1
|
Müller L, Di Benedetto S. Neuroimmune crosstalk in chronic neuroinflammation: microglial interactions and immune modulation. Front Cell Neurosci 2025; 19:1575022. [PMID: 40260075 PMCID: PMC12009833 DOI: 10.3389/fncel.2025.1575022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Accepted: 03/25/2025] [Indexed: 04/23/2025] Open
Abstract
Neuroinflammation is a fundamental feature of many chronic neurodegenerative diseases, where it contributes to disease onset, progression, and severity. This persistent inflammatory state arises from the activation of innate and adaptive immune responses within the central nervous system (CNS), orchestrated by a complex interplay of resident immune cells, infiltrating peripheral immune cells, and an array of molecular mediators such as cytokines, chemokines, and extracellular vesicles. Among CNS-resident cells, microglia play a central role, exhibiting a dynamic spectrum of phenotypes ranging from neuroprotective to neurotoxic. In chronic neurodegenerative diseases, sustained microglial activation often leads to the amplification of inflammatory cascades, reinforcing a pathogenic cycle of immune-mediated damage. Intercellular communication within the inflamed CNS is central to the persistence and progression of neuroinflammation. Microglia engage in extensive crosstalk with astrocytes, neurons, oligodendrocytes, and infiltrating immune cells, shaping both local and systemic inflammatory responses. These interactions influence key processes such as synaptic pruning, phagocytosis, blood-brain barrier integrity, and cytokine-mediated signaling. Understanding the mechanisms of cell-cell signaling in this context is critical for identifying therapeutic strategies to modulate the immune response and restore homeostasis. This review explores the key players in CNS neuroinflammation, with a focus on the role of microglia, the molecular pathways underlying intercellular communication, and potential therapeutic approaches to mitigate neuroinflammatory damage in chronic neurodegenerative diseases.
Collapse
Affiliation(s)
- Ludmila Müller
- Max Planck Institute for Human Development, Center for Lifespan Psychology, Berlin, Germany
| | | |
Collapse
|
2
|
Izumi Y, O'Dell KA, Mennerick S, Zorumski CF. Effects of acute pro-inflammatory stimulation and 25-hydroxycholesterol on hippocampal plasticity and learning involve NLRP3 inflammasome and cellular stress responses. Sci Rep 2025; 15:6149. [PMID: 39979396 PMCID: PMC11842721 DOI: 10.1038/s41598-025-90149-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Accepted: 02/11/2025] [Indexed: 02/22/2025] Open
Abstract
Neuroinflammation is an increasingly important target for therapeutics in neuropsychiatry and contributes to cognitive dysfunction, disability and death across a range of illnesses. We previously found that acute effects of pro-inflammatory stimulation with lipopolysaccharide (LPS) on hippocampal long-term potentiation (LTP), a form of synaptic plasticity involved in learning and memory, requires synthesis of the oxysterol, 25-hydroxycholesterol (25HC) and exogenous 25HC mimics effects of LPS. However, downstream mechanisms engaged by LPS and 25HC remain uncertain. Here we use rat hippocampal slices and in vivo behavioral studies to provide evidence that acute modulation of synaptic plasticity by both LPS and 25HC requires activation of the NLRP3 inflammasome, caspase-1 and interleukin-1 receptor. Furthermore, both LPS and 25HC engage cellular stress responses including synthesis of 5α-reduced neurosteroids and effects on plasticity are prevented by modulators of these responses. In studies of acute learning using a one-trial inhibitory avoidance task, inhibition of learning by LPS and 25HC are prevented by pre-treatment with an inhibitor of NLRP3. The present studies provide strong support for the role of 25HC as a mediator of pro-inflammatory stimulation on hippocampal synaptic plasticity and for the importance of NLRP3 inflammasome and caspase-1 activation in the deleterious effects of acute inflammation.
Collapse
Affiliation(s)
- Yukitoshi Izumi
- Department of Psychiatry, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO, 63110, USA
- Taylor Family Institute for Innovative Psychiatric Research, Washington University School of Medicine, St. Louis, MO, USA
| | - Kazuko A O'Dell
- Department of Psychiatry, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO, 63110, USA
| | - Steven Mennerick
- Department of Psychiatry, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO, 63110, USA
- Taylor Family Institute for Innovative Psychiatric Research, Washington University School of Medicine, St. Louis, MO, USA
| | - Charles F Zorumski
- Department of Psychiatry, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO, 63110, USA.
- Taylor Family Institute for Innovative Psychiatric Research, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
3
|
De Felice E, Bobotis BC, Rigillo G, Khakpour M, Gonçalves de Andrade E, Benatti C, Vilella A, Tascedda F, Limatola C, Tremblay MÈ, Alboni S, Maggi L. Female mice exhibit similar long-term plasticity and microglial properties between the dorsal and ventral hippocampal poles. Brain Behav Immun 2025; 124:192-204. [PMID: 39617070 DOI: 10.1016/j.bbi.2024.11.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 11/13/2024] [Accepted: 11/27/2024] [Indexed: 12/09/2024] Open
Abstract
The hippocampus is a heterogenous structure that exhibits functional segregation along its longitudinal axis. We recently showed that in male mice, microglia, the brain's resident immune cells, differ between the dorsal (DH) and ventral (VH) hippocampus, impacting long-term potentiation (LTP) mainly through the CX3CL1-CX3CR1 signaling. Here, we assessed the specific features of the hippocampal poles in female mice, demonstrating a similar LTP amplitude in VH and DH in both control and Cx3cr1 knock-out mice. In addition, the expression levels of Cx3cr1 and Cx3cl1 mRNA do not differ between the two poles in control mice. These data support the critical role of the CX3CL1-CX3CR1 signaling in setting the physiological amount of plasticity, equally between poles in females. Although BDNF is higher in DH compared to VH, the expression levels of inflammatory markers involved in plasticity and of phagocytosis markers in microglia are comparable between the two poles. In accordance, microglia soma and arborization area/perimeter, and microglial ultrastructure are similar across regions, with the exception of microglial density, cells arborization solidity and circularity that are higher in DH. Understanding the molecular processes underlying microglial sex differences and their potential implications for plasticity in specific brain regions is of major importance in physiological and pathological conditions.
Collapse
Affiliation(s)
- Eleonora De Felice
- Department of Physiology and Pharmacology, Sapienza University of Rome, Piazzale Aldo Moro, 5, 00185 Rome, Italy.
| | - Bianca Caroline Bobotis
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada; Centre for Advanced Materials and Related Technology (CAMTEC), University of Victoria, Victoria, BC, Canada
| | - Giovanna Rigillo
- Centre of Neuroscience and Neurotechnology, University of Modena and Reggio Emilia, Modena, Italy; Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | | | | | - Cristina Benatti
- Centre of Neuroscience and Neurotechnology, University of Modena and Reggio Emilia, Modena, Italy; Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Antonietta Vilella
- Centre of Neuroscience and Neurotechnology, University of Modena and Reggio Emilia, Modena, Italy; Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Fabio Tascedda
- Centre of Neuroscience and Neurotechnology, University of Modena and Reggio Emilia, Modena, Italy; Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Cristina Limatola
- IRCCS Neuromed, Pozzilli, Italy; Department of Physiology and Pharmacology, Laboratory Affiliated to Institute Pasteur, Sapienza University, Rome, Italy
| | - Marie-Ève Tremblay
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada; Centre for Advanced Materials and Related Technology (CAMTEC), University of Victoria, Victoria, BC, Canada; Institute On Aging and Lifelong Health (IALH), University of Victoria, Victoria, BC, Canada; Department of Biochemistry and Molecular Biology, The University of British Columbia Vancouver, BC, Canada; Department of Neurology and Neurosurgery, McGill University, Montréal, QC, Canada
| | - Silvia Alboni
- Centre of Neuroscience and Neurotechnology, University of Modena and Reggio Emilia, Modena, Italy; Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Laura Maggi
- Department of Physiology and Pharmacology, Sapienza University of Rome, Piazzale Aldo Moro, 5, 00185 Rome, Italy
| |
Collapse
|
4
|
Müller L, Di Benedetto S, Müller V. From Homeostasis to Neuroinflammation: Insights into Cellular and Molecular Interactions and Network Dynamics. Cells 2025; 14:54. [PMID: 39791755 PMCID: PMC11720143 DOI: 10.3390/cells14010054] [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: 11/27/2024] [Revised: 12/26/2024] [Accepted: 01/03/2025] [Indexed: 01/12/2025] Open
Abstract
Neuroinflammation is a complex and multifaceted process that involves dynamic interactions among various cellular and molecular components. This sophisticated interplay supports both environmental adaptability and system resilience in the central nervous system (CNS) but may be disrupted during neuroinflammation. In this article, we first characterize the key players in neuroimmune interactions, including microglia, astrocytes, neurons, immune cells, and essential signaling molecules such as cytokines, neurotransmitters, extracellular matrix (ECM) components, and neurotrophic factors. Under homeostatic conditions, these elements promote cellular cooperation and stability, whereas in neuroinflammatory states, they drive adaptive responses that may become pathological if dysregulated. We examine how neuroimmune interactions, mediated through these cellular actors and signaling pathways, create complex networks that regulate CNS functionality and respond to injury or inflammation. To further elucidate these dynamics, we provide insights using a multilayer network (MLN) approach, highlighting the interconnected nature of neuroimmune interactions under both inflammatory and homeostatic conditions. This perspective aims to enhance our understanding of neuroimmune communication and the mechanisms underlying shifts from homeostasis to neuroinflammation. Applying an MLN approach offers a more integrative view of CNS resilience and adaptability, helping to clarify inflammatory processes and identify novel intervention points within the layered landscape of neuroinflammatory responses.
Collapse
Affiliation(s)
- Ludmila Müller
- Max Planck Institute for Human Development, Lentzeallee 94, 14195 Berlin, Germany (V.M.)
| | | | | |
Collapse
|
5
|
Vella VR, Holman PJ, Bodnar TS, Raineki C. Ontogenetic Neuroimmune Changes Following Prenatal Alcohol Exposure: Implications for Neurobehavioral Function. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1473:15-39. [PMID: 40128473 DOI: 10.1007/978-3-031-81908-7_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 03/26/2025]
Abstract
This chapter reviews the enduring effects of prenatal alcohol exposure (PAE) on neuroimmune function across the lifespan, including discussion of associated neurobehavioral alterations. Alcohol has potent teratogenic effects, with a large body of work linking PAE to perturbations in neuroimmune function. These PAE-related neuroimmune disturbances may have downstream effects on neurobehavioral function given the critical role of the neuroimmune system in central nervous system development. The neuroimmune system matures over time, playing distinct roles depending on the developmental processes occurring within that maturational stage. This chapter thus takes an ontogenetic approach to understanding how PAE induces unique neuroimmune changes across the lifespan, beginning with a review of changes in early life before moving into adolescence and ending in adulthood. The focus will be on work utilizing rodent models, which allow for more tightly controlled conditions than are possible in human research. The chapter concludes with a discussion of possible mechanisms underlying the developmental changes in neuroimmune function following PAE, with a specific focus on the role of the gut microbiota.
Collapse
Affiliation(s)
- Victoria R Vella
- Department of Psychology, Brock University, St. Catharines, ON, Canada
| | - Parker J Holman
- Department of Psychology, Brock University, St. Catharines, ON, Canada
| | - Tamara S Bodnar
- Department of Biological Sciences, University of Calgary, Calgary, AB, Canada
- Alberta Children's Hospital Research Institute, Calgary, AB, Canada
- Hotchkiss Brain Institute, Calgary, AB, Canada
| | - Charlis Raineki
- Department of Psychology, Brock University, St. Catharines, ON, Canada.
- Centre for Neuroscience, Brock University, St. Catharines, ON, Canada.
| |
Collapse
|
6
|
Besedovsky H, Del Rey A. A Glucocorticoid-Mediated Immunoregulatory Circuit Integrated at Brain Levels: Our Early Studies and a Present View. Neuroimmunomodulation 2024; 31:230-245. [PMID: 39504948 DOI: 10.1159/000542401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 10/29/2024] [Indexed: 11/08/2024] Open
Abstract
BACKGROUND It was known since the 1940s that pharmacological administration of glucocorticoids can inhibit inflammatory and immune processes, and these hormones are still today among the most widely used therapeutic tools to treat diseases with immune components. However, it became clear later that endogenous glucocorticoids can either support or restrain immune processes. SUMMARY Early studies showed that (a) endogenous levels of glucocorticoids can modulate immune cell activity; (b) the immune response itself can stimulate the hypothalamus-pituitary-adrenal (HPA) axis to release glucocorticoids to levels that can exert immunoregulatory effects; (c) immune products, later identified as cytokines, mediate this effect. On these bases, the existence of a glucocorticoid-mediated immunoregulatory circuit was proposed. It was also shown that increased levels of endogenous glucocorticoids exert protective effects during infections and other diseases with immune components. However, it was found in animal models and in humans that these effects can be blunted in several immune-linked diseases by defects at several levels, for example, by glucocorticoid resistance or by adrenal insufficiency. Evidence was later provided that the glucocorticoid-mediated immunoregulatory circuit can also be activated by cytokines produced not only as consequence of immune stimulation but also following psycho/sensorial and physical stimuli. Thus, this circuit can be integrated at brain levels and, besides stimulating the HPA axis, cytokines can also affect synaptic plasticity, most likely via a tripartite synapse, with astrocytes as neuro-immune cells acting as the third component. KEY MESSAGES It is now well established that the glucocorticoid-mediated immunoregulatory circuit plays a central role in maintaining health. However, several variables can condition the efficacy of the effect of endogenous glucocorticoids. Furthermore, since cytokines and other immune products have many other neuroendocrine and metabolic effects, other neuroendocrine-immune circuits could simultaneously operate or become predominant during different pathologies. The consideration of these aspects might help to implement strategies to eventually decrease therapeutic doses of exogenous glucocorticoids.
Collapse
Affiliation(s)
- Hugo Besedovsky
- Research Group Immunophysiology, Department Neurophysiology, Institute of Physiology and Pathophysiology, Marburg, Germany
| | - Adriana Del Rey
- Research Group Immunophysiology, Department Neurophysiology, Institute of Physiology and Pathophysiology, Marburg, Germany
| |
Collapse
|
7
|
Khan H, Naseem T, Kaushik P, Narang J, Khan R, Panwar S, Parvez S. Decoding paradoxical links of cytokine markers in cognition: Cross talk between physiology, inflammaging, and Alzheimer's disease- related cognitive decline. Ageing Res Rev 2024; 101:102535. [PMID: 39374831 DOI: 10.1016/j.arr.2024.102535] [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: 07/27/2024] [Revised: 09/29/2024] [Accepted: 10/01/2024] [Indexed: 10/09/2024]
Abstract
Recent research has revolutionized our understanding of memory consolidation by emphasizing the critical role of astrocytes, microglia, and immune cells in through cytokine signaling. Cytokines, compact proteins, play pivotal roles in neuronal development, synaptic transmission, and normal aging. This review explores the cellular mechanisms contributing to cognitive decline in inflammaging and Alzheimer's disease, highlighting the paradoxical effects of most studied cytokines (IL-1, IL-6, TNF-α) in brain function, which act as a double-edged sword in brain physiology, acting both as facilitators of healthy cognitive function and as a potential contributor to cognitive decline.
