1
|
Campos JMB, de Aguiar da Costa M, de Rezende VL, Costa RRN, Ebs MFP, Behenck JP, de Roch Casagrande L, Venturini LM, Silveira PCL, Réus GZ, Gonçalves CL. Animal Model of Autism Induced by Valproic Acid Combined with Maternal Deprivation: Sex-Specific Effects on Inflammation and Oxidative Stress. Mol Neurobiol 2025; 62:3653-3672. [PMID: 39316355 DOI: 10.1007/s12035-024-04491-z] [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: 09/15/2023] [Accepted: 09/08/2024] [Indexed: 09/25/2024]
Abstract
Autism spectrum disorder (ASD) etiology probably involves a complex interplay of both genetic and environmental risk factors, which includes pre- and perinatal exposure to environmental stressors. Thus, this study evaluated the effects of prenatal exposure to valproic acid (VPA) combined with maternal deprivation (MD) on behavior, oxidative stress parameters, and inflammatory state at a central and systemic level in male and female rats. Pregnant Wistar rats were exposed to VPA during gestation, and the offspring were submitted to MD. Offspring were tested for locomotor and social behavior; rats were euthanized, where the cerebellum, posterior cortex, prefrontal cortex, and peripheric blood were collected for oxidative stress and inflammatory analysis. It was observed that young rats (25-30 days old) exposed only to VPA presented a lower social approach when compared to the control group. VPA + MD rats did not present the same deficit. Female rats exposed to VPA + MD presented oxidative stress in all brain areas analyzed. Male rats in the VPA and VPA + MD groups presented oxidative stress only in the cerebellum. Regarding inflammatory parameters, male rats exposed only to MD exhibited an increase in pro-inflammatory cytokines in the blood and in the cortex total. The same was observed in females exposed only to VPA. Animals exposed to VPA + MD showed no alterations in the cytokines analyzed. In summary, gestational (VPA) and perinatal (MD) insults can affect molecular mechanisms such as oxidative stress and inflammation differently depending on the sex and brain area analyzed. Combined exposition to VPA and MD triggers oxidative stress especially in female brains without evoking an inflammatory response.
Collapse
Affiliation(s)
- José Marcelo Botancin Campos
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), 1105, Criciúma, SC, 88806-000, Brazil
| | - Maiara de Aguiar da Costa
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), 1105, Criciúma, SC, 88806-000, Brazil
| | - Victória Linden de Rezende
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), 1105, Criciúma, SC, 88806-000, Brazil
| | - Rosiane Ronchi Nascimento Costa
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), 1105, Criciúma, SC, 88806-000, Brazil
| | - Maria Fernanda Pedro Ebs
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), 1105, Criciúma, SC, 88806-000, Brazil
| | - João Paulo Behenck
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Laura de Roch Casagrande
- Laboratory of Experimental Physiopathology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Ligia Milanez Venturini
- Laboratory of Experimental Physiopathology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Paulo Cesar Lock Silveira
- Laboratory of Experimental Physiopathology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Gislaine Zilli Réus
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Cinara Ludvig Gonçalves
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), 1105, Criciúma, SC, 88806-000, Brazil.
| |
Collapse
|
2
|
Mohammadi S, Bashghareh A, Karimi-Zandi L, Mokhtari T. Understanding Role of Maternal Separation in Depression, Anxiety,and Pain Behaviour: A Mini Review of Preclinical Research With Focus on Neuroinflammatory Pathways. Int J Dev Neurosci 2025; 85:e70002. [PMID: 39895419 DOI: 10.1002/jdn.70002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 01/14/2025] [Accepted: 01/16/2025] [Indexed: 02/04/2025] Open
Abstract
Contact between mother and child is essential for the proper development of an infant's physiological systems, brain maturation and behavioural outcomes. Early life stress (ELS), which includes factors such as inadequate parental care and childhood abuse, significantly increases the risk of developing neuropsychiatric disorders, including anxiety and depression. This review examines the impact of maternal separation (MS) on depression, anxiety and pain behaviour, with a particular emphasis on neuroinflammatory pathways. Experiences of ELS can adversely affect the maturation of neurotransmitter systems and associated neural circuits that are crucial for processing painful stimuli and regulating anxiety and depression. Stressful experiences trigger inflammatory processes in the brain, initiating immune responses in neural cells and stimulating the production of pro-inflammatory cytokines. In mammals, MS serves as a significant stressor that activates the hypothalamic-pituitary-adrenal (HPA) axis and other stress-related systems, leading to increased immune challenges and heightened pain sensitivity in adulthood due to systemic inflammation. Key inflammatory mediators, such as IL-1β, IL-6 and TNF-α, play critical roles in the development of pathological pain, while the activation of microglia releases inflammatory mediators that contribute to neurological dysfunction and the pathophysiology of stress, depression and anxiety. Moreover, therapeutics targeting oxidative stress and inflammation have shown promise in alleviating affective disorders following MS. This review discusses potential pathways, with a primary focus on neuroinflammatory mechanisms and the therapeutic strategies that may mitigate the adverse effects of MS. There is a pressing need for further research to elucidate the underlying pathways and identify effective interventions to improve mental health outcomes in individuals affected by MS.
Collapse
Affiliation(s)
- Shima Mohammadi
- Department of Neuroscience, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Alieh Bashghareh
- Department of Anatomy, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Leila Karimi-Zandi
- School of Advanced Technologies in Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Tahmineh Mokhtari
- Department of Entomology and Nematology and UCD Comprehensive Cancer Center, University of California, Davis, California, USA
| |
Collapse
|
3
|
Sgro M, Kodila Z, Salberg S, Li CN, Smith MJ, Freeman J, Vlassopoulos E, Harris S, Shultz SR, Yamakawa GR, Noel M, Mychasiuk R. Exposure to perinatal trauma modifies nociception and gene expression in the prefrontal cortex and hypothalamus of adolescent rats. THE JOURNAL OF PAIN 2024; 28:104762. [PMID: 39730020 DOI: 10.1016/j.jpain.2024.104762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 12/12/2024] [Accepted: 12/18/2024] [Indexed: 12/29/2024]
Abstract
The perinatal period encompasses a critical window for neurodevelopment that renders the brain highly responsive to experience. Trauma, such as intimate partner violence (IPV) and early life stress/neglect, during this period negatively affects physical and mental health outcomes, including increasing ones risk for chronic pain. Although epigenetic programming likely contributes, the mechanisms that drive the relationship between perinatal trauma and adverse health outcomes, are not fully understood. Therefore, we explored the relationship between perinatal trauma (in utero exposure to IPV and/or early life neglect) and socio-emotional functioning, nociceptive sensitivity, and transcriptomic changes within the prefrontal cortex (PFC) and hypothalamus in dams and their adolescent offspring. Rat dams were randomly assigned to an IPV (i.e., combined mild traumatic brain injury and strangulation) or sham procedure during pregnancy. Following birth, offspring were subsequently assigned the early life neglect or control paradigm. In adolescence, offspring received a plantar incision or sham injury. Perinatal trauma altered nociception and emotional functioning in a sex-dependent manner when combined with the surgical procedure. We identified transcriptomic changes related to DNA transcription and expression within the PFC and hypothalamus of the dams. Examination of the offspring transcriptome highlighted impairment in immune regulation, dysfunction in stress-reactivity, as well as microglia activation. We also identified altered expression of genes associated with chronic pain. This demonstrates that perinatal trauma modifies offspring behaviour, including nociceptive sensitivity. We provide insight into the mechanisms that contribute to the chronification of pain, thereby informing future research targeted at the generation of prevention and therapeutic strategies. PERSPECTIVE: Perinatal trauma impaired cognitive, socio-emotional, and pain processing in offspring, while also inducing changes in gene expression, in both mothers and offspring. The findings highlight possible mechanisms responsible for intergenerational transmission of risk for chronic pain and provide targets for therapeutics which could potentially reverse perinatal-trauma induced epigenetic change.
