intra-amniotic lipopolysaccharide (LPS) 7 days prior to delivery at ~125 days of gestation (85% of gestation), whereupon lambs were delivered and randomised to receive an injurious (LPS + INJ, n = 6) or protective (LPS + PROT, n = 6) ventilation strategy. MRI of the brain was conducted 90 min after preterm delivery, using structural, diffusion tensor imaging (DTI), and magnetic resonance spectroscopy (MRS) techniques. A colour map threshold technique was utilised to compare distributions of low diffusivity voxels in the brains of LPS-exposed lambs with those not exposed to LPS (PROT, n = 7 PROT and INJ, n = 10). RESULTS No overt cerebral injury was identified on structural MRI images of any lamb. However, on DTI, axial diffusivity, radial diffusivity, and mean diffusivity values were lower and significantly more heterogeneous in specific brain regions of lambs in the LPS + INJ group compared to the LPS + PROT group. Colour mapping revealed lower diffusivity in the thalamus, periventricular white matter, internal capsule, and frontal white matter in the LPS + INJ group compared to LPS + PROT group. The MRS peak area ratios of lactate, relative to those for the metabolites creatine, choline, and N-acetylaspartate, were not different between LPS-exposed groups. Lambs exposed to LPS had lower diffusivity within the white matter regions assessed than non-LPS-treated control lambs. CONCLUSION DTI colour map threshold techniques detected early brain injury in preterm lambs exposed to intrauterine inflammation and detected differences between injurious and protective ventilation strategies. DTI mapping approaches are potentially useful for early detection of subtle brain injury in premature infants.
Collapse
Affiliation(s)
- Dhafer M Alahmari
- Department of Medical Imaging and Radiation Sciences, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia.,Monash Biomedical Imaging, Monash University, Clayton, VIC, Australia
| | - Beatrice Skiöld
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Samantha K Barton
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia
| | - Ilias Nitsos
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia
| | - Courtney McDonald
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia
| | - Suzanne L Miller
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia.,Department of Obstetrics and Gynaecology and Paediatrics, Monash University, Clayton, VIC, Australia
| | - Valerie Zahra
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia
| | - Robert Galinsky
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia.,Department of Physiology, University of Auckland, Grafton, New Zealand
| | - Qizhu Wu
- Monash Biomedical Imaging, Monash University, Clayton, VIC, Australia
| | - Michael John Farrell
- Department of Medical Imaging and Radiation Sciences, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia.,Monash Biomedical Imaging, Monash University, Clayton, VIC, Australia
| | - Timothy J Moss
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia.,Department of Obstetrics and Gynaecology and Paediatrics, Monash University, Clayton, VIC, Australia
| | - Stuart B Hooper
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia.,Department of Obstetrics and Gynaecology and Paediatrics, Monash University, Clayton, VIC, Australia
| | - James T Pearson
- Monash Biomedical Imaging, Monash University, Clayton, VIC, Australia.,Department of Physiology, Monash University, Clayton, VIC, Australia.,Department of Cardiac Physiology, National Cerebral and Cardiovascular Center, Suita, Osaka, Japan
| | - Graeme R Polglase
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia.,Department of Obstetrics and Gynaecology and Paediatrics, Monash University, Clayton, VIC, Australia
| |
Collapse
|
37
|
Nitsos I, Rees SM, Duncan J, Kramer BW, Harding R, Newnham JP, Moss TJM. Chronic Exposure to Intra-Amniotic Lipopolysaccharide Affects the Ovine Fetal Brain. ACTA ACUST UNITED AC 2016; 13:239-47. [PMID: 16697939 DOI: 10.1016/j.jsgi.2006.02.011] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2005] [Indexed: 11/23/2022]
Abstract