Collapse
Affiliation(s)
- Hiba Khan
- Department of Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi 110062, India
| | - Talib Naseem
- Department of Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi 110062, India
| | - Pooja Kaushik
- Department of Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi 110062, India
| | - Jagriti Narang
- Department of Biotechnology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi 110062, India
| | - Rehan Khan
- Chemical Biology Unit, Institute of Nano Science and Technology, Sector 81, Knowledge City, Sahibzada Ajit Singh Nagar, Mohali, Punjab 140306, India
| | - Siddharth Panwar
- School of Computing and Electrical Engineering, Indian Institute of Technology, Mandi, Himachal Pradesh 175075, India
| | - Suhel Parvez
- Department of Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi 110062, India.
| |
Collapse
|
8
|
Benarroch E. What Is the Role of Cytokines in Synaptic Transmission? Neurology 2024; 103:e209928. [PMID: 39303183 DOI: 10.1212/wnl.0000000000209928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/22/2024] Open
|
9
|
Subramani M, Lambrecht B, Ahmad I. Human microglia-derived proinflammatory cytokines facilitate human retinal ganglion cell development and regeneration. Stem Cell Reports 2024; 19:1092-1106. [PMID: 39059376 PMCID: PMC11368696 DOI: 10.1016/j.stemcr.2024.06.009] [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: 03/01/2024] [Revised: 06/21/2024] [Accepted: 06/23/2024] [Indexed: 07/28/2024] Open
Abstract
Microglia (μG), the resident immune cells in the central nervous system, surveil the parenchyma to maintain the structural and functional homeostasis of neurons. Besides, they influence neurogenesis and synaptogenesis through complement-mediated phagocytosis. Emerging evidence suggests that μG may also influence development through proinflammatory cytokines. Here, we examined the premise that tumor necrosis factor alpha (TNF-α) and interleukin-1β (IL-1β), the two most prominent components of the μG secretome, influence retinal development, specifically the morphological and functional differentiation of human retinal ganglion cells (hRGCs). Using controlled generation of hRGCs and human μG (hμG) from pluripotent stem cells, we demonstrate that TNF-α and IL-1β secreted by unchallenged hμG did not influence hRGC generation. However, their presence significantly facilitated neuritogenesis along with the basal function of hRGCs, which involved the recruitment of the AKT/mTOR pathway. We present ex vivo evidence that proinflammatory cytokines may play an important role in the morphological and physiological maturation of hRGCs, which may be recapitulated for regeneration.
Collapse
Affiliation(s)
- Murali Subramani
- Department of Ophthalmology and Visual Science, University of Nebraska Medical Center, Omaha, NE, USA
| | - Brandon Lambrecht
- Department of Ophthalmology and Visual Science, University of Nebraska Medical Center, Omaha, NE, USA
| | - Iqbal Ahmad
- Department of Ophthalmology and Visual Science, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
10
|
Talebi V, Alamdari KA, Patel DI. Simple and Complex Wheel Running Effect on Depression, Memory, Neuroinflammation, and Neurogenesis in Alzheimer's Rat Model. Med Sci Sports Exerc 2024; 56:1159-1167. [PMID: 38227543 DOI: 10.1249/mss.0000000000003394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2024]
Abstract
INTRODUCTION The aim of this study was to investigate 12 wk of simple and complex voluntary wheel running on Alzheimer's disease (AD), associated biomarkers, and behaviors. METHODS Sixty male Wistar rats were randomly divided into six groups: healthy control (Con-Sed), AD only (AD-Sed), simple wheel control (SWC), complex wheel control (CWC), simple wheel AD (SWAD), and complex wheel AD (CWAD). Novelty-suppressed feeding test and the Morris water maze test were used to evaluate depression and memory, respectively. Ki67 was measured in the hippocampus, whereas interleukin (IL)-1β and neural/glial antigen 2 (NG2) were measured in both the hippocampus and the prefrontal cortex. One-way ANOVA with Tukey's post hoc test was performed. RESULTS AD-Sed group had significantly lower spacial memory ( P < 0.001) compared with Con-Sed. Simple and complex wheel running attenuated these deficits in the SWAD and CWAD groups, respectively ( P < 0.001). Only the CWAD group had significantly improved novelty-suppressed feeding test time compared with AD-Sed ( P < 0.001), equivalent to the healthy wheel running groups. AD-Sed has significantly higher hippocampal concentrations of Ki67 ( P = 0.01) compared with the Con-Sed. Both SWAD and CWAD had significantly reduced Ki67 with similar concentrations compared with the SWC and CWC groups ( P > 0.05). AD-Sed animals also presented with significantly higher hippocampal and prefrontal cortex concentrations of IL-1β compared with Con-Sed ( P < 0.001). SWAD and CWAD had no effect in changing these concentrations. Complex wheel running significantly increased NG2 in the healthy control and AD models, whereas simple wheel running significantly increased NG2 in the AD model. CONCLUSIONS The results of our study suggest that complex wheel running might be more advantageous in promoting memory and neuroplasticity while reducing depression that is associated with AD.
Collapse
Affiliation(s)
- Vahid Talebi
- Department of Sports Science, Faculty of Educational Sciences and Psychology, Azarbaijan Shahid Madani University, Tabriz, IRAN
| | - Karim Azali Alamdari
- Department of Sports Science, Faculty of Educational Sciences and Psychology, Azarbaijan Shahid Madani University, Tabriz, IRAN
| | - Darpan I Patel
- School of Nursing, University of Texas Medical Branch at Galveston, Galveston, TX
| |
Collapse
|
11
|
Namba MD, Xie Q, Park K, Jackson JG, Barker JM. EcoHIV Infection Modulates the Effects of Cocaine Exposure Pattern and Abstinence on Cocaine Seeking and Neuroimmune Protein Expression in Male Mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.15.589615. [PMID: 38659915 PMCID: PMC11042347 DOI: 10.1101/2024.04.15.589615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
Cocaine use disorders (CUDs) and human immunodeficiency virus (HIV) remain persistent public health dilemmas throughout the world. One major hurdle for treating CUD is the increase in cocaine craving and seeking behavior that occurs over a protracted period of abstinence, an effect known as the incubation of craving. Little is known about how HIV may modulate this process. Thus, we sought to examine the impact of chronic HIV infection on the incubation of cocaine craving and associated changes in the central and peripheral immune systems. Here, mice were inoculated with EcoHIV, which is a chimeric HIV-1 construct that produces chronic HIV infection in mice. EcoHIV- and sham-infected mice were conditioned with cocaine daily or intermittently in a conditioned place preference (CPP) paradigm, followed by 1 or 21 days of forced abstinence prior to assessing preference for the cocaine-paired chamber. Under both conditioning regimens, sham mice exhibited incubation of cocaine CPP after 21 days of abstinence. EcoHIV-infected mice conditioned daily with cocaine showed enhanced cocaine seeking at both abstinence timepoints, whereas infected mice conditioned intermittently showed a reversal of the incubation effect, with higher cocaine seeking after 1 day of abstinence compared to 21 days. Analysis of corticolimbic CX3CL1-CX3CR1 and glutamate receptor expression revealed alterations in medial prefrontal cortex (mPFC) CX3CL1 and nucleus accumbens (NAc) GluN2A receptors that correlated with cocaine seeking following daily cocaine exposure. Moreover, examination of peripheral immune markers showed that the effect of abstinence and EcoHIV infection on these measures depended on the cocaine exposure regimen. Altogether, these results highlight the importance of cocaine abstinence and exposure pattern as critical variables that modulate HIV-associated neuroimmune outcomes and relapse vulnerability.
Collapse
Affiliation(s)
- Mark D. Namba
- Department of Pharmacology & Physiology, College of Medicine, Drexel University, Philadelphia, PA, USA
| | - Qiaowei Xie
- Department of Pharmacology & Physiology, College of Medicine, Drexel University, Philadelphia, PA, USA
- Graduate Program in Pharmacology and Physiology, College of Medicine, Drexel University
| | - Kyewon Park
- Center for AIDS Research (CFAR), University of Pennsylvania, Philadelphia, PA, USA
| | - Joshua G. Jackson
- Department of Pharmacology & Physiology, College of Medicine, Drexel University, Philadelphia, PA, USA
| | - Jacqueline M. Barker
- Department of Pharmacology & Physiology, College of Medicine, Drexel University, Philadelphia, PA, USA
| |
Collapse
|
12
|
Zhu Q, Wan L, Huang H, Liao Z. IL-1β, the first piece to the puzzle of sepsis-related cognitive impairment? Front Neurosci 2024; 18:1370406. [PMID: 38665289 PMCID: PMC11043581 DOI: 10.3389/fnins.2024.1370406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Accepted: 04/02/2024] [Indexed: 04/28/2024] Open
Abstract
Sepsis is a leading cause of death resulting from an uncontrolled inflammatory response to an infectious agent. Multiple organ injuries, including brain injuries, are common in sepsis. The underlying mechanism of sepsis-associated encephalopathy (SAE), which is associated with neuroinflammation, is not yet fully understood. Recent studies suggest that the release of interleukin-1β (IL-1β) following activation of microglial cells plays a crucial role in the development of long-lasting neuroinflammation after the initial sepsis episode. This review provides a comprehensive analysis of the recent literature on the molecular signaling pathways involved in microglial cell activation and interleukin-1β release. It also explores the physiological and pathophysiological role of IL-1β in cognitive function, with a particular focus on its contribution to long-lasting neuroinflammation after sepsis. The findings from this review may assist healthcare providers in developing novel interventions against SAE.
Collapse
Affiliation(s)
- Qing Zhu
- Department of Anesthesiology, Key Laboratory of Birth Defects and Related Diseases of Women and Children, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Li Wan
- Department of Medical Genetics/Prenatal Diagnostic Center Nursing and Key Laboratory of Birth Defects and Related Diseases of Women and Children, West China Second University Hospital of Sichuan University, Chengdu, China
| | - Han Huang
- Department of Anesthesiology, Key Laboratory of Birth Defects and Related Diseases of Women and Children, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Zhimin Liao
- Department of Anesthesiology, Key Laboratory of Birth Defects and Related Diseases of Women and Children, West China Second University Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
13
|
Borreca A, Mantovani C, Desiato G, Corradini I, Filipello F, Elia CA, D'Autilia F, Santamaria G, Garlanda C, Morini R, Pozzi D, Matteoli M. Loss of interleukin 1 signaling causes impairment of microglia- mediated synapse elimination and autistic-like behaviour in mice. Brain Behav Immun 2024; 117:493-509. [PMID: 38307446 DOI: 10.1016/j.bbi.2024.01.221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 01/11/2024] [Accepted: 01/25/2024] [Indexed: 02/04/2024] Open
Abstract
In the last years, the hypothesis that elevated levels of proinflammatory cytokines contribute to the pathogenesis of neurodevelopmental diseases has gained popularity. IL-1 is one of the main cytokines found to be elevated in Autism spectrum disorder (ASD), a complex neurodevelopmental condition characterized by defects in social communication and cognitive impairments. In this study, we demonstrate that mice lacking IL-1 signaling display autistic-like defects associated with an excessive number of synapses. We also show that microglia lacking IL-1 signaling at early neurodevelopmental stages are unable to properly perform the process of synapse engulfment and display excessive activation of mammalian target of rapamycin (mTOR) signaling. Notably, even the acute inhibition of IL-1R1 by IL-1Ra is sufficient to enhance mTOR signaling and reduce synaptosome phagocytosis in WT microglia. Finally, we demonstrate that rapamycin treatment rescues the defects in IL-1R deficient mice. These data unveil an exclusive role of microglial IL-1 in synapse refinement via mTOR signaling and indicate a novel mechanism possibly involved in neurodevelopmental disorders associated with defects in the IL-1 pathway.
Collapse
Affiliation(s)
- Antonella Borreca
- Institute of Neuroscience (IN-CNR), Consiglio Nazionale delle Ricerche, Milan, Italy; IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Cristina Mantovani
- IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Rozzano, Milan, Italy; Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20072 Pieve Emanuele, Milan, Italy
| | - Genni Desiato
- IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Irene Corradini
- Institute of Neuroscience (IN-CNR), Consiglio Nazionale delle Ricerche, Milan, Italy; IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Fabia Filipello
- IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Chiara Adriana Elia
- IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Francesca D'Autilia
- IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Giulia Santamaria
- IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Cecilia Garlanda
- IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Rozzano, Milan, Italy; Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20072 Pieve Emanuele, Milan, Italy
| | - Raffaella Morini
- IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Davide Pozzi
- IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Rozzano, Milan, Italy; Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20072 Pieve Emanuele, Milan, Italy.
| | - Michela Matteoli
- Institute of Neuroscience (IN-CNR), Consiglio Nazionale delle Ricerche, Milan, Italy; IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Rozzano, Milan, Italy.
| |
Collapse
|
14
|
Šimončičová E, Henderson Pekarik K, Vecchiarelli HA, Lauro C, Maggi L, Tremblay MÈ. Adult Neurogenesis, Learning and Memory. ADVANCES IN NEUROBIOLOGY 2024; 37:221-242. [PMID: 39207695 DOI: 10.1007/978-3-031-55529-9_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Neural plasticity can be defined as the ability of neural circuits to be shaped by external and internal factors. It provides the brain with a capacity for functional and morphological remodelling, with many lines of evidence indicating that these changes are vital for learning and memory formation. The basis of this brain plasticity resides in activity- and experience-driven modifications of synaptic strength, including synaptic formation, elimination or weakening, as well as of modulation of neuronal population, which drive the structural reorganization of neural networks. Recent evidence indicates that brain-resident glial cells actively participate in these processes, suggesting that mechanisms underlying plasticity in the brain are multifaceted. Establishing the 'tripartite' synapse, the role of astrocytes in modulating synaptic transmission in response to neuronal activity was recognized first. Further redefinition of the synapse as 'quad-partite' followed to acknowledge the contribution of microglia which were revealed to affect numerous brain functions via dynamic interactions with synapses, acting as 'synaptic sensors' that respond to neuronal activity and neurotransmitter release, as well as crosstalk with astrocytes. Early studies identified microglial ability to dynamically survey their local brain environment and established their integral role in the active interfacing of environmental stimuli (both internal and external), with brain plasticity and remodelling. Following the introduction to neurogenesis, this chapter details the role that microglia play in regulating neurogenesis in adulthood, specifically as it relates to learning and memory, as well as factors involved in modulation of microglia. Further, a microglial perspective is introduced for the context of environmental enrichment impact on neurogenesis, learning and memory across states of stress, ageing, disease and injury.