Collapse
Affiliation(s)
- Marissa Sgro
- Department of Neuroscience, School of Translational Medicine, Monash University,Melbourne, Victoria, Australia
| | - Zoe Kodila
- Department of Neuroscience, School of Translational Medicine, Monash University,Melbourne, Victoria, Australia
| | - Sabrina Salberg
- Department of Neuroscience, School of Translational Medicine, Monash University,Melbourne, Victoria, Australia
| | - Crystal N Li
- Department of Neuroscience, School of Translational Medicine, Monash University,Melbourne, Victoria, Australia
| | - Madeleine J Smith
- Department of Neuroscience, School of Translational Medicine, Monash University,Melbourne, Victoria, Australia
| | - James Freeman
- Department of Neuroscience, School of Translational Medicine, Monash University,Melbourne, Victoria, Australia
| | - Elaina Vlassopoulos
- Department of Neuroscience, School of Translational Medicine, Monash University,Melbourne, Victoria, Australia
| | - Sydney Harris
- Department of Neuroscience, School of Translational Medicine, Monash University,Melbourne, Victoria, Australia
| | - Sandy R Shultz
- Department of Neuroscience, School of Translational Medicine, Monash University,Melbourne, Victoria, Australia; Centre for Trauma and Mental Health Research, Vancouver Island University, Nanaimo, B.C., Canada
| | - Glenn R Yamakawa
- Department of Neuroscience, School of Translational Medicine, Monash University,Melbourne, Victoria, Australia
| | - Melanie Noel
- Department of Psychology, Alberta Children's Hospital, Hotchkiss Brain Institute, University of Calgary,AB, Canada
| | - Richelle Mychasiuk
- Department of Neuroscience, School of Translational Medicine, Monash University,Melbourne, Victoria, Australia.
| |
Collapse
|
4
|
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
|
5
|
Fanikos M, Kohn SA, Stamato R, Brenhouse HC, Gildawie KR. Impacts of age and environment on postnatal microglial activity: Consequences for cognitive function following early life adversity. PLoS One 2024; 19:e0306022. [PMID: 38917075 PMCID: PMC11198844 DOI: 10.1371/journal.pone.0306022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 06/10/2024] [Indexed: 06/27/2024] Open
Abstract
Early life adversity (ELA) increases the likelihood of later-life neuropsychiatric disorders and cognitive dysfunction. Importantly, ELA, neuropsychiatric disorders, and cognitive deficits all involve aberrant immune signaling. Microglia are the primary neuroimmune cells and regulate brain development. Microglia are particularly sensitive to early life insults, which can program their responses to future challenges. ELA in the form of maternal separation (MS) in rats alters later-life microglial morphology and the inflammatory profile of the prefrontal cortex, a region important for cognition. However, the role of microglial responses during MS in the development of later cognition is not known. Therefore, here we aimed to determine whether the presence of microglia during MS mediates long-term impacts on adult working memory. Clodronate liposomes were used to transiently deplete microglia from the brain, while empty liposomes were used as a control. We hypothesized that if microglia mediate the long-term impacts of ELA on working memory in adulthood, then depleting microglia during MS would prevent these deficits. Importantly, microglial function shifts throughout the neonatal period, so an exploratory investigation assessed whether depletion during the early versus late neonatal period had different effects on adult working memory. Surprisingly, empty liposome treatment during the early, but not late, postnatal period induced microglial activity changes that compounded with MS to impair working memory in females. In contrast, microglial depletion later in infancy impaired later life working memory in females, suggesting that microglial function during late infancy plays an important role in the development of cognitive function. Together, these findings suggest that microglia shift their sensitivity to early life insults across development. Our findings also highlight the potential for MS to impact some developmental processes only when compounded with additional neuroimmune challenges in a sex-dependent manner.
Collapse
Affiliation(s)
- Michaela Fanikos
- Department of Psychology, Northeastern University, Boston, Massachusetts, United States of America
| | - Skylar A. Kohn
- Department of Psychology, Northeastern University, Boston, Massachusetts, United States of America
| | - Rebecca Stamato
- Department of Psychology, Northeastern University, Boston, Massachusetts, United States of America
| | - Heather C. Brenhouse
- Department of Psychology, Northeastern University, Boston, Massachusetts, United States of America
| | - Kelsea R. Gildawie
- Department of Psychology, Northeastern University, Boston, Massachusetts, United States of America
| |
Collapse
|
6
|
Bris ÁG, MacDowell KS, Ulecia-Morón C, Martín-Hernández D, Moreno B, Madrigal JLM, García-Bueno B, Caso JR, Leza JC. Differential regulation of innate immune system in frontal cortex and hippocampus in a "double-hit" neurodevelopmental model in rats. Neurotherapeutics 2024; 21:e00300. [PMID: 38241165 PMCID: PMC10903097 DOI: 10.1016/j.neurot.2023.10.010] [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: 10/17/2023] [Accepted: 10/28/2023] [Indexed: 01/21/2024] Open
Abstract
Neurodevelopmental disorders (NDs) are neuropsychiatric conditions affecting central nervous system development, characterized by cognitive and behavioural alterations. Inflammation has been recently linked to NDs. Animal models are essential for understanding their pathophysiology and identifying therapeutic targets. Double-hit models can reproduce neurodevelopmental and neuroinflammatory impairments. Sixty-seven newborn rats were assigned to four groups: Control, Maternal deprivation (MD, 24-h-deprivation), Isolation (Iso, 5 weeks), and Maternal deprivation + Isolation (MD + Iso, also known as double-hit). Cognitive dysfunction was assessed using behavioural tests. Inflammasome, MAPKs, and TLRs inflammatory elements expression in the frontal cortex (FC) and hippocampus (HP) was analysed through western blot and qRT-PCR. Oxidative/nitrosative (O/N) evaluation and corticosterone levels were measured in plasma samples. Double-hit group was affected in executive and working memory. Most inflammasomes and TLRs inflammatory responses were increased in FC compared to the control group, whilst MAPKs were downregulated. Conversely, hippocampal inflammasome and inflammatory components were reduced after the double-hit exposure, while MAPKs were elevated. Our findings reveal differential regulation of innate immune system components in FC and HP in the double-hit group. Further investigations on MAPKs are necessary to understand their role in regulating HP neuroinflammatory status, potentially linking our MAPKs results to cognitive impairments through their proliferative and anti-inflammatory activity.
Collapse
Affiliation(s)
- Álvaro G Bris
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid. Centro de Investigación Biomédica en Red de Salud Mental, Instituto de Salud Carlos III (CIBERSAM, ISCIII). Instituto de Investigación Sanitaria Hospital 12 de Octubre (Imas12) e Instituto Universitario de Investigación en Neuroquímica (IUIN), Spain
| | - Karina S MacDowell
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid. Centro de Investigación Biomédica en Red de Salud Mental, Instituto de Salud Carlos III (CIBERSAM, ISCIII). Instituto de Investigación Sanitaria Hospital 12 de Octubre (Imas12) e Instituto Universitario de Investigación en Neuroquímica (IUIN), Spain
| | - Cristina Ulecia-Morón
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid. Centro de Investigación Biomédica en Red de Salud Mental, Instituto de Salud Carlos III (CIBERSAM, ISCIII). Instituto de Investigación Sanitaria Hospital 12 de Octubre (Imas12) e Instituto Universitario de Investigación en Neuroquímica (IUIN), Spain
| | - David Martín-Hernández
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid. Centro de Investigación Biomédica en Red de Salud Mental, Instituto de Salud Carlos III (CIBERSAM, ISCIII). Instituto de Investigación Sanitaria Hospital 12 de Octubre (Imas12) e Instituto Universitario de Investigación en Neuroquímica (IUIN), Spain
| | - Beatriz Moreno
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid. Centro de Investigación Biomédica en Red de Salud Mental, Instituto de Salud Carlos III (CIBERSAM, ISCIII). Instituto de Investigación Sanitaria Hospital 12 de Octubre (Imas12) e Instituto Universitario de Investigación en Neuroquímica (IUIN), Spain
| | - José L M Madrigal
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid. Centro de Investigación Biomédica en Red de Salud Mental, Instituto de Salud Carlos III (CIBERSAM, ISCIII). Instituto de Investigación Sanitaria Hospital 12 de Octubre (Imas12) e Instituto Universitario de Investigación en Neuroquímica (IUIN), Spain
| | - Borja García-Bueno
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid. Centro de Investigación Biomédica en Red de Salud Mental, Instituto de Salud Carlos III (CIBERSAM, ISCIII). Instituto de Investigación Sanitaria Hospital 12 de Octubre (Imas12) e Instituto Universitario de Investigación en Neuroquímica (IUIN), Spain
| | - Javier R Caso
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid. Centro de Investigación Biomédica en Red de Salud Mental, Instituto de Salud Carlos III (CIBERSAM, ISCIII). Instituto de Investigación Sanitaria Hospital 12 de Octubre (Imas12) e Instituto Universitario de Investigación en Neuroquímica (IUIN), Spain
| | - Juan C Leza
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid. Centro de Investigación Biomédica en Red de Salud Mental, Instituto de Salud Carlos III (CIBERSAM, ISCIII). Instituto de Investigación Sanitaria Hospital 12 de Octubre (Imas12) e Instituto Universitario de Investigación en Neuroquímica (IUIN), Spain.