OBJECTIVE Fetal brain injury is associated with chorioamnionitis, which is often present without signs of overt infection or fetal compromise. We aimed to determine if prolonged exposure to intrauterine inflammation caused by intra-amniotic infusion of lipopolysaccharide (LPS) would affect the fetal brain. METHODS At 80 days of pregnancy ewes bearing singletons had osmotic pumps implanted intra-amniotically to infuse Escherichia coli LPS (055:B5; n = 8) or saline (n = 7) for 28 days. At delivery (110 days), umbilical arterial blood and chorioamnion were assessed for inflammation; cytokine concentrations (interleukin [IL]-6 and IL-8) in amniotic fluid and fetal and maternal plasma were measured. The fetal cerebral hemispheres were examined for gross anatomical changes and the number of activated microglia/macrophages, astrocytes, and oligodendrocytes estimated after immunohistochemical staining. RESULTS Intra-amniotic administration of LPS caused chorioamnionitis, fetal leucocytosis, and a moderate to extensive infiltration of activated microglia/macrophages in the subcortical white matter in six of eight fetuses; the remaining two fetuses were less affected. Within these focal regions of damage there was an attenuation of astrocytic processes, axonal injury, and a reduction in the number of 2',3'-cyclic nucleotide 3'-phosphodiesterase (CNPase) immunoreactive oligodendrocytes in areas of extensive focal damage. In control fetuses there was mild (3/7) or no infiltration of activated microglia/macrophages in the subcortical white matter. Overall the infiltration of activated microglia/macrophages in the white matter was significantly greater in LPS-exposed fetuses compared to controls. In regions devoid of injury, the number of oligodendrocytes and astrocytes was not different between groups, nor was there a difference in the volume of cerebral white matter or density of blood vessels within the white matter. Amniotic fluid IL-6 and IL-8, and maternal plasma IL-8 concentrations were significantly increased by LPS infusion. CONCLUSIONS An increase in inflammatory cells and axonal disruption in the subcortical white matter of the fetal brain can accompany chorioamnionitis induced by intra-amniotic administration of LPS, but cystic lesions do not occur. Thus, the effect on the fetal brain is milder than that reported from animal models of acute fetal/intrauterine infection.
Collapse
Affiliation(s)
- Ilias Nitsos
- School of Women's and Infants' Health, The University of Western Australia, Crawley, Western Australia, Australia.
| | | | | | | | | | | | | |
Collapse
|
38
|
Garnier Y, Coumans ABC, Jensen A, Hasaart THM, Berger R. Infection-Related Perinatal Brain Injury: The Pathogenic Role of Impaired Fetal Cardiovascular Control. ACTA ACUST UNITED AC 2016. [DOI: 10.1016/s1071-55760300150-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
| | | | | | - Tom H. M. Hasaart
- Department of Obstetrics and Gynecology, University of Bochum, Bochum, Germany; Department of obstetrics and Gynecology, University of Maastricht, Maastricht, The Netherlands
| | - Richard Berger
- Department of Obstetrics and Gynecology, University of Bochum, Bochum, Germany; Department of obstetrics and Gynecology, University of Maastricht, Maastricht, The Netherlands; Universitätsfrauenklinik Bochum, Knappschaftskrankenhaus, In der Schornau 23-25, 44982 Bochum
| |
Collapse
|
39
|
Coumans ABC, Garnier Y, Supçun S, Jensen A, Berger R, Hasaart THM. The Effects of Low-Dose Endotoxin on the Umbilicoplacental Circulation in Preterm Sheep. ACTA ACUST UNITED AC 2016; 11:289-93. [PMID: 15219882 DOI: 10.1016/j.jsgi.2003.12.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
OBJECTIVE In the present study we examined the effects of low-dose endotoxin (lipopolysaccharides, LPS) on continuously recorded umbilical blood flow. METHODS Twenty fetal sheep were catheterized at a gestational age of 107 +/- 1 days. A flow probe was placed around either the common umbilical artery or one single umbilical artery. Three days later fetuses received either 100 or 500 nanograms of LPS (n = 14) or 2 mL saline (n = 6) intravenously. Six fetuses died within 12 hours after LPS. Fetal heart rate (FHR), mean arterial pressure (MAP), and umbilical blood flow (Q(umb)) were monitored for 3 days. RESULTS FHR increased by 25 +/- 4% at 4-5 hours after LPS (P <.01) and was elevated for 15 hours after LPS. MAP increased by 18 +/- 5% 1 hour after LPS (P <.01) and returned to control value 4-5 hours after LPS. Q(umb) began to decrease 1 hour after LPS and was minimal (-30 +/- 7%, P <.001) at 4-5 hours after LPS. Q(umb) slowly returned to the control value at 12 hours after LPS. Placental vascular resistance increased by 73 +/- 37% (P <.01), whereas pH did not appreciably change. CONCLUSION Intravenous application of endotoxin caused a substantial and long-lasting decrease in umbilical blood flow resulting in fetal hypoxemia without acidemia. These effects may be of significance in the development of fetal brain damage associated with intrauterine infection.
