122 results on '"Lauren L. Jantzie"'
Search Results
52. Contributors
- Author
-
Erica T. Akhter, Tenpei Akita, Sana Al Awabdh, Francisco J. Alvarez, Yehezkel Ben-Ari, Mohammad Iqbal H. Bhuiyan, Maria Bolla, Laura Cancedda, Shao-Rui Chen, Quentin Chevy, Bice Chini, Marie-Pascale Côté, Arthur W. English, Diana C. Ferrari, Atsuo Fukuda, Nouchine Hadjikhani, Knut Holthoff, Saiyun Hou, Huachen Huang, Lori L. Isom, Lauren L. Jantzie, Anass Jawhari, Frances E. Jensen, Tong Jiang, Nicholas J. Justice, Shilpa D. Kadam, Werner Kilb, Knut Kirmse, Eric Lemonnier, Sabine Lévi, Wolfgang Liedtke, Olaya Llano, Anastasia Ludwig, Jamie Maguire, Vivek Mahadevan, Igor Medina, Jessie C. Newville, Heather A. O'Malley, Akosua Y. Oppong, Hui-Lin Pan, Lucie I. Pisella, Jean Christophe Poncer, Davide Pozzi, Jessica C. Pressey, Denis Ravel, Claudio Rivera, Shenandoah Robinson, Annalisa Savardi, Clémence Simonnet, Yeri J. Song, Brennan J. Sullivan, Dandan Sun, Taraneh Taheri, Delia M. Talos, Miho Watanabe, Melanie A. Woodin, Li Yang, Michele Yeo, Zhongling Zhang, and Ilias Ziogas
- Published
- 2020
- Full Text
- View/download PDF
53. Placenta and perinatal brain injury: the gateway to individualized therapeutics and precision neonatal medicine
- Author
-
Lauren L, Jantzie and Shenandoah, Robinson
- Subjects
Pregnancy ,Brain Injuries ,Placenta ,Hypoxia-Ischemia, Brain ,Infant, Newborn ,Brain ,Humans ,Female ,Precision Medicine ,Erythropoietin ,Magnetic Resonance Imaging - Abstract
Wu and colleagues analyzed the placental pathology from a subset of the neonates in the NEATO trial who had reports available and correlated the placental pathology findings with outcomes. This study highlights the importance of placental pathology, and its potential to bring precision medicine to critically-ill neonates. Placental pathology will likely aid stratification of neonates for clinical trials and accelerate progress for neurorepair.
- Published
- 2019
54. Prenatal opioid exposure: The next neonatal neuroinflammatory disease
- Author
-
Tracylyn R. Yellowhair, Yuma Kitase, Ludmila N. Bakhireva, Jessie Newville, Lorraine Milio, Jessie R. Maxwell, Gwendolyn Gerner, Jonathan L. Brigman, Andrea M. Allan, Lauren L. Jantzie, Shenandoah Robinson, Nethra K. Madurai, Aylin Tekes, Frances J. Northington, and Sindhu Ramachandra
- Subjects
0301 basic medicine ,Male ,Neuroimmunomodulation ,Immunology ,Central nervous system ,Disease ,Bioinformatics ,Article ,Rats, Sprague-Dawley ,03 medical and health sciences ,Behavioral Neuroscience ,0302 clinical medicine ,Pregnancy ,medicine ,Animals ,Neuroinflammation ,Inflammation ,Endocrine and Autonomic Systems ,business.industry ,Opioid use disorder ,medicine.disease ,Associative learning ,Rats ,Analgesics, Opioid ,Disease Models, Animal ,030104 developmental biology ,medicine.anatomical_structure ,Diffusion Tensor Imaging ,Opioid ,Prenatal Exposure Delayed Effects ,Biomarker (medicine) ,Female ,business ,030217 neurology & neurosurgery ,Methadone ,medicine.drug - Abstract
The rates of opioid use disorder during pregnancy have more than quadrupled in the last decade, resulting in numerous infants suffering exposure to opioids during the perinatal period, a critical period of central nervous system (CNS) development. Despite increasing use, the characterization and definition of the molecular and cellular mechanisms of the long-term neurodevelopmental impacts of opioid exposure commencing in utero remains incomplete. Thus, in consideration of the looming public health crisis stemming from the multitude of infants with prenatal opioid exposure entering school age, we undertook an investigation of the effects of perinatal methadone exposure in a novel preclinical model. Specifically, we examined the effects of opioids on the developing brain to elucidate mechanisms of putative neural cell injury, to identify diagnostic biomarkers and to guide clinical studies of outcome and follow-up. We hypothesized that methadone would induce a pronounced inflammatory profile in both dams and their pups, and be associated with immune system dysfunction, sustained CNS injury, and altered cognition and executive function into adulthood. This investigation was conducted using a combination of cellular, molecular, biochemical, and clinically translatable biomarker, imaging and cognitive assessment platforms. Data reveal that perinatal methadone exposure increases inflammatory cytokines in the neonatal peripheral circulation, and reprograms and primes the immune system through sustained peripheral immune hyperreactivity. In the brain, perinatal methadone exposure not only increases chemokines and cytokines throughout a crucial developmental period, but also alters microglia morphology consistent with activation, and upregulates TLR4 and MyD88 mRNA. This increase in neuroinflammation coincides with reduced myelin basic protein and altered neurofilament expression, as well as reduced structural coherence and significantly decreased fractional anisotropy on diffusion tensor imaging. In addition to this microstructural brain injury, adult rats exposed to methadone in the perinatal period have significant impairment in associative learning and executive control as assessed using touchscreen technology. Collectively, these data reveal a distinct systemic and neuroinflammatory signature associated with prenatal methadone exposure, suggestive of an altered CNS microenvironment, dysregulated developmental homeostasis, complex concurrent neural injury, and imaging and cognitive findings consistent with clinical literature. Further investigation is required to define appropriate therapies targeted at the neural injury and improve the long-term outcomes for this exceedingly vulnerable patient population.
- Published
- 2019
55. LFA-1 antagonist (BIRT377) similarly reverses peripheral neuropathic pain in male and female mice with underlying sex divergent peripheral immune proinflammatory phenotypes
- Author
-
Mara A. Havard, Erin D. Milligan, Jacob E. Sanchez, Harrison T. West, Nikolaos Mellios, Nathan W. Harris, Arden G. Vanderwall, Jeffrey P. Norenberg, Shahani Noor, Melody S. Sun, Carsten R. Wagner, Lauren L. Jantzie, and Monique Nysus
- Subjects
neuroimmune ,glia ,peripheral immune ,Lymphocyte ,medicine.medical_treatment ,T cell ,Immunology ,T cells ,Neuropathic pain ,Article ,Proinflammatory cytokine ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Medicine ,business.industry ,FOXP3 ,Allodynia ,medicine.anatomical_structure ,Cytokine ,Neurology ,Peripheral nerve injury ,Neurology (clinical) ,medicine.symptom ,business ,030217 neurology & neurosurgery ,030215 immunology - Abstract
Aim The majority of preclinical studies investigating aberrant glial-neuroimmune actions underlying neuropathic pain have focused on male rodent models. Recently, studies have shown peripheral immune cells play a more prominent role than glial cells in mediating pathological pain in females. Here, we compared the onset and duration of allodynia in males and females, and the anti-allodynic action of a potentially novel therapeutic drug (BIRT377) that not only antagonizes the action of lymphocyte function-associated antigen-1 (LFA-1) to reduce cell migration in the periphery, but may also directly alter the cellular inflammatory bias. Methods Male and female mice were subjected to peripheral nerve injury chronic constriction injury (CCI) applying two methods, using either 4-0 or 5-0 chromic gut suture material, to examine potential sex differences in the onset, magnitude and duration of allodynia. Hindpaw sensitivity before and after CCI and application of intravenous BIRT377 was assessed. Peripheral and spinal tissues were analyzed for protein (multiplex electrochemiluminescence technology) and mRNA expression (quantitative real-time PCR). The phenotype of peripheral T cells was determined using flow cytometry. Results Sex differences in proinflammatory CCL2 and IL-1β and the anti-inflammatory IL-10 were observed from a set of cytokines analyzed. A profound proinflammatory T cell (Th17) response in the periphery and spinal cord was also observed in neuropathic females. BIRT377 reversed pain, reduced IL-1β and TNF, and increased IL-10 and transforming growth factor (TGF)-β1, also an anti-inflammatory cytokine, in both sexes. However, female-derived T cell cytokines are transcriptionally regulated by BIRT377, as demonstrated by reducing proinflammatory IL-17A production with concurrent increases in IL-10, TGF-β1 and the anti-inflammatory regulatory T cell-related factor, FOXP3. Conclusion This study supports that divergent peripheral immune and neuroimmune responses during neuropathy exists between males and females. Moreover, the modulatory actions of BIRT377 on T cells during neuropathy are predominantly specific to females. These data highlight the necessity of including both sexes for studying drug efficacy and mechanisms of action in preclinical studies and clinical trials.
- Published
- 2019
- Full Text
- View/download PDF
56. Chorioamnionitis Precipitates Perinatal Alterations of Heme-Oxygenase-1 (HO-1) Homeostasis in the Developing Rat Brain
- Author
-
Yuma Kitase, Shenandoah Robinson, Lauren L. Jantzie, Christopher Burkhardt, Sindhu Ramachandra, Vikram Vasan, and Maide Ozen
- Subjects
TfR1 ,Placenta ,T-Lymphocytes ,medicine.disease_cause ,Chorioamnionitis ,neuroinflammation ,Pregnancy ,Homeostasis ,Medicine ,Biology (General) ,Spectroscopy ,chemistry.chemical_classification ,neurodevelopment ,Brain ,General Medicine ,Computer Science Applications ,Chemistry ,Premature Birth ,Female ,medicine.symptom ,medicine.medical_specialty ,QH301-705.5 ,Iron ,HO-1 ,neural-immune ,Inflammation ,Peripheral blood mononuclear cell ,Article ,Catalysis ,Inorganic Chemistry ,Internal medicine ,Receptors, Transferrin ,Animals ,RNA, Messenger ,Physical and Theoretical Chemistry ,QD1-999 ,Molecular Biology ,Neuroinflammation ,business.industry ,Organic Chemistry ,medicine.disease ,Rats ,Heme oxygenase ,Oxidative Stress ,Endocrinology ,chemistry ,Transferrin ,Brain Injuries ,peripheral immune activation ,Heme Oxygenase (Decyclizing) ,perinatal brain injury ,business ,Heme Oxygenase-1 ,Oxidative stress - Abstract
Chorioamnionitis (CHORIO), placental insufficiency, and preterm birth are well-known antecedents of perinatal brain injury (PBI). Heme-oxygenase-1 (HO-1) is an important inducible enzyme in oxidative and inflammatory conditions. In the brain, HO-1 and the iron regulatory receptor, transferrin receptor-1 (TfR1), are known to be involved in iron homeostasis, oxidative stress, and cellular adaptive mechanisms. However, the role of HO pathway in the pathophysiology of PBI has not been previously studied. In this study, we set out to define the ontogeny of the HO pathway in the brain and determine if CHORIO changed its normal developmental regulation. We also aimed to determine the role of HO-1/TfR1 in CHORIO-induced neuroinflammation and peripheral inflammation in a clinically relevant rat model of PBI. We show that HO-1, HO-2, and TfR1 expression are developmentally regulated in the brain during the perinatal period. CHORIO elevates HO-1 and TfR1 mRNA expression in utero and in the early postnatal period and results in sustained increase in HO-1/TfR1 ratios in the brain. This is associated with neuroinflammatory and peripheral immune phenotype supported by a significant increase in brain mononuclear cells and peripheral blood double negative T cells suggesting a role of HO-1/TfR1 pathway dysregulation in CHORIO-induced neuroinflammation.
- Published
- 2021
- Full Text
- View/download PDF
57. Microstructural and microglial changes after repetitive mild traumatic brain injury in mice
- Author
-
Jesse L. Winer, Rebekah Mannix, Shenandoah Robinson, Justin Berkner, Jesse L. Denson, Christopher V. Anstine, Jianhua Qiu, Lauren L. Jantzie, Laurel O. Sillerud, Jessie R. Maxwell, Jacqueline Berglass, Yirong Yang, and William P. Meehan
- Subjects
0301 basic medicine ,Pathology ,medicine.medical_specialty ,medicine.diagnostic_test ,Traumatic brain injury ,Magnetic resonance imaging ,Stereology ,Inflammation ,medicine.disease ,Microgliosis ,Pathophysiology ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,030104 developmental biology ,0302 clinical medicine ,Closed head injury ,medicine ,medicine.symptom ,Psychology ,Pathological ,030217 neurology & neurosurgery - Abstract
Traumatic brain injury (TBI) is a major public health issue, with recently increased awareness of the potential long-term sequelae of repetitive injury. Although TBI is common, objective diagnostic tools with sound neurobiological predictors of outcome are lacking. Indeed, such tools could help to identify those at risk for more severe outcomes after repetitive injury and improve understanding of biological underpinnings to provide important mechanistic insights. We tested the hypothesis that acute and subacute pathological injury, including the microgliosis that results from repeated mild closed head injury (rmCHI), is reflected in susceptibility-weighted magnetic resonance imaging and diffusion-tensor imaging microstructural abnormalities. Using a combination of high-resolution magnetic resonance imaging, stereology, and quantitative PCR, we studied the pathophysiology of male mice that sustained seven consecutive mild traumatic brain injuries over 9 days in acute (24 hr) and subacute (1 week) time periods. rmCHI induced focal cortical microhemorrhages and impaired axial diffusivity at 1 week postinjury. These microstructural abnormalities were associated with a significant increase in microglia. Notably, microgliosis was accompanied by a change in inflammatory microenvironment defined by robust spatiotemporal alterations in tumor necrosis factor-α receptor mRNA. Together these data contribute novel insight into the fundamental biological processes associated with repeated mild brain injury concomitant with subacute imaging abnormalities in a clinically relevant animal model of repeated mild TBI. These findings suggest new diagnostic techniques that can be used as biomarkers to guide the use of future protective or reparative interventions. © 2016 Wiley Periodicals, Inc.