| |
Collapse
|
7
|
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
|
8
|
Liu R, Gong Y, Xia C, Cao Y, Zhao C, Zhou M. Itaconate: A promising precursor for treatment of neuroinflammation associated depression. Biomed Pharmacother 2023; 167:115521. [PMID: 37717531 DOI: 10.1016/j.biopha.2023.115521] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 09/11/2023] [Accepted: 09/14/2023] [Indexed: 09/19/2023] Open
Abstract
Neuroinflammation triggers the production of inflammatory factors, influences neuron generation and synaptic plasticity, thus playing an important role in the pathogenesis of depression and becoming an important direction of depression prevention and treatment. Itaconate is a metabolite secreted by macrophages in immunomodulatory responses, that has potent immunomodulatory effects and has been proven to exert anti-inflammatory effects in a variety of diseases. Microglia are mononuclear macrophages that reside in the central nervous system (CNS), and may be the source of endogenous itaconate in the brain. Itaconate can directly inhibit succinate dehydrogenase (SDH), reduce the production of NOD-like receptor thermal protein domain associated protein 3 (NLRP3), activate nuclear factor erythroid-2 related factor 2 (Nrf2), and block glycolysis, and thereby improving the depressive symptoms associated with the above mechanisms. Notably, itaconate also indirectly ameliorates the depressive symptoms associated with some inflammatory diseases. With the optimization of the structure and the development of new delivery systems, the application value and therapeutic potential of itaconate have been significantly improved. Dimethyl itaconate (DI) and 4-octyl itaconate (4-OI), cell-permeable derivatives of itaconate, are more suitable for crossing the blood-brain barrier (BBB), exhibiting therapeutic effects in the research of multiple diseases. This article provides an overview of the immunomodulatory effects of itaconate and its potential therapeutic efficacy in inflammatory depression, focusing on the promising application of itaconate as a precursor of antidepressants.
Collapse
Affiliation(s)
- Ruisi Liu
- Shanghai Traditional Chinese Medicine Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200082, China; Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yueling Gong
- Shanghai Traditional Chinese Medicine Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200082, China; Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Chenyi Xia
- Department of Physiology, School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yemin Cao
- Shanghai Traditional Chinese Medicine Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200082, China
| | - Cheng Zhao
- Shanghai Traditional Chinese Medicine Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200082, China.
| | - Mingmei Zhou
- Shanghai Traditional Chinese Medicine Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200082, China; Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
9
|
Li X, Wang S, Duan S, Long L, Zhuo L, Peng Y, Xiong Y, Li S, Peng X, Yan Y, Wang Z, Jiang W. Exploring the Therapeutic Effects of Multifunctional N-Salicylic Acid Tryptamine Derivative against Parkinson's Disease. ACS OMEGA 2023; 8:28910-28923. [PMID: 37576637 PMCID: PMC10413456 DOI: 10.1021/acsomega.3c04277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 07/14/2023] [Indexed: 08/15/2023]
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease worldwide. Neuroinflammation and oxidative stress play an important role in the whole course of PD, which have been the focus of PD drug development. In our previous research, a series of N-salicylic acid tryptamine derivatives were synthesized, and the biological evaluation showed that the compound LZWL02003 has good anti-neuroinflammatory activity and displayed great therapeutic potency for neurodegenerative disease models. In this work, the neuroprotective efficiency of LZWL02003 against PD in vitro and in vivo has been explored. It was found that LZWL02003 could protect human neuron cells SH-SY5Y from MPP+-induced neuronal damage by inhibiting ROS generation, mitochondrial dysfunction, and cellular apoptosis. Moreover, LZWL02003 could improve cognition, memory, learning, and athletic ability in a rotenone-induced PD rat model. In general, our study has demonstrated that LZWL02003 has good activity against PD in in vitro and in vivo experiments, which can potentially be developed into a therapeutic candidate for PD.
Collapse
Affiliation(s)
- Xuelin Li
- School
of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
- The
First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
| | - Shuzhi Wang
- School
of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
| | - Shan Duan
- The
First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
| | - Lin Long
- School
of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
| | - Linsheng Zhuo
- School
of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
| | - Yan Peng
- School
of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
| | - Yongxia Xiong
- School
of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
| | - Shuang Li
- School
of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
| | - Xue Peng
- School
of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
| | - Yiguo Yan
- The
First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
| | - Zhen Wang
- School
of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
- The
First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
| | - Weifan Jiang
- School
of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
| |
Collapse
|
10
|
Cruz KLO, Salla DH, Oliveira MP, Silva LE, Vedova LMD, Mendes TF, Bressan CBC, Silva MR, Santos SML, Soares HJ, Mendes RL, Vernke CN, Silva MG, Laurentino AOM, Medeiros FD, Vilela TC, Lemos I, Bitencourt RM, Réus GZ, Streck EL, Mello AH, Rezin GT. Energy metabolism and behavioral parameters in female mice subjected to obesity and offspring deprivation stress. Behav Brain Res 2023; 451:114526. [PMID: 37271313 DOI: 10.1016/j.bbr.2023.114526] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 05/20/2023] [Accepted: 06/01/2023] [Indexed: 06/06/2023]
Abstract
This study aimed to evaluate the behavioral and energy metabolism parameters in female mice subjected to obesity and offspring deprivation (OD) stress. Eighty female Swiss mice, 40 days old, were weighed and divided into two groups: Control group (control diet, n = 40) and Obese group (high-fat diet, n = 40), for induction of the animal model of obesity, the protocol was based on the consumption of a high-fat diet and lasted 8 weeks. Subsequently, the females were subjected to pregnancy, after the birth of the offspring, were divided again into the following groups (n = 20): Control non-deprived (ND), Control + OD, Obese ND, and Obese + OD, for induction of the stress protocol by OD. After the offspring were 21 days old, weaning was performed and the dams were subjected to behavioral tests. The animals were humanely sacrificed, the brain was removed, and brain structures were isolated to assess energy metabolism. Both obesity and OD led to anhedonia in the dams. It was shown that the structures most affected by obesity and OD are the hypothalamus and hippocampus, as evidenced by the mitochondrial dysfunction found in these structures. When analyzing the groups separately, it was observed that OD led to more pronounced mitochondrial damage; however, the association of obesity with OD, as well as obesity alone, also generated damage. Thus, it is concluded that obesity and OD lead to anhedonia in animals and to mitochondrial dysfunction in the hypothalamus and hippocampus, which may lead to losses in feeding control and cognition of the dams.
Collapse
Affiliation(s)
- Kenia L O Cruz
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Brazil
| | - Daniele H Salla
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Brazil
| | - Mariana P Oliveira
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Brazil
| | - Larissa E Silva
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Brazil.
| | - Larissa M D Vedova
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Brazil
| | - Talita F Mendes
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Brazil
| | - Catarina B C Bressan
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Brazil
| | - Mariella R Silva
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Brazil
| | - Sheila M L Santos
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Brazil
| | - Hevylin J Soares
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Brazil
| | - Rayane L Mendes
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Brazil
| | - Camila N Vernke
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Brazil
| | - Marina G Silva
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Brazil
| | - Ana O M Laurentino
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Brazil
| | - Fabiana D Medeiros
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Brazil
| | - Thais C Vilela
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Brazil
| | - Isabela Lemos
- Laboratory of Experimental Neurology, Postgraduate Program in Health Sciences, University of the Extreme South of Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Rafael M Bitencourt
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Brazil
| | - Gislaine Z Réus
- Translational Psychiatry Laboratory, Postgraduate Program in Health Sciences, University of the Extreme South of Santa Catarina (UNESC), Criciúma, Brazil
| | - Emilio L Streck
- Laboratory of Experimental Neurology, Postgraduate Program in Health Sciences, University of the Extreme South of Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Aline H Mello
- Department of Pediatrics, The University of Texas Medical Branch, Galveston, TX, USA
| | - Gislaine T Rezin
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Brazil
| |
Collapse
|
11
|
Shin SH, Kim YK. Early Life Stress, Neuroinflammation, and Psychiatric Illness of Adulthood. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1411:105-134. [PMID: 36949308 DOI: 10.1007/978-981-19-7376-5_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/24/2023]
Abstract
Stress exposure during early stages of life elevates the risk of developing psychopathologies and psychiatric illness in later life. The brain and immune system are not completely developed by birth and therefore continue develop after birth; this post birth development is influenced by several psychosocial factors; hence, early life stress (ELS) exposure can alter brain structural development and function. A growing number of experimental animal and observational human studies have investigated the link between ELS exposure and increased risk of psychopathology through alternations in the immune system, by evaluating inflammation biomarkers. Recent studies, including brain imaging, have also shed light on the mechanisms by which both the innate and adaptive immune systems interact with neural circuits and neurotransmitters, which affect psychopathology. Herein, we discuss the link between the experience of stress in early life and lifelong alterations in the immune system, which subsequently lead to the development of various psychiatric illnesses.