Collapse
Affiliation(s)
- A B C Coumans
- Department of Obstetrics and Gynecology, University of Maastricht, Maastricht, The Netherlands
| | | | | | | | | | | |
Collapse
|
40
|
Labus KM, Puttlitz CM. An anisotropic hyperelastic constitutive model of brain white matter in biaxial tension and structural-mechanical relationships. J Mech Behav Biomed Mater 2016; 62:195-208. [PMID: 27214689 DOI: 10.1016/j.jmbbm.2016.05.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Revised: 04/26/2016] [Accepted: 05/03/2016] [Indexed: 10/21/2022]
Abstract
Computational models of the brain require accurate and robust constitutive models to characterize the mechanical behavior of brain tissue. The anisotropy of white matter has been previously demonstrated; however, there is a lack of data describing the effects of multi-axial loading, even though brain tissue experiences multi-axial stress states. Therefore, a biaxial tensile experiment was designed to more fully characterize the anisotropic behavior of white matter in a quasi-static loading state, and the mechanical data were modeled with an anisotropic hyperelastic continuum model. A probabilistic analysis was used to quantify the uncertainty in model predictions because the mechanical data of brain tissue can show a high degree of variability, and computational studies can benefit from reporting the probability distribution of model responses. The axonal structure in white matter can be heterogeneous and regionally dependent, which can affect computational model predictions. Therefore, corona radiata and corpus callosum regions were tested, and histology and transmission electron microscopy were performed on tested specimens to relate the distribution of axon orientations and the axon volume fraction to the mechanical behavior. These measured properties were implemented into a structural constitutive model. Results demonstrated a significant, but relatively low anisotropic behavior, yet there were no conclusive mechanical differences between the two regions tested. The inclusion of both biaxial and uniaxial tests in model fits improved the accuracy of model predictions. The mechanical anisotropy of individual specimens positively correlated with the measured axon volume fraction, and, accordingly, the structural model exhibited slightly decreased uncertainty in model predictions compared to the model without structural properties.
Collapse
Affiliation(s)
- Kevin M Labus
- School of Biomedical Engineering, Colorado State University, Fort Collins, CO, USA
| | - Christian M Puttlitz
- School of Biomedical Engineering, Colorado State University, Fort Collins, CO, USA; Department of Mechanical Engineering, Colorado State University, Fort Collins, CO, USA; Department of Clinical Sciences, Colorado State University, Fort Collins, CO, USA.
| |
Collapse
|
41
|
Jenkins DD, Wiest DB, Mulvihill DM, Hlavacek AM, Majstoravich SJ, Brown TR, Taylor JJ, Buckley JR, Turner RP, Rollins LG, Bentzley JP, Hope KE, Barbour AB, Lowe DW, Martin RH, Chang EY. Fetal and Neonatal Effects of N-Acetylcysteine When Used for Neuroprotection in Maternal Chorioamnionitis. J Pediatr 2016; 168:67-76.e6. [PMID: 26545726 PMCID: PMC4698030 DOI: 10.1016/j.jpeds.2015.09.076] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Revised: 08/25/2015] [Accepted: 09/29/2015] [Indexed: 01/24/2023]
Abstract
OBJECTIVE To evaluate the clinical safety of antenatal and postnatal N-acetylcysteine (NAC) as a neuroprotective agent in maternal chorioamnionitis in a randomized, controlled, double-blinded trial. STUDY DESIGN Twenty-two mothers >24 weeks gestation presenting within 4 hours of diagnosis of clinical chorioamnionitis were randomized with their 24 infants to NAC or saline treatment. Antenatal NAC (100 mg/kg/dose) or saline was given intravenously every 6 hours until delivery. Postnatally, NAC (12.5-25 mg/kg/dose, n = 12) or saline (n = 12) was given every 12 hours for 5 doses. Doppler studies of fetal umbilical and fetal and infant cerebral blood flow, cranial ultrasounds, echocardiograms, cerebral oxygenation, electroencephalograms, and serum cytokines were evaluated before and after treatment, and 12, 24, and 48 hours after birth. Magnetic resonance spectroscopy and diffusion imaging were performed at term age equivalent. Development was followed for cerebral palsy or autism to 4 years of age. RESULTS Cardiovascular measures, cerebral blood flow velocity and vascular resistance, and cerebral oxygenation did not differ between treatment groups. Cerebrovascular coupling was disrupted in infants with chorioamnionitis treated with saline but preserved in infants treated with NAC, suggesting improved vascular regulation in the presence of neuroinflammation. Infants treated with NAC had higher serum anti-inflammatory interleukin-1 receptor antagonist and lower proinflammatory vascular endothelial growth factor over time vs controls. No adverse events related to NAC administration were noted. CONCLUSIONS In this cohort of newborns exposed to chorioamnionitis, antenatal and postnatal NAC was safe, preserved cerebrovascular regulation, and increased an anti-inflammatory neuroprotective protein. TRIAL REGISTRATION ClinicalTrials.gov: NCT00724594.
