1
|
San Felipe D, Martín-Sánchez B, Zekri-Nechar K, Moya M, Llorente R, Zamorano-León JJ, Marco EM, López-Gallardo M. Consequences of Early Maternal Deprivation on Neuroinflammation and Mitochondrial Dynamics in the Central Nervous System of Male and Female Rats. BIOLOGY 2024; 13:1011. [PMID: 39765678 PMCID: PMC11672930 DOI: 10.3390/biology13121011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 12/02/2024] [Accepted: 12/03/2024] [Indexed: 01/11/2025]
Abstract
Early life stress (ELS) is associated with an increased risk for neuropsychiatric disorders, and both neuroinflammation and mitochondrial dysfunction seem to be central to mental health. Herein, using an animal model of ELS, a single episode of maternal deprivation (MD, 24 h on pnd 9) extensively documented to elicit behavioural anomalies in male and female Wistar rats, we investigated its consequences in terms of neuroinflammation and mitochondrial dynamics in the prefrontal cortex (PFC) and the hippocampal formation (HCF). MD differentially affected the brain content of cytokines: MD induced a transient increase in pro-inflammatory cytokines (IL-1β and IL-6) in the PFC, as well as in the levels of the anti-inflammatory cytokine IL-10 in the HCF. MD also induced a significant decrease mitochondria citrate synthase activity, but MD did not exert significant changes in mitochondria Complex IV activity, revealing a generalized decrease in mitochondrial density without any change in mitochondrial respiration. In the present study, we demonstrate that MD induces neuroinflammatory processes in specific brain regions. Additional research is needed to better understand the temporal pattern of such changes, their impact on the developing brain, and their participation in the already well-known behavioural consequences of MD.
Collapse
Affiliation(s)
- Diego San Felipe
- Department of Physiology, School of Medicine, Complutense University of Madrid, Pza. Ramón y Cajal s/n, Ciudad Universitaria, 28040 Madrid, Spain; (D.S.F.); (B.M.-S.); (M.M.); (R.L.); (M.L.-G.)
| | - Beatriz Martín-Sánchez
- Department of Physiology, School of Medicine, Complutense University of Madrid, Pza. Ramón y Cajal s/n, Ciudad Universitaria, 28040 Madrid, Spain; (D.S.F.); (B.M.-S.); (M.M.); (R.L.); (M.L.-G.)
| | - Khaoula Zekri-Nechar
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Department of Public Health and Maternal-Child Health, School of Medicine, Complutense University of Madrid, Pza. Ramón y Cajal s/n, Ciudad Universitaria, 28040 Madrid, Spain;
| | - Marta Moya
- Department of Physiology, School of Medicine, Complutense University of Madrid, Pza. Ramón y Cajal s/n, Ciudad Universitaria, 28040 Madrid, Spain; (D.S.F.); (B.M.-S.); (M.M.); (R.L.); (M.L.-G.)
| | - Ricardo Llorente
- Department of Physiology, School of Medicine, Complutense University of Madrid, Pza. Ramón y Cajal s/n, Ciudad Universitaria, 28040 Madrid, Spain; (D.S.F.); (B.M.-S.); (M.M.); (R.L.); (M.L.-G.)
| | - Jose J. Zamorano-León
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Department of Public Health and Maternal-Child Health, School of Medicine, Complutense University of Madrid, Pza. Ramón y Cajal s/n, Ciudad Universitaria, 28040 Madrid, Spain;
| | - Eva M. Marco
- Department of Genetics, Physiology and Microbiology, Faculty of Biological Sciences, Complutense University of Madrid, C/José Antonio Novais 12, 28040 Madrid, Spain
| | - Meritxell López-Gallardo
- Department of Physiology, School of Medicine, Complutense University of Madrid, Pza. Ramón y Cajal s/n, Ciudad Universitaria, 28040 Madrid, Spain; (D.S.F.); (B.M.-S.); (M.M.); (R.L.); (M.L.-G.)
| |
Collapse
|
2
|
Bertollo AG, Mingoti MED, de Medeiros J, da Silva GB, Capoani GT, Lindemann H, Cassol J, Manica D, de Oliveira T, Garcez ML, Bagatini MD, Bohnen LC, Junior WAR, Ignácio ZM. Hydroalcoholic Extract of Centella asiatica and Madecassic Acid Reverse Depressive-Like Behaviors, Inflammation and Oxidative Stress in Adult Rats Submitted to Stress in Early Life. Mol Neurobiol 2024; 61:10182-10197. [PMID: 38703344 DOI: 10.1007/s12035-024-04198-1] [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: 12/24/2023] [Accepted: 04/22/2024] [Indexed: 05/06/2024]
Abstract
Major depressive disorder (MDD) is a severe disorder that causes enormous loss of quality of life, and among the factors underlying MDD is stress in maternal deprivation (MD). In addition, classic pharmacotherapy has presented severe adverse effects. Centella asiatica (C. asiatica) demonstrates a potential neuroprotective effect but has not yet been evaluated in MD models. This study aimed to evaluate the effect of C. asiatica extract and the active compound madecassic acid on possible depressive-like behavior, inflammation, and oxidative stress in the hippocampus and serum of young rats submitted to MD in the first days of life. Rats (after the first day of birth) were separated from the mother for 3 h a day for 10 days. When adults, these animals were divided into groups and submitted to treatment for 14 days. After subjecting the animals to protocols of locomotor activity in the open field and behavioral despair in the forced swimming test, researchers then euthanized the animals. The hippocampus and serum were collected and analyzed for the inflammatory cytokines and oxidative markers. The C. asiatica extract and active compound reversed or reduced depressive-like behaviors, inflammation in the hippocampus, and oxidative stress in serum and hippocampus. These results suggest that C. asiatica and madecassic acid have potential antidepressant action, at least partially, through anti-inflammatory and antioxidant profiles.
Collapse
Affiliation(s)
- Amanda Gollo Bertollo
- Laboratory of Physiology Pharmacology and Psychopathology, Graduate Program in Biomedical Sciences, Federal University of Fronteira Sul, Chapecó, SC, 89815-899, Brazil
| | - Maiqueli Eduarda Dama Mingoti
- Laboratory of Physiology Pharmacology and Psychopathology, Graduate Program in Biomedical Sciences, Federal University of Fronteira Sul, Chapecó, SC, 89815-899, Brazil
| | - Jesiel de Medeiros
- Laboratory of Physiology Pharmacology and Psychopathology, Graduate Program in Biomedical Sciences, Federal University of Fronteira Sul, Chapecó, SC, 89815-899, Brazil
| | - Gilnei Bruno da Silva
- Multicentric Postgraduate Program in Biochemistry and Molecular Biology, State University of Santa Catarina, Lages, SC, Brazil
| | - Giovana Tamara Capoani
- Laboratory of Pharmacognosy, Community University of Chapecó Region, Unochapecó, Chapecó, SC, Brazil
| | - Heloisa Lindemann
- Laboratory of Pharmacognosy, Community University of Chapecó Region, Unochapecó, Chapecó, SC, Brazil
| | - Joana Cassol
- Laboratory of Physiology Pharmacology and Psychopathology, Graduate Program in Biomedical Sciences, Federal University of Fronteira Sul, Chapecó, SC, 89815-899, Brazil
| | - Daiane Manica
- Laboratory of Physiology Pharmacology and Psychopathology, Graduate Program in Biomedical Sciences, Federal University of Fronteira Sul, Chapecó, SC, 89815-899, Brazil
| | - Tacio de Oliveira
- Laboratory of Physiology Pharmacology and Psychopathology, Graduate Program in Biomedical Sciences, Federal University of Fronteira Sul, Chapecó, SC, 89815-899, Brazil
| | - Michelle Lima Garcez
- Department of Clinical Analysis, Federal University of Santa Catarina, Florianópolis, SC, Brazil
| | - Margarete Dulce Bagatini
- Laboratory of Physiology Pharmacology and Psychopathology, Graduate Program in Biomedical Sciences, Federal University of Fronteira Sul, Chapecó, SC, 89815-899, Brazil
| | - Lilian Caroline Bohnen
- Postgraduate Program in Health Sciences, Community University of Chapecó Region, Unochapecó, Chapecó, SC, Brazil
| | - Walter Antônio Roman Junior
- Postgraduate Program in Health Sciences, Community University of Chapecó Region, Unochapecó, Chapecó, SC, Brazil
| | - Zuleide Maria Ignácio
- Laboratory of Physiology Pharmacology and Psychopathology, Graduate Program in Biomedical Sciences, Federal University of Fronteira Sul, Chapecó, SC, 89815-899, Brazil.
| |
Collapse
|
3
|
She K, Yuan N, Huang M, Zhu W, Tang M, Ma Q, Chen J. Emerging role of microglia in the developing dopaminergic system: perturbation by early life stress. Neural Regen Res 2024; 21:01300535-990000000-00587. [PMID: 39589170 PMCID: PMC12094535 DOI: 10.4103/nrr.nrr-d-24-00742] [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: 08/06/2024] [Revised: 09/13/2024] [Accepted: 10/15/2024] [Indexed: 11/27/2024] Open
Abstract
Early life stress correlates with a higher prevalence of neurological disorders, including autism, attention-deficit/hyperactivity disorder, schizophrenia, depression, and Parkinson's disease. These conditions, primarily involving abnormal development and damage of the dopaminergic system, pose significant public health challenges. Microglia, as the primary immune cells in the brain, are crucial in regulating neuronal circuit development and survival. From the embryonic stage to adulthood, microglia exhibit stage-specific gene expression profiles, transcriptome characteristics, and functional phenotypes, enhancing the susceptibility to early life stress. However, the role of microglia in mediating dopaminergic system disorders under early life stress conditions remains poorly understood. This review presents an up-to-date overview of preclinical studies elucidating the impact of early life stress on microglia, leading to dopaminergic system disorders, along with the underlying mechanisms and therapeutic potential for neurodegenerative and neurodevelopmental conditions. Impaired microglial activity damages dopaminergic neurons by diminishing neurotrophic support (e.g., insulin-like growth factor-1) and hinders dopaminergic axon growth through defective phagocytosis and synaptic pruning. Furthermore, blunted microglial immunoreactivity suppresses striatal dopaminergic circuit development and reduces neuronal transmission. Furthermore, inflammation and oxidative stress induced by activated microglia can directly damage dopaminergic neurons, inhibiting dopamine synthesis, reuptake, and receptor activity. Enhanced microglial phagocytosis inhibits dopamine axon extension. These long-lasting effects of microglial perturbations may be driven by early life stress-induced epigenetic reprogramming of microglia. Indirectly, early life stress may influence microglial function through various pathways, such as astrocytic activation, the hypothalamic-pituitary-adrenal axis, the gut-brain axis, and maternal immune signaling. Finally, various therapeutic strategies and molecular mechanisms for targeting microglia to restore the dopaminergic system were summarized and discussed. These strategies include classical antidepressants and antipsychotics, antibiotics and anti-inflammatory agents, and herbal-derived medicine. Further investigations combining pharmacological interventions and genetic strategies are essential to elucidate the causal role of microglial phenotypic and functional perturbations in the dopaminergic system disrupted by early life stress.
Collapse
Affiliation(s)
- Kaijie She
- Guangzhou Key Laboratory of Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, Guangdong Province, China
| | - Naijun Yuan
- Guangzhou Key Laboratory of Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, Guangdong Province, China
- Shenzhen People’s Hospital, The 2 Clinical Medical College, Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Shenzhen, Guangdong Province, China
| | - Minyi Huang
- Guangzhou Key Laboratory of Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, Guangdong Province, China
| | - Wenjun Zhu
- Guangzhou Key Laboratory of Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, Guangdong Province, China
| | - Manshi Tang
- Guangzhou Key Laboratory of Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, Guangdong Province, China
| | - Qingyu Ma
- Guangzhou Key Laboratory of Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, Guangdong Province, China
| | - Jiaxu Chen
- Guangzhou Key Laboratory of Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, Guangdong Province, China
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
4
|
Smail MA, Lenz KM. Developmental functions of microglia: Impact of psychosocial and physiological early life stress. Neuropharmacology 2024; 258:110084. [PMID: 39025401 PMCID: PMC12051134 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
|
Baram TZ, Birnie MT. Enduring memory consequences of early-life stress / adversity: Structural, synaptic, molecular and epigenetic mechanisms. Neurobiol Stress 2024; 33:100669. [PMID: 39309367 PMCID: PMC11415888 DOI: 10.1016/j.ynstr.2024.100669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 08/13/2024] [Accepted: 08/28/2024] [Indexed: 09/25/2024] Open
Abstract
Adverse early life experiences are strongly associated with reduced cognitive function throughout life. The link is strong in many human studies, but these do not enable assigning causality, and the limited access to the live human brain can impede establishing the mechanisms by which early-life adversity (ELA) may induce cognitive problems. In experimental models, artificially imposed chronic ELA/stress results in deficits in hippocampus dependent memory as well as increased vulnerability to the deleterious effects of adult stress on memory. This causal relation of ELA and life-long memory impairments provides a framework to probe the mechanisms by which ELA may lead to human cognitive problems. Here we focus on the consequences of a one-week exposure to adversity during early postnatal life in the rodent, the spectrum of the ensuing memory deficits, and the mechanisms responsible. We highlight molecular, cellular and circuit mechanisms using convergent trans-disciplinary approaches aiming to enable translation of the discoveries in experimental models to the clinic.
Collapse
Affiliation(s)
- Tallie Z. Baram
- Department of Pediatrics, University of California-Irvine, Irvine, CA, USA
- Department of Anatomy/Neurobiology, University of California-Irvine, Irvine, CA, USA
- Department of Neurology, University of California-Irvine, Irvine, CA, USA
| | - Matthew T. Birnie
- Department of Pediatrics, University of California-Irvine, Irvine, CA, USA
| |
Collapse
|
6
|
Gonzales J, Dharshika C, Mazhar K, Morales-Soto W, McClain JL, Moeser AJ, Nault R, Price TJ, Gulbransen BD. Early life adversity promotes gastrointestinal dysfunction through a sex-dependent phenotypic switch in enteric glia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.31.596805. [PMID: 38895433 PMCID: PMC11185517 DOI: 10.1101/2024.05.31.596805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Irritable bowel syndrome and related disorders of gut-brain interaction (DGBI) are common and exhibit a complex, poorly understood etiology that manifests as abnormal gut motility and pain. Risk factors such as biological sex, stressors during critical periods, and inflammation are thought to influence DGBI vulnerability by reprogramming gut-brain circuits, but the specific cells affected are unclear. Here, we used a model of early life stress to understand cellular mechanisms in the gut that produce DGBIs. Our findings identify enteric glia as a key cellular substrate in which stress and biological sex converge to dictate DGBI susceptibility. Enteric glia exhibit sexual dimorphism in genes and functions related to cellular communication, inflammation, and disease susceptibility. Experiencing early life stress has sex-specific effects on enteric glia that cause a phenotypic switch in male glia toward a phenotype normally observed in females. This phenotypic transformation is followed by physiological changes in the gut, mirroring those observed in DGBI in humans. These effects are mediated, in part, by alterations to glial prostaglandin and endocannabinoid signaling. Together, these data identify enteric glia as a cellular integration site through which DGBI risk factors produce changes in gut physiology and suggest that manipulating glial signaling may represent an attractive target for sex-specific therapeutic strategies in DGBIs.