Collapse
Affiliation(s)
- Eva Šimončičová
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | | | | | - Clotilde Lauro
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy
| | - Laura Maggi
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy
| | - Marie-Ève Tremblay
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada.
| |
Collapse
|
15
|
Bellingacci L, Canonichesi J, Mancini A, Parnetti L, Di Filippo M. Cytokines, synaptic plasticity and network dynamics: a matter of balance. Neural Regen Res 2023; 18:2569-2572. [PMID: 37449591 DOI: 10.4103/1673-5374.371344] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/18/2023] Open
Abstract
The modern view of the immune system as a sensitizing and modulating machinery of the central nervous system is now well recognized. However, the specific mechanisms underlying this fine crosstalk have yet to be fully disentangled. To control cognitive function and behavior, the two systems are engaged in a subtle interacting act. In this scenario, a dual action of pro-inflammatory cytokines in the modulation of brain network connections is emerging. Pro-inflammatory cytokines are indeed required to express physiological plasticity in the hippocampal network while being detrimental when over-expressed during uncontrolled inflammatory processes. In this dynamic equilibrium, synaptic functioning and the performance of neural networks are ensured by maintaining an appropriate balance between pro- and anti-inflammatory molecules in the central nervous system microenvironment.
Collapse
Affiliation(s)
- Laura Bellingacci
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Jacopo Canonichesi
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Andrea Mancini
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Lucilla Parnetti
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Massimiliano Di Filippo
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| |
Collapse
|
16
|
Singh J, Habean ML, Panicker N. Inflammasome assembly in neurodegenerative diseases. Trends Neurosci 2023; 46:814-831. [PMID: 37633753 PMCID: PMC10530301 DOI: 10.1016/j.tins.2023.07.009] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 06/26/2023] [Accepted: 07/31/2023] [Indexed: 08/28/2023]
Abstract
Neurodegenerative disorders are characterized by the progressive dysfunction and death of selectively vulnerable neuronal populations, often associated with the accumulation of aggregated host proteins. Sustained brain inflammation and hyperactivation of inflammasome complexes have been increasingly demonstrated to contribute to neurodegenerative disease progression. Here, we review molecular mechanisms leading to inflammasome assembly in neurodegeneration. We focus primarily on four degenerative brain disorders in which inflammasome hyperactivation has been well documented: Alzheimer's disease (AD), Parkinson's disease (PD), multiple sclerosis (MS), and the spectrum of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). We discuss shared and divergent principles of inflammasome assembly across these disorders, and underscore the differences between neurodegeneration-associated inflammasome activation pathways and their peripheral-immune counterparts. We examine how aberrant assembly of inflammasome complexes may amplify pathology in neurodegeneration, including misfolded protein aggregation, and highlight prospects for neurotherapeutic interventions based on targeting inflammasome pathways.
Collapse
Affiliation(s)
- Jagjit Singh
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Maria L Habean
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA; Biomedical Scientist Training Program (Department of Neurosciences), Case Western Reserve University, Cleveland, OH, USA
| | - Nikhil Panicker
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA; Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH 44106, USA; Kent State University, Neurosciences, School of Biomedical Sciences, Cleveland, OH, USA.
| |
Collapse
|
17
|
González Olmo BM, Bettes MN, DeMarsh JW, Zhao F, Askwith C, Barrientos RM. Short-term high-fat diet consumption impairs synaptic plasticity in the aged hippocampus via IL-1 signaling. NPJ Sci Food 2023; 7:35. [PMID: 37460765 DOI: 10.1038/s41538-023-00211-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 07/06/2023] [Indexed: 07/20/2023] Open
Abstract
More Americans are consuming diets higher in saturated fats and refined sugars than ever before. These trends could have serious consequences for the older population because high-fat diet (HFD) consumption, known to induce neuroinflammation, has been shown to accelerate and aggravate memory declines. We have previously demonstrated that short-term HFD consumption, which does not evoke obesity-related comorbidities, produced profound impairments to hippocampal-dependent memory in aged rats. These impairments were precipitated by increases in proinflammatory cytokines, primarily interleukin-1 beta (IL-1β). Here, we explored the extent to which short-term HFD consumption disrupts hippocampal synaptic plasticity, as measured by long-term potentiation (LTP), in young adult and aged rats. We demonstrated that (1) HFD disrupted late-phase LTP in the hippocampus of aged, but not young adult rats, (2) HFD did not disrupt early-phase LTP, and (3) blockade of the IL-1 receptor rescued L-LTP in aged HFD-fed rats. These findings suggest that hippocampal memory impairments in aged rats following HFD consumption occur through the deterioration of synaptic plasticity and that IL-1β is a critical driver of that deterioration.
Collapse
Affiliation(s)
- Brigitte M González Olmo
- Department of Biomedical Education & Anatomy, Ohio State University, Columbus, OH, USA
- Institute for Behavioral Medicine Research, Ohio State University, Columbus, OH, USA
| | - Menaz N Bettes
- Institute for Behavioral Medicine Research, Ohio State University, Columbus, OH, USA
| | - James W DeMarsh
- Institute for Behavioral Medicine Research, Ohio State University, Columbus, OH, USA
| | - Fangli Zhao
- Department of Neuroscience, The Ohio State University, Columbus, OH, USA
| | - Candice Askwith
- Department of Neuroscience, The Ohio State University, Columbus, OH, USA
| | - Ruth M Barrientos
- Institute for Behavioral Medicine Research, Ohio State University, Columbus, OH, USA.
- Department of Neuroscience, The Ohio State University, Columbus, OH, USA.
- Department of Psychiatry and Behavioral Health, Ohio State University, Columbus, OH, USA.
- Chronic Brain Injury Program, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
18
|
Fingolimod prevents cognitive impairments following hypoxia-induced neonatal seizure by ameliorating the inflammation and oxidative stress in male and female juvenile rats. LEARNING AND MOTIVATION 2023. [DOI: 10.1016/j.lmot.2023.101874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
|
19
|
Mancini M, Natoli S, Gardoni F, Di Luca M, Pisani A. Dopamine Transmission Imbalance in Neuroinflammation: Perspectives on Long-Term COVID-19. Int J Mol Sci 2023; 24:ijms24065618. [PMID: 36982693 PMCID: PMC10056044 DOI: 10.3390/ijms24065618] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Revised: 03/09/2023] [Accepted: 03/13/2023] [Indexed: 03/17/2023] Open
Abstract
Dopamine (DA) is a key neurotransmitter in the basal ganglia, implicated in the control of movement and motivation. Alteration of DA levels is central in Parkinson’s disease (PD), a common neurodegenerative disorder characterized by motor and non-motor manifestations and deposition of alpha-synuclein (α-syn) aggregates. Previous studies have hypothesized a link between PD and viral infections. Indeed, different cases of parkinsonism have been reported following COVID-19. However, whether SARS-CoV-2 may trigger a neurodegenerative process is still a matter of debate. Interestingly, evidence of brain inflammation has been described in postmortem samples of patients infected by SARS-CoV-2, which suggests immune-mediated mechanisms triggering the neurological sequelae. In this review, we discuss the role of proinflammatory molecules such as cytokines, chemokines, and oxygen reactive species in modulating DA homeostasis. Moreover, we review the existing literature on the possible mechanistic interplay between SARS-CoV-2-mediated neuroinflammation and nigrostriatal DAergic impairment, and the cross-talk with aberrant α-syn metabolism.
Collapse
Affiliation(s)
- Maria Mancini
- Department of Brain and Behavioral Sciences, University of Pavia, 27100 Pavia, Italy;
- IRCCS Mondino Foundation, 27100 Pavia, Italy
| | - Silvia Natoli
- Department of Clinical Science and Translational Medicine, University of Rome Tor Vergata, 00133 Rome, Italy;
- IRCCS Maugeri Pavia, 27100 Pavia, Italy
| | - Fabrizio Gardoni
- Department of Pharmacological and Biomolecular Sciences “Rodolfo Paoletti”, University of Milan, 20133 Milan, Italy; (F.G.); (M.D.L.)
| | - Monica Di Luca
- Department of Pharmacological and Biomolecular Sciences “Rodolfo Paoletti”, University of Milan, 20133 Milan, Italy; (F.G.); (M.D.L.)
| | - Antonio Pisani
- Department of Brain and Behavioral Sciences, University of Pavia, 27100 Pavia, Italy;
- IRCCS Mondino Foundation, 27100 Pavia, Italy
- Correspondence: ; Tel.: +39-0382-380-247
| |
Collapse
|
20
|
Pascoal VDB, Marchesini RB, Athié MCP, Matos AHB, Conte FF, Pereira TC, Secolin R, Gilioli R, Malheiros JM, Polli RS, Tannús A, Covolan L, Pascoal LB, Vieira AS, Cavalheiro EA, Cendes F, Lopes-Cendes I. Modulating Expression of Endogenous Interleukin 1 Beta in the Acute Phase of the Pilocarpine Model of Epilepsy May Change Animal Survival. Cell Mol Neurobiol 2023; 43:367-380. [PMID: 35061107 DOI: 10.1007/s10571-022-01190-y] [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: 06/04/2021] [Accepted: 01/05/2022] [Indexed: 01/07/2023]
Abstract
The pilocarpine-induced (PILO) model has helped elucidate the electrophysiological and molecular aspects related to mesial temporal lobe epilepsy. It has been suggested that the extensive cell death and edema observed in the brains of these animals could be induced by increased inflammatory responses, such as the rapid release of the inflammatory cytokine interleukin 1 beta (Il1b). In this study, we investigate the role of endogenous Il1b in the acute phase of the PILO model. Our aim is twofold. First, we want to determine whether it is feasible to silence Il1b in the central nervous system using a non-invasive procedure. Second, we aim to investigate the effect of silencing endogenous Il1b and its antagonist, Il1rn.We used RNA interference applied non-invasively to knockdown Il1b and its endogenous antagonist Il1rn. We found that knocking down Il1b prior to pilocarpine injection increased the mortality rate of treated animals. Furthermore, we observed that, when exposing the animals to more Il1b by silencing its endogenous antagonist Il1rn, there was a better response to status epilepticus with decreased animal mortality in the acute phase of the PILO model. Thus, we show the feasibility of using a novel, less invasive approach to study genes involved in the inflammatory response in the central nervous system. Furthermore, our results provide suggestive evidence that modulating endogenous Il1b improves animal survival in the acute phase of the PILO model and may have effects that extend into the chronic phase.
Collapse
Affiliation(s)
- V D B Pascoal
- Department of Translational Medicine, School of Medical Sciences, University of Campinas (UNICAMP), and the Brazilian Institute of Neuroscience and Neurotechnology (BRAINN), Tessália Vieira de Camargo, 126, Cidade Universitária "Zeferino Vaz", Campinas, SP, 13083-887, Brazil
- Department of Basic Science, Fluminense Federal University, Nova Friburgo, RJ, Brazil
| | - R B Marchesini
- Department of Translational Medicine, School of Medical Sciences, University of Campinas (UNICAMP), and the Brazilian Institute of Neuroscience and Neurotechnology (BRAINN), Tessália Vieira de Camargo, 126, Cidade Universitária "Zeferino Vaz", Campinas, SP, 13083-887, Brazil
| | - M C P Athié
- Department of Translational Medicine, School of Medical Sciences, University of Campinas (UNICAMP), and the Brazilian Institute of Neuroscience and Neurotechnology (BRAINN), Tessália Vieira de Camargo, 126, Cidade Universitária "Zeferino Vaz", Campinas, SP, 13083-887, Brazil
| | - A H B Matos
- Department of Translational Medicine, School of Medical Sciences, University of Campinas (UNICAMP), and the Brazilian Institute of Neuroscience and Neurotechnology (BRAINN), Tessália Vieira de Camargo, 126, Cidade Universitária "Zeferino Vaz", Campinas, SP, 13083-887, Brazil
| | - F F Conte
- Department of Translational Medicine, School of Medical Sciences, University of Campinas (UNICAMP), and the Brazilian Institute of Neuroscience and Neurotechnology (BRAINN), Tessália Vieira de Camargo, 126, Cidade Universitária "Zeferino Vaz", Campinas, SP, 13083-887, Brazil
| | - T C Pereira
- Department of Translational Medicine, School of Medical Sciences, University of Campinas (UNICAMP), and the Brazilian Institute of Neuroscience and Neurotechnology (BRAINN), Tessália Vieira de Camargo, 126, Cidade Universitária "Zeferino Vaz", Campinas, SP, 13083-887, Brazil
- Department of Biology, Faculty of Philosophy, Sciences and Letters at Ribeirao Preto, University of Sao Paulo (USP), Ribeirao Preto, SP, Brazil
| | - R Secolin
- Department of Translational Medicine, School of Medical Sciences, University of Campinas (UNICAMP), and the Brazilian Institute of Neuroscience and Neurotechnology (BRAINN), Tessália Vieira de Camargo, 126, Cidade Universitária "Zeferino Vaz", Campinas, SP, 13083-887, Brazil
- Department of Basic Science, Fluminense Federal University, Nova Friburgo, RJ, Brazil
| | - R Gilioli
- Multidisciplinary Centre for Biological Investigation (CEMIB), University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - J M Malheiros
- Centro de Imagens e Espectroscopia por Ressonancia Magnetica (CIERMag), Institute of Physics, University of Sao Paulo (USP), Sao Carlos, SP, Brazil
- Department of Physiology, Federal University of Sao Paulo (UNIFESP), Sao Paulo, SP, Brazil
| | - R S Polli
- Institute of Science and Technology, Federal University of São Paulo, São José dos Campos, SP, Brazil
| | - A Tannús
- Centro de Imagens e Espectroscopia por Ressonancia Magnetica (CIERMag), Institute of Physics, University of Sao Paulo (USP), Sao Carlos, SP, Brazil
| | - L Covolan
- Department of Physiology, Federal University of Sao Paulo (UNIFESP), Sao Paulo, SP, Brazil
| | - L B Pascoal
- Laboratory of Cell Signaling, School of Medical Sciences, University of Campinas - (UNICAMP), Campinas, SP, Brazil
| | - A S Vieira
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas - (UNICAMP), Campinas, SP, Brazil
| | - E A Cavalheiro
- Department of Neurology and Neurosurgery, Federal University of Sao Paulo, (UNIFESP), Sao Paulo, SP, Brazil
| | - F Cendes
- Department of Neurology, School of Medical Sciences, University of Campinas - (UNICAMP); and the Brazilian Institute of Neuroscience and Neurotechnology (BRAINN), Campinas, SP, Brazil
| | - I Lopes-Cendes
- Department of Translational Medicine, School of Medical Sciences, University of Campinas (UNICAMP), and the Brazilian Institute of Neuroscience and Neurotechnology (BRAINN), Tessália Vieira de Camargo, 126, Cidade Universitária "Zeferino Vaz", Campinas, SP, 13083-887, Brazil.
