1
|
Li Z, Zhang F, Huang L, Deng J, Pan Y, Xu T, Liu J, Gao N, Duan R, Shao C, Wu C, Wang M, Lu L. Akt/mTOR Pathway Agonist SC79 Inhibits Autophagy and Apoptosis of Oligodendrocyte Precursor Cells Associated with Neonatal White Matter Dysplasia. Neurochem Res 2024; 49:670-683. [PMID: 38015410 PMCID: PMC10884134 DOI: 10.1007/s11064-023-04057-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 10/18/2023] [Accepted: 10/25/2023] [Indexed: 11/29/2023]
Abstract
White matter dysplasia (WMD) in preterm infants due to intrauterine inflammation is caused by excessive apoptosis of oligodendrocyte precursor cells (OPCs). In recent years, studies have found that excessive autophagy and apoptosis are highly interconnected and important in infection and inflammatory diseases in general. Therefore, in this study, we aimed to confirm whether regulation of autophagy by using the Akt phosphorylation agonist SC79 can inhibit abnormal apoptosis of OPCs and promote myelin maturation and white matter development in neonatal rats with WMD. We investigated the effect of inflammation on oligodendrocyte development in P0 neonatal rats by intracerebellar injection of LPS, and collected brain tissue at P2 and P5. Immunohistochemical and immunofluorescence staining were used to evaluate white matter damage, while immunofluorescence staining, terminal deoxynucleotidyl transferase dUTP nick end labeling analysis (TUNEL), and western blotting were used to evaluate autophagy and apoptosis. First, we observed that white matter development was arrested and white matter fiber maturation was impaired in LPS-inflicted pups compared with those in the sham-operated group. Second, treatment with SC79 reduced the levels of LC3II, caspase 3, caspase 9, and Bax/Bcl-2 and increased the levels of p62, p-Akt, and p-mTOR in the brain tissue of neonatal rats. Finally, SC79 treatment inhibited OPC apoptosis by increasing the binding of Beclin 1 to Bcl-2, which promoted OPC differentiation and maturation. However, the opposite results were observed after rapamycin administration. Taken together, our results suggest that SC79 can inhibit the abnormal apoptosis of OPCs caused by excessive autophagy through the Akt/mTOR pathway and that SC79 is a potential therapeutic agent for WMD in preterm infants.
Collapse
Affiliation(s)
- Zhongni Li
- Department of Pediatrics, The First Affiliated Hospital of Chengdu Medical College, No. 278, Middle Section of Baoguang Avenue, Xindu District, Chengdu, 610500, Sichuan Province, People's Republic of China
- Clinic Medical College, Chengdu Medical College, No. 783 Xindu Avenue, Xindu District, Chengdu, 610500, Sichuan Province, People's Republic of China
| | - Feng Zhang
- Department of Pediatrics, The First Affiliated Hospital of Chengdu Medical College, No. 278, Middle Section of Baoguang Avenue, Xindu District, Chengdu, 610500, Sichuan Province, People's Republic of China
- Clinic Medical College, Chengdu Medical College, No. 783 Xindu Avenue, Xindu District, Chengdu, 610500, Sichuan Province, People's Republic of China
| | - Li Huang
- Department of Pediatrics, The First Affiliated Hospital of Chengdu Medical College, No. 278, Middle Section of Baoguang Avenue, Xindu District, Chengdu, 610500, Sichuan Province, People's Republic of China
- Clinic Medical College, Chengdu Medical College, No. 783 Xindu Avenue, Xindu District, Chengdu, 610500, Sichuan Province, People's Republic of China
| | - Jiehong Deng
- Department of Pediatrics, The First Affiliated Hospital of Chengdu Medical College, No. 278, Middle Section of Baoguang Avenue, Xindu District, Chengdu, 610500, Sichuan Province, People's Republic of China
- Clinic Medical College, Chengdu Medical College, No. 783 Xindu Avenue, Xindu District, Chengdu, 610500, Sichuan Province, People's Republic of China
| | - Yutong Pan
- Department of Pediatrics, The First Affiliated Hospital of Chengdu Medical College, No. 278, Middle Section of Baoguang Avenue, Xindu District, Chengdu, 610500, Sichuan Province, People's Republic of China
- Clinic Medical College, Chengdu Medical College, No. 783 Xindu Avenue, Xindu District, Chengdu, 610500, Sichuan Province, People's Republic of China
| | - Ting Xu
- Department of Pediatrics, The First Affiliated Hospital of Chengdu Medical College, No. 278, Middle Section of Baoguang Avenue, Xindu District, Chengdu, 610500, Sichuan Province, People's Republic of China
- Clinic Medical College, Chengdu Medical College, No. 783 Xindu Avenue, Xindu District, Chengdu, 610500, Sichuan Province, People's Republic of China
| | - Jingyi Liu
- Department of Pediatrics, The First Affiliated Hospital of Chengdu Medical College, No. 278, Middle Section of Baoguang Avenue, Xindu District, Chengdu, 610500, Sichuan Province, People's Republic of China
- Clinic Medical College, Chengdu Medical College, No. 783 Xindu Avenue, Xindu District, Chengdu, 610500, Sichuan Province, People's Republic of China
| | - Na Gao
- Department of Pediatrics, The First Affiliated Hospital of Chengdu Medical College, No. 278, Middle Section of Baoguang Avenue, Xindu District, Chengdu, 610500, Sichuan Province, People's Republic of China
- Clinic Medical College, Chengdu Medical College, No. 783 Xindu Avenue, Xindu District, Chengdu, 610500, Sichuan Province, People's Republic of China
| | - Rongrong Duan
- Department of Pediatrics, The First Affiliated Hospital of Chengdu Medical College, No. 278, Middle Section of Baoguang Avenue, Xindu District, Chengdu, 610500, Sichuan Province, People's Republic of China
- Clinic Medical College, Chengdu Medical College, No. 783 Xindu Avenue, Xindu District, Chengdu, 610500, Sichuan Province, People's Republic of China
| | - Chunyan Shao
- Department of Pediatrics, The First Affiliated Hospital of Chengdu Medical College, No. 278, Middle Section of Baoguang Avenue, Xindu District, Chengdu, 610500, Sichuan Province, People's Republic of China
- Clinic Medical College, Chengdu Medical College, No. 783 Xindu Avenue, Xindu District, Chengdu, 610500, Sichuan Province, People's Republic of China
| | - Chan Wu
- Department of Pediatrics, The First Affiliated Hospital of Chengdu Medical College, No. 278, Middle Section of Baoguang Avenue, Xindu District, Chengdu, 610500, Sichuan Province, People's Republic of China
- Clinic Medical College, Chengdu Medical College, No. 783 Xindu Avenue, Xindu District, Chengdu, 610500, Sichuan Province, People's Republic of China
| | - Minrong Wang
- Department of Pediatrics, The First Affiliated Hospital of Chengdu Medical College, No. 278, Middle Section of Baoguang Avenue, Xindu District, Chengdu, 610500, Sichuan Province, People's Republic of China
- Clinic Medical College, Chengdu Medical College, No. 783 Xindu Avenue, Xindu District, Chengdu, 610500, Sichuan Province, People's Republic of China
| | - Liqun Lu
- Department of Pediatrics, The First Affiliated Hospital of Chengdu Medical College, No. 278, Middle Section of Baoguang Avenue, Xindu District, Chengdu, 610500, Sichuan Province, People's Republic of China.
- Clinic Medical College, Chengdu Medical College, No. 783 Xindu Avenue, Xindu District, Chengdu, 610500, Sichuan Province, People's Republic of China.
| |
Collapse
|
2
|
Zhang F, Zhang Z, Alt J, Kambhampati SP, Sharma A, Singh S, Nance E, Thomas AG, Rojas C, Rais R, Slusher BS, Kannan RM, Kannan S. Dendrimer-enabled targeted delivery attenuates glutamate excitotoxicity and improves motor function in a rabbit model of cerebral palsy. J Control Release 2023; 358:27-42. [PMID: 37054778 PMCID: PMC10330216 DOI: 10.1016/j.jconrel.2023.04.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 03/17/2023] [Accepted: 04/10/2023] [Indexed: 04/15/2023]
Abstract
Glutamate carboxypeptidase II (GCPII), localized on the surface of astrocytes and activated microglia, regulates extracellular glutamate concentration in the central nervous system (CNS). We have previously shown that GCPII is upregulated in activated microglia in the presence of inflammation. Inhibition of GCPII activity could reduce glutamate excitotoxicity, which may decrease inflammation and promote a 'normal' microglial phenotype. 2-(3-Mercaptopropyl) pentanedioic acid (2-MPPA) is the first GCPII inhibitor that underwent clinical trials. Unfortunately, immunological toxicities have hindered 2-MPPA clinical translation. Targeted delivery of 2-MPPA specifically to activated microglia and astrocytes that over-express GCPII has the potential to mitigate glutamate excitotoxicity and attenuate neuroinflammation. In this study, we demonstrate that 2-MPPA when conjugated to generation-4, hydroxyl-terminated polyamidoamine (PAMAM) dendrimers (D-2MPPA) localize specifically in activated microglia and astrocytes only in newborn rabbits with cerebral palsy (CP), not in controls. D-2MPPA treatment led to higher 2-MPPA levels in the injured brain regions compared to 2-MPPA treatment, and the extent of D-2MPPA uptake correlated with the injury severity. D-2MPPA was more efficacious than 2-MPPA in decreasing extracellular glutamate level in ex vivo brain slices of CP kits, and in increasing transforming growth factor beta 1 (TGF-β1) level in primary mixed glial cell cultures. A single systemic intravenous dose of D-2MPPA on postnatal day 1 (PND1) decreased microglial activation and resulted in a change in microglial morphology to a more ramified form along with amelioration of motor deficits by PND5. These results indicate that targeted dendrimer-based delivery specifically to activated microglia and astrocytes can improve the efficacy of 2-MPPA by attenuating glutamate excitotoxicity and microglial activation.