Collapse
Affiliation(s)
- Dorothea D. Jenkins
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC
| | - Donald B. Wiest
- Department of Clinical Pharmacy and Outcome Science, Medical University of South Carolina, Charleston, SC
| | - Denise M. Mulvihill
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC
| | - Anthony M. Hlavacek
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC
| | | | - Truman R. Brown
- Department of Neuroscience’s Center for Advanced Imaging Research, Medical University of South Carolina, Charleston, SC
| | - Joseph J. Taylor
- Department of Neuroscience’s Center for Advanced Imaging Research, Medical University of South Carolina, Charleston, SC,Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC
| | - Jason R. Buckley
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC
| | - Robert P. Turner
- Department of Clinical Pediatrics and Neurology, University of South Carolina School of Medicine and Palmetto Health Richland Children’s Hospital, Columbia, SC
| | | | - Jessica P. Bentzley
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC
| | - Kathryn E. Hope
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC
| | - Andrew B. Barbour
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC
| | - Danielle W. Lowe
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC
| | - Renee H. Martin
- Department of Public Health Sciences, Medical University of South Carolina, Charleston, SC
| | - Eugene Y. Chang
- Department of Obstetrics and Gynecology, Medical University of South Carolina, Charleston, SC
| |
Collapse
|
42
|
Subclinical decelerations during developing hypotension in preterm fetal sheep after acute on chronic lipopolysaccharide exposure. Sci Rep 2015; 5:16201. [PMID: 26537688 PMCID: PMC4633652 DOI: 10.1038/srep16201] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Accepted: 10/12/2015] [Indexed: 12/16/2022] Open
Abstract
Subclinical (shallow) heart rate decelerations occur during neonatal sepsis, but there is limited information on their relationship with hypotension or whether they occur before birth. We examined whether subclinical decelerations, a fall in fetal heart rate (FHR) that remained above 100 bpm, were associated with hypotension in preterm fetal sheep exposed to lipopolysaccharide (LPS). Chronically-instrumented fetal sheep at 0.7 gestation received continuous low-dose LPS infusions (n = 15, 100 ng/kg over 24 h, followed by 250 ng/kg/24 h for 96 h) or saline (n = 8). Boluses of 1 μg LPS or saline were given at 48 and 72 h. FHR variability (FHRV) was calculated, and sample asymmetry was used to assess the severity and frequency of decelerations. Low-dose LPS infusion did not affect FHR. After the first LPS bolus, 7 fetuses remained normotensive, while 8 developed hypotension (a fall in mean arterial blood pressure of ≥5 mmHg). Developing hypotension was associated with subclinical decelerations, with a corresponding increase in sample asymmetry and FHRV (p < 0.05). The second LPS bolus was associated with similar but attenuated changes in FHR and blood pressure (p < 0.05). In conclusion, subclinical decelerations are not consistently seen during prenatal exposure to LPS, but may be a useful marker of developing inflammation-related hypotension before birth.