Collapse
Affiliation(s)
- Sang Ho Shin
- Department of Psychiatry, College of Medicine, Korea University Ansan Hospital, Korea University, Ansan, Republic of Korea
| | - Yong-Ku Kim
- Department of Psychiatry, College of Medicine, Korea University Ansan Hospital, Korea University, Ansan, Republic of Korea.
| |
Collapse
|
12
|
Ji N, Lei M, Chen Y, Tian S, Li C, Zhang B. How Oxidative Stress Induces Depression? ASN Neuro 2023; 15:17590914231181037. [PMID: 37331994 DOI: 10.1177/17590914231181037] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/20/2023] Open
Abstract
Depression increasingly affects a wide range and a large number of people worldwide, both physically and psychologically, which makes it a social problem requiring prompt attention and management. Accumulating clinical and animal studies have provided us with substantial insights of disease pathogenesis, especially central monoamine deficiency, which considerably promotes antidepressant research and clinical treatment. The first-line antidepressants mainly target the monoamine system, whose drawbacks mainly include slow action and treatment resistant. The novel antidepressant esketamine, targeting on central glutamatergic system, rapidly and robustly alleviates depression (including treatment-resistant depression), whose efficiency is shadowed by potential addictive and psychotomimetic side effects. Thus, exploring novel depression pathogenesis is necessary, for seeking more safe and effective therapeutic methods. Emerging evidence has revealed vital involvement of oxidative stress (OS) in depression, which inspires us to pursue antioxidant pathway for depression prevention and treatment. Fully uncovering the underlying mechanisms of OS-induced depression is the first step towards the avenue, thus we summarize and expound possible downstream pathways of OS, including mitochondrial impairment and related ATP deficiency, neuroinflammation, central glutamate excitotoxicity, brain-derived neurotrophic factor/tyrosine receptor kinase B dysfunction and serotonin deficiency, the microbiota-gut-brain axis disturbance and hypothalamic-pituitary-adrenocortical axis dysregulation. We also elaborate on the intricate interactions between the multiple aspects, and molecular mechanisms mediating the interplay. Through reviewing the related research progress in the field, we hope to depict an integral overview of how OS induces depression, in order to provide fresh ideas and novel targets for the final goal of efficient treatment of the disease.
Collapse
Affiliation(s)
- Na Ji
- The School of Public Health, Faculty of Basic Medical Sciences, Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin Guangxi, China
| | - Mengzhu Lei
- The School of Public Health, Faculty of Basic Medical Sciences, Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin Guangxi, China
| | - Yating Chen
- The School of Public Health, Faculty of Basic Medical Sciences, Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin Guangxi, China
| | - Shaowen Tian
- The School of Public Health, Faculty of Basic Medical Sciences, Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin Guangxi, China
| | - Chuanyu Li
- The School of Public Health, Faculty of Basic Medical Sciences, Guilin Medical University, Guilin Guangxi, China
| | - Bo Zhang
- The School of Public Health, Faculty of Basic Medical Sciences, Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin Guangxi, China
| |
Collapse
|
13
|
Polydatin Prevents Neuroinflammation and Relieves Depression via Regulating Sirt1/HMGB1/NF-κB Signaling in Mice. Neurotox Res 2022; 40:1393-1404. [PMID: 35986876 DOI: 10.1007/s12640-022-00553-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 07/22/2022] [Accepted: 07/29/2022] [Indexed: 12/21/2022]
|
14
|
Döhne N, Falck A, Janach GMS, Byvaltcev E, Strauss U. Interferon-γ augments GABA release in the developing neocortex via nitric oxide synthase/soluble guanylate cyclase and constrains network activity. Front Cell Neurosci 2022; 16:913299. [PMID: 36035261 PMCID: PMC9401097 DOI: 10.3389/fncel.2022.913299] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 07/13/2022] [Indexed: 11/13/2022] Open
Abstract
Interferon-γ (IFN-γ), a cytokine with neuromodulatory properties, has been shown to enhance inhibitory transmission. Because early inhibitory neurotransmission sculpts functional neuronal circuits, its developmental alteration may have grave consequences. Here, we investigated the acute effects of IFN-γ on γ-amino-butyric acid (GABA)ergic currents in layer 5 pyramidal neurons of the somatosensory cortex of rats at the end of the first postnatal week, a period of GABA-dependent cortical maturation. IFN-γ acutely increased the frequency and amplitude of spontaneous/miniature inhibitory postsynaptic currents (s/mIPSC), and this could not be reversed within 30 min. Neither the increase in amplitude nor frequency of IPSCs was due to upregulated interneuron excitability as revealed by current clamp recordings of layer 5 interneurons labeled with VGAT-Venus in transgenic rats. As we previously reported in more mature animals, IPSC amplitude increase upon IFN-γ activity was dependent on postsynaptic protein kinase C (PKC), indicating a similar activating mechanism. Unlike augmented IPSC amplitude, however, we did not consistently observe an increased IPSC frequency in our previous studies on more mature animals. Focusing on increased IPSC frequency, we have now identified a different activating mechanism-one that is independent of postsynaptic PKC but is dependent on inducible nitric oxide synthase (iNOS) and soluble guanylate cyclase (sGC). In addition, IFN-γ shifted short-term synaptic plasticity toward facilitation as revealed by a paired-pulse paradigm. The latter change in presynaptic function was not reproduced by the application of a nitric oxide donor. Functionally, IFN-γ-mediated alterations in GABAergic transmission overall constrained early neocortical activity in a partly nitric oxide-dependent manner as revealed by microelectrode array field recordings in brain slices analyzed with a spike-sorting algorithm. In summary, with IFN-γ-induced, NO-dependent augmentation of spontaneous GABA release, we have here identified a mechanism by which inflammation in the central nervous system (CNS) plausibly modulates neuronal development.
Collapse
Affiliation(s)
- Noah Döhne
- Institute of Cell Biology and Neurobiology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Alice Falck
- Institute of Cell Biology and Neurobiology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Gabriel M. S. Janach
- Institute of Cell Biology and Neurobiology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Egor Byvaltcev
- Institute of Cell Biology and Neurobiology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
- Institute of Neuroscience, Lobachevsky State, University of Nizhny Novgorod, Nizhny Novgorod, Russia
| | - Ulf Strauss
- Institute of Cell Biology and Neurobiology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| |
Collapse
|
15
|
Kumar Palepu MS, Dandekar MP. Remodeling of microbiota gut-brain axis using psychobiotics in depression. Eur J Pharmacol 2022; 931:175171. [PMID: 35926568 DOI: 10.1016/j.ejphar.2022.175171] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 07/14/2022] [Accepted: 07/21/2022] [Indexed: 12/11/2022]
Abstract
Depression is a multifaceted psychiatric disorder mainly orchestrated by dysfunction of neuroendocrine, neurochemical, immune, and metabolic systems. The interconnection of gut microbiota perturbation with the central nervous system disorders has been well documented in recent times. Indeed, alteration of commensal intestinal microflora is noted in several psychiatric disorders such as anxiety and depression, which are presumed to be routed through the enteric nervous system, autonomic nervous system, endocrine, and immune system. This review summarises the new mechanisms underlying the crosstalk between gut microbiota and brain involved in the management of depression. Depression-induced changes in the commensal intestinal microbiota are majorly linked with the disruption of gut integrity, hyperinflammation, and modulation of short-chain fatty acids, neurotransmitters, kynurenine metabolites, endocannabinoids, brain-derived neurotropic factors, hypothalamic-pituitary-adrenal axis, and gut peptides. The restoration of gut microbiota with prebiotics, probiotics, postbiotics, synbiotics, and fermented foods (psychobiotics) has gained a considerable attention for the management of depression. Recent evidence also propose the role of gut microbiota in the process of treatment-resistant depression. Thus, remodeling of the microbiota-gut-brain axis using psychobiotics appears to be a promising therapeutic approach for the reversal of psychiatric disorders, and it is imperative to decipher the underlying mechanisms for gut-brain crosstalk.
