1
|
Khodir SA, Faried MA, Abd-Elhafiz HI, Sweed EM. Sitagliptin Attenuates the Cognitive Deficits in L-Methionine-Induced Vascular Dementia in Rats. BIOMED RESEARCH INTERNATIONAL 2022; 2022:7222590. [PMID: 35265716 PMCID: PMC8898801 DOI: 10.1155/2022/7222590] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 01/29/2022] [Indexed: 02/05/2023]
Abstract
Vascular dementia (VaD) is the second most prevalent type of dementia characterized by progressive cognitive deficits and is a major risk factor for the development of Alzheimer's disease and other neurodegenerative disorders. This study is aimed at determining the potential neuroprotective effect of sitagliptin (STG) on cognitive deficits in L-methionine-induced VaD in rats and the possible underlying mechanisms. 30 adult male Wistar albino rats were divided equally (n = 10) into three groups: control, VaD, and VaD + STG groups. The cognitive performance of the animals was conducted by open field, elevated plus maze, Y-maze, novel object recognition, and Morris water maze tests. Serum homocysteine, TNF-α, IL-6, IL-10, total cholesterol, and triglycerides levels were assessed together with hippocampal MDA, SOD, and BDNF. Histopathological and immunohistochemical assessments of the thoracic aorta and hippocampus (CA1 region) were also performed. Chronic L-methionine administration impaired memory and learning and induced anxiety. On the other hand, STG protected against cognitive deficits through improving oxidative stress biomarkers, inflammatory mediators, lipid profiles, and hippocampus level of BDNF as well as decreasing caspase-3 and GFAP and increasing Ki-67 immunoreactions in the hippocampus. Also, STG improved the endothelial dysfunction via upregulation of aortic eNOS immunoreaction. STG improved the cognitive deficits of L-methionine-induced VaD by its antioxidant, anti-inflammatory, antiapoptotic, and neurotrophic effects. These findings suggest that STG may be a promising future agent for protection against VaD.
Collapse
Affiliation(s)
- Suzan A. Khodir
- Medical Physiology Department, Faculty of Medicine, Menoufia University, Menoufia 32511, Egypt
| | - Manar A. Faried
- Human Anatomy and Embryology, Faculty of Medicine, Menoufia University, Menoufia 32511, Egypt
| | - Huda I. Abd-Elhafiz
- Clinical Pharmacology Department, Faculty of Medicine, Menoufia University, Menoufia 32511, Egypt
| | - Eman M. Sweed
- Clinical Pharmacology Department, Faculty of Medicine, Menoufia University, Menoufia 32511, Egypt
| |
Collapse
|
2
|
Muk T, Stensballe A, Dmytriyeva O, Brunse A, Jiang PP, Thymann T, Sangild PT, Pankratova S. Differential Brain and Cerebrospinal Fluid Proteomic Responses to Acute Prenatal Endotoxin Exposure. Mol Neurobiol 2022; 59:2204-2218. [PMID: 35064541 DOI: 10.1007/s12035-022-02753-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 01/17/2022] [Indexed: 12/12/2022]
Abstract
Chorioamnionitis (CA) is a risk factor for preterm birth and is associated with neurodevelopmental delay and cognitive disorders. Prenatal inflammation-induced brain injury may resolve during the immediate postnatal period when rapid brain remodeling occurs. Cerebrospinal fluid (CSF) collected at birth may be a critical source of predictive biomarkers. Using pigs as a model of preterm infants exposed to CA, we hypothesized that prenatal lipopolysaccharide (LPS) exposure induces proteome changes in the CSF and brain at birth and postnatally. Fetal piglets (103 days gestation of full-term at 117 days) were administered intra-amniotic (IA) lipopolysaccharide (LPS) 3 days before preterm delivery by caesarian section. CSF and brain tissue were collected on postnatal Days 1 and 5 (P1 and P5). CSF and hippocampal proteins were profiled by LC-MS-based quantitative proteomics. Neuroinflammatory responses in the cerebral cortex, periventricular white matter and hippocampus were evaluated by immunohistochemistry, and gene expression was evaluated by qPCR. Pigs exposed to LPS in utero showed changes in CSF protein levels at birth but not at P5. Complement protein C3, hemopexin, vasoactive intestinal peptide, carboxypeptidase N subunit 2, ITIH1, and plasminogen expression were upregulated in the CSF, while proteins associated with axon growth and synaptic functions (FGFR1, BASP1, HSPD1, UBER2N, and RCN2), adhesion (talin1), and neuronal survival (Atox1) were downregulated. Microglia, but not astrocytes, were activated by LPS at P5 in the hippocampus but not in other brain regions. At this time, marginal increases in complement protein C3, LBP, HIF1a, Basp1, Minpp1, and FGFR1 transcription indicated hippocampal proinflammatory responses. In conclusion, few days exposure to endotoxin prenatally induce proteome changes in the CSF and brain at birth, but most changes resolve a few days later. The developing hippocampus has high neuronal plasticity in response to perinatal inflammation. Changes in CSF protein expression at birth may predict later structural brain damage in preterm infants exposed to variable types and durations of CA-related inflammation in utero.
Collapse
Affiliation(s)
- Tik Muk
- Section of Comparative Pediatrics and Nutrition, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Allan Stensballe
- Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Oksana Dmytriyeva
- Faculty of Health and Medical Sciences, Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Anders Brunse
- Section of Comparative Pediatrics and Nutrition, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ping-Ping Jiang
- Section of Comparative Pediatrics and Nutrition, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Thomas Thymann
- Section of Comparative Pediatrics and Nutrition, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Per Torp Sangild
- Section of Comparative Pediatrics and Nutrition, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Department of Pediatrics and Adolescent Medicine, Rigshospitalet, Copenhagen, Denmark.,Department of Paediatrics, Odense University Hospital, Odense, Denmark
| | - Stanislava Pankratova
- Section of Comparative Pediatrics and Nutrition, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark. .,Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, University of Copenhagen, 1870, Frederiksberg C, Denmark.