Collapse
|
43
|
Ahmad AS, Satriotomo I, Fazal J, Nadeau SE, Doré S. Considerations for the Optimization of Induced White Matter Injury Preclinical Models. Front Neurol 2015; 6:172. [PMID: 26322013 PMCID: PMC4532913 DOI: 10.3389/fneur.2015.00172] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Accepted: 07/20/2015] [Indexed: 11/13/2022] Open
Abstract
White matter (WM) injury in relation to acute neurologic conditions, especially stroke, has remained obscure until recently. Current advances in imaging technologies in the field of stroke have confirmed that WM injury plays an important role in the prognosis of stroke and suggest that WM protection is essential for functional recovery and post-stroke rehabilitation. However, due to the lack of a reproducible animal model of WM injury, the pathophysiology and mechanisms of this injury are not well studied. Moreover, producing selective WM injury in animals, especially in rodents, has proven to be challenging. Problems associated with inducing selective WM ischemic injury in the rodent derive from differences in the architecture of the brain, most particularly, the ratio of WM to gray matter in rodents compared to humans, the agents used to induce the injury, and the location of the injury. Aging, gender differences, and comorbidities further add to this complexity. This review provides a brief account of the techniques commonly used to induce general WM injury in animal models (stroke and non-stroke related) and highlights relevance, optimization issues, and translational potentials associated with this particular form of injury.
Collapse
Affiliation(s)
- Abdullah Shafique Ahmad
- Department of Anesthesiology, Center for Translational Research in Neurodegenerative Disease, University of Florida , Gainesville, FL , USA
| | - Irawan Satriotomo
- Department of Anesthesiology, Center for Translational Research in Neurodegenerative Disease, University of Florida , Gainesville, FL , USA
| | - Jawad Fazal
- Department of Anesthesiology, Center for Translational Research in Neurodegenerative Disease, University of Florida , Gainesville, FL , USA
| | - Stephen E Nadeau
- Research Service, Brain Rehabilitation Research Center, Malcom Randall Veterans Affairs Medical Center , Gainesville, FL , USA ; Department of Neurology, University of Florida , Gainesville, FL , USA
| | - Sylvain Doré
- Department of Anesthesiology, Center for Translational Research in Neurodegenerative Disease, University of Florida , Gainesville, FL , USA ; Research Service, Brain Rehabilitation Research Center, Malcom Randall Veterans Affairs Medical Center , Gainesville, FL , USA ; Department of Neurology, University of Florida , Gainesville, FL , USA ; Department of Neuroscience, University of Florida , Gainesville, FL , USA ; Department of Neurology, University of Florida , Gainesville, FL , USA ; Department of Pharmaceutics, University of Florida , Gainesville, FL , USA ; Department of Psychology, University of Florida , Gainesville, FL , USA ; Department of Psychiatry, University of Florida , Gainesville, FL , USA
| |
Collapse
|
44
|
Hagberg H, Mallard C, Ferriero DM, Vannucci SJ, Levison SW, Vexler ZS, Gressens P. The role of inflammation in perinatal brain injury. Nat Rev Neurol 2015; 11:192-208. [PMID: 25686754 PMCID: PMC4664161 DOI: 10.1038/nrneurol.2015.13] [Citation(s) in RCA: 597] [Impact Index Per Article: 59.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Inflammation is increasingly recognized as being a critical contributor to both normal development and injury outcome in the immature brain. The focus of this Review is to highlight important differences in innate and adaptive immunity in immature versus adult brain, which support the notion that the consequences of inflammation will be entirely different depending on context and stage of CNS development. Perinatal brain injury can result from neonatal encephalopathy and perinatal arterial ischaemic stroke, usually at term, but also in preterm infants. Inflammation occurs before, during and after brain injury at term, and modulates vulnerability to and development of brain injury. Preterm birth, on the other hand, is often a result of exposure to inflammation at a very early developmental phase, which affects the brain not only during fetal life, but also over a protracted period of postnatal life in a neonatal intensive care setting, influencing critical phases of myelination and cortical plasticity. Neuroinflammation during the perinatal period can increase the risk of neurological and neuropsychiatric disease throughout childhood and adulthood, and is, therefore, of concern to the broader group of physicians who care for these individuals.
