1
|
Frasch MG, Wakefield C, Janoschek B, Frank YS, Karp F, Reyes N, Desrochers A, Wallingford MC, Antonelli MC, Metz GAS. Perinatal Psychoneuroimmunology of Prenatal Stress and Its Effects on Fetal and Postnatal Brain Development. Methods Mol Biol 2025; 2868:303-332. [PMID: 39546237 DOI: 10.1007/978-1-0716-4200-9_16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2024]
Abstract
Prenatal stress (PS) impacts early behavioral, neuroimmune, and cognitive development. Pregnant rat models have been very valuable in examining the mechanisms of such fetal programming. A pregnant sheep model of maternal stress offers the unique advantages of chronic in utero monitoring and manipulation. This chapter presents the techniques used to model single and multigenerational stress exposures and their pleiotropic effects on the offspring.
Collapse
Affiliation(s)
- Martin G Frasch
- Department of Obstetrics and Gynecology and Institute on Human Development and Disability, University of Washington, Seattle, WA, USA.
| | - Colin Wakefield
- Department of Obstetrics and Gynecology and Institute on Human Development and Disability, University of Washington, Seattle, WA, USA
| | - Ben Janoschek
- Department of Obstetrics and Gynecology and Institute on Human Development and Disability, University of Washington, Seattle, WA, USA
| | - Yael S Frank
- Department of Obstetrics and Gynecology and Institute on Human Development and Disability, University of Washington, Seattle, WA, USA
| | - Floyd Karp
- Departments of Pharmacy and Bioengineering, University of Washington, Seattle, WA, USA
| | - Nicholas Reyes
- Department of Comparative Medicine, University of Washington, Seattle, WA, USA
| | - Andre Desrochers
- Department of Clinical Sciences, Faculty of Veterinary Medicine, University of Montreal, Saint-Hyacinthe, QC, Canada
| | - Mary C Wallingford
- Mother Infant Research Institute, Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA, USA
- Department of Obstetrics and Gynecology, Tufts University School of Medicine, Boston, MA, USA
| | - Marta C Antonelli
- Department of Obstetrics and Gynecology, Klinikum Rechts Der Isar, Technical University of Munich, Munich, Germany
- Instituto de Biología Celular y Neurociencia "Prof. Eduardo De Robertis", Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Gerlinde A S Metz
- Canadian Centre for Behavioural Neuroscience, Department of Neuroscience, University of Lethbridge, Lethbridge, AB, Canada
| |
Collapse
|
2
|
Ponciano R, Hallak JEC, Crippa JA, Guimarães FS, Bel EAD. Cannabigerol Mitigates Haloperidol-Induced Vacuous Chewing Movements in Mice. Neurotox Res 2024; 43:2. [PMID: 39699828 DOI: 10.1007/s12640-024-00724-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 12/10/2024] [Accepted: 12/11/2024] [Indexed: 12/20/2024]
Abstract
Chronic use of typical antipsychotics can lead to varying motor effects depending on the timing of analysis. Acute treatment typically induces hypokinesia, resembling parkinsonism, while repeated use can result in tardive dyskinesia, a hyperkinetic syndrome marked by involuntary orofacial movements, such as vacuous chewing movements in mice. Tardive dyskinesia is particularly concerning due to its potential irreversibility and associated motor discomfort. One prevailing theory suggests that tardive dyskinesia arises from hypersensitivity of D2-type dopaminergic receptors caused by continuous blockade from typical antipsychotics like haloperidol. Additionally, increased inflammation, oxidative stress, and elevated FosB protein expression in the dorsolateral striatum are implicated in its pathophysiology. Current treatments for tardive dyskinesia often lack clear efficacy and may lead to significant side effects. Cannabigerol, a non-psychotomimetic cannabinoid with antioxidant and anti-inflammatory properties, has been investigated for its potential antidyskinetic effects. In this study, mice were treated with cannabigerol at doses of 3 and 10 mg/kg to evaluate its ability to prevent, ameliorate, or reverse haloperidol-induced vacuous chewing movements. Cannabigerol successfully reduced vacuous chewing movements without affecting normal motor activity, exacerbating haloperidol-induced hypokinesia, or inducing dyskinetic effects on its own. However, no significant reversal of the haloperidol-induced motor effects was observed under the current protocol. Furthermore, cannabigerol did not alter FosB expression or microglia morphology. These findings underscore the need for further research to explore cannabigerol's therapeutic potential and contribute to our understanding of its possible clinical applications in managing tardive dyskinesia.
Collapse
Affiliation(s)
- R Ponciano
- Department of Physiology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - J E C Hallak
- Department of Neurosciences and Behavioral Sciences, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - J A Crippa
- Department of Neurosciences and Behavioral Sciences, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - F S Guimarães
- Department of Pharmacology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Elaine Ap Del Bel
- Department of Physiology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil.
- Department of Neurosciences and Behavioral Sciences, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil.
- Department of Basic and Oral Biology, School of Dentistry of Ribeirão Preto, University of São Paulo, Av. Do Café s/n, Ribeirão Preto, São Paulo, Ribeirão Preto, SP, 14049-900, Brazil.
| |
Collapse
|
3
|
Zamudio-Flores J, Cerqueda D, Phillips-Farfán B, Guerrero-Flores S, Salinas-García AF, Meléndez-Herrera E, Sélem-Mojica N, Kline AE, Lajud N. Environmental enrichment-induced cognitive recovery after a moderate pediatric traumatic brain injury is associated with the gut microbiota and neuroinflammation. Exp Neurol 2024; 385:115109. [PMID: 39662794 DOI: 10.1016/j.expneurol.2024.115109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 11/11/2024] [Accepted: 12/04/2024] [Indexed: 12/13/2024]
Abstract
Pediatric traumatic brain injury (TBI) is a significant health concern, yet access to rehabilitation therapies for children remains limited. Environmental enrichment (EE) is a preclinical model of neurorehabilitation that promotes behavioral recovery and reduces neuroinflammation after TBI. While the gut microbiota has recently emerged as a potential therapeutic target for treating TBI sequelae in adults, its role in recovery after pediatric TBI remains unclear. Therefore, our aim was to assess the effect of EE on gut microbiota and its correlation with cognition as well as microglial morphology in a preclinical model of pediatric TBI. Male rats underwent a controlled cortical impact of moderate severity or sham injury at postnatal day 21 and were then randomly assigned to either EE or standard (STD) housing. Cognition was evaluated using the Morris water maze (MWM) on post-injury days 14-19. Microglial morphology and caecum microbiota was characterized on post-injury day 21. Cognitive deficits and increased microglial activation in the ipsilateral cortex were observed in the STD-housed TBI rats but not those in EE. TBI decreased microbiota α-diversity, while PERMANOVA analysis showed that both TBI and EE modified microbiota β-diversity. Furthermore, regression models indicated that microglial morphology in the ipsilateral cortex and Lactobacillus reuteri predicted behavioral outcomes, while Prevotellaceae NK3B31 was associated with microglial morphology. The data suggest that EE mitigates TBI-induced alterations in gut microbiota and that there is a complex interplay between EE, microbiota and microglial morphology that predicts behavioral recovery in pediatric rats.
Collapse
Affiliation(s)
- Jonathan Zamudio-Flores
- División de Neurociencias, Centro de Investigación Biomédica de Michoacán - Instituto Mexicano del Seguro Social, Morelia, Michoacán, Mexico; Instituto de Investigaciones sobre los Recursos Naturales - Universidad Michoacana de San Nicolás de Hidalgo, Morelia, Michoacán, Mexico
| | - Daniel Cerqueda
- Red de Manejo Biorracional de Plagas y Vectores, Instituto de Ecología, A. C., Xalapa, Mexico
| | | | | | - Ana Fernanda Salinas-García
- División de Neurociencias, Centro de Investigación Biomédica de Michoacán - Instituto Mexicano del Seguro Social, Morelia, Michoacán, Mexico; Instituto de Investigaciones sobre los Recursos Naturales - Universidad Michoacana de San Nicolás de Hidalgo, Morelia, Michoacán, Mexico
| | - Esperanza Meléndez-Herrera
- Instituto de Investigaciones sobre los Recursos Naturales - Universidad Michoacana de San Nicolás de Hidalgo, Morelia, Michoacán, Mexico
| | - Nelly Sélem-Mojica
- Centro de Ciencias Matemáticas, Universidad Nacional Autónoma de, Mexico
| | - Anthony E Kline
- Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, PA, United States of America; Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA, United States of America; Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, United States of America; Center for the Neural Basis of Cognition, University of Pittsburgh, Pittsburgh, PA, United States of America; Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America; Psychology, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Naima Lajud
- División de Neurociencias, Centro de Investigación Biomédica de Michoacán - Instituto Mexicano del Seguro Social, Morelia, Michoacán, Mexico.
| |
Collapse
|
4
|
Smail MA, Lenz KM. Developmental functions of microglia: Impact of psychosocial and physiological early life stress. Neuropharmacology 2024; 258:110084. [PMID: 39025401 DOI: 10.1016/j.neuropharm.2024.110084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 07/03/2024] [Accepted: 07/15/2024] [Indexed: 07/20/2024]
Abstract
Microglia play numerous important roles in brain development. From early embryonic stages through adolescence, these immune cells influence neuronal genesis and maturation, guide connectivity, and shape brain circuits. They also interact with other glial cells and structures, influencing the brain's supportive microenvironment. While this central role makes microglia essential, it means that early life perturbations to microglia can have widespread effects on brain development, potentially resulting in long-lasting behavioral impairments. Here, we will focus on the effects of early life psychosocial versus physiological stressors in rodent models. Psychosocial stress refers to perceived threats that lead to stress axes activation, including prenatal stress, or chronic postnatal stress, including maternal separation and resource scarcity. Physiological stress refers to physical threats, including maternal immune activation, postnatal infection, and traumatic brain injury. Differing sources of early life stress have varied impacts on microglia, and these effects are moderated by factors such as developmental age, brain region, and sex. Overall, these stressors appear to either 1) upregulate basal microglia numbers and activity throughout the lifespan, while possibly blunting their responsivity to subsequent stressors, or 2) shift the developmental curve of microglia, resulting in differential timing and function, impacting the critical periods they govern. Either could contribute to behavioral dysfunctions that occur after the resolution of early life stress. Exploring how different stressors impact microglia, as well as how multiple stressors interact to alter microglia's developmental functions, could deepen our understanding of how early life stress changes the brain's developmental trajectory. This article is part of the Special Issue on "Microglia".
Collapse
Affiliation(s)
- Marissa A Smail
- Department of Psychology, Ohio State University, Columbus, OH, USA.
| | - Kathryn M Lenz
- Department of Psychology, Ohio State University, Columbus, OH, USA; Department of Neuroscience, Ohio State University, Columbus, OH, USA; Institute for Behavioral Medicine Research, Ohio State University, Columbus, OH, USA; Chronic Brain Injury Program, Ohio State University, Columbus, OH, USA
| |
Collapse
|
5
|
Perez M, Barroso Spejo A, Bortolança Chiarotto G, Silveira Guimarães F, Leite Rodrigues de Oliveira A, Politti Cartarozzi L. Selective blockade of cannabinoid receptors influences motoneuron survival and glial responses after neonatal axotomy. Neuroscience 2024; 565:265-276. [PMID: 39481830 DOI: 10.1016/j.neuroscience.2024.10.051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 10/23/2024] [Accepted: 10/28/2024] [Indexed: 11/03/2024]
Abstract
Sciatic nerve crush in neonatal rats leads to an extensive death of motor and sensory neurons, serving as a platform to develop new neuroprotective approaches. The endocannabinoid system plays important neuromodulatory roles and has been involved in neurodevelopment and neuroprotection. The present work investigated the role of the cannabinoid receptors CB1 and CB2 in the neuroprotective response after neonatal axotomy. CB1 and CB2 antagonists (AM251 and AM630, respectively) were used after sciatic nerve crush in 2-day-old Wistar rats. Five days after lesion and treatment, the rats were perfused, and the spinal cords and dorsal root ganglia (DRG) were obtained and processed to investigate neuronal survival and immunohistochemistry changes, or RT-qPCR analysis. Motoneuron survival analysis showed that blocking CB2 alone or in combination with CB1 was neuroprotective. This effect was associated with a decrease in astrogliosis and microglial reaction. Interestingly, Cnr1 (CB1) and Bdnf gene transcripts were downregulated in the spinal cords of the antagonist-treated groups. Despite no intergroup difference regarding neuronal survival in the DRG, the simultaneous blockade of CB1 and CB2 receptors led to an increased expression of both Cnr1 and Cnr2, combined with Gdnf upregulation. The results indicate that the selective antagonism of cannabinoid receptors facilitates neuroprotection and decreases glial reactivity, suggesting new potential treatment approaches.
Collapse
Affiliation(s)
- Matheus Perez
- Laboratory of Nerve Regeneration, University of Campinas - UNICAMP, Cidade Universitaria "Zeferino Vaz", Rua Monteiro Lobato, 255, 13083-862, Campinas, SP, Brazil; Center for Studies in Anatomy, São Francisco University - USF, Av. São Francisco de Assis, 218 14049-900, Bragança Paulista, SP, Brazil
| | - Aline Barroso Spejo
- Laboratory of Nerve Regeneration, University of Campinas - UNICAMP, Cidade Universitaria "Zeferino Vaz", Rua Monteiro Lobato, 255, 13083-862, Campinas, SP, Brazil
| | - Gabriela Bortolança Chiarotto
- Laboratory of Nerve Regeneration, University of Campinas - UNICAMP, Cidade Universitaria "Zeferino Vaz", Rua Monteiro Lobato, 255, 13083-862, Campinas, SP, Brazil
| | - Francisco Silveira Guimarães
- Department of Pharmacology, School of Medicine of Ribeirão Preto, University of São Paulo, Av. Bandeirantes, 3900 14040-907, Ribeirão Preto, SP, Brazil
| | - Alexandre Leite Rodrigues de Oliveira
- Laboratory of Nerve Regeneration, University of Campinas - UNICAMP, Cidade Universitaria "Zeferino Vaz", Rua Monteiro Lobato, 255, 13083-862, Campinas, SP, Brazil
| | - Luciana Politti Cartarozzi
- Laboratory of Nerve Regeneration, University of Campinas - UNICAMP, Cidade Universitaria "Zeferino Vaz", Rua Monteiro Lobato, 255, 13083-862, Campinas, SP, Brazil.
| |
Collapse
|
6
|
Issy AC, Pedrazzi JF, Nascimento GC, Faccioli LH, Del Bel E. Impact of 5-Lipoxygenase Deficiency on Dopamine-Mediated Behavioral Responses. Neurotox Res 2024; 42:42. [PMID: 39365372 DOI: 10.1007/s12640-024-00720-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 08/26/2024] [Accepted: 09/24/2024] [Indexed: 10/05/2024]
Abstract
The 5-lipoxygenase/leukotriene system has been implicated in both physiological and pathological states within the central nervous system. Understanding how this system interacts with the dopaminergic system could provide valuable insights into dopamine-related pathologies. This study focused on examining both motor and non-motor dopamine-related responses in 5-lipoxygenase/leukotriene-deficient mice. We used pharmacological agents such as amphetamine, apomorphine, and reserpine to challenge the dopaminergic system, evaluating their effects on prepulse inhibition reaction (PPI), general motor activity, and oral involuntary movements. Additionally, we analyzed striatal glial marker expression (GFAP and Iba-1) in reserpine-treated mice. The 5-lipoxygenase/leukotriene-deficient mice exhibited increased spontaneous locomotor activity, including both horizontal and vertical exploration, along with stereotyped behavior compared to wild-type mice. This hyperactivity was reduced by acute apomorphine treatment. Although basal PPI responses were unchanged, 5-lipoxygenase/leukotriene-deficient mice displayed a significant reduction in susceptibility to amphetamine-induced PPI disruption. Conversely, these mice were more vulnerable to reserpine-induced involuntary movements. There were no significant differences in the basal expression of striatal GFAP and Iba-1 positive cells between 5-lipoxygenase/leukotriene-deficient and wild-type mice. However, reserpine treatment significantly increased GFAP immunoreactivity in wild-type mice, an effect not observed in 5-lipoxygenase-deficient mice. Additionally, the percentage of activated microglia was significantly higher in reserpine-treated wild-type mice, an effect absents in 5-lipoxygenase/leukotriene-deficient mice. Our findings suggest that 5-lipoxygenase/leukotriene deficiency leads to a distinctive dopaminergic phenotype, indicating that leukotrienes may influence the modulation of dopamine-mediated responses.