| |
Collapse
|
3
|
Najafian SA, Farbood Y, Sarkaki A, Ghafouri S. FTY720 administration following hypoxia-induced neonatal seizure reverse cognitive impairments and severity of seizures in male and female adult rats: The role of inflammation. Neurosci Lett 2021; 748:135675. [PMID: 33516800 DOI: 10.1016/j.neulet.2021.135675] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 12/05/2020] [Accepted: 01/21/2021] [Indexed: 11/15/2022]
Abstract
Hypoxia-induced neonatal seizure mainly leads to deleterious effects on brain function, especially cognitive impairments and increased susceptibility to epilepsy later in life. Early inflammation plays an important role in the pathology of these consequences. Therefore, we explored the long-term outcomes of Fingolimod treatment as an anti-inflammatory and neuroprotective agent in a rat model of HINS. Seizures were induced in rats (postnatal day 10) by 5% O2 exposure for 15 min. Sixty minutes after the onset of hypoxia, pups received FTY720 (0.3 mg.kg-1) or normal saline for 12 consecutive days (lactation period), and they were used at P60-P63 for behavioral tests, ELISA and Pentylenetetrazole kindling model. The results of open field, novel object recognition and elevated plus maze tasks showed that Fingolimod prevents hippocampal memory dysfunction and anxiety-like behavior in both male and female hypoxic groups, which was accompanied with decreased TNF-α level in hippocampus. In addition, FTY720 postponed epileptogenesis just in female hypoxic + FTY group and decreased severity of seizures in both genders. Our results suggest that, FTY720 treatment in immature rats, which were previously subjected to HINS, prevented the long-lasting deficits, like cognitive impairments, decreased the severity of seizures and related inflammation. In addition, FTY720 did not show significant interaction with gender in most of the experiments, except the average day to reach fully kindled state. Taken together, FTY720 has therapeutic potential for long lasting effects of HINS in both male and female animals at puberty.
Collapse
Affiliation(s)
- Seyed Ahmad Najafian
- Department of Physiology, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Yaghoob Farbood
- Department of Physiology, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran; Persian Gulf Physiology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Alireza Sarkaki
- Department of Physiology, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran; Persian Gulf Physiology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Samireh Ghafouri
- Department of Physiology, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran; Persian Gulf Physiology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| |
Collapse
|
4
|
Özevren H, Deveci E, Tuncer MC. The effect of rosmarinic acid on deformities occurring in brain tissue by craniectomy method. Histopathological evaluation of IBA-1 and GFAP expressions. Acta Cir Bras 2020; 35:e202000406. [PMID: 32578724 PMCID: PMC7307720 DOI: 10.1590/s0102-865020200040000006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 02/09/2020] [Accepted: 03/04/2020] [Indexed: 12/20/2022] Open
Abstract
PURPOSE To investigate the role of Rosmarinic acid (RA) in the prevention of traumatic brain injury and the immunohistochemical analysis of IBA-1 and GFAP expressions. METHODS Healthy male rats were randomly divided into 3 groups consisting of 10 rats. Groups were as follows; control group, traumatic brain injury (TBI) group, and TBI+RA group. After traumatic brain injury, blood samples were taken from the animals and analyzed with various biochemical markers. And then IBA-1 and GFAP expressions were evaluated immunohistochemically. RESULTS Significant results were obtained in all biochemical parameters between groups. Immunohistochemical sections showed IBA-1 not only in microglia and macrophage activity but also in degenerative neurons in blood vessel endothelial cells. However, GFAP reaction and post-traumatic rosmarinic acid administration showed positive expression in astrocytes with regular structure around the blood vessel. CONCLUSION Rosmarinic acid in blood vessel endothelial cells showed that preserving the integrity of astrocytic structure in the blood brain barrier may be an important antioxidant.
Collapse
Affiliation(s)
- Hüseyin Özevren
- Associate Professor, Department of Neurosurgery , Faculty of
Medicine , Dicle University , Diyarbakır , Turkey . Technical procedures, manuscript
preparation and writing, final approval
| | - Engin Deveci
- PhD, Professor, Department of Histology and Embryology , Faculty
of Medicine , Dicle University , Diyarbakır , Turkey . Technical procedures,
histopathological examinations, manuscript preparation and writing, final
approval
| | - Mehmet Cudi Tuncer
- PhD, Professor, Department of Anatomy , Faculty of Medicine ,
Dicle University , Diyarbakır , Turkey . Technical procedures, histopathological
examinations, manuscript preparation and writing, final approval
| |
Collapse
|
5
|
Quinlan S, Merino-Serrais P, Di Grande A, Dussmann H, Prehn JHM, Ní Chonghaile T, Henshall DC, Jimenez-Mateos EM. The Anti-inflammatory Compound Candesartan Cilexetil Improves Neurological Outcomes in a Mouse Model of Neonatal Hypoxia. Front Immunol 2019; 10:1752. [PMID: 31396238 PMCID: PMC6667988 DOI: 10.3389/fimmu.2019.01752] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 07/10/2019] [Indexed: 11/15/2022] Open
Abstract
Recent studies suggest that mild hypoxia-induced neonatal seizures can trigger an acute neuroinflammatory response leading to long-lasting changes in brain excitability along with associated cognitive and behavioral deficits. The cellular elements and signaling pathways underlying neuroinflammation in this setting remain incompletely understood but could yield novel therapeutic targets. Here we show that brief global hypoxia-induced neonatal seizures in mice result in transient cytokine production, a selective expansion of microglia and long-lasting changes to the neuronal structure of pyramidal neurons in the hippocampus. Treatment of neonatal mice after hypoxia-seizures with the novel anti-inflammatory compound candesartan cilexetil suppressed acute seizure-damage and mitigated later-life aggravated seizure responses and hippocampus-dependent learning deficits. Together, these findings improve our understanding of the effects of neonatal seizures and identify potentially novel treatments to protect against short and long-lasting harmful effects.