Collapse
Affiliation(s)
- Mani Surya Kumar Palepu
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana, India
| | - Manoj P Dandekar
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana, India.
| |
Collapse
|
16
|
Effects of early life stress on brain cytokines: A systematic review and meta-analysis of rodent studies. Neurosci Biobehav Rev 2022; 139:104746. [PMID: 35716876 DOI: 10.1016/j.neubiorev.2022.104746] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 05/13/2022] [Accepted: 06/11/2022] [Indexed: 12/21/2022]
Abstract
Exposure to early life stress (ELS) may lead to long-lasting neurobiological and behavioral impairments. Alterations in the immune system and neuroinflammatory state induced by ELS exposure are considered risk factors for developing psychiatric disorders. Here, we performed a systematic review and meta-analysis of rodent studies investigating the short and long-term effects of ELS exposure on anti and pro-inflammatory cytokines in brain tissues. Our analysis shows that animals exposed to ELS present an increase in pro-inflammatory cytokines IL-1β, IL-6, and TNF-α. On the other hand, no alteration was observed in the anti-inflammatory cytokine IL-10. Meta-regression revealed that alterations were more prominent in the hippocampus of adult animals that were exposed to more extended periods of ELS. These inflammatory effects were not permanent since few alterations were identified in aged animals. Our findings suggest that ELS exposure alters pro-inflammatory cytokines expression and may act as a primer for a secondary challenge that may induce lifelong immune alterations. Moreover, the actual evidence is insufficient to comprehend the relationship between anti-inflammatory cytokines and ELS fully.
Collapse
|
17
|
da Cruz KLDO, Salla DH, de Oliveira MP, da Silva LE, Dela Vedova LM, Mendes TF, Bressan CBC, Costa AB, da Silva MR, Réus GZ, de Mello AH, Rezin GT. The impact of obesity-related neuroinflammation on postpartum depression: A narrative review. Int J Dev Neurosci 2022; 82:375-384. [PMID: 35595536 DOI: 10.1002/jdn.10198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 04/29/2022] [Accepted: 05/16/2022] [Indexed: 11/10/2022] Open
Abstract
Obesity is currently one of the most serious health problems, affecting 13% of the world's adult population. Obesity is characterized by persistent low-grade chronic inflammation that assumes systemic proportions and triggers several associated metabolic diseases. Furthermore, obesity has been associated with an increased occurrence of central disorders such as impaired cognitive function, reward system dysfunction, and depression. In summary, there is a quantitative reduction in the release of neurotransmitters in depression. Postsynaptic cells capture lower concentrations of neurotransmitters, which leads to a functional reduction in the central nervous system (CNS). Globally, approximately 15-65% of women experience depressive symptoms during pregnancy, depending on their location. Depressive symptoms persist in some women, leading to postpartum depression (PPD). Thus, obesity may be considered a risk factor for PPD development. This study aimed to synthesize studies on the impact of obesity-related neuroinflammation and PPD. We conducted a narrative review of the relevant literature. The search was performed in electronic databases, specifically PubMed, selecting articles in English published from 2014 to 2021 using the narrative review methodology.
Collapse
Affiliation(s)
- Kenia Lourdes de Oliveira da Cruz
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Graduate Program in Health Sciences, University of Southern Santa Catarina, Tubarao, Brazil
| | - Daniele Hendler Salla
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Graduate Program in Health Sciences, University of Southern Santa Catarina, Tubarao, Brazil
| | - Mariana Pacheco de Oliveira
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Graduate Program in Health Sciences, University of Southern Santa Catarina, Tubarao, Brazil
| | - Larissa Espindola da Silva
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Graduate Program in Health Sciences, University of Southern Santa Catarina, Tubarao, Brazil
| | - Larissa Marques Dela Vedova
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Graduate Program in Health Sciences, University of Southern Santa Catarina, Tubarao, Brazil
| | - Talita Farias Mendes
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Graduate Program in Health Sciences, University of Southern Santa Catarina, Tubarao, Brazil
| | - Catarina Barbosa Chaves Bressan
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Graduate Program in Health Sciences, University of Southern Santa Catarina, Tubarao, Brazil
| | - Ana Beatriz Costa
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Graduate Program in Health Sciences, University of Southern Santa Catarina, Tubarao, Brazil
| | - Mariella Reinol da Silva
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Graduate Program in Health Sciences, University of Southern Santa Catarina, Tubarao, Brazil
| | - Gislaine Zilli Réus
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina, Criciuma, Brazil
| | - Aline Haas de Mello
- Department of Pediatrics, The University of Texas Medical Branch, Galveston, Texas, USA
| | - Gislaine Tezza Rezin
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Graduate Program in Health Sciences, University of Southern Santa Catarina, Tubarao, Brazil
| |
Collapse
|
18
|
Zhang L, Verwer RWH, Zhao J, Huitinga I, Lucassen PJ, Swaab DF. Changes in glial gene expression in the prefrontal cortex in relation to major depressive disorder, suicide and psychotic features. J Affect Disord 2021; 295:893-903. [PMID: 34706460 DOI: 10.1016/j.jad.2021.08.098] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 07/17/2021] [Accepted: 08/28/2021] [Indexed: 01/12/2023]
Abstract
BACKGROUND To establish whether major depressive disorder (MDD), suicidal behaviors and psychotic features contribute to glial alterations in the human prefrontal cortex. MATERIALS AND METHODS We compared mRNA expression using real-time qPCR of 17 glia related genes in the dorsolateral prefrontal cortex (DLPFC) and the anterior cingulate cortex (ACC) between 24 patients with MDD and 12 well-matched controls without psychiatric or neurological diseases. The MDD group was subdivided into i) MDD who died of suicide (MDD-S) or natural causes (MDD-NS) and ii) MDD with or without psychotic features (MDD-P and MDD-NP). The results were followed up with confounder factor analysis. RESULTS Astrocyte gene aldehyde dehydrogenase-1 L1 (ALDH1L1) showed an increased expression in the DLPFC of MDD-NS and the ACC of MDD-NP. S100 calcium-binding protein B (S100B) was upregulated in the DLPFC of MDD compared to the controls. Microglial markers CD11B and purinergic receptor 12 (P2RY12) both showed decreased expression in the ACC of MDD-NS. CD68 was increased in the DLPFC of MDD in both, MDD-S and MDD-P, compared to the controls. In addition, there was increased translocator protein (TSPO) expression in the DLPFC of MDD, especially MDD-NS. In the ACC, this gene had a lower expression in MDD-P than in MDD-NP. Myelin basic protein (MBP) mRNA in the DLPFC increased in MDD, in relation to psychotic features, but not to suicide. LIMITATIONS Sample volumes are relatively small. CONCLUSIONS Different glial functions in MDD were related to specific brain area, suicide or psychotic features.
Collapse
Affiliation(s)
- Lin Zhang
- Neuropsychiatric Disorders Group, Netherlands Institute for Neuroscience, an institute of the Royal Netherlands Academy of Arts and Sciences, University of Amsterdam, Meibergdreef 47, Amsterdam 1105 BA, the Netherlands
| | - Ronald W H Verwer
- Neuropsychiatric Disorders Group, Netherlands Institute for Neuroscience, an institute of the Royal Netherlands Academy of Arts and Sciences, University of Amsterdam, Meibergdreef 47, Amsterdam 1105 BA, the Netherlands
| | - Juan Zhao
- Neuropsychiatric Disorders Group, Netherlands Institute for Neuroscience, an institute of the Royal Netherlands Academy of Arts and Sciences, University of Amsterdam, Meibergdreef 47, Amsterdam 1105 BA, the Netherlands
| | - Inge Huitinga
- Neuroimmunology Group, Netherlands Institute for Neuroscience, an Institute of the Royal Netherlands Academy of Arts and Sciences, Amsterdam, the Netherlands; Brain Plasticity Group, Swammerdam Institute for Life Sciences, Faculty of Science, University of Amsterdam, Amsterdam, the Netherlands
| | - Paul J Lucassen
- Brain Plasticity Group, Swammerdam Institute for Life Sciences, Faculty of Science, University of Amsterdam, Amsterdam, the Netherlands
| | - Dick F Swaab
- Neuropsychiatric Disorders Group, Netherlands Institute for Neuroscience, an institute of the Royal Netherlands Academy of Arts and Sciences, University of Amsterdam, Meibergdreef 47, Amsterdam 1105 BA, the Netherlands.