Collapse
Affiliation(s)
- Fan Zhang
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Zhi Zhang
- Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Jesse Alt
- Johns Hopkins Drug Discovery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Siva P Kambhampati
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Anjali Sharma
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Sarabdeep Singh
- Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Elizabeth Nance
- Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Ajit G Thomas
- Johns Hopkins Drug Discovery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Camilo Rojas
- Johns Hopkins Drug Discovery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Rana Rais
- Johns Hopkins Drug Discovery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Barbara S Slusher
- Johns Hopkins Drug Discovery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Rangaramanujam M Kannan
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA.
| | - Sujatha Kannan
- Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.
| |
Collapse
|
3
|
Zhang F, Gou Z, Zhou Y, Huang L, Shao C, Wang M, Wu C, Lu L. MicroRNA-21-5p agomir inhibits apoptosis of oligodendrocyte precursor cell and attenuates white matter injury in neonatal rats. Brain Res Bull 2022; 189:139-150. [PMID: 35985609 DOI: 10.1016/j.brainresbull.2022.08.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 08/10/2022] [Accepted: 08/13/2022] [Indexed: 11/25/2022]
Abstract
BACKGROUND AND RESEARCH QUESTION/HYPOTHESIS Excessive oligodendrocyte precursor cell (OPC) apoptosis occurs during intrauterine infection-induced white matter injury (WMI) in premature infants, preventing excessive apoptosis of OPCs is one of the mechanisms protecting WMI. Micro-RNA-21-5p (miR-21-5p) mediating anti-apoptotic activity was observed in other diseases. Therefore, the aim of this study was to determine whether miR-21-5p protects against WMI by modulating phosphatase and tensin homolog deleted on chromosome 10/phosphatidylinositol-3-kinase/protein kinase B (PTEN/PI3K/Akt) signaling pathway. METHODS A lipopolysaccharide (LPS)-induced neonatal Sprague-Dawley (SD) rat model of preterm WMI was established. To explore the effect of miR-21-5p on WMI, we intraventricularly injected miR-21-5p agomir and miR-21-5p antagomir to activate or inhibit endogenous miR-21-5p. Immunofluorescent labelling of myelin basic protein, immunohistochemical labelling of 2',3'-cyclic-nucleotide 3'-phosphodiesterase (CNPase), and terminal deoxynucleotidyl transferase dUTP nick end labelling assays were conducted to observe pathological white matter changes. The antibody of anti-oligodendrocyte marker 4 (O4) was used to specifically recognise OPCs. The expressions of miR-21-5p and PTEN mRNA in the brain were detected with quantitative real-time polymerase chain reaction (qRT-PCR). PTEN, Akt, and phosphorylated Akt (p-Akt) protein levels were assayed with western blotting, and apoptotic proteins associated with PI3K/Akt signalling were quantified. RESULTS Intense white matter dysplasia and excessive OPC apoptosis were observed in the brains of rats with WMI. When the miR-21-5p agonist miR-21-5p agomir was used in the WMI group, apoptosis of OPCs was significantly reduced, and myelin maturation increased. MiR-21-5p agomir relieved WMI. MiR-21-5p agomir inhibited the mRNA and protein expression of PTEN, increased p-Akt phosphorylation, and decreased the expression and activation of related apoptotic proteins.On the other hand, the administration of miR-21-5p specific blocker, miR-21-5p antagomir, reduced the level of p-AKT, increased OPC apoptosis, and worsened WMI. INTERPRETATION Our findings revealed that miR-21-5p agomir had anti-OPC over-apoptotic effects and enhanced myelin development in WMI by modulating the PTEN/Akt signaling pathway. DATA AVAILABILITY STATEMENT The datasets used and or/analysed in the current study are available from the corresponding author on reasonable request.
Collapse
Affiliation(s)
- Feng Zhang
- Clinic Medical College, Chengdu Medical College, No. 783 Xindu Avenue, Xindu District, Chengdu, Sichuan Province 610500, P.R. China; Department of Pediatrics, The First Affiliated Hospital of Chengdu Medical College, No. 278, Middle Section of Baoguang Avenue, Xindu District, Chengdu, Sichuan Province 610500, P.R. China
| | - Zhixian Gou
- Department of Pediatrics, The First Affiliated Hospital of Chengdu Medical College, No. 278, Middle Section of Baoguang Avenue, Xindu District, Chengdu, Sichuan Province 610500, P.R. China
| | - Yue Zhou
- Department of Pediatrics, The First Affiliated Hospital of Chengdu Medical College, No. 278, Middle Section of Baoguang Avenue, Xindu District, Chengdu, Sichuan Province 610500, P.R. China
| | - Lin Huang
- Clinic Medical College, Chengdu Medical College, No. 783 Xindu Avenue, Xindu District, Chengdu, Sichuan Province 610500, P.R. China; Department of Pediatrics, The First Affiliated Hospital of Chengdu Medical College, No. 278, Middle Section of Baoguang Avenue, Xindu District, Chengdu, Sichuan Province 610500, P.R. China
| | - Chunyan Shao
- Clinic Medical College, Chengdu Medical College, No. 783 Xindu Avenue, Xindu District, Chengdu, Sichuan Province 610500, P.R. China; Department of Pediatrics, The First Affiliated Hospital of Chengdu Medical College, No. 278, Middle Section of Baoguang Avenue, Xindu District, Chengdu, Sichuan Province 610500, P.R. China
| | - Minrong Wang
- Clinic Medical College, Chengdu Medical College, No. 783 Xindu Avenue, Xindu District, Chengdu, Sichuan Province 610500, P.R. China; Department of Pediatrics, The First Affiliated Hospital of Chengdu Medical College, No. 278, Middle Section of Baoguang Avenue, Xindu District, Chengdu, Sichuan Province 610500, P.R. China
| | - Chan Wu
- Clinic Medical College, Chengdu Medical College, No. 783 Xindu Avenue, Xindu District, Chengdu, Sichuan Province 610500, P.R. China; Department of Pediatrics, The First Affiliated Hospital of Chengdu Medical College, No. 278, Middle Section of Baoguang Avenue, Xindu District, Chengdu, Sichuan Province 610500, P.R. China
| | - Liqun Lu
- Clinic Medical College, Chengdu Medical College, No. 783 Xindu Avenue, Xindu District, Chengdu, Sichuan Province 610500, P.R. China; Department of Pediatrics, The First Affiliated Hospital of Chengdu Medical College, No. 278, Middle Section of Baoguang Avenue, Xindu District, Chengdu, Sichuan Province 610500, P.R. China.
| |
Collapse
|
4
|
Ardalan M, Chumak T, Quist A, Jabbari Shiadeh SM, Mallard AJ, Rafati AH, Mallard C. Sex dependent glio-vascular interface abnormality in the hippocampus following postnatal immune activation in mice. Dev Neurosci 2022; 44:320-330. [PMID: 35705008 PMCID: PMC9533445 DOI: 10.1159/000525478] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 06/06/2022] [Indexed: 11/19/2022] Open
Abstract
The neuro-gliovascular unit is a crucial structure for providing a balanced well-functioning environment for neurons and their synapses. Activation of the immune system during the developmental period is believed to affect the gliovascular unit, which may trigger neurodevelopmental and neurological/neuropsychiatric diseases. In this study, we hypothesized that vulnerability of the male brain to a neonatal insult was conditioned by sex-dependent differences in the impairment of the hippocampal gliovascular unit. Male and female C57BL/6J pups received lipopolysaccharide (LPS) (1 mg/kg) or saline on postnatal day (P) 5. Brains were collected at P12 and morphological quantifications of hippocampal fibrillary glial acid protein (GFAP<sup>+</sup>) astrocytes and ionized calcium-binding adaptor molecule 1 protein (Iba1+) microglia were performed by using 3-D image analysis together with measuring the length of CD31<sup>+</sup> and aquaporin-4 (AQP4<sup>+</sup>) vessels. We found a significant increase in the length of CD31<sup>+</sup> capillaries in the male LPS group compared to the saline group; however, coverage of capillaries by astrocytic end-feet (AQP4<sup>+</sup>) was significantly reduced. In contrast, there was a significant increase in AQP4<sup>+</sup> capillary length in female pups 1 week after LPS injection. GFAP<sup>+</sup> astrocytes via morphological changes in the hippocampus showed significant enhancement in the activity 1 week following LPS injection in male mice. We propose that neonatal inflammation could induce susceptibility to neurodevelopmental disorders through modification of hippocampal gliovascular interface in a sex-dependent manner.
Collapse
Affiliation(s)
- Maryam Ardalan
- Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- *Maryam Ardalan,
| | - Tetyana Chumak
- Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Alexandra Quist
- Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Seyedeh Marziyeh Jabbari Shiadeh
- Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Anna-Jean Mallard
- Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Ali Hoseinpoor Rafati
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Carina Mallard
- Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
5
|
Gall AR, Amoah SK, Kitase Y, Jantzie LL. Placental mediated mechanisms of perinatal brain injury: Evolving inflammation and exosomes. Exp Neurol 2022; 347:113914. [PMID: 34752783 PMCID: PMC8712107 DOI: 10.1016/j.expneurol.2021.113914] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Revised: 10/29/2021] [Accepted: 11/01/2021] [Indexed: 01/03/2023]
Abstract
Pregnancy is an inflammatory process that is carefully regulated by the placenta via immunomodulation and cell-to-cell communication of maternal and fetal tissues. Exosomes, types of extracellular vesicles, facilitate the intercellular communication and traffic biologically modifying cargo within the maternal-placental-fetal axis in normal and pathologic pregnancies. Chorioamnionitis is characterized by inflammation of chorioamniotic membranes that produces systemic maternal and fetal inflammatory responses of cytokine dysregulation and has been associated with brain injury and neurodevelopmental disorders. This review focuses on how pathologic placental exosomes propagate acute and chronic inflammation leading to brain injury. The evidence reviewed here highlights the need to investigate exosomes from pathologic pregnancies and those with known brain injury to identify new diagnostics, biomarkers, and potential therapeutic targets.