| |
Collapse
|
21
|
Zhang Y, Zhang M. Systemic inflammatory response syndrome-mediated neuronal plasticity in the central nervous system contributes to neurocognitive complications of extracorporeal membrane oxygenation. ALL LIFE 2022. [DOI: 10.1080/26895293.2022.2154857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Affiliation(s)
- Ying Zhang
- Department of Cardiovascular Surgery, Xi’an International Medical Center Hospital, Xi’an, People’s Republic of China
| | - Ming Zhang
- Department of Basic Medical Laboratory, The General Hospital of Western Theater Command, Chengdu, People’s Republic of China
| |
Collapse
|
22
|
De Felice E, Gonçalves de Andrade E, Golia MT, González Ibáñez F, Khakpour M, Di Castro MA, Garofalo S, Di Pietro E, Benatti C, Brunello N, Tascedda F, Kaminska B, Limatola C, Ragozzino D, Tremblay ME, Alboni S, Maggi L. Microglial diversity along the hippocampal longitudinal axis impacts synaptic plasticity in adult male mice under homeostatic conditions. J Neuroinflammation 2022; 19:292. [PMID: 36482444 PMCID: PMC9730634 DOI: 10.1186/s12974-022-02655-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 11/22/2022] [Indexed: 12/13/2022] Open
Abstract
The hippocampus is a plastic brain area that shows functional segregation along its longitudinal axis, reflected by a higher level of long-term potentiation (LTP) in the CA1 region of the dorsal hippocampus (DH) compared to the ventral hippocampus (VH), but the mechanisms underlying this difference remain elusive. Numerous studies have highlighted the importance of microglia-neuronal communication in modulating synaptic transmission and hippocampal plasticity, although its role in physiological contexts is still largely unknown. We characterized in depth the features of microglia in the two hippocampal poles and investigated their contribution to CA1 plasticity under physiological conditions. We unveiled the influence of microglia in differentially modulating the amplitude of LTP in the DH and VH, showing that minocycline or PLX5622 treatment reduced LTP amplitude in the DH, while increasing it in the VH. This was recapitulated in Cx3cr1 knockout mice, indicating that microglia have a key role in setting the conditions for plasticity processes in a region-specific manner, and that the CX3CL1-CX3CR1 pathway is a key element in determining the basal level of CA1 LTP in the two regions. The observed LTP differences at the two poles were associated with transcriptional changes in the expression of genes encoding for Il-1, Tnf-α, Il-6, and Bdnf, essential players of neuronal plasticity. Furthermore, microglia in the CA1 SR region showed an increase in soma and a more extensive arborization, an increased prevalence of immature lysosomes accompanied by an elevation in mRNA expression of phagocytic markers Mertk and Cd68 and a surge in the expression of microglial outward K+ currents in the VH compared to DH, suggesting a distinct basal phenotypic state of microglia across the two hippocampal poles. Overall, we characterized the molecular, morphological, ultrastructural, and functional profile of microglia at the two poles, suggesting that modifications in hippocampal subregions related to different microglial statuses can contribute to dissect the phenotypical aspects of many diseases in which microglia are known to be involved.
Collapse
Affiliation(s)
- E. De Felice
- grid.7841.aDepartment of Physiology and Pharmacology, Sapienza University of Rome, Piazzale Aldo Moro, 5, 00185 Rome, Italy
| | - E. Gonçalves de Andrade
- grid.143640.40000 0004 1936 9465Division of Medical Sciences, University of Victoria, Victoria, Canada
| | - M. T. Golia
- grid.7841.aDepartment of Physiology and Pharmacology, Sapienza University of Rome, Piazzale Aldo Moro, 5, 00185 Rome, Italy
| | - F. González Ibáñez
- grid.143640.40000 0004 1936 9465Division of Medical Sciences, University of Victoria, Victoria, Canada ,grid.411081.d0000 0000 9471 1794Faculté de Médecine and Centre de Recherche, CHU de Québec-Université Laval, Quebec, Canada
| | - M. Khakpour
- grid.143640.40000 0004 1936 9465Division of Medical Sciences, University of Victoria, Victoria, Canada
| | - M. A. Di Castro
- grid.7841.aDepartment of Physiology and Pharmacology, Sapienza University of Rome, Piazzale Aldo Moro, 5, 00185 Rome, Italy
| | - S. Garofalo
- grid.7841.aDepartment of Physiology and Pharmacology, Sapienza University of Rome, Piazzale Aldo Moro, 5, 00185 Rome, Italy
| | - E. Di Pietro
- grid.7841.aDepartment of Physiology and Pharmacology, Sapienza University of Rome, Piazzale Aldo Moro, 5, 00185 Rome, Italy
| | - C. Benatti
- grid.7548.e0000000121697570Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy ,grid.7548.e0000000121697570Centre of Neuroscience and Neurotechnology, University of Modena and Reggio Emilia, Modena, Italy
| | - N. Brunello
- grid.7548.e0000000121697570Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - F. Tascedda
- grid.7548.e0000000121697570Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy ,grid.7548.e0000000121697570Centre of Neuroscience and Neurotechnology, University of Modena and Reggio Emilia, Modena, Italy
| | - B. Kaminska
- grid.419305.a0000 0001 1943 2944Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology of the Polish Academy of Sciences, Warsaw, Poland
| | - C. Limatola
- grid.419543.e0000 0004 1760 3561IRCCS Neuromed, Pozzilli, Italy ,grid.7841.aDepartment of Physiology and Pharmacology, Laboratory Affiliated to Istituto Pasteur, Sapienza University, Rome, Italy
| | - D. Ragozzino
- grid.7841.aDepartment of Physiology and Pharmacology, Sapienza University of Rome, Piazzale Aldo Moro, 5, 00185 Rome, Italy ,grid.417778.a0000 0001 0692 3437Santa Lucia Foundation (IRCCS Fondazione Santa Lucia), Rome, Italy
| | - M. E. Tremblay
- grid.143640.40000 0004 1936 9465Division of Medical Sciences, University of Victoria, Victoria, Canada ,grid.411081.d0000 0000 9471 1794Faculté de Médecine and Centre de Recherche, CHU de Québec-Université Laval, Quebec, Canada
| | - S. Alboni
- grid.7548.e0000000121697570Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy ,grid.7548.e0000000121697570Centre of Neuroscience and Neurotechnology, University of Modena and Reggio Emilia, Modena, Italy
| | - L. Maggi
- grid.7841.aDepartment of Physiology and Pharmacology, Sapienza University of Rome, Piazzale Aldo Moro, 5, 00185 Rome, Italy
| |
Collapse
|
23
|
Artık A, Öztelcan Gündüz B, Mızrak S, Işık Ü. Increased serum levels of tumour necrosis factor-like weak inducer of apoptosis in children with autism spectrum disorder. INTERNATIONAL JOURNAL OF DEVELOPMENTAL DISABILITIES 2022; 69:611-616. [PMID: 37346259 PMCID: PMC10281418 DOI: 10.1080/20473869.2022.2143033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Revised: 10/10/2022] [Accepted: 10/30/2022] [Indexed: 06/23/2023]
Abstract
Introduction: A previous study has evaluated the association between serum tumour necrosis factor-like weak apoptosis inducer (TWEAK) levels and autism spectrum disorder (ASD). In line with this investigation, the present study aimed to measure serum TWEAK levels to determine whether their eventual alteration might have etiopathogenetic significance in children with ASD. Methods: A total of 40 treatment-naive children with ASD and 40 healthy children as controls were included in the present study. The Schedule for Affective Disorders and Schizophrenia for School-Aged Children-Present and Lifetime Version, DSM-5 was used by a psychiatrist to screen the healthy controls for psychiatric disorders after a physical examination by a paediatrician. The clinical severity of the ASD symptoms was assessed by the Childhood Autism Rating Scale. Venous blood samples were collected, and serum TWEAK levels were measured. Results: This study included 80 children in total, with 40 (50.0%) in the patient group and 40 (50.0%) in the healthy control group. Thirty four (85.0%) of the participants in the patient group, and 31 (77.5%) in the healthy control group, were male, and the remainder were female. The distribution of the gender ratio was statistically similar between groups (p = 0.568). The volunteers were between 36 and 59 months old. The average age in the patient group was 46.0 ± 6.5, while that in the healthy control group was 45.2 ± 6.7. The ages were also statistically similar between groups (p = 0.615). The TWEAK values of the patient group were found to be statistically higher than those of the healthy control group (p < 0.001). Discussion: This study examined whether serum TWEAK levels were related to ASD in childhood. Our findings indicate that children with ASD have higher TWEAK levels when compared to other children. The findings further indicate that serum TWEAK levels could be related to ASD etiopathogenesis independent of ASD symptom severity.
Collapse
Affiliation(s)
- Abdülbaki Artık
- Faculty of Medicine, Child and Adolescent Mental Health Department, Uşak University, Ankara, Turkey
| | | | - Soycan Mızrak
- Faculty of Medicine, Department of Medical Biochemistry, Uşak University, Ankara, Turkey
| | - Ümit Işık
- Faculty of Medicine, Child and Adolescent Mental Health Department, Süleyman Demirel University, Ankara, Turkey
| |
Collapse
|
24
|
Patel P, Faldu K, Borisa A, Bhatt H, Shah J. Insights of Valacyclovir in Treatment of Alzheimer's Disease: Computational Docking Studies and Scopolamine Rat Model. Curr Neurovasc Res 2022; 19:344-357. [PMID: 36089794 DOI: 10.2174/1567202619666220908125125] [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: 07/05/2022] [Revised: 07/26/2022] [Accepted: 08/01/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND Alzheimer's Disease (AD) impairs memory and cognitive functions in the geriatric population and is characterized by intracellular deposition of neurofibrillary tangles, extracellular deposition of amyloid plaques, and neuronal degeneration. Literature suggests that latent viral infections in the brain act as prions and promote neurodegeneration. Memantine possesses both anti-viral and N-methyl-D-aspartate (NMDA) receptor antagonistic activity. OBJECTIVES This research was designed to evaluate the efficacy of antiviral agents, especially valacyclovir, a prodrug of acyclovir in ameliorating the pathology of AD based on the presumption that anti-viral agents targeting the Herpes Simplex Virus (HSV) can have a protective effect on neurodegenerative diseases like Alzheimer's disease. METHODS Thus, we evaluated acyclovir's potential activity by in-silico computational docking studies against acetylcholinesterase (AChE), butyrylcholinesterase (BuChE), and beta-secretase 1 (BACE-1). These findings were further evaluated by in-vivo scopolamine-induced cognitive impairment in rats. Two doses of valacyclovir, a prodrug of acyclovir (100 mg/kg and 150 mg/kg orally) were tested. RESULTS Genetic Optimisation for Ligand Docking scores and fitness scores of acyclovir were comparable to donepezil. Valacyclovir improved neurobehavioral markers. It inhibited AChE and BuChE (p<0.001) enzymes. It also possessed disease-modifying efficacy as it decreased the levels of BACE-1 (p<0.001), amyloid beta 1-42 (p<0.001), amyloid beta 1-40 (p<0.001), phosphorylatedtau (p<0.001), neprilysin (p<0.01), and insulin-degrading enzyme. It ameliorated neuroinflammation through decreased levels of tumour necrosis factor α (p<0.001), nuclear factor-kappa B (p<0.001), interleukin 6 (p<0.001), interleukin 1 beta (p<0.001), and interferon-gamma (p<0.001). It also maintained synaptic plasticity and consolidated memory. Histopathology showed that valacyclovir could restore cellular density and also preserve the dentate gyrus. CONCLUSION Valacyclovir showed comparable activity to donepezil and thus can be further researched for the treatment of Alzheimer's disease.
Collapse
Affiliation(s)
- Parmi Patel
- Department of Pharmacology, Institute of Pharmacy, Nirma University, Ahmedabad, Gujarat, India
| | - Khushboo Faldu
- Department of Pharmacology, Institute of Pharmacy, Nirma University, Ahmedabad, Gujarat, India
| | - Ankit Borisa
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Nirma University, Ahmedabad, Gujarat, India
| | - Hardik Bhatt
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Nirma University, Ahmedabad, Gujarat, India
| | - Jigna Shah
- Department of Pharmacology, Institute of Pharmacy, Nirma University, Ahmedabad, Gujarat, India
| |
Collapse
|
25
|
Acrylamide inhibits long-term potentiation and learning involving microglia and pro-inflammatory signaling. Sci Rep 2022; 12:12429. [PMID: 35858988 PMCID: PMC9300615 DOI: 10.1038/s41598-022-16762-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 07/14/2022] [Indexed: 11/16/2022] Open
Abstract
Acrylamide is a chemical used in various industries and a product following high-temperature cooking of vegetables containing asparagine. Environmental or dietary exposure to acrylamide could impair cognitive function because of its neurotoxicity. Using rat hippocampal slices, we tested whether acrylamide alters induction of long-term potentiation (LTP), a cellular model of learning and memory. We hypothesized that acrylamide impairs cognitive function via activation of pro-inflammatory cytokines because robust upregulation of NLRP3 inflammasome has been reported. Although acrylamide up to 3 mM did not alter basal synaptic transmission, incubation with 10 μM or acute administration of 100 μM acrylamide inhibited induction of LTP. Inhibitors of toll-like receptor 4 (TLR4), and minocycline, an inhibitor of microglial activation, overcame the effects of acrylamide on LTP induction. Furthermore, we observed that acrylamide failed to inhibit LTP after administration of MCC950, an inhibitor of NLRP3, or in the presence of Interleukin-1 receptor antagonist (IL-1Ra). We also found that in vivo acrylamide injection transiently impaired body weight gain and impaired one-trial inhibitory avoidance learning. This learning deficit was overcome by MCC950. These results indicate that cognitive impairment by acrylamide is mediated by mechanisms involving microglia and release of cytokines via NLRP3 activation.
Collapse
|
26
|
Baek SH, Kim H, Kim JW, Ryu S, Lee JY, Kim JM, Shin IS, Kim SW. Association between Peripheral Inflammatory Cytokines and Cognitive Function in Patients with First-Episode Schizophrenia. J Pers Med 2022; 12:jpm12071137. [PMID: 35887634 PMCID: PMC9317024 DOI: 10.3390/jpm12071137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 07/07/2022] [Accepted: 07/10/2022] [Indexed: 11/25/2022] Open
Abstract
In this study, we investigated the impact of inflammatory cytokines on the cognitive performance of patients with schizophrenia. The included patients met the criteria for schizophrenia spectrum disorder and were aged between 15 and 40 years, with a duration of illness ≤1 year. Plasma tumor necrosis factor (TNF)-α; interferon-γ; and interleukin (IL)-1β, IL-6, IL-8, IL-10, and IL-12 levels were measured. A computerized neurocognitive battery, measures for social cognitive function, and clinical measures were administered. A total of 174 patients with first-episode psychosis were enrolled. The TNF-α level was negatively correlated with scores on the digit span, verbal learning, and Wisconsin card sorting tests, and the number of correct responses on the continuous performance test (CR-CPT), whereas a positive correlation was detected with the trail making test (TMT)-B time. The interferon-γ level was negatively correlated with performance on the false belief and visual learning tests. The IL-1β level was positively correlated with the TMT-A time and CPT reaction time, whereas it was negatively correlated with the CR-CPT and performance on the visual learning and social cognitive tests. The IL-12 level was negatively correlated with the CR-CPT and false belief test. Our results suggest that proinflammatory cytokines are associated with cognitive impairment in patients with schizophrenia.