Collapse
|
7
|
Andersen SL. Increasing CB2 Receptor Activity after Early Life Stress Prevents Depressive Behavior in Female Rats. Biomolecules 2024; 14:464. [PMID: 38672480 PMCID: PMC11047932 DOI: 10.3390/biom14040464] [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: 01/10/2024] [Revised: 02/28/2024] [Accepted: 04/07/2024] [Indexed: 04/28/2024] Open
Abstract
Early adversity, the loss of the inhibitory GABAergic interneuron parvalbumin, and elevated neuroinflammation are associated with depression. Individuals with a maltreatment history initiate medicinal cannabis use earlier in life than non-maltreated individuals, suggesting self-medication. Female rats underwent maternal separation (MS) between 2 and 20 days of age to model early adversity or served as colony controls. The prelimbic cortex and behavior were examined to determine whether MS alters the cannabinoid receptor 2 (CB2), which has anti-inflammatory properties. A reduction in the CB2-associated regulatory enzyme MARCH7 leading to increased NLRP3 was observed with Western immunoblots in MS females. Immunohistochemistry with stereology quantified numbers of parvalbumin-immunoreactive cells and CB2 at 25, 40, and 100 days of age, revealing that the CB2 receptor associated with PV neurons initially increases at P25 and subsequently decreases by P40 in MS animals, with no change in controls. Confocal and triple-label microscopy suggest colocalization of these CB2 receptors to microglia wrapped around the parvalbumin neuron. Depressive-like behavior in MS animals was elevated at P40 and reduced with the CB2 agonist HU-308 or a CB2-overexpressing lentivirus microinjected into the prelimbic cortex. These results suggest that increasing CB2 expression by P40 in the prelimbic cortex prevents depressive behavior in MS female rats.
Collapse
Affiliation(s)
- Susan L Andersen
- Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
8
|
Kouba BR, de Araujo Borba L, Borges de Souza P, Gil-Mohapel J, Rodrigues ALS. Role of Inflammatory Mechanisms in Major Depressive Disorder: From Etiology to Potential Pharmacological Targets. Cells 2024; 13:423. [PMID: 38474387 PMCID: PMC10931285 DOI: 10.3390/cells13050423] [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: 01/18/2024] [Revised: 02/20/2024] [Accepted: 02/26/2024] [Indexed: 03/14/2024] Open
Abstract
The involvement of central and peripheral inflammation in the pathogenesis and prognosis of major depressive disorder (MDD) has been demonstrated. The increase of pro-inflammatory cytokines (interleukin (IL)-1β, IL-6, IL-18, and TNF-α) in individuals with depression may elicit neuroinflammatory processes and peripheral inflammation, mechanisms that, in turn, can contribute to gut microbiota dysbiosis. Together, neuroinflammation and gut dysbiosis induce alterations in tryptophan metabolism, culminating in decreased serotonin synthesis, impairments in neuroplasticity-related mechanisms, and glutamate-mediated excitotoxicity. This review aims to highlight the inflammatory mechanisms (neuroinflammation, peripheral inflammation, and gut dysbiosis) involved in the pathophysiology of MDD and to explore novel anti-inflammatory therapeutic approaches for this psychiatric disturbance. Several lines of evidence have indicated that in addition to antidepressants, physical exercise, probiotics, and nutraceuticals (agmatine, ascorbic acid, and vitamin D) possess anti-inflammatory effects that may contribute to their antidepressant properties. Further studies are necessary to explore the therapeutic benefits of these alternative therapies for MDD.
Collapse
Affiliation(s)
- Bruna R. Kouba
- Department of Biochemistry, Center of Biological Sciences, Universidade Federal de Santa Catarina, Florianópolis 88040-900, SC, Brazil; (B.R.K.); (L.d.A.B.); (P.B.d.S.)
| | - Laura de Araujo Borba
- Department of Biochemistry, Center of Biological Sciences, Universidade Federal de Santa Catarina, Florianópolis 88040-900, SC, Brazil; (B.R.K.); (L.d.A.B.); (P.B.d.S.)
| | - Pedro Borges de Souza
- Department of Biochemistry, Center of Biological Sciences, Universidade Federal de Santa Catarina, Florianópolis 88040-900, SC, Brazil; (B.R.K.); (L.d.A.B.); (P.B.d.S.)
| | - Joana Gil-Mohapel
- Island Medical Program, Faculty of Medicine, University of British Columbia, Victoria, BC V8P 5C2, Canada
- Division of Medical Sciences, University of Victoria, Victoria, BC V8P 5C2, Canada
| | - Ana Lúcia S. Rodrigues
- Department of Biochemistry, Center of Biological Sciences, Universidade Federal de Santa Catarina, Florianópolis 88040-900, SC, Brazil; (B.R.K.); (L.d.A.B.); (P.B.d.S.)
| |
Collapse
|
9
|
De Santa F, Strimpakos G, Marchetti N, Gargari G, Torcinaro A, Arioli S, Mora D, Petrella C, Farioli-Vecchioli S. Effect of a multi-strain probiotic mixture consumption on anxiety and depression symptoms induced in adult mice by postnatal maternal separation. MICROBIOME 2024; 12:29. [PMID: 38369490 PMCID: PMC10875865 DOI: 10.1186/s40168-024-01752-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 01/04/2024] [Indexed: 02/20/2024]
Abstract
BACKGROUND Intestinal microbial composition not only affects the health of the gut but also influences centrally mediated systems involved in mood, through the "gut-brain" axis, a bidirectional communication between gut microbiota and the brain. In this context, the modulation of intestinal microbiota and its metabolites through the administration of probiotics seems to represent a very promising approach in the treatment of the central nervous system alterations. Early postnatal life is a critical period during which the brain undergoes profound and essential modulations in terms of maturation and plasticity. Maternal separation (MS), i.e., the disruption of the mother-pup interaction, represents a pivotal paradigm in the study of stress-related mood disorders, by inducing persistent changes in the immune system, inflammatory processes, and emotional behavior in adult mammals. RESULTS We conducted experiments to investigate whether sustained consumption of a multi-strain probiotic formulation by adult male mice could mitigate the effects of maternal separation. Our data demonstrated that the treatment with probiotics was able to totally reverse the anxiety- and depressive-like behavior; normalize the neuro-inflammatory state, by restoring the resting state of microglia; and finally induce a proneurogenic effect. Mice subjected to maternal separation showed changes in microbiota composition compared to the control group that resulted in permissive colonization by the administered multi-strain probiotic product. As a consequence, the probiotic treatment also significantly affected the production of SCFA and in particular the level of butyrate. CONCLUSION Gut microbiota and its metabolites mediate the therapeutic action of the probiotic mix on MS-induced brain dysfunctions. Our findings extend the knowledge on the use of probiotics as a therapeutic tool in the presence of alterations of the emotional sphere that significantly impact on gut microbiota composition. Video Abstract.
Collapse
Affiliation(s)
- Francesca De Santa
- Institute of Biochemistry and Cell Biology, IBBC, CNR, Via E. Ramarini, 32, Monterotondo, Rome, 00015, Italy
| | - Georgios Strimpakos
- Institute of Biochemistry and Cell Biology, IBBC, CNR, Via E. Ramarini, 32, Monterotondo, Rome, 00015, Italy
| | - Nicole Marchetti
- Institute of Biochemistry and Cell Biology, IBBC, CNR, Via E. Ramarini, 32, Monterotondo, Rome, 00015, Italy
- Sciences of Nutrition, Aging, Metabolism and Gender Pathologies, Catholic University of Roma, Rome, 00100, Italy
| | - Giorgio Gargari
- Department of Food Environmental and Nutritional Sciences (DeFENS), University of Milan, Milan, Italy
| | - Alessio Torcinaro
- Institute of Biochemistry and Cell Biology, IBBC, CNR, Via E. Ramarini, 32, Monterotondo, Rome, 00015, Italy
| | - Stefania Arioli
- Department of Food Environmental and Nutritional Sciences (DeFENS), University of Milan, Milan, Italy
| | - Diego Mora
- Department of Food Environmental and Nutritional Sciences (DeFENS), University of Milan, Milan, Italy
| | - Carla Petrella
- Institute of Biochemistry and Cell Biology, IBBC, CNR, Policlinico Umberto I, Rome, Italy
| | - Stefano Farioli-Vecchioli
- Institute of Biochemistry and Cell Biology, IBBC, CNR, Via E. Ramarini, 32, Monterotondo, Rome, 00015, Italy.
| |
Collapse
|
10
|
Shukla P, Akotkar L, Aswar U. Resveratrol attenuates early life stress induced depression in rats: Behavioural and neurochemical evidence. Neurosci Lett 2024; 820:137606. [PMID: 38110147 DOI: 10.1016/j.neulet.2023.137606] [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: 08/23/2023] [Revised: 11/27/2023] [Accepted: 12/15/2023] [Indexed: 12/20/2023]
Abstract
PURPOSE Maternal deprivation (MD), a severe naturalistic type of stress in the early postnatal days, is a well-established model of early life stress (ELS) that models juvenile adversity and may result in significant depressive disease in adults. In order to analyze the behavioural, brain monoamine level and HPA axis dysregulations caused by ELS and to determine whether Resveratrol (Res) could counteract these effects, Wistar rat pups were subjected to the MD paradigm, which simulated the consequences of depression. METHODS The pups on their postnatal day 1-10 were divided in 5 groups (n = 8); nondeprived (ND), maternally deprived (DC), standard fluoxetine (FLX) (5 mg/kg i.p), Res (20, 40 mg/kg i.p). Excluding the ND group, other pups were separated from dam for 3hr/day from day 1 to 10th day. Treatment was initiated from 50th day and was given for 12 days. The behaviour parameters light/dark test, sucrose preference, and resident intruder test were employed. Serum cortisol levels, brain antioxidant activity, monoamine levels and neuronal morphology in the hippocampus were assessed. RESULTS The MD rats showed altered behaviour, including more light-dark transitions, less desire for sucrose, and lower attack latencies. MD influenced the release of serum cortisol and interfered with monoamine, antioxidant levels as well as reduced Nissl bodies in the hippocampus. Treatment with Res led to improved behavioural functions also restored monoamine levels, reduced cortisol release, oxidative stress and prevented histopathological alterations in the rat hippocampus. CONCLUSION Res showed neuroprotective effects by improving the brain antioxidants and monoamine levels and HPA axis dysregulation and thus improves MD induced depression like behaviour in Wistar rats.
Collapse
Affiliation(s)
- Pavan Shukla
- Department of Pharmacology, Poona College of Pharmacy, Bharati Vidyapeeth (Deemed to be University), Pune, Maharashtra, India
| | - Likhit Akotkar
- Department of Pharmacology, Poona College of Pharmacy, Bharati Vidyapeeth (Deemed to be University), Pune, Maharashtra, India
| | - Urmila Aswar
- Department of Pharmacology, Poona College of Pharmacy, Bharati Vidyapeeth (Deemed to be University), Pune, Maharashtra, India.
| |
Collapse
|
11
|
Rahmatinia M, Mohseni-Bandpei A, Khodagholi F, Abdollahifar MA, Amouei Torkmahalleh M, Hassani Moghaddam M, Hopke PK, Ghavimehr E, Bazzazpour S, Shahsavani A. Exposure to different PM 2.5 extracts induces gliosis and changes behavior in male rats similar to autism spectrum disorders features. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 340:122804. [PMID: 37907193 DOI: 10.1016/j.envpol.2023.122804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 10/20/2023] [Accepted: 10/23/2023] [Indexed: 11/02/2023]
Abstract
Epidemiological studies have documented that exposure to fine particulate matter (PM2.5) could affect neurodevelopment, thereby leading to autism spectrum disorders (ASD). Nevertheless, there is little laboratory data to support this epidemiological evidence. In the current study, we carried out a series of experiments to assess whether developmental exposures to different extracts of PM2.5 can result in ASD-like behavioral, biochemical, and immunohistochemical characteristics in male rat offspring. PM2.5 samples were collected daily for a year, and monthly composites were extracted with an acetone-hexane mixture. The extracts were analyzed for their chemical constituents. Three groups of rats were exposed to the different PM2.5 extracts during pre- and postnatal periods. All exposed groups of rats exhibited typical behavioral features of ASD, including increased repetitive and depression-related behaviors. We also found microglia and astrocytes activation and decreased concentrations of oxytocin (OXT) in the brain regions of exposed rats compared with control rats. Comparing the current results with a prior study, the induced biological effects followed a sequence of whole particles of PM2.5 > organic extract > inorganic extract. These findings indicated that exposure to PM2.5 can elicit ASD-like features in rats and raise concerns about particulate matter as a possible trigger for the induction of ASD in humans; therefore, mitigating the contents of the PAHs and metals could reduce the PM2.5 neurotoxicity.
Collapse
Affiliation(s)
- Masoumeh Rahmatinia
- Department of Environmental Health Engineering, School of Public Health and Safety, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Air Quality and Climate Change Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Anoushiravan Mohseni-Bandpei
- Department of Environmental Health Engineering, School of Public Health and Safety, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Air Quality and Climate Change Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fariba Khodagholi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad-Amin Abdollahifar
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mehdi Amouei Torkmahalleh
- Division of Environmental and Occupational Health Sciences, School of Public Health, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Meysam Hassani Moghaddam
- Department of Anatomical Sciences, Faculty of Medicine, AJA University of Medical Sciences, Tehran, Iran
| | - Philip K Hopke
- Department of Public Health Sciences, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - Ehsan Ghavimehr
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Shahriyar Bazzazpour
- Department of Environmental Health Engineering, School of Public Health and Safety, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abbas Shahsavani
- Department of Environmental Health Engineering, School of Public Health and Safety, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Air Quality and Climate Change Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
12
|
Gore IR, Gould E. Developmental and adult stress: effects of steroids and neurosteroids. Stress 2024; 27:2317856. [PMID: 38563163 PMCID: PMC11046567 DOI: 10.1080/10253890.2024.2317856] [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: 07/03/2023] [Accepted: 02/03/2024] [Indexed: 04/04/2024] Open
Abstract
In humans, exposure to early life adversity has profound implications for susceptibility to developing neuropsychiatric disorders later in life. Studies in rodents have shown that stress experienced during early postnatal life can have lasting effects on brain development. Glucocorticoids and sex steroids are produced in endocrine glands and the brain from cholesterol; these molecules bind to nuclear and membrane-associated steroid receptors. Unlike other steroids that can also be made in the brain, neurosteroids bind specifically to neurotransmitter receptors, not steroid receptors. The relationships among steroids, neurosteroids, and stress are multifaceted and not yet fully understood. However, studies demonstrating altered levels of progestogens, androgens, estrogens, glucocorticoids, and their neuroactive metabolites in both developmental and adult stress paradigms strongly suggest that these molecules may be important players in stress effects on brain circuits and behavior. In this review, we discuss the influence of developmental and adult stress on various components of the brain, including neurons, glia, and perineuronal nets, with a focus on sex steroids and neurosteroids. Gaining an enhanced understanding of how early adversity impacts the intricate systems of brain steroid and neurosteroid regulation could prove instrumental in identifying novel therapeutic targets for stress-related conditions.