| |
Collapse
|
19
|
Duan J, Li W, Li W, Liu Q, Tian M, Chen C, Zhang L, Zhang M. Quantitative Proteomics Analysis of Susceptibility and Resilience to Stress in a Rat model of PTSD. Behav Brain Res 2021; 415:113509. [PMID: 34358573 DOI: 10.1016/j.bbr.2021.113509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 07/31/2021] [Accepted: 08/01/2021] [Indexed: 11/16/2022]
Abstract
Posttraumatic stress disorder (PTSD) is a prevalent psychiatric disorder and sometimes deadly consequence of exposure to severe psychological trauma. However, there has been little known about the definitive molecular changes involved in determining vulnerability to PTSD. In the current study, we used proteomics to quantify protein changes in the hippocampus of foot shocks rats. A total of 6151 proteins were quantified and 97 proteins were significantly differentially expressed. The protein-protein interaction (PPI) analysis showed that oxidation-reduction process and glutathione homeostasis may be the potential key progress of being vulnerable to PTSD. The Gene Ontology analysis revealed enriched GO terms in the protein groups of Susceptible group vs Control group rats for glutathione binding,oligopeptide binding,modified amino acid binding,and glutathione transferase activity for their molecular functions (MF) and in the process of cellular response to toxic substance,xenobiotic metabolic process, urea metabolic process, and response to drug for the biological process (BP).SIGNIFICANCE:In recent years, there has been a growing interest in mental illness associated with trauma exposure. We found that stress susceptibility was associated with increased expression of arginase 1 indicated as a potential treatment target. Our results also proposed that carbonic anhydrases 3 could be a biomarker for the development of PTSD. This research helps to explain the potential molecular mechanism in PTSD and supply a new method for ameliorating PTSD.
Collapse
Affiliation(s)
- Jiao Duan
- Department of Anesthesiology, Guangdong Women and Children Hospital, Guangzhou, China
| | - Wenjun Li
- Department of Anesthesiology, The Third Affiliated Hospital of Southern Medical University, China
| | - Weiyan Li
- Department of Anesthesiology, Jinling Hospital, the first School of Clinical Medicine, Southern Medical University, China
| | - Qingzhen Liu
- Department of Anesthesiology, Jinling Hospital, the first School of Clinical Medicine, Southern Medical University, China
| | - Mi Tian
- Department of Anesthesiology, Jinling Hospital, the first School of Clinical Medicine, Southern Medical University, China
| | - Chunlong Chen
- Department of Anesthesiology, Jinling Hospital, the first School of Clinical Medicine, Southern Medical University, China
| | - Lidong Zhang
- Department of Anesthesiology, Jinling Hospital, the first School of Clinical Medicine, Southern Medical University, China
| | - Minhao Zhang
- Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, China.
| |
Collapse
|
20
|
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
|
21
|
Wu Q, Wang B, Ntim M, Zhang X, Na XY, Yuan YH, Wu XF, Yang JY, Li S. SRC-1 Deficiency Increases Susceptibility of Mice to Depressive-Like Behavior After Exposure to CUMS. Neurochem Res 2021; 46:1830-1843. [PMID: 33881662 DOI: 10.1007/s11064-021-03316-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 03/24/2021] [Accepted: 03/29/2021] [Indexed: 11/24/2022]
Abstract
Steroid receptor coactivator 1 (SRC-1) is one of the coactivators recruited by the nuclear receptors (NRs) when NRs are activated by steroid hormones, such as glucocorticoid. SRC-1 is abundant in hippocampus and hypothalamus and is also related to some major risk factors for depression, implicated by its reduced expression after stress and its effect on hypothalamus-pituitary-adrenal gland axis function. However, whether SRC-1 is involved in the formation of depression remains unclear. In this study, we firstly established chronic unpredictable stress (CUS) to induce depressive-like behaviors in mice and found that SRC-1 expression was reduced by CUS. A large number of studies have shown that neuroinflammation is associated with stress-induced depression and lipopolysaccharide (LPS) injection can lead to neuroinflammation and depressive-like behaviors in mice. Our result indicated that LPS treatment also decreased SRC-1 expression in mouse brain, implying the involvement of SRC-1 in the process of inflammation and depression. Next, we showed that the chronic unpredictable mild stress (CUMS) failed to elicit the depressive-like behaviors and dramatically promoted the expression of SRC-1 in brain of wild type mice. What's more, the SRC-1 knockout mice were more susceptible to CUMS to develop depressive-like behaviors and presented the changed expression of glucocorticoid receptor. However, SRC-1 deficiency did not affect the microglia activation induced by CUMS. Altogether, these results indicate a correlation between SRC-1 level and depressive-like behaviors, suggesting that SRC-1 might be involved in the development of depression induced by stress.
Collapse
Affiliation(s)
- Qiong Wu
- Liaoning Provincial Key Laboratory of Cerebral Diseases, Department of Physiology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Bin Wang
- Liaoning Provincial Key Laboratory of Cerebral Diseases, Department of Physiology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Michael Ntim
- Liaoning Provincial Key Laboratory of Cerebral Diseases, Department of Physiology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Xuan Zhang
- National-Local Joint Engineering Research Center for Drug-Research and Development (R & D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, China
| | - Xue-Yan Na
- Liaoning Provincial Key Laboratory of Cerebral Diseases, Department of Physiology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Yu-Hui Yuan
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Xue-Fei Wu
- Liaoning Provincial Key Laboratory of Cerebral Diseases, Department of Physiology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China.
| | - Jin-Yi Yang
- Department of Urology, Affiliated Dalian Friendship Hospital of Dalian Medical University, Dalian, China.
| | - Shao Li
- Liaoning Provincial Key Laboratory of Cerebral Diseases, Department of Physiology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China.
| |
Collapse
|
22
|
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
|
23
|
Ji Y, Luo J, Zeng J, Fang Y, Liu R, Luan F, Zeng N. Xiaoyao Pills Ameliorate Depression-like Behaviors and Oxidative Stress Induced by Olfactory Bulbectomy in Rats via the Activation of the PIK3CA-AKT1-NFE2L2/BDNF Signaling Pathway. Front Pharmacol 2021; 12:643456. [PMID: 33935736 PMCID: PMC8082504 DOI: 10.3389/fphar.2021.643456] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 03/02/2021] [Indexed: 12/28/2022] Open
Abstract
Numerous studies have revealed that oxidative stress is closely associated with the occurrence and development of depression. Xiaoyao Pills (XYW) are included in the Chinese Pharmacopoeia and are frequently used for treating anxiety and depression by smoothing the liver, strengthening the spleen, and nourishing the blood. However, the antidepressant effects of XYW have not yet been thoroughly investigated. The objective of our study was to investigate the antidepressant-like effects of XYW and the underlying molecular mechanism in the olfactory bulbectomized (OB) rat model of depression using the open field test (OFT), sucrose preference test (SPT), splash test (ST), and novelty suppressed feeding test (NSFT). Results showed that XYW (0.93 and 1.86 g·kg−1) significantly alleviated depression-like behaviors in rats, which was indicated by increased sucrose preference in the SPT, prolonged grooming time in the ST, decreased horizontal movement in the OFT, and shorter feeding latency in the NSFT. In addition, XYW treatment dramatically reversed the reduced activity of superoxide dismutase and the decreased level of glutathione, while also lowering levels of malondialdehyde, an inflammatory mediator (nitric oxide), and pro-inflammatory cytokines (interleukin-6 and 1β) in the serum and cortex of OB rats. Mechanistically, XYW induced marked upregulation of mRNA and protein expression levels of NFE2L2, KEAP1, GPX3, HMOX1, SOD1, NQO1, OGG1, PIK3CA, p-AKT1/AKT1, NTRK2, and BDNF, and downregulation of ROS in the cortex and hippocampus via the activation of the NFE2L2/KEAP1, PIK3CA/AKT1, and NTRK2/BDNF pathways. These findings suggest that XYW exert antidepressant-like effects in OB rats with depression-like symptoms, and these effects are mediated by the alleviation of oxidative stress and the enhancement of neuroprotective effects through the activation of the PIK3CA-AKT1-NFE2L2/BDNF signaling pathways.