Collapse
Affiliation(s)
- Seon-Hwa Baek
- Department of Psychiatry, Chonnam National University Medical School, Gwangju 61419, Korea; (S.-H.B.); (H.K.); (J.-W.K.); (S.R.); (J.-Y.L.); (J.-M.K.); (I.-S.S.)
| | - Honey Kim
- Department of Psychiatry, Chonnam National University Medical School, Gwangju 61419, Korea; (S.-H.B.); (H.K.); (J.-W.K.); (S.R.); (J.-Y.L.); (J.-M.K.); (I.-S.S.)
| | - Ju-Wan Kim
- Department of Psychiatry, Chonnam National University Medical School, Gwangju 61419, Korea; (S.-H.B.); (H.K.); (J.-W.K.); (S.R.); (J.-Y.L.); (J.-M.K.); (I.-S.S.)
| | - Seunghyong Ryu
- Department of Psychiatry, Chonnam National University Medical School, Gwangju 61419, Korea; (S.-H.B.); (H.K.); (J.-W.K.); (S.R.); (J.-Y.L.); (J.-M.K.); (I.-S.S.)
| | - Ju-Yeon Lee
- Department of Psychiatry, Chonnam National University Medical School, Gwangju 61419, Korea; (S.-H.B.); (H.K.); (J.-W.K.); (S.R.); (J.-Y.L.); (J.-M.K.); (I.-S.S.)
| | - Jae-Min Kim
- Department of Psychiatry, Chonnam National University Medical School, Gwangju 61419, Korea; (S.-H.B.); (H.K.); (J.-W.K.); (S.R.); (J.-Y.L.); (J.-M.K.); (I.-S.S.)
| | - Il-Seon Shin
- Department of Psychiatry, Chonnam National University Medical School, Gwangju 61419, Korea; (S.-H.B.); (H.K.); (J.-W.K.); (S.R.); (J.-Y.L.); (J.-M.K.); (I.-S.S.)
| | - Sung-Wan Kim
- Department of Psychiatry, Chonnam National University Medical School, Gwangju 61419, Korea; (S.-H.B.); (H.K.); (J.-W.K.); (S.R.); (J.-Y.L.); (J.-M.K.); (I.-S.S.)
- Mindlink, Gwangju Bukgu Mental Health Center, Gwangju 61220, Korea
- Correspondence: ; Tel.: +82-62-220-6148
| |
Collapse
|
27
|
Darwish B, Chamaa F, Awada B, Lawand N, Saadé NE, Abou Fayad AG, Abou-Kheir W. Urinary Tract Infections Impair Adult Hippocampal Neurogenesis. BIOLOGY 2022; 11:891. [PMID: 35741412 PMCID: PMC9220213 DOI: 10.3390/biology11060891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 04/11/2022] [Accepted: 04/12/2022] [Indexed: 11/20/2022]
Abstract
Previous studies have suggested a link between urinary tract infections (UTIs) and cognitive impairment. One possible contributing factor for UTI-induced cognitive changes that has not yet been investigated is a potential alteration in hippocampal neurogenesis. In this study, we aim to investigate the effect of UTI on brain plasticity by specifically examining alterations in neurogenesis. Adult male Sprague Dawley rats received an intra-urethral injection of an Escherichia coli (E. coli) clinical isolate (108 CFU/mL). We found that rats with a UTI (CFU/mL ≥ 105) had reduced proliferation of neural stem cells (NSCs) at an early time point post infection (day 4) and neurogenesis at a later time point (day 34). This was associated with the decreased expression in mRNA of BDNF, NGF, and FGF2, and elevated expression of IL-1β in the hippocampus at 6 h post infection, but with no changes in optical intensity of the microglia and astrocytes. In addition, infected rats spent less time exploring a novel arm in the Y-maze test. Treatment with an anti-inflammatory drug did not revert the effect on NSCs, while treatment with antibiotics further decreased the basal level of their proliferation. This study presents novel findings on the impact of urinary tract infections on hippocampal neurogenesis that could be correlated with cognitive impairment.
Collapse
Affiliation(s)
- Batoul Darwish
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut P.O. Box 11-0236, Lebanon; (B.D.); (F.C.); (N.L.); (N.E.S.)
| | - Farah Chamaa
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut P.O. Box 11-0236, Lebanon; (B.D.); (F.C.); (N.L.); (N.E.S.)
- Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology, Thuwal 23955, Saudi Arabia
| | - Bassel Awada
- Department of Experimental Pathology, Immunology and Microbiology, Faculty of Medicine, American University of Beirut, Beirut P.O. Box 11-0236, Lebanon; (B.A.); (A.G.A.F.)
- Center for Infectious Diseases Research, American University of Beirut, Beirut P.O. Box 11-0236, Lebanon
| | - Nada Lawand
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut P.O. Box 11-0236, Lebanon; (B.D.); (F.C.); (N.L.); (N.E.S.)
- Department of Neurology, Faculty of Medicine, American University of Beirut, Beirut P.O. Box 11-0236, Lebanon
| | - Nayef E. Saadé
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut P.O. Box 11-0236, Lebanon; (B.D.); (F.C.); (N.L.); (N.E.S.)
| | - Antoine G. Abou Fayad
- Department of Experimental Pathology, Immunology and Microbiology, Faculty of Medicine, American University of Beirut, Beirut P.O. Box 11-0236, Lebanon; (B.A.); (A.G.A.F.)
- Center for Infectious Diseases Research, American University of Beirut, Beirut P.O. Box 11-0236, Lebanon
| | - Wassim Abou-Kheir
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut P.O. Box 11-0236, Lebanon; (B.D.); (F.C.); (N.L.); (N.E.S.)
| |
Collapse
|
28
|
Role of Cannabinoid CB2 Receptor in Alcohol Use Disorders: From Animal to Human Studies. Int J Mol Sci 2022; 23:ijms23115908. [PMID: 35682586 PMCID: PMC9180470 DOI: 10.3390/ijms23115908] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 05/22/2022] [Accepted: 05/23/2022] [Indexed: 12/04/2022] Open
Abstract
Cumulative evidence has pointed out cannabinoid CB2 receptors (CB2r) as a potential therapeutic key target for treating alcohol use disorder (AUD). This review provides the most relevant results obtained from rodent and human studies, including an integrative section focused on the involvement of CB2r in the neurobiology of alcohol addiction. A literature search was conducted using the electronic databases Medline and Scopus for articles. The search strategy was as follows: “Receptor, Cannabinoid, CB2” AND “Alcohol-Related Disorders” AND “human/or patients”; “Receptor, Cannabinoid, CB2” AND “Alcohol” OR “Ethanol” AND “rodents/or mice/or rats”. Pharmacological approaches demonstrated that the activation or blockade of CB2r modulated different alcohol-addictive behaviors. Rodent models of alcoholism revealed significant alterations of CB2r in brain areas of the reward system. In addition, mice lacking CB2r (CB2KO) show increased alcohol consumption, motivation, and relapse alterations. It has been stressed that the potential neurobiological mechanisms underlying their behavioral effects involve critical elements of the alcohol reward system. Interestingly, recent postmortem studies showed CNR2 alterations in brain areas of alcoholic patients. Moreover, although the number of studies is limited, the results revealed an association between some genetic alterations of the CNR2 and an increased risk for developing AUD. This review provides evidence that CB2r may play a role in alcohol addiction. Clinical studies are necessary to figure out whether CB2r ligands may prove useful for the treatment of AUD in humans.
Collapse
|
29
|
Hippocampal Cytokine Release in Experimental Epileptogenesis—A Longitudinal In Vivo Microdialysis Study. Brain Sci 2022; 12:brainsci12050677. [PMID: 35625063 PMCID: PMC9139593 DOI: 10.3390/brainsci12050677] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 05/13/2022] [Accepted: 05/18/2022] [Indexed: 12/27/2022] Open
Abstract
Background: Inflammation, particularly cytokine release, contributes to epileptogenesis by influencing the cerebral tissue remodeling and neuronal excitability that occurs after a precipitating epileptogenic insult. While several cytokines have been explored in this process, release kinetics are less well investigated. Determining the time course of cytokine release in the epileptogenic zone is necessary for precisely timed preventive or therapeutic anti-inflammatory interventions. Methods: Hippocampal extracellular levels of six cytokines and chemokines (IL-1β, IL-6, IL-10, CCL2, CCL3, and CCL5) were quantified at various time points during epileptogenesis in a rat model of mesial temporal lobe epilepsy with hippocampal sclerosis (mTLE-HS) using microdialysis (MD). Results: The analysis of microdialysates demonstrated consistent elevation at all time points during epileptogenesis for IL-1β and IL-10. IL-10 release was maximal on day 1, IL-1β release peaked at day 8. No correlation between local hippocampal IL-1β concentrations and IL-1β blood levels was found. Conclusion: The release kinetics of IL-1β are consistent with its established pro-epileptogenic properties, while the kinetics of IL-10 suggest a counter-regulatory effect. This proof-of-concept study demonstrates the feasibility of intraindividual longitudinal monitoring of hippocampal molecular inflammatory processes via repetitive MD over several weeks and sheds light on the kinetics of hippocampal cytokine release during epileptogenesis.
Collapse
|
30
|
Cathomas F, Holt LM, Parise EM, Liu J, Murrough JW, Casaccia P, Nestler EJ, Russo SJ. Beyond the neuron: Role of non-neuronal cells in stress disorders. Neuron 2022; 110:1116-1138. [PMID: 35182484 PMCID: PMC8989648 DOI: 10.1016/j.neuron.2022.01.033] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 12/15/2021] [Accepted: 01/24/2022] [Indexed: 12/11/2022]
Abstract
Stress disorders are leading causes of disease burden in the U.S. and worldwide, yet available therapies are fully effective in less than half of all individuals with these disorders. Although to date, much of the focus has been on neuron-intrinsic mechanisms, emerging evidence suggests that chronic stress can affect a wide range of cell types in the brain and periphery, which are linked to maladaptive behavioral outcomes. Here, we synthesize emerging literature and discuss mechanisms of how non-neuronal cells in limbic regions of brain interface at synapses, the neurovascular unit, and other sites of intercellular communication to mediate the deleterious, or adaptive (i.e., pro-resilient), effects of chronic stress in rodent models and in human stress-related disorders. We believe that such an approach may one day allow us to adopt a holistic "whole body" approach to stress disorder research, which could lead to more precise diagnostic tests and personalized treatment strategies. Stress is a major risk factor for many psychiatric disorders. Cathomas et al. review new insight into how non-neuronal cells mediate the deleterious effects, as well as the adaptive, protective effects, of stress in rodent models and human stress-related disorders.
Collapse
Affiliation(s)
- Flurin Cathomas
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Leanne M Holt
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Eric M Parise
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jia Liu
- Neuroscience Initiative, Advanced Science Research Center, Program in Biology and Biochemistry at The Graduate Center of The City University of New York, New York, NY, USA
| | - James W Murrough
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Patrizia Casaccia
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Neuroscience Initiative, Advanced Science Research Center, Program in Biology and Biochemistry at The Graduate Center of The City University of New York, New York, NY, USA
| | - Eric J Nestler
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Scott J Russo
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
31
|
Zheng Y, Reiner B, Liu J, Xu L, Xiong H. Methamphetamine augments HIV-1 gp120 inhibition of synaptic transmission and plasticity in rat hippocampal slices: Implications for methamphetamine exacerbation of HIV-associated neurocognitive disorders. Neurobiol Dis 2022; 168:105712. [PMID: 35337950 PMCID: PMC9150446 DOI: 10.1016/j.nbd.2022.105712] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 03/16/2022] [Accepted: 03/18/2022] [Indexed: 11/30/2022] Open
Abstract
Methamphetamine (Meth) abuse and human immunodeficiency virus type 1 (HIV-1) infection are two major public health problems worldwide. Being frequently comorbid with HIV-1 infection, Meth abuse exacerbates neurocognitive impairment in HIV-1-infected individuals even in the era of combined antiretroviral therapy. While a large body of research have studied the individual effects of Meth and HIV-1 envelope glycoprotein 120 (gp120) in the brain, far less has focused on their synergistic influence. Moreover, it is well-documented that the hippocampus is the primary site of spatial learning and long-term memory formation. Dysregulation of activity-dependent synaptic transmission and plasticity in the hippocampus is believed to impair neurocognitive function. To uncover the underlying mechanisms for increased incidence and severity of HIV-1-associated neurocognitive disorders (HAND) in HIV-1-infected patients with Meth abuse, we investigated acute individual and combined effects of Meth (20 μM) and gp120 (200 pM) on synaptic transmission and plasticity in the CA1 region of young adult male rat hippocampus, a brain region known to be vulnerable to HIV-1 infection. Our results showed that acute localized application of Meth and gp120 each alone onto the CA1 region reduced short-term dynamics of input-output responses and frequency facilitation, and attenuated long-term potentiation (LTP) induced by either high frequency stimulation or theta burst stimulation. A synergistic augmentation on activity-dependent synaptic plasticity was observed when Meth and gp120 were applied in combination. Paired-pulse facilitation results exhibited an altered facilitation ratio, suggesting a presynaptic site of action. Further studies revealed an involvement of microglia NLRP3 inflammasome activation in Meth augmentation of gp120-mediated attenuation of LTP. Taken together, our results demonstrated Meth augmented gp120 attenuation of LTP in the hippocampus. Since LTP is the accepted experimental analog of learning at the synaptic level, such augmentation may underlie Meth exacerbation of HAND observed clinically.
Collapse
Affiliation(s)
- Ya Zheng
- The Neurophysiology Laboratory, Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5880, USA
| | - Benjamin Reiner
- The Neurophysiology Laboratory, Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5880, USA
| | - Jianuo Liu
- The Neurophysiology Laboratory, Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5880, USA
| | - Linda Xu
- The Neurophysiology Laboratory, Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5880, USA
| | - Huangui Xiong
- The Neurophysiology Laboratory, Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5880, USA.
| |
Collapse
|
32
|
Welch C, Mulligan K. Does Bisphenol A Confer Risk of Neurodevelopmental Disorders? What We Have Learned from Developmental Neurotoxicity Studies in Animal Models. Int J Mol Sci 2022; 23:2894. [PMID: 35270035 PMCID: PMC8910940 DOI: 10.3390/ijms23052894] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 03/02/2022] [Accepted: 03/05/2022] [Indexed: 02/01/2023] Open
Abstract
Substantial evidence indicates that bisphenol A (BPA), a ubiquitous environmental chemical used in the synthesis of polycarbonate plastics and epoxy resins, can impair brain development. Clinical and epidemiological studies exploring potential connections between BPA and neurodevelopmental disorders in humans have repeatedly identified correlations between early BPA exposure and developmental disorders, such as attention deficit/hyperactivity disorder and autism spectrum disorder. Investigations using invertebrate and vertebrate animal models have revealed that developmental exposure to BPA can impair multiple aspects of neuronal development, including neural stem cell proliferation and differentiation, synapse formation, and synaptic plasticity-neuronal phenotypes that are thought to underpin the fundamental changes in behavior-associated neurodevelopmental disorders. Consistent with neuronal phenotypes caused by BPA, behavioral analyses of BPA-treated animals have shown significant impacts on behavioral endophenotypes related to neurodevelopmental disorders, including altered locomotor activity, learning and memory deficits, and anxiety-like behavior. To contextualize the correlations between BPA and neurodevelopmental disorders in humans, this review summarizes the current literature on the developmental neurotoxicity of BPA in laboratory animals with an emphasis on neuronal phenotypes, molecular mechanisms, and behavioral outcomes. The collective works described here predominantly support the notion that gestational exposure to BPA should be regarded as a risk factor for neurodevelopmental disorders.