Collapse
Affiliation(s)
- Sean Quinlan
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Paula Merino-Serrais
- Division for Neurogeriatrics, Department of Neurobiology Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden.,Departamento de Neurobiologia Funcional y de Sistemas, Instituto Cajal, Consejo Superior de Investigaciones Cientificas, Madrid, Spain
| | - Alessandra Di Grande
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Heiko Dussmann
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Jochen H M Prehn
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland.,FutureNeuro Research Centre, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Tríona Ní Chonghaile
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - David C Henshall
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland.,FutureNeuro Research Centre, Royal College of Surgeons in Ireland, Dublin, Ireland.,INFANT Research Centre, UCC, Cork, Ireland
| | - Eva M Jimenez-Mateos
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland.,Department of Physiology, School of Medicine, Trinity College Dublin, The University of Dublin, Dublin, Ireland
| |
Collapse
|
6
|
Berkiks I, Garcia-Segura L, Nassiri A, Mesfioui A, Ouichou A, Boulbaroud S, Bahbiti Y, Lopez-Rodriguez A, Hasnaoui E, El Hessni A. The sex differences of the behavior response to early Life immune stimulation: Microglia and astrocytes involvement. Physiol Behav 2019; 199:386-394. [DOI: 10.1016/j.physbeh.2018.11.037] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 10/20/2018] [Accepted: 11/28/2018] [Indexed: 01/01/2023]
|
7
|
Izvolskaia M, Sharova V, Zakharova L. Prenatal Programming of Neuroendocrine System Development by Lipopolysaccharide: Long-Term Effects. Int J Mol Sci 2018; 19:ijms19113695. [PMID: 30469423 PMCID: PMC6274672 DOI: 10.3390/ijms19113695] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 11/16/2018] [Accepted: 11/19/2018] [Indexed: 12/12/2022] Open
Abstract
Various stress factors during critical periods of fetal development modulate the epigenetic mechanisms controlling specific genes, which can affect the structure and function of physiological systems. Maternal immune stress by bacterial infection simulated by lipopolysaccharide (LPS) in an experiment is considered to be a powerful programming factor of fetal development. Studies of the molecular mechanisms controlling the formation and functioning of physiological systems are in the pilot stage. LPSs are the most potent natural inflammation factors. LPS-induced increases in fetal levels of pro- and anti-inflammatory cytokines can affect brain development and have long-term effects on behavior and neuroendocrine functions. The degradation of serotonergic neurons induced by LPS in the fetus is attributed to the increased levels of interleukin (IL)-6 and tumor necrosis factor (TNFα) as well as to anxiety and depression in children. Dopamine deficiency causes dysthymia, learning disability, and Parkinson’s disease. According to our data, an LPS-induced increase in the levels of IL-6, leukemia inhibitory factor (LIF), and monocyte chemotactic protein (MCP-1) in maternal and fetal rats during early pregnancy disturbs the development and functioning of gonadotropin-releasing hormone production and reproductive systems. It is important to note the high responsiveness of epigenetic developmental mechanisms to many regulatory factors, which offers opportunities to correct the defects.
Collapse
Affiliation(s)
- Marina Izvolskaia
- Koltsov Institute of Developmental Biology, Russian Academy of Sciences, 119334 Moscow, Russia.
| | - Viktoria Sharova
- Koltsov Institute of Developmental Biology, Russian Academy of Sciences, 119334 Moscow, Russia.
| | - Liudmila Zakharova
- Koltsov Institute of Developmental Biology, Russian Academy of Sciences, 119334 Moscow, Russia.
| |
Collapse
|
8
|
Edlow AG, Glass RM, Smith CJ, Tran PK, James K, Bilbo S. Placental Macrophages: A Window Into Fetal Microglial Function in Maternal Obesity. Int J Dev Neurosci 2018; 77:60-68. [PMID: 30465871 DOI: 10.1016/j.ijdevneu.2018.11.004] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 11/05/2018] [Accepted: 11/14/2018] [Indexed: 01/18/2023] Open
Abstract
Fetal placental macrophages and microglia (resident brain macrophages) have a common origin in the fetal yolk sac. Yolk-sac-derived macrophages comprise the permanent pool of brain microglia throughout an individual's lifetime. Inappropriate fetal microglial priming may therefore have lifelong neurodevelopmental consequences, but direct evaluation of microglial function in a living fetus or neonate is impossible. We sought to test the hypothesis that maternal obesity would prime both placental macrophages and fetal brain microglia to overrespond to an immune challenge, thus providing a window into microglial function using placental cells. Obesity was induced in C57BL/6 J mice using a 60% high-fat diet. On embryonic day 17.5, fetal brain microglia and corresponding CD11b + placental cells were isolated from fresh tissue. Cells were treated with media or lipopolysaccharide (LPS). Tumor necrosis factor-alpha (TNF-α) production by stimulated and unstimulated cells was quantified via ELISA. We demonstrate for the first time that the proinflammatory cytokine production of CD11b + placental cells is strongly correlated with that of brain microglia (Spearman's ρ = 0.73, p = 0.002) in the setting of maternal obesity. Maternal obesity-exposed CD11b + cells had an exaggerated response to LPS compared to controls, with a 5.1-fold increase in TNF-α production in placentas (p = 0.003) and 3.8-fold increase in TNF-α production in brains (p = 0.002). In sex-stratified analyses, only male obesity-exposed brains and placentas had significant increase in TNF-α production in response to LPS. Taken together, these data suggest that maternal obesity primes both placental macrophages and fetal brain microglia to overproduce a proinflammatory cytokine in response to immune challenge. Male brain and placental immune response is more marked than female in this setting. Given that fetal microglial priming may impact neuroimmune function throughout the lifespan, these data could provide insight into the male predominance of certain neurodevelopmental morbidities linked to maternal obesity, including cognitive dysfunction, autism spectrum disorder, and ADHD. Placental CD11b+ macrophages may have the potential to serve as an accessible biomarker of aberrant fetal brain immune activation in maternal obesity. This finding may have broader implications for assaying the impact of other maternal exposures on fetal brain development.