- Published
- 2016
- Full Text
- View/download PDF
58. Imaging and serum biomarkers reflecting the functional efficacy of extended erythropoietin treatment in rats following infantile traumatic brain injury
- Author
-
Jessie R. Maxwell, Jesse L. Denson, Lindsay A.S. Chan, Yirong Yang, Justin Berkner, Jesse L. Winer, Robert C. Tasker, William P. Meehan, Shenandoah Robinson, Lauren L. Jantzie, Laurel O. Sillerud, and Rebekah Mannix
- Subjects
Male ,medicine.medical_specialty ,Pathology ,Time Factors ,Traumatic brain injury ,Receptor expression ,Statistics, Nonparametric ,Article ,White matter ,03 medical and health sciences ,0302 clinical medicine ,030225 pediatrics ,Internal medicine ,Brain Injuries, Traumatic ,Glial Fibrillary Acidic Protein ,Image Processing, Computer-Assisted ,Receptors, Erythropoietin ,medicine ,Animals ,Erythropoietin ,Gait Disorders, Neurologic ,Cerebral Cortex ,Symporters ,biology ,Glial fibrillary acidic protein ,Calpain ,business.industry ,Age Factors ,Gene Expression Regulation, Developmental ,General Medicine ,medicine.disease ,Rats ,Epoetin Alfa ,CXCL1 ,Disease Models, Animal ,Diffusion Magnetic Resonance Imaging ,medicine.anatomical_structure ,Endocrinology ,Animals, Newborn ,Cerebral cortex ,biology.protein ,Cytokines ,Female ,business ,Biomarkers ,030217 neurology & neurosurgery ,medicine.drug - Abstract
OBJECTIVE Traumatic brain injury (TBI) is a leading cause of death and severe morbidity for otherwise healthy full-term infants around the world. Currently, the primary treatment for infant TBI is supportive, as no targeted therapies exist to actively promote recovery. The developing infant brain, in particular, has a unique response to injury and the potential for repair, both of which vary with maturation. Targeted interventions and objective measures of therapeutic efficacy are needed in this special population. The authors hypothesized that MRI and serum biomarkers can be used to quantify outcomes following infantile TBI in a preclinical rat model and that the potential efficacy of the neuro-reparative agent erythropoietin (EPO) in promoting recovery can be tested using these biomarkers as surrogates for functional outcomes. METHODS With institutional approval, a controlled cortical impact (CCI) was delivered to postnatal Day (P)12 rats of both sexes (76 rats). On postinjury Day (PID)1, the 49 CCI rats designated for chronic studies were randomized to EPO (3000 U/kg/dose, CCI-EPO, 24 rats) or vehicle (CCI-veh, 25 rats) administered intraperitoneally on PID1–4, 6, and 8. Acute injury (PID3) was evaluated with an immunoassay of injured cortex and serum, and chronic injury (PID13–28) was evaluated with digitized gait analyses, MRI, and serum immunoassay. The CCI-veh and CCI-EPO rats were compared with shams (49 rats) primarily using 2-way ANOVA with Bonferroni post hoc correction. RESULTS Following CCI, there was 4.8% mortality and 55% of injured rats exhibited convulsions. Of the injured rats designated for chronic analyses, 8.1% developed leptomeningeal cyst–like lesions verified with MRI and were excluded from further study. On PID3, Western blot showed that EPO receptor expression was increased in the injured cortex (p = 0.008). These Western blots also showed elevated ipsilateral cortex calpain degradation products for αII-spectrin (αII-SDPs; p < 0.001), potassium chloride cotransporter 2 (KCC2-DPs; p = 0.037), and glial fibrillary acidic protein (GFAP-DPs; p = 0.002), as well as serum GFAP (serum GFAP-DPs; p = 0.001). In injured rats multiplex electrochemiluminescence analyses on PID3 revealed elevated serum tumor necrosis factor alpha (TNFα p = 0.01) and chemokine (CXC) ligand 1 (CXCL1). Chronically, that is, in PID13–16 CCI-veh rats, as compared with sham rats, gait deficits were demonstrated (p = 0.033) but then were reversed (p = 0.022) with EPO treatment. Diffusion tensor MRI of the ipsilateral and contralateral cortex and white matter in PID16–23 CCI-veh rats showed widespread injury and significant abnormalities of functional anisotropy (FA), mean diffusivity (MD), axial diffusivity (AD), and radial diffusivity (RD); MD, AD, and RD improved after EPO treatment. Chronically, P13–P28 CCI-veh rats also had elevated serum CXCL1 levels, which normalized in CCI-EPO rats. CONCLUSIONS Efficient translation of emerging neuro-reparative interventions dictates the use of age-appropriate preclinical models with human clinical trial–compatible biomarkers. In the present study, the authors showed that CCI produced chronic gait deficits in P12 rats that resolved with EPO treatment and that chronic imaging and serum biomarkers correlated with this improvement.
- Published
- 2016
- Full Text
- View/download PDF
59. Neonatal opioid exposure: public health crisis and novel neuroinflammatory disease
- Author
-
Yuma Kitase, V Joanna Burton, Shenandoah Robinson, Gwendolyn Gerner, Vikram Vasan, Lauren L. Jantzie, and Jessie Newville
- Subjects
cognition ,medicine.medical_specialty ,biomarker ,buprenorphine ,executive function ,magnetic resonance imaging ,methadone ,neonate ,Brain Structure and Function ,Review ,Disease ,lcsh:RC346-429 ,Developmental Neuroscience ,Medicine ,Medical prescription ,Intensive care medicine ,lcsh:Neurology. Diseases of the nervous system ,business.industry ,Public health ,Opioid ,In utero ,business ,Buprenorphine ,medicine.drug ,Methadone - Abstract
Substance use, specifically the use of prescription and non-prescription opioids among pregnant women, is a major public health issue and chief contributor to the opioid crisis. The prevalence of Neonatal Opioid Withdrawal Syndrome has risen 5-fold in the past decade, and is a well-recognized consequence of perinatal opioid exposure. By contrast, the long-term damage to the developing brain from opioid medications is just beginning to be recognized as a serious concern. Published data suggest that opioid exposure commencing in utero negatively affects the maturation of the neural-immune system, and trajectory of central nervous system development. Methadone induces peripheral immune hyper-reactivity, lasting structural and microstructural brain injury, and significant deficits in executive function and cognitive control in adult animals following in utero exposure. Thus, to address the cascading public health crisis stemming from the multitude of infants with in utero opioid exposure who will grow up with altered neurodevelopmental trajectories, rigorous preclinical, mechanistic studies are required. Such studies will define the long-term sequelae of prenatal opioid exposure in an effort to develop appropriate and targeted interventions. Specifically, the development of novel fluid, neuroimaging and biobehavioral biomarkers will be the most useful to aid in early identification and treatment of opioid exposed infants with the greatest risk of poor clinical outcomes. These studies will be essential to understand how in utero insults determine brain structure and function in adulthood, and what targeted interventions will be required to improve long-term outcomes in the countless children being born exposed to opioids each year.
- Published
- 2021
- Full Text
- View/download PDF
60. Time to reconsider extended erythropoietin treatment for infantile traumatic brain injury?
- Author
-
Lauren L. Jantzie, Nagat El Demerdash, Shenandoah Robinson, and Jessie Newville
- Subjects
0301 basic medicine ,Multiple stages ,Phase iii trials ,Traumatic brain injury ,medicine.medical_treatment ,Central nervous system ,Bioinformatics ,03 medical and health sciences ,0302 clinical medicine ,Developmental Neuroscience ,Perinatal Brain Injury ,Brain Injuries, Traumatic ,Medicine ,Animals ,Humans ,Erythropoietin ,Independent research ,business.industry ,Infant, Newborn ,Infant ,medicine.disease ,030104 developmental biology ,Cytokine ,medicine.anatomical_structure ,Neuroprotective Agents ,Neurology ,Child, Preschool ,business ,030217 neurology & neurosurgery ,medicine.drug - Abstract
Pediatric traumatic brain injury (TBI) remains a leading cause of childhood morbidity and mortality worldwide. Most efforts to reduce the chronic impact of pediatric TBI involve prevention and minimization of secondary injury. Currently, no treatments are used in routine clinical care during the acute and subacute phases to actively repair injury to the developing brain. The endogenous pluripotent cytokine erythropoietin (EPO) holds promise as an emerging neuroreparative agent in perinatal brain injury (PBI). EPO signaling in the central nervous system (CNS) is essential for multiple stages of neurodevelopment, including the genesis, survival and differentiation of multiple lineages of neural cells. Postnatally, EPO signaling decreases markedly as the CNS matures. Importantly, high-dose, extended EPO regimens have shown efficacy in preclinical controlled cortical impact (CCI) models of infant TBI at two different, early ages by independent research groups. Specifically, extended high-dose EPO treatment after infantile CCI prevents long-term cognitive deficits in adult rats. Because of the striking differences in the molecular and cellular responses to both injury and recovery in the developing and mature CNS, and the excellent safety profile of EPO in infants and children, extended courses of EPO are currently in Phase III trials for neonates with PBI. Extended, high-dose EPO may also warrant testing for infants and young children with TBI.
- Published
- 2019
61. Evidence for Sexual Dimorphism in the Response to TLR3 Activation in the Developing Neonatal Mouse Brain: A Pilot Study
- Author
-
Carina Mallard, Lauren L. Jantzie, Raul Chavez-Valdez, Amin Mottahedin, Tracylyn R. Yellowhair, Frances J. Northington, and Linnea Stridh
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Chemokine ,Physiology ,medicine.medical_treatment ,caspase ,Intraperitoneal injection ,Hippocampus ,microglia ,Caspase 3 ,poly I:C ,lcsh:Physiology ,03 medical and health sciences ,0302 clinical medicine ,Downregulation and upregulation ,Internal medicine ,Physiology (medical) ,Gene expression ,medicine ,Receptor ,Original Research ,biology ,lcsh:QP1-981 ,astroglia ,Fas receptor ,cytokines ,030104 developmental biology ,Endocrinology ,inflammation ,biology.protein ,endoplasmic reticulum stress ,030217 neurology & neurosurgery - Abstract
Toll-like receptor (TLR)3 activation during the neonatal period produces responses linked to the origins of neuropsychiatric disorders. Although there is sexual dimorphism in neuropsychiatric disorders, it is unknown if brain responses to TLR3 activation are sex-specific. We hypothesized that poly I:C in a post-natal day (P)8 model induces a sexually dimorphic inflammatory responses. C57BL6 mice received intraperitoneal injection of poly I:C (10 mg/kg) or vehicle [normal saline (NS)] at P8. Pups were killed at 6 or 14 h for caspase 3 and 8 activity assays, NFkB ELISA, IRF3, AP1, and GFAP western blotting and cytokines/chemokines gene expression real time qRT-PCR (4–6/group). A second group of pups were killed at 24 h (P9) or 7 days (P15) after poly I:C to assess astrocytic (GFAP) and microglia (Iba1) activation in the hippocampus, thalamus and cortex using immunohistochemistry, and gene and protein expression of cytokines/chemokines using real time RT-PCR and MSD, respectively (4–6/group). Non-parametric analysis was applied. Six hours after poly I:C, caspase-3 and -8 activities in cytosolic fractions were 1.6 and 2.8-fold higher in poly I:C-treated than in NS-treated female mice, respectively, while gene expressions of pro-inflammatory cytokines were upregulated in both sexes. After poly I:C, IRF3 nuclear translocation occurred earlier (6 h) in female mice and later (14 h) in male mice. At 14 h after poly I:C, only male mice also had increased nuclear NFκB levels (88%, p < 0.001) and GFAP expression coinciding with persistent IL-6 and FAS gene upregulation (110 and 77%, respectively; p < 0.001) and IL-10 gene downregulation (-42%, p < 0.05). At 24 h after poly I:C, IL-1β, CXCL-10, TNF-α, and MCP-1 were similarly increased in both sexes but at 7 days after exposure, CXCL-10 and INFγ were increased and IL-10 was decreased only in female mice. Accordingly, microglial activation persisted at 7 days after poly I:C in the hippocampus, thalamus and cortex of female mice. This preliminary study suggests that TLR3 activation may produce in the developing neonatal mouse brain a sexually dimorphic response with early activation of caspase-dependent pathways in female mice, activation of inflammatory cascades in both sexes, which then persists in female mice. Further well-powered studies are essential to confirm these sex-specific findings.
- Published
- 2018
62. T123. A Psychosis-Altered Glial-Produced miRNA Downregulates Neuronal Gene Expression via Exosomes
- Author
-
Jason P. Weick, Tracylyn R. Yellowhair, Brian A. Rodriguez, Lauren L. Jantzie, Steven D. Sheridan, Marree Webster, Stephen K. Amoah, Begűm Alural, Jasmin Lelonde, Stephen J. Haggarty, Roy H. Perlis, Constantine N. Logothetis, Crina M. Floruta, Nikolaos Mellios, and Carl M. Sellgren
- Subjects
Psychosis ,microRNA ,Gene expression ,medicine ,Biology ,medicine.disease ,Biological Psychiatry ,Microvesicles ,Cell biology - Published
- 2019
- Full Text
- View/download PDF
63. Combined in utero hypoxia-ischemia and lipopolysaccharide administration in rats induces chorioamnionitis and a fetal inflammatory response syndrome
- Author
-
Jessie R. Maxwell, Lauren L. Jantzie, Nancy E. Joste, Jesse L. Denson, and Shenandoah Robinson
- Subjects
Lipopolysaccharides ,Pathology ,medicine.medical_specialty ,Placenta ,Central nervous system ,Ischemia ,Inflammation ,Fetal Hypoxia ,Chorioamnionitis ,Rats, Sprague-Dawley ,Pregnancy ,medicine ,Animals ,Neuroinflammation ,Fetus ,business.industry ,Brain ,Obstetrics and Gynecology ,medicine.disease ,Rats ,medicine.anatomical_structure ,Animals, Newborn ,Reproductive Medicine ,Immunology ,Cytokines ,Female ,Tumor necrosis factor alpha ,medicine.symptom ,business ,Developmental Biology - Abstract
Introduction Preterm birth is a major cause of infant morbidity and long-term disability, and is associated with numerous central nervous system (CNS) deficits. Infants exposed to intrauterine inflammation, specifically chorioamnionitis, are at risk for very early preterm birth and neurological complications including cerebral palsy, epilepsy, and behavioral and cognitive deficits. However, placenta-brain axis abnormalities and their relationship to subsequent permanent CNS injury remain poorly defined. Methods Intrauterine injury was induced in rats on embryonic day 18 (E18) by transient systemic hypoxia-ischemia (TSHI) and intra-amniotic lipopolysaccharide (LPS) injection. Placenta, brain and serum were collected from E19 to postnatal day 0 (P0). Histology, TUNEL staining, western blot and multiplex immunoassays were used to quantify placental and brain abnormalities, and fetal serum cytokine levels. Results Prenatal TSHI + LPS caused acute and subacute placental injury hallmarked by inflammatory infiltrate, edema, hemorrhage and cell death along with placental increases in IL-1β and TNFα. TSHI + LPS increased a diverse array of circulating inflammatory proteins including IL-1β, TNFα, IL-6, IL-10, IL-4, IFNγ and CXCL1, both immediately after TSHI + LPS and in live born pups. CNS inflammation was characterized by increased CXCL1. Discussion Prenatal TSHI + LPS in rats induces placental injury and inflammation histologically consistent with chorioamnionitis, concomitant with elevated serum and CNS pro-inflammatory cytokines. This model accurately recapitulates key pathophysiological processes observed in extremely preterm infants including placental, fetal, and CNS inflammation. Further investigation into the mechanism of CNS injury following chorioamnionitis and the placental-brain axis will guide the use of future interventions.