Collapse
Affiliation(s)
- Alexander R Gall
- Division of Neonatology, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Stephen K Amoah
- Division of Neonatology, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Yuma Kitase
- Division of Neonatology, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Lauren L Jantzie
- Division of Neonatology, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA,Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA,Kennedy Krieger Institute, Baltimore, MD, USA,Corresponding author at: 600 N. Wolfe Street, CMSC Building, 6-104A, Baltimore, MD 21287, USA. (L.L. Jantzie)
| |
Collapse
|
6
|
Van der Veeken L, Emam D, Bleeser T, Valenzuela I, Van der Merwe J, Rex S, Deprest J. Fetal surgery has no additional effect to general anesthesia on brain development in neonatal rabbits. Am J Obstet Gynecol MFM 2022; 4:100513. [PMID: 34706302 DOI: 10.1016/j.ajogmf.2021.100513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 10/08/2021] [Accepted: 10/10/2021] [Indexed: 11/29/2022]
Abstract
BACKGROUND Fetal surgery is part of modern fetal medicine, and some procedures, such as fetal spina bifida repair, are performed under general anesthesia. Fetuses are operated on in a time window when the developing brain is extremely vulnerable to external, potentially harmful factors. To date, little is known about the effect of fetal surgery on fetal brain development. OBJECTIVE This study aimed to assess the effect of fetal surgery on the developing fetal brain in the rabbit model. STUDY DESIGN This was a randomized, sham-controlled study in time-mated pregnant does at 28 days' gestation (term, 31 days), which corresponds to the start of the peak of brain development and end of the second trimester of pregnancy in humans. We included 4 different groups in this experiment: no-surgery, general anesthesia, general anesthesia+hysterotomy, and general anesthesia+fetal surgery. In 11 does, anesthesia was induced using propofol and maintained for 75 minutes with 3.6 vol% (4% is the equivalent of 1 minimum alveolar concentration) sevoflurane. Maternal blood pressure, heart rate, oxygen saturation, temperature, end-tidal CO2 were continuously monitored. For each operated doe, 6 fetuses were part of the experiment. Randomization determined which cornual sac and what opposing third sac were assigned to fetal surgery: hysterotomy, fetal injection (atropine, fentanyl, and cisatracurium), fetal skin incision, and suturing. Only hysterotomy was performed on the opposing cornual and third amniotic sacs of the does. The fetus in these experimental sacs was used as internal unmanipulated control (general anesthesia). All fetuses (n=38) from unmanipulated does (n=4) served as external controls (no-surgery). At term, the does were delivered by cesarean delivery under ketamine-medetomidine sedation and local anesthesia. The pups underwent standardized motoric and sensory neurologic testing on day 1 followed by euthanasia and brain harvesting for histologic assessment of neurons, synapses, proliferation, and glial cells. RESULTS Maternal vital signs were stable during surgery. Survival was similar in the 4 groups (75%-94%), and brain-to-body weight ratio was comparable; only the no-surgery pups had a higher brain weight. On postnatal day 1, the pups in the 4 groups had a comparable neurobehavioral outcome on both motoric and sensory testing. In the prefrontal cortex, no-surgery pups had significantly higher neuron density than pups who underwent maternal surgery, but there was no difference among pups that underwent general anesthesia, hysterotomy, or fetal surgery. The measurements of proliferation had a similar outcome: a higher proliferation rate in the prefrontal cortex of no-surgery pups. Moreover, synaptic density values were higher in the no-surgery pups, but there was no difference observed among pups who underwent general anesthesia, hysterotomy, and fetal surgery. Lastly, there was no difference in gliosis among the 4 groups. CONCLUSION In rabbits, fetal surgery through hysterotomy under maternal general anesthesia did not affect brain development, in addition to the effects of general anesthesia per se.
Collapse
Affiliation(s)
- Lennart Van der Veeken
- Department of Development and Regeneration, Cluster Woman and Child, Group Biomedical Sciences, Catholic University of Leuven, Leuven, Belgium (Drs Van der Veeken, Emam, and Valenzuela, Dr Van der Merwe, and Dr Deprest); Department of Obstetrics and Gynaecology, University Hospitals Leuven, Leuven, Belgium (Drs Van der Veeken and Valenzuela, Dr Van der Merwe, and Dr Deprest); Departement of Obstetrics and Gynecology, University Hospital Antwerp, Belgium (Dr Van der Veeken)
| | - Doaa Emam
- Department of Development and Regeneration, Cluster Woman and Child, Group Biomedical Sciences, Catholic University of Leuven, Leuven, Belgium (Drs Van der Veeken, Emam, and Valenzuela, Dr Van der Merwe, and Dr Deprest); Department Obstetrics and Gynaecology, University Hospitals Tanta, Tanta, Egypt (Dr Emam)
| | - Tom Bleeser
- Department of Anesthesiology, University Hospitals Leuven, Leuven, Belgium (Drs Bleeser and Rex); Department of Cardiovascular Sciences, Group Biomedical Sciences, Catholic University of Leuven, Leuven, Belgium (Drs Bleeser and Rex)
| | - Ignacio Valenzuela
- Department of Development and Regeneration, Cluster Woman and Child, Group Biomedical Sciences, Catholic University of Leuven, Leuven, Belgium (Drs Van der Veeken, Emam, and Valenzuela, Dr Van der Merwe, and Dr Deprest); Department of Obstetrics and Gynaecology, University Hospitals Leuven, Leuven, Belgium (Drs Van der Veeken and Valenzuela, Dr Van der Merwe, and Dr Deprest)
| | - Johannes Van der Merwe
- Department of Development and Regeneration, Cluster Woman and Child, Group Biomedical Sciences, Catholic University of Leuven, Leuven, Belgium (Drs Van der Veeken, Emam, and Valenzuela, Dr Van der Merwe, and Dr Deprest); Department of Obstetrics and Gynaecology, University Hospitals Leuven, Leuven, Belgium (Drs Van der Veeken and Valenzuela, Dr Van der Merwe, and Dr Deprest)
| | - Steffen Rex
- Department of Anesthesiology, University Hospitals Leuven, Leuven, Belgium (Drs Bleeser and Rex); Department of Cardiovascular Sciences, Group Biomedical Sciences, Catholic University of Leuven, Leuven, Belgium (Drs Bleeser and Rex)
| | - Jan Deprest
- Department of Development and Regeneration, Cluster Woman and Child, Group Biomedical Sciences, Catholic University of Leuven, Leuven, Belgium (Drs Van der Veeken, Emam, and Valenzuela, Dr Van der Merwe, and Dr Deprest); Department of Obstetrics and Gynaecology, University Hospitals Leuven, Leuven, Belgium (Drs Van der Veeken and Valenzuela, Dr Van der Merwe, and Dr Deprest); Institute for Women's Health, University College London, London, United Kingdom (Dr Deprest).
| |
Collapse
|
7
|
Van Camp N, Lavisse S, Roost P, Gubinelli F, Hillmer A, Boutin H. TSPO imaging in animal models of brain diseases. Eur J Nucl Med Mol Imaging 2021; 49:77-109. [PMID: 34245328 PMCID: PMC8712305 DOI: 10.1007/s00259-021-05379-z] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 04/25/2021] [Indexed: 12/19/2022]
Abstract
Over the last 30 years, the 18-kDa TSPO protein has been considered as the PET imaging biomarker of reference to measure increased neuroinflammation. Generally assumed to image activated microglia, TSPO has also been detected in endothelial cells and activated astrocytes. Here, we provide an exhaustive overview of the recent literature on the TSPO-PET imaging (i) in the search and development of new TSPO tracers and (ii) in the understanding of acute and chronic neuroinflammation in animal models of neurological disorders. Generally, studies testing new TSPO radiotracers against the prototypic [11C]-R-PK11195 or more recent competitors use models of acute focal neuroinflammation (e.g. stroke or lipopolysaccharide injection). These studies have led to the development of over 60 new tracers during the last 15 years. These studies highlighted that interpretation of TSPO-PET is easier in acute models of focal lesions, whereas in chronic models with lower or diffuse microglial activation, such as models of Alzheimer's disease or Parkinson's disease, TSPO quantification for detection of neuroinflammation is more challenging, mirroring what is observed in clinic. Moreover, technical limitations of preclinical scanners provide a drawback when studying modest neuroinflammation in small brains (e.g. in mice). Overall, this review underlines the value of TSPO imaging to study the time course or response to treatment of neuroinflammation in acute or chronic models of diseases. As such, TSPO remains the gold standard biomarker reference for neuroinflammation, waiting for new radioligands for other, more specific targets for neuroinflammatory processes and/or immune cells to emerge.
Collapse
Affiliation(s)
- Nadja Van Camp
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, 92265, Fontenay-aux-Roses, France
| | - Sonia Lavisse
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, 92265, Fontenay-aux-Roses, France
| | - Pauline Roost
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, 92265, Fontenay-aux-Roses, France
| | - Francesco Gubinelli
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, 92265, Fontenay-aux-Roses, France
| | - Ansel Hillmer
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
- Department of Radiology & Biomedical Imaging, Yale School of Medicine, New Haven, CT, USA
- Department of Biomedical Engineering, Yale School of Engineering & Applied Science, New Haven, CT, USA
| | - Hervé Boutin
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Brain and Mental Health, University of Manchester, M13 9PL, Manchester, UK.
- Wolfson Molecular Imaging Centre, University of Manchester, 27 Palatine Road, M20 3LJ, Manchester, UK.
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance & University of Manchester, Manchester, UK.
| |
Collapse
|
8
|
Wu J, Zhou M, Qin K, Liao S, Tang C, Ruan Y, Hu X, Long F, Mo K, Kuang H, Deng R. Microscopic anatomical atlas study on the lateral ventricles of the rabbit cerebrum and its related structures. TRANSLATIONAL RESEARCH IN ANATOMY 2021. [DOI: 10.1016/j.tria.2021.100140] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
|
9
|
Guma E, Snook E, Spring S, Lerch JP, Nieman BJ, Devenyi GA, Chakravarty MM. Subtle alterations in neonatal neurodevelopment following early or late exposure to prenatal maternal immune activation in mice. Neuroimage Clin 2021; 32:102868. [PMID: 34749289 PMCID: PMC8573196 DOI: 10.1016/j.nicl.2021.102868] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 10/25/2021] [Accepted: 10/26/2021] [Indexed: 11/21/2022]
Abstract
Prenatal exposure to maternal immune activation (MIA) is a risk factor for a variety of neurodevelopmental and psychiatric disorders. The timing of MIA-exposure has been shown to affect adolescent and adult offspring neurodevelopment, however, less is known about these effects in the neonatal period. To better understand the impact of MIA-exposure on neonatal brain development in a mouse model, we assess neonate communicative abilities with the ultrasonic vocalization task, followed by high-resolution ex vivo magnetic resonance imaging (MRI) on the neonatal (postnatal day 8) mouse brain. Early exposed offspring displayed decreased communicative ability, while brain anatomy appeared largely unaffected, apart from some subtle alterations. By integrating MRI and behavioural assays to investigate the effects of MIA-exposure on neonatal neurodevelopment we show that offspring neuroanatomy and behaviour are only subtly affected by both early and late exposure. This suggests that the deficits often observed in later stages of life may be dormant, not yet developed in the neonatal period, or not as easily detectable using a cross-sectional approach.
