1
|
Pauletti A, Gurlo P, Weiß E, DePaula-Silva AB, Wilcox KS, Bröer S. Viral encephalitis and seizures cause rapid depletion of neuronal progenitor cells and alter neurogenesis in the adult mouse dentate gyrus. Front Cell Neurosci 2025; 18:1528918. [PMID: 39876841 PMCID: PMC11772278 DOI: 10.3389/fncel.2024.1528918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Accepted: 12/23/2024] [Indexed: 01/31/2025] Open
Abstract
Infections impacting the central nervous system (CNS) constitute a substantial predisposing factor for the emergence of epileptic seizures. Given that epilepsy conventionally correlates with hippocampal sclerosis and neuronal degeneration, a potentially innovative avenue for therapeutic intervention involves fostering adult neurogenesis, a process primarily occurring within the subgranular zone of the dentate gyrus (DG) through the differentiation of neural stem cells (NSC). While experimental seizures induced by chemoconvulsants or electrical stimulation transiently enhance neurogenesis, the effects of encephalitis and the resultant virus-induced seizures remain inadequately understood. Thus, this study employed the Theiler's Murine Encephalomyelitis Virus (TMEV) model of virus-induced seizures in adult C57BL/6J mice to investigate the impact of infection-induced seizures on neurogenesis at three distinct time points [3, 7, and 14 days post-infection (dpi)]. Immunohistochemical analysis revealed a reduction in the overall number of proliferating cells post-infection. More notably, the specific cell types exhibiting proliferation diverged between TMEV and control (CTR) mice: (1) Neuronal progenitors (doublecortin, DCX+) were almost entirely absent at 3 dpi in the dorsal DG. They resumed proliferation at 14 dpi, but, did not recover to CTR levels, and displayed aberrant migration patterns. (2) The number of proliferating NSCs significantly decreased within the dorsal DG of TMEV mice at 14 dpi compared to CTR, while (3) a heightened population of proliferating astrocytes was observed. Most observed changes were not different between seizing and non-seizing infected mice. In summary, our findings demonstrate that viral infection rapidly depletes neuronal progenitor cells and causes aberrant migration of the remaining ones, potentially contributing to hyperexcitability. Additionally, the increased differentiation toward glial cell fates in infected mice emerges as a possible additional pro-epileptogenic mechanism.
Collapse
Affiliation(s)
- Alberto Pauletti
- School of Veterinary Medicine, Institute of Pharmacology and Toxicology, Freie Universität Berlin, Berlin, Germany
| | - Polina Gurlo
- School of Veterinary Medicine, Institute of Pharmacology and Toxicology, Freie Universität Berlin, Berlin, Germany
| | - Edna Weiß
- School of Veterinary Medicine, Institute of Pharmacology and Toxicology, Freie Universität Berlin, Berlin, Germany
| | | | - Karen S. Wilcox
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, UT, United States
| | - Sonja Bröer
- School of Veterinary Medicine, Institute of Pharmacology and Toxicology, Freie Universität Berlin, Berlin, Germany
| |
Collapse
|
2
|
Savoca G, Gianfredi A, Bartolini L. The Development of Epilepsy Following CNS Viral Infections: Mechanisms. Curr Neurol Neurosci Rep 2024; 25:2. [PMID: 39549124 DOI: 10.1007/s11910-024-01393-4] [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] [Accepted: 10/08/2024] [Indexed: 11/18/2024]
Abstract
PURPOSE OF REVIEW This review examines the role of different viral infections in epileptogenesis, with a focus on Herpesviruses such as Human Herpesvirus 6 (HHV-6) and Epstein Barr Virus (EBV), Flaviviruses, Picornaviruses, Human Immunodeficiency Virus (HIV), Influenzavirus and Severe Acute Respiratory Syndrome CoronaVirus 2 (SARS-CoV-2). RECENT FINDINGS A growing literature on animal models, such as the paradigmatic Theiler's murine encephalomyelitis virus (TMEV) model, and clinical investigations in patients with epilepsy have started to elucidate cellular mechanisms implicated in seizure initiation and development of epilepsy following viral infections. A central role of neuroinflammation has emerged, with evidence of activation of the innate and adaptive immunity, dysregulation of microglial and astrocytic activity and production of multiple cytokines and other inflammatory mediators. Several chronic downstream effects result in increased blood-brain barrier permeability, direct neuronal damage, and modifications of ion channels ultimately leading to altered neuronal excitability and seizure generation. Key findings underscore the complex interplay between initial viral infection, neuroinflammation, and later development of epilepsy. Further research is needed to elucidate these mechanisms and develop targeted interventions.