- Published
- 2015
- Full Text
- View/download PDF
64. Erythropoietin Modulates Cerebral and Serum Degradation Products from Excess Calpain Activation following Prenatal Hypoxia-Ischemia
- Author
-
Jesse L. Winer, Christopher J. Corbett, Shenandoah Robinson, and Lauren L. Jantzie
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Central nervous system ,Apoptosis ,Neuroprotection ,Article ,Rats, Sprague-Dawley ,03 medical and health sciences ,0302 clinical medicine ,Developmental Neuroscience ,Internal medicine ,Animals ,Medicine ,Erythropoietin ,Caspase ,Calpastatin ,biology ,Calpain ,business.industry ,Calcium-Binding Proteins ,Membrane Proteins ,Myelin Basic Protein ,Axons ,Myelin basic protein ,Enzyme Activation ,030104 developmental biology ,medicine.anatomical_structure ,Endocrinology ,Animals, Newborn ,Neurology ,Brain Injuries ,Caspases ,Hypoxia-Ischemia, Brain ,Immunology ,biology.protein ,Female ,business ,030217 neurology & neurosurgery ,medicine.drug - Abstract
Preterm infants suffer central nervous system (CNS) injury from hypoxia-ischemia and inflammation - termed encephalopathy of prematurity. Mature CNS injury activates caspase and calpain proteases. Erythropoietin (EPO) limits apoptosis mediated by activated caspases, but its role in modulating calpain activation has not yet been investigated extensively following injury to the developing CNS. We hypothesized that excess calpain activation degrades developmentally regulated molecules essential for CNS circuit formation, myelination and axon integrity, including neuronal potassium-chloride co-transporter (KCC2), myelin basic protein (MBP) and phosphorylated neurofilament (pNF), respectively. Further, we predicted that post-injury EPO treatment could mitigate CNS calpain-mediated degradation. Using prenatal transient systemic hypoxia-ischemia (TSHI) in rats to mimic CNS injury from extreme preterm birth, and postnatal EPO treatment with a clinically relevant dosing regimen, we found sustained postnatal excess cortical calpain activation following prenatal TSHI, as shown by the cleavage of alpha II-spectrin (αII-spectrin) into 145-kDa αII-spectrin degradation products (αII-SDPs) and p35 into p25. Postnatal expression of the endogenous calpain inhibitor calpastatin was also reduced following prenatal TSHI. Calpain substrate expression following TSHI, including cortical KCC2, MBP and NF, was modulated by postnatal EPO treatment. Calpain activation was reflected in serum levels of αII-SDPs and KCC2 fragments, and notably, EPO treatment also modulated KCC2 fragment levels. Together, these data indicate that excess calpain activity contributes to the pathogenesis of encephalopathy of prematurity. Serum biomarkers of calpain activation may detect ongoing cerebral injury and responsiveness to EPO or similar neuroprotective strategies.
- Published
- 2015
- Full Text
- View/download PDF
65. Chloride cotransporter NKCC1 inhibitor bumetanide protects against white matter injury in a rodent model of periventricular leukomalacia
- Author
-
Jenny Y. Yu, Lauren L. Jantzie, Michele Jackson, Jessie R. Maxwell, Melody Y. Hu, Hyun Kyung Park, and Frances E. Jensen
- Subjects
Male ,inorganic chemicals ,congenital, hereditary, and neonatal diseases and abnormalities ,medicine.medical_specialty ,Leukomalacia, Periventricular ,Mice, Transgenic ,Chloride ,Mice ,Sodium Potassium Chloride Symporter Inhibitors ,Ischemia ,Internal medicine ,medicine ,Animals ,Solute Carrier Family 12, Member 2 ,Rats, Long-Evans ,cardiovascular diseases ,Hypoxia ,Bumetanide ,Cerebral Cortex ,Neurons ,Periventricular leukomalacia ,urogenital system ,Chemistry ,White Matter Injury ,Rodent model ,Anatomy ,Long evans ,medicine.disease ,White Matter ,Rats ,nervous system diseases ,Disease Models, Animal ,Oligodendroglia ,Endocrinology ,Gene Expression Regulation ,Pediatrics, Perinatology and Child Health ,Solute Carrier Family 12 ,Cotransporter ,medicine.drug - Abstract
Periventricular leukomalacia (PVL) is a major form of preterm brain injury. Na(+)-K(+)-Cl(-) 1 cotransporter (NKCC1) expression on neurons and astrocytes is developmentally regulated and mediates Cl(-) reversal potential. We hypothesized that NKCC1 is highly expressed on oligodendrocytes (OLs) and increases vulnerability to hypoxia-ischemia (HI) mediated white matter injury, and that the NKCC1 inhibitor bumetanide would be protective in a rodent PVL model.Immunohistochemistry in Long-Evans rats and PLP-EGFP transgenic mice was used to establish cell-specific expression of NKCC1 in the immature rodent brain. HI was induced on postnatal day 6 (P6) in rats and the protective efficacy of bumetanide (0.3 mg/kg/i.p. q12h × 60 h) established.NKCC1 was expressed on OLs and subplate neurons through the first 2 postnatal weeks, peaking in white matter and the subplate between P3-7. Following HI, NKCC1 is expressed on OLs and neurons. Bumetanide treatment significantly attenuates myelin basic protein loss and neuronal degeneration 7 d post-HI.Presence and relative overexpression of NKCC1 in rodent cerebral cortex coincides with a period of developmental vulnerability to HI white matter injury in the immature prenatal brain. The protective efficacy of bumetanide in this model of preterm brain injury suggests that Cl(-) transport is a factor in PVL and that its inhibition may have clinical application in premature human infants.
- Published
- 2015
- Full Text
- View/download PDF
66. Embracing oligodendrocyte diversity in the context of perinatal injury
- Author
-
Jessie Newville, Lee Anna Cunningham, and Lauren L. Jantzie
- Subjects
0301 basic medicine ,Lineage (genetic) ,glia ,oligodendrogenesis ,oligodendrocyte progenitor cell ,myelination ,central nervous system development ,ontogenetic origin ,white matter ,white matter injury ,preterm birth ,macroglia ,media_common.quotation_subject ,Population ,Context (language use) ,Biology ,lcsh:RC346-429 ,Developmental psychology ,White matter ,03 medical and health sciences ,Myelin ,0302 clinical medicine ,Developmental Neuroscience ,030225 pediatrics ,Perinatal Brain Injury ,medicine ,education ,lcsh:Neurology. Diseases of the nervous system ,media_common ,education.field_of_study ,Invited Review ,Oligodendrocyte ,030104 developmental biology ,medicine.anatomical_structure ,Neuroscience ,Diversity (politics) - Abstract
Emerging evidence is fueling a new appreciation of oligodendrocyte diversity that is overturning the traditional view that oligodendrocytes are a homogenous cell population. Oligodendrocytes of distinct origins, maturational stages, and regional locations may differ in their functional capacity or susceptibility to injury. One of the most unique qualities of the oligodendrocyte is its ability to produce myelin. Myelin abnormalities have been ascribed to a remarkable array of perinatal brain injuries, with concomitant oligodendrocyte dysregulation. Within this review, we discuss new insights into the diversity of the oligodendrocyte lineage and highlight their relevance in paradigms of perinatal brain injury. Future therapeutic development will be informed by comprehensive knowledge of oligodendrocyte pathophysiology that considers the particular facets of heterogeneity that this lineage exhibits.
- Published
- 2017
67. Effects of spinal non-viral interleukin-10 gene therapy formulated with d-mannose in neuropathic interleukin-10 deficient mice: Behavioral characterization, mRNA and protein analysis in pain relevant tissues
- Author
-
Nikolaos Mellios, Lauren L. Jantzie, Erin D. Milligan, Shahani Noor, Melody S. Sun, Xuexian O. Yang, Jacob E. Sanchez, and Arden G. Vanderwall
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Immunology ,Central nervous system ,Article ,03 medical and health sciences ,Behavioral Neuroscience ,Mice ,0302 clinical medicine ,Internal medicine ,Spinal Cord Dorsal Horn ,medicine ,Animals ,Mice, Knockout ,Behavior, Animal ,Endocrine and Autonomic Systems ,business.industry ,Cauda equina ,Genetic Therapy ,Spinal cord ,Interleukin-10 ,Lumbar Spinal Cord ,030104 developmental biology ,medicine.anatomical_structure ,Allodynia ,Endocrinology ,Spinal Cord ,Anesthesia ,Neuropathic pain ,Neuralgia ,medicine.symptom ,business ,Mannose ,030217 neurology & neurosurgery ,Astrocyte - Abstract
Studies show that spinal (intrathecal; i.t.) interleukin-10 (IL-10) gene therapy reverses neuropathic pain in animal models, and co-administration with the mannose receptor (MR; CD206) ligand d-mannose (DM) greatly improves therapeutic efficacy. However, the actions of endogenous IL-10 may be required for enduring pain control observed following i.t. IL-10 gene therapy, potentially narrowing the application of this non-viral transgene delivery approach. Here, we show that i.t. application of naked plasmid DNA expressing the IL-10 transgene co-injected with DM (DM/pDNA-IL-10) for the treatment of peripheral neuropathic pain in IL-10 deficient (IL-10 KO) mice results in a profound and prolonged bilateral pain suppression. Neuropathic pain is induced by unilateral sciatic chronic constriction injury (CCI), and while enduring relief of light touch sensitivity (mechanical allodynia) in both wild type (WT) and IL-10 KO mice was observed following DM/pDNA-IL-10 co-therapy, transient reversal from allodynia was observed following i.t. DM alone. In stably pain-relieved IL-10 KO mice given DM/pDNA-IL-10, mRNA for the IL-10 transgene is detected in the cauda equina and ipsilateral dorsal root ganglia (DRG), but not the lumbar spinal cord. Further, DM/pDNA-IL-10 application increases anti-inflammatory TGF-β1 and decreases pro-inflammatory TNF mRNA in the ipsilateral DRG compared to allodynic controls. Additionally, DM/pDNA-IL-10 treated mice exhibit decreased spinal pro-inflammatory mRNA expression for TNF, CCL2 (MCP-1), and for the microglial-specific marker TMEM119. Similarly, DM/pDNA-IL-10 treatment decreases immunoreactivity for the astrocyte activation marker GFAP in lumbar spinal cord dorsal horn. Despite transient reversal and early return to allodynia in DM-treated mice, lumbar spinal cord revealed elevated TNF, CCL2 and TMEM119 mRNA levels. Both MR (CD206) and IL-10 receptor mRNAs are increased in the DRG following CCI manipulation independent of injection treatment, suggesting that pathological conditions stimulate upregulation and availability of relevant receptors in critical anatomical regions required for the therapeutic actions of the DM/pDNA-IL-10 co-therapy. Taken together, the current report demonstrates that non-viral DM/pDNA-IL-10 gene therapy does not require endogenous IL-10 for enduring relief of peripheral neuropathic pain and does not require direct contact with the spinal cord dorsal horn for robust and enduring relief of neuropathic pain. Spinal non-viral DM/pDNA-IL-10 co-therapy may offer a framework for the development of non-viral gene therapeutic approaches for other diseases of the central nervous system.
- Published
- 2017
68. Cognitive development in preterm infants: multifaceted deficits reflect vulnerability of rigorous neurodevelopmental pathways
- Author
-
Akosua Y. Oppong, Jean R. Lowe, Lauren L. Jantzie, Robin K. Ohls, Jessie R. Maxwell, Tracylyn R. Yellowhair, and Jenny E. Camacho
- Subjects
Pediatrics ,medicine.medical_specialty ,Developmental Disabilities ,Vulnerability ,Gestational Age ,Infant, Premature, Diseases ,03 medical and health sciences ,Cognition ,0302 clinical medicine ,Social cognition ,030225 pediatrics ,Intervention (counseling) ,Cognitive development ,Humans ,Medicine ,Erythropoietin ,business.industry ,Infant, Newborn ,Gestational age ,Preterm Births ,Adult life ,Pediatrics, Perinatology and Child Health ,Cognition Disorders ,business ,Infant, Premature ,030217 neurology & neurosurgery - Abstract
Prematurity remains the major cause of neonatal morbidity and mortality, with 15 million preterm births occurring worldwide in 2010. Infants born less than 37 weeks gestation are at high risk of abnormal neurodevelopmental outcomes, given that the central nervous system is extremely sensitive to an abnormal intra- and extra-uterine environment. Children born preterm have multiple neurodevelopmental sequelae involving dynamic and complex cognitive deficits. Former preterm infants have difficulty with each domain of cognition, including executive function, language, learning and memory, complex attention, perceptual-motor function and social cognition when compared to children born at term. Although deficits are not always severe, even mild delays can be impactful, resulting in a spectrum of outcomes from difficulties in school to an inability to lead an independent adult life. Here, we review current literature on the cognitive outcomes of infants born preterm with a focus on how specific disruption in crucial neurodevelopmental pathways render these children vulnerable to dynamic deficits in cognition as they mature. Further, we highlight promising therapies and intervention strategies aimed at mitigating these deficits, including the use of erythropoietin. With an increasing number of preterm infants surviving, understanding developmental deficits will allow therapies to be developed and optimized, in order to ensure the best outcome for this vulnerable patient population.