Collapse
Affiliation(s)
- Henrik Hagberg
- 1] Centre for the Developing Brain, Division of Imaging Sciences and Biomedical Engineering, King's College London, King's Health Partners, St Thomas' Hospital, London SE1 7EH, UK. [2] Perinatal Center, Institute of Physiology and Neurosciences and Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, 435 43 Gothenburg, Sweden
| | - Carina Mallard
- Perinatal Center, Institute of Physiology and Neurosciences and Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, 435 43 Gothenburg, Sweden
| | - Donna M Ferriero
- Departments of Neurology and Pediatrics, University of California San Francisco, San Francisco, CA 94158, USA
| | - Susan J Vannucci
- Department of Pediatrics/Newborn Medicine, Weill Cornell Medical College, 1300 York Avenue, New York, NY 10065, USA
| | - Steven W Levison
- Department of Neurology and Neuroscience, Rutgers University, RBHS-New Jersey Medical School, Cancer Center, H-1226 205 South Orange Avenue, Newark, NJ 07103, USA
| | - Zinaida S Vexler
- Departments of Neurology and Pediatrics, University of California San Francisco, San Francisco, CA 94158, USA
| | | |
Collapse
|
45
|
Bonestroo HJC, Heijnen CJ, Groenendaal F, van Bel F, Nijboer CH. Development of cerebral gray and white matter injury and cerebral inflammation over time after inflammatory perinatal asphyxia. Dev Neurosci 2015; 37:78-94. [PMID: 25634435 DOI: 10.1159/000368770] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Accepted: 10/01/2014] [Indexed: 11/19/2022] Open
Abstract
Antenatal inflammation is associated with increased severity of hypoxic-ischemic (HI) encephalopathy and adverse outcome in human neonates and experimental rodents. We investigated the effect of lipopolysaccharide (LPS) on the timing of HI-induced cerebral tissue loss and gray matter injury, white matter injury and integrity, and the cerebral inflammatory response. On postnatal day 9, mice underwent HI by unilateral carotid artery occlusion followed by systemic hypoxia which resulted in early neuronal damage (MAP2 loss) at 3 h that did not increase up to day 15. LPS injection 14 h before HI (LPS+HI) significantly and gradually aggravated MAP2 loss from 3 h up to day 15, resulting in an acellular cystic lesion. LPS+HI increased white matter damage, reduced myelination in the corpus callosum and increased white matter fiber coherency in the cingulum. The number of oligodendrocytes throughout the lineage (Olig2-positive) was increased whereas more mature myelinating (CNPase-positive) oligodendrocytes were strongly decreased after LPS+HI. LPS+HI induced an increased and prolonged expression of cerebral cytokines/chemokines compared to HI. Additionally, LPS+HI increased macrophage/microglia activation and influx of neutrophils in the brain compared to HI. This study demonstrates the sensitizing effect of LPS on neonatal HI brain injury for an extended time-frame up to 15 days postinsult. LPS before HI induced a gradual increase in gray and white matter deficits, including reduced numbers of more mature myelinating oligodendrocytes and a decrease in white matter integrity. Moreover, LPS+HI prolonged and intensified the cerebral inflammatory response, including cellular infiltration. In conclusion, as the timing of damage and/or involved pathways are changed when HI is preceded by inflammation, experimental therapies might require modifications in the time window, dosage or combinations of therapies for efficacious neuroprotection.