Collapse
Affiliation(s)
- Elisa Guma
- Computational Brain Anatomy Laboratory, Cerebral Imaging Center, Douglas Mental Health University Institute, Montreal, Quebec, Canada; Integrated Program in Neuroscience, McGill University, Montreal, Quebec, Canada.
| | - Emily Snook
- Computational Brain Anatomy Laboratory, Cerebral Imaging Center, Douglas Mental Health University Institute, Montreal, Quebec, Canada; Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Shoshana Spring
- Mouse Imaging Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Jason P Lerch
- Mouse Imaging Centre, The Hospital for Sick Children, Toronto, Ontario, Canada; Department of Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, Ontario, Canada; Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada; Wellcome Centre for Integrative Neuroimaging, University of Oxford, Oxford, United Kingdom
| | - Brian J Nieman
- Mouse Imaging Centre, The Hospital for Sick Children, Toronto, Ontario, Canada; Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada; Translational Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada; Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Gabriel A Devenyi
- Computational Brain Anatomy Laboratory, Cerebral Imaging Center, Douglas Mental Health University Institute, Montreal, Quebec, Canada; Department of Psychiatry, McGill University, Montreal, Quebec, Canada
| | - M Mallar Chakravarty
- Computational Brain Anatomy Laboratory, Cerebral Imaging Center, Douglas Mental Health University Institute, Montreal, Quebec, Canada; Integrated Program in Neuroscience, McGill University, Montreal, Quebec, Canada; Department of Psychiatry, McGill University, Montreal, Quebec, Canada; Department of Biological and Biomedical Engineering, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
10
|
Guma E, Bordignon PDC, Devenyi GA, Gallino D, Anastassiadis C, Cvetkovska V, Barry AD, Snook E, Germann J, Greenwood CMT, Misic B, Bagot RC, Chakravarty MM. Early or Late Gestational Exposure to Maternal Immune Activation Alters Neurodevelopmental Trajectories in Mice: An Integrated Neuroimaging, Behavioral, and Transcriptional Study. Biol Psychiatry 2021; 90:328-341. [PMID: 34053674 DOI: 10.1016/j.biopsych.2021.03.017] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 02/23/2021] [Accepted: 03/15/2021] [Indexed: 02/06/2023]
Abstract
BACKGROUND Exposure to maternal immune activation (MIA) in utero is a risk factor for neurodevelopmental disorders later in life. The impact of the gestational timing of MIA exposure on downstream development remains unclear. METHODS We characterized neurodevelopmental trajectories of mice exposed to the viral mimetic poly I:C (polyinosinic:polycytidylic acid) either on gestational day 9 (early) or on day 17 (late) using longitudinal structural magnetic resonance imaging from weaning to adulthood. Using multivariate methods, we related neuroimaging and behavioral variables for the time of greatest alteration (adolescence/early adulthood) and identified regions for further investigation using RNA sequencing. RESULTS Early MIA exposure was associated with accelerated brain volume increases in adolescence/early adulthood that normalized in later adulthood in the striatum, hippocampus, and cingulate cortex. Similarly, alterations in anxiety-like, stereotypic, and sensorimotor gating behaviors observed in adolescence normalized in adulthood. MIA exposure in late gestation had less impact on anatomical and behavioral profiles. Multivariate maps associated anxiety-like, social, and sensorimotor gating deficits with volume of the dorsal and ventral hippocampus and anterior cingulate cortex, among others. The most transcriptional changes were observed in the dorsal hippocampus, with genes enriched for fibroblast growth factor regulation, autistic behaviors, inflammatory pathways, and microRNA regulation. CONCLUSIONS Leveraging an integrated hypothesis- and data-driven approach linking brain-behavior alterations to the transcriptome, we found that MIA timing differentially affects offspring development. Exposure in late gestation leads to subthreshold deficits, whereas exposure in early gestation perturbs brain development mechanisms implicated in neurodevelopmental disorders.
Collapse
Affiliation(s)
- Elisa Guma
- Integrated Program in Neuroscience, McGill University, Montreal, Quebec, Canada; Computational Brain Imaging Lab, Cerebral Imaging Center, Douglas Mental Health University Institute, Montreal, Quebec, Canada.
| | - Pedro do Couto Bordignon
- Department of Psychology, McGill University, Montreal, Quebec, Canada; Ludmer Center for Neuroinformatics and Mental Health, Montreal, Quebec, Canada
| | - Gabriel A Devenyi
- Department of Psychiatry, McGill University, Montreal, Quebec, Canada; Computational Brain Imaging Lab, Cerebral Imaging Center, Douglas Mental Health University Institute, Montreal, Quebec, Canada
| | - Daniel Gallino
- Computational Brain Imaging Lab, Cerebral Imaging Center, Douglas Mental Health University Institute, Montreal, Quebec, Canada
| | - Chloe Anastassiadis
- Computational Brain Imaging Lab, Cerebral Imaging Center, Douglas Mental Health University Institute, Montreal, Quebec, Canada; Institute of Medical Science & Collaborative Program in Neuroscience, University of Toronto, Toronto, Ontario, Canada
| | | | - Amadou D Barry
- Departments of Human Genetics and Epidemiology, Biostatistics and Occupational Health, McGill University, Montreal, Quebec, Canada; Ludmer Center for Neuroinformatics and Mental Health, Montreal, Quebec, Canada
| | - Emily Snook
- Computational Brain Imaging Lab, Cerebral Imaging Center, Douglas Mental Health University Institute, Montreal, Quebec, Canada; Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Jurgen Germann
- Computational Brain Imaging Lab, Cerebral Imaging Center, Douglas Mental Health University Institute, Montreal, Quebec, Canada; University Health Network, Toronto, Ontario, Canada
| | - Celia M T Greenwood
- Gerald Bronfman Department of Oncology, McGill University, Montreal, Quebec, Canada; Lady Davis Institute for Medical Research, Jewish General Hospital, McGill University, Montreal, Quebec, Canada; Departments of Human Genetics and Epidemiology, Biostatistics and Occupational Health, McGill University, Montreal, Quebec, Canada; Ludmer Center for Neuroinformatics and Mental Health, Montreal, Quebec, Canada
| | - Bratislav Misic
- Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Rosemary C Bagot
- Department of Psychology, McGill University, Montreal, Quebec, Canada; Ludmer Center for Neuroinformatics and Mental Health, Montreal, Quebec, Canada
| | - M Mallar Chakravarty
- Integrated Program in Neuroscience, McGill University, Montreal, Quebec, Canada; Department of Psychiatry, McGill University, Montreal, Quebec, Canada; Department of Biological and Biomedical Engineering, McGill University, Montreal, Quebec, Canada; Computational Brain Imaging Lab, Cerebral Imaging Center, Douglas Mental Health University Institute, Montreal, Quebec, Canada.
| |
Collapse
|
11
|
Herrera-Ruiz M, Jiménez-Ferrer E, Tortoriello J, Zamilpa A, Alegría-Herrera E, Jiménez-Aparicio AR, Arenas-Ocampo ML, Martínez-Duncker I, Monterrosas-Brisson N. Anti-neuroinflammatory effect of agaves and cantalasaponin-1 in a model of LPS-induced damage. Nat Prod Res 2021; 35:884-887. [PMID: 31084220 DOI: 10.1080/14786419.2019.1608537] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 03/26/2019] [Accepted: 04/06/2019] [Indexed: 12/28/2022]
Abstract
Chronic neuroinflammation is a key component of many neurodegenerative disorders. Chronic activation of this process produces pro-inflammatory cytokines, prostaglandins and reactive oxygen species that induce brain injury and neuronal dysfunction. Agave species contain saponins, compounds with anti-inflammatory activity. Extracts from A. tequilana (At), A. angustifolia (Aan), A. Americana (Aam) (125 mg/kg) and cantalasaponin-1 (5 and 10 mg/kg, isolated from Aam) were administered to male ICR mice with lipopolysaccharide (LPS)-induced neuroinflammation, after which inflammatory cytokines were measured in brain homogenates by using an enzyme-linked immunoassay (ELISA) test. All agave extracts and cantalasaponin-1, reduced brain concentration of LPS-induced pro-inflammatory cytokines IL-6 and TNF-α. Moreover, Cantalasaponin-1 increased the brain concentration of the anti-inflammatory cytokine IL-10. Agave extracts and derived compounds show promising results in the development of novel drugs for neuroinflammatory disease therapy.
Collapse
Affiliation(s)
- Maribel Herrera-Ruiz
- Instituto Mexicano del Seguro Social (IMSS), Centro de Investigación Biomédica del Sur, Xochitepec, Morelos, Mexico
| | - Enrique Jiménez-Ferrer
- Instituto Mexicano del Seguro Social (IMSS), Centro de Investigación Biomédica del Sur, Xochitepec, Morelos, Mexico
| | - Jaime Tortoriello
- Instituto Mexicano del Seguro Social (IMSS), Centro de Investigación Biomédica del Sur, Xochitepec, Morelos, Mexico
| | - Alejandro Zamilpa
- Instituto Mexicano del Seguro Social (IMSS), Centro de Investigación Biomédica del Sur, Xochitepec, Morelos, Mexico
| | - Elian Alegría-Herrera
- Instituto Mexicano del Seguro Social (IMSS), Centro de Investigación Biomédica del Sur, Xochitepec, Morelos, Mexico
| | | | - Martha L Arenas-Ocampo
- Centro de Desarrollo de Productos Bióticos, Instituto Politécnico Nacional, Yautepec, Morelos, Mexico
| | - Iván Martínez-Duncker
- Centro de Investigación en Dinámica Celular, Universidad Autónoma del Estado de Morelos (UAEM), Cuernavaca, Morelos, Mexico
| | - Nayeli Monterrosas-Brisson
- Facultad de Ciencias Biológicas, Universidad Autónoma del Estado de Morelos (UAEM), Cuernavaca, Morelos, Mexico
| |
Collapse
|
12
|
Bleeser T, Van Der Veeken L, Devroe S, Vergote S, Emam D, van der Merwe J, Ghijsens E, Joyeux L, Basurto D, Van de Velde M, Deprest J, Rex S. Effects of Maternal Abdominal Surgery on Fetal Brain Development in the Rabbit Model. Fetal Diagn Ther 2021; 48:189-200. [PMID: 33631746 PMCID: PMC7613467 DOI: 10.1159/000512489] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 10/22/2020] [Indexed: 11/19/2022]
Abstract
INTRODUCTION Anesthesia during pregnancy can impair fetal neurodevelopment, but effects of surgery remain unknown. The aim is to investigate effects of abdominal surgery on fetal brain development. Hypothesis is that surgery impairs outcome. METHODS Pregnant rabbits were randomized at 28 days of gestation to 2 h of general anesthesia (sevoflurane group, n = 6) or to anesthesia plus laparoscopic appendectomy (surgery group, n = 13). On postnatal day 1, neurobehavior of pups was assessed and brains harvested. Primary outcome was neuron density in the frontal cortex, and secondary outcomes included neurobehavioral assessment and other histological parameters. RESULTS Fetal survival was lower in the surgery group: 54 versus 100% litters alive at birth (p = 0.0442). In alive litters, pup survival until harvesting was 50 versus 69% (p = 0.0352). No differences were observed for primary outcome (p = 0.5114) for surviving pups. Neuron densities were significantly lower in the surgery group in the caudate nucleus (p = 0.0180), but not different in other regions. No differences were observed for secondary outcomes. Conclusions did not change after adjustment for mortality. CONCLUSION Abdominal surgery in pregnant rabbits at a gestational age corresponding to the end of human second trimester results in limited neurohistological changes but not in neurobehavioral impairments. High intrauterine mortality limits translation to clinical scenario, where fetal mortality is close to zero.