Collapse
Affiliation(s)
- Ana Carolina Issy
- Department of Basic and Oral Biology, Ribeirão Preto School of Dentistry, University of São Paulo (USP), Ribeirão Preto, São Paulo, Brazil
| | - João Francisco Pedrazzi
- Department of Neuroscience and Behavior Sciences, Medical School of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, São Paulo, Brazil
| | - Glauce Crivelaro Nascimento
- Department of Basic and Oral Biology, Ribeirão Preto School of Dentistry, University of São Paulo (USP), Ribeirão Preto, São Paulo, Brazil
| | - Lúcia Helena Faccioli
- Department of Clinical Analyses, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Elaine Del Bel
- Department of Basic and Oral Biology, Ribeirão Preto School of Dentistry, University of São Paulo (USP), Ribeirão Preto, São Paulo, Brazil.
| |
Collapse
|
7
|
Peralta F, Vidal Escobedo AA, Hanotte JL, Avallone M, Björklund T, Reggiani PC, Pardo J. Preventive cognitive protection based on AAV9 overexpression of IGF1 in hippocampal astrocytes. Neurobiol Dis 2024; 200:106612. [PMID: 39032798 DOI: 10.1016/j.nbd.2024.106612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 07/05/2024] [Accepted: 07/16/2024] [Indexed: 07/23/2024] Open
Abstract
Astrocytes play key roles in the brain. When astrocyte support fails, neurological disorders follow, resulting in disrupted synaptic communication, neuronal degeneration, and cell death. We posit that astrocytes overexpressing neurotrophic factors, such as Insulin Like Growth Factor 1 (IGF1), prevent the onset of neurodegeneration. We overexpressed IGF1 and the reporter TdTomato (TOM) in hippocampal astrocytes with bicistronic Adeno-Associated Virus (AAV) harboring the Glial Fibrillary Acidic Protein (GFAP) promoter and afterwards induced neurodegeneration by the intracerebroventricular (ICV) injection of streptozotocin (STZ), a rat model of behavioral impairment, neuroinflammation and shortening of hippocampal astrocytes. We achieved a thorough transgene expression along the hippocampus with a single viral injection. Although species typical behavior was impaired, memory deficit was prevented by IGF1. STZ prompted astrocyte shortening, albeit the length of these cells in animals injected with GFP and IGF1 vectors did not statistically differ from the other groups. In STZ control animals, hippocampal microglial reactive cells increased dramatically, but this was alleviated in IGF1 rats. We conclude that overexpression of IGF1 in astrocytes prevents neurodegeneration onset. Hence, individuals with early neurotrophic exhaustion would be vulnerable to age-related neurodegeneration.
Collapse
Affiliation(s)
- Facundo Peralta
- Instituto de Investigaciones Bioquímicas de La Plata "Profesor Doctor Rodolfo R. Brenner". Facultad de Ciencias Médicas. Universidad Nacional de La Plata. Buenos Aires, Argentina
| | - Ana Abril Vidal Escobedo
- Instituto de Investigaciones Bioquímicas de La Plata "Profesor Doctor Rodolfo R. Brenner". Facultad de Ciencias Médicas. Universidad Nacional de La Plata. Buenos Aires, Argentina
| | - Juliette López Hanotte
- Instituto de Investigaciones Bioquímicas de La Plata "Profesor Doctor Rodolfo R. Brenner". Facultad de Ciencias Médicas. Universidad Nacional de La Plata. Buenos Aires, Argentina
| | - Martino Avallone
- Molecular Neuromodulation, Wallenberg Neuroscience Center, Lund University, Lund, Sweden
| | - Tomas Björklund
- Molecular Neuromodulation, Wallenberg Neuroscience Center, Lund University, Lund, Sweden
| | - Paula Cecilia Reggiani
- Instituto de Investigaciones Bioquímicas de La Plata "Profesor Doctor Rodolfo R. Brenner". Facultad de Ciencias Médicas. Universidad Nacional de La Plata. Buenos Aires, Argentina
| | - Joaquín Pardo
- Instituto de Investigaciones Bioquímicas de La Plata "Profesor Doctor Rodolfo R. Brenner". Facultad de Ciencias Médicas. Universidad Nacional de La Plata. Buenos Aires, Argentina; Molecular Neuromodulation, Wallenberg Neuroscience Center, Lund University, Lund, Sweden.
| |
Collapse
|
8
|
López Hanotte J, Peralta F, Reggiani PC, Zappa Villar MF. Investigating the Impact of Intracerebroventricular Streptozotocin on Female Rats with and without Ovaries: Implications for Alzheimer's Disease. Neurochem Res 2024; 49:2785-2802. [PMID: 38985243 DOI: 10.1007/s11064-024-04204-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 06/12/2024] [Accepted: 06/25/2024] [Indexed: 07/11/2024]
Abstract
To contribute to research on female models of Alzheimer's disease (AD), our aim was to study the effect of intracerebroventricular (ICV) injection of streptozotocin (STZ) in female rats, and to evaluate a potential neuroprotective action of ovarian steroids against STZ. Female rats were either ovariectomized (OVX) or kept with ovaries (Sham) two weeks before ICV injections. Animals were injected with either vehicle (artificial cerebrospinal fluid, aCSF) or STZ (3 mg/kg) and separated into four experimental groups: Sham + aCSF, Sham + STZ, OVX + aCSF and OVX + STZ. Nineteen days post-injection, we assessed different behavioral aspects: burying, anxiety and exploration, object recognition memory, spatial memory, and depressive-like behavior. Immunohistochemistry and Immunoblot analyses were performed in the hippocampus to examine changes in AD-related proteins and neuronal and microglial populations. STZ affected burying and exploratory behavior depending on ovarian status, and impaired recognition but not spatial memory. STZ and ovariectomy increased depressive-like behavior. Interestingly, STZ did not alter the expression of β-amyloid peptide or Tau phosphorylated forms. STZ affected the neuronal population from the Dentate Gyrus, where immature neurons were more vulnerable to STZ in OVX rats. Regarding microglia, STZ increased reactive cells, and the OVX + STZ group showed an increase in the total cell number. In sum, STZ partially affected female rats, compared to what was previously reported for males. Although AD is more frequent in women, reports about the effect of ICV-STZ in female rats are scarce. Our work highlights the need to deepen into the effects of STZ in the female brain and study possible sex differences.
Collapse
Affiliation(s)
- Juliette López Hanotte
- Instituto de Investigaciones Bioquímicas de La Plata "Profesor Doctor Rodolfo R. Brenner", Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Facultad de Ciencias Médicas, Universidad Nacional de La Plata (UNLP), La Plata, Argentina
| | - Facundo Peralta
- Instituto de Investigaciones Bioquímicas de La Plata "Profesor Doctor Rodolfo R. Brenner", Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Facultad de Ciencias Médicas, Universidad Nacional de La Plata (UNLP), La Plata, Argentina
| | - Paula Cecilia Reggiani
- Instituto de Investigaciones Bioquímicas de La Plata "Profesor Doctor Rodolfo R. Brenner", Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Facultad de Ciencias Médicas, Universidad Nacional de La Plata (UNLP), La Plata, Argentina.
- Cátedra de Citología, Histología y Embriología, Facultad de Ciencias Médicas, Universidad Nacional de La Plata (UNLP), La Plata, Argentina.
| | - María Florencia Zappa Villar
- Instituto de Investigaciones Bioquímicas de La Plata "Profesor Doctor Rodolfo R. Brenner", Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Facultad de Ciencias Médicas, Universidad Nacional de La Plata (UNLP), La Plata, Argentina.
| |
Collapse
|
9
|
Noel SC, Madranges JF, Gothié JDM, Ewald J, Milnerwood AJ, Kennedy TE, Scott ME. Maternal gastrointestinal nematode infection alters hippocampal neuroimmunity, promotes synaptic plasticity, and improves resistance to direct infection in offspring. Sci Rep 2024; 14:10773. [PMID: 38730262 PMCID: PMC11087533 DOI: 10.1038/s41598-024-60865-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 04/29/2024] [Indexed: 05/12/2024] Open
Abstract
The developing brain is vulnerable to maternal bacterial and viral infections which induce strong inflammatory responses in the mother that are mimicked in the offspring brain, resulting in irreversible neurodevelopmental defects, and associated cognitive and behavioural impairments. In contrast, infection during pregnancy and lactation with the immunoregulatory murine intestinal nematode, Heligmosomoides bakeri, upregulates expression of genes associated with long-term potentiation (LTP) of synaptic networks in the brain of neonatal uninfected offspring, and enhances spatial memory in uninfected juvenile offspring. As the hippocampus is involved in spatial navigation and sensitive to immune events during development, here we assessed hippocampal gene expression, LTP, and neuroimmunity in 3-week-old uninfected offspring born to H. bakeri infected mothers. Further, as maternal immunity shapes the developing immune system, we assessed the impact of maternal H. bakeri infection on the ability of offspring to resist direct infection. In response to maternal infection, we found an enhanced propensity to induce LTP at Schaffer collateral synapses, consistent with RNA-seq data indicating accelerated development of glutamatergic synapses in uninfected offspring, relative to those from uninfected mothers. Hippocampal RNA-seq analysis of offspring of infected mothers revealed increased expression of genes associated with neurogenesis, gliogenesis, and myelination. Furthermore, maternal infection improved resistance to direct infection of H. bakeri in offspring, correlated with transfer of parasite-specific IgG1 to their serum. Hippocampal immunohistochemistry and gene expression suggest Th2/Treg biased neuroimmunity in offspring, recapitulating peripheral immunoregulation of H. bakeri infected mothers. These findings indicate maternal H. bakeri infection during pregnancy and lactation alters peripheral and neural immunity in uninfected offspring, in a manner that accelerates neural maturation to promote hippocampal LTP, and upregulates the expression of genes associated with neurogenesis, gliogenesis, and myelination.
Collapse
Affiliation(s)
- Sophia C Noel
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, 3801 University Street, Montreal, QC, H3A 2B4, Canada.
- Institute of Parasitology, McGill University (Macdonald Campus), 21,111 Lakeshore Road, Sainte-Anne de Bellevue, QC, H9X 3V9, Canada.
| | - Jeanne F Madranges
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, 3801 University Street, Montreal, QC, H3A 2B4, Canada
| | - Jean-David M Gothié
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, 3801 University Street, Montreal, QC, H3A 2B4, Canada
| | - Jessica Ewald
- Institute of Parasitology, McGill University (Macdonald Campus), 21,111 Lakeshore Road, Sainte-Anne de Bellevue, QC, H9X 3V9, Canada
| | - Austen J Milnerwood
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, 3801 University Street, Montreal, QC, H3A 2B4, Canada
| | - Timothy E Kennedy
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, 3801 University Street, Montreal, QC, H3A 2B4, Canada
| | - Marilyn E Scott
- Institute of Parasitology, McGill University (Macdonald Campus), 21,111 Lakeshore Road, Sainte-Anne de Bellevue, QC, H9X 3V9, Canada.
| |
Collapse
|
10
|
Gouveia HJCB, Manhães-de-Castro R, Costa-de-Santana BJR, Vasconcelos EEM, Silva ER, Roque A, Torner L, Guzmán-Quevedo O, Toscano AE. Creatine supplementation increases postnatal growth and strength and prevents overexpression of pro-inflammatory interleukin 6 in the hippocampus in an experimental model of cerebral palsy. Nutr Neurosci 2024; 27:425-437. [PMID: 37141266 DOI: 10.1080/1028415x.2023.2206688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
ABSTRACTObjectives: The aim of this study was thus to evaluate the effect of Cr supplementation on morphological changes and expression of pro-inflammatory cytokines in the hippocampus and on developmental parameters. Methods: Male Wistar rat pups were submitted to an experimental model of CP. Cr was administered via gavage from the 21st to the 28th postnatal day, and in water after the 28th, until the end of the experiment. Body weight (BW), food consumption (FC), muscle strength, and locomotion were evaluated. Expression of interleukin-1β (IL-1β), interleukin-6 (IL-6), and tumor necrosis factor α (TNF-α) were assessed in the hippocampus by quantitative real-time polymerase chain reaction. Iba1 immunoreactivity was assessed by immunocytochemistry in the hippocampal hilus. Results: Experimental CP caused increased density and activation of microglial cells, and overexpression of IL-6. The rats with CP also presented abnormal BW development and impairment of strength and locomotion. Cr supplementation was able to reverse the overexpression of IL-6 in the hippocampus and mitigate the impairments observed in BW, strength, and locomotion. Discussion: Future studies should evaluate other neurobiological characteristics, including changes in neural precursor cells and other cytokines, both pro- and anti-inflammatory.
Collapse
Affiliation(s)
- Henrique J C B Gouveia
- Postgraduate Program in Nutrition, Health Sciences Center, Federal University of Pernambuco, Recife, Brazil
- Studies in Nutrition and Phenotypic Plasticity Unit, Department of Nutrition, Federal University of Pernambuco, Recife, Brazil
- Tecnológico Nacional de México (TECNM) - Instituto Tecnológico Superior de Tacámbaro, Tacámbaro, Michoacán, Mexico
- Centro de Investigación Biomédica de Michoacán, Instituto Mexicano del Seguro Social, Morelia, Michoacán, Mexico
| | - Raul Manhães-de-Castro
- Studies in Nutrition and Phenotypic Plasticity Unit, Department of Nutrition, Federal University of Pernambuco, Recife, Brazil
| | - Bárbara J R Costa-de-Santana
- Studies in Nutrition and Phenotypic Plasticity Unit, Department of Nutrition, Federal University of Pernambuco, Recife, Brazil
- Postgraduate Program in Neuropsychiatry and Behavioral Sciences, Federal University of Pernambuco, Recife, Brazil
| | - Emanuel Ewerton M Vasconcelos
- Studies in Nutrition and Phenotypic Plasticity Unit, Department of Nutrition, Federal University of Pernambuco, Recife, Brazil
| | - Eliesly Roberto Silva
- Studies in Nutrition and Phenotypic Plasticity Unit, Department of Nutrition, Federal University of Pernambuco, Recife, Brazil
| | - Angélica Roque
- Centro de Investigación Biomédica de Michoacán, Instituto Mexicano del Seguro Social, Morelia, Michoacán, Mexico
| | - Luz Torner
- Centro de Investigación Biomédica de Michoacán, Instituto Mexicano del Seguro Social, Morelia, Michoacán, Mexico
| | - Omar Guzmán-Quevedo
- Tecnológico Nacional de México (TECNM) - Instituto Tecnológico Superior de Tacámbaro, Tacámbaro, Michoacán, Mexico
- Centro de Investigación Biomédica de Michoacán, Instituto Mexicano del Seguro Social, Morelia, Michoacán, Mexico
- Postgraduate Program in Neuropsychiatry and Behavioral Sciences, Federal University of Pernambuco, Recife, Brazil
| | - Ana E Toscano
- Studies in Nutrition and Phenotypic Plasticity Unit, Department of Nutrition, Federal University of Pernambuco, Recife, Brazil
- Postgraduate Program in Neuropsychiatry and Behavioral Sciences, Federal University of Pernambuco, Recife, Brazil
- Department of Nursing, CAV, Federal University of Pernambuco, Recife, Brazil
| |
Collapse
|
11
|
Christoforidou E, Moody L, Joilin G, Simoes FA, Gordon D, Talbot K, Hafezparast M. An ALS-associated mutation dysregulates microglia-derived extracellular microRNAs in a sex-specific manner. Dis Model Mech 2024; 17:dmm050638. [PMID: 38721655 PMCID: PMC11152562 DOI: 10.1242/dmm.050638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 04/29/2024] [Indexed: 05/30/2024] Open
Abstract
Evidence suggests the presence of microglial activation and microRNA (miRNA) dysregulation in amyotrophic lateral sclerosis (ALS), the most common form of adult motor neuron disease. However, few studies have investigated whether the miRNA dysregulation originates from microglia. Furthermore, TDP-43 (encoded by TARDBP), involved in miRNA biogenesis, aggregates in tissues of ∼98% of ALS cases. Thus, this study aimed to determine whether expression of the ALS-linked TDP-43M337V mutation in a transgenic mouse model dysregulates microglia-derived miRNAs. RNA sequencing identified several dysregulated miRNAs released by transgenic microglia and a differential miRNA release by lipopolysaccharide-stimulated microglia, which was more pronounced in cells from female mice. We validated the downregulation of three candidate miRNAs, namely, miR-16-5p, miR-99a-5p and miR-191-5p, by reverse transcription quantitative polymerase chain reaction (RT-qPCR) and identified their predicted targets, which primarily include genes involved in neuronal development and function. These results suggest that altered TDP-43 function leads to changes in the miRNA population released by microglia, which may in turn be a source of the miRNA dysregulation observed in the disease. This has important implications for the role of neuroinflammation in ALS pathology and could provide potential therapeutic targets.