Collapse
Affiliation(s)
- Hilde J C Bonestroo
- Laboratory of Neuroimmunology and Developmental Origins of Disease (NIDOD), University Medical Center Utrecht, Utrecht, The Netherlands
| | | | | | | | | |
Collapse
|
46
|
Pearson C, Littlewood E, Douglas P, Robertson S, Gant TW, Hansell AL. Exposures and health outcomes in relation to bioaerosol emissions from composting facilities: a systematic review of occupational and community studies. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART B, CRITICAL REVIEWS 2015; 18:43-69. [PMID: 25825807 PMCID: PMC4409048 DOI: 10.1080/10937404.2015.1009961] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
The number of composting sites in Europe is rapidly increasing, due to efforts to reduce the fraction of waste destined for landfill, but evidence on possible health impacts is limited. This article systematically reviews studies related to bioaerosol exposures within and near composting facilities and associated health effects in both community and occupational health settings. Six electronic databases and bibliographies from January 1960 to July 2014 were searched for studies reporting on health outcomes and/or bioaerosol emissions related to composting sites. Risk of bias was assessed using a customized score. Five hundred and thirty-six papers were identified and reviewed, and 66 articles met the inclusion criteria (48 exposure studies, 9 health studies, 9 health and exposure studies). Exposure information was limited, with most measurements taken in occupational settings and for limited time periods. Bioaerosol concentrations were highest on-site during agitation activities (turning, shredding, and screening). Six studies detected concentrations of either Aspergillus fumigatus or total bacteria above the English Environment Agency's recommended threshold levels beyond 250 m from the site. Occupational studies of compost workers suggested elevated risks of respiratory illnesses with higher bioaerosol exposures. Elevated airway irritation was reported in residents near composting sites, but this may have been affected by reporting bias. The evidence base on health effects of bioaerosol emissions from composting facilities is still limited, although there is sufficient evidence to support a precautionary approach for regulatory purposes. While data to date are suggestive of possible respiratory effects, further study is needed to confirm this and to explore other health outcomes.
Collapse
Affiliation(s)
- Clare Pearson
- Small Area Health Statistics Unit, MRC-PHE Centre for Environment and Health & NIHR HPRU in Health Impact of Environmental Hazards, Imperial College London, London, United Kingdom
| | - Emma Littlewood
- Small Area Health Statistics Unit, MRC-PHE Centre for Environment and Health & NIHR HPRU in Health Impact of Environmental Hazards, Imperial College London, London, United Kingdom
| | - Philippa Douglas
- Small Area Health Statistics Unit, MRC-PHE Centre for Environment and Health & NIHR HPRU in Health Impact of Environmental Hazards, Imperial College London, London, United Kingdom
| | - Sarah Robertson
- Centre for Radiation, Chemical and Environmental Hazards, Public Health England, Harwell Campus, Didcot, Oxfordshire, United Kingdom
| | - Timothy W. Gant
- Centre for Radiation, Chemical and Environmental Hazards, Public Health England, Harwell Campus, Didcot, Oxfordshire, United Kingdom
| | - Anna L. Hansell
- Small Area Health Statistics Unit, MRC-PHE Centre for Environment and Health & NIHR HPRU in Health Impact of Environmental Hazards, Imperial College London, London, United Kingdom
- Public Health and Primary Care, Imperial College Healthcare NHS Trust, London, United Kingdom
| |
Collapse
|
47
|
Back SA, Riddle A, Hohimer AR. The Sheep as a Model of Brain Injury in the Premature Infant. ANIMAL MODELS OF NEURODEVELOPMENTAL DISORDERS 2015. [DOI: 10.1007/978-1-4939-2709-8_8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
|
48
|
Kemp MW. Preterm birth, intrauterine infection, and fetal inflammation. Front Immunol 2014; 5:574. [PMID: 25520716 PMCID: PMC4249583 DOI: 10.3389/fimmu.2014.00574] [Citation(s) in RCA: 118] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2014] [Accepted: 10/27/2014] [Indexed: 01/07/2023] Open
Abstract
Preterm birth (PTB) (delivery before 37 weeks’ gestation) is a leading cause of neonatal death and disease in industrialized and developing countries alike. Infection (most notably in high-risk deliveries occurring before 28 weeks’ gestation) is hypothesized to initiate an intrauterine inflammatory response that plays a key role in the premature initiation of labor as well as a host of the pathologies associated with prematurity. As such, a better understanding of intrauterine inflammation in pregnancy is critical to our understanding of preterm labor and fetal injury, as well as on-going efforts to prevent PTB. Focusing on the fetal innate immune system responses to intrauterine infection, the present paper will review clinical and experimental studies to discuss the capacity for a fetal contribution to the intrauterine inflammation associated with PTB. Evidence from experimental studies to suggest that the fetus has the capacity to elicit a pro-inflammatory response to intrauterine infection is highlighted, with reference to the contribution of the lung, skin, and gastrointestinal tract. The paper will conclude that pathological intrauterine inflammation is a complex process that is modified by multiple factors including time, type of agonist, host genetics, and tissue.