- Published
- 2017
- Full Text
- View/download PDF
69. 45: Microstructural thalamic injury in a rat model of chorioamnionitis: potential avenues for neurorepair
- Author
-
Shenandoah Robinson, Conrad R. Chao, Jessie Newville, Tracylyn R. Yellowhair, Jenny E. Camacho, Vivek R. Katukuri, Jessie R. Maxwell, and Lauren L. Jantzie
- Subjects
Pathology ,medicine.medical_specialty ,business.industry ,Rat model ,medicine ,Obstetrics and Gynecology ,Chorioamnionitis ,medicine.disease ,business - Published
- 2019
- Full Text
- View/download PDF
70. Chronic gliosis and behavioral deficits in mice following repetitive mild traumatic brain injury
- Author
-
Philippe Moleus, Lauren L. Jantzie, Jianhua Qiu, Georgia Gunner, Nick Andrews, Jacqueline Berglass, Rebekah Mannix, William P. Meehan, Laura Berglass, Justin Berkner, and Shenandoah Robinson
- Subjects
medicine.medical_specialty ,Traumatic brain injury ,business.industry ,Morris water navigation task ,Brain damage ,medicine.disease ,Open field ,Physical medicine and rehabilitation ,Gliosis ,Concussion ,Closed head injury ,medicine ,Anxiety ,medicine.symptom ,business ,Neuroscience - Abstract
Object With the recent increasing interest in outcomes after repetitive mild traumatic brain injury (rmTBI; e.g., sports concussions), several models of rmTBI have been established. Characterizing these models in terms of behavioral and histopathological outcomes is vital to assess their clinical translatability. The purpose of this study is to provide an in-depth behavioral and histopathological phenotype of a clinically relevant model of rmTBI. Methods The authors used a previously published weight-drop model of rmTBI (7 injuries in 9 days) in 2- to 3-month-old mice that produces cognitive deficits without persistent loss of consciousness, seizures, gross structural imaging findings, or microscopic evidence of structural brain damage. Injured and sham-injured (anesthesia only) mice were subjected to a battery of behavioral testing, including tests of balance (rotarod), spatial memory (Morris water maze), anxiety (open field plus maze), and exploratory behavior (hole-board test). After behavioral testing, brains were assessed for histopathological outcomes, including brain volume and microglial and astrocyte immunolabeling. Results Compared with sham-injured mice, mice subjected to rmTBI showed increased exploratory behavior and had impaired balance and worse spatial memory that persisted up to 3 months after injury. Long-term behavioral deficits were associated with chronic increased astrocytosis and microgliosis but no volume changes. Conclusions The authors demonstrate that their rmTBI model results in a characteristic behavioral phenotype that correlates with the clinical syndrome of concussion and repetitive concussion. This model offers a platform from which to study therapeutic interventions for rmTBI.
- Published
- 2014
- Full Text
- View/download PDF
71. Sex differences in the effect of progesterone after controlled cortical impact in adolescent mice: a preliminary study
- Author
-
William P. Meehan, Rachel M. Stanley, Shenandoah Robinson, Jianhua Qiu, Justin Berkner, Jacqueline Berglass, Rebekah Mannix, Philippe Moleus, and Lauren L. Jantzie
- Subjects
medicine.medical_specialty ,Experimental model ,business.industry ,Traumatic brain injury ,Anesthesia ,Public health ,Psychological intervention ,Medicine ,Progesterone treatment ,business ,medicine.disease ,Pediatric population - Abstract
Object While progesterone has been well studied in experimental models of adult traumatic brain injury (TBI), it has not been evaluated in pediatric models. The study of promising interventions in pediatric TBI is important because children have the highest public health burden of such injuries. Therapies that are beneficial in adults may not necessarily be effective in the pediatric population. The purpose of this study was to evaluate whether progesterone treatment improves outcomes in an experimental model of pediatric TBI. Methods The authors determined whether progesterone administered after controlled cortical impact (CCI) improves functional and histopathological outcomes in 4-week-old mice. Both male and female mice (58 mice total) were included in this study, as the majority of prior studies have used only male and/or reproductively senescent females. Mice were randomized to treatment with progesterone or vehicle and to CCI injury or sham injury. Motor (wire grip test) and memory (Morris water maze) testing were performed to determine the effect of progesterone on TBI. Lesion volume was also assessed. Results Compared with their vehicle-treated counterparts, the progesterone-treated CCI-injured male mice had improved motor performance (p < 0.001). In contrast, progesterone-treated CCI-injured female mice had a worse performance than their vehicle-treated counterparts (p = 0.001). Progesterone treatment had no effect on spatial memory performance or lesion volume in injured male or female mice. Conclusions These data suggest a sex-specific effect of progesterone treatment after CCI in adolescent mice and could inform clinical trials in children.
- Published
- 2014
- Full Text
- View/download PDF
72. Erythropoietin attenuates loss of potassium chloride co-transporters following prenatal brain injury
- Author
-
Shenandoah Robinson, Lauren L. Jantzie, Daniel J. Firl, Christopher G. Wilson, Paulina M. Getsy, and Robert H. Miller
- Subjects
Male ,N-Methylaspartate ,Carbazoles ,Hippocampus ,In Vitro Techniques ,Pharmacology ,Inhibitory postsynaptic potential ,Epileptogenesis ,Article ,Indole Alkaloids ,Rats, Sprague-Dawley ,Cellular and Molecular Neuroscience ,Downregulation and upregulation ,Pregnancy ,Excitatory Amino Acid Agonists ,medicine ,Animals ,Enzyme Inhibitors ,Erythropoietin ,Molecular Biology ,Brain-derived neurotrophic factor ,Symporters ,biology ,Brain-Derived Neurotrophic Factor ,Age Factors ,Gene Expression Regulation, Developmental ,Calpain ,Cell Biology ,Rats ,Neuroprotective Agents ,Animals, Newborn ,Ischemic Attack, Transient ,biology.protein ,Excitatory postsynaptic potential ,Female ,Neuroscience ,medicine.drug - Abstract
Therapeutic agents that restore the inhibitory actions of γ-amino butyric acid (GABA) by modulating intracellular chloride concentrations will provide novel avenues to treat stroke, chronic pain, epilepsy, autism, and neurodegenerative and cognitive disorders. During development, upregulation of the potassium-chloride co-transporter KCC2, and the resultant switch from excitatory to inhibitory responses to GABA guide the formation of essential inhibitory circuits. Importantly, maturation of inhibitory mechanisms is also central to the development of excitatory circuits and proper balance between excitatory and inhibitory networks in the developing brain. Loss of KCC2 expression occurs in postmortem samples from human preterm infant brains with white matter lesions. Here we show that late gestation brain injury in a rat model of extreme prematurity impairs the developmental upregulation of potassium chloride co-transporters during a critical postnatal period of circuit maturation in CA3 hippocampus by inducing a sustained loss of oligomeric KCC2 via a calpain-dependent mechanism. Further, administration of erythropoietin (EPO) in a clinically relevant postnatal dosing regimen following the prenatal injury protects the developing brain by reducing calpain activity, restoring oligomeric KCC2 expression and attenuating KCC2 fragmentation, thus providing the first report of a safe therapy to address deficits in KCC2 expression. Together, these data indicate it is possible to reverse abnormalities in KCC2 expression during the postnatal period, and potentially reverse deficits in inhibitory circuit formation central to cognitive impairment and epileptogenesis.
- Published
- 2014
- Full Text
- View/download PDF
73. Developmental Expression of N-Methyl-d-Aspartate (NMDA) Receptor Subunits in Human White and Gray Matter: Potential Mechanism of Increased Vulnerability in the Immature Brain
- Author
-
Mirna Lechpammer, Dionne A. Graham, Frances E. Jensen, Rebecca D. Folkerth, Lauren L. Jantzie, Hyun Kyung Park, Michele Jackson, Joseph J. Volpe, and Delia M. Talos
- Subjects
Adult ,Male ,Leukomalacia, Periventricular ,Cognitive Neuroscience ,Excitotoxicity ,Biology ,medicine.disease_cause ,Receptors, N-Methyl-D-Aspartate ,White matter ,Cellular and Molecular Neuroscience ,medicine ,Humans ,Gray Matter ,Child ,Receptor ,Periventricular leukomalacia ,musculoskeletal, neural, and ocular physiology ,Infant, Newborn ,Glutamate receptor ,Brain ,Infant ,Articles ,Human brain ,Middle Aged ,medicine.disease ,White Matter ,medicine.anatomical_structure ,nervous system ,Child, Preschool ,NMDA receptor ,Neuroglia ,Female ,Neuroscience - Abstract
The pathophysiology of perinatal brain injury is multifactorial and involves hypoxia-ischemia (HI) and inflammation. N-methyl-d-aspartate receptors (NMDAR) are present on neurons and glia in immature rodents, and NMDAR antagonists are protective in HI models. To enhance clinical translation of rodent data, we examined protein expression of 6 NMDAR subunits in postmortem human brains without injury from 20 postconceptional weeks through adulthood and in cases of periventricular leukomalacia (PVL). We hypothesized that the developing brain is intrinsically vulnerable to excitotoxicity via maturation-specific NMDAR levels and subunit composition. In normal white matter, NR1 and NR2B levels were highest in the preterm period compared with adult. In gray matter, NR2A and NR3A expression were highest near term. NR2A was significantly elevated in PVL white matter, with reduced NR1 and NR3A in gray matter compared with uninjured controls. These data suggest increased NMDAR-mediated vulnerability during early brain development due to an overall upregulation of individual receptors subunits, in particular, the presence of highly calcium permeable NR2B-containing and magnesium-insensitive NR3A NMDARs. These data improve understanding of molecular diversity and heterogeneity of NMDAR subunit expression in human brain development and supports an intrinsic prenatal vulnerability to glutamate-mediated injury; validating NMDAR subunit-specific targeted therapies for PVL.
- Published
- 2013
- Full Text
- View/download PDF
74. Adolescent Mice Demonstrate a Distinct Pattern of Injury after Repetitive Mild Traumatic Brain Injury
- Author
-
William P. Meehan, Jumana Hashim, Lauren L. Jantzie, Sasha Alcon, Justin Berkner, Shenandoah Robinson, Zhengrong Mei, Jianhua Qiu, and Rebekah Mannix
- Subjects
0301 basic medicine ,Male ,Elevated plus maze ,Pathology ,medicine.medical_specialty ,Traumatic brain injury ,Excitotoxicity ,Hippocampus ,Physiology ,Morris water navigation task ,Unconsciousness ,Impulsivity ,medicine.disease_cause ,Receptors, N-Methyl-D-Aspartate ,03 medical and health sciences ,Mice ,Random Allocation ,0302 clinical medicine ,medicine ,Animals ,Young adult ,Maze Learning ,Brain Concussion ,Age Factors ,Original Articles ,Recovery of Function ,medicine.disease ,Mice, Inbred C57BL ,030104 developmental biology ,Neurology (clinical) ,medicine.symptom ,Psychology ,030217 neurology & neurosurgery - Abstract
Recently, there has been increasing interest in outcomes after repetitive mild traumatic brain injury (rmTBI) (e.g., sports concussions). Although most of the scientific attention has focused on elite athlete populations, the sequelae of rmTBI in children and young adults have not been well studied. Prior TBI studies have suggested that developmental differences in response to injury, including differences in excitotoxicity and inflammation, could result in differences in functional and histopathological outcomes after injury. The purpose of this study is to compare outcomes in adolescent (5-week-old) versus adult (4-month-old) mice in a clinically relevant model of rmTBI. We hypothesized that functional and histopathological outcomes after rmTBI would differ in developing adolescent brains compared with mature adult brains. Male adolescent and adult (C57Bl/6) mice were subjected to a weight drop model of rmTBI (n = 10-16/group). Loss of consciousness (LOC) after each injury was measured. Functional outcomes were assessed including tests of balance (rotorod), spatial memory (Morris water maze), and impulsivity (elevated plus maze). After behavioral testing, brains were assessed for histopathological outcomes including microglial immunolabeling and N-methyl-d-aspartate (NMDA) receptor subunit expression. Injured adolescent mice had longer LOC than injured adult mice compared with their respective sham controls. Compared with sham mice, adolescent and adult mice subjected to rmTBI had impaired balance, increased impulsivity, and worse spatial memory that persisted up to 3 months after injury, and the effect of injury was worse in adolescent than in adult mice in terms of spatial memory. Three months after injury, adolescent and adult mice demonstrated increased ionized calcium binding adaptor 1 (IbA1) immunolabeling compared with sham controls. Compared with sham controls, NMDA receptor subtype 2B (NR2B) expression in the hippocampus was reduced by ∼20% in both adolescent and adult injured mice. The data suggest that injured adolescent mice may show a distinct pattern of functional deficits after injury that warrants further mechanistic studies.