Collapse
Affiliation(s)
- Isha R Gore
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, USA
| | - Elizabeth Gould
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, USA
| |
Collapse
|
13
|
Li ZA, Cai Y, Taylor RL, Eisenstein SA, Barch DM, Marek S, Hershey T. Associations Between Socioeconomic Status, Obesity, Cognition, and White Matter Microstructure in Children. JAMA Netw Open 2023; 6:e2320276. [PMID: 37368403 DOI: 10.1001/jamanetworkopen.2023.20276] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/28/2023] Open
Abstract
Importance Lower neighborhood and household socioeconomic status (SES) are associated with negative health outcomes and altered brain structure in children. It is unclear whether such findings extend to white matter and via what mechanisms. Objective To assess whether and how neighborhood and household SES are independently associated with children's white matter microstructure and examine whether obesity and cognitive performance (reflecting environmental cognitive and sensory stimulation) are plausible mediators. Design, Setting, and Participants This cross-sectional study used baseline data from participants in the Adolescent Brain Cognitive Development (ABCD) study. Data were collected at 21 US sites, and school-based recruitment was used to represent the US population. Children aged 9 to 11 years and their parents or caregivers completed assessments between October 1, 2016, and October 31, 2018. After exclusions, 8842 of 11 875 children in the ABCD study were included in the analyses. Data analysis was conducted from July 11 to December 19, 2022. Exposures Neighborhood disadvantage was derived from area deprivation indices at participants' primary residence. Household SES factors were total income and highest parental educational attainment. Main Outcomes and Measures A restriction spectrum imaging (RSI) model was used to quantify restricted normalized directional (RND; reflecting oriented myelin organization) and restricted normalized isotropic (RNI; reflecting glial and neuronal cell bodies) diffusion in 31 major white matter tracts. The RSI measurements were scanner harmonized. Obesity was assessed through body mass index (BMI; calculated as weight in kilograms divided by height in meters squared), age- and sex-adjusted BMI z scores, and waist circumference, and cognition was assessed through the National Institutes of Health Toolbox Cognition Battery. Analyses were adjusted for age, sex, pubertal development stage, intracranial volume, mean head motion, and twin or siblingship. Results Among 8842 children, 4543 (51.4%) were boys, and the mean (SD) age was 9.9 (0.7) years. Linear mixed-effects models revealed that greater neighborhood disadvantage was associated with lower RSI-RND in the left superior longitudinal fasciculus (β = -0.055; 95% CI, -0.081 to -0.028) and forceps major (β = -0.040; 95% CI, -0.067 to -0.013). Lower parental educational attainment was associated with lower RSI-RND in the bilateral superior longitudinal fasciculus (eg, right hemisphere: β = 0.053; 95% CI, 0.025-0.080) and bilateral corticospinal or pyramidal tract (eg, right hemisphere: β = 0.042; 95% CI, 0.015-0.069). Structural equation models revealed that lower cognitive performance (eg, lower total cognition score and higher neighborhood disadvantage: β = -0.012; 95% CI, -0.016 to -0.009) and greater obesity (eg, higher BMI and higher neighborhood disadvantage: β = -0.004; 95% CI, -0.006 to -0.001) partially accounted for the associations between SES and RSI-RND. Lower household income was associated with higher RSI-RNI in most tracts (eg, right inferior longitudinal fasciculus: β = -0.042 [95% CI, -0.073 to -0.012]; right anterior thalamic radiations: β = -0.045 [95% CI, -0.075 to -0.014]), and greater neighborhood disadvantage had similar associations in primarily frontolimbic tracts (eg, right fornix: β = 0.046 [95% CI, 0.019-0.074]; right anterior thalamic radiations: β = 0.045 [95% CI, 0.018-0.072]). Lower parental educational attainment was associated with higher RSI-RNI in the forceps major (β = -0.048; 95% CI, -0.077 to -0.020). Greater obesity partially accounted for these SES associations with RSI-RNI (eg, higher BMI and higher neighborhood disadvantage: β = 0.015; 95% CI, 0.011-0.020). Findings were robust in sensitivity analyses and were corroborated using diffusion tensor imaging. Conclusions and Relevance In this cross-sectional study, both neighborhood and household contexts were associated with white matter development in children, and findings suggested that obesity and cognitive performance were possible mediators in these associations. Future research on children's brain health may benefit from considering these factors from multiple socioeconomic perspectives.
Collapse
Affiliation(s)
- Zhaolong Adrian Li
- Department of Psychiatry, Washington University in St Louis School of Medicine, St Louis, Missouri
- Department of Psychological and Brain Sciences, Washington University in St Louis, St Louis, Missouri
| | - Yuqi Cai
- Department of Psychological and Brain Sciences, Washington University in St Louis, St Louis, Missouri
- Now with Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Rita L Taylor
- Department of Psychological and Brain Sciences, Washington University in St Louis, St Louis, Missouri
| | - Sarah A Eisenstein
- Department of Psychiatry, Washington University in St Louis School of Medicine, St Louis, Missouri
- Mallinckrodt Institute of Radiology, Washington University in St Louis School of Medicine, St Louis, Missouri
| | - Deanna M Barch
- Department of Psychiatry, Washington University in St Louis School of Medicine, St Louis, Missouri
- Department of Psychological and Brain Sciences, Washington University in St Louis, St Louis, Missouri
- Mallinckrodt Institute of Radiology, Washington University in St Louis School of Medicine, St Louis, Missouri
| | - Scott Marek
- Department of Psychiatry, Washington University in St Louis School of Medicine, St Louis, Missouri
- Mallinckrodt Institute of Radiology, Washington University in St Louis School of Medicine, St Louis, Missouri
| | - Tamara Hershey
- Department of Psychiatry, Washington University in St Louis School of Medicine, St Louis, Missouri
- Department of Psychological and Brain Sciences, Washington University in St Louis, St Louis, Missouri
- Mallinckrodt Institute of Radiology, Washington University in St Louis School of Medicine, St Louis, Missouri
- Department of Neurology, Washington University in St Louis School of Medicine, St Louis, Missouri
| |
Collapse
|
14
|
Warhaftig G, Almeida D, Turecki G. Early life adversity across different cell- types in the brain. Neurosci Biobehav Rev 2023; 148:105113. [PMID: 36863603 DOI: 10.1016/j.neubiorev.2023.105113] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 02/13/2023] [Accepted: 02/24/2023] [Indexed: 03/04/2023]
Abstract
Early life adversity (ELA)- which includes physical, psychological, emotional, and sexual abuse is one of the most common predictors to diverse psychopathologies later in adulthood. As ELA has a lasting impact on the brain at a developmental stage, recent findings from the field highlighted the specific contributions of different cell types to ELA and their association with long lasting consequences. In this review we will gather recent findings describing morphological, transcriptional and epigenetic alterations within neurons, glia and perineuronal nets and their associated cellular subpopulation. The findings reviewed and summarized here highlight important mechanisms underlying ELA and point to therapeutic approaches for ELA and related psychopathologies later in life.
Collapse
Affiliation(s)
- Gal Warhaftig
- McGill Group for Suicide Studies, Douglas Hospital Research Center, Montreal QC H4H 1R3, Canada
| | - Daniel Almeida
- McGill Group for Suicide Studies, Douglas Hospital Research Center, Montreal QC H4H 1R3, Canada
| | - Gustavo Turecki
- McGill Group for Suicide Studies, Douglas Hospital Research Center, Montreal QC H4H 1R3, Canada; Department of Psychiatry, McGill University, Montreal QC H3A 1A1, Canada.
| |
Collapse
|
15
|
Li ZA, Cai Y, Taylor RL, Eisenstein SA, Barch DM, Marek S, Hershey T. Associations between socioeconomic status and white matter microstructure in children: indirect effects via obesity and cognition. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.02.09.23285150. [PMID: 36798149 PMCID: PMC9934783 DOI: 10.1101/2023.02.09.23285150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/12/2023]
Abstract
Importance Both neighborhood and household socioeconomic disadvantage relate to negative health outcomes and altered brain structure in children. It is unclear whether such findings extend to white matter development, and via what mechanisms socioeconomic status (SES) influences the brain. Objective To test independent associations between neighborhood and household SES indicators and white matter microstructure in children, and examine whether body mass index and cognitive function (a proxy of environmental cognitive/sensory stimulation) may plausibly mediate these associations. Design This cross-sectional study used baseline data from the Adolescent Brain Cognitive Development (ABCD) Study, an ongoing 10-year cohort study tracking child health. Setting School-based recruitment at 21 U.S. sites. Participants Children aged 9 to 11 years and their parents/caregivers completed baseline assessments between October 1 st , 2016 and October 31 st , 2018. Data analysis was conducted from July to December 2022. Exposures Neighborhood disadvantage was derived from area deprivation indices at primary residence. Household SES indicators were total income and the highest parental education attainment. Main Outcomes and Measures Thirty-one major white matter tracts were segmented from diffusion-weighted images. The Restriction Spectrum Imaging (RSI) model was implemented to measure restricted normalized directional (RND; reflecting oriented myelin organization) and isotropic (RNI; reflecting glial/neuronal cell bodies) diffusion in each tract. Obesity-related measures were body mass index (BMI), BMI z -scores, and waist circumference, and cognitive performance was assessed using the NIH Toolbox Cognition Battery. Linear mixed-effects models tested the associations between SES indicators and scanner-harmonized RSI metrics. Structural equation models examined indirect effects of obesity and cognitive performance in the significant associations between SES and white mater microstructure summary principal components. Analyses were adjusted for age, sex, pubertal development stage, intracranial volume, and head motion. Results The analytical sample included 8842 children (4299 [48.6%] girls; mean age [SD], 9.9 [0.7] years). Greater neighborhood disadvantage and lower parental education were independently associated with lower RSI-RND in forceps major and corticospinal/pyramidal tracts, and had overlapping associations in the superior longitudinal fasciculus. Lower cognition scores and greater obesity-related measures partially accounted for these SES associations with RSI-RND. Lower household income was related to higher RSI-RNI in almost every tract, and greater neighborhood disadvantage had similar effects in primarily frontolimbic tracts. Lower parental education was uniquely linked to higher RSI-RNI in forceps major. Greater obesity-related measures partially accounted for these SES associations with RSI-RNI. Findings were robust in sensitivity analyses and mostly corroborated using traditional diffusion tensor imaging (DTI). Conclusions and Relevance These cross-sectional results demonstrate that both neighborhood and household contexts are relevant to white matter development in children, and suggest cognitive performance and obesity as possible pathways of influence. Interventions targeting obesity reduction and improving cognition from multiple socioeconomic angles may ameliorate brain health in low-SES children. Key Points Question: Are neighborhood and household socioeconomic levels associated with children’s brain white matter microstructure, and if so, do obesity and cognitive performance (reflecting environmental stimulation) mediate the associations?Findings: In a cohort of 8842 children, higher neighborhood disadvantage, lower household income, and lower parental education had independent and overlapping associations with lower restricted directional diffusion and greater restricted isotropic diffusion in white matter. Greater body mass index and poorer cognitive performance partially mediated these associations.Meaning: Both neighborhood and household poverty may contribute to altered white matter development in children. These effects may be partially explained by obesity incidence and poorer cognitive performance.
Collapse
Affiliation(s)
- Zhaolong Adrian Li
- Department of Psychiatry, Washington University in St. Louis School of Medicine, St. Louis, MO 63130, USA
- Department of Psychological & Brain Sciences, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Yuqi Cai
- Department of Psychological & Brain Sciences, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Rita L. Taylor
- Department of Psychological & Brain Sciences, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Sarah A. Eisenstein
- Department of Psychiatry, Washington University in St. Louis School of Medicine, St. Louis, MO 63130, USA
- Mallinckrodt Institute of Radiology, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
| | - Deanna M. Barch
- Department of Psychiatry, Washington University in St. Louis School of Medicine, St. Louis, MO 63130, USA
- Department of Psychological & Brain Sciences, Washington University in St. Louis, St. Louis, MO 63130, USA
- Mallinckrodt Institute of Radiology, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
| | - Scott Marek
- Mallinckrodt Institute of Radiology, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
| | - Tamara Hershey
- Department of Psychiatry, Washington University in St. Louis School of Medicine, St. Louis, MO 63130, USA
- Department of Psychological & Brain Sciences, Washington University in St. Louis, St. Louis, MO 63130, USA
- Mallinckrodt Institute of Radiology, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
- Department of Neurology, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
16
|
Sex-specific effects of neonatal paternal deprivation on microglial cell density in adult California mouse (Peromyscus californicus) dentate gyrus. Brain Behav Immun 2022; 106:1-10. [PMID: 35908654 DOI: 10.1016/j.bbi.2022.07.161] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 07/20/2022] [Accepted: 07/26/2022] [Indexed: 11/20/2022] Open
Abstract
Adverse early-life experiences are risk factors for psychiatric disease development, resulting in stress-related neuronal modeling and neurobehavioral changes. Stressful experiences modulate the immune system, contributing to neuronal damage in higher cortical regions, like the hippocampus. Moreover, early-life stressors dysregulate the function of microglia, the resident immune cells of the brain, in the developing hippocampus. Paternal deprivation, an early-life stressor in many biparental species, facilitates sex-dependent inhibitions in hippocampal plasticity, but parental contributors to these sex-specific outcomes are unknown. Also, neurobiological mechanisms contributing to impairments in hippocampal neuroplasticity are less known. Thus, our goals were to 1) determine whether parental behavior is altered in maternal females following removal of the paternal male, 2) assess the effects of paternal deprivation on dentate gyrus (DG) volume and microglia proliferation, and 3) determine if early-life experimental handling mitigates sex-specific reductions in DG cell survival. California mice were born to multiparous breeders and reared by both parents (biparental care) or by their mother alone (i.e., father removed on postnatal day 1; paternal deprivation). One cohort of offspring underwent offspring retrieval tests for eight days beginning on postnatal day 2. On PND 68, these offspring (and a second cohort of mice without behavioral testing) were euthanized and brains visualized for bromodeoxyuridine (BrdU) and neuron-specific class III beta-tubulin (TuJ-1) or ionized calcium binding adaptor molecule 1 (Iba1). While mate absence did not impair maternal retrieval, paternal deprivation reduced DG volume, but Iba1+ cell density was only higher in paternally-deprived females. Neither sex or paternal deprivation significantly altered the number of BrdU+ or Tuj1+ cells in the DG - an absence of a reduction in cell survival may be related to daily handing during early offspring retrieval tests. Together, these data suggest that paternal deprivation impairs hippocampal plasticity; however, sex and early environment may influence the magnitude of these outcomes.
Collapse
|
17
|
Carlessi AS, Botelho MEM, Manosso LM, Borba LA, Maciel LR, Andrade NM, Martinello NS, Padilha APZ, Generoso CM, Bencke CV, de Moura AB, Lodetti BF, Collodel A, Joaquim L, Bonfante S, Biehl E, Generoso JS, Arent CO, Barichello T, Petronilho F, Quevedo J, Réus GZ. Sex differences on the response to antidepressants and psychobiotics following early life stress in rats. Pharmacol Biochem Behav 2022; 220:173468. [PMID: 36174752 DOI: 10.1016/j.pbb.2022.173468] [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: 07/06/2022] [Revised: 09/20/2022] [Accepted: 09/21/2022] [Indexed: 01/06/2023]
Abstract
Major depressive disorder (MDD) is the most prevalent mood disorder globally. Most antidepressants available for the treatment of MDD increase the concentration of monoamines in the synaptic cleft. However, such drugs have a high latency time to obtain benefits. Thus, new antidepressants with fast action and robust efficacy are very important. This study evaluated the effects of escitalopram, ketamine, and probiotic Bifidobacterium infantis in rats submitted to the maternal deprivation (MD). MD rats received saline, escitalopram, ketamine, or probiotic for 10, 30, or 50 days, depending on the postnatal day (PND):21, 41, and 61. Following behavior, this study examined the integrity of the blood-brain barrier (BBB) and oxidative stress markers. MD induced depressive-like behavior in females with PND21 and males with PND61. All treatments reversed depressive-like behavior in females and escitalopram and ketamine in males. MD induced an increase in the permeability of the BBB, an imbalance between oxidative stress and antioxidant defenses. Treatments regulated the oxidative damage and the integrity of the BBB induced by MD. The treatment with escitalopram, ketamine, or probiotics may prevent behavioral and neurochemical changes associated with MDD, depending on the developmental period and gender.