Collapse
Affiliation(s)
- Tom Bleeser
- Department of Anesthesiology, UZ Leuven, Leuven, Belgium
- Department of Cardiovascular Sciences, Group Biomedical Sciences, KU Leuven, Leuven, Belgium
- Department of Development and Regeneration, My FetUZ Fetal Research Center, KU Leuven, Leuven, Belgium
| | - Lennart Van Der Veeken
- Department of Development and Regeneration, My FetUZ Fetal Research Center, KU Leuven, Leuven, Belgium
- Center for Surgical Technologies, Faculty of Medicine, KU Leuven, Leuven, Belgium
- Department of Obstetrics and Gynecology, UZ Leuven, Leuven, Belgium
| | - Sarah Devroe
- Department of Anesthesiology, UZ Leuven, Leuven, Belgium
- Department of Cardiovascular Sciences, Group Biomedical Sciences, KU Leuven, Leuven, Belgium
- Department of Development and Regeneration, My FetUZ Fetal Research Center, KU Leuven, Leuven, Belgium
| | - Simen Vergote
- Department of Development and Regeneration, My FetUZ Fetal Research Center, KU Leuven, Leuven, Belgium
- Center for Surgical Technologies, Faculty of Medicine, KU Leuven, Leuven, Belgium
- Department of Obstetrics and Gynecology, UZ Leuven, Leuven, Belgium
| | - Doaa Emam
- Department of Development and Regeneration, My FetUZ Fetal Research Center, KU Leuven, Leuven, Belgium
- Center for Surgical Technologies, Faculty of Medicine, KU Leuven, Leuven, Belgium
- Department of Obstetrics and Gynecology, UZ Leuven, Leuven, Belgium
- Department Obstetrics and Gynecology, University Hospitals Tanta, Tanta, Egypt
| | - Johannes van der Merwe
- Department of Development and Regeneration, My FetUZ Fetal Research Center, KU Leuven, Leuven, Belgium
- Center for Surgical Technologies, Faculty of Medicine, KU Leuven, Leuven, Belgium
- Department of Obstetrics and Gynecology, UZ Leuven, Leuven, Belgium
| | - Elina Ghijsens
- Center for Surgical Technologies, Faculty of Medicine, KU Leuven, Leuven, Belgium
| | - Luc Joyeux
- Department of Development and Regeneration, My FetUZ Fetal Research Center, KU Leuven, Leuven, Belgium
- Center for Surgical Technologies, Faculty of Medicine, KU Leuven, Leuven, Belgium
- Department of Obstetrics and Gynecology, UZ Leuven, Leuven, Belgium
- Department of Pediatric Surgery, Great Ormond Street Hospital, University College London Hospitals, London, United Kingdom
| | - David Basurto
- Department of Development and Regeneration, My FetUZ Fetal Research Center, KU Leuven, Leuven, Belgium
- Center for Surgical Technologies, Faculty of Medicine, KU Leuven, Leuven, Belgium
- Department of Obstetrics and Gynecology, UZ Leuven, Leuven, Belgium
| | - Marc Van de Velde
- Department of Anesthesiology, UZ Leuven, Leuven, Belgium
- Department of Cardiovascular Sciences, Group Biomedical Sciences, KU Leuven, Leuven, Belgium
| | - Jan Deprest
- Department of Development and Regeneration, My FetUZ Fetal Research Center, KU Leuven, Leuven, Belgium
- Center for Surgical Technologies, Faculty of Medicine, KU Leuven, Leuven, Belgium
- Department of Obstetrics and Gynecology, UZ Leuven, Leuven, Belgium
- Institute for Women's Health, University College London, London, United Kingdom
| | - Steffen Rex
- Department of Anesthesiology, UZ Leuven, Leuven, Belgium,
- Department of Cardiovascular Sciences, Group Biomedical Sciences, KU Leuven, Leuven, Belgium,
| |
Collapse
|
13
|
Qiu H, Qian T, Wu T, Wang X, Zhu C, Chen C, Wang L. Umbilical cord blood cells for the treatment of preterm white matter injury: Potential effects and treatment options. J Neurosci Res 2020; 99:778-792. [PMID: 33207392 DOI: 10.1002/jnr.24751] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 10/20/2020] [Accepted: 10/21/2020] [Indexed: 12/31/2022]
Abstract
Preterm birth is a global public health problem. A large number of preterm infants survive with preterm white matter injury (PWMI), which leads to neurological deficits, and has multifaceted etiology, clinical course, monitoring, and outcomes. The principal upstream insults leading to PWMI initiation are hypoxia-ischemia and infection and/or inflammation and the key target cells are late oligodendrocyte precursor cells. Current PWMI treatments are mainly supportive, and thus have little effect in terms of protecting the immature brain or repairing injury to improve long-term outcomes. Umbilical cord blood (UCB) cells comprise abundant immunomodulatory and stem cells, which have the potential to reduce brain injury, mainly due to anti-inflammatory and immunomodulatory mechanisms, and also through their release of neurotrophic or growth factors to promote endogenous neurogenesis. In this review, we briefly summarize PWMI pathogenesis and pathophysiology, and the specific properties of different cell types in UCB. We further explore the potential mechanism by which UCB can be used to treat PWMI, and discuss the advantages of and potential issues related to UCB cell therapy. Finally, we suggest potential future studies of UCB cell therapy in preterm infants.
Collapse
Affiliation(s)
- Han Qiu
- Key Laboratory of Neonatal Diseases of Health Commission of the People's Republic of China, Shanghai, China.,Department of Neonatology, National Children's Medical Center/Children's Hospital of Fudan University, Shanghai, China
| | - Tianyang Qian
- Key Laboratory of Neonatal Diseases of Health Commission of the People's Republic of China, Shanghai, China.,Department of Neonatology, National Children's Medical Center/Children's Hospital of Fudan University, Shanghai, China
| | - Tong Wu
- Key Laboratory of Neonatal Diseases of Health Commission of the People's Republic of China, Shanghai, China.,Department of Neonatology, National Children's Medical Center/Children's Hospital of Fudan University, Shanghai, China
| | - Xiaoyang Wang
- Center of Perinatal Medicine and Health, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Henan Key Laboratory of Child Brain Injury, Institute of Neuroscience and Third Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| | - Changlian Zhu
- Center of Perinatal Medicine and Health, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Henan Key Laboratory of Child Brain Injury, Institute of Neuroscience and Third Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| | - Chao Chen
- Key Laboratory of Neonatal Diseases of Health Commission of the People's Republic of China, Shanghai, China.,Department of Neonatology, National Children's Medical Center/Children's Hospital of Fudan University, Shanghai, China
| | - Laishuan Wang
- Key Laboratory of Neonatal Diseases of Health Commission of the People's Republic of China, Shanghai, China.,Department of Neonatology, National Children's Medical Center/Children's Hospital of Fudan University, Shanghai, China
| |
Collapse
|
14
|
Zhang Z, Lin YA, Kim SY, Su L, Liu J, Kannan RM, Kannan S. Systemic dendrimer-drug nanomedicines for long-term treatment of mild-moderate cerebral palsy in a rabbit model. J Neuroinflammation 2020; 17:319. [PMID: 33100217 PMCID: PMC7586697 DOI: 10.1186/s12974-020-01984-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 10/05/2020] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Neuroinflammation mediated by microglia plays a central role in the pathogenesis of perinatal/neonatal brain injury, including cerebral palsy (CP). Therapeutics mitigating neuroinflammation potentially provide an effective strategy to slow the disease progression and rescue normal brain development. Building on our prior results which showed that a generation-4 hydroxyl poly(amidoamine) (PAMAM) dendrimer could deliver drugs specifically to activated glia from systemic circulation, we evaluated the sustained efficacy of a generation-6 (G6) hydroxyl-terminated PAMAM dendrimer that showed a longer blood circulation time and increased brain accumulation. N-acetyl-L-cysteine (NAC), an antioxidant and anti-inflammatory agent that has high plasma protein binding properties and poor brain penetration, was conjugated to G6-PAMAM dendrimer-NAC (G6D-NAC). The efficacy of microglia-targeted G6D-NAC conjugate was evaluated in a clinically relevant rabbit model of CP, with a mild/moderate CP phenotype to provide a longer survival of untreated CP kits, enabling the assessment of sustained efficacy over 15 days of life. METHODS G6D-NAC was conjugated and characterized. Cytotoxicity and anti-inflammatory assays were performed in BV-2 microglial cells. The efficacy of G6D-NAC was evaluated in a rabbit model of CP. CP kits were randomly divided into 5 groups on postnatal day 1 (PND1) and received an intravenous injection of a single dose of PBS, or G6D-NAC (2 or 5 mg/kg), or NAC (2 or 5 mg/kg). Neurobehavioral tests, microglia morphology, and neuroinflammation were evaluated at postnatal day 5 (PND5) and day 15 (PND15). RESULTS A single dose of systemic 'long circulating' G6D-NAC showed a significant penetration across the impaired blood-brain-barrier (BBB), delivered NAC specifically to activated microglia, and significantly reduced microglia-mediated neuroinflammation in both the cortex and cerebellum white matter areas. Moreover, G6D-NAC treatment significantly improved neonatal rabbit survival rate and rescued motor function to nearly healthy control levels at least up to 15 days after birth (PND15), while CP kits treated with free NAC died before PND9. CONCLUSIONS Targeted delivery of therapeutics to activated microglia in neonatal brain injury can ameliorate pro-inflammatory microglial responses to injury, promote survival rate, and improve neurological outcomes that can be sustained for a long period. Appropriate manipulation of activated microglia enabled by G6D-NAC can impact the injury significantly beyond inflammation.
Collapse
Affiliation(s)
- Zhi Zhang
- Department of Anesthesiology and Critical Care, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Present address: Department of Natural Sciences, University of Michigan-Dearborn, Dearborn, MI, USA
| | - Yi-An Lin
- Center for Nanomedicine, Wilmer Eye Institute, Johns Hopkins School of Medicine, 400 North Broadway, Baltimore, MD, 21287, USA
| | - Soo-Young Kim
- Center for Nanomedicine, Wilmer Eye Institute, Johns Hopkins School of Medicine, 400 North Broadway, Baltimore, MD, 21287, USA
| | - Lilly Su
- Department of Anesthesiology and Critical Care, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Jinhuan Liu
- Department of Anesthesiology and Critical Care, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Rangaramanujam M Kannan
- Center for Nanomedicine, Wilmer Eye Institute, Johns Hopkins School of Medicine, 400 North Broadway, Baltimore, MD, 21287, USA.
| | - Sujatha Kannan
- Department of Anesthesiology and Critical Care, Johns Hopkins School of Medicine, Baltimore, MD, USA.