- Published
- 2017
75. The Unifying Effects of Maternal-Placental-Fetal Axis Dysregulation on Neurodevelopment Following Infectious and Toxic In Utero Insults
- Author
-
Shenandoah Robinson, Fatu S. Conteh, Lauren L. Jantzie, Akosua Y. Oppong, Teresa Easwaran, Danny A Rogers, Jessie Newville, Tracylyn R. Yellowhair, and Jessie R. Maxwell
- Subjects
Fetus ,Pregnancy ,biology ,business.industry ,Central nervous system ,medicine.disease ,Bioinformatics ,biology.organism_classification ,Diagnostic tools ,Prenatal development ,Alcohol consumption during pregnancy ,Zika virus ,medicine.anatomical_structure ,In utero ,Immunology ,medicine ,business - Abstract
Homeostasis during pregnancy and in the in utero environment is essential for prenatal development. Prenatal maturation is hallmarked by an orchestrated and rigorous developmental program characterized by critical cascades in multiple organ systems. During these critical periods of development, however, potential vulnerability to injury exists throughout pregnancy. Indeed, the developing central nervous system (CNS) is extremely vulnerable to environmental insults throughout the entirety of gestation. These insults can adversely affect the developing brain and spinal cord, and permanently alter the neurodevelopmental trajectory. Specifically, in utero insults and dysregulation of the maternal-placental-fetal axis can change molecular, cellular, structural, and functional development of the CNS, culminating in adverse outcomes and neurological disorders throughout postnatal life. In this review, we will discuss common infectious and toxin-induced in utero insults that have recently garnered attention, including Zika virus, prenatal opioid and alcohol exposure, and chorioamnionitis. The goals are to identify common pathophysiological mechanisms, to emphasize the urgent need for new diagnostic tools, and to promote a broader understanding of the diverse array of neurological outcomes presenting in these children throughout their lifespan. With an increasing number of infants exposed to in utero infections and toxins, and the expanding public health awareness of the consequences of Zika infection, the opioid crisis, alcohol consumption during pregnancy, and the frequency of preterm birth in the United States, familiarity with the underlying mechanisms of each of these insults is paramount to improve the diagnosis and treatment for this exceedingly vulnerable patient population.
- Published
- 2017
- Full Text
- View/download PDF
76. A time for cocktails and inclusion
- Author
-
Lauren L. Jantzie, Shenandoah Robinson, and Frances J. Northington
- Subjects
Information retrieval ,business.industry ,MEDLINE ,lcsh:RC346-429 ,03 medical and health sciences ,0302 clinical medicine ,Developmental Neuroscience ,030225 pediatrics ,Perspective ,Medicine ,business ,Inclusion (education) ,030217 neurology & neurosurgery ,lcsh:Neurology. Diseases of the nervous system - Published
- 2018
77. Doxycycline inhibits proinflammatory cytokines but not acute cerebral cytogenesis after hypoxia–ischemia in neonatal rats
- Author
-
Lauren L. Jantzie and Kathryn G. Todd
- Subjects
medicine.medical_specialty ,Time Factors ,Neuroimmunomodulation ,Interleukin-1beta ,Subventricular zone ,Neuroprotection ,Rats, Sprague-Dawley ,Brain ischemia ,Neurotrophic factors ,Internal medicine ,medicine ,Animals ,Pharmacology (medical) ,Longitudinal Studies ,Biological Psychiatry ,Neuroinflammation ,Cell Proliferation ,Antibacterial agent ,Neurons ,Brain-derived neurotrophic factor ,Tumor Necrosis Factor-alpha ,business.industry ,Brain-Derived Neurotrophic Factor ,Dentate gyrus ,Brain ,medicine.disease ,Rats ,Psychiatry and Mental health ,Neuroprotective Agents ,medicine.anatomical_structure ,Endocrinology ,Animals, Newborn ,Matrix Metalloproteinase 9 ,Doxycycline ,Hypoxia-Ischemia, Brain ,Immunology ,Cytokines ,Matrix Metalloproteinase 2 ,business ,Research Paper - Abstract
Background Neonatal hypoxia-ischemia (HI) is a major cause of perinatal brain injury and is associated with a spectrum of neuropsychiatric disorders. Although very few treatment options are currently available, doxycycline (DOXY) has been reported to be neuroprotective in neontatal HI. Our objective was to investigate the effects of DOXY on neonatal brain development in normal and HI rat pups. We hypothesized that DOXY would inhibit microglial activation but that developmentally important processes, including cytogenesis and trophic responses, would not be impaired. Methods To investigate the putative neurodevelopmental consequences of DOXY administration in a clinically relevant animal model of HI, we performed a time-course analysis such that postnatal rat pups received DOXY (10mg/kg) or vehicle immediately before HI (n >or= 6). We then assessed cytogenesis, proinflammatory cytokines, brain-derived neurotrophic factor (BDNF) and matrix metalloproteinases regionally and longitudinally. Results We found that DOXY significantly inhibits neuroinflammation in the frontal cortex, striatum and hippocampus; decreases interleukin-1Beta (IL-1Beta) and tumour necrosis factor-alpha (TNF-alpha); and augments BDNF following HI. In addition, DOXY-treated pups have significantly fewer 2-bromo-5-deoxyuridine (BrdU)-positive cells in the subventricular zone 6 hours post-HI. However, DOXY does not persistently affect cytogenesis in the subventricular zone or dentate gyrus up to 7 days post-HI. The BrdU-positive cells not expressing markers for mature neurons colabel with nestin, an intermediate filament protein typical of neuronal precursors. Limitations Our study investigates "acute" neurodevelopment over the first 7 days of life after HI injury. Further long-term investigations into adulthood are underway. Conclusion Taken together, our results suggest the putative clinical potential of DOXY in the management of neonatal cerebral HI injury.
- Published
- 2010
- Full Text
- View/download PDF
78. Cerebral Amino Acid Profiles after Hypoxia-Reoxygenation and N-Acetylcysteine Treatment in the Newborn Piglet
- Author
-
Kathryn G. Todd, Lauren L. Jantzie, Scott T. Johnson, David L. Bigam, and Po-Yin Cheung
- Subjects
Resuscitation ,Swine ,Radical ,Pharmacology ,Hypoxia ischemia ,Antioxidants ,Drug Administration Schedule ,Acetylcysteine ,medicine ,Animals ,Humans ,Amino Acids ,Hypoxia, Brain ,Cerebrum ,chemistry.chemical_classification ,Asphyxia Neonatorum ,Chemistry ,Infant, Newborn ,Free Radical Scavengers ,Hypoxia (medical) ,Amino acid ,Oxygen ,Disease Models, Animal ,Animals, Newborn ,Biochemistry ,Reperfusion Injury ,Hypoxia-Ischemia, Brain ,Pediatrics, Perinatology and Child Health ,Metabolome ,Hypoxia reoxygenation ,medicine.symptom ,Developmental Biology ,medicine.drug - Abstract
Background: Neonatal hypoxia-ischemia (HI) is a common clinical occurrence. Recently, much evidence has been gathered to suggest that oxygen free radicals are implicated in the pathogenesis of hypoxia-reoxygenation injury through the initiation and propagation of toxic cascades including glutamate excitotoxicity and the manifestation of post-HI neurologic disorders. Following HI, excessive free radicals are formed and antioxidant defenses are diminished. N-acetylcysteine (NAC) is a clinically available antioxidant and has been previously shown to reduce oxidative stress and scavenge free radicals in multiple models of brain injury. Objectives: Using an acutely instrumented swine model of neonatal hypoxia-reoxygenation, the objective of the present study was to examine the neurochemical effects of NAC administration in 5 brain regions exquisitely vulnerable to severe hypoxia. Methods: In a blinded fashion, newborn piglets (1–4 d, 1.4–2.2 kg) were block randomized into surgical sham (SHAM), hypoxic control (HC) and NAC-treated (H-NAC) groups. Both HC and H-NAC piglets were subject to 2 h of alveolar hypoxia (paO2 = 20–40 mm Hg) and then resuscitated with 100% O2 for 1 h followed by 21% for an additional 3 h. Results: Our results show that two hours of severe hypoxemia causes metabolic acidosis and significant changes in cerebral amino acids including glutamate, aspartate and alanine, in all brain regions investigated including the cortex, basal ganglia and thalamus. The administration of NAC 10 min into the reoxygenation period and subsequently continued as an infusion, maintains post-resuscitation amino acid neurochemistry at the levels observed in SHAM piglets. Conclusions: In newborn piglets that have sustained brain injury related to hypoxia/reoxygenation, the administration of NAC does not disrupt cerebral amino acid balance and maintains cerebral amino acid homeostasis.
- Published
- 2009
- Full Text
- View/download PDF
79. Persistent neurochemical changes in neonatal piglets after hypoxia–ischemia and resuscitation with 100%, 21% or 18% oxygen
- Author
-
David L. Bigam, Marwan Emara, Laila Obaid, Lauren L. Jantzie, Po-Yin Cheung, Kathryn G. Todd, and Scott T. Johnson
- Subjects
Resuscitation ,medicine.medical_specialty ,Swine ,Ischemia ,Emergency Nursing ,Neurochemical ,Intensive care ,Internal medicine ,medicine ,Animals ,Amino Acids ,Chromatography, High Pressure Liquid ,chemistry.chemical_classification ,Analysis of Variance ,business.industry ,Oxygen Inhalation Therapy ,Glutamate receptor ,Hypoxia (medical) ,medicine.disease ,Amino acid ,Endocrinology ,Animals, Newborn ,chemistry ,Anesthesia ,Hypoxia-Ischemia, Brain ,Emergency Medicine ,medicine.symptom ,Cardiology and Cardiovascular Medicine ,business ,Neonatal resuscitation - Abstract
Summary Background Neonatal hypoxia–ischemia (HI) is a common complication of pregnancy and delivery. Conventional clinical practice is to resuscitate neonates with 100% O 2 , and evidence is building to suggest resuscitation with lower O 2 concentrations is safer. Significant neurochemical changes are associated with HI injury and persistent changes in amino acids are related to cell death, therefore we used a swine survival model of neonatal HI-reoxygenation (HI/R) to investigate the effects of resuscitation with 100%, 21% or 18% O 2 on amino acid neurotransmitters. Methods In a blinded randomized fashion, following permanent ligation of the left common carotid artery, newborn pigs (1–4 d, 1.7–2.5kg) received alveolar normocapnic hypoxia (FiO 2 =0.15, 2h) and were reoxygenated with 18%, 21% or 100% O 2 . After a 4-day survival period, brain regions were processed for amino acid levels using high-performance liquid chromatography (HPLC). Results Results showed that resuscitation with different O 2 concentrations caused hemispheric and regional changes in all amino acids investigated including glutamate, alanine, γ-amino butyric acid, glycine and aspartate, 4 days post-HI. Resuscitation with 100% O 2 significantly increased glutamate and glycine in the dorsal cortex contralateral to the ligated common carotid artery, compared to piglets resuscitated with 21% O 2 . Additionally, piglets resuscitated with 21% O 2 had significantly lower alanine levels than those resuscitated with 18% O 2 . Conclusion Significant resuscitation-dependent changes in amino acid neurotransmitters are still evident 4 days post-HI in the newborn piglet. These data suggest that persistent changes in neurochemistry occur 4 days after HI/R and further studies are warranted to elucidate the consequences of this on neonatal brain development.
- Published
- 2008
- Full Text
- View/download PDF
80. Neonatal ischemic stroke: a hypoxic–ischemic injury to the developing brain
- Author
-
Kathryn G. Todd, Lauren L. Jantzie, and Po-Yin Cheung
- Subjects
Cell type ,Fetus ,business.industry ,Lissencephaly ,medicine.disease ,Cerebral palsy ,Developmental disorder ,Neurology ,Schizophrenia ,medicine ,Neurology (clinical) ,business ,Stroke ,Neuroscience ,Neonatal stroke - Abstract
Neonatal stroke is a major cause of neonatal death and pediatric disability. The type of stroke and the etiology depend on the age of the fetus and infant, but arterial ischemic stroke is the most common entity [1]. Despite being relatively small for the body’s mass, the brain receives 15% of total cardiac output and 20% of the body’s oxygen supply. As such, the brain is extraordinarily vulnerable to changes in blood flow and oxygen content. While arterial ischemic stroke and hypoxia–ischemia (HI) share some common pathogenetic features, the term HI is commonly used to reflect the alterations in blood flow and oxygen delivery to the brain and may offer greater specificity in designing management strategies. Ischemic stroke in neonates has a wide range of presentations and manifestations and, thus, is very difficult to diagnose. Although HI injury is detrimental to almost every organ of the body, the brain is amongst the most vulnerable. HI injury to the brain may result in devastating consequences. Common diseases such as cerebral palsy, seizure disorders, hearing and vision loss, mental retardation, learning disabilities, schizophrenia, attention-deficit hyperactivity disorder, conduct disorders and developmental diseases, including neuronal migration disorders (lissencephaly and heterotopias), can all have some degree of cerebral HI linked to their etiologies. As the neonatal brain is constantly undergoing natural programmed cell death (apoptosis), it is inherently primed for cell death. Thus, cerebral HI itself may be viewed as a developmental disorder. The fetal brain develops in a stepwise, organized fashion, involving discrete and sequential processes including division, migration and differentiation of diverse cell types [2]. As this occurs and as neurotransmitter systems mature, behavioral patterns emerge that reflect the integration of complex cerebral networks [2]. Cerebral apoptosis is a required developmental process for axonal outgrowth, synaptic consolidation and to establish the appropriate number of neurons and eliminate cells with DNA damage. The fetus has twice as many neurons as required in a mature brain [2], and the developmental ‘pruning’ that occurs is vital and occurs in all cell types. By contrast with the other organs, which are fully formed by the 12th week of gestation, the CNS continues to develop throughout pregnancy and early childhood. After birth, the pruning of excess cells occurs as myelination, neuronal migration and synapse formation are part of postnatal brain development. As the window for potential insult is quite large, it is intuitive that dysregulation of the molecular and cellular events of apoptotic cell death results in detrimental developmental brain pathology.