Collapse
Affiliation(s)
- Andrea G Edlow
- Obstetrics, Gynecology, and Reproductive Biology, Harvard Medical School, Massachusetts General Hospital, Vincent Center for Reproductive Biology
| | - Ruthy M Glass
- Obstetrics, Gynecology, and Reproductive Biology, Harvard Medical School, Massachusetts General Hospital, Vincent Center for Reproductive Biology
| | - Caroline J Smith
- Pediatrics and Program in Neuroscience, Harvard Medical School, Lurie Center for Autism, MassGeneral Hospital for Children
| | - Phuong Kim Tran
- Pediatrics and Program in Neuroscience, Harvard Medical School, Lurie Center for Autism, MassGeneral Hospital for Children
| | - Kaitlyn James
- Massachusetts General Hospital, Deborah Kelly Center for Outcomes Research
| | - Staci Bilbo
- Pediatrics and Program in Neuroscience, Harvard Medical School, Lurie Center for Autism, MassGeneral Hospital for Children
| |
Collapse
|
9
|
Thymelaea lythroides extract attenuates microglial activation and depressive-like behavior in LPS-induced inflammation in adult male rats. Biomed Pharmacother 2018; 99:655-663. [PMID: 29710462 DOI: 10.1016/j.biopha.2018.01.125] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Revised: 01/13/2018] [Accepted: 01/24/2018] [Indexed: 12/29/2022] Open
Abstract
Thymelaea lythroides extract is widely used as a traditional folk medicine in Morocco, especially for the treatment of diabetes, rheumatism and Inflammatory disease. The aim of the study is to evaluate the possible effect of methanolic extract of Thymelaea lythroides in repressing the inflammatory responses and long-lasting depression-like behavior associated with neuroinflammation in adult rats after neonatal LPS exposure. Male rat pups were treated systemically with either LPS (250??g/kg) or vehicle (phosphate buffer saline) on postnatal day 14. Six hours later, the LPS groups were assigned to intraperitoneal (ip) injection of Minocycline (50?mg/kg) or Thymelaea lythroides (200?mg/kg). Thereafter, in adulthood (postnatal days 90-97), the spontaneous locomotor activity and depression-like behavior were assessed successively in open field and forced swim tests. The levels of proinflammatory cytokines, oxidative damage, and activation of microglia were determined in the hippocampus (HP) of male rats on (PND90-97). Our results showed that open field hypoactivity and increased immobility period in LPS-induced adult rats were normalized on treatment with Thymelaea lythroides and minocycline. Both treatments attenuate the overactivated microglial cells in the CA1 and CA3 of hippocampus (HP) and significantly reduced the oxidative-nitrosative stress markers and cytokine (TNF ?) production in the HP. Thymelaea lythroides seems to have similar neuroprotective effects to Minocycline, and such protection may be due to: reduction of oxidative stress, upregulation of inflammatory mediators production, antidepressant behavior which all are associated with neuroinflammation.
Collapse
|
10
|
Bilbo SD, Block CL, Bolton JL, Hanamsagar R, Tran PK. Beyond infection - Maternal immune activation by environmental factors, microglial development, and relevance for autism spectrum disorders. Exp Neurol 2017; 299:241-251. [PMID: 28698032 DOI: 10.1016/j.expneurol.2017.07.002] [Citation(s) in RCA: 207] [Impact Index Per Article: 25.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 06/26/2017] [Accepted: 07/05/2017] [Indexed: 12/17/2022]
Abstract
Immune molecules such as cytokines and chemokines and the cells that produce them within the brain, notably microglia, are critical for normal brain development. This recognition has in recent years led to the working hypothesis that inflammatory events during pregnancy, e.g. in response to infection, may disrupt the normal expression of immune molecules during critical stages of neural development and thereby contribute to the risk for neurodevelopmental disorders such as autism spectrum disorder (ASD). This hypothesis has in large part been shepherded by the work of Dr. Paul Patterson and colleagues, which has elegantly demonstrated that a single viral infection or injection of a viral mimetic to pregnant mice significantly and persistently impacts offspring immune and nervous system function, changes that underlie ASD-like behavioral dysfunction including social and communication deficits. Subsequent studies by many labs - in humans and in non-human animal models - have supported the hypothesis that ongoing disrupted immune molecule expression and/or neuroinflammation contributes to at least a significant subset of ASD. The heterogeneous clinical and biological phenotypes observed in ASD strongly suggest that in genetically susceptible individuals, environmental risk factors combine or synergize to create a tipping or threshold point for dysfunction. Importantly, animal studies showing a link between maternal immune activation (MIA) and ASD-like outcomes in offspring involve different species and diverse environmental factors associated with ASD in humans, beyond infection, including toxin exposures, maternal stress, and maternal obesity, all of which impact inflammatory or immune pathways. The goal of this review is to highlight the broader implications of Dr. Patterson's work for the field of autism, with a focus on the impact that MIA by diverse environmental factors has on fetal brain development, immune system development, and the pathophysiology of ASD.
Collapse
Affiliation(s)
- Staci D Bilbo
- Pediatrics and Neuroscience, Harvard Medical School, Lurie Center for Autism, Massachusetts General Hospital for Children, Boston, MA 02126, United States.
| | - Carina L Block
- Psychology and Neuroscience, Duke University, Durham, NC 27708, United States
| | - Jessica L Bolton
- Pediatrics and Anatomy/Neurobiology, University of California-Irvine, Irvine, CA 92697, United States
| | - Richa Hanamsagar
- Pediatrics and Neuroscience, Harvard Medical School, Lurie Center for Autism, Massachusetts General Hospital for Children, Boston, MA 02126, United States
| | - Phuong K Tran
- Pediatrics and Neuroscience, Harvard Medical School, Lurie Center for Autism, Massachusetts General Hospital for Children, Boston, MA 02126, United States
| |
Collapse
|
11
|
Experimental Zika virus infection induces spinal cord injury and encephalitis in newborn Swiss mice. ACTA ACUST UNITED AC 2016; 69:63-71. [PMID: 27899230 DOI: 10.1016/j.etp.2016.11.004] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Revised: 11/02/2016] [Accepted: 11/21/2016] [Indexed: 11/20/2022]
Abstract
A widespread epidemic of Zika virus (ZIKV) infection was reported in 2015 in South and Central America, with neurological symptons including meningoencephalitis and Guillain-Barré syndrome in adults, besides an apparent increased incidence of microcephaly in infants born to infected mothers. It is becoming a necessity to have a trustworthy animal model to better understand ZIKV infection. In this study we used newborn white Swiss mice as a model to investigate the ZIKV strain recently isolated in Brazil. ZIKV was inoculated via intracerebral and subcutaneous routes and analysed through gross histopathology and immunohistochemistry. Here we demonstrated first that the intracerebral group (ICG) displayed severe cerebral lesions, with neuronal death, presence of apoptotic bodies, white matter degeneration and neutrophil perivascular cuffing. In the subcutaneous group (SCG), we observed moderate cerebral lesions, morphologically similar to that found in ICG and additional myelopathy, with architectural loss, marked by neuronal death and apoptotic bodies. Interestingly, we found an intense astrogliosis in brain of both groups, with increased immunoexpression of GFAP (glial fibrillary acidic protein) and presence of hypertrophic astrocytes. The spinal cord of subcutaneous group (SCG) exhibited reduction of astrocytes, but those positive for GFAP were hypertrophic and presented prolonged cellular processes. Finally significant lesions in the central nervous system (CNS) were present in newborn mice inoculated by both routes, but SCG method led to an important neurological manifestations (including myelopathy), during a longer period of time and appears for us to be a better model for ZIKV infection.