Collapse
Affiliation(s)
- Eleni Christoforidou
- Sussex Neuroscience, School of Life Sciences, University of Sussex, Brighton, BN1 9QG, UK
| | - Libby Moody
- Sussex Neuroscience, School of Life Sciences, University of Sussex, Brighton, BN1 9QG, UK
| | - Greig Joilin
- Sussex Neuroscience, School of Life Sciences, University of Sussex, Brighton, BN1 9QG, UK
| | - Fabio A. Simoes
- Sussex Neuroscience, School of Life Sciences, University of Sussex, Brighton, BN1 9QG, UK
| | - David Gordon
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, OX3 9DU, UK
| | - Kevin Talbot
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, OX3 9DU, UK
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, OX1 3QU, UK
| | - Majid Hafezparast
- Sussex Neuroscience, School of Life Sciences, University of Sussex, Brighton, BN1 9QG, UK
| |
Collapse
|
12
|
Gumusoglu SB. The role of the placenta-brain axis in psychoneuroimmune programming. Brain Behav Immun Health 2024; 36:100735. [PMID: 38420039 PMCID: PMC10900837 DOI: 10.1016/j.bbih.2024.100735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 01/06/2024] [Accepted: 02/04/2024] [Indexed: 03/02/2024] Open
Abstract
Gestational exposures have enduring impacts on brain and neuroimmune development and function. Perturbations of pregnancy leading to placental structure/function deficits, cell stress, immune activation, and endocrine changes (metabolic, growth factors, etc.) all increase neuropsychiatric risk in offspring. The existing literature links obstetric diseases with placental involvement to offspring neuroimmune outcomes and neurodevelopmental risk. Psychoneuroimmune outcomes in offspring brain include changes to microglia, cytokine/chemokine production, cell stress, and long-term immunoreactivity. These outcomes are altered by structural, anti-angiogenic/hypoxic, inflammatory, and metabolic diseases of the placenta. This fetal programming occurs via direct placental passage or production of factors which can act directly on fetal brain substrates, or indirectly via action of circulating factors on intermediates in the placenta. Placental neuroendocrine, vascular/angiogenic, immune, and extracellular vesicular mechanisms are detailed. These mechanisms interact within various placental and pregnancy conditions. An increased understanding of the placental origins of psychoneuroimmunology will yield dividends for human health. Identifying maternal and placental biomarkers for fetal neuroimmune health may also revolutionize early diagnosis and precision psychiatry, empowering patients to make the best healthcare decisions for their families. Targeting placental mechanisms may be a valuable approach for the prevention and mitigation of intergenerational, lifelong neuropathology.
Collapse
Affiliation(s)
- Serena B. Gumusoglu
- Department of Obstetrics and Gynecology, University of Iowa Carver College of Medicine, 200 Hawkins Dr. Iowa City, IA, 52327, USA
- Department of Psychiatry, University of Iowa Carver College of Medicine, USA
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, USA
| |
Collapse
|
13
|
Reemst K, Lopizzo N, Abbink MR, Engelenburg HJ, Cattaneo A, Korosi A. Molecular underpinnings of programming by early-life stress and the protective effects of early dietary ω6/ω3 ratio, basally and in response to LPS: Integrated mRNA-miRNAs approach. Brain Behav Immun 2024; 117:283-297. [PMID: 38242369 DOI: 10.1016/j.bbi.2024.01.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 12/22/2023] [Accepted: 01/14/2024] [Indexed: 01/21/2024] Open
Abstract
Early-life stress (ELS) exposure increases the risk for mental disorders, including cognitive impairments later in life. We have previously demonstrated that an early diet with low ω6/ω3 polyunsaturated fatty acid (PUFA) ratio protects against ELS-induced cognitive impairments. Several studies have implicated the neuroimmune system in the ELS and diet mediated effects, but currently the molecular pathways via which ELS and early diet exert their long-term impact are not yet fully understood. Here we study the effects of ELS and dietary PUFA ratio on hippocampal mRNA and miRNA expression in adulthood, both under basal as well as inflammatory conditions. Male mice were exposed to chronic ELS by the limiting bedding and nesting material paradigm from postnatal day(P)2 to P9, and provided with a diet containing a standard (high (15:1.1)) or protective (low (1.1:1)) ω6 linoleic acid to ω3 alpha-linolenic acid ratio from P2 to P42. At P120, memory was assessed using the object location task. Subsequently, a single lipopolysaccharide (LPS) injection was given and 24 h later hippocampal genome-wide mRNA and microRNA (miRNA) expression was measured using microarray. Spatial learning deficits induced by ELS in mice fed the standard (high ω6/ω3) diet were reversed by the early-life protective (low ω6/ω3) diet. An integrated miRNA - mRNA analysis revealed that ELS and early diet induced miRNA driven mRNA expression changes into adulthood. Under basal conditions both ELS and the diet affected molecular pathways related to hippocampal plasticity, with the protective (low ω6/ω3 ratio) diet leading to activation of molecular pathways associated with improved hippocampal plasticity and learning and memory in mice previously exposed to ELS (e.g., CREB signaling and endocannabinoid neuronal synapse pathway). LPS induced miRNA and mRNA expression was strongly dependent on both ELS and early diet. In mice fed the standard (high ω6/ω3) diet, LPS increased miRNA expression leading to activation of inflammatory pathways. In contrast, in mice fed the protective diet, LPS reduced miRNA expression and altered target mRNA expression inhibiting inflammatory signaling pathways and pathways associated with hippocampal plasticity, which was especially apparent in mice previously exposed to ELS. This data provides molecular insights into how the protective (low ω6/ω3) diet during development could exert its long-lasting beneficial effects on hippocampal plasticity and learning and memory especially in a vulnerable population exposed to stress early in life, providing the basis for the development of intervention strategies.
Collapse
Affiliation(s)
- Kitty Reemst
- Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Science park 904, Amsterdam, 1098 XH, the Netherlands
| | - Nicola Lopizzo
- Biological Psychiatry Unit, Istituto di Recupero e Cura a Carattere Scientifico (IRCCS) Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy; Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Maralinde R Abbink
- Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Science park 904, Amsterdam, 1098 XH, the Netherlands
| | - Hendrik J Engelenburg
- Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Science park 904, Amsterdam, 1098 XH, the Netherlands
| | - Annamaria Cattaneo
- Biological Psychiatry Unit, Istituto di Recupero e Cura a Carattere Scientifico (IRCCS) Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy; Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Aniko Korosi
- Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Science park 904, Amsterdam, 1098 XH, the Netherlands.
| |
Collapse
|
14
|
Masai K, Nakayama Y, Shin K, Sugahara C, Miyazaki I, Yasuhara T, Date I, Asanuma M. Neurogenesis impairment with glial activation in the hippocampus-connected regions of intracerebroventricular streptozotocin-injected mice. Neurosci Lett 2024; 820:137598. [PMID: 38110145 DOI: 10.1016/j.neulet.2023.137598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 12/11/2023] [Accepted: 12/13/2023] [Indexed: 12/20/2023]
Abstract
Adult neurogenesis in the hippocampus and subventricular zone (SVZ) is impaired by intracerebroventricular administration of streptozotocin (icv-STZ) to rodents. Although neural cells in the several brain regions which connect with the hippocampus or SVZ is thought to be involved in the adult neurogenesis, few studies have investigated morphological alterations of glial cells in these areas. The present study revealed that icv-STZ induces reduction of neural progenitor cells and a dramatic increase in reactive astrocytes and microglia especially in the hippocampus and various hippocampus-connected brain areas. In contrast, there was no significant neuronal damage excluding demyelination of the stria medullaris. The results indicate the hippocampal neurogenesis impairment of this model might be occurred by activated glial cells in the hippocampus, or hippocampus-connected regions.
Collapse
Affiliation(s)
- Kaori Masai
- Department of Medical Neurobiology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1, Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| | - Yuta Nakayama
- Department of Medical Neurobiology, Okayama University Medical School, 2-5-1, Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| | - Kotaro Shin
- Department of Medical Neurobiology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1, Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| | - Chiaki Sugahara
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1, Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| | - Ikuko Miyazaki
- Department of Medical Neurobiology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1, Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| | - Takao Yasuhara
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1, Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| | - Isao Date
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1, Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| | - Masato Asanuma
- Department of Medical Neurobiology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1, Shikata-cho, Kita-ku, Okayama 700-8558, Japan.
| |
Collapse
|
15
|
Musillo C, Creutzberg KC, Collacchi B, Ajmone-Cat MA, De Simone R, Lepre M, Amrein I, Riva MA, Berry A, Cirulli F. Bdnf-Nrf-2 crosstalk and emotional behavior are disrupted in a sex-dependent fashion in adolescent mice exposed to maternal stress or maternal obesity. Transl Psychiatry 2023; 13:399. [PMID: 38105264 PMCID: PMC10725882 DOI: 10.1038/s41398-023-02701-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 11/28/2023] [Accepted: 12/04/2023] [Indexed: 12/19/2023] Open
Abstract
Maternal obesity has been recognized as a stressor affecting the developing fetal brain, leading to long-term negative outcomes comparable to those resulting from maternal psychological stress, although the mechanisms have not been completely elucidated. In this study, we tested the hypothesis that adverse prenatal conditions as diverse as maternal stress and maternal obesity might affect emotional regulation and stress response in the offspring through common pathways, with a main focus on oxidative stress and neuroplasticity. We contrasted and compared adolescent male and female offspring in two mouse models of maternal psychophysical stress (restraint during pregnancy - PNS) and maternal obesity (high-fat diet before and during gestation - mHFD) by combining behavioral assays, evaluation of the hypothalamic-pituitary-adrenal (HPA) axis reactivity, immunohistochemistry and gene expression analysis of selected markers of neuronal function and neuroinflammation in the hippocampus, a key region involved in stress appraisal. Prenatal administration of the antioxidant N-acetyl-cysteine (NAC) was used as a strategy to protect fetal neurodevelopment from the negative effects of PNS and mHFD. Our findings show that these two stressors produce overlapping effects, reducing brain anti-oxidant defenses (Nrf-2) and leading to sex-dependent impairments of hippocampal Bdnf expression and alterations of the emotional behavior and HPA axis functionality. Prenatal NAC administration, by restoring the redox balance, was able to exert long-term protective effects on brain development, suggesting that the modulation of redox pathways might be an effective strategy to target common shared mechanisms between different adverse prenatal conditions.
Collapse
Affiliation(s)
- Chiara Musillo
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
- Ph.D. Program in Behavioral Neuroscience, Department of Psychology, Sapienza University of Rome, 00185, Rome, Italy
| | - Kerstin C Creutzberg
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Barbara Collacchi
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Maria Antonietta Ajmone-Cat
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Roberta De Simone
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Marcello Lepre
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Irmgard Amrein
- Functional Neuroanatomy, Institute of Anatomy, University of Zürich, Zurich, Switzerland
| | - Marco A Riva
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
- Biological Psychiatry Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Alessandra Berry
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy.
| | - Francesca Cirulli
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy.
| |
Collapse
|
16
|
Hisada C, Kajimoto K, Tsugane H, Mitsuo I, Azuma K, Kubo KY. Maternal chewing alleviates prenatal stress-related neuroinflammation mediated by microglia in the hippocampus of the mouse offspring. J Prosthodont Res 2023; 67:588-594. [PMID: 36792221 DOI: 10.2186/jpr.jpr_d_22_00255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
PURPOSE Prenatal stress affects the hippocampal structure and function in pups. Maternal chewing ameliorates hippocampus-dependent cognitive impairments induced by prenatal stress. In this study, we investigated hippocampal microglia-mediated neuroinflammation in pups of dams exposed to prenatal stress with or without chewing during gestation. METHODS Pregnant mice were randomly assigned to control, stress, and stress/chewing groups. Stress and stress/chewing animals were subjected to restraint stress for 45 min three times daily from gestation day 12 to parturition, and were given a wooden stick to chew during the stress period. Four-month-old male pups were intraperitoneally administered with lipopolysaccharide (LPS). Serum corticosterone levels were determined 24 h after administration. The expression levels of hippocampal inflammatory cytokines were measured, and the microglia were analyzed morphologically. RESULTS Prenatal stress increased serum corticosterone levels, induced hippocampal microglia priming, and facilitated the release of interleukin-1β and tumor necrosis factor-α in the offspring. LPS treatment significantly increased the effects of prenatal stress on serum corticosterone levels, hippocampal microglial activation, and hippocampal neuroinflammation. Maternal chewing significantly inhibited the increase in serum corticosterone levels, suppressed microglial overactivation, and normalized inflammatory cytokine levels under basal prenatal stress conditions as well as after LPS administration. CONCLUSIONS Our findings indicate that maternal chewing can alleviate the increase in corticosterone levels and inhibit hippocampal microglia-mediated neuroinflammation induced by LPS administration and prenatal stress in adult offspring.
Collapse
Affiliation(s)
- Chie Hisada
- Departments of Pediatric Dentistry, Asahi University School of Dentistry, Gifu, Japan
| | - Kyoko Kajimoto
- Departments of Pediatric Dentistry, Asahi University School of Dentistry, Gifu, Japan
| | - Hiroko Tsugane
- Departments of Pediatric Dentistry, Asahi University School of Dentistry, Gifu, Japan
| | - Iinuma Mitsuo
- Departments of Pediatric Dentistry, Asahi University School of Dentistry, Gifu, Japan
| | - Kagaku Azuma
- Department of Anatomy, School of Medicine, University of Occupational and Environmental Health, Kitakyusyu, Japan
| | - Kin-Ya Kubo
- Graduate School of Human Life Science, Nagoya Women's University, Aichi, Japan
| |
Collapse
|
17
|
Juacy Rodrigues Costa-de-Santana B, Manhães-de-Castro R, José Cavalcanti Bezerra Gouveia H, Roberto Silva E, Antônio da Silva Araújo M, Cabral Lacerda D, Guzmán-Quevedo O, Torner L, Elisa Toscano A. Motor deficits are associated with increased glial cell activation in the hypothalamus and cerebellum of young rats subjected to cerebral palsy. Brain Res 2023; 1814:148447. [PMID: 37301423 DOI: 10.1016/j.brainres.2023.148447] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 05/27/2023] [Accepted: 06/04/2023] [Indexed: 06/12/2023]
Abstract
Cerebral palsy (CP) is a syndrome characterized by a wide range of sensory and motor damage, associated with behavioral and cognitive deficits. The aim of the present study was to investigate the potential of a model of CP using a combination of perinatal anoxia and sensorimotor restriction of hind paws to replicate motor, behavioral and neural deficits. A total of 30 of male Wistar rats were divided into Control (C, n = 15), and CP (CP, n = 15) groups. The potential of the CP model was assessed by evaluating food intake, the behavioral satiety sequence, performance on the CatWalk and parallel bars, muscle strength, and locomotor activity. The weight of the encephalon, soleus, and extensor digitorum longus (EDL) muscles, and the activation of glial cells (microglia and astrocytes) were also measured. The CP animals showed delayed satiety, impaired locomotion on the CatWalk and open field test, reduced muscle strength, and reduced motor coordination. CP also reduced the weight of the soleus and muscles, brain weight, liver weight, and quantity of fat in various parts of the body. There was also found to be an increase in astrocyte and microglia activation in the cerebellum and hypothalamus (arcuate nucleus, ARC) of animals subjected to CP.