Collapse
Affiliation(s)
- Yafei Ji
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China.,Department of Pharmacology, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jie Luo
- Department of Pharmacology, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jiuseng Zeng
- Department of Pharmacology, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yang Fang
- Department of Pharmacology, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Rong Liu
- Department of Pharmacology, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Fei Luan
- Department of Pharmacology, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Nan Zeng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China.,Department of Pharmacology, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
24
|
Manosso LM, Lin J, Carlessi AS, Recco KCC, Quevedo J, Gonçalves CL, Réus GZ. Sex-related patterns of the gut-microbiota-brain axis in the neuropsychiatric conditions. Brain Res Bull 2021; 171:196-208. [PMID: 33838211 DOI: 10.1016/j.brainresbull.2021.04.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 03/30/2021] [Accepted: 04/02/2021] [Indexed: 12/19/2022]
Abstract
Sex differences are often observed in psychiatric patients, especially major depressive disorders (MDD), schizophrenia, and developmental disorders, including autism spectrum disorders (ASDs). The prevalence rates between males and females seem variate according to the clinical condition. Although the findings are still incipient, it is suggested that these differences can involve neuroanatomical, neurochemical, and physiological sex differences. In this context, the microbiota-gut-brain axis hypothesis arises to explain some aspects of the complex pathophysiology of neuropsychiatric disorders. The microbiota composition is host-specific and can change conforming to age, sex, diet, medication, exercise, and others. The communication between the brain and the gut is bidirectional and may impact the entire system homeostasis. Many pathways appear to be involved, including neuroanatomic communication, neuroendocrine pathways, immune system, bacteria-derived metabolites, hormones, neurotransmitters, and neurotrophic factors. Although the clinical and preclinical studies are sparse and not very consistent, they suggest that sex differences in the gut microbiota may play an essential role in some neuropsychiatric conditions. Thus, this narrative review has as a mainly aim to show the points sex-related patterns associated to the gut-microbiota-brain axis in the MDD, ASDs, and schizophrenia.
Collapse
Affiliation(s)
- Luana M Manosso
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Jaime Lin
- Experimental Neurology Laboratory, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciúma, SC, Brazil
| | - Anelise S Carlessi
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Kelen C C Recco
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - João Quevedo
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, 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
| | - Cinara L Gonçalves
- Experimental Neurology Laboratory, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciúma, SC, Brazil
| | - Gislaine Z Réus
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil.
| |
Collapse
|
25
|
Rebai R, Jasmin L, Boudah A. Agomelatine effects on fat-enriched diet induced neuroinflammation and depression-like behavior in rats. Biomed Pharmacother 2021; 135:111246. [PMID: 33453676 DOI: 10.1016/j.biopha.2021.111246] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Revised: 11/24/2020] [Accepted: 01/03/2021] [Indexed: 12/12/2022] Open
Abstract
Growing evidence suggests that a high fat diet (HFD) induces oxidative stress on the central nervous system (CNS), which predisposes to mood disorders and neuroinflammation. In this study we postulated that in addition to improving mood, antidepressant therapy would reverse inflammatory changes in the brain of rats exposed to a HFD. To test our hypothesis, we measured the effect of the antidepressant agomelatine (AGO) on anxiety- and depressive-like behaviors, as well as on CNS markers of inflammation in rats rendered obese. Agomelatine is an agonist of the melatonin receptors MT1 and MT2 and an antagonist of the serotonin receptors 5HT2B and 5HT2C. A subset of rats was also treated with lipopolysaccharides (LPS) to determine how additional neuroinflammation alters behavior and affects the response to the antidepressant. Specifically, rats were subjected to a 14-week HFD, during which time behavior was evaluated twice, first at the 10th week prior to LPS and/or agomelatine, and then at the 14th week after a bi-weekly exposure to LPS (250 μg/kg) and daily treatment with agomelatine (40 mg/kg). Immediately after the second behavioral testing we measured the proinflammatory cytokines tumor necrosis factor alpha (TNF-α), interleukin 6 (IL-6) and interleukin 1 beta (IL-1β), markers of oxidative stress thiobarbituric acid reactive substances (TABRS), catalase (CAT) and glutathione peroxidase (GPx), the growth factor BDNF, as well as the apoptosis marker caspase-3. Our results show that a HFD induced an anxiety-like behavior in the open field test (OFT) at the 10th week, followed by a depressive-like behavior in the forced swim test (FST) at the 14th week. In the prefrontal and hippocampal cortices of rats exposed to a HFD we noted an overproduction of TNF-α, IL-6, IL-1β, and TABRS, together with an increase in caspase-3 activity. We also observed a decrease in BDNF, as well as reduced CAT and GPx activity in the same brain areas. Treatment with agomelatine reversed the signs of anxiety and depression, and decreased the cytokines (TNF-α, IL-6 and IL-1β), TABRS, as well as caspase-3 activity. Agomelatine also restored BDNF levels and the activity of antioxidant enzymes CAT and GPx. Our findings suggest that the anxiolytic/antidepressant effect of agomelatine in obese rats could result from a reversal of the inflammatory and oxidative stress brought about by their diet.
Collapse
Affiliation(s)
- Redouane Rebai
- Department of Natural and Life Sciences, Faculty of Exact Sciences and Natural and Life Sciences, University Mohamed Khider of Biskra, BP 145 RP, 07000, Biskra, Algeria; Laboratory of Biotechnology, National Higher School of Biotechnology, Ville universitaire Ali Mendjeli, BP E66 25100, Constantine, Algeria.
| | - Luc Jasmin
- Department of Oral and Maxillofacial Surgery, University of California, San Francisco, 707 Parnassus Ave Suite D-1201, San Francisco, CA, 94143, USA.
| | - Abdennacer Boudah
- Laboratory of Biotechnology, National Higher School of Biotechnology, Ville universitaire Ali Mendjeli, BP E66 25100, Constantine, Algeria.
| |
Collapse
|
26
|
Wu Y, Zhang H, Wang C, Broekman BFP, Chong YS, Shek LP, Gluckman PD, Meaney MJ, Fortier MV, Qiu A. Inflammatory modulation of the associations between prenatal maternal depression and neonatal brain. Neuropsychopharmacology 2021; 46:470-477. [PMID: 32688365 PMCID: PMC7852623 DOI: 10.1038/s41386-020-0774-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 06/26/2020] [Accepted: 07/13/2020] [Indexed: 11/09/2022]
Abstract
Inflammatory signaling has a role in sensing intrauterine environment, which may be moderators in altering fetal brain development upon maternal environment. This study integrated cytokine transcriptome of post-mortem fetal brains, neonatal brain imaging and genetic variants (n = 161) to examine whether cytokines are candidates for modulating the relationship between prenatal maternal depression and fetal brain development. This study obtained the transcriptome data of 208 cytokine genes in 12 fetal brain regions from the BrainSpan database. We also included 161 mother-child dyads with prenatal maternal depressive symptoms assessed at 26 weeks of gestation, cytokine genotype data extracted from umbilical cord specimens, and neonatal brain images from a longitudinal prospective birth cohort. We revealed that 22 cytokine genes are expressed in specific brain regions in utero, whose variants have roles in modulating the effects of the prenatal environment on the accelerated fetal development of the hippocampus, auditory, parietal, orbitofrontal, and dorsal prefrontal cortex. Neonates high in the genetic expression score (GES) of TNFRSF19 and IL17RB showed a larger right hippocampal volume, high in the GES of BMPR1B showed the thicker thickness of the sensorimotor cortex, and high in the GES of IL1RAP and CXCR4 demonstrated the thicker thickness of the dorsal and orbital prefrontal cortex in relation with greater prenatal maternal depressive symptoms. Our findings suggest that in humans, the cytokine genes are expressed in a brain region-specific manner in utero and may have potential roles in modulating the fetal development of the corresponding brain regions in response to the maternal environment.