- Published
- 2008
- Full Text
- View/download PDF
81. Postresuscitation N-acetylcysteine treatment reduces cerebral hydrogen peroxide in the hypoxic piglet brain
- Author
-
Tze Fun Lee, Lauren L. Jantzie, Kathryn G. Todd, and Po-Yin Cheung
- Subjects
Taurine ,medicine.medical_specialty ,Antioxidant ,Swine ,Resuscitation ,medicine.medical_treatment ,Critical Care and Intensive Care Medicine ,medicine.disease_cause ,Acetylcysteine ,chemistry.chemical_compound ,Internal medicine ,Intensive care ,medicine ,Animals ,Prospective Studies ,Hypoxia ,chemistry.chemical_classification ,Reactive oxygen species ,business.industry ,Brain ,Free Radical Scavengers ,Hydrogen Peroxide ,Glutathione ,Hypoxia (medical) ,Oxidative Stress ,Endocrinology ,chemistry ,Anesthesia ,medicine.symptom ,business ,Oxidative stress ,medicine.drug - Abstract
Reactive oxygen species have been implicated in the pathogenesis of hypoxia–reoxygenation injury. However, little information is known regarding the temporal profile of cerebral hydrogen peroxide (HPO) production and its response to N-acetylcysteine (an antioxidant) administration during neonatal hypoxia–reoxygenation. Using an acute swine model of neonatal hypoxia–reoxygenation, we examined the short-term neuroprotective effects of N-acetylcysteine on cerebral HPO production and oxidative stress in the brain. Controlled, block-randomized animal study. University animal research laboratory. Newborn piglets (1–3 days, 1.7–2.1 kg). At 5 min after reoxygenation, piglets were given either saline or N-acetylcysteine (20 or 100 mg/kg/h) in a blinded, randomized fashion. Newborn piglets were block-randomized into a sham-operated group (without hypoxia–reoxygenation, n = 5) and three hypoxic–reoxygenated groups (2 h of normocapnic alveolar hypoxia followed by 2 h of reoxygenation, n = 7/group). Heart rate, mean arterial pressure, cortical HPO concentration, amino acid levels in cerebral microdialysate, and cerebral tissue glutathione and lipid hydroperoxide levels were examined. Hypoxic piglets were hypotensive and acidotic, and they recovered similarly in all hypoxic–reoxygenated groups. In hypoxic–reoxygenated control piglets, the cortical HPO concentration gradually increased during reoxygenation. Both doses of N-acetylcysteine abolished the increased HPO concentration and oxidized glutathione levels and tended to reduce the glutathione ratio and lipid hydroperoxide levels in the cerebral cortex (p = 0.08 and p = 0.1 vs. controls, respectively). N-acetylcysteine at 100 mg/kg/h also increased the cerebral extracellular taurine levels. In newborn piglets with hypoxia–reoxygenation, postresuscitation administration of N-acetylcysteine reduces cerebral HPO production and oxidative stress, probably through a taurine-related mechanism.
- Published
- 2007
- Full Text
- View/download PDF
82. Neuroprotection and Drugs Used to Treat Psychiatric Disorders
- Author
-
Glen B. Baker, Kathryn G. Todd, and Lauren L. Jantzie
- Subjects
medicine.medical_specialty ,business.industry ,Immunology ,Neurodegeneration ,Ischemia ,Disease ,medicine.disease ,Neuroprotection ,Mechanism of action ,Mood disorders ,Neuroimaging ,Schizophrenia ,Medicine ,medicine.symptom ,business ,Psychiatry - Abstract
Traditionally, research on mechanisms of action of psychiatric drugs has focused on their effects on a relatively small number of neurotranmitters/neuromodulators. Over the years, hypotheses related to these mechanisms have been challenged. Neuroimaging literature has suggested that some psychiatric illnesses may have neurodegenerative features and has stimulated innovative and exciting research on the mechanism of action of psychiatric drugs. Neurodegeneration, neuronal cell loss, neuronal atrophy, and glial activation are common histological features of certain psychiatric and neurologic disorders. Research in clinical and basic neuroscience has highlighted common pathways in diseases such as cerebral ischemia, Parkinson’s disease and Alzheimer’s disease that are crucial to neurodegeneration and may be altered in some psychiatric disorders. Thus, drugs that target one or more of the cellular processes that lead to neuronal cell death or dysfunction may be useful to treat both symptoms and underlying pathology in mood disorders and schizophrenia, and may improve the prognoses of these illnesses.
- Published
- 2007
- Full Text
- View/download PDF
83. Modeling Encephalopathy of Prematurity Using Prenatal Hypoxia-ischemia with Intra-amniotic Lipopolysaccharide in Rats
- Author
-
Lindsay A.S. Chan, Jesse L. Winer, Lauren L. Jantzie, Shenandoah Robinson, and Jessie R. Maxwell
- Subjects
Pathology ,medicine.medical_specialty ,Fetus ,General Immunology and Microbiology ,business.industry ,Obstetrics ,General Chemical Engineering ,General Neuroscience ,Encephalopathy ,Central nervous system ,Inflammation ,medicine.disease ,Chorioamnionitis ,General Biochemistry, Genetics and Molecular Biology ,White matter ,medicine.anatomical_structure ,Placenta ,Subplate ,medicine ,Medicine ,medicine.symptom ,business - Abstract
Encephalopathy of prematurity (EoP) is a term that encompasses the central nervous system (CNS) abnormalities associated with preterm birth. To best advance translational objectives and uncover new therapeutic strategies for brain injury associated with preterm birth, preclinical models of EoP must include similar mechanisms of prenatal global injury observed in humans and involve multiple components of the maternal-placental-fetal system. Ideally, models should produce a similar spectrum of functional deficits in the mature animal and recapitulate multiple aspects of the pathophysiology. To mimic human systemic placental perfusion defects, placental underperfusion and/or chorioamnionitis associated with pathogen-induced inflammation in early preterm birth, we developed a model of prenatal transient systemic hypoxia-ischemia (TSHI) combined with intra-amniotic lipopolysaccharide (LPS). In pregnant Sprague Dawley rats, TSHI via uterine artery occlusion on embryonic day 18 (E18) induces a graded placental underperfusion defect associated with increasing CNS damage in the fetus. When combined with intra-amniotic LPS injections, placental inflammation is increased and CNS damage is compounded with associated white matter, gait and imaging abnormalities. Prenatal TSHI and TSHI+LPS prenatal insults meet several of the criteria of an EoP model including recapitulating the intrauterine insult, causing loss of neurons, oligodendrocytes and axons, loss of subplate, and functional deficits in adult animals that mimic those observed in children born extremely preterm. Moreover, this model allows for the dissection of inflammation induced by divergent injury types.
- Published
- 2015
- Full Text
- View/download PDF
84. Microstructural and microglial changes after repetitive mild traumatic brain injury in mice
- Author
-
Shenandoah, Robinson, Jacqueline B, Berglass, Jesse L, Denson, Justin, Berkner, Christopher V, Anstine, Jesse L, Winer, Jessie R, Maxwell, Jianhua, Qiu, Yirong, Yang, Laurel O, Sillerud, William P, Meehan, Rebekah, Mannix, and Lauren L, Jantzie
- Subjects
Male ,Calcium-Binding Proteins ,Microfilament Proteins ,Brain ,Nitric Oxide Synthase Type II ,Nerve Fibers, Myelinated ,Statistics, Nonparametric ,Article ,Mice, Inbred C57BL ,Disease Models, Animal ,Mice ,Diffusion Tensor Imaging ,Gene Expression Regulation ,Brain Injuries, Traumatic ,Animals ,Cytokines ,Microglia ,RNA, Messenger ,Intracranial Hemorrhages - Abstract
Traumatic brain injury (TBI) is a major public health issue, with recently increased awareness of the potential long-term sequelae of repetitive injury. Although TBI is common, objective diagnostic tools with sound neurobiological predictors of outcome are lacking. Indeed, such tools could help to identify those at risk for more severe outcomes after repetitive injury and improve understanding of biological underpinnings to provide important mechanistic insights. We tested the hypothesis that acute and subacute pathological injury, including the microgliosis that results from repeated mild closed head injury (rmCHI), is reflected in susceptibility-weighted magnetic resonance imaging and diffusion-tensor imaging microstructural abnormalities. Using a combination of high-resolution magnetic resonance imaging, stereology, and quantitative PCR, we studied the pathophysiology of male mice that sustained seven consecutive mild traumatic brain injuries over 9 days in acute (24 hr) and subacute (1 week) time periods. rmCHI induced focal cortical microhemorrhages and impaired axial diffusivity at 1 week postinjury. These microstructural abnormalities were associated with a significant increase in microglia. Notably, microgliosis was accompanied by a change in inflammatory microenvironment defined by robust spatiotemporal alterations in tumor necrosis factor-α receptor mRNA. Together these data contribute novel insight into the fundamental biological processes associated with repeated mild brain injury concomitant with subacute imaging abnormalities in a clinically relevant animal model of repeated mild TBI. These findings suggest new diagnostic techniques that can be used as biomarkers to guide the use of future protective or reparative interventions. © 2016 Wiley Periodicals, Inc.
- Published
- 2015
85. Prenatal hypoxia-ischemia induces abnormalities in CA3 microstructure, potassium chloride cotransporter 2 expression and inhibitory tone
- Author
-
Daniel J. Firl, Christopher G. Wilson, Jesse L. Denson, Danny A Rogers, Paulina M. Getsy, Jessie R. Maxwell, Lauren L. Jantzie, and Shenandoah Robinson
- Subjects
KCC2 ,seizure ,Encephalopathy ,microstructure ,Hippocampus ,CA3 ,Hippocampal formation ,Inhibitory postsynaptic potential ,lcsh:RC321-571 ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,Epilepsy ,0302 clinical medicine ,Downregulation and upregulation ,030225 pediatrics ,Biological neural network ,medicine ,lcsh:Neurosciences. Biological psychiatry. Neuropsychiatry ,Original Research ,prematurity ,medicine.disease ,inhibition ,nervous system ,Excitatory postsynaptic potential ,Psychology ,Neuroscience ,030217 neurology & neurosurgery - Abstract
Infants who suffer perinatal brain injury, including those with encephalopathy of prematurity, are prone to chronic neurological deficits, including epilepsy, cognitive impairment, and behavioral problems, such as anxiety, inattention, and poor social interaction. These deficits, especially in combination, pose the greatest hindrance to these children becoming independent adults. Cerebral function depends on adequate development of essential inhibitory neural circuits and the appropriate amount of excitation and inhibition at specific stages of maturation. Early neuronal synaptic responses to γ-amino butyric acid (GABA) are initially excitatory. During the early postnatal period, GABAAR responses switch to inhibitory with the upregulation of potassium-chloride co-transporter KCC2. With extrusion of chloride by KCC2, the Cl(-) reversal potential shifts and GABA and glycine responses become inhibitory. We hypothesized that prenatal hypoxic-ischemic brain injury chronically impairs the developmental upregulation of KCC2 that is essential for cerebral circuit formation. Following late gestation hypoxia-ischemia (HI), diffusion tensor imaging in juvenile rats shows poor microstructural integrity in the hippocampal CA3 subfield, with reduced fractional anisotropy and elevated radial diffusivity. The loss of microstructure correlates with early reduced KCC2 expression on NeuN-positive pyramidal neurons, and decreased monomeric and oligomeric KCC2 protein expression in the CA3 subfield. Together with decreased inhibitory post-synaptic currents during a critical window of development, we document for the first time that prenatal transient systemic HI in rats impairs hippocampal CA3 inhibitory tone. Failure of timely development of inhibitory tone likely contributes to a lower seizure threshold and impaired cognitive function in children who suffer perinatal brain injury.
- Published
- 2015
- Full Text
- View/download PDF
86. Dysregulation of FMRP/mTOR signaling cascade in hypoxic-ischemic injury of premature human brain
- Author
-
Frances E. Jensen, Pia Wintermark, Mirna Lechpammer, Katherine Merry, Michele Jackson, and Lauren L. Jantzie
- Subjects
Male ,medicine.medical_specialty ,congenital, hereditary, and neonatal diseases and abnormalities ,Encephalopathy ,Blotting, Western ,Pilot Projects ,Biology ,Article ,Pathogenesis ,03 medical and health sciences ,Fragile X Mental Retardation Protein ,0302 clinical medicine ,Downregulation and upregulation ,030225 pediatrics ,Internal medicine ,medicine ,Humans ,TOR Serine-Threonine Kinases ,Infant, Newborn ,Brain ,Human brain ,Hypoxia (medical) ,Middle Aged ,medicine.disease ,Immunohistochemistry ,nervous system diseases ,Endocrinology ,medicine.anatomical_structure ,Pediatrics, Perinatology and Child Health ,Hypoxia-Ischemia, Brain ,Autism ,Female ,Neurology (clinical) ,medicine.symptom ,Signal transduction ,Neuroscience ,030217 neurology & neurosurgery ,Signal Transduction - Abstract
In this study the authors investigated whether dysregulation of the fragile X mental retardation protein and mammalian target of rapamycin signaling cascade can have a role in the pathogenesis of encephalopathy of prematurity following perinatal hypoxia-ischemia. The authors examined the brain tissue of newborns with encephalopathy and compared it to age-matched controls with normal brain development and adults. In normal controls, the fragile X mental retardation protein expression in cortical gray matter spiked 4-fold during 36-39 gestational weeks compared to the adult, with a concomitant suppression of p70S6K and S6. In encephalopathy cases, the developmental spike of fragile X mental retardation protein was not observed, and fragile X mental retardation protein levels remained significantly lower than in normal controls. Importantly, this fragile X mental retardation protein downregulation was followed by a significant overexpression of p70S6K and S6. These novel findings thus suggest that premature hypoxic-ischemic brain injury can affect the fragile X mental retardation protein/mammalian target of rapamycin pathway, as otherwise observed in inherited syndromes of cognitive disability and autism spectrum disorders.