Collapse
Affiliation(s)
- Giulia Savoca
- Neuroscience Department, Meyer Children's Hospital IRCCS, Viale Pieraccini 24, 50139, Florence, Italy
- University of Florence School of Medicine, Florence, Italy
| | - Arianna Gianfredi
- Neuroscience Department, Meyer Children's Hospital IRCCS, Viale Pieraccini 24, 50139, Florence, Italy
- University of Florence School of Medicine, Florence, Italy
| | - Luca Bartolini
- Neuroscience Department, Meyer Children's Hospital IRCCS, Viale Pieraccini 24, 50139, Florence, Italy.
- Department of Neuroscience, Psychology, Pharmacology and Child Health (NEUROFARBA), University of Florence, Florence, Italy.
| |
Collapse
|
3
|
Zierath DK, Davidson S, Manoukian J, Knox KM, White HS, Meeker S, Ericsson A, Barker-Haliski M. Diet composition and sterilization modifies intestinal microbiome diversity and burden of Theiler's virus infection-induced acute seizures. Epilepsia 2024; 65:1777-1790. [PMID: 38491947 DOI: 10.1111/epi.17946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 02/26/2024] [Accepted: 02/27/2024] [Indexed: 03/18/2024]
Abstract
OBJECTIVE Brain infection with Theiler's murine encephalomyelitis virus (TMEV) in C57BL/6J mice can induce acquired epileptogenesis. Diet alters acute seizure incidence in TMEV-infected mice; yet it is unclear whether intestinal dysbiosis may also impact acute or chronic behavioral comorbidities. This study thus assessed the impact of diet formulation and sterilization on acute seizure presentation, gut microbiome composition, and epilepsy-related chronic behavioral comorbidities. METHODS Baseline fecal samples were collected from male C57BL/6J mice (4- to 5-weeks-old; Jackson Labs) upon facility arrival. Mice were randomized to either autoclaved (AC) or irradiated diet (IR) (Prolab RMH 3000) or IR (Picolab 5053). Three days later, mice underwent intracerebral TMEV or phosphate-buffered saline (PBS) injection. Fecal samples were collected from a subset of mice at infection (Day 0) and Day 7 post-infection. Epilepsy-related working memory deficits and seizure threshold were assessed 6 weeks post-infection. Gut microbiome diversity was determined by 16S rRNA amplicon sequencing of fecal samples. RESULTS TMEV-infected mice displayed acute handling-induced seizures, regardless of diet: 28 of 57 IR Picolab 5053 (49.1%), 30 of 41 IR Prolab RMH 3000 (73.2%), and 47 of 77 AC Prolab RMH 3000 (61%) mice displayed seizures. The number of observed seizures differed significantly by diet: IR Picolab 5053 diet-fed mice had 2.2 ± 2.8 seizures (mean ± standard deviation), IR Prolab RMH 3000 diet-fed mice had 3.5 ± 2.9 seizures, and AC Prolab RMH 3000 diet-fed mice had 4.4 ± 3.8 seizures during the 7-day monitoring period. Gut microbiome composition differed significantly in TMEV-infected mice fed the AC Prolab RMH 3000 diet, with measured differences in gram-positive bacteria. These mice also displayed worsened long-term working memory deficits. SIGNIFICANCE Diet-induced differences in intestinal dysbiosis in the TMEV model are associated with marked changes in acute seizure presentation, symptomatic recovery, and onset of chronic behavioral comorbidities of epilepsy. Our study reveals a novel disease-modifying impact of dietary manipulation on intestinal bacterial species after TMEV-induced acute seizures.