Collapse
Affiliation(s)
- Anelise S Carlessi
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciuma, SC, Brazil
| | - Maria Eduarda M Botelho
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciuma, SC, Brazil
| | - Luana M Manosso
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciuma, SC, Brazil
| | - Laura A Borba
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciuma, SC, Brazil
| | - Larissa R Maciel
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciuma, SC, Brazil
| | - Natalia M Andrade
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciuma, SC, Brazil
| | - Nicoly S Martinello
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciuma, SC, Brazil
| | - Alex Paulo Z Padilha
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciuma, SC, Brazil
| | - Camille M Generoso
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciuma, SC, Brazil
| | - Clara Vitória Bencke
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciuma, SC, Brazil
| | - Airam B de Moura
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciuma, SC, Brazil
| | - Bruna F Lodetti
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Allan Collodel
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Larissa Joaquim
- Laboratory of Clinical and Experimental Pathophysiology, Postgraduate Program in Health Sciences, University of Southern Santa Catarina (UNISUL), Tubarão, SC, Brazil
| | - Sandra Bonfante
- Laboratory of Clinical and Experimental Pathophysiology, Postgraduate Program in Health Sciences, University of Southern Santa Catarina (UNISUL), Tubarão, SC, Brazil
| | - Erica Biehl
- Laboratory of Clinical and Experimental Pathophysiology, Postgraduate Program in Health Sciences, University of Southern Santa Catarina (UNISUL), Tubarão, SC, Brazil
| | - Jaqueline S Generoso
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Camila O Arent
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciuma, SC, Brazil
| | - Tatiana Barichello
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil; Laboratory of Clinical and Experimental Pathophysiology, Postgraduate Program in Health Sciences, University of Southern Santa Catarina (UNISUL), Tubarão, SC, Brazil
| | - Fabricia Petronilho
- Laboratory of Clinical and Experimental Pathophysiology, Postgraduate Program in Health Sciences, University of Southern Santa Catarina (UNISUL), Tubarão, SC, Brazil
| | - João Quevedo
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciuma, SC, Brazil; Translational Psychiatry Program, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA; Center of Excellence on Mood Disorders, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA; Neuroscience Graduate Program, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Gislaine Z Réus
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciuma, SC, Brazil.
| |
Collapse
|
18
|
Dandekar MP, Palepu MSK, Satti S, Jaiswal Y, Singh AA, Dash SP, Gajula SNR, Sonti R. Multi-strain Probiotic Formulation Reverses Maternal Separation and Chronic Unpredictable Mild Stress-Generated Anxiety- and Depression-like Phenotypes by Modulating Gut Microbiome-Brain Activity in Rats. ACS Chem Neurosci 2022; 13:1948-1965. [PMID: 35735411 DOI: 10.1021/acschemneuro.2c00143] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Depression is a debilitating mental disorder that affects >322 million people worldwide. Despite the availability of several antidepressant agents, many patients remain treatment refractory. A growing literature study has indicated the role of gut microbiota in neuropsychiatric disorders. Herein, we examined the psychobiotic-like activity of multi-strain probiotic formulation in maternal separation (MS) and chronic unpredictable mild stress (CUMS) models of anxiety- and depression-like phenotypes in Sprague-Dawley rats. Early- and late-life stress was employed in both male and female rats by exposing them to MS and CUMS. The multi-strain probiotic formulation (Cognisol) containing Bacillus coagulans Unique IS-2, Lactobacillus plantarum UBLP-40, Lactobacillus rhamnosus UBLR-58, Bifidobacterium lactis UBBLa-70, Bifidobacterium breve UBBr-01, and Bifidobacterium infantis UBBI-01 at a total strength of 10 billion cfu along with l-glutamine was administered for 6 weeks via drinking water. Neurobehavioral assessment was done using the forced swim test (FST), sucrose preference test (SPT), elevated plus maze (EPM), and open field test (OFT). Animals were sacrificed after behavioral assessment, and blood, brain, and intestine samples were collected to analyze the levels of cytokines, metabolites, and neurotransmitters and histology. Animals exposed to stress showed increased passivity, consumed less sucrose solution, and minimally explored the open arms in the FST, SPT, and EPM, respectively. Administration of multi-strain probiotics along with l-glutamine for 6 weeks ameliorated the behavioral abnormalities. The locomotor activity of animals in the OFT and their body weight remained unchanged across the groups. Cognisol treatment reversed the decreased BDNF and serotonin levels and increased CRP, TNF-α, and dopamine levels in the hippocampus and/or frontal cortex. Administration of Cognisol also restored the plasma levels of l-tryptophan, l-kynurenine, kynurenic-acid, and 3-hydroxyanthranilic acid; the Firmicutes-to-Bacteroides ratio; the levels of acetate, propionate, and butyrate in fecal samples; the villi/crypt ratio; and the goblet cell count, which manifested in the restoration of intestinal functions. We suggest that the multi-strain probiotic and glutamine formulation (Cognisol) ameliorated the MS + UCMS-generated anxiety- and depression-like phenotypes by reshaping the gut microbiome-brain activity in both sexes.
Collapse
Affiliation(s)
- Manoj P Dandekar
- Department of Biological Sciences, Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, India
| | - Mani Surya Kumar Palepu
- Department of Biological Sciences, Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, India
| | - Srilakshmi Satti
- Department of Biological Sciences, Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, India
| | - Yash Jaiswal
- Department of Biological Sciences, Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, India
| | - Aditya A Singh
- Department of Biological Sciences, Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, India
| | - Surya Prakash Dash
- Department of Biological Sciences, Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, India
| | - Siva Nageswara Rao Gajula
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, India
| | - Rajesh Sonti
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, India
| |
Collapse
|
19
|
Early Life Events and Maturation of the Dentate Gyrus: Implications for Neurons and Glial Cells. Int J Mol Sci 2022; 23:ijms23084261. [PMID: 35457079 PMCID: PMC9031216 DOI: 10.3390/ijms23084261] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 04/08/2022] [Accepted: 04/11/2022] [Indexed: 12/15/2022] Open
Abstract
The dentate gyrus (DG), an important part of the hippocampus, plays a significant role in learning, memory, and emotional behavior. Factors potentially influencing normal development of neurons and glial cells in the DG during its maturation can exert long-lasting effects on brain functions. Early life stress may modify maturation of the DG and induce lifelong alterations in its structure and functioning, underlying brain pathologies in adults. In this paper, maturation of neurons and glial cells (microglia and astrocytes) and the effects of early life events on maturation processes in the DG have been comprehensively reviewed. Early postnatal interventions affecting the DG eventually result in an altered number of granule neurons in the DG, ectopic location of neurons and changes in adult neurogenesis. Adverse events in early life provoke proinflammatory changes in hippocampal glia at cellular and molecular levels immediately after stress exposure. Later, the cellular changes may disappear, though alterations in gene expression pattern persist. Additional stressful events later in life contribute to manifestation of glial changes and behavioral deficits. Alterations in the maturation of neuronal and glial cells induced by early life stress are interdependent and influence the development of neural nets, thus predisposing the brain to the development of cognitive and psychiatric disorders.
Collapse
|
20
|
Chaudhari PR, Singla A, Vaidya VA. Early Adversity and Accelerated Brain Aging: A Mini-Review. Front Mol Neurosci 2022; 15:822917. [PMID: 35392273 PMCID: PMC8980717 DOI: 10.3389/fnmol.2022.822917] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Accepted: 02/25/2022] [Indexed: 11/13/2022] Open
Abstract
Early adversity is an important risk factor that influences brain aging. Diverse animal models of early adversity, including gestational stress and postnatal paradigms disrupting dam-pup interactions evoke not only persistent neuroendocrine dysfunction and anxio-depressive behaviors, but also perturb the trajectory of healthy brain aging. The process of brain aging is thought to involve hallmark features such as mitochondrial dysfunction and oxidative stress, evoking impairments in neuronal bioenergetics. Furthermore, brain aging is associated with disrupted proteostasis, progressively defective epigenetic and DNA repair mechanisms, the build-up of neuroinflammatory states, thus cumulatively driving cellular senescence, neuronal and cognitive decline. Early adversity is hypothesized to evoke an “allostatic load” via an influence on several of the key physiological processes that define the trajectory of healthy brain aging. In this review we discuss the evidence that animal models of early adversity impinge on fundamental mechanisms of brain aging, setting up a substratum that can accelerate and compromise the time-line and nature of brain aging, and increase risk for aging-associated neuropathologies.
Collapse
|
21
|
Combination of electroconvulsive stimulation with ketamine or escitalopram protects the brain against inflammation and oxidative stress induced by maternal deprivation and is critical for associated behaviors in male and female rats. Mol Neurobiol 2022; 59:1452-1475. [PMID: 34994953 DOI: 10.1007/s12035-021-02718-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 12/23/2021] [Indexed: 12/24/2022]
Abstract
This study aimed at evaluating the treatment effects with ketamine, electroconvulsive stimulation (ECS), escitalopram, alone or in combination in adult rats of both sexes, subjected to the animal model of maternal deprivation (MD). All groups were subjected to the forced swimming test (FST), splash and open field tests. The prefrontal cortex (PFC), hippocampus and serum were collected to analyze oxidative stress and inflammatory parameters. MD induced depressive-like behavior in the FST test in males and reduced grooming time in male and female rats. The treatments alone or combined reversed depressive and anhedonic behavior in females. In males, all treatments increased grooming time, except for ECS + escitalopram + ketamine. MD increased lipid peroxidation and protein carbonylation, nitrite/nitrate concentration and myeloperoxidase activity in the PFC and hippocampus of males and females. However, the treatment's response was sex dependent. Catalase activity decreased in the PFC of males and the PFC and hippocampus of females, and most treatments were not able to reverse it. MD increased the inflammation biomarkers levels in the PFC and hippocampus of males and females, and most treatments were able to reverse this increase. In all groups, a reduction in the interleukin-10 levels in the PFC and hippocampus of female and male rats was observed. Our study shows different responses between the sexes in the patterns evaluated and reinforces the use of the gender variable as a biological factor in MDD related to early stress and in the response of the therapeutic strategies used.
Collapse
|
22
|
Nicolas S, McGovern AJ, Hueston CM, O'Mahony SM, Cryan JF, O'Leary OF, Nolan YM. Prior maternal separation stress alters the dendritic complexity of new hippocampal neurons and neuroinflammation in response to an inflammatory stressor in juvenile female rats. Brain Behav Immun 2022; 99:327-338. [PMID: 34732365 DOI: 10.1016/j.bbi.2021.10.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 10/19/2021] [Accepted: 10/23/2021] [Indexed: 12/11/2022] Open
Abstract
Stress during critical periods of neurodevelopment is associated with an increased risk of developing stress-related psychiatric disorders, which are more common in women than men. Hippocampal neurogenesis (the birth of new neurons) is vulnerable to maternal separation (MS) and inflammatory stressors, and emerging evidence suggests that hippocampal neurogenesis is more sensitive to stress in the ventral hippocampus (vHi) than in the dorsal hippocampus (dHi). Although research into the effects of MS stress on hippocampal neurogenesis is well documented in male rodents, the effect in females remains underexplored. Similarly, reports on the impact of inflammatory stressors on hippocampal neurogenesis in females are limited, especially when female bias in the prevalence of stress-related psychiatric disorders begins to emerge. Thus, in this study we investigated the effects of MS followed by an inflammatory stressor (lipopolysaccharide, LPS) in early adolescence on peripheral and hippocampal inflammatory responses and hippocampal neurogenesis in juvenile female rats. We show that MS enhanced an LPS-induced increase in the pro-inflammatory cytokine IL-1β in the vHi but not in the dHi. However, microglial activation was similar following LPS alone or MS alone in both hippocampal regions, while MS prior to LPS reduced microglial activation in both dHi and vHi. The production of new neurons was unaffected by MS and LPS. MS and LPS independently reduced the dendritic complexity of new neurons, and MS exacerbated LPS-induced reductions in the complexity of distal dendrites of new neurons in the vHi but not dHi. These data highlight that MS differentially primes the physiological response to LPS in the juvenile female rat hippocampus.
Collapse
Affiliation(s)
- Sarah Nicolas
- Department of Anatomy and Neuroscience, University College Cork, Ireland
| | - Andrew J McGovern
- Department of Anatomy and Neuroscience, University College Cork, Ireland
| | - Cara M Hueston
- Department of Anatomy and Neuroscience, University College Cork, Ireland
| | - Siobhain M O'Mahony
- Department of Anatomy and Neuroscience, University College Cork, Ireland; APC Microbiome Ireland, University College Cork, Ireland
| | - John F Cryan
- Department of Anatomy and Neuroscience, University College Cork, Ireland; APC Microbiome Ireland, University College Cork, Ireland
| | - Olivia F O'Leary
- Department of Anatomy and Neuroscience, University College Cork, Ireland; APC Microbiome Ireland, University College Cork, Ireland
| | - Yvonne M Nolan
- Department of Anatomy and Neuroscience, University College Cork, Ireland; APC Microbiome Ireland, University College Cork, Ireland.
| |
Collapse
|
23
|
Milbocker KA, Campbell TS, Collins N, Kim S, Smith IF, Roth TL, Klintsova AY. Glia-Driven Brain Circuit Refinement Is Altered by Early-Life Adversity: Behavioral Outcomes. Front Behav Neurosci 2021; 15:786234. [PMID: 34924972 PMCID: PMC8678604 DOI: 10.3389/fnbeh.2021.786234] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 11/12/2021] [Indexed: 12/12/2022] Open
Abstract
Early-life adversity (ELA), often clinically referred to as "adverse childhood experiences (ACE)," is the exposure to stress-inducing events in childhood that can result in poor health outcomes. ELA negatively affects neurodevelopment in children and adolescents resulting in several behavioral deficits and increasing the risk of developing a myriad of neuropsychiatric disorders later in life. The neurobiological mechanisms by which ELA alters neurodevelopment in childhood have been the focus of numerous reviews. However, a comprehensive review of the mechanisms affecting adolescent neurodevelopment (i.e., synaptic pruning and myelination) is lacking. Synaptic pruning and myelination are glia-driven processes that are imperative for brain circuit refinement during the transition from adolescence to adulthood. Failure to optimize brain circuitry between key brain structures involved in learning and memory, such as the hippocampus and prefrontal cortex, leads to the emergence of maladaptive behaviors including increased anxiety or reduced executive function. As such, we review preclinical and clinical literature to explore the immediate and lasting effects of ELA on brain circuit development and refinement. Finally, we describe a number of therapeutic interventions best-suited to support adolescent neurodevelopment in children with a history of ELA.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Anna Y. Klintsova
- Department of Psychological and Brain Sciences, University of Delaware, Newark, DE, United States
| |
Collapse
|
24
|
Poleksic J, Aksic M, Kapor S, Aleksic D, Stojkovic T, Radovic M, Djulejic V, Markovic B, Stamatakis A. Effects of Maternal Deprivation on the Prefrontal Cortex of Male Rats: Cellular, Neurochemical, and Behavioral Outcomes. Front Behav Neurosci 2021; 15:666547. [PMID: 34819843 PMCID: PMC8606589 DOI: 10.3389/fnbeh.2021.666547] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 10/08/2021] [Indexed: 01/13/2023] Open
Abstract
Stressful events experienced during early life are associated with increased vulnerability of developing psychopathology in adulthood. In the present study, we exposed 9-day-old Wistar rats to 24 h maternal deprivation (MD) with the aim to investigate the impact of early life stress (ELS) on morphological, biochemical, and functional aspects of the prefrontal cortex (PFC), a brain region particularly sensitive to stress. We found that in the superficial medial orbital cortex (MO), young adult male rats had reduced density of GAD67 and CCK immunopositive cells, while the rostral part of the ventral lateral orbital cortex (roVLO) showed a decrease in the density of GAD67 immunopositive cells in both superficial and deep layers. In addition, the superficial rostral part of area 1 of the cingulate cortex (roCg1) and deep prelimbic cortex (PrL) was also affected by MD indicated by the reduction in PV immunopositive cellular density. Furthermore, MD induced upregulation of brain-derived neurotrophic factor (BDNF), while it did not affect the overall expression of Iba1 in neonatal or young adult PFC as measured by Western blot, however, microglial activation in young adult MD rats was detected immunohistochemically in deep layers of MO and infralimbic cortex (IL). Interestingly, when young adult male rats were subjected to a behavioral flexibility test in a T-maze, MD rats showed a subtle impairment in T-maze reversal learning indicating a mildly affected PFC function. Taken together, our findings demonstrated that MD reduced the density of interneurons and induced microglial activation, in particular, PFC areas at young adulthood, and could alter synaptic plasticity accompanied by PFC dysfunction.