Collapse
|
12
|
Jiang P, Zhu T, Xia Z, Gao F, Gu W, Chen X, Yuan T, Yu H. Inhibition of MAPK/ERK signaling blocks hippocampal neurogenesis and impairs cognitive performance in prenatally infected neonatal rats. Eur Arch Psychiatry Clin Neurosci 2015; 265:497-509. [PMID: 25721317 DOI: 10.1007/s00406-015-0588-y] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2014] [Accepted: 02/19/2015] [Indexed: 12/20/2022]
Abstract
Hippocampus endogenous neurogenesis has been postulated to play a favorable role in brain restoration after injury. However, the underlying molecular mechanisms have been insufficiently deciphered. Here we investigated the potential regulatory capacity of MAPK/ERK signaling on neurogenesis and the associated cognitive performance in prenatally infected neonatal rats. From our data, intrauterine infection could induce hippocampal neuronal apoptosis and promote endogenous repair by evoking neural stem cell proliferation and survival. We also found intrauterine infection could induce increased levels of p-ERK, p-CREB and BDNF, which might be responsible for the potential endogenous rescue system. Furthermore, inhibition of MAPK/ERK signaling could aggravate hippocampal neuronal apoptosis, decrease neurogenesis, and impair the offspring's cognitive performances and could also down-regulate the levels of p-ERK, p-CREB and BDNF. Our data strongly suggest that the activation of MAPK/ERK signaling may play a significant role in promoting survival of newly generated neural stem cells via an anti-apoptotic mechanism, which may be particularly important in endogenous neuroprotection associated with cognitive performance development in prenatally infected rats.
Collapse
Affiliation(s)
- Peifang Jiang
- Department of Neurology, Children's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | | | | | | | | | | | | | | |
Collapse
|
13
|
Retraction notice to “THE EFFECT OF MATERNAL INFECTION ON COGNITIVE DEVELOPMENT AND HIPPOCAMPUS NEURONAL APOPTOSIS, PROLIFERATION AND DIFFERENTIATION IN THE NEONATAL RATS” [Neuroscience 246 (2013) 422–434]. Neuroscience 2013; 246:422-34. [DOI: 10.1016/j.neuroscience.2013.04.021] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2013] [Revised: 04/04/2013] [Accepted: 04/05/2013] [Indexed: 01/29/2023]
|
14
|
The persistent effects of maternal infection on the offspring's cognitive performance and rates of hippocampal neurogenesis. Prog Neuropsychopharmacol Biol Psychiatry 2013; 44:279-89. [PMID: 23562668 DOI: 10.1016/j.pnpbp.2013.03.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2012] [Revised: 02/27/2013] [Accepted: 03/26/2013] [Indexed: 01/20/2023]
Abstract
Accumulating evidence indicates that perinatal infection is a major cause of neonatal neurologic morbidity. Here we explored the effects of maternal infection on the offspring's cognitive performance and hippocampal neurogenesis. Pregnant rats were treated with Escherichia coli suspension and allowed to deliver. Proliferating cells in the hippocampus were examined at postnatal (P) 3, 7, 14, and 28 days and neuronal survival/differentiation was assessed at P28. Additionally, we examined the expressions of BDNF, TrkB and Akt. The cognitive performance of the offspring was assessed by the Morris water maze test. We found that maternal infection significantly impaired the offspring's spatial learning ability and spatial memory, thus could delay the cognitive performance development. Maternal infection significantly increased the number of proliferating cells in the offspring's hippocampus at postnatal 3, 7 and 14 days, accompanied by significantly increased expressions of BDNF, TrkB and p-Akt at postnatal 3 and 7 days. On postnatal 28 days, maternal infection did not significantly affect the neuronal and glial differentiation, nor any significant changes in the expression levels of BDNF and TrkB in the hippocampus. Our result suggests that the hippocampal neurogenesis level may increase during early postnatal period after maternal infection. Increase of BDNF/TrkB expression and Akt activity may be the contributing molecular mechanism.
Collapse
|
15
|
Jiang P, Sun Y, Zhu T, Zhan C, Gu W, Yuan T, Yu H. Endogenous neurogenesis in the hippocampus of developing rat after intrauterine infection. Brain Res 2012; 1459:1-14. [DOI: 10.1016/j.brainres.2012.03.058] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2011] [Revised: 02/28/2012] [Accepted: 03/25/2012] [Indexed: 11/17/2022]
|
16
|
Bilbo SD, Smith SH, Schwarz JM. A lifespan approach to neuroinflammatory and cognitive disorders: a critical role for glia. J Neuroimmune Pharmacol 2012; 7:24-41. [PMID: 21822589 PMCID: PMC3267003 DOI: 10.1007/s11481-011-9299-y] [Citation(s) in RCA: 89] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2011] [Accepted: 07/14/2011] [Indexed: 12/15/2022]
Abstract
Cognitive decline is a common problem of aging. Whereas multiple neural and glial mechanisms may account for these declines, microglial sensitization and/or dystrophy has emerged as a leading culprit in brain aging and dysfunction. However, glial activation is consistently observed in normal brain aging as well, independent of frank neuroinflammation or functional impairment. Such variability suggests the existence of additional vulnerability factors that can impact neuronal-glial interactions and thus overall brain and cognitive health. The goal of this review is to elucidate our working hypothesis that an individual's risk or resilience to neuroinflammatory disorders and poor cognitive aging may critically depend on their early life experience, which can change immune reactivity within the brain for the remainder of the lifespan. For instance, early-life infection in rats can profoundly disrupt memory function in young adulthood, as well as accelerate age-related cognitive decline, both of which are linked to enduring changes in glial function that occur in response to the initial infection. We discuss these findings within the context of the growing literature on the role of immune molecules and neuroimmune crosstalk in normal brain development. We highlight the intrinsic factors (e.g., chemokines, hormones) that regulate microglial development and their colonization of the embryonic and postnatal brain, and the capacity for disruption or "re-programming" of this crucial process by external events (e.g., stress, infection). An impact on glia, which in turn alters neural development, has the capacity to profoundly impact cognitive and mental health function at all stages of life.