Collapse
Affiliation(s)
- Chloe Welch
- Division of Biological Sciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA;
| | - Kimberly Mulligan
- Department of Biological Sciences, California State University, Sacramento, 6000 J Street, Sacramento, CA 95819, USA
| |
Collapse
|
33
|
Dutta SS, Andonova AA, Wöllert T, Hewett SJ, Hewett JA. P2X7-dependent constitutive Interleukin-1β release from pyramidal neurons of the normal mouse hippocampus: Evidence for a role in maintenance of the innate seizure threshold. Neurobiol Dis 2022; 168:105689. [DOI: 10.1016/j.nbd.2022.105689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 03/02/2022] [Accepted: 03/07/2022] [Indexed: 11/30/2022] Open
|
34
|
Cytokines in the Brain and Neuroinflammation: We Didn’t Starve the Fire! Pharmaceuticals (Basel) 2022; 15:ph15020140. [PMID: 35215252 PMCID: PMC8878213 DOI: 10.3390/ph15020140] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 01/23/2022] [Accepted: 01/24/2022] [Indexed: 12/19/2022] Open
Abstract
In spite of the brain-protecting tissues of the skull, meninges, and blood-brain barrier, some forms of injury to or infection of the CNS can give rise to cerebral cytokine production and action and result in drastic changes in brain function and behavior. Interestingly, peripheral infection-induced systemic inflammation can also be accompanied by increased cerebral cytokine production. Furthermore, it has been recently proposed that some forms of psychological stress may have similar CNS effects. Different conditions of cerebral cytokine production and action will be reviewed here against the background of neuroinflammation. Within this context, it is important to both deepen our understanding along already taken paths as well as to explore new ways in which neural functioning can be modified by cytokines. This, in turn, should enable us to put forward different modes of cerebral cytokine production and action in relation to distinct forms of neuroinflammation.
Collapse
|
35
|
Stampanoni Bassi M, Iezzi E, Centonze D. Multiple sclerosis: Inflammation, autoimmunity and plasticity. HANDBOOK OF CLINICAL NEUROLOGY 2022; 184:457-470. [PMID: 35034754 DOI: 10.1016/b978-0-12-819410-2.00024-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
In recent years, experimental studies have clarified that immune system influences the functioning of the central nervous system (CNS) in both physiologic and pathologic conditions. The neuro-immune crosstalk plays a crucial role in neuronal development and may be critically involved in mediating CNS response to neuronal damage. Multiple sclerosis (MS) represents a good model to investigate how the immune system regulates neuronal activity. Accordingly, a growing body of evidence has demonstrated that increased levels of pro-inflammatory mediators may significantly impact synaptic mechanisms, influencing overall neuronal excitability and synaptic plasticity expression. In this chapter, we provide an overview of preclinical data and clinical studies exploring synaptic functioning noninvasively with transcranial magnetic stimulation (TMS) in patients with MS. Moreover, we examine how inflammation-driven synaptic dysfunction could affect synaptic plasticity expression, negatively influencing the MS course. Contrasting CSF inflammation together with pharmacologic enhancement of synaptic plasticity and application of noninvasive brain stimulation, alone or in combination with rehabilitative treatments, could improve the clinical compensation and prevent the accumulating deterioration in MS.
Collapse
Affiliation(s)
| | - Ennio Iezzi
- Unit of Neurology & Neurorehabilitation, IRCCS Neuromed, Pozzilli, Italy
| | - Diego Centonze
- Unit of Neurology & Neurorehabilitation, IRCCS Neuromed, Pozzilli, Italy; Laboratory of Synaptic Immunopathology, Department of Systems Medicine, Tor Vergata University, Rome, Italy.
| |
Collapse
|
36
|
Costa A, Rani B, Bastiaanssen TFS, Bonfiglio F, Gunnigle E, Provensi G, Rossitto M, Boehme M, Strain C, Martínez CS, Blandina P, Cryan JF, Layé S, Corradetti R, Passani MB. Diet Prevents Social Stress-Induced Maladaptive Neurobehavioural and Gut Microbiota Changes in a Histamine-Dependent Manner. Int J Mol Sci 2022; 23:862. [PMID: 35055048 PMCID: PMC8775792 DOI: 10.3390/ijms23020862] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/11/2022] [Accepted: 01/12/2022] [Indexed: 12/30/2022] Open
Abstract
Exposure to repeated social stress may cause maladaptive emotional reactions that can be reduced by healthy nutritional supplementation. Histaminergic neurotransmission has a central role in orchestrating specific behavioural responses depending on the homeostatic state of a subject, but it remains to be established if it participates in the protective effects against the insults of chronic stress afforded by a healthy diet. By using C57BL/6J male mice that do not synthesize histamine (Hdc-/-) and their wild type (Hdc+/+) congeners we evaluated if the histaminergic system participates in the protective action of a diet enriched with polyunsaturated fatty acids and vitamin A on the deleterious effect of chronic stress. Behavioural tests across domains relevant to cognition and anxiety were performed. Hippocampal synaptic plasticity, cytokine expression, hippocampal fatty acids, oxylipins and microbiota composition were also assessed. Chronic stress induced social avoidance, poor recognition memory, affected hippocampal long-term potentiation, changed the microbiota profile, brain cytokines, fatty acid and oxylipins composition of both Hdc-/- and Hdc+/+ mice. Dietary enrichment counteracted stress-induced deficits only in Hdc+/+ mice as histamine deficiency prevented almost all the diet-related beneficial effects. Interpretation: Our results reveal a previously unexplored and novel role for brain histamine as a mediator of many favorable effects of the enriched diet. These data present long-reaching perspectives in the field of nutritional neuropsychopharmacology.
Collapse
Affiliation(s)
- Alessia Costa
- Dipartimento di Scienze della Salute, Universitá di Firenze, Viale Pieraccini 6, 50139 Firenze, Italy; (A.C.); (B.R.)
| | - Barbara Rani
- Dipartimento di Scienze della Salute, Universitá di Firenze, Viale Pieraccini 6, 50139 Firenze, Italy; (A.C.); (B.R.)
| | - Thomaz F. S. Bastiaanssen
- APC Microbiome Ireland, University College Cork, T12 YT20 Cork, Ireland; (T.F.S.B.); (E.G.); (M.B.); (C.S.); (C.S.M.); (J.F.C.)
- Department of Anatomy and Neuroscience, University College Cork, T12 YT20 Cork, Ireland
| | - Francesco Bonfiglio
- Dipartimento di Neuroscienze, Psicologia, Area del Farmaco e Salute del Bambino (NEUROFARBA), Universitá di Firenze, Viale Pieraccini 6, 50139 Firenze, Italy; (F.B.); (G.P.); (P.B.)
| | - Eoin Gunnigle
- APC Microbiome Ireland, University College Cork, T12 YT20 Cork, Ireland; (T.F.S.B.); (E.G.); (M.B.); (C.S.); (C.S.M.); (J.F.C.)
| | - Gustavo Provensi
- Dipartimento di Neuroscienze, Psicologia, Area del Farmaco e Salute del Bambino (NEUROFARBA), Universitá di Firenze, Viale Pieraccini 6, 50139 Firenze, Italy; (F.B.); (G.P.); (P.B.)
| | - Moira Rossitto
- Laboratoire NutriNeuro, UMR INRAE, Bordeaux INP, Université de Bordeaux, 146 Rue Léo Saignat, 33076 Bordeaux, France; (M.R.); (S.L.)
| | - Marcus Boehme
- APC Microbiome Ireland, University College Cork, T12 YT20 Cork, Ireland; (T.F.S.B.); (E.G.); (M.B.); (C.S.); (C.S.M.); (J.F.C.)
| | - Conall Strain
- APC Microbiome Ireland, University College Cork, T12 YT20 Cork, Ireland; (T.F.S.B.); (E.G.); (M.B.); (C.S.); (C.S.M.); (J.F.C.)
| | - Clara S. Martínez
- APC Microbiome Ireland, University College Cork, T12 YT20 Cork, Ireland; (T.F.S.B.); (E.G.); (M.B.); (C.S.); (C.S.M.); (J.F.C.)
| | - Patrizio Blandina
- Dipartimento di Neuroscienze, Psicologia, Area del Farmaco e Salute del Bambino (NEUROFARBA), Universitá di Firenze, Viale Pieraccini 6, 50139 Firenze, Italy; (F.B.); (G.P.); (P.B.)
| | - John F. Cryan
- APC Microbiome Ireland, University College Cork, T12 YT20 Cork, Ireland; (T.F.S.B.); (E.G.); (M.B.); (C.S.); (C.S.M.); (J.F.C.)
- Department of Anatomy and Neuroscience, University College Cork, T12 YT20 Cork, Ireland
| | - Sophie Layé
- Laboratoire NutriNeuro, UMR INRAE, Bordeaux INP, Université de Bordeaux, 146 Rue Léo Saignat, 33076 Bordeaux, France; (M.R.); (S.L.)
| | - Renato Corradetti
- Dipartimento di Neuroscienze, Psicologia, Area del Farmaco e Salute del Bambino (NEUROFARBA), Universitá di Firenze, Viale Pieraccini 6, 50139 Firenze, Italy; (F.B.); (G.P.); (P.B.)
| | - Maria Beatrice Passani
- Dipartimento di Scienze della Salute, Universitá di Firenze, Viale Pieraccini 6, 50139 Firenze, Italy; (A.C.); (B.R.)
| |
Collapse
|
37
|
Fighting fire with fire: the immune system might be key in our fight against Alzheimer's disease. Drug Discov Today 2022; 27:1261-1283. [PMID: 35032668 DOI: 10.1016/j.drudis.2022.01.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 11/25/2021] [Accepted: 01/06/2022] [Indexed: 12/13/2022]
Abstract
The ultimate cause of Alzheimer's disease (AD) is still unknown and no disease-modifying treatment exists. Emerging evidence supports the concept that the immune system has a key role in AD pathogenesis. This awareness leads to the idea that specific parts of the immune system must be engaged to ward off the disease. Immunotherapy has dramatically improved the management of several previously untreatable cancers and could hold similar promise as a novel therapy for treating AD. However, before potent immunotherapies can be rationally designed as treatment against AD, we need to fully understand the dynamic interplay between AD and the different parts of our immune system. Accordingly, here we review the most important aspects of both the innate and adaptive immune system in relation to AD pathology. Teaser: Emerging results support the concept that Alzheimer's disease is affected by the inability of the immune system to contain the pathology of the brain. Here, we discuss how we can engage our immune system to fight this devastating disease.
Collapse
|
38
|
Amin MM, Futrawan R, Husada MS. Correlation Between Schizophrenia and Coronavirus Disease in North Sumatera, Indonesia: A Correlative Analytical Study. Front Psychiatry 2022; 13:896623. [PMID: 35722585 PMCID: PMC9198631 DOI: 10.3389/fpsyt.2022.896623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 04/28/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND In the first quarter of 2020, two cases of coronavirus disease (COVID-19) were reported in Indonesia, approximately 4 months after the first case was reported in China. The numbers continued to increase following the introduction of many variants of the virus. The pandemic may have an impact on the community's mental health, particularly on those with mental illnesses. Therefore, this study aimed to determine the correlation between schizophrenia and COVID-19 based on demographic characteristics. METHODS This nominal-nominal and numerical-nominal correlative analytical study used a cross-sectional approach and was conducted at a psychiatric hospital in North Sumatra. The sample population consisted of 48 patients and 48 healthy controls, who were selected using a non-probability consecutive sampling method. RESULTS The analysis showed that there were correlations between schizophrenia and COVID-19 (r = 0.417, p < 0.001) and between the age of patients with schizophrenia and COVID-19 with (r = 0.544). CONCLUSIONS COVID-19 is correlated with schizophrenia and the age of patients with schizophrenia. We recommend that patients with schizophrenia follow the same health guidelines as the clinical high-risk group for COVID-19 and receive the same treatment. Physicians that treat patients with COVID-19 should pay close attention to those with schizophrenia because they may underestimate their condition.
Collapse
Affiliation(s)
- Mustafa M Amin
- Department of Psychiatry, Faculty of Medicine, Universitas Sumatera Utara, Medan, Indonesia
| | - Richie Futrawan
- Department of Psychiatry, Faculty of Medicine, Universitas Sumatera Utara, Medan, Indonesia
| | - Muhammad Surya Husada
- Department of Psychiatry, Faculty of Medicine, Universitas Sumatera Utara, Medan, Indonesia
| |
Collapse
|
39
|
Di Filippo M, Mancini A, Bellingacci L, Gaetani L, Mazzocchetti P, Zelante T, La Barbera L, De Luca A, Tantucci M, Tozzi A, Durante V, Sciaccaluga M, Megaro A, Chiasserini D, Salvadori N, Lisetti V, Portaccio E, Costa C, Sarchielli P, Amato MP, Parnetti L, Viscomi MT, Romani L, Calabresi P. Interleukin-17 affects synaptic plasticity and cognition in an experimental model of multiple sclerosis. Cell Rep 2021; 37:110094. [PMID: 34879272 DOI: 10.1016/j.celrep.2021.110094] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 08/06/2021] [Accepted: 11/12/2021] [Indexed: 12/20/2022] Open
Abstract
Cognitive impairment (CI) is a disabling concomitant of multiple sclerosis (MS) with a complex and controversial pathogenesis. The cytokine interleukin-17A (IL-17A) is involved in the immune pathogenesis of MS, but its possible effects on synaptic function and cognition are still largely unexplored. In this study, we show that the IL-17A receptor (IL-17RA) is highly expressed by hippocampal neurons in the CA1 area and that exposure to IL-17A dose-dependently disrupts hippocampal long-term potentiation (LTP) through the activation of its receptor and p38 mitogen-activated protein kinase (MAPK). During experimental autoimmune encephalomyelitis (EAE), IL-17A overexpression is paralleled by hippocampal LTP dysfunction. An in vivo behavioral analysis shows that visuo-spatial learning abilities are preserved when EAE is induced in mice lacking IL-17A. Overall, this study suggests a key role for the IL-17 axis in the neuro-immune cross-talk occurring in the hippocampal CA1 area and its potential involvement in synaptic dysfunction and MS-related CI.