Collapse
Affiliation(s)
- Joko Poleksic
- Institute of Anatomy "Niko Miljanic", School of Medicine, University of Belgrade, Belgrade, Serbia
| | - Milan Aksic
- Institute of Anatomy "Niko Miljanic", School of Medicine, University of Belgrade, Belgrade, Serbia
| | - Slobodan Kapor
- Institute of Anatomy "Niko Miljanic", School of Medicine, University of Belgrade, Belgrade, Serbia
| | - Dubravka Aleksic
- Institute of Anatomy "Niko Miljanic", School of Medicine, University of Belgrade, Belgrade, Serbia
| | - Tihomir Stojkovic
- Institute of Clinical and Medical Biochemistry, School of Medicine, University of Belgrade, Belgrade, Serbia
| | - Marina Radovic
- Institute of Physiology and Biochemistry "Ivan Djaja", Faculty of Biology, University of Belgrade, Belgrade, Serbia
| | - Vuk Djulejic
- Institute of Anatomy "Niko Miljanic", School of Medicine, University of Belgrade, Belgrade, Serbia
| | - Branka Markovic
- Faculty of Sport and Physical Education, University of Belgrade, Belgrade, Serbia
| | - Antonios Stamatakis
- Biology-Biochemistry Lab, School of Health Sciences, Faculty of Nursing, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
25
|
FKBP5 and early life stress affect the hippocampus by an age-dependent mechanism. Brain Behav Immun Health 2021; 9:100143. [PMID: 34589890 PMCID: PMC8474669 DOI: 10.1016/j.bbih.2020.100143] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 09/13/2020] [Indexed: 01/30/2023] Open
Abstract
Early life stress (ELS) adversely affects the brain and is commonly associated with the etiology of mental health disorders, like depression. In addition to the mood-related symptoms, patients with depression show dysregulation of the hypothalamic-pituitary-adrenal (HPA) axis, increased peripheral inflammation, and structural brain alterations. Although the underlying causes are unknown, polymorphisms in the FK506-binding protein 5 (FKBP5) gene, a regulator of glucocorticoid receptor (GR) activity, interact with childhood adversities to increase vulnerability to depressive disorders. We hypothesized that high FKBP5 protein levels combined with early life stress (ELS) would alter the HPA axis and brain, promoting depressive-like behaviors. To test this, we exposed males and females of a mouse model overexpressing FKBP5 in the brain (rTgFKBP5 mice), or littermate controls, to maternal separation for 14 days after birth. Then, we evaluated neuroendocrine, behavioral, and brain changes in young adult and aged mice. We observed lower basal corticosterone (CORT) levels in rTgFKBP5 mice, which was exacerbated in females. Aged, but not young, rTgFKBP5 mice showed increased depressive-like behaviors. Moreover, FKBP5 overexpression reduced hippocampal neuron density in aged mice, while promoting markers of microglia expression, but these effects were reversed by ELS. Together, these results demonstrate that high FKBP5 affects basal CORT levels, depressive-like symptoms, and numbers of neurons and microglia in the hippocampus in an age-dependent manner. High FKBP5 reduces basal corticosterone levels in mice, especially in females. ELS prevents FKBP5-induced susceptibility to depressive-like behavior in aged mice. FKBP5 overexpression reduces hippocampal neuron density in aged mice, while increasing microglial markers.
Collapse
|
26
|
Réus GZ, Giridharan VV, de Moura AB, Borba LA, Botelho MEM, Behenck JP, Generoso JS, Selvaraj S, Bhatti G, Barichello T, Quevedo J. The impact of early life stress and immune challenge on behavior and glia cells alteration in late adolescent rats. Int J Dev Neurosci 2021; 81:407-415. [PMID: 33788296 DOI: 10.1002/jdn.10108] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 03/08/2021] [Accepted: 03/25/2021] [Indexed: 01/06/2023] Open
Abstract
Maternal deprivation (MD) is known to be related to long-term changes that could influence the onset of psychiatric disorders. Studies have demonstrated that early life stress makes the cells in the brain more susceptible to subsequent stressors. To test it, we used an animal model of MD conducted from postnatal day (PND) 1 to 10. Deprived and non-deprived rats (control) were randomized to receive or not lipopolysaccharide (LPS) at 5 mg/kg on PND 50. The behavior and glial cells activation were evaluated in all groups from 51 to 53 PND. There was an increase in the immobility time in the MD and MD+LPS groups. The spontaneous locomotor activity was not changed between groups. We found elevated ionized calcium-binding adapter molecule 1 (Iba-1)-positive cells levels in the control+LPS and MD+LPS groups. In the MD+LPS group, it was found an increase in Iba-positive cells compared to the MD+sal group. The glial fibrillary acidic protein (GFAP)-positive cells were also increased in the MD+LPS, compared to control+sal, control+LPS, and MD+sal groups. Immune challenge by LPS in late adolescence, which was subjected to MD, did not influence the depressive-like behavior but exerted a pronounced effect in the microglial activation and astrocyte atrophy.
Collapse
Affiliation(s)
- Gislaine Z Réus
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, Brazil
| | - Vijayasree V Giridharan
- Translational Psychiatry Program, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Airam B de Moura
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, Brazil
| | - Laura A Borba
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, Brazil
| | - Maria Eduarda M Botelho
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, Brazil
| | - João Paulo Behenck
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, Brazil
| | - Jaqueline S Generoso
- Laboratory of Experimental Microbiology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, Brazil
| | - Sudhakar Selvaraj
- Louis Faillace, MD, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Gursimrat Bhatti
- Translational Psychiatry Program, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Tatiana Barichello
- Translational Psychiatry Program, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
- Laboratory of Experimental Microbiology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, Brazil
| | - João Quevedo
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, Brazil
- Translational Psychiatry Program, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
- Center of Excellence on Mood Disorders, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
- Neuroscience Graduate Program, The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, TX, USA
| |
Collapse
|
27
|
Abelaira HM, Veron DC, de Moura AB, Carlessi AS, Borba LA, Botelho MEM, Andrade NM, Martinello NS, Zabot GC, Joaquim L, Biehl E, Bonfante S, Budni J, Petronilho F, Quevedo J, Réus GZ. Sex differences on the behavior and oxidative stress after ketamine treatment in adult rats subjected to early life stress. Brain Res Bull 2021; 172:129-138. [PMID: 33932489 PMCID: PMC10464594 DOI: 10.1016/j.brainresbull.2021.04.021] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 04/25/2021] [Accepted: 04/26/2021] [Indexed: 01/22/2023]
Abstract
This study aimed to evaluate the effects of ketamine, on behavioral parameters, oxidative stress, and inflammation in the brain of male and female rats submitted to the animal model of maternal deprivation (MD). Wistar rats were deprived of maternal care in the first 10 days of life (three hours daily). As adults, male and female rats were divided: control + saline deprived + saline and deprived + ketamine (15 mg/kg). The behavior was evaluated through the open field and forced swimming tests. Then brain was removed for analysis of oxidative damage, the activity of superoxide dismutase (SOD), catalase (CAT), and myeloperoxidase (MPO) activity, and levels of interleukin-6 (IL-6). MD induced depressive behavior in males and ketamine reversed these changes. MD induced an increase in lipid peroxidation in males and females; ketamine reversed these effects in males. Protein carbonylation was increased in males and females, with ketamine decreasing such effects. The concentration of nitrite/nitrate increased in males and females, whereas ketamine decreased this in the PFC of males. SOD and CAT activities were decreased in male and female deprived groups and deprived groups treated with ketamine. MPO activity and IL-6 levels increased in males subjected to MD and ketamine reversed this effect. The results suggest that stressful events in early life can induce behavioral, neuroimmune changes, and oxidative stress, however, such effects depend on sex and brain area. Ketamine presents anti-inflammatory and antioxidant properties and could be considered an alternative for individuals who are resistant to classical treatments.
Collapse
Affiliation(s)
- Helena M Abelaira
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Deise Cristina Veron
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Airam B de Moura
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Anelise S Carlessi
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Laura A Borba
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Maria Eduarda M Botelho
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Natalia M Andrade
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Nicolly S Martinello
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Gabriel C Zabot
- Experimental Neurology Laboratory, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciúma, SC, Brazil
| | - Larissa Joaquim
- Laboratory of Clinical and Experimental Pathophysiology, Postgraduate Program in Health Sciences, University of Southern Santa Catarina (UNISUL), Tubarão, SC, Brazil
| | - Erica Biehl
- Laboratory of Clinical and Experimental Pathophysiology, Postgraduate Program in Health Sciences, University of Southern Santa Catarina (UNISUL), Tubarão, SC, Brazil
| | - Sandra Bonfante
- Laboratory of Clinical and Experimental Pathophysiology, Postgraduate Program in Health Sciences, University of Southern Santa Catarina (UNISUL), Tubarão, SC, Brazil
| | - Josiane Budni
- Experimental Neurology Laboratory, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciúma, SC, Brazil
| | - Fabricia Petronilho
- Laboratory of Clinical and Experimental Pathophysiology, Postgraduate Program in Health Sciences, University of Southern Santa Catarina (UNISUL), Tubarão, SC, Brazil
| | - João Quevedo
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil; Center of Excellence on Mood Disorders, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA; Neuroscience Graduate Program, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA; Translational Psychiatry Program, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Gislaine Z Réus
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil.
| |
Collapse
|
28
|
Tovar R, Vargas A, Aranda J, Sánchez-Salido L, González-González L, Chowen JA, Rodríguez de Fonseca F, Suárez J, Rivera P. Analysis of Both Lipid Metabolism and Endocannabinoid Signaling Reveals a New Role for Hypothalamic Astrocytes in Maternal Caloric Restriction-Induced Perinatal Programming. Int J Mol Sci 2021; 22:ijms22126292. [PMID: 34208173 PMCID: PMC8230792 DOI: 10.3390/ijms22126292] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 06/07/2021] [Accepted: 06/08/2021] [Indexed: 12/29/2022] Open
Abstract
Maternal malnutrition in critical periods of development increases the risk of developing short- and long-term diseases in the offspring. The alterations induced by this nutritional programming in the hypothalamus of the offspring are of special relevance due to its role in energy homeostasis, especially in the endocannabinoid system (ECS), which is involved in metabolic functions. Since astrocytes are essential for neuronal energy efficiency and are implicated in brain endocannabinoid signaling, here we have used a rat model to investigate whether a moderate caloric restriction (R) spanning from two weeks prior to the start of gestation to its end induced changes in offspring hypothalamic (a) ECS, (b) lipid metabolism (LM) and/or (c) hypothalamic astrocytes. Monitorization was performed by analyzing both the gene and protein expression of proteins involved in LM and ECS signaling. Offspring born from caloric-restricted mothers presented hypothalamic alterations in both the main enzymes involved in LM and endocannabinoids synthesis/degradation. Furthermore, most of these changes were similar to those observed in hypothalamic offspring astrocytes in culture. In conclusion, a maternal low caloric intake altered LM and ECS in both the hypothalamus and its astrocytes, pointing to these glial cells as responsible for a large part of the alterations seen in the total hypothalamus and suggesting a high degree of involvement of astrocytes in nutritional programming.
Collapse
Affiliation(s)
- Rubén Tovar
- Instituto de Investigación Biomédica de Málaga-IBIMA, 29010 Málaga, Spain; (R.T.); (A.V.); (J.A.); (L.S.-S.); (L.G.-G.); (F.R.d.F.)
- UGC Salud Mental, Hospital Regional Universitario de Málaga, 29010 Málaga, Spain
- Andalucia Tech, Facultad de Medicina, Universidad de Málaga, Campus de Teatinos s/n, 29071 Málaga, Spain
| | - Antonio Vargas
- Instituto de Investigación Biomédica de Málaga-IBIMA, 29010 Málaga, Spain; (R.T.); (A.V.); (J.A.); (L.S.-S.); (L.G.-G.); (F.R.d.F.)
- UGC Salud Mental, Hospital Regional Universitario de Málaga, 29010 Málaga, Spain
| | - Jesús Aranda
- Instituto de Investigación Biomédica de Málaga-IBIMA, 29010 Málaga, Spain; (R.T.); (A.V.); (J.A.); (L.S.-S.); (L.G.-G.); (F.R.d.F.)
- Andalucia Tech, Facultad de Medicina, Universidad de Málaga, Campus de Teatinos s/n, 29071 Málaga, Spain
| | - Lourdes Sánchez-Salido
- Instituto de Investigación Biomédica de Málaga-IBIMA, 29010 Málaga, Spain; (R.T.); (A.V.); (J.A.); (L.S.-S.); (L.G.-G.); (F.R.d.F.)
- UGC Salud Mental, Hospital Regional Universitario de Málaga, 29010 Málaga, Spain
| | - Laura González-González
- Instituto de Investigación Biomédica de Málaga-IBIMA, 29010 Málaga, Spain; (R.T.); (A.V.); (J.A.); (L.S.-S.); (L.G.-G.); (F.R.d.F.)
| | - Julie A. Chowen
- Department of Endocrinology, Instituto de Investigación Biomédica la Princesa, Fundación Investigación Biomédica del Hospital Infantil Universitario Niño Jesús, 28009 Madrid, Spain;
- CIBEROBN (Centro de Investigación Biomédica en Red Sobre Fisiopatología de la Obesidad y Nutrición), Instituto de Salud Carlos III, 28009 Madrid, Spain
- IMDEA Food Institute, CEI UAM + CSIC, 28009 Madrid, Spain
| | - Fernando Rodríguez de Fonseca
- Instituto de Investigación Biomédica de Málaga-IBIMA, 29010 Málaga, Spain; (R.T.); (A.V.); (J.A.); (L.S.-S.); (L.G.-G.); (F.R.d.F.)
- UGC Salud Mental, Hospital Regional Universitario de Málaga, 29010 Málaga, Spain
| | - Juan Suárez
- Instituto de Investigación Biomédica de Málaga-IBIMA, 29010 Málaga, Spain; (R.T.); (A.V.); (J.A.); (L.S.-S.); (L.G.-G.); (F.R.d.F.)
- Departamento de Anatomía Humana, Medicina Legal e Historia de la Ciencia, Universidad de Málaga, 29071 Málaga, Spain
- Correspondence: (J.S.); (P.R.); Tel.: +34-952614012 (J.S.); +34-952614012 (P.R.)
| | - Patricia Rivera
- Instituto de Investigación Biomédica de Málaga-IBIMA, 29010 Málaga, Spain; (R.T.); (A.V.); (J.A.); (L.S.-S.); (L.G.-G.); (F.R.d.F.)