Collapse
Affiliation(s)
- Bárbara Juacy Rodrigues Costa-de-Santana
- Graduate Program in Neuropsychiatry and Behavioral Sciences, Center for Medical Sciences, Federal University of Pernambuco, Recife-Pernambuco, 50670-901, Brazil; Studies in Nutrition and Phenotypic Plasticity Unit, Center for Health Sciences, Federal University of Pernambuco, Recife-Pernambuco, 50670-420, Brazil; Laboratory of Experimental Neuronutriton and Food Engineering, Tecnológico Nacional de México (TECNM)/Instituto Tecnológico Superior de Tacámbaro, Tacámbaro, Michoacán, Mexico
| | - Raul Manhães-de-Castro
- Graduate Program in Neuropsychiatry and Behavioral Sciences, Center for Medical Sciences, Federal University of Pernambuco, Recife-Pernambuco, 50670-901, Brazil; Studies in Nutrition and Phenotypic Plasticity Unit, Center for Health Sciences, Federal University of Pernambuco, Recife-Pernambuco, 50670-420, Brazil; Graduate Program in Nutrition, Center for Health Sciences, Federal University of Pernambuco, Recife-Pernambuco, 50670-420, Brazil
| | - Henrique José Cavalcanti Bezerra Gouveia
- Studies in Nutrition and Phenotypic Plasticity Unit, Center for Health Sciences, Federal University of Pernambuco, Recife-Pernambuco, 50670-420, Brazil; Graduate Program in Nutrition, Center for Health Sciences, Federal University of Pernambuco, Recife-Pernambuco, 50670-420, Brazil
| | - Eliesly Roberto Silva
- Studies in Nutrition and Phenotypic Plasticity Unit, Center for Health Sciences, Federal University of Pernambuco, Recife-Pernambuco, 50670-420, Brazil
| | - Marcos Antônio da Silva Araújo
- Studies in Nutrition and Phenotypic Plasticity Unit, Center for Health Sciences, Federal University of Pernambuco, Recife-Pernambuco, 50670-420, Brazil
| | - Diego Cabral Lacerda
- Studies in Nutrition and Phenotypic Plasticity Unit, Center for Health Sciences, Federal University of Pernambuco, Recife-Pernambuco, 50670-420, Brazil
| | - Omar Guzmán-Quevedo
- Graduate Program in Neuropsychiatry and Behavioral Sciences, Center for Medical Sciences, Federal University of Pernambuco, Recife-Pernambuco, 50670-901, Brazil; Laboratory of Experimental Neuronutriton and Food Engineering, Tecnológico Nacional de México (TECNM)/Instituto Tecnológico Superior de Tacámbaro, Tacámbaro, Michoacán, Mexico; Centro de Investigación Biomédica de Michoacán, Instituto Mexicano del Seguro Social, Morelia, Michoacán, Mexico
| | - Luz Torner
- Centro de Investigación Biomédica de Michoacán, Instituto Mexicano del Seguro Social, Morelia, Michoacán, Mexico
| | - Ana Elisa Toscano
- Graduate Program in Neuropsychiatry and Behavioral Sciences, Center for Medical Sciences, Federal University of Pernambuco, Recife-Pernambuco, 50670-901, Brazil; Studies in Nutrition and Phenotypic Plasticity Unit, Center for Health Sciences, Federal University of Pernambuco, Recife-Pernambuco, 50670-420, Brazil; Graduate Program in Nutrition, Center for Health Sciences, Federal University of Pernambuco, Recife-Pernambuco, 50670-420, Brazil; Nursing Unit, Vitória Academic Center, Federal University of Pernambuco, Vitória de Santo Antão-Pernambuco, 55608-680, Brazil.
| |
Collapse
|
18
|
Tomiyama ALMR, Cartarozzi LP, de Oliveira Coser L, Chiarotto GB, Oliveira ALR. Neuroprotection by upregulation of the major histocompatibility complex class I (MHC I) in SOD1 G93A mice. Front Cell Neurosci 2023; 17:1211486. [PMID: 37711512 PMCID: PMC10498468 DOI: 10.3389/fncel.2023.1211486] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 08/07/2023] [Indexed: 09/16/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease that progressively affects motoneurons, causing muscle atrophy and evolving to death. Astrocytes inhibit the expression of MHC-I by neurons, contributing to a degenerative outcome. The present study verified the influence of interferon β (IFN β) treatment, a proinflammatory cytokine that upregulates MHC-I expression, in SOD1G93A transgenic mice. For that, 17 days old presymptomatic female mice were subjected to subcutaneous application of IFN β (250, 1,000, and 10,000 IU) every other day for 20 days. Rotarod motor test, clinical score, and body weight assessment were conducted every third day throughout the treatment period. No significant intergroup variations were observed in such parameters during the pre-symptomatic phase. All mice were then euthanized, and the spinal cords collected for comparative analysis of motoneuron survival, reactive gliosis, synapse coverage, microglia morphology classification, cytokine analysis by flow cytometry, and RT-qPCR quantification of gene transcripts. Additionally, mice underwent Rotarod motor assessment, weight monitoring, and neurological scoring. The results show that IFN β treatment led to an increase in the expression of MHC-I, which, even at the lowest dose (250 IU), resulted in a significant increase in neuronal survival in the ALS presymptomatic period which lasted until the onset of the disease. The treatment also influenced synaptic preservation by decreasing excitatory inputs and upregulating the expression of AMPA receptors by astrocytes. Microglial reactivity quantified by the integrated density of pixels did not decrease with treatment but showed a less activated morphology, coupled with polarization to an M1 profile. Disease progression upregulated gene transcripts for pro- and anti-inflammatory cytokines, and IFN β treatment significantly decreased mRNA expression for IL4. Overall, the present results demonstrate that a low dosage of IFN β shows therapeutic potential by increasing MHC-I expression, resulting in neuroprotection and immunomodulation.
Collapse
Affiliation(s)
| | | | | | | | - Alexandre L. R. Oliveira
- Department of Structural and Functional Biology, Institute of Biology—University of Campinas (UNICAMP), Campinas, Brazil
| |
Collapse
|
19
|
Reddaway J, Richardson PE, Bevan RJ, Stoneman J, Palombo M. Microglial morphometric analysis: so many options, so little consistency. Front Neuroinform 2023; 17:1211188. [PMID: 37637472 PMCID: PMC10448193 DOI: 10.3389/fninf.2023.1211188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 07/05/2023] [Indexed: 08/29/2023] Open
Abstract
Quantification of microglial activation through morphometric analysis has long been a staple of the neuroimmunologist's toolkit. Microglial morphological phenomics can be conducted through either manual classification or constructing a digital skeleton and extracting morphometric data from it. Multiple open-access and paid software packages are available to generate these skeletons via semi-automated and/or fully automated methods with varying degrees of accuracy. Despite advancements in methods to generate morphometrics (quantitative measures of cellular morphology), there has been limited development of tools to analyze the datasets they generate, in particular those containing parameters from tens of thousands of cells analyzed by fully automated pipelines. In this review, we compare and critique the approaches using cluster analysis and machine learning driven predictive algorithms that have been developed to tackle these large datasets, and propose improvements for these methods. In particular, we highlight the need for a commitment to open science from groups developing these classifiers. Furthermore, we call attention to a need for communication between those with a strong software engineering/computer science background and neuroimmunologists to produce effective analytical tools with simplified operability if we are to see their wide-spread adoption by the glia biology community.
Collapse
Affiliation(s)
- Jack Reddaway
- Division of Neuroscience, School of Biosciences, Cardiff University, Cardiff, United Kingdom
- Hodge Centre for Neuropsychiatric Immunology, Neuroscience and Mental Health Innovation Institute (NMHII), Cardiff University, Cardiff, United Kingdom
| | | | - Ryan J. Bevan
- UK Dementia Research Institute, Cardiff University, Cardiff, United Kingdom
| | - Jessica Stoneman
- Division of Neuroscience, School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | - Marco Palombo
- Cardiff University Brain Research Imaging Centre (CUBRIC), School of Psychology, Cardiff University, Cardiff, United Kingdom
- School of Computer Science and Informatics, Cardiff University, Cardiff, United Kingdom
| |
Collapse
|
20
|
Mawson ER, Morris BJ. A consideration of the increased risk of schizophrenia due to prenatal maternal stress, and the possible role of microglia. Prog Neuropsychopharmacol Biol Psychiatry 2023; 125:110773. [PMID: 37116354 DOI: 10.1016/j.pnpbp.2023.110773] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 04/07/2023] [Accepted: 04/18/2023] [Indexed: 04/30/2023]
Abstract
Schizophrenia is caused by interaction of a combination of genetic and environmental factors. Of the latter, prenatal exposure to maternal stress is reportedly associated with elevated disease risk. The main orchestrators of inflammatory processes within the brain are microglia, and aberrant microglial activation/function has been proposed to contribute to the aetiology of schizophrenia. Here, we evaluate the epidemiological and preclinical evidence connecting prenatal stress to schizophrenia risk, and consider the possible mediating role of microglia in the prenatal stress-schizophrenia relationship. Epidemiological findings are rather consistent in supporting the association, albeit they are mitigated by effects of sex and gestational timing, while the evidence for microglial activation is more variable. Rodent models of prenatal stress generally report lasting effects on offspring neurobiology. However, many uncertainties remain as to the mechanisms underlying the influence of maternal stress on the developing foetal brain. Future studies should aim to characterise the exact processes mediating this aspect of schizophrenia risk, as well as focussing on how prenatal stress may interact with other risk factors.
Collapse
Affiliation(s)
- Eleanor R Mawson
- School of Psychology and Neuroscience, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK
| | - Brian J Morris
- School of Psychology and Neuroscience, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK.
| |
Collapse
|
21
|
Costello A, Linning-Duffy K, Vandenbrook C, Lonstein JS, Yan L. Daytime Light Deficiency Leads to Sex- and Brain Region-Specific Neuroinflammatory Responses in a Diurnal Rodent. Cell Mol Neurobiol 2023; 43:1369-1384. [PMID: 35864429 PMCID: PMC10635710 DOI: 10.1007/s10571-022-01256-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 07/07/2022] [Indexed: 11/30/2022]
Abstract
Seasonal changes in peripheral inflammation are well documented in both humans and animal models, but seasonal changes in neuroinflammation, especially the impact of seasonal lighting environment on neuroinflammation remain unclear. To address this question, the present study examined the effects of environmental lighting conditions on neuroinflammation in a diurnal rodent model, Nile grass rats (Arvicanthis niloticus). Male and female grass rats were housed in either bright (brLD) or dim (dimLD) light during the day to simulate a summer or winter light condition, respectively. After 4 weeks, microglia markers Iba-1 and CD11b, as well as pro-inflammatory cytokines TNF-α and IL-6, were examined in the anterior cingulate cortex (ACC), basolateral amygdala (BLA), and dorsal hippocampus (dHipp). The results revealed that winter-like dim light during the day leads to indicators of increased neuroinflammation in a brain site- and sex-specific manner. Specifically, relatively few changes in the neuroinflammatory markers were observed in the ACC, while numerous changes were found in the BLA and dHipp. In the BLA, winter-like dimLD resulted in hyper-ramified microglia morphology and increased expression of the pro-inflammatory cytokine IL-6, but only in males. In the dHipp, dimLD led to a higher number and hyper-ramified morphology of microglia as well as increased expression of CD11b and TNF-α, but only in females. Neuroinflammatory state is thus influenced by environmental light, differently in males and females, and could play a role in sex differences in the prevalence and symptoms of psychiatric or neurological disorders that are influenced by season or other environmental light conditions. Diurnal Nile grass rats were housed under bright or dim light during the day for 4 weeks, simulating seasonal fluctuations in daytime lighting environment. Dim light housing resulted in hyper-ramified morphology of microglia (scale bar, 15 μm) and altered expression of pro-inflammatory cytokines (TNF-α) in a sex- and brain region-specific manner.
Collapse
Affiliation(s)
- Allison Costello
- Behavioral Neuroscience Program, Department of Psychology, Michigan State University, 766, Service Road, East Lansing, MI, 48824, USA
| | - Katrina Linning-Duffy
- Behavioral Neuroscience Program, Department of Psychology, Michigan State University, 766, Service Road, East Lansing, MI, 48824, USA
| | - Carleigh Vandenbrook
- Behavioral Neuroscience Program, Department of Psychology, Michigan State University, 766, Service Road, East Lansing, MI, 48824, USA
| | - Joseph S Lonstein
- Behavioral Neuroscience Program, Department of Psychology, Michigan State University, 766, Service Road, East Lansing, MI, 48824, USA
- Neuroscience Program, Michigan State University, East Lansing, MI, 48824, USA
| | - Lily Yan
- Behavioral Neuroscience Program, Department of Psychology, Michigan State University, 766, Service Road, East Lansing, MI, 48824, USA.
- Neuroscience Program, Michigan State University, East Lansing, MI, 48824, USA.
| |
Collapse
|
22
|
Romero-Juárez PA, Visco DB, Manhães-de-Castro R, Urquiza-Martínez MV, Saavedra LM, González-Vargas MC, Mercado-Camargo R, Aquino JDS, Toscano AE, Torner L, Guzmán-Quevedo O. Dietary flavonoid kaempferol reduces obesity-associated hypothalamic microglia activation and promotes body weight loss in mice with obesity. Nutr Neurosci 2023; 26:25-39. [PMID: 34905445 DOI: 10.1080/1028415x.2021.2012629] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
BACKGROUND Obesity results from an unbalance in the ingested and burned calories. Energy balance (EB) is critically regulated by the hypothalamic arcuate nucleus (ARC) by promoting appetite or anorectic actions. Hypothalamic inflammation, driven by high activation of the microglia, has been reported as a key mechanism involved in the development of diet-induced obesity. Kaempferol (KF), a flavonoid-type polyphenol present in a large number of fruits and vegetables, was shown to regulate both energy metabolism and inflammation. OBJECTIVES In this work, we studied the effects of both the central and peripheral treatment with KF on hypothalamic inflammation and EB regulation in mice with obesity. METHODS Obese adult mice were chronically (40 days) treated with KF (0.5 mg/kg/day, intraperitoneally). During the treatment, body weight, food intake (FI), feed efficiency (FE), glucose tolerance, and insulin sensitivity were determined. Analysis of microglia activation in the ARC of the hypothalamus at the end of the treatment was also performed. Body weight, FI, and FE changes were also evaluated in response to 5µg KF, centrally administrated. RESULTS Chronic administration of KF decreased ∼43% of the density, and ∼30% of the ratio, of activated microglia in the arcuate nucleus. These changes were accompanied by body weight loss, decreased FE, reduced fasting blood glucose, and a tendency to improve insulin sensitivity. Finally, acute central administration of KF reproduced the effects on EB triggered by peripheral administration. CONCLUSION These findings suggest that KF might fight obesity by regulating central processes related to EB regulation and hypothalamic inflammation.
Collapse
Affiliation(s)
- Pedro A Romero-Juárez
- Instituto Tecnológico Superior de Tacámbaro, Michoacán, México.,Facultad de Químico-Farmacobiología, Universidad Michoacana de San Nicolás de Hidalgo, Morelia, México.,Centro de Investigación Biomédica de Michoacán, Instituto Mexicano del Seguro Social, Morelia, México
| | - Diego Bulcão Visco
- Instituto Tecnológico Superior de Tacámbaro, Michoacán, México.,Centro de Investigación Biomédica de Michoacán, Instituto Mexicano del Seguro Social, Morelia, México.,Departamento de Nutrição, Universidade Federal de Pernambuco, Recife, Brasil.,Unidade de Estudos em Nutrição e Plasticidade Fenotípica do Departamento de Nutrição, Universidade Federal de Pernambuco, Recife, Brazil
| | - Raul Manhães-de-Castro
- Departamento de Nutrição, Universidade Federal de Pernambuco, Recife, Brasil.,Unidade de Estudos em Nutrição e Plasticidade Fenotípica do Departamento de Nutrição, Universidade Federal de Pernambuco, Recife, Brazil
| | - Mercedes V Urquiza-Martínez
- Facultad de Químico-Farmacobiología, Universidad Michoacana de San Nicolás de Hidalgo, Morelia, México.,Centro de Investigación Biomédica de Michoacán, Instituto Mexicano del Seguro Social, Morelia, México
| | - Luis Miguel Saavedra
- Centro de Investigación Biomédica de Michoacán, Instituto Mexicano del Seguro Social, Morelia, México
| | - Mari C González-Vargas
- Instituto Tecnológico Superior de Tacámbaro, Michoacán, México.,Centro de Investigación Biomédica de Michoacán, Instituto Mexicano del Seguro Social, Morelia, México
| | - Rosalio Mercado-Camargo
- Facultad de Químico-Farmacobiología, Universidad Michoacana de San Nicolás de Hidalgo, Morelia, México
| | - Jailane de Souza Aquino
- Laboratório de Nutrição Experimental, Departamento de Nutrição, Universidade Federal da Paraíba, João Pessoa, Brazil
| | - Ana E Toscano
- Unidade de Estudos em Nutrição e Plasticidade Fenotípica do Departamento de Nutrição, Universidade Federal de Pernambuco, Recife, Brazil.,Departmento de Enfermagem, Universidade Federal de Pernambuco, Recife, Brasil.,Pós-Graduação em Neuropsiquiatria e Ciências do Comportamento, Universidade Federal de Pernambuco, Recife, Brasil
| | - Luz Torner
- Centro de Investigación Biomédica de Michoacán, Instituto Mexicano del Seguro Social, Morelia, México
| | - Omar Guzmán-Quevedo
- Instituto Tecnológico Superior de Tacámbaro, Michoacán, México.,Centro de Investigación Biomédica de Michoacán, Instituto Mexicano del Seguro Social, Morelia, México.,Pós-Graduação em Neuropsiquiatria e Ciências do Comportamento, Universidade Federal de Pernambuco, Recife, Brasil
| |
Collapse
|
23
|
Hall MB, Willis DE, Rodriguez EL, Schwarz JM. Maternal immune activation as an epidemiological risk factor for neurodevelopmental disorders: Considerations of timing, severity, individual differences, and sex in human and rodent studies. Front Neurosci 2023; 17:1135559. [PMID: 37123361 PMCID: PMC10133487 DOI: 10.3389/fnins.2023.1135559] [Citation(s) in RCA: 34] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Accepted: 03/13/2023] [Indexed: 05/02/2023] Open
Abstract
Epidemiological evidence suggests that one's risk of being diagnosed with a neurodevelopmental disorder (NDD)-such as autism, ADHD, or schizophrenia-increases significantly if their mother had a viral or bacterial infection during the first or second trimester of pregnancy. Despite this well-known data, little is known about how developing neural systems are perturbed by events such as early-life immune activation. One theory is that the maternal immune response disrupts neural processes important for typical fetal and postnatal development, which can subsequently result in specific and overlapping behavioral phenotypes in offspring, characteristic of NDDs. As such, rodent models of maternal immune activation (MIA) have been useful in elucidating neural mechanisms that may become dysregulated by MIA. This review will start with an up-to-date and in-depth, critical summary of epidemiological data in humans, examining the association between different types of MIA and NDD outcomes in offspring. Thereafter, we will summarize common rodent models of MIA and discuss their relevance to the human epidemiological data. Finally, we will highlight other factors that may interact with or impact MIA and its associated risk for NDDs, and emphasize the importance for researchers to consider these when designing future human and rodent studies. These points to consider include: the sex of the offspring, the developmental timing of the immune challenge, and other factors that may contribute to individual variability in neural and behavioral responses to MIA, such as genetics, parental age, the gut microbiome, prenatal stress, and placental buffering.