Collapse
Affiliation(s)
- Staci D Bilbo
- Department of Psychology & Neuroscience, Duke University, Durham, NC 27708, USA.
| | | | | |
Collapse
|
17
|
Prenatal exposure to lipopolysaccharide results in neurodevelopmental damage that is ameliorated by zinc in mice. Brain Behav Immun 2012; 26:326-36. [PMID: 22024135 DOI: 10.1016/j.bbi.2011.10.002] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2011] [Revised: 09/22/2011] [Accepted: 10/09/2011] [Indexed: 01/24/2023] Open
Abstract
There is converging evidence that during pregnancy a maternal immune response to infection can cause neurodevelopmental damage. Lipopolysaccharide (LPS)-mediated induction of metallothionein (MT) and subsequent hypozincaemia has been linked to fetal brain damage. Our group has demonstrated that Zn, when co-administered with LPS in early pregnancy in mice (gestation day (GD) 8), prevents fetal malformations and neurodevelopmental deficits in offspring. Others demonstrating fetal brain lesions have administered LPS much later in gestation (after GD 16), when the influence of LPS-mediated MT-induction on maternal plasma Zn levels, and the effect of Zn co-administration with LPS, are unknown. The aims of this study are firstly to examine whether LPS causes MT induction and maternal hypozincaemia in mid-to-late pregnancy, and secondly to determine if histochemical markers of inflammatory damage in fetal brain are affected by LPS and whether this damage can be alleviated with Zn treatment. Pregnant mice were injected with LPS (5 mg/kgbodywt.) or saline vehicle on GD 16 and then humanely killed at 8, 16 and 24 h for Zn and MT measurements, or concomitantly injected subcutaneously with Zn (2 mg/kgbodywt.) or saline and then killed on GD 18 and immunohistochemistry performed on fetal brain. Maternal hepatic MT was markedly induced after LPS-challenge and this was associated with a 38% reduction in maternal plasma Zn concentrations. Coincidentally, the fetuses of LPS-treated dams showed astrogliosis, extensive cell death and an increased number of cells producing TNF-α which was prevented with concomitant Zn treatment. These results support the premise that in mid-to-late pregnancy, an infection-mediated activation of a maternal immune response can cause MT induction that redistributes Zn in the mother, restricting fetal Zn supply, causing neurodevelopmental damage.
Collapse
|
18
|
Chronic fetal hypoxia produces selective brain injury associated with altered nitric oxide synthases. Am J Obstet Gynecol 2011; 204:254.e16-28. [PMID: 21272843 DOI: 10.1016/j.ajog.2010.11.032] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2010] [Revised: 10/18/2010] [Accepted: 11/09/2010] [Indexed: 11/21/2022]
Abstract
OBJECTIVE The purpose of this study was to investigate the impact of chronic hypoxia on the nitric oxide synthase isoenzymes in specific brain structures. STUDY DESIGN Time-mated pregnant guinea pigs were exposed to 10.5% molecular oxygen for 14 days (animals with chronic fetal hypoxia; HPX) or room air (control animals; NMX); L-N6-(1-iminoethyl)-lysine (L-NIL; an inducible nitric oxide synthase inhibitor, 1 mg/kg/d) was administered to HPX group for 14 days (L-NIL + HPX). Fetal brains were harvested at term. Multilabeled immunofluorescence was used to generate a brain injury map. Laser capture microdissection and quantitative polymerase chain reaction were applied; cell injury markers, apoptosis activation, neuron loss, total nitric oxide, and the levels of individual nitric oxide synthase isoenzymes were quantified. RESULTS Chronic hypoxia causes selective fetal brain injury rather than global. Injury is associated with differentially affected nitric oxide synthases in both neurons and glial cells, with inducible macrophage-type nitric oxide synthase up-regulated at all injury sites. L-NIL attenuated the injury, despite continued hypoxia. CONCLUSION These studies demonstrate that chronic hypoxia selectively injures the fetal brain in part by the differential regulation of nitric oxide synthase isoenzymes in an anatomic- and cell-specific manner.
Collapse
|
19
|
|
20
|
Izvol’skaya MS, Sharova VS, Zakharova LA. Mechanisms of hypothalamic-pituitary and immune system regulation: The role of gonadotropin-releasing hormone and immune mediators. BIOL BULL+ 2010. [DOI: 10.1134/s1062359010040084] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
21
|
Rico JLR, Ferraz DB, Ramalho-Pinto FJ, Morato S. Neonatal exposure to LPS leads to heightened exploratory activity in adolescent rats. Behav Brain Res 2010; 215:102-9. [PMID: 20620170 DOI: 10.1016/j.bbr.2010.07.001] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2010] [Revised: 06/23/2010] [Accepted: 07/02/2010] [Indexed: 01/30/2023]
Abstract
Although several reports have demonstrated physiological and behavioral changes in adult rats due to neonatal immune challenges, little is known about their effects in adolescence. Since neonatal exposure to lipopolysaccharide (LPS) alters the neural substrates involved in cognitive disorders, we tested the hypothesis that it may also alter the response to novel environments in adolescent rats. At 3 and 5 days of age, male Wistar rats received intraperitoneal injections of either vehicle solution or E. coli LPS (0.05mg/kg) or were left undisturbed. In the mid-adolescent period, between 40 and 46 days of age, the rats were exposed to the following behavioral tests: elevated plus-maze, open-field, novel-object exploration task, hole-board and the modified Porsolt forced swim test. The results showed that, in comparison with control animals, LPS-treated rats exhibited (1) less anxiety-related behaviors and enhanced patterns of locomotion and rearing in the plus-maze and the open-field tests, (2) high levels of exploration of both objects in the novel-object task and of corner and central holes in hole-board test, and (3) more time spent diving, an active behavior in the forced swim test. The present findings suggest that neonatal LPS exposure has long-lasting effects on the behavior profile adolescent rats exhibit in response to novelty. This behavioral pattern, characterized by heightened exploratory activity in novel environments, also suggests that early immune stimulation may contribute to the development of impulsive behavior in adolescent rats.