- UGC Salud Mental, Hospital Regional Universitario de Málaga, 29010 Málaga, Spain
- Correspondence: (J.S.); (P.R.); Tel.: +34-952614012 (J.S.); +34-952614012 (P.R.)
| |
Collapse
|
29
|
Hammond BP, Manek R, Kerr BJ, Macauley MS, Plemel JR. Regulation of microglia population dynamics throughout development, health, and disease. Glia 2021; 69:2771-2797. [PMID: 34115410 DOI: 10.1002/glia.24047] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 05/20/2021] [Accepted: 05/28/2021] [Indexed: 12/11/2022]
Abstract
The dynamic expansions and contractions of the microglia population in the central nervous system (CNS) to achieve homeostasis are likely vital for their function. Microglia respond to injury or disease but also help guide neurodevelopment, modulate neural circuitry throughout life, and direct regeneration. Throughout these processes, microglia density changes, as does the volume of area that each microglia surveys. Given that microglia are responsible for sensing subtle alterations to their environment, a change in their density could affect their capacity to mobilize rapidly. In this review, we attempt to synthesize the current literature on the ligands and conditions that promote microglial proliferation across development, adulthood, and neurodegenerative conditions. Microglia display an impressive proliferative capacity during development and in neurodegenerative diseases that is almost completely absent at homeostasis. However, the appropriate function of microglia in each state is critically dependent on density fluctuations that are primarily induced by proliferation. Proliferation is a natural microglial response to insult and often serves neuroprotective functions. In contrast, inappropriate microglial proliferation, whether too much or too little, often precipitates undesirable consequences for nervous system health. Thus, fluctuations in the microglia population are tightly regulated to ensure these immune cells can execute their diverse functions.
Collapse
Affiliation(s)
- Brady P Hammond
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Rupali Manek
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Bradley J Kerr
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada.,Department of Anesthesiology & Pain Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Matthew S Macauley
- Department of Chemistry, University of Alberta, Edmonton, Alberta, Canada.,Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Alberta, Canada
| | - Jason R Plemel
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada.,Department of Medicine, Division of Neurology, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
30
|
Interrelationships Between Oxidative Stress, Cytokines, and Psychotic Symptoms and Executive Functions in Patients With Chronic Schizophrenia. Psychosom Med 2021; 83:485-491. [PMID: 34080586 DOI: 10.1097/psy.0000000000000931] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
OBJECTIVE Accumulating evidence has demonstrated that the pathophysiology of schizophrenia is involved in various abnormalities in oxidative stress markers and cytokines closely related to synaptic plasticity. However, the interactive effects among key cytokines, oxidative stress, and executive dysfunction and symptoms of schizophrenia have not been investigated yet. METHODS A total of 189 patients with chronic schizophrenia and 60 controls were recruited in the current study. Tumor necrosis factor α (TNF-α), interleukin (IL)-8, IL-6, and IL-2 levels; catalase, glutathione peroxidase, and superoxide dismutase (SOD) activities; and malondialdehyde (MDA) levels were determined in patients and controls. Executive function was evaluated by the Wisconsin card sorting tests, the verbal fluency tests, and the Stroop word-color test. Clinical symptoms were evaluated by the Positive and Negative Syndrome Scale. RESULTS Relative to the controls, the patients had lower activities of SOD and glutathione peroxidase and levels of TNF-α, but higher levels of MDA, IL-8, IL-6, and IL-2 (all p values < .05). A significant negative relationship between SOD activity and IL-8 levels was found only in patients (β = -0.44, p = .008). Furthermore, we found that an interactive effect of low TNF-α level and high MDA level was associated with negative symptoms (β = -0.02, p = .01). Moreover, the interactive effects of IL-8 and MDA or IL-8 and SOD were correlated with executive function only in patients (β = 0.23, p = .02; β = 0.09, p = .03). CONCLUSIONS Our findings suggest that the interrelationships between oxidative stress markers and cytokines occur in schizophrenia patients, which may be the basis of their pathological mechanisms underlying clinical symptoms and cognitive dysfunction.
Collapse
|
31
|
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: 4.8] [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
|
32
|
Catale C, Bisicchia E, Carola V, Viscomi MT. Early life stress exposure worsens adult remote microglia activation, neuronal death, and functional recovery after focal brain injury. Brain Behav Immun 2021; 94:89-103. [PMID: 33677027 DOI: 10.1016/j.bbi.2021.02.032] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 02/25/2021] [Accepted: 02/28/2021] [Indexed: 01/08/2023] Open
Abstract
Trauma to the central nervous system (CNS) is a devastating condition resulting in severe functional impairments that strongly vary among patients. Patients' features, such as age, social and cultural environment, and pre-existing psychiatric conditions may be particularly relevant for determining prognosis after CNS trauma. Although several studies demonstrated the impact of adult psycho-social stress exposure on functional recovery after CNS damage, no data exist regarding the long-term effects of the exposure to such experience at an early age. Here, we assessed whether early life stress (ELS) hampers the neuroinflammatory milieuand the functional recovery after focal brain injury in adulthood by using a murine model of ELS exposure combined with hemicerebellectomy (HCb), a model of remote damage. We found that ELS permanently altered microglia responses such that, once experienced HCb, they produced an exaggerated remote inflammatory response - consistent with a primed phenotype - associated with increased cell death and worse functional recovery. Notably, prevention of microglia/macrophages activation by GW2580 treatment during ELS exposure significantly reduced microglia responses, cell death and improved functional recovery. Conversely, GW2580 treatment administered in adulthood after HCb was ineffective in reducing inflammation and cell death or improving functional recovery. Our findings highlight that ELS impacts the immune system maturation producing permanent changes, and that it is a relevant factor modulating the response to a CNS damage. Further studies are needed to clarify the mechanisms underlying the interaction between ELS and brain injury with the aim of developing targeted treatments to improve functional recovery after CNS damage.
Collapse
Affiliation(s)
- Clarissa Catale
- Department of Psychology, Ph.D. Program in "Behavioral Neuroscience", Sapienza University of Rome, Rome, Italy
| | | | - Valeria Carola
- IRCCS Santa Lucia Foundation, Rome, Italy; Department of Dynamic and Clinical Psychology, and Health Studies, Sapienza University of Rome, Rome, Italy.
| | - Maria Teresa Viscomi
- Department of Life Science and Public Health, Section of Histology and Embryology, University "Cattolica Del S. Cuore", Rome, Italy.
| |
Collapse
|
33
|
Brain CHID1 Expression Correlates with NRGN and CALB1 in Healthy Subjects and AD Patients. Cells 2021; 10:cells10040882. [PMID: 33924468 PMCID: PMC8069241 DOI: 10.3390/cells10040882] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 04/06/2021] [Accepted: 04/12/2021] [Indexed: 12/14/2022] Open
Abstract
Alzheimer’s disease is a progressive, devastating, and irreversible brain disorder that, day by day, destroys memory skills and social behavior. Despite this, the number of known genes suitable for discriminating between AD patients is insufficient. Among the genes potentially involved in the development of AD, there are the chitinase-like proteins (CLPs) CHI3L1, CHI3L2, and CHID1. The genes of the first two have been extensively investigated while, on the contrary, little information is available on CHID1. In this manuscript, we conducted transcriptome meta-analysis on an extensive sample of brains of healthy control subjects (n = 1849) (NDHC) and brains of AD patients (n = 1170) in order to demonstrate CHID1 involvement. Our analysis revealed an inverse correlation between the brain CHID1 expression levels and the age of NDHC subjects. Significant differences were highlighted comparing CHID1 expression of NDHC subjects and AD patients. Exclusive in AD patients, the CHID1 expression levels were correlated positively to calcium-binding adapter molecule 1 (IBA1) levels. Furthermore, both in NDHC and in AD patient’s brains, the CHID1 expression levels were directly correlated with calbindin 1 (CALB1) and neurogranin (NRGN). According to brain regions, correlation differences were shown between the expression levels of CHID1 in prefrontal, frontal, occipital, cerebellum, temporal, and limbic system. Sex-related differences were only highlighted in NDHC. CHID1 represents a new chitinase potentially involved in the principal processes underlying Alzheimer’s disease.
Collapse
|
34
|
Behavior and oxidative stress parameters in rats subjected to the animal's models induced by chronic mild stress and 6-hydroxydopamine. Behav Brain Res 2021; 406:113226. [PMID: 33684423 DOI: 10.1016/j.bbr.2021.113226] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 03/01/2021] [Accepted: 03/02/2021] [Indexed: 12/16/2022]
Abstract
Major depressive disorder (MDD) is one of the most prevalent forms of mental illness also affecting older adults. Recent evidence suggests a relationship between MDD and neurodegenerative diseases, including Parkinson's disease (PD). Individuals with PD have a predisposition to developing MDD, and both neurobiological conditions are associated with oxidative stress. Thus, we conducted this study to investigate depressive-like behavior and oxidative stress parameters using both animal models of PD and stress. Adult Wistar rats were subjected to chronic mild stress (CMS) protocol by 40 days and then it was used 6-hydroxydopamine (6-OHDA) as a model of PD, into the striatum. The experimental groups were: Control + Sham, Stress + Sham, Control+6-OHDA, and Stress+6-OHDA. Depressive like-behavior was evaluated by the forced swimming test (FST) and spontaneous locomotor activity by open-field test. Oxidative stress parameters were measured in the striatum, hippocampus, and prefrontal cortex (PFC). The results showed effects to increase immobility and decrease climbing times in the FST in Stress + Sham, Control+6-OHDA, and Stress+6-OHDA groups. The number of crossings and rearings were decreased in the Stress+6-OHDA group. The lipid peroxidation was increased in the PFC of Stress + Sham, and the hippocampus and striatum of Stress + Sham and Control+6-OHDA groups. Carbonyl protein levels increased in the PFC of Stress + Sham and striatum in Control+6-OHDA. Nitrite/Nitrate concentration was elevated in the PFC of Stress + Sham, in the hippocampus of Control+6-OHDA, the striatum of Stress + Sham, and Control+6-OHDA groups. Myeloperoxidase (MPO) activity was increased in the PFC and hippocampus of Stress + Sham and Control+6-OHDA groups. The activity of catalase decreased in the PFC of the Stress + Sham group. The activity of the superoxide dismutase (SOD) was decreased in the PFC of the Stress + Sham group, in the hippocampus of Stress + Sham and Control+6-OHDA groups, and the striatum of Control+6-OHDA group. These findings suggest that both stress and 6-OHDA induce depressive-like behavior and oxidative stress in the brain. The joining models have little evidence of the effects. Thus these findings suggest that other pathways are involved in the common point of the pathophysiology of PD and MDD.
Collapse
|
35
|
Gonçalves CL, Abelaira HM, Rosa T, de Moura AB, Veron DC, Borba LA, Botelho MEM, Goldim MP, Garbossa L, Fileti ME, Petronilho F, Ignácio ZM, Quevedo J, Réus GZ. Ketamine treatment protects against oxidative damage and the immunological response induced by electroconvulsive therapy. Pharmacol Rep 2021; 73:525-535. [PMID: 33393059 DOI: 10.1007/s43440-020-00200-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 11/17/2020] [Accepted: 11/21/2020] [Indexed: 12/27/2022]
Abstract
BACKGROUND Electroconvulsive therapy (ECT) is often recommended for major depressive disorder (MDD) for those who do not respond to the first and second antidepressant trials. A combination of two therapies could improve antidepressant efficacy. Thus, this study aimed to investigate the synergistic effects of ECT combined to antidepressants with a different mechanism of action. METHODS Rats were treated once a day, for five days with ketamine (5 mg/kg), fluoxetine (1 mg/kg), and bupropion (4 mg/kg) alone or in combination with ECT (1 mA; 100 V). After, oxidative damage and antioxidant capacity were assessed in the prefrontal cortex (PFC) and hippocampus, and pro-inflammatory cytokines levels were evaluated in the serum. RESULTS ECT alone increased lipid peroxidation in the PFC and hippocampus. In the PFC of rats treated with ECT in combination with fluoxetine and bupropion, and in the hippocampus of rats treated with ECT combined with ketamine and bupropion there was a reduction in the lipid peroxidation. The nitrite/nitrate was increased by ECT alone but reverted by combination with ketamine in the hippocampus. Superoxide dismutase (SOD) was increased by ECT and maintained by fluoxetine and bupropion in the PFC. ECT alone increased interleukin-1β (IL-1β) and the administration of ketamine was able to revert this increase showing a neuroprotective effect of this drug when in combination with ECT. CONCLUSION The treatment with ECT leads to an increase in oxidative damage and alters the immunological system. The combination with ketamine was able to protect against oxidative damage and the immunological response induced by ECT.
Collapse
Affiliation(s)
- Cinara Ludvig Gonçalves
- Experimental Neurology Laboratory, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciúma, SC, Brazil
| | - Helena Mendes Abelaira
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciúma, SC, 88806-000, Brazil
| | - Thayse Rosa
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciúma, SC, 88806-000, Brazil
| | - Airam Barbosa de Moura
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciúma, SC, 88806-000, Brazil
| | - Deise Cristina Veron
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciúma, SC, 88806-000, Brazil
| | - Laura Araújo Borba
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciúma, SC, 88806-000, Brazil
| | - Maria Eduarda Mendes Botelho
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciúma, SC, 88806-000, Brazil
| | - Mariana Pereira Goldim
- Neurobiology of Metabolic and Inflammatory Processes Laboratory, Graduate Program in Health Sciences, University of South Santa Catarina, Tubarão, SC, Brazil
| | - Leandro Garbossa
- Neurobiology of Metabolic and Inflammatory Processes Laboratory, Graduate Program in Health Sciences, University of South Santa Catarina, Tubarão, SC, Brazil
| | - Maria Eduarda Fileti
- Neurobiology of Metabolic and Inflammatory Processes Laboratory, Graduate Program in Health Sciences, University of South Santa Catarina, Tubarão, SC, Brazil
| | - Fabricia Petronilho
- Neurobiology of Metabolic and Inflammatory Processes Laboratory, Graduate Program in Health Sciences, University of South Santa Catarina, Tubarão, SC, Brazil
| | - Zuleide Maria Ignácio
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciúma, SC, 88806-000, Brazil.,Laboratory of Physiology, Pharmacology and Psychopathology, Campus Chapecó, Federal University of South Frontier (UFFS), Chapecó, Santa Catarina, Brazil.,State Secretary for Justice and Citizenship of Santa Catarina, Chapecó, Brazil
| | - João Quevedo
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciúma, SC, 88806-000, Brazil.,Department of Psychiatry and Behavioral Sciences, Center of Excellence On Mood Disorders, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA.,Translational Psychiatry Program, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA.,Neuroscience Graduate Program, Graduate School of Biomedical Sciences, The University of Texas Health Science Center At Houston (UTHealth), Houston, TX, USA
| | - Gislaine Zilli Réus
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciúma, SC, 88806-000, Brazil.
| |
Collapse
|
36
|
Alviña K, Jodeiri Farshbaf M, Mondal AK. Long term effects of stress on hippocampal function: Emphasis on early life stress paradigms and potential involvement of neuropeptide Y. J Neurosci Res 2021; 99:57-66. [PMID: 32162350 DOI: 10.1002/jnr.24614] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 02/06/2020] [Accepted: 02/27/2020] [Indexed: 01/06/2023]
Abstract
The brain is both central in orchestrating the response to stress, and, a very sensitive target when such response is not controlled. In fact, stress has long been associated with the onset and/or exacerbation of several neuropsychiatric disorders such as anxiety, depression, and drug addiction. The hippocampus is a key brain region involved in the response to stress, not only due to its anatomical connections with the hypothalamic-pituitary-adrenal axis but also as a major target of stress mediators. The hippocampal dentate gyrus (DG)-CA3 circuit, composed of DG granule cells axons (mossy fibers) synapsing onto CA3 pyramidal cells, plays an essential role in memory encoding and retrieval, functions that are vulnerable to stress. Although naturally excitatory, this circuit is under the inhibitory control of GABAergic interneurons that maintain the excitation/inhibition balance. One subgroup of such interneurons produces neuropeptide Y (NPY), which has emerged as a promising endogenous stress "resilience molecule" due to its anxiolytic and anti-epileptic properties. Here we examine existing evidence that reveals a potential role for hilar NPY+ interneurons in mediating stress-induced changes in hippocampal function. We will focus specifically on rodent models of early life stress (ELS), defined as adverse conditions during the early postnatal period that can have profound consequences for neurodevelopment. Collectively, these findings suggest that the long-lasting effects of ELS might stem from the loss of GABAergic NPY+ cells, which then can lead to reduced inhibition in the DG-CA3 pathway. Such change might then lead to hyperexcitability and concomitant hippocampal-dependent behavioral deficits.