Collapse
|
24
|
Reemst K, Kracht L, Kotah JM, Rahimian R, van Irsen AAS, Congrains Sotomayor G, Verboon LN, Brouwer N, Simard S, Turecki G, Mechawar N, Kooistra SM, Eggen BJL, Korosi A. Early-life stress lastingly impacts microglial transcriptome and function under basal and immune-challenged conditions. Transl Psychiatry 2022; 12:507. [PMID: 36481769 PMCID: PMC9731997 DOI: 10.1038/s41398-022-02265-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 11/18/2022] [Accepted: 11/23/2022] [Indexed: 12/13/2022] Open
Abstract
Early-life stress (ELS) leads to increased vulnerability to psychiatric disorders including depression later in life. Neuroinflammatory processes have been implicated in ELS-induced negative health outcomes, but how ELS impacts microglia, the main tissue-resident macrophages of the central nervous system, is unknown. Here, we determined the effects of ELS-induced by limited bedding and nesting material during the first week of life (postnatal days [P]2-9) on microglial (i) morphology; (ii) hippocampal gene expression; and (iii) synaptosome phagocytic capacity in male pups (P9) and adult (P200) mice. The hippocampus of ELS-exposed adult mice displayed altered proportions of morphological subtypes of microglia, as well as microglial transcriptomic changes related to the tumor necrosis factor response and protein ubiquitination. ELS exposure leads to distinct gene expression profiles during microglial development from P9 to P200 and in response to an LPS challenge at P200. Functionally, synaptosomes from ELS-exposed mice were phagocytosed less by age-matched microglia. At P200, but not P9, ELS microglia showed reduced synaptosome phagocytic capacity when compared to control microglia. Lastly, we confirmed the ELS-induced increased expression of the phagocytosis-related gene GAS6 that we observed in mice, in the dentate gyrus of individuals with a history of child abuse using in situ hybridization. These findings reveal persistent effects of ELS on microglial function and suggest that altered microglial phagocytic capacity is a key contributor to ELS-induced phenotypes.
Collapse
Affiliation(s)
- Kitty Reemst
- grid.7177.60000000084992262Swammerdam Institute for Life Sciences, Center for Neuroscience, Brain Plasticity Group, University of Amsterdam, Amsterdam, Science Park 904, 1098 XH The Netherlands
| | - Laura Kracht
- grid.4494.d0000 0000 9558 4598Department of Biomedical Sciences of Cells & Systems, Section Molecular Neurobiology, University of Groningen, University Medical Center Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Janssen M. Kotah
- grid.7177.60000000084992262Swammerdam Institute for Life Sciences, Center for Neuroscience, Brain Plasticity Group, University of Amsterdam, Amsterdam, Science Park 904, 1098 XH The Netherlands
| | - Reza Rahimian
- grid.412078.80000 0001 2353 5268McGill Group for Suicide Studies, Douglas Hospital Research Center, Montreal, QC H4H 1R3 Canada ,grid.14709.3b0000 0004 1936 8649Department of Psychiatry, McGill University, Montreal, QC H3A 1A1 Canada
| | - Astrid A. S. van Irsen
- grid.7177.60000000084992262Swammerdam Institute for Life Sciences, Center for Neuroscience, Brain Plasticity Group, University of Amsterdam, Amsterdam, Science Park 904, 1098 XH The Netherlands
| | - Gonzalo Congrains Sotomayor
- grid.7177.60000000084992262Swammerdam Institute for Life Sciences, Center for Neuroscience, Brain Plasticity Group, University of Amsterdam, Amsterdam, Science Park 904, 1098 XH The Netherlands
| | - Laura N. Verboon
- grid.7177.60000000084992262Swammerdam Institute for Life Sciences, Center for Neuroscience, Brain Plasticity Group, University of Amsterdam, Amsterdam, Science Park 904, 1098 XH The Netherlands
| | - Nieske Brouwer
- grid.4494.d0000 0000 9558 4598Department of Biomedical Sciences of Cells & Systems, Section Molecular Neurobiology, University of Groningen, University Medical Center Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Sophie Simard
- grid.412078.80000 0001 2353 5268McGill Group for Suicide Studies, Douglas Hospital Research Center, Montreal, QC H4H 1R3 Canada ,grid.14709.3b0000 0004 1936 8649Department of Psychiatry, McGill University, Montreal, QC H3A 1A1 Canada
| | - Gustavo Turecki
- grid.412078.80000 0001 2353 5268McGill Group for Suicide Studies, Douglas Hospital Research Center, Montreal, QC H4H 1R3 Canada ,grid.14709.3b0000 0004 1936 8649Department of Psychiatry, McGill University, Montreal, QC H3A 1A1 Canada
| | - Naguib Mechawar
- grid.412078.80000 0001 2353 5268McGill Group for Suicide Studies, Douglas Hospital Research Center, Montreal, QC H4H 1R3 Canada ,grid.14709.3b0000 0004 1936 8649Department of Psychiatry, McGill University, Montreal, QC H3A 1A1 Canada
| | - Susanne M. Kooistra
- grid.4494.d0000 0000 9558 4598Department of Biomedical Sciences of Cells & Systems, Section Molecular Neurobiology, University of Groningen, University Medical Center Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Bart J. L. Eggen
- grid.4494.d0000 0000 9558 4598Department of Biomedical Sciences of Cells & Systems, Section Molecular Neurobiology, University of Groningen, University Medical Center Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Aniko Korosi
- Swammerdam Institute for Life Sciences, Center for Neuroscience, Brain Plasticity Group, University of Amsterdam, Amsterdam, Science Park 904, 1098 XH, The Netherlands.
| |
Collapse
|
25
|
Majerczyk D, Ayad E, Brewton K, Saing P, Hart P. Systemic maternal inflammation promotes ASD via IL-6 and IFN-γ. Biosci Rep 2022; 42:BSR20220713. [PMID: 36300375 PMCID: PMC9670245 DOI: 10.1042/bsr20220713] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 09/30/2022] [Accepted: 10/26/2022] [Indexed: 07/25/2023] Open
Abstract
Autism spectrum disorder (ASD) is a neurological disorder that manifests during early development, impacting individuals through their ways of communicating, social behaviors, and their ability to perform day-to-day activities. There have been different proposed mechanisms on how ASD precipitates within a patient, one of which being the impact cytokines have on fetal development once a mother's immune system has been activated (referred to as maternal immune activation, MIA). The occurrence of ASD has long been associated with elevated levels of several cytokines, including interleukin-6 (IL-6) and interferon gamma (IFN-γ). These proinflammatory cytokines can achieve high systemic levels in response to immune activating pathogens from various extrinsic sources. Transfer of cytokines such as IL-6 across the placental barrier allows accumulation in the fetus, potentially inducing neuroinflammation and consequently altering neurodevelopmental processes. Individuals who have been later diagnosed with ASD have been observed to have elevated levels of IL-6 and other proinflammatory cytokines during gestation. Moreover, the outcome of MIA has been associated with neurological effects such as impaired social interaction and an increase in repetitive behavior in animal models, supporting a mechanistic link between gestational inflammation and development of ASD-like characteristics. The present review attempts to provide a concise overview of the available preclinical and clinical data that suggest cross-talk between IL-6 and IFN-γ through both extrinsic and intrinsic factors as a central mechanism of MIA that may promote the development of ASD.
Collapse
Affiliation(s)
- Daniel Majerczyk
- College of Science, Health and Pharmacy, Roosevelt University, Illinois 60173, U.S.A
- Loyola Medicine, Berwyn, Illinois 60402, U.S.A
| | - Elizabeth G. Ayad
- College of Science, Health and Pharmacy, Roosevelt University, Illinois 60173, U.S.A
| | - Kari L. Brewton
- College of Science, Health and Pharmacy, Roosevelt University, Illinois 60173, U.S.A
| | - Pichrasmei Saing
- College of Science, Health and Pharmacy, Roosevelt University, Illinois 60173, U.S.A
| | - Peter C. Hart
- College of Science, Health and Pharmacy, Roosevelt University, Illinois 60173, U.S.A
| |
Collapse
|
26
|
The Time Course of MHC-I Expression in C57BL/6J and A/J Mice Correlates with the Degree of Retrograde Gliosis in the Spinal Cord following Sciatic Nerve Crush. Cells 2022; 11:cells11233710. [PMID: 36496969 PMCID: PMC9740909 DOI: 10.3390/cells11233710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 11/02/2022] [Accepted: 11/03/2022] [Indexed: 11/23/2022] Open
Abstract
The pleiotropic role of the major histocompatibility complex class I (MHC-I) reflects the close association between the nervous and immune systems. In turn, MHC-I upregulation postinjury is associated with a better regenerative outcome in isogenic mice following peripheral nerve damage. In the present work, we compared the time course of neuronal, glial, and sensorimotor recovery (1, 3, 5, 7, and 28 days after lesion—dal) following unilateral sciatic nerve crush in A/J and C57BL/6J mice. The A/J strain showed higher expression of MHC-I (7 dal, ** p < 0.01), Iba-1 (microglial reaction, 7 dal, *** p < 0.001), and GFAP (astrogliosis, 5 dal, * p < 0.05) than the C57BL/6J counterpart. Synaptic coverage (synaptophysin) was equivalent in both strains over time. In addition, mRNA expression of microdissected spinal motoneurons revealed an increase in cytoskeleton-associated molecules (cofilin, shp2, and crmp2, * p < 0.05), but not trkB, in C57BL/6J mice. Gait recovery, studied by the sciatic functional index, was faster in the A/J strain, despite the equivalent results of C57BL/6J at 28 days after injury. A similar recovery was also seen for the nociceptive threshold (von Frey test). Interestingly, when evaluating proprioceptive recovery, C57BL/6J animals showed an enlarged base of support, indicating abnormal ambulation postinjury. Overall, the present results reinforce the role of MHC-I expression in the plasticity of the nervous system following axotomy, which in turn correlates with the variable recovery capacity among strains of mice.
Collapse
|
27
|
Long-term effects of prenatal stress on the development of prefrontal cortex in the adolescent offspring. J Chem Neuroanat 2022; 125:102169. [DOI: 10.1016/j.jchemneu.2022.102169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 09/25/2022] [Accepted: 10/06/2022] [Indexed: 11/23/2022]
|
28
|
Smiley CE, Wood SK. Stress- and drug-induced neuroimmune signaling as a therapeutic target for comorbid anxiety and substance use disorders. Pharmacol Ther 2022; 239:108212. [PMID: 35580690 DOI: 10.1016/j.pharmthera.2022.108212] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 05/10/2022] [Accepted: 05/10/2022] [Indexed: 10/18/2022]
Abstract
Stress and substance use disorders remain two of the most highly prevalent psychiatric conditions and are often comorbid. While individually these conditions have a debilitating impact on the patient and a high cost to society, the symptomology and treatment outcomes are further exacerbated when they occur together. As such, there are few effective treatment options for these patients, and recent investigation has sought to determine the neural processes underlying the co-occurrence of these disorders to identify novel treatment targets. One such mechanism that has been linked to stress- and addiction-related conditions is neuroimmune signaling. Increases in inflammatory factors across the brain have been heavily implicated in the etiology of these disorders, and this review seeks to determine the nature of this relationship. According to the "dual-hit" hypothesis, also referred to as neuroimmune priming, prior exposure to either stress or drugs of abuse can sensitize the neuroimmune system to be hyperresponsive when exposed to these insults in the future. This review completes an examination of the literature surrounding stress-induced increases in inflammation across clinical and preclinical studies along with a summarization of the evidence regarding drug-induced alterations in inflammatory factors. These changes in neuroimmune profiles are also discussed within the context of their impact on the neural circuitry responsible for stress responsiveness and addictive behaviors. Further, this review explores the connection between neuroimmune signaling and susceptibility to these conditions and highlights the anti-inflammatory pharmacotherapies that may be used for the treatment of stress and substance use disorders.
Collapse
Affiliation(s)
- Cora E Smiley
- Department of Pharmacology, Physiology, and Neuroscience; University of South Carolina School of Medicine, Columbia, SC 29209, United States of America; WJB Dorn Veterans Administration Medical Center, Columbia, SC 29209, United States of America.
| | - Susan K Wood
- Department of Pharmacology, Physiology, and Neuroscience; University of South Carolina School of Medicine, Columbia, SC 29209, United States of America; WJB Dorn Veterans Administration Medical Center, Columbia, SC 29209, United States of America.
| |
Collapse
|
29
|
Navaei F, Fathabadi FF, Moghaddam MH, Fathi M, Vakili K, Abdollahifar MA, Boroujeni ME, Zamani N, Zamani N, Norouzian M, Aliaghaei A. Chronic exposure to methadone impairs memory, induces microgliosis, astrogliosis and neuroinflammation in the hippocampus of adult male rats. J Chem Neuroanat 2022; 125:102139. [PMID: 35872237 DOI: 10.1016/j.jchemneu.2022.102139] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 07/18/2022] [Accepted: 07/19/2022] [Indexed: 01/15/2023]
Abstract
Methadone is a centrally-acting synthetic opioid analgesic widely used in methadone maintenance therapy (MMT) programs throughout the world. Given its neurotoxic effects, particularly on the hippocampus, this study aims to address the behavioral and histological alterations in the hippocampus associated with methadone administration. To do so, twenty-four adult male albino rats were randomized into two groups, methadone treatment and control. Methadone was administered subcutaneously (2.5-10 mg/kg) once a day for two consecutive weeks. A comparison was drawn with behavioral and structural changes recorded in the control group. The results showed that methadone administration interrupted spatial learning and memory function. Accordingly, treating rats with methadone not only induced cell death but also prompted the actuation of microgliosis, astrogliosis, and apoptotic biomarkers. Furthermore, the results demonstrated that treating rats with methadone decreased the complexity of astrocyte processes and the complexity of microglia processes. These findings suggest that methadone altered the special distribution of neurons. Also, a substantial increase was observed in the expression of TNF-α due to methadone. According to the findings, methadone administration exerts a neurodegenerative effect on the hippocampus via dysregulation of microgliosis, astrogliosis, apoptosis, and neuro-inflammation.
Collapse
Affiliation(s)
- Fatemeh Navaei
- Hearing Disorders Research Center, Loghman-Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, the Islamic Republic of Iran; Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, the Islamic Republic of Iran
| | - Fatemeh Fadaei Fathabadi
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, the Islamic Republic of Iran
| | - Meysam Hassani Moghaddam
- Department of Anatomical Sciences, Faculty of Medicine, AJA University of Medical Sciences, Tehran, the Islamic Republic of Iran
| | - Mobina Fathi
- Student Research Committee, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, the Islamic Republic of Iran
| | - Kimia Vakili
- Student Research Committee, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, the Islamic Republic of Iran
| | - Mohammad-Amin Abdollahifar
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, the Islamic Republic of Iran
| | - Mahdi Eskandarian Boroujeni
- Department of Human Molecular Genetics, Institute of Molecular Biology and Biotechnology, Faculty of Biology, Adam Mickiewicz University, Poznan, Poland
| | - Naghmeh Zamani
- Department of Biology, Tehran North Branch, Islamic Azad University, Tehran, the Islamic Republic of Iran
| | - Nasim Zamani
- Department of Clinical Toxicology, Loghman-Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, the Islamic Republic of Iran
| | - Mohsen Norouzian
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, the Islamic Republic of Iran.
| | - Abbas Aliaghaei
- Hearing Disorders Research Center, Loghman-Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, the Islamic Republic of Iran; Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, the Islamic Republic of Iran.
| |
Collapse
|
30
|
Souza LC, Andrade MK, Azevedo EM, Ramos DC, Bail EL, Vital MABF. Andrographolide Attenuates Short-Term Spatial and Recognition Memory Impairment and Neuroinflammation Induced by a Streptozotocin Rat Model of Alzheimer's Disease. Neurotox Res 2022; 40:1440-1454. [PMID: 36029454 DOI: 10.1007/s12640-022-00569-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 08/12/2022] [Accepted: 08/22/2022] [Indexed: 11/28/2022]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder clinically manifested by a gradual cognitive decline. Intracerebroventricular injection (ICV) of streptozotocin (STZ), a model of sporadic AD (sAD), shows many aspects of sAD abnormalities (i.e., neuroinflammation, oxidative stress, protein aggregation), resulting in memory impairment. Andrographolide (ANDRO), a natural diterpene lactone, has numerous bioactivities including anti-inflammatory and antioxidant properties. Studies in rodents revealed that ANDRO has neuroprotective properties and restores cognitive impairment. In the present study, we investigated the effects of ANDRO in the ICV-STZ model relative to short-term spatial memory (object location test (OLT) and Y maze test), short-term recognition memory (object recognition test (ORT)), locomotor activity (open field test (OFT)), expression of amyloid precursor protein (APP), and activation of astrocytes (glial fibrillary acidic protein (GFAP) expression) and microglia (ionized calcium-binding adapter molecule-1 (Iba-1) immunohistochemistry) in the prefrontal cortex (PFC) and hippocampus (HIP). Wistar rats were injected ICV with STZ (3 mg/kg) or vehicle and treated with ANDRO (2 mg/kg, i.p.; three times per week). After four weeks, ANDRO attenuated the impairments of the Y maze and ORT performances, and the increase of astrocyte activation in the PFC induced by the ICV-STZ model. In addition, ANDRO decreased the number of activated microglia cells in the HIP of STZ-injected rats. The APP expression was not altered, neither by the STZ nor ANDRO. ANDRO showed a beneficial effect on memory impairment and neuroinflammation in the STZ model of AD.