- Published
- 2015
87. Inferior olivary neurons innervate multiple zones of the flocculus in pigeons (Columba livia)
- Author
-
Lauren L. Jantzie, Angela P. Nguyen, Douglas R. Wylie, Kathryn G. Todd, Janelle M.P. Pakan, Peter L. Hurd, and Ian R. Winship
- Subjects
Neurons ,Horizontal axis ,General Neuroscience ,Vertical axis ,Fixation, Ocular ,Reflex, Vestibulo-Ocular ,Flocculus ,Optokinetic reflex ,Climbing fiber ,Anatomy ,Olivary Nucleus ,Biology ,Axons ,Cerebellar Cortex ,medicine.anatomical_structure ,Vestibular nuclei ,Cerebellar cortex ,Neural Pathways ,medicine ,Animals ,Columbidae ,Psychomotor Performance ,Fluorescent Dyes ,Optokinetic stimulation - Abstract
Complex spike activity of floccular Purkinje cells responds to patterns of rotational optic flow about the vertical axis (rVA neurons) or a horizontal axis 45 degrees to the midline (rH45 neurons). The pigeon flocculus is organized into four parasagittal zones: two rVA zones (zones 0 and 2) interdigitated with two rH45 zones (zones 1 and 3). Climbing fiber input to the rVA and rH45 zones arises in the caudal and rostral regions of the medial column of the inferior olive (mcIO), respectively. To determine whether the two rVA zones and the two rH45 zones receive input from different areas of the caudal and rostral mcIO and whether individual neurons project to both zones of the same rotational preference, different colors of fluorescent retrograde tracer were injected into the two rVA or two rH45 zones. For the rVA injections, retrogradely labeled cells from the two zones were intermingled in the caudal mcIO, but the distribution of cells labeled from zone 0 was slightly caudal to that from zone 2. On average, 18% of neurons were double labeled. For the rH45 injections, cells retrogradely labeled from the two zones were intermingled in the rostral mcIO, but the distribution of cells labeled from zone 1 was slightly rostral to that from zone 3. On average, 22% of neurons were double labeled. In sum, each of the two rVA zones and the two rH45 zones receives input from slightly different regions of the mcIO, and about 20% of the neurons project to both zones.
- Published
- 2005
- Full Text
- View/download PDF
88. 131 Neonatal Treatment with Endogenous Repair Agents Reverses Abnormal Thermal Sensation in a Preclinical Model of Cerebral Palsy
- Author
-
Jessie R. Maxwell, Tracylyn R. Yellowhair, Shenandoah Robinson, Akosua Y. Oppong, Lauren L. Jantzie, and Fatu S. Conteh
- Subjects
business.industry ,Chronic pain ,Endogeny ,medicine.disease ,Spinal cord ,Chorioamnionitis ,Cerebral palsy ,medicine.anatomical_structure ,Allodynia ,Erythropoietin ,Anesthesia ,Neuropathic pain ,Medicine ,Surgery ,Neurology (clinical) ,medicine.symptom ,business ,medicine.drug - Published
- 2017
- Full Text
- View/download PDF
89. Sex differences in the effect of progesterone after controlled cortical impact in adolescent mice: a preliminary study
- Author
-
Rebekah, Mannix, Jacqueline, Berglass, Justin, Berkner, Philippe, Moleus, Jianhua, Qiu, Lauren L, Jantzie, William P, Meehan, Rachel M, Stanley, and Shenandoah, Robinson
- Subjects
Male ,Sex Characteristics ,Age Factors ,Pilot Projects ,Recovery of Function ,Motor Activity ,Article ,Mice, Inbred C57BL ,Disease Models, Animal ,Random Allocation ,Treatment Outcome ,Brain Injuries ,Animals ,Female ,Progestins ,Maze Learning ,Progesterone ,Spatial Memory - Abstract
While progesterone has been well studied in experimental models of adult traumatic brain injury (TBI), it has not been evaluated in pediatric models. The study of promising interventions in pediatric TBI is important because children have the highest public health burden of such injuries. Therapies that are beneficial in adults may not necessarily be effective in the pediatric population. The purpose of this study was to evaluate whether progesterone treatment improves outcomes in an experimental model of pediatric TBI.The authors determined whether progesterone administered after controlled cortical impact (CCI) improves functional and histopathological outcomes in 4-week-old mice. Both male and female mice (58 mice total) were included in this study, as the majority of prior studies have used only male and/or reproductively senescent females. Mice were randomized to treatment with progesterone or vehicle and to CCI injury or sham injury. Motor (wire grip test) and memory (Morris water maze) testing were performed to determine the effect of progesterone on TBI. Lesion volume was also assessed.Compared with their vehicle-treated counterparts, the progesterone-treated CCI-injured male mice had improved motor performance (p0.001). In contrast, progesterone-treated CCI-injured female mice had a worse performance than their vehicle-treated counterparts (p = 0.001). Progesterone treatment had no effect on spatial memory performance or lesion volume in injured male or female mice.These data suggest a sex-specific effect of progesterone treatment after CCI in adolescent mice and could inform clinical trials in children.
- Published
- 2014
90. Preclinical Models of Encephalopathy of Prematurity
- Author
-
Shenandoah Robinson and Lauren L. Jantzie
- Subjects
Brain Diseases ,business.industry ,Encephalopathy ,Central nervous system ,Cognition ,Context (language use) ,Infant, Premature, Diseases ,medicine.disease ,Bioinformatics ,Article ,Cerebral palsy ,Cns injury ,Disease Models, Animal ,Developmental trajectory ,medicine.anatomical_structure ,Developmental Neuroscience ,Neurology ,Multiple Models ,Anesthesia ,medicine ,Animals ,Humans ,business - Abstract
Encephalopathy of prematurity (EoP) encompasses the central nervous system (CNS) abnormalities associated with injury from preterm birth. Although rapid progress is being made, limited understanding exists of how cellular and molecular CNS injury from early birth manifests as the myriad of neurological deficits in children who are born preterm. More importantly, this lack of direct insight into the pathogenesis of these deficits hinders both our ability to diagnose those infants who are at risk in real time and could potentially benefit from treatment and our ability to develop more effective interventions. Current barriers to clarifying the pathophysiology, developmental trajectory, injury timing, and evolution include preclinical animal models that only partially recapitulate the molecular, cellular, histological, and functional abnormalities observed in the mature CNS following EoP. Inflammation from hypoxic-ischemic and/or infectious injury induced in utero in lower mammals, or actual prenatal delivery of more phylogenetically advanced mammals, are likely to be the most clinically relevant EOP models, facilitating translation to benefit infants. Injury timing, type, severity, and pathophysiology need to be optimized to address the specific hypothesis being tested. Functional assays of the mature animal following perinatal injury to mimic EoP should ideally test for the array of neurological deficits commonly observed in preterm infants, including gait, seizure threshold and cognitive and behavioral abnormalities. Here, we review the merits of various preclinical models, identify gaps in knowledge that warrant further study and consider challenges that animal researchers may face in embarking on these studies. While no one model system is perfect, insights relevant to the clinical problem can be gained with interpretation of experimental results within the context of inherent limitations of the chosen model system. Collectively, optimal use of multiple models will address a major challenge facing the field today - to identify the type and severity of CNS injury these vulnerable infants suffer in a safe and timely manner, such that emerging neurointerventions can be tailored to specifically address individual reparative needs.
- Published
- 2014
91. Complex pattern of interaction between in uterohypoxia-ischemia and intra-amniotic inflammation disrupts brain development and motor function
- Author
-
Daniel J. Firl, Julian Flores, Chris Corbett, Jacqueline Berglass, Lauren L. Jantzie, Rebekah Mannix, and Shenandoah Robinson
- Subjects
Lipopolysaccharides ,Pathology ,Neurofilament ,Rats, Sprague-Dawley ,Myelin ,0302 clinical medicine ,Leukoencephalopathies ,Pregnancy ,Receptors, Erythropoietin ,Hypoxia-ischemia ,Gait ,Glial fibrillary acidic protein ,General Neuroscience ,Microfilament Proteins ,Brain ,Gene Expression Regulation, Developmental ,3. Good health ,medicine.anatomical_structure ,Neurology ,In utero ,Prenatal Exposure Delayed Effects ,Hypoxia-Ischemia, Brain ,Female ,medicine.symptom ,Prematurity ,medicine.drug ,medicine.medical_specialty ,Immunology ,Lipopolysaccharide ,Inflammation ,Motor deficit ,Biology ,White matter ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,Preterm ,030225 pediatrics ,Internal medicine ,Glial Fibrillary Acidic Protein ,medicine ,Animals ,Erythropoietin ,Research ,Calcium-Binding Proteins ,Myelin Basic Protein ,Embryo, Mammalian ,Axons ,Rats ,Myelin basic protein ,Disease Models, Animal ,Endocrinology ,Animals, Newborn ,Gliosis ,biology.protein ,030217 neurology & neurosurgery - Abstract
Background: Infants born preterm commonly suffer from a combination of hypoxia-ischemia (HI) and infectious perinatal inflammatory insults that lead to cerebral palsy, cognitive delay, behavioral issues and epilepsy. Using a novel rat model of combined late gestation HI and lipopolysaccharide (LPS)-induced inflammation, we tested our hypothesis that inflammation from HI and LPS differentially affects gliosis, white matter development and motor impairment during the first postnatal month. Methods: Pregnant rats underwent laparotomy on embryonic day 18 and transient systemic HI (TSHI) and/ or intra-amniotic LPS injection. Shams received laparotomy and anesthesia only. Pups were born at term. Immunohistochemistry with stereological estimates was performed to assess regional glial loads, and western blots were performed for protein expression. Erythropoietin ligand and receptor levels were quantified using quantitative PCR. Digigait analysis detected gait deficits. Statistical analysis was performed with one-way analysis of variance and post-hoc Bonferonni correction. Results: Microglial and astroglial immunolabeling are elevated in TSHI + LPS fimbria at postnatal day 2 compared to sham (both P < 0.03). At postnatal day 15, myelin basic protein expression is reduced by 31% in TSHI + LPS pups compared to shams (P < 0.05). By postnatal day 28, white matter injury shifts from the acute injury pattern to a chronic injury pattern in TSHI pups only. Both myelin basic protein expression (P < 0.01) and the phosphoneurofilament/ neurofilament ratio, a marker of axonal dysfunction, are reduced in postnatal day 28 TSHI pups (P < 0.001). Erythropoietin ligand to receptor ratios differ between brains exposed to TSHI and LPS. Gait analyses reveal that all groups (TSHI, LPS and TSHI + LPS) are ataxic with deficits in stride, paw placement, gait consistency and coordination (all P < 0.001). Conclusions: Prenatal TSHI and TSHI + LPS lead to different patterns of injury with respect to myelination, axon integrity and gait deficits. Dual injury leads to acute alterations in glial response and cellular inflammation, while TSHI alone causes more prominent chronic white matter and axonal injury. Both injuries cause significant gait deficits. Further study will contribute to stratification of injury mechanisms in preterm infants, and guide the use of promising therapeutic interventions.
- Published
- 2014
- Full Text
- View/download PDF
92. Postnatal Erythropoietin Mitigates Impaired Cerebral Cortical Development Following Subplate Loss from Prenatal Hypoxia–Ischemia
- Author
-
Lauren L. Jantzie, Chris Corbett, Shenandoah Robinson, and Daniel J. Firl
- Subjects
Cognitive Neuroscience ,Biology ,In Vitro Techniques ,Motor Activity ,Neuroprotection ,Rats, Sprague-Dawley ,Cellular and Molecular Neuroscience ,Epilepsy ,Downregulation and upregulation ,Subplate ,Nuclear Receptor Subfamily 4, Group A, Member 2 ,medicine ,Animals ,Receptor ,Erythropoietin ,Cerebral Cortex ,Cell Death ,Symporters ,Age Factors ,Gene Expression Regulation, Developmental ,Articles ,medicine.disease ,Embryo, Mammalian ,Receptors, GABA-A ,Rats ,Disease Models, Animal ,Fetal Diseases ,medicine.anatomical_structure ,Animals, Newborn ,Apoptosis ,Cerebral cortex ,Brain Injuries ,Hypoxia-Ischemia, Brain ,Neuroscience ,medicine.drug - Abstract
Preterm birth impacts brain development and leads to chronic deficits including cognitive delay, behavioral problems, and epilepsy. Premature loss of the subplate, a transient subcortical layer that guides development of the cerebral cortex and axonal refinement, has been implicated in these neurological disorders. Subplate neurons influence postnatal upregulation of the potassium chloride co-transporter KCC2 and maturation of γ-amino-butyric acid A receptor (GABAAR) subunits. We hypothesized that prenatal transient systemic hypoxia-ischemia (TSHI) in Sprague-Dawley rats that mimic brain injury from extreme prematurity in humans would cause premature subplate loss and affect cortical layer IV development. Further, we predicted that the neuroprotective agent erythropoietin (EPO) could attenuate the injury. Prenatal TSHI induced subplate neuronal loss via apoptosis. TSHI impaired cortical layer IV postnatal upregulation of KCC2 and GABAAR subunits, and postnatal EPO treatment mitigated the loss (n ≥ 8). To specifically address how subplate loss affects cortical development, we used in vitro mechanical subplate ablation in slice cultures (n ≥ 3) and found EPO treatment attenuates KCC2 loss. Together, these results show that subplate loss contributes to impaired cerebral development, and EPO treatment diminishes the damage. Limitation of premature subplate loss and the resultant impaired cortical development may minimize cerebral deficits suffered by extremely preterm infants.
- Published
- 2014
93. Erythropoietin Signaling Promotes Oligodendrocyte Development Following Prenatal Systemic Hypoxic-Ischemic Brain Injury
- Author
-
Lauren L. Jantzie, Shenandoah Robinson, and Robert H. Miller
- Subjects
Neurogenesis ,Hypoxic ischemic brain injury ,Biology ,Hypoxia ischemia ,Article ,Rats, Sprague-Dawley ,Pregnancy ,medicine ,Animals ,Base sequence ,Erythropoietin ,DNA Primers ,Base Sequence ,Extramural ,Oligodendrocyte ,Recombinant Proteins ,Rats ,Sprague dawley ,Oligodendroglia ,medicine.anatomical_structure ,Brain Injuries ,Pediatrics, Perinatology and Child Health ,Immunology ,Hypoxia-Ischemia, Brain ,Cancer research ,Female ,Signal transduction ,Cell Division ,medicine.drug ,Signal Transduction - Abstract
Brain injury from preterm birth causes white matter injury (WMI), and it leads to chronic neurological deficits including cerebral palsy, epilepsy, cognitive, and behavioral delay. Immature O4+ oligodendrocytes are particularly vulnerable to WMI. Understanding how the developing brain recovers after injury is essential to finding more effective therapeutic strategies. Erythropoietin (EPO) promotes neuronal recovery after injury; however, its role in enhancing oligodendroglial lineage recovery is unclear. Previously, we found that recombinant EPO (rEPO) treatment enhances myelin basic protein (MBP) expression and functional recovery in adult rats after prenatal transient systemic hypoxia-ischemia (TSHI). We hypothesized that after injury, rEPO would enhance oligodendroglial lineage cell genesis, survival, maturation, and myelination.In vitro assays were used to define how rEPO contributes to specific stages of oligodendrocyte development and recovery after TSHI.After prenatal TSHI injury, rEPO promotes genesis of oligodendrocyte progenitors from oligodendrospheres, survival of oligodendrocyte precursor cells (OPCs) and O4+ immature oligodendrocytes, O4+ cell process extension, and MBP expression. rEPO did not alter OPC proliferation.Together, these studies demonstrate that EPO signaling promotes critical stages of oligodendroglial lineage development and recovery after prenatal TSHI injury. EPO treatment may be beneficial to preterm and other infant patient populations with developmental brain injury hallmarked by WMI.