Collapse
MESH Headings
- Animals
- Behavior, Animal
- CA1 Region, Hippocampal/metabolism
- CA1 Region, Hippocampal/pathology
- CA1 Region, Hippocampal/physiopathology
- Cognition
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Encephalomyelitis, Autoimmune, Experimental/physiopathology
- Encephalomyelitis, Autoimmune, Experimental/psychology
- Interleukin-17/genetics
- Interleukin-17/metabolism
- Long-Term Potentiation
- Male
- Mice, Biozzi
- Mice, Inbred C57BL
- Mice, Knockout
- Neuronal Plasticity
- Receptors, Interleukin-17/genetics
- Receptors, Interleukin-17/metabolism
- Signal Transduction
- Spatial Learning
- Synapses/metabolism
- Synapses/pathology
- p38 Mitogen-Activated Protein Kinases
- Mice
Collapse
Affiliation(s)
- Massimiliano Di Filippo
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy.
| | - Andrea Mancini
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Laura Bellingacci
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Lorenzo Gaetani
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Petra Mazzocchetti
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Teresa Zelante
- Section of Pathology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Livia La Barbera
- Unit of Molecular Neurosciences, Department of Medicine, University Campus-Biomedico, Rome, Italy
| | - Antonella De Luca
- Section of Pathology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Michela Tantucci
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Alessandro Tozzi
- Section of Physiology and Biochemistry, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Valentina Durante
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Miriam Sciaccaluga
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Alfredo Megaro
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Davide Chiasserini
- Section of Physiology and Biochemistry, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Nicola Salvadori
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Viviana Lisetti
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Emilio Portaccio
- Department of NEUROFARBA, University of Florence, Florence, Italy
| | - Cinzia Costa
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Paola Sarchielli
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Maria Pia Amato
- Department of NEUROFARBA, University of Florence, Florence, Italy; IRCCS Fondazione Don Carlo Gnocchi, Florence, Italy
| | - Lucilla Parnetti
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Maria Teresa Viscomi
- Section of Histology and Embryology, Department of Life Science and Public Health, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Luigina Romani
- Section of Pathology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Paolo Calabresi
- Neurology, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS, Rome, Italy; Section of Neurology, Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
40
|
Morcuende A, Navarrete F, Nieto E, Manzanares J, Femenía T. Inflammatory Biomarkers in Addictive Disorders. Biomolecules 2021; 11:biom11121824. [PMID: 34944470 PMCID: PMC8699452 DOI: 10.3390/biom11121824] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Revised: 11/23/2021] [Accepted: 11/30/2021] [Indexed: 12/21/2022] Open
Abstract
Substance use disorders are a group of diseases that are associated with social, professional, and family impairment and that represent a high socio-economic impact on the health systems of countries around the world. These disorders present a very complex diagnosis and treatment regimen due to the lack of suitable biomarkers supporting the correct diagnosis and classification and the difficulty of selecting effective therapies. Over the last few years, several studies have pointed out that these addictive disorders are associated with systemic and central nervous system inflammation, which could play a relevant role in the onset and progression of these diseases. Therefore, identifying different immune system components as biomarkers of such addictive disorders could be a crucial step to promote appropriate diagnosis and treatment. Thus, this work aims to provide an overview of the immune system alterations that may be biomarkers of various addictive disorders.
Collapse
Affiliation(s)
- Alvaro Morcuende
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, Avda. de Ramón y Cajal s/n, San Juan de Alicante, 03550 Alicante, Spain; (A.M.); (F.N.); (E.N.); (J.M.)
| | - Francisco Navarrete
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, Avda. de Ramón y Cajal s/n, San Juan de Alicante, 03550 Alicante, Spain; (A.M.); (F.N.); (E.N.); (J.M.)
- Red Temática de Investigación Cooperativa en Salud (RETICS), Red de Trastornos Adictivos, Instituto de Salud Carlos III, MICINN and FEDER, 28029 Madrid, Spain
| | - Elena Nieto
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, Avda. de Ramón y Cajal s/n, San Juan de Alicante, 03550 Alicante, Spain; (A.M.); (F.N.); (E.N.); (J.M.)
| | - Jorge Manzanares
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, Avda. de Ramón y Cajal s/n, San Juan de Alicante, 03550 Alicante, Spain; (A.M.); (F.N.); (E.N.); (J.M.)
- Red Temática de Investigación Cooperativa en Salud (RETICS), Red de Trastornos Adictivos, Instituto de Salud Carlos III, MICINN and FEDER, 28029 Madrid, Spain
| | - Teresa Femenía
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, Avda. de Ramón y Cajal s/n, San Juan de Alicante, 03550 Alicante, Spain; (A.M.); (F.N.); (E.N.); (J.M.)
- Red Temática de Investigación Cooperativa en Salud (RETICS), Red de Trastornos Adictivos, Instituto de Salud Carlos III, MICINN and FEDER, 28029 Madrid, Spain
- Correspondence: ; Tel.: +34-965-919-553
| |
Collapse
|
41
|
Urdinguio RG, Tejedor JR, Fernández-Sanjurjo M, Pérez RF, Peñarroya A, Ferrero C, Codina-Martínez H, Díez-Planelles C, Pinto-Hernández P, Castilla-Silgado J, Coto-Vilcapoma A, Díez-Robles S, Blanco-Agudín N, Tomás-Zapico C, Iglesias-Gutiérrez E, Fernández-García B, Fernandez AF, Fraga MF. Physical exercise shapes the mouse brain epigenome. Mol Metab 2021; 54:101398. [PMID: 34801767 PMCID: PMC8661702 DOI: 10.1016/j.molmet.2021.101398] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 11/09/2021] [Accepted: 11/14/2021] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVE To analyze the genome-wide epigenomic and transcriptomic changes induced by long term resistance or endurance training in the hippocampus of wild-type mice. METHODS We performed whole-genome bisulfite sequencing (WGBS) and RNA sequencing (RNA-seq) of mice hippocampus after 4 weeks of specific training. In addition, we used a novel object recognition test before and after the intervention to determine whether the exercise led to an improvement in cognitive function. RESULTS Although the majority of DNA methylation changes identified in this study were training-model specific, most were associated with hypomethylation and were enriched in similar histone marks, chromatin states, and transcription factor biding sites. It is worth highlighting the significant association found between the loss of DNA methylation in Tet1 binding sites and gene expression changes, indicating the importance of these epigenomic changes in transcriptional regulation. However, endurance and resistance training activate different gene pathways, those being associated with neuroplasticity in the case of endurance exercise, and interferon response pathways in the case of resistance exercise, which also appears to be associated with improved learning and memory functions. CONCLUSIONS Our results help both understand the molecular mechanisms by which different exercise models exert beneficial effects for brain health and provide new potential therapeutic targets for future research.
Collapse
Affiliation(s)
- Rocío G Urdinguio
- Nanomaterials and Nanotechnology Research Center (CINN-CSIC), Health Research Institute of Asturias (ISPA), Institute of Oncology of Asturias (IUOPA), Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 33011 Oviedo, Asturias, Spain
| | - Juan Ramon Tejedor
- Nanomaterials and Nanotechnology Research Center (CINN-CSIC), Health Research Institute of Asturias (ISPA), Institute of Oncology of Asturias (IUOPA), Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 33011 Oviedo, Asturias, Spain
| | - Manuel Fernández-Sanjurjo
- Departamento de Biología Funcional, Fisiología, Universidad de Oviedo, Oviedo 33006, Spain; Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo 33011, Spain
| | - Raúl F Pérez
- Nanomaterials and Nanotechnology Research Center (CINN-CSIC), Health Research Institute of Asturias (ISPA), Institute of Oncology of Asturias (IUOPA), Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 33011 Oviedo, Asturias, Spain
| | - Alfonso Peñarroya
- Nanomaterials and Nanotechnology Research Center (CINN-CSIC), Health Research Institute of Asturias (ISPA), Institute of Oncology of Asturias (IUOPA), Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 33011 Oviedo, Asturias, Spain
| | - Cecilia Ferrero
- Nanomaterials and Nanotechnology Research Center (CINN-CSIC), Health Research Institute of Asturias (ISPA), Institute of Oncology of Asturias (IUOPA), Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 33011 Oviedo, Asturias, Spain
| | - Helena Codina-Martínez
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo 33011, Spain; Departamento de Morfología y Biología Celular, Universidad de Oviedo, Oviedo 33006, Spain
| | - Carlos Díez-Planelles
- Departamento de Biología Funcional, Fisiología, Universidad de Oviedo, Oviedo 33006, Spain
| | - Paola Pinto-Hernández
- Departamento de Biología Funcional, Fisiología, Universidad de Oviedo, Oviedo 33006, Spain; Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo 33011, Spain
| | - Juan Castilla-Silgado
- Departamento de Biología Funcional, Fisiología, Universidad de Oviedo, Oviedo 33006, Spain; Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo 33011, Spain
| | - Almudena Coto-Vilcapoma
- Departamento de Biología Funcional, Fisiología, Universidad de Oviedo, Oviedo 33006, Spain; Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo 33011, Spain
| | - Sergio Díez-Robles
- Departamento de Biología Funcional, Fisiología, Universidad de Oviedo, Oviedo 33006, Spain
| | - Noelia Blanco-Agudín
- Departamento de Biología Funcional, Fisiología, Universidad de Oviedo, Oviedo 33006, Spain
| | - Cristina Tomás-Zapico
- Departamento de Biología Funcional, Fisiología, Universidad de Oviedo, Oviedo 33006, Spain; Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo 33011, Spain
| | - Eduardo Iglesias-Gutiérrez
- Departamento de Biología Funcional, Fisiología, Universidad de Oviedo, Oviedo 33006, Spain; Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo 33011, Spain.
| | - Benjamín Fernández-García
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo 33011, Spain; Departamento de Morfología y Biología Celular, Universidad de Oviedo, Oviedo 33006, Spain
| | - Agustin F Fernandez
- Nanomaterials and Nanotechnology Research Center (CINN-CSIC), Health Research Institute of Asturias (ISPA), Institute of Oncology of Asturias (IUOPA), Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 33011 Oviedo, Asturias, Spain.
| | - Mario F Fraga
- Nanomaterials and Nanotechnology Research Center (CINN-CSIC), Health Research Institute of Asturias (ISPA), Institute of Oncology of Asturias (IUOPA), Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 33011 Oviedo, Asturias, Spain; Department of Organisms and Systems Biology (B.O.S), University of Oviedo, 33011 Oviedo, Asturias, Spain.
| |
Collapse
|
42
|
Wang Q, Yao H, Liu W, Ya B, Cheng H, Xing Z, Wu Y. Microglia Polarization in Alzheimer's Disease: Mechanisms and a Potential Therapeutic Target. Front Aging Neurosci 2021; 13:772717. [PMID: 34819850 PMCID: PMC8606412 DOI: 10.3389/fnagi.2021.772717] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 10/07/2021] [Indexed: 11/13/2022] Open
Abstract
Neuroinflammation regulated by microglia is one of the important factors involved in the pathogenesis of Alzheimer’s disease (AD). Activated microglia exhibited phenotypes termed as M1 and M2 phenotypes separately. M1 microglia contribute to the development of inflammation via upregulating pro-inflammatory cytokines, while M2 microglia exert anti-inflammation effects through enhancing the expression of anti-inflammation factors. Moreover, M1 and M2 microglia could be mutually transformed under various conditions. Both M1 and M2 microglia are implicated in AD. Amyloid-β (Aβ) and hyperphosphorylated tau are two major components of AD pathological hallmarks, neuritic plaques, and neurofibrillary tangles. Both Aβ and hyperphosphorylated tau were involved in microglial activation and subsequent inflammation, which further contribute to neuronal and synaptic loss in AD. In this review, we summarized the roles of M1 and M2 microglia in AD and underlying mechanisms, which will provide an insight into the role of microglia in the pathogenesis of AD and highlight the therapeutic potential of modulating microglia.
Collapse
Affiliation(s)
- Qinqin Wang
- Shandong Collaborative Innovation Center for Diagnosis, Treatment and Behavioral Interventions of Mental Disorders, Institute of Mental Health, Jining Medical University, Jining, China
| | - Hongmei Yao
- Affiliated Hospital of Jining Medical University, Jining, China
| | - Wenyan Liu
- Department of Physiology, College of Basic Medicine, Jining Medical University, Jining, China
| | - Bailiu Ya
- Department of Physiology, College of Basic Medicine, Jining Medical University, Jining, China
| | - Hongju Cheng
- Department of Physiology, College of Basic Medicine, Jining Medical University, Jining, China
| | - Zhenkai Xing
- Shandong Collaborative Innovation Center for Diagnosis, Treatment and Behavioral Interventions of Mental Disorders, Institute of Mental Health, Jining Medical University, Jining, China
| | - Yili Wu
- The Affiliated Kangning Hospital, Wenzhou Medical University, Wenzhou, China.,Key Laboratory of Alzheimer's Disease of Zhejiang Province, Institute of Aging, School of Mental Health, Wenzhou Medical University, Wenzhou, China.,Oujiang Laboratory, Wenzhou, China
| |
Collapse
|
43
|
Pan JX, Sun D, Lee D, Xiong L, Ren X, Guo HH, Yao LL, Lu Y, Jung C, Xiong WC. Osteoblastic Swedish mutant APP expedites brain deficits by inducing endoplasmic reticulum stress-driven senescence. Commun Biol 2021; 4:1326. [PMID: 34824365 PMCID: PMC8617160 DOI: 10.1038/s42003-021-02843-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 11/03/2021] [Indexed: 11/09/2022] Open
Abstract
Patients with Alzheimer’s disease (AD) often have osteoporosis or osteopenia. However, their direct link and relationship remain largely unclear. Previous studies have detected osteoporotic deficits in young adult Tg2576 and TgAPPsweOCN mice, which express APPswe (Swedish mutant) ubiquitously and selectively in osteoblast (OB)-lineage cells. This raises the question, whether osteoblastic APPswe contributes to AD development. Here, we provide evidence that TgAPPsweOCN mice also exhibit AD-relevant brain pathologies and behavior phenotypes. Some brain pathologies include age-dependent and regional-selective increases in glial activation and pro-inflammatory cytokines, which are accompanied by behavioral phenotypes such as anxiety, depression, and altered learning and memory. Further cellular studies suggest that APPswe, but not APPwt or APPlon (London mutant), in OB-lineage cells induces endoplasmic reticulum-stress driven senescence, driving systemic and cortex inflammation as well as behavioral changes in 6-month-old TgAPPsweOCN mice. These results therefore reveal an unrecognized function of osteoblastic APPswe to brain axis in AD development. Jin-Xiu Pan et al. report that an osteoblast-specific expression of Swedish mutant amyloid precursor protein (APPswe) induces ER stress-driven senescence, leading to systemic inflammation and inflammation in the cortex that drives behavioral changes. The results demonstrate a previously unrecognized function of osteoblastic APPswe to brain axis in AD development.