Collapse
Affiliation(s)
- Leonardo C Souza
- Department of Pharmacology, Federal University of Paraná, Curitiba, PR, Brazil.
| | - Marcos K Andrade
- Department of Pharmacology, Federal University of Paraná, Curitiba, PR, Brazil
| | - Evellyn M Azevedo
- Department of Physiology, Federal University of Paraná, Curitiba, PR, Brazil
| | - Daniele C Ramos
- Department of Pharmacology, Federal University of Paraná, Curitiba, PR, Brazil
| | - Ellen L Bail
- Department of Physiology, Federal University of Paraná, Curitiba, PR, Brazil
| | - Maria A B F Vital
- Department of Pharmacology, Federal University of Paraná, Curitiba, PR, Brazil
| |
Collapse
|
31
|
Zhao F, Li B, Yang W, Ge T, Cui R. Brain-immune interaction mechanisms: Implications for cognitive dysfunction in psychiatric disorders. Cell Prolif 2022; 55:e13295. [PMID: 35860850 PMCID: PMC9528770 DOI: 10.1111/cpr.13295] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 04/28/2022] [Accepted: 06/07/2022] [Indexed: 12/15/2022] Open
Abstract
Objectives Cognitive dysfunction has been identified as a major symptom of a series of psychiatric disorders. Multidisciplinary studies have shown that cognitive dysfunction is monitored by a two‐way interaction between the neural and immune systems. However, the specific mechanisms of cognitive dysfunction in immune response and brain immune remain unclear. Materials and methods In this review, we summarized the relevant research to uncover our comprehension of the brain–immune interaction mechanisms underlying cognitive decline. Results The pathophysiological mechanisms of brain‐immune interactions in psychiatric‐based cognitive dysfunction involve several specific immune molecules and their associated signaling pathways, impairments in neural and synaptic plasticity, and the potential neuro‐immunological mechanism of stress. Conclusions Therefore, this review may provide a better theoretical basis for integrative therapeutic considerations for psychiatric disorders associated with cognitive dysfunction.
Collapse
Affiliation(s)
- Fangyi Zhao
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| | - Bingjin Li
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| | - Wei Yang
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| | - Tongtong Ge
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| | - Ranji Cui
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
32
|
Nahum K, Todder D, Zohar J, Cohen H. The Role of Microglia in the (Mal)adaptive Response to Traumatic Experience in an Animal Model of PTSD. Int J Mol Sci 2022; 23:ijms23137185. [PMID: 35806185 PMCID: PMC9266429 DOI: 10.3390/ijms23137185] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 06/22/2022] [Accepted: 06/23/2022] [Indexed: 01/06/2023] Open
Abstract
The present study investigates whether predator scent-stress (PSS) shifts the microglia from a quiescent to a chronically activated state and whether morphological alterations in microglial activation differ between individuals displaying resilient vs. vulnerable phenotypes. In addition, we examined the role that GC receptors play during PSS exposure in the impairment of microglial activation and thus in behavioral response. Adult male Sprague Dawley rats were exposed to PSS or sham-PSS for 15 min. Behaviors were assessed with the elevated plus-maze (EPM) and acoustic startle response (ASR) paradigms 7 days later. Localized brain expression of Iba-1 was assessed, visualized, and classified based on their morphology and stereological counted. Hydrocortisone and RU486 were administered systemically 10 min post PSS exposure and behavioral responses were measured on day 7 and hippocampal expression of Ionized calcium-binding adaptor molecule 1 (Iba-1) was subsequently evaluated. Animals whose behavior was extremely disrupted (PTSD-phenotype) selectively displayed excessive expression of Iba-1 with concomitant downregulation in the expression of CX3C chemokine receptor 1 (CX3CR1) in hippocampal structures as compared with rats whose behavior was minimally or partially disrupted. Changes in microglial morphology have also been related only to the PTSD-phenotype group. These data indicate that PSS-induced microglia activation in the hippocampus serves as a critical mechanistic link between the HPA-axis and PSS-induced impairment in behavioral responses.
Collapse
Affiliation(s)
- Kesem Nahum
- Department of Psychology Experimental Psychology, Brain and Cognition, Faculty of Humanities and Social Sciences, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel;
| | - Doron Todder
- Beer-Sheva Mental Health Center, Ministry of Health, Anxiety and Stress Research Unit, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 8461144, Israel;
| | - Joseph Zohar
- Post-Trauma Center, Sheba Medical Center, Tel Aviv University, Tel Aviv 52621, Israel;
| | - Hagit Cohen
- Department of Psychology Experimental Psychology, Brain and Cognition, Faculty of Humanities and Social Sciences, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel;
- Beer-Sheva Mental Health Center, Ministry of Health, Anxiety and Stress Research Unit, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 8461144, Israel;
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
- Correspondence: ; Tel.: +972-8-6401742
| |
Collapse
|
33
|
Olude MA, Mouihate A, Mustapha OA, Farina C, Quintana FJ, Olopade JO. Astrocytes and Microglia in Stress-Induced Neuroinflammation: The African Perspective. Front Immunol 2022; 13:795089. [PMID: 35707531 PMCID: PMC9190229 DOI: 10.3389/fimmu.2022.795089] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 04/13/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Africa is laden with a youthful population, vast mineral resources and rich fauna. However, decades of unfortunate historical, sociocultural and leadership challenges make the continent a hotspot for poverty, indoor and outdoor pollutants with attendant stress factors such as violence, malnutrition, infectious outbreaks and psychological perturbations. The burden of these stressors initiate neuroinflammatory responses but the pattern and mechanisms of glial activation in these scenarios are yet to be properly elucidated. Africa is therefore most vulnerable to neurological stressors when placed against a backdrop of demographics that favor explosive childbearing, a vast population of unemployed youths making up a projected 42% of global youth population by 2030, repressive sociocultural policies towards women, poor access to healthcare, malnutrition, rapid urbanization, climate change and pollution. Early life stress, whether physical or psychological, induces neuroinflammatory response in developing nervous system and consequently leads to the emergence of mental health problems during adulthood. Brain inflammatory response is driven largely by inflammatory mediators released by glial cells; namely astrocytes and microglia. These inflammatory mediators alter the developmental trajectory of fetal and neonatal brain and results in long-lasting maladaptive behaviors and cognitive deficits. This review seeks to highlight the patterns and mechanisms of stressors such as poverty, developmental stress, environmental pollutions as well as malnutrition stress on astrocytes and microglia in neuroinflammation within the African context.
Collapse
Affiliation(s)
- Matthew Ayokunle Olude
- Vertebrate Morphology, Environmental Toxicology and Neuroscience Unit, College of Veterinary Medicine, Federal University of Agriculture, Abeokuta, Nigeria
- *Correspondence: Matthew Ayokunle Olude,
| | - Abdeslam Mouihate
- Department of Physiology, Faculty of Medicine, Health Sciences Centre, Kuwait University, Kuwait City, Kuwait
| | - Oluwaseun Ahmed Mustapha
- Vertebrate Morphology, Environmental Toxicology and Neuroscience Unit, College of Veterinary Medicine, Federal University of Agriculture, Abeokuta, Nigeria
| | - Cinthia Farina
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCSS) San Raffaele Scientific Institute, Institute of Experimental Neurology (INSPE) and Division of Neuroscience, Milan, Italy
| | - Francisco Javier Quintana
- Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - James Olukayode Olopade
- Neuroscience Unit, Department of Veterinary Anatomy, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| |
Collapse
|
34
|
The maternal-placental-fetal interface: Adaptations of the HPA axis and immune mediators following maternal stress and prenatal alcohol exposure. Exp Neurol 2022; 355:114121. [DOI: 10.1016/j.expneurol.2022.114121] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Revised: 05/15/2022] [Accepted: 05/17/2022] [Indexed: 11/18/2022]
|
35
|
Davis JLB, O’Connor M, Erlbacher H, Schlichte SL, Stevens HE. The Impact of Maternal Antioxidants on Prenatal Stress Effects on Offspring Neurobiology and Behavior. THE YALE JOURNAL OF BIOLOGY AND MEDICINE 2022; 95:87-104. [PMID: 35370489 PMCID: PMC8961714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Prenatal stress is a neuropsychiatric risk factor, and effects may be mediated by prenatal oxidative stress. Cell types in the brain sensitive to oxidative stress-cortical microglia and cortical and hippocampal interneurons-may be altered by oxidative stress generated during prenatal stress and may be neurobiological substrates for altered behavior. Our objective was to determine the critical nature of oxidative stress in prenatal stress effects by manipulating prenatal antioxidants. CD1 mouse dams underwent restraint embryonic day 12 to 18 three times daily or no stress and received intraperitoneal injections before each stress period of vehicle, N-acetylcysteine (200 mg/kg daily), or astaxanthin (30 mg/kg before first daily stress, 10 mg/kg before second/third stresses). Adult male and female offspring behavior, microglia, and interneurons were assessed. Results supported the hypothesis that prenatal stress-induced oxidative stress affects microglia; microglia ramification increased after prenatal stress, and both antioxidants prevented these effects. In addition, N-acetylcysteine or astaxanthin was effective in preventing distinct male and female interneuron changes; decreased female medial frontal cortical parvalbumin interneurons was prevented by either antioxidant; increased male medial frontal cortical parvalbumin interneurons was prevented by N-acetylcysteine and decreased male hippocampal GAD67GFP+ cells prevented by astaxanthin. Prenatal stress-induced increased anxiety-like behavior and decreased sociability were not prevented by prenatal antioxidants. Sensorimotor gating deficits in males was partially prevented by prenatal astaxanthin. This study demonstrates the importance of oxidative stress for persistent impacts on offspring cortical microglia and interneurons, but did not link these changes with anxiety-like, social, and sensorimotor gating behaviors.
Collapse
Affiliation(s)
- Jada L-B Davis
- Department of Psychiatry, University of Iowa, Iowa
City, IA, USA
- Interdisciplinary Graduate Program in Neuroscience,
University of Iowa, Iowa City, IA, USA
| | - Mara O’Connor
- Department of Psychiatry, University of Iowa, Iowa
City, IA, USA
| | - Hannah Erlbacher
- Department of Psychiatry, University of Iowa, Iowa
City, IA, USA
| | | | - Hanna E. Stevens
- Department of Psychiatry, University of Iowa, Iowa
City, IA, USA
- Interdisciplinary Graduate Program in Neuroscience,
University of Iowa, Iowa City, IA, USA
| |
Collapse
|
36
|
Cartarozzi LP, Perez M, Fernandes GG, Chiarotto GB, Luzo ÂCM, Campos AC, Kirchhoff F, de Oliveira ALR. Neuroprotection and gliosis attenuation by intravenous application of human mesenchymal stem cells (hMSC) following ventral root crush in mice. Mol Cell Neurosci 2021; 118:103694. [PMID: 34954382 DOI: 10.1016/j.mcn.2021.103694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 12/13/2021] [Accepted: 12/17/2021] [Indexed: 11/15/2022] Open
Abstract
Rupture and stretching of spinal roots are common incidents that take place in high-energy accidents. The proximal axotomy of motoneurons by crushing of ventral roots is directly related to the degeneration of half of the lesioned population within the first two weeks. Moreover, only a small percentage of surviving motoneurons can successfully achieve regeneration after such a proximal lesion, and new treatments are necessary to improve this scenario. In this sense, mesenchymal stem cells (MSC) are of great interest once they secrete a broad spectrum of bioactive molecules that are immunomodulatory and can restore the environment after a lesion. The present work aimed at studying the effects of human mesenchymal stem cells (hMSC) therapy after ventral root crush (VRC) in mice. We evaluated motoneuron survival, glial reaction, and synapse preservation at the ventral horn. For this purpose, C57BL/6 J were submitted to a crush procedure of L4 to L6 ventral roots and treated with a single intravenous injection of adipose-derived hMSC. Evaluation of the results was carried out at 7, 14, and 28 days after injury. Analysis of motoneuron survival and astrogliosis showed that hMSC treatment resulted in higher motoneuron preservation (motoneuron survival ipsi/contralateral ratio: VRC group = 53%, VRC + hMSC group = 66%; p < 0.01), combined with reduction of astrogliosis (ipsi/contralateral GFAP immunolabeling: VRC group = 470%, VRC + hMSC group = 250%; p < 0.001). The morphological classification and Sholl analysis of microglial activation revealed that hMSC treatment reduced type V and increased type II profiles, indicating an enhancement of surveying over activated microglial cells. The glial reactivity modulation directly influenced synaptic inputs in apposition to axotomized motoneurons. In the hMSC-treated group, synaptic maintenance was increased (ipsi/contralateral synaptophysin immunolabeling: VRC group = 53%, VRC + hMSC group = 64%; p < 0.05). Overall, the present data show that intravenous injection of hMSC has neuroprotective and anti-inflammatory effects, decreasing reactive astrogliosis, and microglial reaction. Also, such cell therapy results in motoneuron preservation, combined with significant maintenance of spinal cord circuits, in particular those related to the ventral horn.
Collapse
Affiliation(s)
- Luciana Politti Cartarozzi
- Laboratory of Nerve Regeneration, University of Campinas - UNICAMP, Cidade Universitaria "Zeferino Vaz", Rua Monteiro Lobato, 255, 13083-970 Campinas, SP, Brazil
| | - Matheus Perez
- School of Physical Education and Sport of Ribeirão Preto, University of São Paulo, Av. Bandeirantes, 3900, 14040-907 Ribeirão Preto, SP, Brazil
| | - Gabriel Gripp Fernandes
- Department of Pharmacology, School of Medicine of Ribeirão Preto, University of São Paulo, Av. Bandeirantes, 3900, 14040-907 Ribeirão Preto, SP, Brazil
| | - Gabriela Bortolança Chiarotto
- Laboratory of Nerve Regeneration, University of Campinas - UNICAMP, Cidade Universitaria "Zeferino Vaz", Rua Monteiro Lobato, 255, 13083-970 Campinas, SP, Brazil
| | - Ângela Cristina Malgeiros Luzo
- Hematology and Hemotherapy Center, University of Campinas/Hemocentro-Unicamp, Instituto Nacional de Ciência e Tecnologia do Sangue, Campinas, São Paulo, Brazil
| | - Alline Cristina Campos
- Department of Pharmacology, School of Medicine of Ribeirão Preto, University of São Paulo, Av. Bandeirantes, 3900, 14040-907 Ribeirão Preto, SP, Brazil
| | - Frank Kirchhoff
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, Building 48, 66421 Homburg, Germany
| | - Alexandre Leite Rodrigues de Oliveira
- Laboratory of Nerve Regeneration, University of Campinas - UNICAMP, Cidade Universitaria "Zeferino Vaz", Rua Monteiro Lobato, 255, 13083-970 Campinas, SP, Brazil.
| |
Collapse
|
37
|
Carloni E, Ramos A, Hayes LN. Developmental Stressors Induce Innate Immune Memory in Microglia and Contribute to Disease Risk. Int J Mol Sci 2021; 22:13035. [PMID: 34884841 PMCID: PMC8657756 DOI: 10.3390/ijms222313035] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 11/23/2021] [Accepted: 11/25/2021] [Indexed: 12/26/2022] Open
Abstract
Many types of stressors have an impact on brain development, function, and disease susceptibility including immune stressors, psychosocial stressors, and exposure to drugs of abuse. We propose that these diverse developmental stressors may utilize a common mechanism that underlies impaired cognitive function and neurodevelopmental disorders such as schizophrenia, autism, and mood disorders that can develop in later life as a result of developmental stressors. While these stressors are directed at critical developmental windows, their impacts are long-lasting. Immune activation is a shared pathophysiology across several different developmental stressors and may thus be a targetable treatment to mitigate the later behavioral deficits. In this review, we explore different types of prenatal and perinatal stressors and their contribution to disease risk and underlying molecular mechanisms. We highlight the impact of developmental stressors on microglia biology because of their early infiltration into the brain, their critical role in brain development and function, and their long-lived status in the brain throughout life. Furthermore, we introduce innate immune memory as a potential underlying mechanism for developmental stressors' impact on disease. Finally, we highlight the molecular and epigenetic reprogramming that is known to underlie innate immune memory and explain how similar molecular mechanisms may be at work for cells to retain a long-term perturbation after exposure to developmental stressors.