Collapse
|
22
|
Guo R, Hou W, Dong Y, Yu Z, Stites J, Weiner CP. Brain injury caused by chronic fetal hypoxemia is mediated by inflammatory cascade activation. Reprod Sci 2010; 17:540-8. [PMID: 20360591 DOI: 10.1177/1933719110364061] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The prevalence of cerebral palsy (CP) shows little temporal or geographic variation and is associated with preterm birth, maternal/fetal infection/inflammation, and fetal growth restriction (IUGR), a potential surrogate for chronic fetal hypoxemia (CHX). We previously demonstrated CHX causes a fetal inflammatory response syndrome (FIRS). Herein, we test the hypothesis that CHX may cause fetal brain injury by upregulating inflammatory cytokine cascades, culminating in apoptosis pathway activation. Time-mated guinea pigs were housed in 12% or 10.5% O(2) for the last 21% of gestation. Chronic fetal hypoxemia increased the lactate/pyruvate and decreased the glutathione (GSH)/oxidized glutathione (GSSH) ratios, confirming a shift to a prooxidant state. The end result was a >30% decrease in hippocampal neuron density. Based on a microarray spotted with 113 cytokines and receptors, 22 genes were upregulated by CHX in proportion to the degree of hypoxia; the findings were confirmed by quantitative polymerase chain reaction (PCR). Thus, CHX triggers fetal brain inflammation inversely proportional to its severity characterized by increased apoptosis and neuronal loss. We suggest CHX fetal brain injury is not directly caused by oxygen deprivation but rather is an adaptive response that becomes maladaptive.
Collapse
Affiliation(s)
- Rong Guo
- Department of Pathophysiology, Xian Jiaotong University School of Medicine, Xian, Shannxi, PR China
| | | | | | | | | | | |
Collapse
|
23
|
Yuan TM, Yu HM. Notch signaling: key role in intrauterine infection/inflammation, embryonic development, and white matter damage? J Neurosci Res 2010; 88:461-8. [PMID: 19768798 DOI: 10.1002/jnr.22229] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The mechanisms or pathophysiologies that lead to cerebral white matter damage during development are complex and not fully understood. It is postulated that exposure of the preterm brain to inflammatory cytokines during intrauterine infection/inflammation contributes to brain white matter damage, and this damage may affect the function and differentiation of progenitor oligodendrocyte cells under physiological conditions. The Notch pathway, an important signaling pathway controlling various cells' differentiation, functions in the timing of oligodendrocyte differentiation, and Notch signaling may contribute to white matter damage and may mediate neurogenesis in a pathophysiological phase. Recent studies have led to recognition of the role of the Notch pathway in neurogenesis in cerebral ischemic damage and in myelination and axonal damage of neurodegenerative diseases. Moreover, Notch plays a critical role in steering an immune response toward inflammation by regulating expression of various cytokines and proinflammatory cytokines resulting in the activation of Notch signaling. Thus, the Notch signaling pathway likely plays a key role in intrauterine infection/inflammation, brain development, and white matter damage, and future research directed toward understanding its role will be important. Insofar as Notch signaling could have an important effect on neurogenesis, mobilization of progenitor cells is one strategy for compensating for the neuronal losses seen in white matter damage after intrauterine infection/inflammation.
Collapse
Affiliation(s)
- Tian-Ming Yuan
- Department of Neonatology, Children's Hospital, Zhejiang University, School of Medicine, Hangzhou, People's Republic of China
| | | |
Collapse
|
24
|
Shen Y, Yu HM, Yuan TM, Gu WZ, Wu YD. Erythropoietin attenuates white matter damage, proinflammatory cytokine and chemokine induction in developing rat brain after intra-uterine infection. Neuropathology 2009; 29:528-35. [DOI: 10.1111/j.1440-1789.2009.01005.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
25
|
Bilbo SD, Schwarz JM. Early-life programming of later-life brain and behavior: a critical role for the immune system. Front Behav Neurosci 2009; 3:14. [PMID: 19738918 PMCID: PMC2737431 DOI: 10.3389/neuro.08.014.2009] [Citation(s) in RCA: 467] [Impact Index Per Article: 29.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2009] [Accepted: 07/22/2009] [Indexed: 12/12/2022] Open
Abstract
The immune system is well characterized for its critical role in host defense. Far beyond this limited role however, there is mounting evidence for the vital role the immune system plays within the brain, in both normal, “homeostatic” processes (e.g., sleep, metabolism, memory), as well as in pathology, when the dysregulation of immune molecules may occur. This recognition is especially critical in the area of brain development. Microglia and astrocytes, the primary immunocompetent cells of the CNS, are involved in every major aspect of brain development and function, including synaptogenesis, apoptosis, and angiogenesis. Cytokines such as tumor necrosis factor (TNF)α, interleukin [IL]-1β, and IL-6 are produced by glia within the CNS, and are implicated in synaptic formation and scaling, long-term potentiation, and neurogenesis. Importantly, cytokines are involved in both injury and repair, and the conditions underlying these distinct outcomes are under intense investigation and debate. Evidence from both animal and human studies implicates the immune system in a number of disorders with known or suspected developmental origins, including schizophrenia, anxiety/depression, and cognitive dysfunction. We review the evidence that infection during the perinatal period of life acts as a vulnerability factor for later-life alterations in cytokine production, and marked changes in cognitive and affective behaviors throughout the remainder of the lifespan. We also discuss the hypothesis that long-term changes in brain glial cell function underlie this vulnerability.