Collapse
Affiliation(s)
- Dannielle K Zierath
- Department of Pharmacy, School of Pharmacy, University of Washington, Seattle, Washington, USA
| | - Stephanie Davidson
- Department of Pharmacy, School of Pharmacy, University of Washington, Seattle, Washington, USA
| | - Jonathan Manoukian
- Department of Pharmacy, School of Pharmacy, University of Washington, Seattle, Washington, USA
| | - Kevin M Knox
- Department of Pharmacy, School of Pharmacy, University of Washington, Seattle, Washington, USA
| | - H Steve White
- Department of Pharmacy, School of Pharmacy, University of Washington, Seattle, Washington, USA
| | - Stacey Meeker
- Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Aaron Ericsson
- Department of Veterinary Pathobiology, University of Missouri, Columbia, Missouri, USA
| | - Melissa Barker-Haliski
- Department of Pharmacy, School of Pharmacy, University of Washington, Seattle, Washington, USA
| |
Collapse
|
4
|
Bröer S, Pauletti A. Microglia and infiltrating macrophages in ictogenesis and epileptogenesis. Front Mol Neurosci 2024; 17:1404022. [PMID: 38873242 PMCID: PMC11171130 DOI: 10.3389/fnmol.2024.1404022] [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: 03/20/2024] [Accepted: 05/15/2024] [Indexed: 06/15/2024] Open
Abstract
Phagocytes maintain homeostasis in a healthy brain. Upon injury, they are essential for repairing damaged tissue, recruiting other immune cells, and releasing cytokines as the first line of defense. However, there seems to be a delicate balance between the beneficial and detrimental effects of their activation in a seizing brain. Blocking the infiltration of peripheral phagocytes (macrophages) or their depletion can partially alleviate epileptic seizures and prevent the death of neurons in experimental models of epilepsy. However, the depletion of resident phagocytes in the brain (microglia) can aggravate disease outcomes. This review describes the role of resident microglia and peripheral infiltrating monocytes in animal models of acutely triggered seizures and epilepsy. Understanding the roles of phagocytes in ictogenesis and the time course of their activation and involvement in epileptogenesis and disease progression can offer us new biomarkers to identify patients at risk of developing epilepsy after a brain insult, as well as provide novel therapeutic targets for treating epilepsy.
Collapse
Affiliation(s)
- Sonja Bröer
- Institute of Pharmacology and Toxicology, School of Veterinary Medicine, Freie Universität Berlin, Berlin, Germany
| | | |
Collapse
|
5
|
Barker-Haliski M, DePaula-Silva AB, Pitsch J, Sontheimer H, Hirsch LJ, Galanopoulou AS, Kearney JA. Brain on Fire: How Brain Infection and Neuroinflammation Drive Worldwide Epilepsy Burden. Epilepsy Curr 2024:15357597241242238. [PMID: 39554268 PMCID: PMC11562294 DOI: 10.1177/15357597241242238] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2024] Open
Abstract
Roughly 80% of the global burden of epilepsy resides in low- and middle-income countries (LMICs; WHO, 2022). Despite numerous new therapies for the treatment of epilepsy, the number of patients who remain resistant to available medications is unchanged. Additionally, no therapy has yet been clinically proven to prevent or attenuate the development of epilepsy in at-risk individuals. Unfortunately, access to next generation therapies in LMICs is low, the stigma associated with epilepsy remains high, and access to adequate resources is unchanged. Thus, the global epilepsy burden disproportionately falls on LMICs such that strategies to conscientiously integrate global epilepsy risk factors into preclinical research may meaningfully advance 21st century epilepsy therapies. Brain infections are one of the main risk factors for epilepsy in resource-poor settings. Further, both infection- and autoimmune-associated encephalitis contribute to worldwide epilepsy risk and remain relatively understudied. For example, clinical SARS CoV-2 infection can induce rare instances of encephalopathy and acute seizures. Among viruses known to cause acute brain infection, enteroviruses increase risk for encephalitis-induced epilepsy, but are not associated with risk for other neurodevelopmental disorders (eg, autism spectrum or attentional deficit hyperactivity disorders). Naturally occurring models of viral infection-induced epilepsy therefore provide an exquisite opportunity to uncover novel contributors to epileptogenesis. Moreover, the convergent neuroinflammatory pathways that are associated with viral infection-induced encephalitis and autoimmune encephalitis reflect an untapped therapeutic opportunity to meaningfully reduce the global burden of epilepsy. This review summarizes the latest advances in translational research integrating encephalitis-induced seizure and epilepsy models, in tandem with progress in clinical diagnosis of inflammation and virally mediated epilepsy. This improved awareness of the shared biological underpinnings of epileptogenesis following brain infection or autoimmune encephalitis is anticipated to beneficially impact the global burden of epilepsy.