- Department of Anesthesiology and Critical Care Medicine, Charlotte Bloomberg Children's Center 6318D, 1800 Orleans Street, Baltimore, MD, 21287, USA.
| |
Collapse
|
15
|
Yap V, Perlman JM. Mechanisms of brain injury in newborn infants associated with the fetal inflammatory response syndrome. Semin Fetal Neonatal Med 2020; 25:101110. [PMID: 32303463 DOI: 10.1016/j.siny.2020.101110] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The fetal inflammatory response syndrome (FIRS) is characterized by umbilical cord inflammation and elevated fetal pro-inflammatory cytokines. Surviving neonates, especially very preterm infants, have increased rates of neonatal morbidity including neurodevelopmental impairment. The mechanism of brain injury in FIRS is complex and may involve "multiple hits." Exposure to in utero inflammation initiates a cascade of the fetal immune response, where pro-inflammatory cytokines can cause direct injury to oligodendrocytes and neurons. Activation of microglia results in further injury to vulnerable pre-myelinating oligodendrocytes and influences the integrity of the fetal and newborn's blood-brain barrier, resulting in further exposure of the brain to developmental insults. Newborns exposed to FIRS are frequently exposed to additional perinatal and postnatal insults that can result in further brain injury. Future directions should include evaluations for new therapeutic interventions aimed at reducing brain injury by dampening FIRS, inhibition of microglial activation, and regeneration of immature oligodendrocytes.
Collapse
Affiliation(s)
- Vivien Yap
- Weill Cornell Medicine - New York Presbyterian Hospital, 525 East 68th Street, Suite N-506, New York, NY, 10065, United States.
| | - Jeffrey M Perlman
- Weill Cornell Medicine - New York Presbyterian Hospital, 525 East 68th Street, Suite N-506, New York, NY, 10065, United States
| |
Collapse
|
16
|
Bertrand SJ, Zhang Z, Patel R, O'Ferrell C, Punjabi NM, Kudchadkar SR, Kannan S. Transient neonatal sleep fragmentation results in long-term neuroinflammation and cognitive impairment in a rabbit model. Exp Neurol 2020; 327:113212. [PMID: 31987835 DOI: 10.1016/j.expneurol.2020.113212] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 01/18/2020] [Accepted: 01/24/2020] [Indexed: 12/29/2022]
Abstract
Sleep fragmentation is an increase in sleep-wake transitions without an overall decrease in total sleep time. Sleep fragmentation is well documented during acute and chronic hospitalization and can result in delirium and memory problems in children. Sleep fragmentation is also often noted in neurodevelopmental disorders. However, it is unclear how sleep fragmentation independent of disease affects brain development and function. We hypothesized that acute sleep fragmentation during the neonatal period in otherwise healthy animals would result in neuroinflammation and would be associated with abnormalities in cognitive development. The orbital shaker method was used to fragment sleep for 72 h in postnatal day 3 New Zealand white rabbit kits (fragmentation group). To control for maternal separation, the sham group was separated from the dam and maintained in the same conditions without undergoing sleep fragmentation. A naïve control group remained with the dam. Kits underwent behavioral testing with novel object recognition and spontaneous alternation T-maze tests at 2-3 weeks post-fragmentation and were sacrificed 3-50 days after fragmentation. Sleep fragmentation resulted in acute and chronic changes in microglial morphology in the hippocampus and cortex, and regional differences in mRNA expression of pro- and anti-inflammatory cytokines at 3, 7 and 50 days post-fragmentation. Impaired novel object recognition and a longer latency in T-maze task completion were noted in the fragmented kits. This was in spite of normalization of sleep architecture noted at 2 months of age in these kits. The results indicate that transient neonatal sleep fragmentation results in short-term and long-term immune alterations in the brain, along with diminished performance in cognitive tasks long-term.
Collapse
Affiliation(s)
- Sarah J Bertrand
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, United States of America
| | - Zhi Zhang
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, United States of America
| | - Ruchit Patel
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, United States of America
| | - Caroline O'Ferrell
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, United States of America
| | - Naresh M Punjabi
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Johns Hopkins University School of Medicine, United States of America
| | - Sapna R Kudchadkar
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, United States of America; Department of Pediatrics, Johns Hopkins University School of Medicine, United States of America; Department of Physical Medicine and Rehabilitation, Johns Hopkins University School of Medicine, United States of America.
| | - Sujatha Kannan
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, United States of America.
| |
Collapse
|
17
|
Lopez-Tello J, Arias-Alvarez M, Gonzalez-Bulnes A, Sferuzzi-Perri AN. Models of Intrauterine growth restriction and fetal programming in rabbits. Mol Reprod Dev 2019; 86:1781-1809. [PMID: 31538701 DOI: 10.1002/mrd.23271] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 09/02/2019] [Indexed: 12/23/2022]
Abstract
Intrauterine growth restriction (IUGR) affects approximately 10% of human pregnancies globally and has immediate and life-long consequences for offspring health. However, the mechanisms underlying the pathogenesis of IUGR and its association with later health and disease outcomes are poorly understood. To address these knowledge gaps, the use of experimental animals is critically important. Since the 50's different environmental, pharmacological, and surgical manipulations have been performed in the rabbit to improve our knowledge of the control of fetal growth, fetal responses to IUGR, and mechanisms by which offspring may be programmed by an adverse gestational environment. The purpose of this review is therefore to summarize the utility of the rabbit as a model for IUGR research. It first summarizes the knowledge of prenatal and postnatal development in the rabbit and how these events relate to developmental milestones in humans. It then describes the methods used to induce IUGR in rabbits and the knowledge gained about the mechanisms determining prenatal and postnatal outcomes of the offspring. Finally, it discusses the application of state of the art approaches in the rabbit, including high-resolution ultrasound, magnetic resonance imaging, and gene targeting, to gain a deeper integrative understanding of the physiological and molecular events governing the development of IUGR. Overall, we hope to engage and inspire investigators to employ the rabbit as a model organism when studying pregnancy physiology so that we may advance our understanding of mechanisms underlying IUGR and its consequences in humans and other mammalian species.
Collapse
Affiliation(s)
- Jorge Lopez-Tello
- Department of Physiology, Development, and Neuroscience, Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
| | - Maria Arias-Alvarez
- Department of Animal Production. Veterinary Faculty, Complutense University of Madrid, Ciudad Universitaria, Madrid, Spain
| | | | - Amanda N Sferuzzi-Perri
- Department of Physiology, Development, and Neuroscience, Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
| |
Collapse
|
18
|
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
|
19
|
The role of maternal immune activation in altering the neurodevelopmental trajectories of offspring: A translational review of neuroimaging studies with implications for autism spectrum disorder and schizophrenia. Neurosci Biobehav Rev 2019; 104:141-157. [DOI: 10.1016/j.neubiorev.2019.06.020] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 05/24/2019] [Accepted: 06/13/2019] [Indexed: 02/01/2023]
|
20
|
Nasr IW, Chun Y, Kannan S. Neuroimmune responses in the developing brain following traumatic brain injury. Exp Neurol 2019; 320:112957. [PMID: 31108085 DOI: 10.1016/j.expneurol.2019.112957] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Revised: 05/10/2019] [Accepted: 05/15/2019] [Indexed: 12/26/2022]
Abstract
Traumatic brain injury (TBI) is one of the leading causes of both acute and long-term morbidity in the pediatric population, leading to a substantial, long-term socioeconomic burden. Despite the increase in the amount of pre-clinical and clinical research, treatment options for TBI rely heavily on supportive care with very limited targeted interventions that improve the acute and chronic sequelae of TBI. Other than injury prevention, not much can be done to limit the primary injury, which consists of tissue damage and cellular destruction. Secondary injury is the result of the ongoing complex inflammatory pathways that further exacerbate tissue damage, resulting in the devastating chronic outcomes of TBI. On the other hand, some level of inflammation is essential for neuronal regeneration and tissue repair. In this review article we discuss the various stages of the neuroimmune response in the immature, pediatric brain in the context of normal maturation and development of the immune system. The developing brain has unique features that distinguish it from the adult brain, and the immune system plays an integral role in CNS development. Those features could potentially make the developing brain more susceptible to worse outcomes, both acutely and in the long-term. The neuroinflammatory reaction which is triggered by TBI can be described as a highly intricate interaction between the cells of the innate and the adaptive immune systems. The innate immune system is triggered by non-specific danger signals that are released from damaged cells and tissues, which in turn leads to neutrophil infiltration, activation of microglia and astrocytes, complement release, as well as histamine release by mast cells. The adaptive immune response is subsequently activated leading to the more chronic effects of neuroinflammation. We will also discuss current attempts at modulating the TBI-induced neuroinflammatory response. A better understanding of the role of the immune system in normal brain development and how immune function changes with age is crucial for designing therapies to appropriately target the immune responses following TBI in order to enhance repair and plasticity.
Collapse
Affiliation(s)
- Isam W Nasr
- Pediatric Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States of America
| | - Young Chun
- Pediatric Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States of America
| | - Sujatha Kannan
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States of America.
| |
Collapse
|
21
|
Xue H, Xu Y, Wang S, Wu ZY, Li XY, Zhang YH, Niu JY, Gao QS, Zhao P. Sevoflurane post-conditioning alleviates neonatal rat hypoxic-ischemic cerebral injury via Ezh2-regulated autophagy. DRUG DESIGN DEVELOPMENT AND THERAPY 2019; 13:1691-1706. [PMID: 31190748 PMCID: PMC6528650 DOI: 10.2147/dddt.s197325] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 03/04/2019] [Indexed: 12/12/2022]
Abstract
Background: When neonatal rats suffer hypoxic-ischemic brain injury (HIBI), autophagy is over-activated in the hippocampus, and inhibition of autophagy provides neuroprotection. The aim of this study was to investigate the possible roles of autophagy and Ezh2-regulated Pten/Akt/mTOR pathway in sevoflurane post-conditioning (SPC)-mediated neuroprotection against HIBI in neonatal rats. Methods: Seven-day-old Sprague–Dawley rats underwent left common artery ligation followed by 2 h hypoxia as described in the Rice–Vannucci model. The roles of autophagy and the Ezh2-regulated Pten/Akt/mTOR signaling pathway in the neuroprotection conferred by SPC were examined by left-side intracerebroventricular injection with the autophagy activator rapamycin and the Ezh2 inhibitor GSK126. Results: SPC was neuroprotective against HIBI through the inhibition of over-activated autophagy in the hippocampus as characterized by the rapamycin-induced reversal of neuronal density, neuronal morphology, cerebral morphology, and the expression of the autophagy markers, LC3B-II and Beclin1. SPC significantly increased the expression of Ezh2, H3K27me3, pAkt, and mTOR and decreased the expression of Pten induced by HI. The Ezh2 inhibitor, GSK126, significantly reversed the SPC-induced changes in expression of H3K27me3, Pten, pAkt, mTOR, LC3B-II, and Beclin1. Ezh2 inhibition also reversed SPC-mediated attenuation of neuronal loss and behavioral improvement in the Morris water maze. Conclusion: These results indicate that SPC inhibits excessive autophagy via the regulation of Pten/Akt/mTOR signaling by Ezh2 to confer neuroprotection against HIBI in neonatal rats.