Collapse
Affiliation(s)
- Staci D Bilbo
- Department of Psychology & Neuroscience, Duke University Durham, NC, USA.
| | | |
Collapse
|
26
|
Shen Y, Yu HM, Yuan TM, Gu WZ, Wu YD. Intrauterine infection induced oligodendrocyte injury and inducible nitric oxide synthase expression in the developing rat brain. J Perinat Med 2007; 35:203-9. [PMID: 17480148 DOI: 10.1515/jpm.2007.058] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
AIMS In order to investigate the neuropathological effect on the developing rat brain after intrauterine infection, 2', 3'-cyclic nucleotide 3'-phosphodiesterase (CNPase) and inducible nitric oxide synthase (iNOS) were evaluated. METHODS Escherichia coli (E. coli) was inoculated into the uterine cervix of the time-pregnant rats and controls were injected with normal saline. Immunohistochemical staining for CNPase was performed to assess oligodendrocyte injury in pup brains at postnatal day 1, 3 and 7 (P1, P3, and P7). Immunohistochemistry was used to evaluate iNOS expression and quantitative reverse transcriptase PCR to analyze iNOS mRNA expression in pup brains at P1, P3 and P7. Nitrate reductase method was used for detection of nitric oxide (NO) concentration in pup brains at P1, P3 and P7. RESULTS The immunohistochemical staining for CNPase in the E. coli-treated group showed a decrease compared with the control in periventricular white matter at P7. Obvious immunohistochemical staining of iNOS was observed in periventricular white matter of the E. coli-treated pup brains at P1. The expression of iNOS mRNA in the E. coli-treated pup brains increased at P1 and P3, but there was no significant difference at P7 compared with controls. Similarly, the NO concentration increased in the E. coli-treated pup brains at P1 and P3, and no significant difference was found at P7 compared with controls. CONCLUSIONS The alteration of CNPase expression indicates that intrauterine infection could cause oligodendrocyte injury in the developing brain. Moreover, the increased expression of iNOS followed by the increasing NO concentration after intrauterine infection suggests that iNOS might be a key mediator between the intrauterine infection and oligodendrocyte injury in the developing brain.
Collapse
Affiliation(s)
- Ying Shen
- Department of Neonatology, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, PR China
| | | | | | | | | |
Collapse
|
27
|
Bilbo SD, Biedenkapp JC, Der-Avakian A, Watkins LR, Rudy JW, Maier SF. Neonatal infection-induced memory impairment after lipopolysaccharide in adulthood is prevented via caspase-1 inhibition. J Neurosci 2006; 25:8000-9. [PMID: 16135757 PMCID: PMC6725459 DOI: 10.1523/jneurosci.1748-05.2005] [Citation(s) in RCA: 193] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
We have reported that neonatal infection leads to memory impairment after an immune challenge in adulthood. Here we explored whether events occurring as a result of early infection alter the response to a subsequent immune challenge in adult rats, which may then impair memory. In experiment 1, peripheral infection with Escherichia coli on postnatal day 4 increased cytokines and corticosterone in the periphery, and cytokine and microglial cell marker gene expression in the hippocampus of neonate pups. Next, rats treated neonatally with E. coli or PBS were injected in adulthood with lipopolysaccharide (LPS) or saline and killed 1-24 h later. Microglial cell marker mRNA was elevated in hippocampus in saline controls infected as neonates. Furthermore, LPS induced a greater increase in glial cell marker mRNA in hippocampus of neonatally infected rats, and this increase remained elevated at 24 h versus controls. After LPS, neonatally infected rats exhibited faster increases in interleukin-1beta (IL-1beta) within the hippocampus and cortex and a prolonged response within the cortex. There were no group differences in peripheral cytokines or corticosterone. In experiment 2, rats treated neonatally with E. coli or PBS received as adults either saline or a centrally administered caspase-1 inhibitor, which specifically prevents the synthesis of IL-1beta, 1 h before a learning event and subsequent LPS challenge. Caspase-1 inhibition completely prevented LPS-induced memory impairment in neonatally infected rats. These data implicate IL-1beta in the set of immune/inflammatory events that occur in the brain as a result of neonatal infection, which likely contribute to cognitive alterations in adulthood.
Collapse
Affiliation(s)
- Staci D Bilbo
- Department of Psychology, University of Colorado, Boulder, Colorado 80309, USA.
| | | | | | | | | | | |
Collapse
|
28
|
Yuan TM, Yu HM, Gu WZ, Li JP. White matter damage and chemokine induction in developing rat brain after intrauterine infection. J Perinat Med 2005; 33:415-22. [PMID: 16238536 DOI: 10.1515/jpm.2005.074] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
In order to investigate the neuropathological effects on the developing rat brain after intrauterine infection, identification of glail fibrillary acidic protein (GFAP), 2', 3'-cyclic nucleotide phosphodiesterase (CNPase), and neurofilament (NF) was observed. Escherichia coli (E. coli) was inoculated into uterine horn of pregnant rats when gestation was 70% complete (15 days) and the control group was inoculated with normal saline. Immunohistochemistry was used for evaluation of GFAP, CNPase, and NF expression in pup brains at postnatal day 7 (P7) and reverse transcriptase-PCR (RT-PCR) to analyze macrophage inflammatory protein-1 alpha mRNA (MIP-1 alpha mRNA), macrophage inflammatory protein-1 beta mRNA (MIP-1beta mRNA), the regulated upon activation normal T expressed and secreted chemokine mRNA (RANTES mRNA) and Eotaxin mRNA expression in pup brains at P1, P3 and P7. The numbers of GFAP-positive cells of the E. coli-treated group pups were marked increased in periventricular white matter and hippocampus at P7 compared with the control group but no significant different levels of GFAP expression in corpus callosum were found between two groups. The integrate density (ID) of CNPase-positive staining of the Escherichia coli-treated group pups were marked decreased in periventricular white matter and corpus callosum at P7 compared with the control group. The ID of NF-positive staining of the Escherichia coli-treated group pups were marked decreased in periventricular white matter at P7 compared with the control group and no significant different levels of NF expression in corpus callosum were found between two groups. The expression of MIP-1 alpha mRNA and MIP-1 beta mRNA in brain of the E. coli-treated pup rat were higher than the control at P1, but the expression of MIP-1 alpha mRNA and MIP-1 beta mRNA in brain of the pup rat at P3 and P7 had no significant difference between two groups. The alteration of expression of GFAP, CNPase, and NF in the brain of neonatal rats after intrauterine infection suggested that intrauterine infection could cause neonatal white matter damage. Moreover, the transient increase in expression of chemokine such as MIP-1 alpha, MIP-1 beta in neonatal brain after intrauterine infection indicated that MIP-1 alpha, MIP-1 beta may be a mechanism mediating between the neonatal white matter damage and the intrauterine infection.
Collapse
Affiliation(s)
- Tian-Ming Yuan
- Department of Neonatology, Laboratory, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, PR China
| | | | | | | |
Collapse
|