Collapse
Affiliation(s)
| | | | - Julika Pitsch
- Department of Epileptology, University Hospital Bonn, Bonn, Germany
| | - Harald Sontheimer
- Department of Neuroscience, School of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Lawrence J. Hirsch
- Department of Neurology, School of Medicine, Yale University, New Haven, CT, USA
| | - Aristea S. Galanopoulou
- Saul R. Korey Department of Neurology, Isabelle Rapin Division of Child Neurology, Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Jennifer A. Kearney
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| |
Collapse
|
6
|
Wallis GJ, Bell LA, Wagner JN, Buxton L, Balachandar L, Wilcox KS. Reactive microglia fail to respond to environmental damage signals in a viral-induced mouse model of temporal lobe epilepsy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.06.583768. [PMID: 38558969 PMCID: PMC10979929 DOI: 10.1101/2024.03.06.583768] [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] [Indexed: 04/04/2024]
Abstract
Microglia are highly adaptable innate immune cells that rapidly respond to damage signals in the brain through adoption of a reactive phenotype and production of defensive inflammatory cytokines. Microglia express a distinct transcriptome, encoding receptors that allow them to dynamically respond to pathogens, damage signals, and cellular debris. Expression of one such receptor, the microglia-specific purinergic receptor P2ry12, is known to be downregulated in reactive microglia. Here, we explore the microglial response to purinergic damage signals in reactive microglia in the TMEV mouse model of viral brain infection and temporal lobe epilepsy. Using two-photon calcium imaging in acute hippocampal brain slices, we found that the ability of microglia to detect damage signals, engage calcium signaling pathways, and chemoattract towards laser-induced tissue damage was dramatically reduced during the peak period of seizures, cytokine production, and infection. Using combined RNAscope in situ hybridization and immunohistochemistry, we found that during this same stage of heightened infection and seizures, microglial P2ry12 expression was reduced, while the pro-inflammatory cytokine TNF-a expression was upregulated in microglia, suggesting that the depressed ability of microglia to respond to new damage signals via P2ry12 occurs during the time when local elevated cytokine production contributes to seizure generation following infection. Therefore, changes in microglial purinergic receptors during infection likely limit the ability of reactive microglia to respond to new threats in the CNS and locally contain the scale of the innate immune response in the brain.
Collapse
Affiliation(s)
- Glenna J. Wallis
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, UT 80904
| | - Laura A. Bell
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, UT 80904
- Interdepartmental Program in Neuroscience, University of Utah, Salt Lake City, UT, 80904
| | - John N. Wagner
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, UT 80904
| | - Lauren Buxton
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, UT 80904
| | - Lakshmini Balachandar
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, UT 80904
| | - Karen S. Wilcox
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, UT 80904
- Interdepartmental Program in Neuroscience, University of Utah, Salt Lake City, UT, 80904
| |
Collapse
|
7
|
DePaula-Silva AB. The Contribution of Microglia and Brain-Infiltrating Macrophages to the Pathogenesis of Neuroinflammatory and Neurodegenerative Diseases during TMEV Infection of the Central Nervous System. Viruses 2024; 16:119. [PMID: 38257819 PMCID: PMC10819099 DOI: 10.3390/v16010119] [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: 12/05/2023] [Revised: 01/06/2024] [Accepted: 01/09/2024] [Indexed: 01/24/2024] Open
Abstract
The infection of the central nervous system (CNS) with neurotropic viruses induces neuroinflammation and is associated with the development of neuroinflammatory and neurodegenerative diseases, including multiple sclerosis and epilepsy. The activation of the innate and adaptive immune response, including microglial, macrophages, and T and B cells, while required for efficient viral control within the CNS, is also associated with neuropathology. Under healthy conditions, resident microglia play a pivotal role in maintaining CNS homeostasis. However, during pathological events, such as CNS viral infection, microglia become reactive, and immune cells from the periphery infiltrate into the brain, disrupting CNS homeostasis and contributing to disease development. Theiler's murine encephalomyelitis virus (TMEV), a neurotropic picornavirus, is used in two distinct mouse models: TMEV-induced demyelination disease (TMEV-IDD) and TMEV-induced seizures, representing mouse models of multiple sclerosis and epilepsy, respectively. These murine models have contributed substantially to our understanding of the pathophysiology of MS and seizures/epilepsy following viral infection, serving as critical tools for identifying pharmacological targetable pathways to modulate disease development. This review aims to discuss the host-pathogen interaction during a neurotropic picornavirus infection and to shed light on our current understanding of the multifaceted roles played by microglia and macrophages in the context of these two complexes viral-induced disease.