Collapse
Affiliation(s)
- Matthew W Kemp
- School of Women's and Infants' Health, The University of Western Australia , Perth, WA , Australia
| |
Collapse
|
49
|
Ma Q, Zhang L. Epigenetic programming of hypoxic-ischemic encephalopathy in response to fetal hypoxia. Prog Neurobiol 2014; 124:28-48. [PMID: 25450949 DOI: 10.1016/j.pneurobio.2014.11.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2014] [Revised: 08/14/2014] [Accepted: 11/02/2014] [Indexed: 12/13/2022]
Abstract
Hypoxia is a major stress to the fetal development and may result in irreversible injury in the developing brain, increased risk of central nervous system (CNS) malformations in the neonatal brain and long-term neurological complications in offspring. Current evidence indicates that epigenetic mechanisms may contribute to the development of hypoxic/ischemic-sensitive phenotype in the developing brain in response to fetal stress. However, the causative cellular and molecular mechanisms remain elusive. In the present review, we summarize the recent findings of epigenetic mechanisms in the development of the brain and their roles in fetal hypoxia-induced brain developmental malformations. Specifically, we focus on DNA methylation and active demethylation, histone modifications and microRNAs in the regulation of neuronal and vascular developmental plasticity, which may play a role in fetal stress-induced epigenetic programming of hypoxic/ischemic-sensitive phenotype in the developing brain.
Collapse
Affiliation(s)
- Qingyi Ma
- Center for Perinatal Biology, Division of Pharmacology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - Lubo Zhang
- Center for Perinatal Biology, Division of Pharmacology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA.
| |
Collapse
|
50
|
Li J, McDonald CA, Fahey MC, Jenkin G, Miller SL. Could cord blood cell therapy reduce preterm brain injury? Front Neurol 2014; 5:200. [PMID: 25346720 PMCID: PMC4191167 DOI: 10.3389/fneur.2014.00200] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2014] [Accepted: 09/19/2014] [Indexed: 12/25/2022] Open
Abstract
Major advances in neonatal care have led to significant improvements in survival rates for preterm infants, but this occurs at a cost, with a strong causal link between preterm birth and neurological deficits, including cerebral palsy (CP). Indeed, in high-income countries, up to 50% of children with CP were born preterm. The pathways that link preterm birth and brain injury are complex and multifactorial, but it is clear that preterm birth is strongly associated with damage to the white matter of the developing brain. Nearly 90% of preterm infants who later develop spastic CP have evidence of periventricular white matter injury. There are currently no treatments targeted at protecting the immature preterm brain. Umbilical cord blood (UCB) contains a diverse mix of stem and progenitor cells, and is a particularly promising source of cells for clinical applications, due to ethical and practical advantages over other potential therapeutic cell types. Recent studies have documented the potential benefits of UCB cells in reducing brain injury, particularly in rodent models of term neonatal hypoxia–ischemia. These studies indicate that UCB cells act via anti-inflammatory and immuno-modulatory effects, and release neurotrophic growth factors to support the damaged and surrounding brain tissue. The etiology of brain injury in preterm-born infants is less well understood than in term infants, but likely results from episodes of hypoperfusion, hypoxia–ischemia, and/or inflammation over a developmental period of white matter vulnerability. This review will explore current knowledge about the neuroprotective actions of UCB cells and their potential to ameliorate preterm brain injury through neonatal cell administration. We will also discuss the characteristics of UCB-derived from preterm and term infants for use in clinical applications.
Collapse
Affiliation(s)
- Jingang Li
- The Ritchie Centre, MIMR-PHI Institute , Clayton, VIC , Australia
| | | | - Michael C Fahey
- The Ritchie Centre, MIMR-PHI Institute , Clayton, VIC , Australia ; Department of Paediatrics, Monash University , Clayton, VIC , Australia
| | - Graham Jenkin
- The Ritchie Centre, MIMR-PHI Institute , Clayton, VIC , Australia ; Department of Obstetrics and Gynaecology, Monash University , Clayton, VIC , Australia
| | - Suzanne L Miller
- The Ritchie Centre, MIMR-PHI Institute , Clayton, VIC , Australia ; Department of Obstetrics and Gynaecology, Monash University , Clayton, VIC , Australia
| |
Collapse
|