Collapse
Affiliation(s)
- Elisa Carloni
- Department of Molecular and Cellular Biology, Dartmouth College, Hanover, NH 03755, USA;
| | - Adriana Ramos
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA;
| | - Lindsay N. Hayes
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| |
Collapse
|
38
|
Guerrin CGJ, Doorduin J, Sommer IE, de Vries EFJ. The dual hit hypothesis of schizophrenia: Evidence from animal models. Neurosci Biobehav Rev 2021; 131:1150-1168. [PMID: 34715148 DOI: 10.1016/j.neubiorev.2021.10.025] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 10/21/2021] [Accepted: 10/24/2021] [Indexed: 12/16/2022]
Abstract
Schizophrenia is a heterogeneous psychiatric disorder, which can severely impact social and professional functioning. Epidemiological and clinical studies show that schizophrenia has a multifactorial aetiology comprising genetic and environmental risk factors. Although several risk factors have been identified, it is still not clear how they result in schizophrenia. This knowledge gap, however, can be investigated in animal studies. In this review, we summarise animal studies regarding molecular and cellular mechanisms through which genetic and environmental factors may affect brain development, ultimately causing schizophrenia. Preclinical studies suggest that early environmental risk factors can affect the immune, GABAergic, glutamatergic, or dopaminergic system and thus increase the susceptibility to another risk factor later in life. A second insult, like social isolation, stress, or drug abuse, can further disrupt these systems and the interactions between them, leading to behavioural abnormalities. Surprisingly, first insults like maternal infection and early maternal separation can also have protective effects. Single gene mutations associated with schizophrenia did not have a major impact on the susceptibility to subsequent environmental hits.
Collapse
Affiliation(s)
- Cyprien G J Guerrin
- Department of Nuclear Medicine and Medical Imaging, University of Groningen, University Medical Centre Groningen, Hanzeplein 1, 9713, GZ, Groningen, the Netherlands
| | - Janine Doorduin
- Department of Nuclear Medicine and Medical Imaging, University of Groningen, University Medical Centre Groningen, Hanzeplein 1, 9713, GZ, Groningen, the Netherlands
| | - Iris E Sommer
- Department of Biomedical Sciences of Cells and Systems, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713, GZ, Groningen, the Netherlands
| | - Erik F J de Vries
- Department of Nuclear Medicine and Medical Imaging, University of Groningen, University Medical Centre Groningen, Hanzeplein 1, 9713, GZ, Groningen, the Netherlands.
| |
Collapse
|
39
|
Kim J, Choi H, Kang EK, Ji GY, Kim Y, Choi IS. In Vitro Studies on Therapeutic Effects of Cannabidiol in Neural Cells: Neurons, Glia, and Neural Stem Cells. Molecules 2021; 26:molecules26196077. [PMID: 34641624 PMCID: PMC8512311 DOI: 10.3390/molecules26196077] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 09/28/2021] [Accepted: 09/30/2021] [Indexed: 11/25/2022] Open
Abstract
(‒)-Cannabidiol (CBD) is one of the major phytocannabinoids extracted from the Cannabis genus. Its non-psychoactiveness and therapeutic potential, partly along with some anecdotal—if not scientific or clinical—evidence on the prevention and treatment of neurological diseases, have led researchers to investigate the biochemical actions of CBD on neural cells. This review summarizes the previously reported mechanistic studies of the CBD actions on primary neural cells at the in vitro cell-culture level. The neural cells are classified into neurons, microglia, astrocytes, oligodendrocytes, and neural stem cells, and the CBD effects on each cell type are described. After brief introduction on CBD and in vitro studies of CBD actions on neural cells, the neuroprotective capability of CBD on primary neurons with the suggested operating actions is discussed, followed by the reported CBD actions on glia and the CBD-induced regeneration from neural stem cells. A summary section gives a general overview of the biochemical actions of CBD on neural cells, with a future perspective. This review will provide a basic and fundamental, but crucial, insight on the mechanistic understanding of CBD actions on neural cells in the brain, at the molecular level, and the therapeutic potential of CBD in the prevention and treatment of neurological diseases, although to date, there seem to have been relatively limited research activities and reports on the cell culture-level, in vitro studies of CBD effects on primary neural cells.
Collapse
Affiliation(s)
- Jungnam Kim
- Department of Chemistry, KAIST, Daejeon 34141, Korea; (J.K.); (H.C.); (E.K.K.)
| | - Hyunwoo Choi
- Department of Chemistry, KAIST, Daejeon 34141, Korea; (J.K.); (H.C.); (E.K.K.)
| | - Eunhye K. Kang
- Department of Chemistry, KAIST, Daejeon 34141, Korea; (J.K.); (H.C.); (E.K.K.)
| | - Gil Yong Ji
- Cannabis Medical, Inc., Sandong-ro 433-31, Eumbong-myeon, Asan-si 31418, Korea; (G.Y.J.); (Y.K.)
| | - Youjeong Kim
- Cannabis Medical, Inc., Sandong-ro 433-31, Eumbong-myeon, Asan-si 31418, Korea; (G.Y.J.); (Y.K.)
| | - Insung S. Choi
- Department of Chemistry, KAIST, Daejeon 34141, Korea; (J.K.); (H.C.); (E.K.K.)
- Correspondence:
| |
Collapse
|
40
|
Bollinger J. Uncovering microglial pathways driving sex-specific neurobiological effects in stress and depression. Brain Behav Immun Health 2021; 16:100320. [PMID: 34589809 PMCID: PMC8474553 DOI: 10.1016/j.bbih.2021.100320] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 08/03/2021] [Accepted: 08/03/2021] [Indexed: 12/13/2022] Open
Abstract
Women suffer from major depressive disorder (MDD) more often than men and report greater MDD symptom severity. Mounting evidence suggests that sex differences in MDD may be driven, in part, by sex-specific neurobiological mechanisms. Chronic stress is a significant risk factor in MDD, and preclinical rodent models show differential patterns of stress-induced neural remodeling and cognitive-behavioral dysfunction in males and females. For instance, chronic stress leads to synapse loss in the medial prefrontal cortex in male rodents yet has either no effect on- or increases-synapse number in females. Recent reports have implicated microglia, the immune cells of the brain, in MDD, and findings demonstrate sex-specific microglial signatures in both preclinical stress models and MDD patients. Given that microglia can remodel neural architecture, modulate synaptic transmission, and affect subsequent changes in behavior, it is plausible that microglial pathways contribute to differential stress effects on neuroplasticity and function in males and females. As such, this review examines the evidence for sex-specific microglia-neuron interactions in preclinical stress models and in patients with MDD. Discoveries highlighted herein demonstrate divergent microglial contributions in males and females and suggest that future studies investigating stress-linked disorders should be guided by sex-dependent neurobiological and behavioral findings. Examining these pathways represents a clear avenue toward both a richer understanding of brain, behavior, and immunity, and innovative psychoneuroimmunology-based applications in personalized medicine.
Collapse
Affiliation(s)
- J.L. Bollinger
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, 2120 East Galbraith Road, Cincinnati, OH, 45237, USA
| |
Collapse
|
41
|
Maternal separation leads to regional hippocampal microglial activation and alters the behavior in the adolescence in a sex-specific manner. Brain Behav Immun Health 2021; 9:100142. [PMID: 34589889 PMCID: PMC8474514 DOI: 10.1016/j.bbih.2020.100142] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 09/04/2020] [Accepted: 09/13/2020] [Indexed: 12/31/2022] Open
Abstract
Early life adversities during childhood (such as maltreatment, abuse, neglect, or parental deprivation) may increase the vulnerability to cognitive disturbances and emotional disorders in both, adolescence and adulthood. Maternal separation (MS) is a widely used model to study stress-related changes in brain and behavior in rodents. In this study, we investigated the effect of MS (postnatal day 2–14, 3 h/day) in both, female and male adolescent mice. Specifically, we evaluated (i) the spatial working memory, anxiety and depressive-like behavior, (ii) the hippocampal synaptic gene expression, and (iii) the hippocampal neuroinflammatory response. Our results show that MS significantly increased depressive-like behavior in adolescent female mice and altered the spatial memory in adolescent male mice. In addition, MS led to decreased expression of genes related to synaptic function (5ht6r, Synaptophysin, and Cox-2) and induced an exacerbated microglial activation in dentate gyrus (DG), CA1, and CA3. However, while the levels of hippocampal inflammatory cytokines were not modified by MS, they did follow a sex-specific expression in adolescent mice. Taken together, our results suggest that MS induces long-term changes in hippocampal microglia and synaptic gene expression, alters the spatial memory, and induces depressive-like behavior in the adolescent mice, in a sex-specific manner. In wildtype adolescent mice (6 weeks of age):Maternal separation alters spatial working memory in males and induces depressive-like behavior in females. Maternal separation changes hippocampal synaptic gene expression. Maternal separation activates microglia in dentate gyrus, CA1, and CA3 but does not affect hippocampal cytokine levels. However, males present higher levels of cytokines compared to females.
Collapse
|
42
|
Roshan-Milani S, Seyyedabadi B, Saboory E, Parsamanesh N, Mehranfard N. Prenatal stress and increased susceptibility to anxiety-like behaviors: role of neuroinflammation and balance between GABAergic and glutamatergic transmission. Stress 2021; 24:481-495. [PMID: 34180763 DOI: 10.1080/10253890.2021.1942828] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
Neuroplasticity during the prenatal period allows neurons to regenerate anatomically and functionally for re-programming the brain development. During this critical period of fetal programming, the fetus phenotype can change in accordance with environmental stimuli such as stress exposure. Prenatal stress (PS) can exert important effects on brain development and result in permanent alterations with long-lasting consequences on the physiology and behavior of the offspring later in life. Neuroinflammation, as well as GABAergic and glutamatergic dysfunctions, has been implicated as potential mediators of behavioral consequences of PS. Hyperexcitation, due to enhanced excitatory transmission or reduced inhibitory transmission, can promote anxiety. Alterations of the GABAergic and/or glutamatergic signaling during fetal development lead to a severe excitatory/inhibitory imbalance in neuronal circuits, a condition that may account for PS-precipitated anxiety-like behaviors. This review summarizes experimental evidence linking PS to an elevated risk to anxiety-like behaviors and interprets the role of the neuroinflammation and alterations of the brain GABAergic and glutamatergic transmission in this phenomenon. We hypothesize this is an imbalance in GABAergic and glutamatergic circuits (as a direct or indirect consequence of neuroinflammation), which at least partially contributes to PS-precipitated anxiety-like behaviors and primes the brain to be vulnerable to anxiety disorders. Therefore, pharmacological interventions with anti-inflammatory activities and with regulatory effects on the excitatory/inhibitory balance can be attributed to the novel therapeutic target for anxiety disorders.
Collapse
Affiliation(s)
- Shiva Roshan-Milani
- Department of Physiology, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
- Neurophysiology Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Urmia, Iran
| | | | - Ehsan Saboory
- Zanjan Metabolic Diseases Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Negin Parsamanesh
- Zanjan Metabolic Diseases Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Nasrin Mehranfard
- Neurophysiology Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Urmia, Iran
| |
Collapse
|
43
|
Highlighting Immune System and Stress in Major Depressive Disorder, Parkinson's, and Alzheimer's Diseases, with a Connection with Serotonin. Int J Mol Sci 2021; 22:ijms22168525. [PMID: 34445231 PMCID: PMC8395198 DOI: 10.3390/ijms22168525] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 08/01/2021] [Accepted: 08/03/2021] [Indexed: 01/10/2023] Open
Abstract
There is recognition that both stress and immune responses are important factors in a variety of neurological disorders. Moreover, there is an important role of several neurotransmitters that connect these factors to several neurological diseases, with a special focus in this paper on serotonin. Accordingly, it is known that imbalances in stressors can promote a variety of neuropsychiatric or neurodegenerative pathologies. Here, we discuss some facts that link major depressive disorder, Alzheimer’s, and Parkinson’s to the stress and immune responses, as well as the connection between these responses and serotonergic signaling. These are important topics of investigation which may lead to new or better treatments, improving the life quality of patients that suffer from these conditions.
Collapse
|
44
|
Microglia react to partner loss in a sex- and brain site-specific manner in prairie voles. Brain Behav Immun 2021; 96:168-186. [PMID: 34058309 PMCID: PMC8319132 DOI: 10.1016/j.bbi.2021.05.026] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 05/25/2021] [Accepted: 05/26/2021] [Indexed: 12/21/2022] Open
Abstract
Positive social relationships are paramount for the survival of mammals and beneficial for mental and physical health, buffer against stressors, and even promote appropriate immune system functioning. By contrast, impaired social relationships, social isolation, or the loss of a bonded partner lead to aggravated physical and mental health. For example, in humans partner loss is detrimental for the functioning of the immune system and heightens the susceptibility for the development of post-traumatic stress disorders, anxiety disorders, and major depressive disorders. To understand potential underlying mechanisms, the monogamous prairie vole can provide important insights. In the present study, we separated pair bonded male and female prairie voles after five days of co-housing, subjected them to the forced swim test on the fourth day following separation, and studied their microglia morphology and activation in specific brain regions. Partner loss increased passive stress-coping in male, but not female, prairie voles. Moreover, partner loss was associated with microglial priming within the parvocellular region of the paraventricular nucleus of the hypothalamus (PVN) in male prairie voles, whereas in female prairie voles the morphological activation within the whole PVN and the prelimbic cortex (PrL) was decreased, marked by a shift towards ramified microglial morphology. Expression of the immediate early protein c-Fos following partner loss was changed within the PrL of male, but not female, prairie voles. However, the loss of a partner did not affect the investigated aspects of the peripheral immune response. These data suggest a potential sex-dependent mechanism for the regulation of microglial activity following the loss of a partner, which might contribute to the observed differences in passive stress-coping. This study furthers our understanding of the effects of partner loss and its short-term impact on the CNS as well as the CNS immune system and the peripheral innate immune system in both male and female prairie voles.
Collapse
|
45
|
Herrera ML, Bandín S, Champarini LG, Hereñú CB, Bellini MJ. Intramuscular insulin-like growth factor-1 gene therapy modulates reactive microglia after traumatic brain injury. Brain Res Bull 2021; 175:196-204. [PMID: 34339780 DOI: 10.1016/j.brainresbull.2021.07.023] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 06/07/2021] [Accepted: 07/27/2021] [Indexed: 12/23/2022]
Abstract
Reactive gliosis is a key feature and an important pathophysiological mechanism underlying chronic neurodegeneration following traumatic brain injury (TBI). In this study, we have explored the effects of intramuscular IGF-1 gene therapy on reactive gliosis and functional outcome after an injury of the cerebral cortex. Young adult male rats were intramuscularly injected with a recombinant adenoviral construct harboring the cDNA of human IGF-1 (RAd-IGF1), with a control vector expressing green fluorescent protein (RAd-GFP) or PBS as control. Three weeks after the intramuscular injections of adenoviral vectors, animals were subjected to a unilateral penetrating brain injury. The data revealed that RAd-IGF1 gene therapy significantly increased serum IGF1 levels and improved working memory performance after one week of TBI as compared to PBS or RAd-GFP lesioned animals. At the same time, when we analyzed the effects of therapy on glial scar formation, the treatment with RAd-IGF1 did not modify the number of glial fibrillary acidic protein (GFAP) positive cells, but we observed a decrease in vimentin immunoreactive astrocytes at 7 days post-lesion in the injured hemisphere compared to RAd-GFP group. Moreover, IGF-1 gene therapy reduced the number of Iba1+ cells with reactive phenotype and the number of MHCII + cells in the injured hemisphere. These results suggest that intramuscular IGF-1 gene therapy may represent a new approach to prevent traumatic brain injury outcomes in rats.