Collapse
Affiliation(s)
- Ana Beatriz DePaula-Silva
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Utah, Salt Lake City, UT 84112, USA
| |
Collapse
|
8
|
Zierath DK, Davidson S, Manoukian J, White HS, Meeker S, Ericsson A, Barker-Haliski M. Diet composition and sterilization modifies intestinal microbiome diversity and burden of Theiler's virus infection-induced acute seizures. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.17.562694. [PMID: 37905123 PMCID: PMC10614857 DOI: 10.1101/2023.10.17.562694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
Objective Central nervous system infection with Theiler's murine encephalomyelitis virus (TMEV) in C57BL/6J mice can model acquired epileptogenesis. Diet alters the acute seizure incidence in TMEV-infected mice; yet it is unclear whether intestinal dysbiosis may also impact acute or chronic behavioral comorbidities. This study thus assessed the impact of diet sterilization in a specific pathogen-free vivarium on acute seizure presentation, the composition of the gut microbiome, and chronic behavioral comorbidities of epilepsy. Methods Baseline fecal samples were collected from male C57BL/6J mice (4-5 weeks-old; Jackson Labs) upon arrival. Mice were randomized to either autoclaved (AC) or irradiated (IR) diet (Prolab RMH 3000 - UU diets) or IR (Picolab 5053 - UW IR diet). Mice then underwent intracerebral TMEV or PBS injection three days later. Fecal samples were collected from a subset of mice at infection (Day 0) and Day 7 post-infection. Epilepsy-related working memory deficits and seizure threshold were assessed 6 weeks post-infection. Gut microbiome diversity was determined by 16S rRNA amplicon sequencing of fecal samples. Results TMEV-infected mice displayed acute handling-induced seizures, regardless of diet: 28/57 UW IR (49.1%), 30/41 UU IR (73.2%), and 47/77 UU AC (61%) mice displayed seizures. The number of observed seizures significantly differed: UW IR mice had 2.2±2.8 seizures (mean±standard deviation), UU IR mice had 3.5±2.9 seizures, and UU AC mice had 4.4±3.8 seizures during the 7-day monitoring period. The composition of the gut microbiome significantly differed in TMEV-infected mice fed the UU AC diet, with most measured differences occurring in Gram-positive bacteria. TMEV-infected mice fed the UU AC diet displayed worsened chronic working memory. Significance Intestinal dysbiosis evokes stark differences in acute seizure presentation in the TMEV model and vastly influences the trajectory of post-TMEV infection-induced behavioral comorbidities of epilepsy. Our study reveals a novel disease-modifying contribution of intestinal bacterial species after TMEV-induced acute seizures.