Collapse
Affiliation(s)
- Yonghui Wu
- grid.4280.e0000 0001 2180 6431Department of Biomedical Engineering, National University of Singapore, Singapore, Singapore
| | - Han Zhang
- grid.4280.e0000 0001 2180 6431Department of Biomedical Engineering, National University of Singapore, Singapore, Singapore
| | - Changqing Wang
- grid.4280.e0000 0001 2180 6431Department of Biomedical Engineering, National University of Singapore, Singapore, Singapore
| | - Birit F. P. Broekman
- grid.452264.30000 0004 0530 269XSingapore Institute for Clinical Sciences, Singapore, Singapore
| | - Yap-Seng Chong
- grid.452264.30000 0004 0530 269XSingapore Institute for Clinical Sciences, Singapore, Singapore ,grid.4280.e0000 0001 2180 6431Department of Obstetrics & Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, National University Health System, Singapore, Singapore
| | - Lynette P. Shek
- grid.4280.e0000 0001 2180 6431Department of Pediatrics, Khoo Teck Puat – National University Children’s Medical Institute, National University of Singapore, Singapore, Singapore
| | - Peter D. Gluckman
- grid.452264.30000 0004 0530 269XSingapore Institute for Clinical Sciences, Singapore, Singapore
| | - Michael J. Meaney
- grid.452264.30000 0004 0530 269XSingapore Institute for Clinical Sciences, Singapore, Singapore ,grid.14709.3b0000 0004 1936 8649Ludmer Centre for Neuroinformatics and Mental Health, Department of Psychiatry, Faculty of Medicine, McGill University, Montreal, Canada
| | - Marielle V. Fortier
- grid.414963.d0000 0000 8958 3388Department of Diagnostic and Interventional Imaging, KK Women’s and Children’s Hospital, Singapore, Singapore
| | - Anqi Qiu
- Department of Biomedical Engineering, National University of Singapore, Singapore, Singapore. .,The N.1 Institute for Health, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
27
|
Kooshki R, Abbasnejad M, Shamsizadeh A, Raoof M, Askari-Zahabi K, Esmaeili-Mahani S. Physical exercise enhances vulnerability to migraine headache associated with CGRP up-expression in trigeminal nucleus caudalis of stressed rats. Neurol Res 2020; 42:952-958. [PMID: 32686605 DOI: 10.1080/01616412.2020.1794243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
OBJECTIVES There is conflicting evidence on the effect of physical exercise on migraine development. Present study investigated the impact of treadmill exercise on migraine - associated symptoms and changes in calcitonin gene-related peptide (CGRP) expression in rats with and without maternal deprivation stress (MD). METHODS Two days after birth, the male Wistar pups were randomly divided into four groups (n = 6) as follows: intact, exercise, MD, and MD plus exercise. The animals in the MD groups were separated from their dams 4 h per day for 2 weeks. At 8 weeks of age, the rats were exercised on a motor-driven treadmill for 4 weeks. Then, nitroglycerin (NTG) (5 mg/kg/IP) was used to induce migraine and pain-related symptoms were recorded for 90 min. NTG-related thermal hyperalgesia was measured by tail flick and hot plate methods. Finally, immunofluorescence staining of CGRP in trigeminal subnucleus caudalis (Vc) was performed. RESULTS NTG - produced a significant headache symptoms and thermal hypersensitivity, which were aggravated following physical exercise in stressed or unstressed groups. Besides, NTG administration increased CGRP expression in the Vc of rats. Such effect was overpowered by treadmill running only in rats exposed to MD stress. CONCLUSION These findings highlight the worsening effects of treadmill exercise for migraine in rats with and without MD stress. However, inflammatory response can further exacerbate in stressed rats.
Collapse
Affiliation(s)
- Razieh Kooshki
- Physiology-pharmacology Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences , Rafsanjan, Iran.,Department of Biology, Faculty of Sciences, Lorestan University , Khorramabad, Iran
| | - Mehdi Abbasnejad
- Department of Biology, Faculty of Sciences, Shahid Bahonar University of Kerman , Kerman, Iran
| | - Ali Shamsizadeh
- Physiology-pharmacology Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences , Rafsanjan, Iran
| | - Maryam Raoof
- Endodontology Research Center, Kerman University of Medical Sciences , Kerman, Iran.,Department of Orofacial Pain and Dysfunction, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam , Amsterdam, The Netherlands
| | - Khadijeh Askari-Zahabi
- Department of Biology, Faculty of Sciences, Shahid Bahonar University of Kerman , Kerman, Iran
| | - Saeed Esmaeili-Mahani
- Department of Biology, Faculty of Sciences, Shahid Bahonar University of Kerman , Kerman, Iran
| |
Collapse
|
28
|
Neuroinflammatory Responses and Parkinson' Disease: Pathogenic Mechanisms and Therapeutic Targets. J Neuroimmune Pharmacol 2020; 15:830-837. [PMID: 32529463 DOI: 10.1007/s11481-020-09926-7] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 05/19/2020] [Indexed: 02/06/2023]
Abstract
Parkinson's disease (PD) is the second most common age-related neurodegenerative disorders of the central nervous system, which mainly impairs the motor system. However, the pathogenic mechanisms are still unclear. Gene-environment complex interaction leads to selective dopaminergic neuron death in PD. Growing evidences supports that neuroinflammatory responses are involved in the pathogenesis of PD. This review critically discusses current studies on the inflammatory response of the pathological process of PD. The mechanisms and strategies of modifying inflammatory responses would be potential treatments for neurodegenerative diseases. Graphical abstract Activated microglia canpromote the damage ofdopaminergic neurons, which inturn aggravates the activation ofmicroglia in the process of PD. Atthe same time, microglia canactivate astrocytes throughproliferation and secretion ofinflammatory factors. The role ofastrocytes on the loss ofdopaminergic neurons is stillcontroversial in PD. (Nonsteroidalanti-inflammatory drugs,NSAIDs. adiposed-derived stemcells, ADSCs.nicotinamideadenine dinucleotide phosphate,NADPH. signal transducers andactivators of transcription,STAT.DJ-1,Aliases forPARK7.mesencephalic astrocytederivedneurotrophic factor,MANF.Ciliary neurotrophicfactor,CNTF.glial cell linederivedneurotrophic factor,GDNF.Wnt Family Member1,Wnt1). Graphical abstract Mitochondrial dysfunction causes neuroinflammation throughDAMPs and a series of factors such as oxidative stress andinflammatory bodies in PD. (Damage-associated molecular patterns,DAMPs. reactive oxygen species, ROS). Graphical abstract Various mechanismsparticipate in NLRP3 activation,causing microglia activation inPD. ( -synuclein, -syn.) TolllikeReceptor 2, TLR2. Toll-likeReceptor 4, TLR4. TumorNecrosis Factor, TNF.Apoptosisassociated speck like proteincontaining a CARD, ASC).
Collapse
|
29
|
Dutcher EG, Pama EC, Lynall ME, Khan S, Clatworthy MR, Robbins TW, Bullmore ET, Dalley JW. Early-life stress and inflammation: A systematic review of a key experimental approach in rodents. Brain Neurosci Adv 2020; 4:2398212820978049. [PMID: 33447663 PMCID: PMC7780197 DOI: 10.1177/2398212820978049] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 11/11/2020] [Indexed: 12/11/2022] Open
Abstract
Repeated maternal separation is the most widely used pre-clinical approach to investigate the relationship between early-life chronic stress and its neuropsychiatric and physical consequences. In this systematic review, we identified 46 studies that conducted repeated maternal separation or single-episode maternal separation and reported measurements of interleukin-1b, interleukin-6, interleukin-10, tumour necrosis factor-alpha, or microglia activation and density. We report that in the short-term and in the context of later-life stress, repeated maternal separation has pro-inflammatory immune consequences in diverse tissues. Repeated maternal separation animals exhibit greater microglial activation and elevated pro-inflammatory cytokine signalling in key brain regions implicated in human psychiatric disorders. Notably, repeated maternal separation generally has no long-term effect on cytokine expression in any tissue in the absence of later-life stress. These observations suggest that the elevated inflammatory signalling that has been reported in humans with a history of early-life stress may be the joint consequence of ongoing stressor exposure together with potentiated neural and/or immune responsiveness to stressors. Finally, our findings provide detailed guidance for future studies interrogating the causal roles of early-life stress and inflammation in disorders such as major depression.
Collapse
Affiliation(s)
- Ethan G. Dutcher
- Department of Psychology, University of Cambridge, Cambridge, UK
| | | | - Mary-Ellen Lynall
- Department of Psychiatry, University of Cambridge, Cambridge, UK
- Molecular Immunity Unit, MRC Laboratory of Molecular Biology, Cambridge, UK
| | - Shahid Khan
- GlaxoSmithKline Research & Development, Stevenage, UK
| | | | | | | | - Jeffrey W. Dalley
- Department of Psychology, University of Cambridge, Cambridge, UK
- Department of Psychiatry, University of Cambridge, Cambridge, UK
| |
Collapse
|
30
|
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: 131] [Impact Index Per Article: 21.8] [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
|
31
|
Geng J, Liu J, Yuan X, Liu W, Guo W. Andrographolide triggers autophagy-mediated inflammation inhibition and attenuates chronic unpredictable mild stress (CUMS)-induced depressive-like behavior in mice. Toxicol Appl Pharmacol 2019; 379:114688. [DOI: 10.1016/j.taap.2019.114688] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 07/17/2019] [Accepted: 07/19/2019] [Indexed: 12/28/2022]
|