Collapse
Affiliation(s)
- Karina Alviña
- Department of Biological Sciences, Texas Tech University, Lubbock, TX, USA
- Department of Neuroscience, University of Florida, Gainesville, FL, USA
| | | | - Amit Kumar Mondal
- Department of Biological Sciences, Texas Tech University, Lubbock, TX, USA
| |
Collapse
|
37
|
Gildawie KR, Orso R, Peterzell S, Thompson V, Brenhouse HC. Sex differences in prefrontal cortex microglia morphology: Impact of a two-hit model of adversity throughout development. Neurosci Lett 2020; 738:135381. [PMID: 32927000 PMCID: PMC7584734 DOI: 10.1016/j.neulet.2020.135381] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 08/14/2020] [Accepted: 09/08/2020] [Indexed: 12/19/2022]
Abstract
Neuroimmune mechanisms play critical roles in brain development and can be impacted by early life adversity. Microglia are the resident immune cells in the brain, with both sex-specific and region-specific developmental profiles. Since early life adversity is associated with several neuropsychiatric disorders with developmental pathogeneses, here we investigated the degree to which maternal separation (MS) impacted microglia over development. Microglia are dynamic cells that alter their morphology in accordance with their functions and in response to stressors. While males and females reportedly display different microglial morphology in several brain regions over development and following immune and psychological challenges, little is known about such differences in the prefrontal cortex (PFC), which regulates several early life adversity-attributable disorders. Additionally, little is known about the potential for early life adversity to prime microglia for later immune challenges. In the current study, male and female rats were exposed to MS followed by lipopolysaccharide administration in juvenility or adolescence. The prelimbic and infralimbic PFC were then separately analyzed for microglial density and morphology. Typically developing males expressed smaller soma and less arborization than females in juvenility, but larger soma than females in adolescence. MS led to fewer microglia in the infralimbic PFC of adolescent males. Both MS and lipopolysaccharide administration affected morphological characteristics in juvenile males and females, with MS exposure leading to a greater increase in soma size following lipopolysaccharide. Interestingly, effects of MS and lipopolysaccharide were not observed in adolescence, while notable sex differences in PFC microglial morphology were apparent. Taken together, these findings provide insight into how PFC microglia may differentially respond to challenges over development in males and females.
Collapse
Affiliation(s)
| | - Rodrigo Orso
- Psychology Department, Northeastern University, Boston, MA, USA; Developmental Cognitive Neuroscience Lab (DCNL), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, RS, Brazil
| | | | | | | |
Collapse
|
38
|
Reddaway J, Brydges NM. Enduring neuroimmunological consequences of developmental experiences: From vulnerability to resilience. Mol Cell Neurosci 2020; 109:103567. [PMID: 33068720 PMCID: PMC7556274 DOI: 10.1016/j.mcn.2020.103567] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 09/14/2020] [Accepted: 10/12/2020] [Indexed: 12/14/2022] Open
Abstract
The immune system is crucial for normal neuronal development and function (neuroimmune system). Both immune and neuronal systems undergo significant postnatal development and are sensitive to developmental programming by environmental experiences. Negative experiences from infection to psychological stress at a range of different time points (in utero to adolescence) can permanently alter the function of the neuroimmune system: given its prominent role in normal brain development and function this dysregulation may increase vulnerability to psychiatric illness. In contrast, positive experiences such as exercise and environmental enrichment are protective and can promote resilience, even restoring the detrimental effects of negative experiences on the neuroimmune system. This suggests the neuroimmune system is a viable therapeutic target for treatment and prevention of psychiatric illnesses, especially those related to stress. In this review we will summarise the main cells, molecules and functions of the immune system in general and with specific reference to central nervous system development and function. We will then discuss the effects of negative and positive environmental experiences, especially during development, in programming the long-term functioning of the neuroimmune system. Finally, we will review the sparse but growing literature on sex differences in neuroimmune development and response to environmental experiences. The immune system is essential for development and function of the central nervous system (neuroimmune system) Environmental experiences can permanently alter neuroimmune function and associated brain development Altered neuroimmune function following negative developmental experiences may play a role in psychiatric illnesses Positive experiences can promote resilience and rescue the effects of negative experiences on the neuroimmune system The neuroimmune system is therefore a viable therapeutic target for preventing and treating psychiatric illnesses
Collapse
Affiliation(s)
- Jack Reddaway
- Neuroscience and Mental Health Research Institute, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff CF24 4HQ, UK
| | - Nichola M Brydges
- Neuroscience and Mental Health Research Institute, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff CF24 4HQ, UK.
| |
Collapse
|
39
|
Molecular Mechanisms of Glial Cells Related Signaling Pathways Involved in the Neuroinflammatory Response of Depression. Mediators Inflamm 2020; 2020:3497920. [PMID: 33100903 PMCID: PMC7569467 DOI: 10.1155/2020/3497920] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 09/17/2020] [Accepted: 09/28/2020] [Indexed: 02/07/2023] Open
Abstract
Dysfunction of the glial cells, such as astrocytes and microglia, is one of the pathological features in many psychiatric disorders, including depression, which emphasizes that glial cells driving neuroinflammation is not only an important pathological change in depression but also a potential therapeutic target. In this review, we summarized a recent update about several signaling pathways in which glial cells may play their roles in depression through neuroinflammatory reactions. We focused on the basic knowledge of these signaling pathways by elaborating each of them. This review may provide an updated image about the recent advances on these signaling pathways that are essential parts of neuroinflammation involved in depression.
Collapse
|
40
|
Comer AL, Carrier M, Tremblay MÈ, Cruz-Martín A. The Inflamed Brain in Schizophrenia: The Convergence of Genetic and Environmental Risk Factors That Lead to Uncontrolled Neuroinflammation. Front Cell Neurosci 2020; 14:274. [PMID: 33061891 PMCID: PMC7518314 DOI: 10.3389/fncel.2020.00274] [Citation(s) in RCA: 130] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 08/03/2020] [Indexed: 12/12/2022] Open
Abstract
Schizophrenia is a disorder with a heterogeneous etiology involving complex interplay between genetic and environmental risk factors. The immune system is now known to play vital roles in nervous system function and pathology through regulating neuronal and glial development, synaptic plasticity, and behavior. In this regard, the immune system is positioned as a common link between the seemingly diverse genetic and environmental risk factors for schizophrenia. Synthesizing information about how the immune-brain axis is affected by multiple factors and how these factors might interact in schizophrenia is necessary to better understand the pathogenesis of this disease. Such knowledge will aid in the development of more translatable animal models that may lead to effective therapeutic interventions. Here, we provide an overview of the genetic risk factors for schizophrenia that modulate immune function. We also explore environmental factors for schizophrenia including exposure to pollution, gut dysbiosis, maternal immune activation and early-life stress, and how the consequences of these risk factors are linked to microglial function and dysfunction. We also propose that morphological and signaling deficits of the blood-brain barrier, as observed in some individuals with schizophrenia, can act as a gateway between peripheral and central nervous system inflammation, thus affecting microglia in their essential functions. Finally, we describe the diverse roles that microglia play in response to neuroinflammation and their impact on brain development and homeostasis, as well as schizophrenia pathophysiology.
Collapse
Affiliation(s)
- Ashley L. Comer
- Graduate Program for Neuroscience, Boston University, Boston, MA, United States
- Department of Biology, Boston University, Boston, MA, United States
- Neurophotonics Center, Boston University, Boston, MA, United States
- Center for Systems Neuroscience, Boston University, Boston, MA, United States
| | - Micaël Carrier
- Axe Neurosciences, Centre de Recherche du CHU de Québec, Université Laval, Québec City, QC, Canada
| | - Marie-Ève Tremblay
- Axe Neurosciences, Centre de Recherche du CHU de Québec, Université Laval, Québec City, QC, Canada
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Department of Biochemistry and Molecular Biology, The University of British Columbia, Vancouver, BC, Canada
| | - Alberto Cruz-Martín
- Graduate Program for Neuroscience, Boston University, Boston, MA, United States
- Department of Biology, Boston University, Boston, MA, United States
- Neurophotonics Center, Boston University, Boston, MA, United States
- Center for Systems Neuroscience, Boston University, Boston, MA, United States
- Department of Pharmacology and Experimental Therapeutics, Boston University, Boston, MA, United States
| |
Collapse
|
41
|
CHI3L2 Expression Levels Are Correlated with AIF1, PECAM1, and CALB1 in the Brains of Alzheimer's Disease Patients. J Mol Neurosci 2020; 70:1598-1610. [PMID: 32705525 DOI: 10.1007/s12031-020-01667-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 07/09/2020] [Indexed: 02/06/2023]
Abstract
Alzheimer's disease (AD) represents one of the main forms of dementia that afflicts our society. The expression of several genes has been associated with disease development. Despite this, the number of genes known to be capable of discriminating between AD patients according to sex remains deficient. In our study, we performed a transcriptomes meta-analysis on a large court of brains of healthy control subjects (n = 2139) (NDHC) and brains of AD patients (n = 1170). Our aim was to verify the brain expression levels of CHI3L2 and its correlation with genes associated with microglia-mediated neuroinflammation (IBA1), alteration of the blood-brain barrier (PECAM1), and neuronal damage (CALB1). We showed that the CHI3L2, IBA1, PECAM1, and CALB1 expression levels were modulated in the brains of patients with AD compared to NDHC subjects. Furthermore, both in NDHC and in AD patient's brains, the CHI3L2 expression levels were directly correlated with IBA1 and PECAM1 and inversely with CALB1. Additionally, the expression levels of CHI3L2, PECAM1, and CALB1 but not of IBA1 were sex-depended. By stratifying the samples according to age and sex, correlation differences emerged between the expression levels of CHI3L2, IBA1, PECAM1, and CALB1 and the age of NDHC subjects and AD patients. CHI3L2 represents a promising gene potentially involved in the key processes underlying Alzheimer's disease. Its expression in the brains of sex-conditioned AD patients opens up new possible sex therapeutic strategies aimed at controlling imbalance in disease progression.
Collapse
|
42
|
Long-Term Impact of Early-Life Stress on Hippocampal Plasticity: Spotlight on Astrocytes. Int J Mol Sci 2020; 21:ijms21144999. [PMID: 32679826 PMCID: PMC7404101 DOI: 10.3390/ijms21144999] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 07/10/2020] [Accepted: 07/13/2020] [Indexed: 12/15/2022] Open
Abstract
Adverse experiences during childhood are among the most prominent risk factors for developing mood and anxiety disorders later in life. Early-life stress interventions have been established as suitable models to study the neurobiological basis of childhood adversity in rodents. Different models such as maternal separation, impaired maternal care and juvenile stress during the postweaning/prepubertal life phase are utilized. Especially within the limbic system, they induce lasting alterations in neuronal circuits, neurotransmitter systems, neuronal architecture and plasticity that are further associated with emotional and cognitive information processing. Recent studies found that astrocytes, a special group of glial cells, have altered functions following early-life stress as well. As part of the tripartite synapse, astrocytes interact with neurons in multiple ways by affecting neurotransmitter uptake and metabolism, by providing gliotransmitters and by providing energy to neurons within local circuits. Thus, astrocytes comprise powerful modulators of neuronal plasticity and are well suited to mediate the long-term effects of early-life stress on neuronal circuits. In this review, we will summarize current findings on altered astrocyte function and hippocampal plasticity following early-life stress. Highlighting studies for astrocyte-related plasticity modulation as well as open questions, we will elucidate the potential of astrocytes as new targets for interventions against stress-induced neuropsychiatric disorders.
Collapse
|
43
|
Han Y, Wang J, Zhao Q, Xie X, Song R, Xiao Y, Kang X, Zhang L, Zhang Y, Peng C, You Z. Pioglitazone alleviates maternal sleep deprivation-induced cognitive deficits in male rat offspring by enhancing microglia-mediated neurogenesis. Brain Behav Immun 2020; 87:568-578. [PMID: 32032783 DOI: 10.1016/j.bbi.2020.02.002] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 02/03/2020] [Accepted: 02/03/2020] [Indexed: 12/12/2022] Open
Abstract
Maternal sleep disturbance in pregnancy causes cognitive impairments and emotional disorders in offspring. Microglia-mediated inflammatory processes contribute to prenatal stress-induced neurodevelopmental deficits. Peroxisome proliferator-activated receptor gamma (PPARγ) activation underlies the switching of microglial activation phenotypes, which has emerged as a pharmacological target for regulating neuroinflammatory responses in the treatment of neuropsychiatric disorders. Here we investigated the effects of PPARγ-dependent microglial activation on neurogenesis and cognitive behavioral outcomes in male rat offspring exposed to maternal sleep deprivation (MSD) for 72 h from days 18-21 of pregnancy. In the Morris water maze test, male MSD rat offspring needed more time than control offspring to escape to the hidden platform and spent less time in the target quadrant when the hidden platform was removed. In MSD rat offspring, microglial density as determined by immunofluorescence was higher, microglia showed fewer and shorter processes, and neurogenesis in the hippocampus was significantly reduced. Levels of mRNA encoding pro-inflammatory markers IL-6, TNFα, and IL-1β were higher in male MSD offspring, whereas levels of anti-inflammatory markers Arg1, IL-4, and IL-10 were lower, as was PPARγ expression in the hippocampus. PPARγ activation by pioglitazone (30 mg/kg/day, i.p., 7 d) mitigated these negative effects of MSD, rescuing hippocampal neurogenesis and improving cognitive function. The PPARγ inhibitor GW9662 (1 mg/kg/day, i.p., 7 d) eliminated the effects of pioglitazone. Conditioned medium from pioglitazone-treated microglia promoted proliferation and differentiation of neural progenitor cells. These results suggest that MSD-induced deficits in spatial learning and memory can be ameliorated through PPARγ-dependent modulation of microglial phenotypes.