Collapse
Affiliation(s)
- Macarena Lorena Herrera
- Universidad Nacional de Córdoba, Facultad de Ciencias Químicas, Departamento de Farmacología, Córdoba, Argentina; Instituto de Farmacología Experimental de Córdoba (IFEC-CONICET), Córdoba, Argentina; Universidad Nacional de La Plata, Facultad de Ciencias Médicas, Buenos Aires, Argentina; Instituto de Investigaciones Bioquímicas de La Plata (INIBIOLP-CONICET), La Plata, Argentina
| | - Sandra Bandín
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), 28002 Madrid, Spain
| | - Leandro Gabriel Champarini
- Universidad Nacional de Córdoba, Facultad de Ciencias Químicas, Departamento de Farmacología, Córdoba, Argentina; Instituto de Farmacología Experimental de Córdoba (IFEC-CONICET), Córdoba, Argentina
| | - Claudia Beatriz Hereñú
- Universidad Nacional de Córdoba, Facultad de Ciencias Químicas, Departamento de Farmacología, Córdoba, Argentina; Instituto de Farmacología Experimental de Córdoba (IFEC-CONICET), Córdoba, Argentina
| | - Maria Jose Bellini
- Universidad Nacional de La Plata, Facultad de Ciencias Médicas, Buenos Aires, Argentina; Instituto de Investigaciones Bioquímicas de La Plata (INIBIOLP-CONICET), La Plata, Argentina.
| |
Collapse
|
46
|
Luft C, Haute GV, Wearick-Silva LE, Antunes KH, da Costa MS, de Oliveira JR, Donadio MVF. Prenatal stress and KCl-induced depolarization modulate cell death, hypothalamic-pituitary-adrenal axis genes, oxidative and inflammatory response in primary cortical neurons. Neurochem Int 2021; 147:105053. [PMID: 33961947 DOI: 10.1016/j.neuint.2021.105053] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 04/01/2021] [Accepted: 04/24/2021] [Indexed: 12/27/2022]
Abstract
Maternal stress has been described as an important component in the offspring's cerebral development, altering the susceptibility to diseases in later life. Moreover, the postnatal period is essential for the development and integration of several peripheral and central systems related to the control of homeostasis. Thus, this study aimed to evaluate the effects of prenatal stress on the activation of cortical neurons, by performing experiments both under basal conditions and after KCl-induced depolarization. Female mice were divided in two groups: control and prenatal restraint stress. Cortical neurons from the offspring were obtained at gestational day 18. The effects of prenatal stress and KCl stimulations on cellular mortality, autophagy, gene expression, oxidative stress, and inflammation were evaluated. We found that neurons from PNS mice have decreased necrosis and autophagy after depolarization. Moreover, prenatal stress modulated the HPA axis, as observed by the increased GR and decreased 5HTr1 mRNA expression. The BDNF is an important factor for neuronal function and results demonstrated that KCl-induced depolarization increased the gene expression of BDNF I, BDNF IV, and TRκB. Furthermore, prenatal stress and KCl treatment induced significant alterations in oxidative and inflammatory markers. In conclusion, prenatal stress and stimulation with KCl may influence several markers related to neurodevelopment in cortical neurons from neonate mice, supporting the well-known long-term effects of maternal stress.
Collapse
Affiliation(s)
- Carolina Luft
- Laboratory of Pediatric Physical Activity, Infant Center, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil; Laboratory of Cellular Biophysics and Inflammation, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Gabriela Viegas Haute
- Laboratory of Pediatric Physical Activity, Infant Center, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Luís Eduardo Wearick-Silva
- Exercise, Behavior and Cognition Research Group, Psychology Department, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Krist Helen Antunes
- Laboratory of Clinical and Experimental Immunology, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Mariana Severo da Costa
- Laboratory of Pediatric Physical Activity, Infant Center, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil; Laboratory of Cellular Biophysics and Inflammation, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Jarbas Rodrigues de Oliveira
- Laboratory of Cellular Biophysics and Inflammation, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Márcio Vinícius Fagundes Donadio
- Laboratory of Pediatric Physical Activity, Infant Center, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil; Laboratory of Cellular Biophysics and Inflammation, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil.
| |
Collapse
|
47
|
Wang M, Feng LR, Li ZL, Ma KG, Chang KW, Chen XL, Yang PB, Ji SF, Ma YB, Han H, Ruganzua JB, Yang WN, Qian YH. Thymosin β4 reverses phenotypic polarization of glial cells and cognitive impairment via negative regulation of NF-κB signaling axis in APP/PS1 mice. J Neuroinflammation 2021; 18:146. [PMID: 34183019 PMCID: PMC8240373 DOI: 10.1186/s12974-021-02166-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 05/04/2021] [Indexed: 01/15/2023] Open
Abstract
Background Thymosin β4 (Tβ4) is the most abundant member of the β-thymosins and plays an important role in the control of actin polymerization in eukaryotic cells. While its effects in multiple organs and diseases are being widely investigated, the safety profile has been established in animals and humans, currently, little is known about its influence on Alzheimer’s disease (AD) and the possible mechanisms. Thus, we aimed to evaluate the effects and mechanisms of Tβ4 on glial polarization and cognitive performance in APP/PS1 transgenic mice. Methods Behavior tests were conducted to assess the learning and memory, anxiety and depression in APP/PS1 mice. Thioflavin S staining, Nissl staining, immunohistochemistry/immunofluorescence, ELISA, qRT-PCR, and immunoblotting were performed to explore Aβ accumulation, phenotypic polarization of glial cells, neuronal loss and function, and TLR4/NF-κB axis in APP/PS1 mice. Results We demonstrated that Tβ4 protein level elevated in all APP/PS1 mice. Over-expression of Tβ4 alone alleviated AD-like phenotypes of APP/PS1 mice, showed less brain Aβ accumulation and more Insulin-degrading enzyme (IDE), reversed phenotypic polarization of microglia and astrocyte to a healthy state, improved neuronal function and cognitive behavior performance, and accidentally displayed antidepressant-like effect. Besides, Tβ4 could downregulate both TLR4/MyD88/NF-κB p65 and p52-dependent inflammatory pathways in the APP/PS1 mice. While combination drug of TLR4 antagonist TAK242 or NF-κB p65 inhibitor PDTC exerted no further effects. Conclusions These results suggest that Tβ4 may exert its function by regulating both classical and non-canonical NF-κB signaling and is restoring its function as a potential therapeutic target against AD. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-021-02166-3.
Collapse
Affiliation(s)
- Meng Wang
- Department of Human Anatomy and Histology-Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, 710061, Shaanxi, China.,Institute of Neuroscience, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Li-Rong Feng
- Department of Human Anatomy and Histology-Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, 710061, Shaanxi, China.,Institute of Neuroscience, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Zi-Long Li
- Department of Human Anatomy and Histology-Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, 710061, Shaanxi, China.,Institute of Neuroscience, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Kai-Ge Ma
- Institute of Neuroscience, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China.,Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, 710061, Shaanxi, China.,Institute of Neurobiology, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Ke-Wei Chang
- Department of Human Anatomy and Histology-Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, 710061, Shaanxi, China.,Institute of Neuroscience, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China.,Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, 710061, Shaanxi, China
| | - Xin-Lin Chen
- Institute of Neuroscience, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China.,Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, 710061, Shaanxi, China.,Institute of Neurobiology, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Peng-Bo Yang
- Department of Human Anatomy and Histology-Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, 710061, Shaanxi, China.,Institute of Neuroscience, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China.,Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, 710061, Shaanxi, China
| | - Sheng-Feng Ji
- Department of Human Anatomy and Histology-Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, 710061, Shaanxi, China.,Institute of Neuroscience, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China.,Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, 710061, Shaanxi, China
| | - Yan-Bing Ma
- Department of Human Anatomy and Histology-Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, 710061, Shaanxi, China.,Institute of Neuroscience, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China.,Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, 710061, Shaanxi, China
| | - Hua Han
- Department of Human Anatomy and Histology-Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, 710061, Shaanxi, China.,Institute of Neuroscience, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China.,Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, 710061, Shaanxi, China
| | - John Bosco Ruganzua
- Department of Human Anatomy and Histology-Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, 710061, Shaanxi, China.,Institute of Neuroscience, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Wei-Na Yang
- Department of Human Anatomy and Histology-Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, 710061, Shaanxi, China.,Institute of Neuroscience, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China.,Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, 710061, Shaanxi, China
| | - Yi-Hua Qian
- Department of Human Anatomy and Histology-Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, 710061, Shaanxi, China. .,Institute of Neuroscience, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China. .,Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, 710061, Shaanxi, China.
| |
Collapse
|
48
|
Yamada A, Hirasawa T, Nishimura K, Shimura C, Kogo N, Fukuda K, Kato M, Yokomori M, Hayashi T, Umeda M, Yoshimura M, Iwakura Y, Nikaido I, Itohara S, Shinkai Y. Derepression of inflammation-related genes link to microglia activation and neural maturation defect in a mouse model of Kleefstra syndrome. iScience 2021; 24:102741. [PMID: 34258564 PMCID: PMC8258976 DOI: 10.1016/j.isci.2021.102741] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 05/02/2021] [Accepted: 06/15/2021] [Indexed: 11/30/2022] Open
Abstract
Haploinsufficiency of EHMT1, which encodes histone H3 lysine 9 (H3K9) methyltransferase G9a-like protein (GLP), causes Kleefstra syndrome (KS), a complex disorder of developmental delay and intellectual disability. Here, we examined whether postnatal supply of GLP can reverse the neurological phenotypes seen in Ehmt1Δ/+ mice as a KS model. Ubiquitous GLP supply from the juvenile stage ameliorated behavioral abnormalities in Ehmt1Δ/+ mice. Postnatal neuron-specific GLP supply was not sufficient for the improvement of abnormal behaviors but still reversed the reduction of H3K9me2 and spine number in Ehmt1Δ/+ mice. Interestingly, some inflammatory genes, including IL-1β (Il1b), were upregulated and activated microglial cells increased in the Ehmt1Δ/+ brain, and such phenotypes were also reversed by neuron-specific postnatal GLP supply. Il1b inactivation canceled the microglial and spine number phenotypes in the Ehmt1Δ/+ mice. Thus, H3K9me2 and some neurological phenotypes are reversible, but behavioral abnormalities are more difficult to improve depending on the timing of GLP supply. Activated microglias increase in a Ehmt1Δ/+ mouse model of Kleefstra syndrome Diminished H3K9me2 in Ehmt1Δ/+ mouse neurons is reversed by post-natal GLP supply GLP supply from the juvenile stage can improve abnormal behaviors of Ehmt1Δ/+ mice Il1b KO cancelles the microglial and spine number phenotypes in the Ehmt1Δ/+ mice
Collapse
Affiliation(s)
- Ayumi Yamada
- Cellular Memory Laboratory, Cluster for Pioneering Research, RIKEN, Wako, Saitama, Japan
| | - Takae Hirasawa
- Department of Biosciences, School of Science and Engineering, Teikyo University, 1-1 Toyosatodai, Utsunomiya, Tochigi, Japan
| | - Kayako Nishimura
- Cellular Memory Laboratory, Cluster for Pioneering Research, RIKEN, Wako, Saitama, Japan
| | - Chikako Shimura
- Cellular Memory Laboratory, Cluster for Pioneering Research, RIKEN, Wako, Saitama, Japan
| | - Naomi Kogo
- Laboratory for Behavioral Genetics, RIKEN Center for Brain Science, Wako, Saitama, Japan
| | - Kei Fukuda
- Cellular Memory Laboratory, Cluster for Pioneering Research, RIKEN, Wako, Saitama, Japan
| | - Madoka Kato
- Department of Biosciences, School of Science and Engineering, Teikyo University, 1-1 Toyosatodai, Utsunomiya, Tochigi, Japan
| | - Masaki Yokomori
- Department of Biosciences, School of Science and Engineering, Teikyo University, 1-1 Toyosatodai, Utsunomiya, Tochigi, Japan
| | - Tetsutaro Hayashi
- Laboratory for Bioinformatics Research, RIKEN Center for Biosystems Dynamics Research, Wako, Saitama, Japan
| | - Mana Umeda
- Laboratory for Bioinformatics Research, RIKEN Center for Biosystems Dynamics Research, Wako, Saitama, Japan
| | - Mika Yoshimura
- Laboratory for Bioinformatics Research, RIKEN Center for Biosystems Dynamics Research, Wako, Saitama, Japan
| | - Yoichiro Iwakura
- Research Institute for Biomedical Sciences, Tokyo University of Science, Noda, Chiba 278-8510, Japan
| | - Itoshi Nikaido
- Laboratory for Bioinformatics Research, RIKEN Center for Biosystems Dynamics Research, Wako, Saitama, Japan.,Functional Genome Informatics, Medical Research Institute, Tokyo Medical and Dental University, Bunkyo, Tokyo, Japan.,Bioinformatics Course, Master's/Doctoral Program in Life Science Innovation (T-LSI), School of Integrative and Global Majors (SIGMA), University of Tsukuba, Wako, Saitama 351-0198, Japan
| | - Shigeyoshi Itohara
- Laboratory for Behavioral Genetics, RIKEN Center for Brain Science, Wako, Saitama, Japan
| | - Yoichi Shinkai
- Cellular Memory Laboratory, Cluster for Pioneering Research, RIKEN, Wako, Saitama, Japan
| |
Collapse
|
49
|
Microglial metabolism is a pivotal factor in sexual dimorphism in Alzheimer's disease. Commun Biol 2021; 4:711. [PMID: 34112929 PMCID: PMC8192523 DOI: 10.1038/s42003-021-02259-y] [Citation(s) in RCA: 75] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 05/21/2021] [Indexed: 12/20/2022] Open
Abstract
Age and sex are major risk factors in Alzheimer's disease (AD) with a higher incidence of the disease in females. Neuroinflammation, which is a hallmark of AD, contributes to disease pathogenesis and is inexorably linked with inappropriate microglial activation and neurodegeneration. We investigated sex-related differences in microglia in APP/PS1 mice and in post-mortem tissue from AD patients. Changes in genes that are indicative of microglial activation were preferentially increased in cells from female APP/PS1 mice and cells from males and females were morphological, metabolically and functionally distinct. Microglia from female APP/PS1 mice were glycolytic and less phagocytic and associated with increased amyloidosis whereas microglia from males were amoeboid and this was also the case in post-mortem tissue from male AD patients, where plaque load was reduced. We propose that the sex-related differences in microglia are likely to explain, at least in part, the sexual dimorphism in AD.
Collapse
|
50
|
Sanguino‐Gómez J, Buurstede JC, Abiega O, Fitzsimons CP, Lucassen PJ, Eggen BJL, Lesuis SL, Meijer OC, Krugers HJ. An emerging role for microglia in stress‐effects on memory. Eur J Neurosci 2021; 55:2491-2518. [PMID: 33724565 PMCID: PMC9373920 DOI: 10.1111/ejn.15188] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 02/13/2021] [Accepted: 03/03/2021] [Indexed: 12/14/2022]
Abstract
Stressful experiences evoke, among others, a rapid increase in brain (nor)epinephrine (NE) levels and a slower increase in glucocorticoid hormones (GCs) in the brain. Microglia are key regulators of neuronal function and contain receptors for NE and GCs. These brain cells may therefore potentially be involved in modulating stress effects on neuronal function and learning and memory. In this review, we discuss that stress induces (1) an increase in microglial numbers as well as (2) a shift toward a pro‐inflammatory profile. These microglia have (3) impaired crosstalk with neurons and (4) disrupted glutamate signaling. Moreover, microglial immune responses after stress (5) alter the kynurenine pathway through metabolites that impair glutamatergic transmission. All these effects could be involved in the impairments in memory and in synaptic plasticity caused by (prolonged) stress, implicating microglia as a potential novel target in stress‐related memory impairments.
Collapse
Affiliation(s)
| | - Jacobus C. Buurstede
- Department of Medicine Division of Endocrinology Leiden University Medical Center Leiden The Netherlands
| | - Oihane Abiega
- Brain Plasticity Group SILS‐CNS University of Amsterdam Amsterdam The Netherlands
| | - Carlos P. Fitzsimons
- Brain Plasticity Group SILS‐CNS University of Amsterdam Amsterdam The Netherlands
| | - Paul J. Lucassen
- Brain Plasticity Group SILS‐CNS University of Amsterdam Amsterdam The Netherlands
| | - Bart J. L. Eggen
- Department of Biomedical Sciences of Cells & Systems Section Molecular Neurobiology University of Groningen University Medical Center Groningen Groningen The Netherlands
| | - Sylvie L. Lesuis
- Brain Plasticity Group SILS‐CNS University of Amsterdam Amsterdam The Netherlands
- Program in Neurosciences and Mental Health Hospital for Sick Children Toronto ON Canada
| | - Onno C. Meijer
- Department of Medicine Division of Endocrinology Leiden University Medical Center Leiden The Netherlands
| | - Harm J. Krugers
- Brain Plasticity Group SILS‐CNS University of Amsterdam Amsterdam The Netherlands
| |
Collapse
|