Collapse
Affiliation(s)
- Hang Xue
- Department of Anesthesiology, Shengjing Hospital, China Medical University, Shenyang 110004, People's Republic of China
| | - Ying Xu
- Department of Anesthesiology, Shengjing Hospital, China Medical University, Shenyang 110004, People's Republic of China
| | - Shuo Wang
- Department of Anesthesiology, Shengjing Hospital, China Medical University, Shenyang 110004, People's Republic of China
| | - Zi-Yi Wu
- Department of Anesthesiology, Shengjing Hospital, China Medical University, Shenyang 110004, People's Republic of China
| | - Xing-Yue Li
- Department of Anesthesiology, Shengjing Hospital, China Medical University, Shenyang 110004, People's Republic of China
| | - Ya-Han Zhang
- Department of Anesthesiology, Shengjing Hospital, China Medical University, Shenyang 110004, People's Republic of China
| | - Jia-Yuan Niu
- Department of Anesthesiology, Shengjing Hospital, China Medical University, Shenyang 110004, People's Republic of China
| | - Qiu-Shi Gao
- Department of Anesthesiology, Shengjing Hospital, China Medical University, Shenyang 110004, People's Republic of China
| | - Ping Zhao
- Department of Anesthesiology, Shengjing Hospital, China Medical University, Shenyang 110004, People's Republic of China
| |
Collapse
|
22
|
Gisslen T, Singh G, Georgieff MK. Fetal inflammation is associated with persistent systemic and hippocampal inflammation and dysregulation of hippocampal glutamatergic homeostasis. Pediatr Res 2019; 85:703-710. [PMID: 30745569 PMCID: PMC6435426 DOI: 10.1038/s41390-019-0330-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2018] [Revised: 12/29/2018] [Accepted: 02/01/2019] [Indexed: 12/22/2022]
Abstract
BACKGROUND Inflammation is a major cause of preterm birth and often results in a fetal inflammatory response syndrome (FIRS). Preterm infants with FIRS have a higher childhood incidence of neurodevelopmental disability than preterm infants without FIRS. The mechanisms connecting FIRS to neurodevelopmental disability in formerly preterm infants are not fully understood, but the effect on premature gray matter may have an important role. METHODS Fetal rats were exposed to intra-amniotic (i.a.) LPS 2 days prior to birth to model FIRS. On postnatal day 7, expression of inflammatory mediators was measured in the liver, lung, and brain. Activation of microglia and expression of glutamatergic receptor subunits and transporters were measured in the hippocampus and cortex. RESULTS LPS caused persistent systemic inflammatory mediators gene expression. In the brain, there was corresponding activation of microglia in the hippocampus and cortex. Expression of inflammatory mediators persisted in the hippocampus, but not the cortex, and was associated with altered glutamatergic receptor subunits and transporters. CONCLUSION Hippocampal inflammation and dysregulation of glutamate metabolism persisted well into the postnatal period following i.a. LPS. Poor neurodevelopmental outcomes after FIRS in preterm infants may result in part through glutamatergically driven gray matter injury to the neonatal hippocampus.
Collapse
Affiliation(s)
- Tate Gisslen
- Division of Neonatology, Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA.
| | - Garima Singh
- Division of Neonatology, Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
| | - Michael K Georgieff
- Division of Neonatology, Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
23
|
Ying YQ, Yan XQ, Jin SJ, Liang Y, Hou L, Niu WT, Luo XP. Inhibitory Effect of LPS on the Proliferation of Oligodendrocyte Precursor Cells through the Notch Signaling Pathway in Intrauterine Infection-induced Rats. Curr Med Sci 2018; 38:840-846. [PMID: 30341518 DOI: 10.1007/s11596-018-1951-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 09/04/2018] [Indexed: 10/28/2022]
Abstract
Periventricular white matter injury (PWMI) is very common in survivors of premature birth, and the final outcomes are a reduction in myelinated neurons leading to white matter hypomyelination. How and (or) why the oligodendrocyte lineage develops abnormally and myelination is reduced is a hot topic in the field. This study focuses on the effect of intrauterine inflammation on the proliferation of oligodendrocyte lineage cells and the underlying mechanisms. Lipopolysaccharide (LPS) (300 μg/kg) was intraperitoneally injected into pregnant Sprague-Dawley rats at embryonic days 19 and 20 to establish a rat model of intrauterine infection-induced white matter injury. Corpus callosum tissues were collected at postnatal day 14 (P14) to quantify the number of oligodendrocytes, the number and proliferation of oligodendrocyte precursor cells (OPCs), and the expression of myelin proteins (MBP and PLP). Furthermore, the expression of Wnt and Notch signaling-related proteins was analyzed. The results showed that the number of oligodendrocytes in the corpus callosum tissues of LPS-treated rats was reduced, and the expression levels of myelinating proteins were down-regulated. Further analysis showed that the Notch signaling pathway was down-regulated in the LPStreated group. These results indicate that intrauterine LPS may inhibit the proliferation of OPCs by down-regulating the Notch rather than the Wnt signaling pathway, leading to hypomyelination of white matter.
Collapse
Affiliation(s)
- Yan-Qin Ying
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xue-Qin Yan
- Department of Children Healthcare, Zhongshan Boai Hospital Affiliated to Southern Medical University, Zhongshan, 528403, China
| | - Sheng-Juan Jin
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yan Liang
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Ling Hou
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Wan-Ting Niu
- VA Boston Healthcare System, Department of Orthopedics, Brigham and Women's Hospital, Harvard Medical School, Boston, 02130, USA
| | - Xiao-Ping Luo
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
24
|
Sharma A, Porterfield JE, Smith E, Sharma R, Kannan S, Kannan RM. Effect of mannose targeting of hydroxyl PAMAM dendrimers on cellular and organ biodistribution in a neonatal brain injury model. J Control Release 2018; 283:175-189. [PMID: 29883694 PMCID: PMC6091673 DOI: 10.1016/j.jconrel.2018.06.003] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 05/21/2018] [Accepted: 06/02/2018] [Indexed: 01/02/2023]
Abstract
Neurotherapeutics for the treatment of central nervous system (CNS) disorders must overcome challenges relating to the blood-brain barrier (BBB), brain tissue penetration, and the targeting of specific cells. Neuroinflammation mediated by activated microglia is a major hallmark of several neurological disorders, making these cells a desirable therapeutic target. Building on the promise of hydroxyl-terminated generation four polyamidoamine (PAMAM) dendrimers (D4-OH) for penetrating the injured BBB and targeting activated glia, we explored if conjugation of targeting ligands would enhance and modify brain and organ uptake. Since mannose receptors [cluster of differentiation (CD) 206] are typically over-expressed on injured microglia, we conjugated mannose to the surface of multifunctional D4-OH using highly efficient, atom-economical, and orthogonal Cu(I)-catalyzed alkyne-azide cycloaddition (CuAAC) click chemistry and evaluated the effect of mannose conjugation on the specific cell uptake of targeted and non-targeted dendrimers both in vitro and in vivo. In vitro results indicate that the conjugation of mannose as a targeting ligand significantly changes the mechanism of dendrimer internalization, giving mannosylated dendrimer a preference for mannose receptor-mediated endocytosis as opposed to non-specific fluid phase endocytosis. We further investigated the brain uptake and biodistribution of targeted and non-targeted fluorescently labeled dendrimers in a maternal intrauterine inflammation-induced cerebral palsy (CP) rabbit model using quantification methods based on fluorescence spectroscopy and confocal microscopy. We found that the conjugation of mannose modified the distribution of D4-OH throughout the body in this neonatal rabbit CP model without lowering the amount of dendrimer delivered to injured glia in the brain, even though significantly higher glial uptake was not observed in this model. Mannose conjugation to the dendrimer modifies the dendrimer's interaction with cells, but does not minimize its inherent inflammation-targeting abilities.
Collapse
Affiliation(s)
- Anjali Sharma
- Center for Nanomedicine, Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Joshua E Porterfield
- Center for Nanomedicine, Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Elizabeth Smith
- Center for Nanomedicine, Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Rishi Sharma
- Center for Nanomedicine, Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Sujatha Kannan
- Center for Nanomedicine, Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; Hugo W. Moser Research Institute at Kennedy Krieger, Inc., Baltimore, MD 21205, USA; Kennedy Krieger Institute - Johns Hopkins University for Cerebral Palsy Research Excellence, Baltimore, MD 21218, USA
| | - Rangaramanujam M Kannan
- Center for Nanomedicine, Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA; Hugo W. Moser Research Institute at Kennedy Krieger, Inc., Baltimore, MD 21205, USA; Kennedy Krieger Institute - Johns Hopkins University for Cerebral Palsy Research Excellence, Baltimore, MD 21218, USA.
| |
Collapse
|
25
|
Cerebellar injury and impaired function in a rabbit model of maternal inflammation induced neonatal brain injury. Neurobiol Learn Mem 2018; 165:106901. [PMID: 30016703 DOI: 10.1016/j.nlm.2018.07.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 07/09/2018] [Accepted: 07/13/2018] [Indexed: 01/01/2023]
Abstract
Cerebellum is involved in higher cognitive functions and plays important roles in neurological disorders. Cerebellar injury has been detected frequently in patients with preterm birth resulting in cognitive dysfunction later in life. Maternal infection and inflammation is associated with preterm birth and in neonatal brain injury. We have previously shown that intrauterine lipopolysaccharide (LPS) exposure induces white matter injury and microglial activation in the cerebral white matter tracts of neonatal rabbits, resulting in motor deficits consistent with the clinical findings of cerebral palsy (CP). Here we investigated whether intrauterine LPS exposure induced cerebellar inflammation and functional impairment. Timed-pregnant New Zealand white rabbits underwent a laparotomy on gestational day 28 (G28) and LPS (3200 EU, endotoxin group) was injected along the wall of the uterus as previously described. Controls did not receive surgical intervention. Kits born to control and endotoxin treated dams were euthanized on postnatal day (PND)1 (3 days post-injury) or PND5 (7 days post-injury) and cerebellum evaluated for presence of inflammation. The microglial morphology in cerebellar white matter areas was analyzed using Neurolucida and Neurolucida Explorer. mRNA expression of inflammatory cytokines was quantified by real-time-PCR. We found that intrauterine exposure to LPS induced intensive microglial activation in cerebellar white matter areas, as evidenced by increased numbers of activated microglia and morphological changes (amoeboid soma and retracted processes) that was accompanied by significant increases in pro-inflammatory cytokines. The Purkinje cell layer was less developed in endotoxin exposed kits than healthy controls. In kits that survived to PND 60, soma size and cell density of Purkinje cells were significantly decreased in endotoxin exposed kits compared to controls. The findings of altered Purkinje cell morphology were consistent with impaired cerebellar function as tested by eye-blink conditioning at 1 month of age. The results indicate that the cerebellum is vulnerable to perinatal insults and that therapies targeting cerebellar inflammation and injury may help in improving outcomes and function.