- Published
- 2013
94. Regional Differences in Susceptibility to Hypoxic-Ischemic Injury in the Preterm Brain: Exploring the Spectrum from White Matter Loss to Selective Grey Matter Injury in a Rat Model
- Author
-
Debra B. Selip, Michele Jackson, Erin C. Fitzgerald, Andrew Murphy, Lauren L. Jantzie, Mun Seog Chang, Frances E. Jensen, and G. Boll
- Subjects
Pathology ,medicine.medical_specialty ,Neocortex ,Periventricular leukomalacia ,Article Subject ,business.industry ,Thalamus ,Grey matter ,medicine.disease ,lcsh:RC346-429 ,White matter ,medicine.anatomical_structure ,Neurology ,Gliosis ,nervous system ,medicine ,Neurology (clinical) ,Astrocytosis ,medicine.symptom ,business ,Infiltration (medical) ,lcsh:Neurology. Diseases of the nervous system ,Research Article - Abstract
Models of premature brain injury have largely focused on the white matter injury thought to underlie periventricular leukomalacia (PVL). However, with increased survival of very low birth weight infants, injury patterns involving grey matter are now recognized. We aimed to determine how grey matter lesions relate to hypoxic-ischemic- (HI) mediated white matter injury by modifying our rat model of PVL. Following HI, microglial infiltration, astrocytosis, and neuronal and axonal degeneration increased in a region-specific manner dependent on the severity of myelin loss in pericallosal white matter. The spectrum of injury ranged from mild, where diffuse white matter abnormalities were dominant and were associated with mild axonal injury and local microglial activation, to severe HI injury characterized by focal MBP loss, widespread neuronal degeneration, axonal damage, and gliosis throughout the neocortex, caudate putamen, and thalamus. In sum, selective regional white matter loss occurs in the preterm rat concomitantly with a clinically relevant spectrum of grey matter injury. These data demonstrate an interspecies similarity of brain injury patterns and further substantiates the reliable use of this model for the study of preterm brain injury.
- Published
- 2012
- Full Text
- View/download PDF
95. Developmental regulation of group I metabotropic glutamate receptors in the premature brain and their protective role in a rodent model of periventricular leukomalacia
- Author
-
Delia M. Talos, Wenbin Deng, Lauren L. Jantzie, Li An, Debra B. Selip, Rebecca D. Folkerth, Frances E. Jensen, and Michele Jackson
- Subjects
medicine.medical_specialty ,Leukomalacia, Periventricular ,Receptor, Metabotropic Glutamate 5 ,Excitotoxicity ,Biology ,medicine.disease_cause ,Receptors, Metabotropic Glutamate ,Article ,White matter ,Cellular and Molecular Neuroscience ,chemistry.chemical_compound ,Fetus ,Internal medicine ,mental disorders ,medicine ,Animals ,Humans ,Rats, Long-Evans ,Periventricular leukomalacia ,Metabotropic glutamate receptor 5 ,Glutamate receptor ,Age Factors ,Infant, Newborn ,Brain ,Gene Expression Regulation, Developmental ,Dioxolanes ,Cell Biology ,medicine.disease ,Rats ,Disease Models, Animal ,Oligodendroglia ,Endocrinology ,medicine.anatomical_structure ,chemistry ,nervous system ,Animals, Newborn ,Metabotropic glutamate receptor ,Purines ,Hypoxia-Ischemia, Brain ,ACPD ,Metabotropic glutamate receptor 1 ,Neuroscience ,Galactosylceramidase - Abstract
Cerebral white matter injury in premature infants, known as periventricular leukomalacia (PVL), is common after hypoxia–ischemia (HI). While ionotropic glutamate receptors (iGluRs) can mediate immature white matter injury, we have previously shown that excitotoxic injury to premyelinating oligodendrocytes (preOLs) in vitro can be attenuated by group I metabotropic glutamate receptor (mGluR) agonists. Thus, we evaluated mGluR expression in developing white matter in rat and human brain, and tested the protective efficacy of a central nervous system (CNS)-penetrating mGluR agonist on injury to developing oligodendrocytes (OLs) in vivo. Group I mGluRs (mGluR1 and mGluR5) were strongly expressed on OLs in neonatal rodent cerebral white matter throughout normal development, with highest expression early in development on preOLs. Specifically at P6, mGluR1 and mGLuR5 were most highly expressed on GalC-positive OLs compared to neurons, axons, astrocytes and microglia. Systemic administration of (1S,3R) 1-aminocyclopentane-trans-1,3,-dicarboxylic acid (ACPD) significantly attenuated the loss of myelin basic protein in the white matter following HI in P6 rats. Assessment of postmortem human tissue showed both mGluR1 and mGluR5 localized on immature OLs in white matter throughout development, with mGluR5 highest in the preterm period. These data indicate group I mGluRs are highly expressed on OLs during the peak period of vulnerability to HI and modulation of mGluRs is protective in a rodent model of PVL. Group I mGluRs may represent important therapeutic targets for protection from HI-mediated white matter injury.
- Published
- 2011
96. Oxidative Stress in Neonatal Hypoxic-Ischemic Encephalopathy
- Author
-
Po-Yin Cheung, Lauren L. Jantzie, and Kathryn G. Todd
- Subjects
Asphyxia ,Periventricular leukomalacia ,business.industry ,Encephalopathy ,Ischemia ,Excitotoxicity ,medicine.disease ,Bioinformatics ,medicine.disease_cause ,Neurochemical ,medicine ,Neurochemistry ,medicine.symptom ,business ,Reperfusion injury - Abstract
Despite improvements in the care of asphyxiated neonates, neonatal or perinatal hypoxia-ischemia remains a challenge to clinical practitioners. In this multisystem dysfunction, hypoxic-ischemic encephalopathy contributes to short- and long-term morbidity of these critically ill neonates. In the developing brain of premature and term neonates, there are immature responses to hypoxia-ischemia in the context of selective vulnerability of different brain structures and neural cells at different stages of development. This difference explains at least in part the diversity of clinical presentations and sequelae of neonatal hypoxic-ischemic brain injury. In addition to hypoxic-ischemic damage, cerebral reoxygenation or reperfusion injury plays an important role in the pathophysiology of hypoxic-ischemic brain injury. Mechanisms of cell death and apoptosis operate at multiple levels including oxygen-derived free radical damage and excitotoxicity. Cerebral oxidative stress and neurochemical changes are related in hypoxia-ischemia of the neonatal brain. Controlled reoxygenation to avoid hyperoxia and its related cerebral damage, and novel antioxidative agents such as N-acetylcysteine, are potential therapeutic interventions that may show promise in the improvement of clinical outcome of these asphyxiated neonates with cerebral hypoxic-ischemic injury. The effects of controlled reoxygenation and N-acetylcysteine on the neurochemistry of asphyxiated neonatal brain are discussed.
- Published
- 2010
- Full Text
- View/download PDF
97. The effects of doxycycline administration on amino acid neurotransmitters in an animal model of neonatal hypoxia-ischemia
- Author
-
Kathryn G. Todd, Gail Rauw, and Lauren L. Jantzie
- Subjects
medicine.medical_specialty ,Taurine ,Hippocampal formation ,Biology ,Neuroprotection ,Rats, Sprague-Dawley ,Cellular and Molecular Neuroscience ,chemistry.chemical_compound ,Internal medicine ,medicine ,Animals ,Amino Acids ,Neurotransmitter ,Chromatography, High Pressure Liquid ,chemistry.chemical_classification ,Neurotransmitter Agents ,Glutamate receptor ,Brain ,Cell Biology ,Immunohistochemistry ,Amino acid ,Rats ,Glutamine ,Disease Models, Animal ,Endocrinology ,chemistry ,Biochemistry ,Animals, Newborn ,Doxycycline ,Hypoxia-Ischemia, Brain ,Systemic administration - Abstract
Neonatal hypoxia-ischemia (HI) is a major contributor to many neurological, psychiatric and behavioral disorders. Previous studies in our laboratory have shown that a one-time dose of doxycycline (DOXY), even when given 3h after HI insult, was neuroprotective and significantly reduced microglial activation and cleaved caspase-3 protein expression in the immature brain. In light of these data, the goal of this study was to investigate the effects of DOXY administration on amino acid neurotransmitters. Post-natal-day 7 rats received DOXY (10mg/kg) or vehicle (VEH) concomitant with the onset of HI, and were euthanized 30 min, 1, 2 or 4h post-HI (nor=6). Extracted brains were either immediately dissected for frontal cortex, striatum and hippocampal regions, or removed in their entirety and flash frozen in isopentane for histological analyses. Dissected regions were homogenized and aliquots were prepared for high performance liquid chromatography (HPLC) analyses of amino acid levels and brain levels of DOXY. HPLC extraction revealed that systemic administration of DOXY resulted in mean drug levels of 867.1+/-376.1 ng/g of brain tissue. Histological analyses revealed microglial activation, caspase-3 activation and neuronal degeneration consistent with a mild injury in the regions most vulnerable to HI. We found that HI caused significant, time-dependent, regional changes in brain amino acids including glutamate, GABA, alanine, aspartate, asparagine, serine, glutamine, glycine and taurine. HI significantly increased glutamate levels in the hippocampus (HI+VEH=15.8+/-3.1 ng/microg versus control=11.8+/-1.4 ng/microg protein) 4h post-HI (p0.05). Pups treated with DOXY had lower glutamate levels (13.1+/-2.4 ng/microg) when compared to VEH-treated pups (15.8+/-3.1 ng/microg), however these values failed to reach significance. In addition, DOXY-treated pups had significantly lower alanine (HI+VEH=1.1+/-0.2 ng/microg versus HI+DOXY=0.5+0.1 ng/microg) and serine (HI+VEH=1.4+/-0.4 ng/microg versus HI+DOXY=0.7+0.1 ng/microg) levels in the hippocampus, 4h post-HI. Similar normalizations and significant reductions in alanine and serine were seen in the cortex and striatum. These results show that in addition to its previously reported and well-documented anti-inflammatory and anti-apoptotic properties, DOXY has significant effects on amino acid neurotransmitters.
- Published
- 2006
98. Doxycycline reduces cleaved caspase-3 and microglial activation in an animal model of neonatal hypoxia-ischemia
- Author
-
Kathryn G. Todd, Lauren L. Jantzie, and Po-Yin Cheung
- Subjects
Pathology ,medicine.medical_specialty ,Ischemia ,Brain damage ,Biology ,Hippocampal formation ,Neuroprotection ,Rats, Sprague-Dawley ,Internal medicine ,medicine ,Animals ,Antibacterial agent ,Doxycycline ,Microglia ,Molecular Structure ,Caspase 3 ,medicine.disease ,Immunohistochemistry ,Rats ,Disease Models, Animal ,medicine.anatomical_structure ,Endocrinology ,Neurology ,Animals, Newborn ,Cerebral cortex ,Caspases ,Hypoxia-Ischemia, Brain ,Neurology (clinical) ,medicine.symptom ,Cardiology and Cardiovascular Medicine ,medicine.drug - Abstract
Neonatal hypoxia-ischemia (HI) is a major contributor to many perinatal neurologic disorders and, thus, the search for therapies and effective treatments for the associated brain damage has become increasingly important. The tetracycline derivative, doxycycline (DOXY), has been reported to be neuroprotective in adult animal models of cerebral ischemia. To investigate the putative neuroprotective effects of DOXY in an animal model of neonatal HI, a time-course study was run such that pups received either DOXY (10 mg/kg) or VEH immediately before hypoxia, 1, 2, or 3 hours after HI ( n=6). At 7 days after injury, the pups were euthanized, and the brains were removed and processed for immunohistochemical and Western blot analyses using antibodies against specific markers for neurons, apoptotic markers, microglia, oligodendrocytes, and astrocytes. Results showed that in vulnerable brain regions including the hippocampal formation, thalamus, striatum, cerebral cortex and white matter tracts, DOXY significantly decreased caspase-3 immunoreactivity (a marker of apoptosis), promoted neuronal survival, inhibited microglial activation and reduced reactive astrocytosis compared with VEH-treated HI pups. These effects were found to occur in a time-dependent manner. Taken together, these results strongly suggest that doxycycline has potential as a pharmacological treatment for mild HI in neonates.
- Published
- 2005
99. Inflammation-induced alterations in maternal-fetal Heme Oxygenase (HO) are associated with sustained innate immune cell dysregulation in mouse offspring.
- Author
-
Maide Ozen, Hui Zhao, Flora Kalish, Yang Yang, Lauren L Jantzie, Ronald J Wong, and David K Stevenson
- Subjects
Medicine ,Science - Abstract
Heme oxygenase-1 (HO-1) is an evolutionarily conserved stress response enzyme and important in pregnancy maintenance, fetal and neonatal outcomes, and a variety of pathologic conditions. Here, we investigated the effects of an exposure to systemic inflammation late in gestation [embryonic day (E)15.5] on wild-type (Wt) and HO-1 heterozygous (Het, HO-1+/-) mothers, fetuses, and offspring. We show that alterations in fetal liver and spleen HO homeostasis during inflammation late in gestation can lead to a sustained dysregulation of innate immune cell populations and intracellular myeloid HO-1 expression in the spleen through young adolescence [postnatal day 25] in mice.
- Published
- 2021
- Full Text
- View/download PDF
100. A time for cocktails and inclusion
- Author
-
Shenandoah Robinson, Frances J Northington, and Lauren L Jantzie
- Subjects
Neurology. Diseases of the nervous system ,RC346-429 - Published
- 2018
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.