Collapse
Affiliation(s)
- Yue Han
- School of Life Science and Technology, Center for Informational Biology, University of Electronic Science and Technology of China, Chengdu 610054, China
| | - Jiutai Wang
- School of Life Science and Technology, Center for Informational Biology, University of Electronic Science and Technology of China, Chengdu 610054, China
| | - Qiuying Zhao
- School of Life Science and Technology, Center for Informational Biology, University of Electronic Science and Technology of China, Chengdu 610054, China
| | - Xiaofang Xie
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Rui Song
- School of Life Science and Technology, Center for Informational Biology, University of Electronic Science and Technology of China, Chengdu 610054, China
| | - Ying Xiao
- School of Life Science and Technology, Center for Informational Biology, University of Electronic Science and Technology of China, Chengdu 610054, China
| | - Xixi Kang
- School of Life Science and Technology, Center for Informational Biology, University of Electronic Science and Technology of China, Chengdu 610054, China
| | - Lijuan Zhang
- School of Life Science and Technology, Center for Informational Biology, University of Electronic Science and Technology of China, Chengdu 610054, China
| | - Yue Zhang
- School of Life Science and Technology, Center for Informational Biology, University of Electronic Science and Technology of China, Chengdu 610054, China
| | - Cheng Peng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Zili You
- School of Life Science and Technology, Center for Informational Biology, University of Electronic Science and Technology of China, Chengdu 610054, China.
| |
Collapse
|
44
|
Mondelli V, Vernon AC. From early adversities to immune activation in psychiatric disorders: the role of the sympathetic nervous system. Clin Exp Immunol 2020; 197:319-328. [PMID: 31319436 DOI: 10.1111/cei.13351] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/14/2019] [Indexed: 02/07/2023] Open
Abstract
Increased peripheral levels of cytokines and central microglial activation have been reported in patients with psychiatric disorders. The degree of both innate and adaptive immune activation is also associated with worse clinical outcomes and poor treatment response in these patients. Understanding the possible causes and mechanisms leading to this immune activation is therefore an important and necessary step for the development of novel and more effective treatment strategies for these patients. In this work, we review the evidence of literature pointing to childhood trauma as one of the main causes behind the increased immune activation in patients with psychiatric disorders. We then discuss the potential mechanisms linking the experience of early life adversity (ELA) to innate immune activation. Specifically, we focus on the innervation of the bone marrow from sympathetic nervous system (SNS) as a new and emerging mechanism that has the potential to bridge the observed increases in both central and peripheral inflammatory markers in patients exposed to ELA. Experimental studies in laboratory rodents suggest that SNS activation following early life stress exposure causes a shift in the profile of innate immune cells, with an increase in proinflammatory monocytes. In turn, these cells traffic to the brain and influence neural circuitry, which manifests as increased anxiety and other relevant behavioural phenotypes. To date, however, very few studies have been conducted to explore this candidate mechanism in humans. Future research is also needed to clarify whether these pathways could be partially reversible to improve prevention and treatment strategies in the future.
Collapse
Affiliation(s)
- V Mondelli
- King's College London, Institute of Psychiatry Psychology and Neuroscience, Department of Psychological Medicine, London, UK.,NIHR Biomedical Research Centre South London and Maudsley NHS Trust, London, UK
| | - A C Vernon
- King's College London, Institute of Psychiatry Psychology and Neuroscience, Department of Basic and Clinical Neuroscience, London, UK.,MRC Centre for Neurodevelopmental Disorders, King's College London, London, UK
| |
Collapse
|
45
|
Abbink MR, Kotah JM, Hoeijmakers L, Mak A, Yvon-Durocher G, van der Gaag B, Lucassen PJ, Korosi A. Characterization of astrocytes throughout life in wildtype and APP/PS1 mice after early-life stress exposure. J Neuroinflammation 2020; 17:91. [PMID: 32197653 PMCID: PMC7083036 DOI: 10.1186/s12974-020-01762-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 02/27/2020] [Indexed: 02/07/2023] Open
Abstract
Background Early-life stress (ES) is an emerging risk factor for later life development of Alzheimer’s disease (AD). We have previously shown that ES modulates amyloid-beta pathology and the microglial response to it in the APPswe/PS1dE9 mouse model. Because astrocytes are key players in the pathogenesis of AD, we studied here if and how ES affects astrocytes in wildtype (WT) and APP/PS1 mice and how these relate to the previously reported amyloid pathology and microglial profile. Methods We induced ES by limiting nesting and bedding material from postnatal days (P) 2–9. We studied in WT mice (at P9, P30, and 6 months) and in APP/PS1 mice (at 4 and 10 months) (i) GFAP coverage, cell density, and complexity in hippocampus (HPC) and entorhinal cortex (EC); (ii) hippocampal gene expression of astrocyte markers; and (iii) the relationship between astrocyte, microglia, and amyloid markers. Results In WT mice, ES increased GFAP coverage in HPC subregions at P9 and decreased it at 10 months. APP/PS1 mice at 10 months exhibited both individual cell as well as clustered GFAP signals. APP/PS1 mice when compared to WT exhibited reduced total GFAP coverage in HPC, which is increased in the EC, while coverage of the clustered GFAP signal in the HPC was increased and accompanied by increased expression of several astrocytic genes. While measured astrocytic parameters in APP/PS1 mice appear not be further modulated by ES, analyzing these in the context of ES-induced alterations to amyloid pathology and microglial shows alterations at both 4 and 10 months of age. Conclusions Our data suggest that ES leads to alterations to the astrocytic response to amyloid-β pathology.
Collapse
Affiliation(s)
- Maralinde R Abbink
- Brain Plasticity Group, Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Science Park 904, 1098 XH, Amsterdam, The Netherlands
| | - Janssen M Kotah
- Brain Plasticity Group, Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Science Park 904, 1098 XH, Amsterdam, The Netherlands
| | - Lianne Hoeijmakers
- Brain Plasticity Group, Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Science Park 904, 1098 XH, Amsterdam, The Netherlands
| | - Aline Mak
- Brain Plasticity Group, Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Science Park 904, 1098 XH, Amsterdam, The Netherlands
| | - Genevieve Yvon-Durocher
- Brain Plasticity Group, Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Science Park 904, 1098 XH, Amsterdam, The Netherlands
| | - Bram van der Gaag
- Brain Plasticity Group, Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Science Park 904, 1098 XH, Amsterdam, The Netherlands
| | - Paul J Lucassen
- Brain Plasticity Group, Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Science Park 904, 1098 XH, Amsterdam, The Netherlands
| | - Aniko Korosi
- Brain Plasticity Group, Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Science Park 904, 1098 XH, Amsterdam, The Netherlands.
| |
Collapse
|
46
|
Cheng JH, Xu X, Li YB, Zhao XD, Aosai F, Shi SY, Jin CH, Piao JS, Ma J, Piao HN, Jin XJ, Piao LX. Arctigenin ameliorates depression-like behaviors in Toxoplasma gondii-infected intermediate hosts via the TLR4/NF-κB and TNFR1/NF-κB signaling pathways. Int Immunopharmacol 2020; 82:106302. [PMID: 32086097 DOI: 10.1016/j.intimp.2020.106302] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2019] [Revised: 01/27/2020] [Accepted: 02/10/2020] [Indexed: 01/23/2023]
Abstract
Toxoplasma gondii (T. gondii) is a known neurotropic protozoan that remains in the central nervous system and induces neuropsychiatric diseases in intermediate hosts. Arctigenin (AG) is one of the major bioactive lignans of the fruit Arctium lappa L. and has a broad spectrum of pharmacological activities such as neuroprotective, anti-inflammatory and anti-T. gondii effects. However, the effect of AG against depressive behaviors observed in T. gondii-infected hosts has not yet been clarified. In the present study, we analyzed the effects of AG against T. gondii-induced depressive behaviors in intermediate hosts using a microglia cell line (BV2 cells) and brain tissues of BALB/c mice during the acute phase of infection with the RH strain of T. gondii. AG attenuated microglial activation and neuroinflammation via the Toll-like receptor/nuclear factor-kappa B (NF-κB) and tumor necrosis factor receptor 1/NF-κB signaling pathways, followed by up-regulating the dopamine and 5-hydroxytryptamine levels and inhibiting the depression-like behaviors of hosts. AG also significantly decreased the T. gondii burden in mouse brain tissues. In conclusion, we elucidated the effects and underlying molecular mechanisms of AG against depressive behaviors induced by T. gondii infection.
Collapse
Affiliation(s)
- Jia-Hui Cheng
- Key Laboratory of Natural Resources of Changbai Mountain & Functional Molecules, Ministry of Education, College of Pharmacy, Yanbian University, Yanji 133002, Jilin, China
| | - Xiang Xu
- Key Laboratory of Natural Resources of Changbai Mountain & Functional Molecules, Ministry of Education, College of Pharmacy, Yanbian University, Yanji 133002, Jilin, China
| | - Ying-Biao Li
- Department of Neurology, Affliated Hospital of Yanbian University, Yanji 133000, Jilin, China
| | - Xu-Dong Zhao
- Key Laboratory of Natural Resources of Changbai Mountain & Functional Molecules, Ministry of Education, College of Pharmacy, Yanbian University, Yanji 133002, Jilin, China
| | - Fumie Aosai
- Department of Infection and Host Defense, Graduate School of Medicine, Shinshu University, Matsumoto, Japan
| | - Su-Yun Shi
- Key Laboratory of Natural Resources of Changbai Mountain & Functional Molecules, Ministry of Education, College of Pharmacy, Yanbian University, Yanji 133002, Jilin, China
| | - Cheng-Hua Jin
- Key Laboratory of Natural Resources of Changbai Mountain & Functional Molecules, Ministry of Education, College of Pharmacy, Yanbian University, Yanji 133002, Jilin, China
| | - Jing-Shu Piao
- Key Laboratory of Natural Resources of Changbai Mountain & Functional Molecules, Ministry of Education, College of Pharmacy, Yanbian University, Yanji 133002, Jilin, China
| | - Juan Ma
- Key Laboratory of Natural Resources of Changbai Mountain & Functional Molecules, Ministry of Education, College of Pharmacy, Yanbian University, Yanji 133002, Jilin, China
| | - Hu-Nan Piao
- Department of Neurology, Affliated Hospital of Yanbian University, Yanji 133000, Jilin, China.
| | - Xue-Jun Jin
- Key Laboratory of Natural Resources of Changbai Mountain & Functional Molecules, Ministry of Education, College of Pharmacy, Yanbian University, Yanji 133002, Jilin, China.
| | - Lian-Xun Piao
- Key Laboratory of Natural Resources of Changbai Mountain & Functional Molecules, Ministry of Education, College of Pharmacy, Yanbian University, Yanji 133002, Jilin, China. https://orcid.org/0000-0002-8315-5918
| |
Collapse
|
47
|
Microglial Function in the Effects of Early-Life Stress on Brain and Behavioral Development. J Clin Med 2020; 9:jcm9020468. [PMID: 32046333 PMCID: PMC7074320 DOI: 10.3390/jcm9020468] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 01/30/2020] [Accepted: 02/05/2020] [Indexed: 02/06/2023] Open
Abstract
The putative effects of early-life stress (ELS) on later behavior and neurobiology have been widely investigated. Recently, microglia have been implicated in mediating some of the effects of ELS on behavior. In this review, findings from preclinical and clinical literature with a specific focus on microglial alterations induced by the exposure to ELS (i.e., exposure to behavioral stressors or environmental agents and infection) are summarized. These studies were utilized to interpret changes in developmental trajectories based on the time at which the stress occurred, as well as the paradigm used. ELS and microglial alterations were found to be associated with a wide array of deficits including cognitive performance, memory, reward processing, and processing of social stimuli. Four general conclusions emerged: (1) ELS interferes with microglial developmental programs, including their proliferation and death and their phagocytic activity; (2) this can affect neuronal and non-neuronal developmental processes, which are dynamic during development and for which microglial activity is instrumental; (3) the effects are extremely dependent on the time point at which the investigation is carried out; and (4) both pre- and postnatal ELS can prime microglial reactivity, indicating a long-lasting alteration, which has been implicated in behavioral abnormalities later in life.
Collapse
|
48
|
Brydges NM, Reddaway J. Neuroimmunological effects of early life experiences. Brain Neurosci Adv 2020; 4:2398212820953706. [PMID: 33015371 PMCID: PMC7513403 DOI: 10.1177/2398212820953706] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 08/07/2020] [Indexed: 12/18/2022] Open
Abstract
Exposure to adverse experiences during development increases the risk of psychiatric illness later in life. Growing evidence suggests a role for the neuroimmune system in this relationship. There is now substantial evidence that the immune system is critical for normal brain development and behaviour, and responds to environmental perturbations experienced early in life. Severe or chronic stress results in dysregulated neuroimmune function, concomitant with abnormal brain morphology and function. Positive experiences including environmental enrichment and exercise exert the opposite effect, promoting normal brain and immune function even in the face of early life stress. The neuroimmune system may therefore provide a viable target for prevention and treatment of psychiatric illness. This review will briefly summarise the neuroimmune system in brain development and function, and review the effects of stress and positive environmental experiences during development on neuroimmune function. There are also significant sex differences in how the neuroimmune system responds to environmental experiences early in life, which we will briefly review.
Collapse
Affiliation(s)
- Nichola M. Brydges
- Neuroscience and Mental Health Research
Institute, Cardiff University, Cardiff, UK
| | - Jack Reddaway
- Neuroscience and Mental Health Research
Institute, Cardiff University, Cardiff, UK
| |
Collapse
|
49
|
Carlessi AS, Borba LA, Zugno AI, Quevedo J, Réus GZ. Gut microbiota-brain axis in depression: The role of neuroinflammation. Eur J Neurosci 2019; 53:222-235. [PMID: 31785168 DOI: 10.1111/ejn.14631] [Citation(s) in RCA: 144] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 11/18/2019] [Accepted: 11/20/2019] [Indexed: 12/17/2022]
Abstract
Major depressive disorder (MDD) is a psychiatric condition that affects a large number of people in the world, and the treatment existents do not work for all individuals affected. Thus, it is believed that other systems or pathways which regulate brain networks involved in mood regulation and cognition are associated with MDD pathogenesis. Studies in humans and animal models have been shown that in MDD there are increased levels of inflammatory mediators, including cytokines and chemokines in both periphery and central nervous system (CNS). In addition, microglial activation appears to be a key event that triggers changes in signaling cascades and gene expression that would be determinant for the onset of depressive symptoms. Recent researches also point out that changes in the gut microbiota would lead to a systemic inflammation that in different ways would reach the CNS modulating inflammatory pathways and especially the microglia, which could influence responses to treatments. Moreover, pre- and probiotics have shown antidepressant responses and anti-inflammatory effects. This review will focus on studies that show the relationship of inflammation with the gut microbiota-brain axis and its relation with MDD.
Collapse
Affiliation(s)
- Anelise S Carlessi
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, Brazil
| | - Laura A Borba
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, Brazil
| | - Alexandra I Zugno
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, Brazil
| | - João Quevedo
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, Brazil.,Translational Psychiatry Program, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA.,Center of Excellence on Mood Disorders, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA.,Neuroscience Graduate Program, The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, TX, USA
| | - Gislaine Z Réus
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, Brazil
| |
Collapse
|
50
|
Antidepressant Potential of Cinnamic Acids: Mechanisms of Action and Perspectives in Drug Development. Molecules 2019; 24:molecules24244469. [PMID: 31817569 PMCID: PMC6943791 DOI: 10.3390/molecules24244469] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 11/28/2019] [Accepted: 11/28/2019] [Indexed: 01/12/2023] Open
Abstract
Depression is a health problem that compromises the quality of life of the world's population. It has different levels of severity and a symptomatic profile that affects social life and performance in work activities, as well as a high number of deaths in certain age groups. In the search for new therapeutic options for the treatment of this behavioral disorder, the present review describes studies on antidepressant activity of cinnamic acids, which are natural products found in medicinal plants and foods. The description of the animal models used and the mechanisms of action of these compounds are discussed.
Collapse
|