Collapse
|
26
|
Sharma R, Sharma A, Kambhampati SP, Reddy RR, Zhang Z, Cleland JL, Kannan S, Kannan RM. Scalable synthesis and validation of PAMAM dendrimer- N-acetyl cysteine conjugate for potential translation. Bioeng Transl Med 2018; 3:87-101. [PMID: 30065965 PMCID: PMC6063872 DOI: 10.1002/btm2.10094] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 04/25/2018] [Accepted: 04/26/2018] [Indexed: 01/13/2023] Open
Abstract
Dendrimer-N-acetyl cysteine (D-NAC) conjugate has shown significant promise in multiple preclinical models of brain injury and is undergoing clinical translation. D-NAC is a generation-4 hydroxyl-polyamidoamine dendrimer conjugate where N-acetyl cysteine (NAC) is covalently bound through disulfide linkages on the surface of the dendrimer. It has shown remarkable potential to selectively target and deliver NAC to activated microglia and astrocytes at the site of brain injury in several animal models, producing remarkable improvements in neurological outcomes at a fraction of the free drug dose. Here we present a highly efficient, scalable, greener, well-defined route to the synthesis of D-NAC, and validate the structure, stability and activity to define the benchmarks for this compound. This newly developed synthetic route has significantly reduced the synthesis time from three weeks to one week, uses industry-friendly solvents/reagents, and involves simple purification procedures, potentially enabling efficient scale up.
Collapse
Affiliation(s)
- Rishi Sharma
- Center for Nanomedicine, Department of OphthalmologyWilmer Eye Institute Johns Hopkins University School of MedicineBaltimoreMD21287
| | - Anjali Sharma
- Center for Nanomedicine, Department of OphthalmologyWilmer Eye Institute Johns Hopkins University School of MedicineBaltimoreMD21287
| | - Siva P. Kambhampati
- Center for Nanomedicine, Department of OphthalmologyWilmer Eye Institute Johns Hopkins University School of MedicineBaltimoreMD21287
| | - Rajsekar Rami Reddy
- Center for Nanomedicine, Department of OphthalmologyWilmer Eye Institute Johns Hopkins University School of MedicineBaltimoreMD21287
| | - Zhi Zhang
- Dept. of Anesthesiology and Critical Care MedicineJohns Hopkins University School of MedicineBaltimoreMD21287
| | | | - Sujatha Kannan
- Center for Nanomedicine, Department of OphthalmologyWilmer Eye Institute Johns Hopkins University School of MedicineBaltimoreMD21287
- Dept. of Anesthesiology and Critical Care MedicineJohns Hopkins University School of MedicineBaltimoreMD21287
- Hugo W. Moser Research Institute at Kennedy Krieger, Inc.BaltimoreMD21205
- Kennedy Krieger Institute – Johns Hopkins University for Cerebral Palsy Research ExcellenceBaltimoreMD21287
| | - Rangaramanujam M. Kannan
- Center for Nanomedicine, Department of OphthalmologyWilmer Eye Institute Johns Hopkins University School of MedicineBaltimoreMD21287
- Hugo W. Moser Research Institute at Kennedy Krieger, Inc.BaltimoreMD21205
- Kennedy Krieger Institute – Johns Hopkins University for Cerebral Palsy Research ExcellenceBaltimoreMD21287
- Dept.of Chemical and Biomolecular EngineeringJohns Hopkins UniversityBaltimoreMD21218
| |
Collapse
|
27
|
Guadagno E, Presta I, Maisano D, Donato A, Pirrone CK, Cardillo G, Corrado SD, Mignogna C, Mancuso T, Donato G, Del Basso De Caro M, Malara N. Role of Macrophages in Brain Tumor Growth and Progression. Int J Mol Sci 2018; 19:ijms19041005. [PMID: 29584702 PMCID: PMC5979398 DOI: 10.3390/ijms19041005] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2018] [Revised: 03/10/2018] [Accepted: 03/23/2018] [Indexed: 12/16/2022] Open
Abstract
The role of macrophages in the growth and the progression of tumors has been extensively studied in recent years. A large body of data demonstrates that macrophage polarization plays an essential role in the growth and progression of brain tumors, such as gliomas, meningiomas, and medulloblastomas. The brain neoplasm cells have the ability to influence the polarization state of the tumor associated macrophages. In turn, innate immunity cells have a decisive role through regulation of the acquired immune response, but also through humoral cross-talking with cancer cells in the tumor microenvironment. Neoangiogenesis, which is an essential element in glial tumor progression, is even regulated by the tumor associated macrophages, whose activity is linked to other factors, such as hypoxia. In addition, macrophages play a decisive role in establishing the entry into the bloodstream of cancer cells. As is well known, the latter phenomenon is also present in brain tumors, even if they only rarely metastasize. Looking ahead in the future, we can imagine that characterizing the relationships between tumor and tumor associated macrophage, as well as the study of circulating tumor cells, could give us useful tools in prognostic evaluation and therapy. More generally, the study of innate immunity in brain tumors can boost the development of new forms of immunotherapy.
Collapse
Affiliation(s)
- Elia Guadagno
- Department of Advanced Biomedical Sciences-Pathology Section, University of Naples "Federico II"-via Pansini 5, 80131 Naples, Italy.
| | - Ivan Presta
- Department of Health Sciences, University of Catanzaro "Magna Græcia"-viale Europa, 88100 Catanzaro, Italy.
| | - Domenico Maisano
- Department of Health Sciences, University of Catanzaro "Magna Græcia"-viale Europa, 88100 Catanzaro, Italy.
| | - Annalidia Donato
- Department of Medical and Surgical Sciences-University of Catanzaro "Magna Graecia"-viale Europa, 88100 Catanzaro, Italy.
| | - Caterina Krizia Pirrone
- Department of Health Sciences, University of Catanzaro "Magna Græcia"-viale Europa, 88100 Catanzaro, Italy.
| | - Gabriella Cardillo
- Department of Health Sciences, University of Catanzaro "Magna Græcia"-viale Europa, 88100 Catanzaro, Italy.
| | - Simona Domenica Corrado
- Department of Health Sciences, University of Catanzaro "Magna Græcia"-viale Europa, 88100 Catanzaro, Italy.
| | - Chiara Mignogna
- Department of Health Sciences, University of Catanzaro "Magna Græcia"-viale Europa, 88100 Catanzaro, Italy.
| | - Teresa Mancuso
- Department of Medical and Surgical Sciences-University of Catanzaro "Magna Graecia"-viale Europa, 88100 Catanzaro, Italy.
| | - Giuseppe Donato
- Department of Health Sciences, University of Catanzaro "Magna Græcia"-viale Europa, 88100 Catanzaro, Italy.
| | - Marialaura Del Basso De Caro
- Department of Advanced Biomedical Sciences-Pathology Section, University of Naples "Federico II"-via Pansini 5, 80131 Naples, Italy.
| | - Natalia Malara
- Department of Clinical and Experimental Medicine-University of Catanzaro "Magna Graecia"-viale Europa, 88100 Catanzaro, Italy.
| |
Collapse
|
28
|
Shi Z, Vasquez-Vivar J, Luo K, Yan Y, Northington F, Mehrmohammadi M, Tan S. Ascending Lipopolysaccharide-Induced Intrauterine Inflammation in Near-Term Rabbits Leading to Newborn Neurobehavioral Deficits. Dev Neurosci 2018; 40:534-546. [PMID: 31163416 PMCID: PMC9873358 DOI: 10.1159/000499960] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Accepted: 03/26/2019] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND Chorioamnionitis from ascending bacterial infection through the endocervix is a potential risk factor for cerebral palsy. Tetrahydrobiopterin, an essential cofactor for nitric oxide synthase (NOS) and amino acid hydroxylases, when augmented in the fetal brain, prevents some of the cerebral palsy-like deficits in a rabbit hypoxia-ischemia model. OBJECTIVES To study the effect of lipopolysaccharide (LPS)-induced intrauterine inflammation in preterm gestation on motor deficits in the newborn, and whether biosynthesis of tetrahydrobiopterin or inflammatory mediators is affected in the fetal brain. METHODS Pregnant rabbits at 28 days gestation (89% term) were administered either saline or LPS into both endocervical openings. One group underwent spontaneous delivery, and neurobehavioral tests were performed at postnatal day (P) 1 and P11, with some kits being sacrificed at P1 for histological analysis. Another group underwent Cesarean section 24 h after LPS administration. Gene sequences for rabbit biosynthetic enzymes of tetra-hydrobiopterin pathways were determined and analyzed in addition to cytokines, using quantitative real-time polymerase chain reaction. RESULTS Exposure to 200 μg/kg/mL LPS caused a locomotion deficit and mild hypertonia at P1. By P11, most animals turned into normal-appearing kits. There was no difference in neuronal cell death in the caudate between hypertonic and nonhypertonic kits at P1 (n = 3-5 in each group). Fetal brain GTP cyclohydrolase I was increased, whereas sepiapterin reductase and 6-pyruvoyltetrahydropterin synthase were decreased, 24 h after LPS administration. Neuronal NOS was also increased. Regardless of the position in the uterus or the brain region, expression of TNF-α and TGF-β was decreased, whereas that of IL-1β, IL-6, and IL-8 was increased (n = 3-4 in each group). CONCLUSIONS This is the first study using an ascending LPS-induced intrauterine inflammation model in rabbits, showing mostly transient hypertonia and mainly locomotor deficits in the kits. Not all proinflammatory cytokines are increased in the fetal brain following LPS administration. Changes in key tetrahydro-biopterin biosynthetic enzymes possibly indicate different effects of the inflammatory insult.
Collapse
Affiliation(s)
- Zhongjie Shi
- Department of Pediatrics, Children’s Hospital of Michigan, Wayne State University, Detroit, MI
| | | | - Kehuan Luo
- Department of Pediatrics, Children’s Hospital of Michigan, Wayne State University, Detroit, MI
| | - Yan Yan
- Department of Biomedical Engineering, Wayne State University, Detroit, MI
| | | | | | - Sidhartha Tan
- Department of Pediatrics, Children’s Hospital of Michigan, Wayne State University, Detroit, MI
| |
Collapse
|