Collapse
Affiliation(s)
- Jin-Xiu Pan
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH, USA.,Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH, USA
| | - Dong Sun
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Daehoon Lee
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH, USA.,Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH, USA
| | - Lei Xiong
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH, USA.,Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH, USA
| | - Xiao Ren
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Hao-Han Guo
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Ling-Ling Yao
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Yuyi Lu
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Caroline Jung
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Wen-Cheng Xiong
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH, USA. .,Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH, USA.
| |
Collapse
|
44
|
Woodburn SC, Bollinger JL, Wohleb ES. The semantics of microglia activation: neuroinflammation, homeostasis, and stress. J Neuroinflammation 2021; 18:258. [PMID: 34742308 PMCID: PMC8571840 DOI: 10.1186/s12974-021-02309-6] [Citation(s) in RCA: 366] [Impact Index Per Article: 91.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 10/28/2021] [Indexed: 02/08/2023] Open
Abstract
Microglia are emerging as critical regulators of neuronal function and behavior in nearly every area of neuroscience. Initial reports focused on classical immune functions of microglia in pathological contexts, however, immunological concepts from these studies have been applied to describe neuro-immune interactions in the absence of disease, injury, or infection. Indeed, terms such as 'microglia activation' or 'neuroinflammation' are used ubiquitously to describe changes in neuro-immune function in disparate contexts; particularly in stress research, where these terms prompt undue comparisons to pathological conditions. This creates a barrier for investigators new to neuro-immunology and ultimately hinders our understanding of stress effects on microglia. As more studies seek to understand the role of microglia in neurobiology and behavior, it is increasingly important to develop standard methods to study and define microglial phenotype and function. In this review, we summarize primary research on the role of microglia in pathological and physiological contexts. Further, we propose a framework to better describe changes in microglia1 phenotype and function in chronic stress. This approach will enable more precise characterization of microglia in different contexts, which should facilitate development of microglia-directed therapeutics in psychiatric and neurological disease.
Collapse
Affiliation(s)
- Samuel C Woodburn
- Department of Pharmacology & Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Justin L Bollinger
- Department of Pharmacology & Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Eric S Wohleb
- Department of Pharmacology & Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| |
Collapse
|
45
|
Posillico CK. Three's Company: Neuroimmune activation, sex, and memory at the tripartite synapse. Brain Behav Immun Health 2021; 16:100326. [PMID: 34589812 PMCID: PMC8474433 DOI: 10.1016/j.bbih.2021.100326] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 08/07/2021] [Accepted: 08/09/2021] [Indexed: 12/30/2022] Open
Abstract
The neuroimmune system is required for normal cognitive functions such as learning and memory in addition to its critical role in detecting and responding to invading pathogens and injury. Understanding the functional convergence of neurons, astrocytes, and microglia at the synapse, particularly in the hippocampus, is key to understanding the nuances of such diverse roles. In the healthy brain, communication between all three cells is important for regulating neuronal activation and synaptic plasticity mechanisms, and during neuroinflammation, the activity and functions of all three cells can produce and be modulated by inflammatory cytokines. An important remaining component to this system is the conclusive evidence of sex differences in hippocampal plasticity mechanisms, hormone modulation of synaptic plasticity, functional properties of hippocampal neurons, and in neuroimmune activation. Sex as a biological variable here is necessary to consider given sex differences in the prevalence of memory-related disorders such as Alzheimer's disease and Post-Traumatic Stress disorder, both of which present with neuroimmune dysregulation. To make meaningful progress towards a deeper understanding of sex biases in memory-related disease prevalence, I propose that the next chapter of psychoneuroimmune research must focus on the signal integration and transduction at the synapse between experience-dependent plasticity mechanisms, neuroimmune activation, and the influence of biological sex.
Collapse
|
46
|
Robinson S, Mogul AS, Taylor-Yeremeeva EM, Khan A, Tirabassi AD, Wang HY. Stress Diminishes BDNF-stimulated TrkB Signaling, TrkB-NMDA Receptor Linkage and Neuronal Activity in the Rat Brain. Neuroscience 2021; 473:142-158. [PMID: 34298123 PMCID: PMC8455453 DOI: 10.1016/j.neuroscience.2021.07.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 07/08/2021] [Accepted: 07/13/2021] [Indexed: 11/21/2022]
Abstract
Exposure to intense or repeated stressors can lead to depression or post-traumatic stress disorder (PTSD). Neurological changes induced by stress include impaired neurotrophin signaling, which is known to influence synaptic integrity and plasticity. The present study used an ex vivo approach to examine the impact of acute or repeated stress on BDNF-stimulated TrkB signaling in hippocampus (HIPPO) and prefrontal cortex (PFC). Rats in an acute multiple stressor group experienced five stressors in one day whereas rats in a repeated unpredictable stressor group experienced 20 stressors across 10 days. After stress exposure, slices were incubated with vehicle or BDNF, followed by immunoprecipitation and immunoblot assays to assess protein levels, activation states and protein-protein linkage associated with BDNF-TrkB signaling. Three key findings are (1) exposure to stressors significantly diminished BDNF-stimulated TrkB signaling in HIPPO and PFC such that reductions in TrkB activation, diminished recruitment of adaptor proteins to TrkB, reduced activation of downstream signaling molecules, disruption of TrkB-NMDAr linkage, and changes in basal and BDNF-stimulated Arc expression were observed. (2) After stress, BDNF stimulation enhanced TrkB-NMDAr linkage in PFC, suggestive of compensatory mechanisms in this region. (3) We discovered an uncoupling between TrkB signaling, TrkB-NMDAr linkage and Arc expression in PFC and HIPPO. In addition, a robust surge in pro-inflammatory cytokines was observed in both regions after repeated exposure to stressors. Collectively, these data provide therapeutic targets for future studies that investigate how to reverse stress-induced downregulation of BDNF-TrkB signaling and underscore the need for functional studies that examine stress-related TrkB-NMDAr activities in PFC.
Collapse
Affiliation(s)
- Siobhan Robinson
- Department of Psychology and Program in Neuroscience, Hamilton College, Clinton, NY, USA.
| | - Allison S Mogul
- Department of Psychology and Program in Neuroscience, Hamilton College, Clinton, NY, USA
| | | | - Amber Khan
- Department of Molecular, Cellular & Biomedical Sciences, The City University of New York School of Medicine, New York, NY, USA; Department of Biology, Neuroscience Program, Graduate School of the City University of New York, New York, NY, USA
| | - Anthony D Tirabassi
- Department of Psychology and Program in Neuroscience, Hamilton College, Clinton, NY, USA
| | - Hoau-Yan Wang
- Department of Molecular, Cellular & Biomedical Sciences, The City University of New York School of Medicine, New York, NY, USA; Department of Biology, Neuroscience Program, Graduate School of the City University of New York, New York, NY, USA
| |
Collapse
|
47
|
Bellingacci L, Mancini A, Gaetani L, Tozzi A, Parnetti L, Di Filippo M. Synaptic Dysfunction in Multiple Sclerosis: A Red Thread from Inflammation to Network Disconnection. Int J Mol Sci 2021; 22:ijms22189753. [PMID: 34575917 PMCID: PMC8469646 DOI: 10.3390/ijms22189753] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 08/31/2021] [Accepted: 09/03/2021] [Indexed: 12/24/2022] Open
Abstract
Multiple sclerosis (MS) has been clinically considered a chronic inflammatory disease of the white matter; however, in the last decade growing evidence supported an important role of gray matter pathology as a major contributor of MS-related disability and the involvement of synaptic structures assumed a key role in the pathophysiology of the disease. Synaptic contacts are considered central units in the information flow, involved in synaptic transmission and plasticity, critical processes for the shaping and functioning of brain networks. During the course of MS, the immune system and its diffusible mediators interact with synaptic structures leading to changes in their structure and function, influencing brain network dynamics. The purpose of this review is to provide an overview of the existing literature on synaptic involvement during experimental and human MS, in order to understand the mechanisms by which synaptic failure eventually leads to brain networks alterations and contributes to disabling MS symptoms and disease progression.
Collapse
Affiliation(s)
- Laura Bellingacci
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, 06132 Perugia, Italy; (L.B.); (A.M.); (L.G.); (L.P.)
| | - Andrea Mancini
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, 06132 Perugia, Italy; (L.B.); (A.M.); (L.G.); (L.P.)
| | - Lorenzo Gaetani
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, 06132 Perugia, Italy; (L.B.); (A.M.); (L.G.); (L.P.)
| | - Alessandro Tozzi
- Section of Physiology and Biochemistry, Department of Medicine and Surgery, University of Perugia, 06132 Perugia, Italy;
| | - Lucilla Parnetti
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, 06132 Perugia, Italy; (L.B.); (A.M.); (L.G.); (L.P.)
| | - Massimiliano Di Filippo
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, 06132 Perugia, Italy; (L.B.); (A.M.); (L.G.); (L.P.)
- Correspondence: ; Tel.: +39-075-578-3830
| |
Collapse
|
48
|
Abstract
Interleukin-1 (IL-1) is an inflammatory cytokine that has been shown to modulate neuronal signaling in homeostasis and diseases. In homeostasis, IL-1 regulates sleep and memory formation, whereas in diseases, IL-1 impairs memory and alters affect. Interestingly, IL-1 can cause long-lasting changes in behavior, suggesting IL-1 can alter neuroplasticity. The neuroplastic effects of IL-1 are mediated via its cognate receptor, Interleukin-1 Type 1 Receptor (IL-1R1), and are dependent on the distribution and cell type(s) of IL-1R1 expression. Recent reports found that IL-1R1 expression is restricted to discrete subpopulations of neurons, astrocytes, and endothelial cells and suggest IL-1 can influence neural circuits directly through neuronal IL-1R1 or indirectly via non-neuronal IL-1R1. In this review, we analyzed multiple mechanisms by which IL-1/IL-1R1 signaling might impact neuroplasticity based upon the most up-to-date literature and provided potential explanations to clarify discrepant and confusing findings reported in the past.
Collapse
Affiliation(s)
- Daniel P. Nemeth
- Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, OH, USA
- Department of Biomedical Science, Charles E. Schmidt College of Medicine and Brain Institute, Florida Atlantic University, Jupiter, FL, USA
| | - Ning Quan
- Department of Biomedical Science, Charles E. Schmidt College of Medicine and Brain Institute, Florida Atlantic University, Jupiter, FL, USA
| |
Collapse
|
49
|
Muscat SM, Barrientos RM. The Perfect Cytokine Storm: How Peripheral Immune Challenges Impact Brain Plasticity & Memory Function in Aging. Brain Plast 2021; 7:47-60. [PMID: 34631420 PMCID: PMC8461734 DOI: 10.3233/bpl-210127] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Precipitous declines in cognitive function can occur in older individuals following a variety of peripheral immune insults, such as surgery, infection, injury, and unhealthy diet. Aging is associated with numerous changes to the immune system that shed some light on why this abrupt cognitive deterioration may occur. Normally, peripheral-to-brain immune signaling is tightly regulated and advantageous; communication between the two systems is bi-directional, via either humoral or neural routes. Following an immune challenge, production, secretion, and translocation of cytokines into the brain is critical to the development of adaptive sickness behaviors. However, aging is normally associated with neuroinflammatory priming, notably microglial sensitization. Microglia are the brain's innate immune cells and become sensitized with advanced age, such that upon immune stimulation they will mount more exaggerated neuroimmune responses. The resultant elevation of pro-inflammatory cytokine expression, namely IL-1β, has profound effects on synaptic plasticity and, consequentially, cognition. In this review, we (1) investigate the processes which lead to aberrantly elevated inflammatory cytokine expression in the aged brain and (2) examine the impact of the pro-inflammatory cytokine IL-1β on brain plasticity mechanisms, including its effects on BDNF, AMPA and NMDA receptor-mediated long-term potentiation.
Collapse
Affiliation(s)
- Stephanie M Muscat
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA
- Biomedical Sciences Graduate Program, The Ohio State University, Columbus, OH, USA
| | - Ruth M Barrientos
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA
- Department of Psychiatry and Behavioral Health, The Ohio State University, Columbus, OH, USA
- Department of Neuroscience, The Ohio State University, Columbus, OH, USA
- Chronic Brain Injury Program, Discovery Themes Initiative, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
50
|
Gupta A, Singh AK, Kumar R, Jamieson S, Pandey AK, Bishayee A. Neuroprotective Potential of Ellagic Acid: A Critical Review. Adv Nutr 2021; 12:1211-1238. [PMID: 33693510 PMCID: PMC8321875 DOI: 10.1093/advances/nmab007] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 09/02/2020] [Accepted: 01/19/2021] [Indexed: 02/06/2023] Open
Abstract
Ellagic acid (EA) is a dietary polyphenol present in various fruits, vegetables, herbs, and nuts. It exists either independently or as part of complex structures, such as ellagitannins, which release EA and several other metabolites including urolithins following absorption. During the past few decades, EA has drawn considerable attention because of its vast range of biological activities as well as its numerous molecular targets. Several studies have reported that the oxidative stress-lowering potential of EA accounts for its broad-spectrum pharmacological attributes. At the biochemical level, several mechanisms have also been associated with its therapeutic action, including its efficacy in normalizing lipid metabolism and lipidemic profile, regulating proinflammatory mediators, such as IL-6, IL-1β, and TNF-α, upregulating nuclear factor erythroid 2-related factor 2 and inhibiting NF-κB action. EA exerts appreciable neuroprotective activity by its free radical-scavenging action, iron chelation, initiation of several cell signaling pathways, and alleviation of mitochondrial dysfunction. Numerous in vivo studies have also explored the neuroprotective attribute of EA against various neurotoxins in animal models. Despite the increasing number of publications with experimental evidence, a critical analysis of available literature to understand the full neuroprotective potential of EA has not been performed. The present review provides up-to-date, comprehensive, and critical information regarding the natural sources of EA, its bioavailability, metabolism, neuroprotective activities, and underlying mechanisms of action in order to encourage further studies to define the clinical usefulness of EA for the management of neurological disorders.
Collapse
Affiliation(s)
- Ashutosh Gupta
- Department of Biochemistry, University of Allahabad, Prayagraj, Uttar Pradesh, India
| | - Amit Kumar Singh
- Department of Biochemistry, University of Allahabad, Prayagraj, Uttar Pradesh, India
| | - Ramesh Kumar
- Department of Biochemistry, University of Allahabad, Prayagraj, Uttar Pradesh, India
| | - Sarah Jamieson
- Lake Erie College of Osteopathic Medicine, Bradenton, FL, USA
| | - Abhay Kumar Pandey
- Department of Biochemistry, University of Allahabad, Prayagraj, Uttar Pradesh, India
| | - Anupam Bishayee
- Lake Erie College of Osteopathic Medicine, Bradenton, FL, USA
| |
Collapse
|