Collapse
Affiliation(s)
- Dannielle K. Zierath
- Department of Pharmacy, School of Pharmacy, University of Washington, Seattle, WA
| | - Stephanie Davidson
- Department of Pharmacy, School of Pharmacy, University of Washington, Seattle, WA
| | - Jonathan Manoukian
- Department of Pharmacy, School of Pharmacy, University of Washington, Seattle, WA
| | - H. Steve White
- Department of Pharmacy, School of Pharmacy, University of Washington, Seattle, WA
| | - Stacey Meeker
- Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, OH
| | - Aaron Ericsson
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO
| | | |
Collapse
|
9
|
Hu A, Yuan H, Qin Y, Zhu Y, Zhang L, Chen Q, Wu L. Lipopolysaccharide (LPS) increases susceptibility to epilepsy via interleukin-1 type 1 receptor signaling. Brain Res 2022; 1793:148052. [PMID: 35970265 DOI: 10.1016/j.brainres.2022.148052] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 07/27/2022] [Accepted: 08/09/2022] [Indexed: 11/18/2022]
Abstract
Epilepsy is the most common disease of the nervous system, characterized by aberrant normal brain activity. Neuroinflammation is a prominent feature in the brain in epileptic humans and animal models of epilepsy. However, it remains elusive as to how peripheral inflammation affects epilepsy. Herein we demonstrated significantly greater seizure susceptibility and severity of epilepsy under kainic acid (KA) via intraperitoneal (i.p.) injection of lipopolysaccharide (LPS) in mouse model of epilepsy. Nissl staining was employed for assessment of the neuronal damage, immunofluorescence for staining of the microglial cells and astrocytes in the mouse brain slices, and ELISA for detection of the changes of inflammatory factors. We observed a smaller population of viable neurons in CA1 and CA3 regions, a greater population of IBA-1-positive and GFAP-positive cells, with a significant upregulation of IL-1β and IL-6 in hippocampus of epileptic mice when treated with LPS, indicating that LPS aggravates hippocampal neuron injury in epilepsy, and induces neuroinflammation in the hippocampus. In addition, we provide an evident increase in BrdU+/DCX+ and Nestin+ cell populations in dentate gyrus (DG) in LPS-treated group, versus saline group on epileptic mouse model, which demonstrated LPS treatment enhanced hippocampal neurogenesis. In order to investigate whether interleukin-1 type 1 (IL-1R1) signaling is involved in this process, we adopted IL-1R1 globally restored mice (IL-1R1GR/GR) as an IL-1R1 reporter to visualize labeling of IL-1R1 mRNA and protein by means of RFP staining. Strikingly, the RFP immunofluorescence revealed increased IL-1R1 expression in LPS-treated group, versus saline group. Further, blockage of central IL-1R1 alleviated seizure susceptibility and severity of epilepsy. In summary, our findings suggested that LPS could enhance central inflammatory response and aggravate the susceptibility to epileptic seizure, which we postulated to be mediated by IL-1R1.
Collapse
Affiliation(s)
- Ankang Hu
- Laboratory Animal Center, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Honghua Yuan
- Laboratory Animal Center, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Ying Qin
- Laboratory Animal Center, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Yuhua Zhu
- Laboratory Animal Center, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Lingzhi Zhang
- Laboratory Animal Center, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Quangang Chen
- Laboratory Animal Center, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Lianlian Wu
- Laboratory Animal Center, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China.
| |
Collapse
|
10
|
Löscher W, Howe CL. Molecular Mechanisms in the Genesis of Seizures and Epilepsy Associated With Viral Infection. Front Mol Neurosci 2022; 15:870868. [PMID: 35615063 PMCID: PMC9125338 DOI: 10.3389/fnmol.2022.870868] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 04/05/2022] [Indexed: 12/16/2022] Open
Abstract
Seizures are a common presenting symptom during viral infections of the central nervous system (CNS) and can occur during the initial phase of infection ("early" or acute symptomatic seizures), after recovery ("late" or spontaneous seizures, indicating the development of acquired epilepsy), or both. The development of acute and delayed seizures may have shared as well as unique pathogenic mechanisms and prognostic implications. Based on an extensive review of the literature, we present an overview of viruses that are associated with early and late seizures in humans. We then describe potential pathophysiologic mechanisms underlying ictogenesis and epileptogenesis, including routes of neuroinvasion, viral control and clearance, systemic inflammation, alterations of the blood-brain barrier, neuroinflammation, and inflammation-induced molecular reorganization of synapses and neural circuits. We provide clinical and animal model findings to highlight commonalities and differences in these processes across various neurotropic or neuropathogenic viruses, including herpesviruses, SARS-CoV-2, flaviviruses, and picornaviruses. In addition, we extensively review the literature regarding Theiler's murine encephalomyelitis virus (TMEV). This picornavirus, although not pathogenic for humans, is possibly the best-characterized model for understanding the molecular mechanisms that drive seizures, epilepsy, and hippocampal damage during viral infection. An enhanced understanding of these mechanisms derived from the TMEV model may lead to novel therapeutic interventions that interfere with ictogenesis and epileptogenesis, even within non-infectious contexts.
Collapse
Affiliation(s)
- Wolfgang Löscher
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine, Hannover, Germany
- Center for Systems Neuroscience, Hannover, Germany
| | - Charles L. Howe
- Division of Experimental Neurology, Department of Neurology, Mayo Clinic, Rochester, MN, United States
- Center for Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
11
|
Sharma R, Casillas-Espinosa PM, Dill LK, Rewell SSJ, Hudson MR, O'Brien TJ, Shultz SR, Semple BD. Pediatric traumatic brain injury and a subsequent transient immune challenge independently influenced chronic outcomes in male mice. Brain Behav Immun 2022; 100:29-47. [PMID: 34808288 DOI: 10.1016/j.bbi.2021.11.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Revised: 10/27/2021] [Accepted: 11/15/2021] [Indexed: 01/30/2023] Open
Abstract
Traumatic brain injury (TBI) is a major contributor to death and disability worldwide. Children are at particularly high risk of both sustaining a TBI and experiencing serious long-term consequences, such as cognitive deficits, mental health problems and post-traumatic epilepsy. Severe TBI patients are highly susceptible to nosocomial infections, which are mostly acquired within the first week of hospitalization post-TBI. Yet the potential chronic impact of such acute infections following pediatric TBI remains unclear. In this study, we hypothesized that a peripheral immune challenge, such as lipopolysaccharide (LPS)-mimicking a hospital-acquired infection-would worsen inflammatory, neurobehavioral, and seizure outcomes after experimental pediatric TBI. To test this, three-week old male C57Bl/6J mice received a moderate controlled cortical impact or sham surgery, followed by 1 mg/kg i.p. LPS (or 0.9% saline vehicle) at 4 days TBI. Mice were randomized to four groups; sham-saline, sham-LPS, TBI-saline or TBI-LPS (n = 15/group). Reduced general activity and increased anxiety-like behavior were observed within 24 h in LPS-treated mice, indicating a transient sickness response. LPS-treated mice also exhibited a reduction in body weights, which persisted chronically. From 2 months post-injury, mice underwent a battery of tests for sensorimotor, cognitive, and psychosocial behaviors. TBI resulted in hyperactivity and spatial memory deficits, independent of LPS; whereas LPS resulted in subtle deficits in spatial memory retention. At 5 months post-injury, video-electroencephalographic recordings were obtained to evaluate both spontaneous seizure activity as well as the evoked seizure response to pentylenetetrazol (PTZ). TBI increased susceptibility to PTZ-evoked seizures; whereas LPS appeared to increase the incidence of spontaneous seizures. Post-mortem analyses found that TBI, but not LPS, resulted in robust glial reactivity and loss of cortical volume. A TBI × LPS interaction in hippocampal volume suggested that TBI-LPS mice had a subtle increase in ipsilateral hippocampus tissue loss; however, this was not reflected in neuronal cell counts. Both TBI and LPS independently had modest effects on chronic hippocampal gene expression. Together, contrary to our hypothesis, we observed minimal synergy between TBI and LPS. Instead, pediatric TBI and a subsequent transient immune challenge independently influenced chronic outcomes. These findings have implications for future preclinical modeling as well as acute post-injury patient management.
Collapse
Affiliation(s)
- Rishabh Sharma
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Pablo M Casillas-Espinosa
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia; Department of Neurology, Alfred Health, Prahran, VIC, Australia; Department of Medicine (Royal Melbourne Hospital), The University of Melbourne, Parkville, VIC, Australia
| | - Larissa K Dill
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia; Department of Neurology, Alfred Health, Prahran, VIC, Australia
| | - Sarah S J Rewell
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia; Department of Neurology, Alfred Health, Prahran, VIC, Australia
| | - Matthew R Hudson
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Terence J O'Brien
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia; Department of Neurology, Alfred Health, Prahran, VIC, Australia; Department of Medicine (Royal Melbourne Hospital), The University of Melbourne, Parkville, VIC, Australia
| | - Sandy R Shultz
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia; Department of Neurology, Alfred Health, Prahran, VIC, Australia; Department of Medicine (Royal Melbourne Hospital), The University of Melbourne, Parkville, VIC, Australia
| | - Bridgette D Semple
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia; Department of Neurology, Alfred Health, Prahran, VIC, Australia; Department of Medicine (Royal Melbourne Hospital), The University of Melbourne, Parkville, VIC, Australia.
| |
Collapse
|
12
|
DePaula-Silva AB, Bell LA, Wallis GJ, Wilcox KS. Inflammation Unleashed in Viral-Induced Epileptogenesis. Epilepsy Curr 2021; 21:433-440. [PMID: 34924851 PMCID: PMC8652320 DOI: 10.1177/15357597211040939] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Viral infection of the central nervous system increasingly places people at risk of developing life-threatening and treatment-resistant acute and chronic seizures (epilepsy). The emergence of new human viruses due to ongoing social, political, and ecological changes places people at risk more than ever before. The development of new preventative or curative strategies is critical to address this burden. However, our understanding of the complex relationship between viruses and the brain has been hindered by the lack of animal models that survive the initial infection and are amenable for long-term mechanistic, behavioral, and pharmacological studies in the process of viral-induced epileptogenesis. In this review, we focus on the Theiler’s murine encephalomyelitis virus (TMEV) mouse model of viral infection–induced epilepsy. The TMEV model has a number of important advantages to address the quintessential processes underlying the development of epilepsy following a viral infection, as well as fuel new therapeutic development. In this review, we highlight the contributions of the TMEV model to our current understanding of the relationship between viral infection, inflammation, and seizures.
Collapse
Affiliation(s)
| | - Laura A. Bell
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, UT, USA
- Interdepartmental Program in Neuroscience, University of Utah, Salt Lake City, UT, USA
| | - Glenna J. Wallis
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, UT, USA
| | - Karen S. Wilcox
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, UT, USA
- Interdepartmental Program in Neuroscience, University of Utah, Salt Lake City, UT, USA
- Karen S. Wilcox, PhD, Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, UT 84112, USA.
| |
Collapse
|
13
|
Sanchez JMS, DePaula-Silva AB, Doty DJ, Hanak TJ, Truong A, Libbey JE, Fujinami RS. The CSF1R-Microglia Axis Has Protective Host-Specific Roles During Neurotropic Picornavirus Infection. Front Immunol 2021; 12:621090. [PMID: 34566948 PMCID: PMC8458822 DOI: 10.3389/fimmu.2021.621090] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Accepted: 08/16/2021] [Indexed: 01/19/2023] Open
Abstract
Viral encephalitis is a major cause of morbidity and mortality, but the manifestation of disease varies greatly between individuals even in response to the same virus. Microglia are professional antigen presenting cells that reside in the central nervous system (CNS) parenchyma that are poised to respond to viral insults. However, the role of microglia in initiating and coordinating the antiviral response is not completely understood. Utilizing Theiler's murine encephalomyelitis virus (TMEV), a neurotropic picornavirus, and PLX5622, a small molecule inhibitor of colony-stimulating factor 1 receptor (CSF1R) signaling that can deplete microglia in the CNS; we investigated the role of the CSF1R-microglia axis in neurotropic picornavirus infection of C57BL/6J and SJL/J mice. These mouse strains differ in their ability to clear TMEV and exhibit different neurological disease in response to TMEV infection. CSF1R antagonism in C57BL/6J mice, which normally clear TMEV in the CNS, led to acute fatal encephalitis. In contrast, CSF1R antagonism in SJL/J mice, which normally develop a chronic CNS TMEV infection, did not result in acute encephalitis, but exacerbated TMEV-induced demyelination. Immunologically, inhibition of CSF1R in C57BL/6J mice reduced major histocompatibility complex II expression in microglia, decreased the proportion of regulatory T cells in the CNS, and upregulated proinflammatory pathways in CNS T cells. Acute CSF1R inhibition in SJL/J mice had no effect on microglial MHC-II expression and upregulated anti-inflammatory pathways in CNS T cells, however chronic CSF1R inhibition resulted in broad immunosuppression. Our results demonstrate strain-specific effects of the CSF1R-microglia axis in the context of neurotropic viral infection as well as inherent differences in microglial antigen presentation and subsequent T cell crosstalk that contribute to susceptibility to neurotropic picornavirus infection.
Collapse
Affiliation(s)
- John Michael S. Sanchez
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT, United States
| | | | - Daniel J. Doty
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT, United States
| | - Tyler J. Hanak
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT, United States
| | - Amanda Truong
- Department of Oncological Sciences, Huntsman Cancer Institute, Salt Lake City, UT, United States
| | - Jane E. Libbey
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT, United States
| | - Robert S. Fujinami
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT, United States
| |
Collapse
|
14
|
Bell LA, Wallis GJ, Wilcox KS. Reactivity and increased proliferation of NG2 cells following central nervous system infection with Theiler's murine encephalomyelitis virus. J Neuroinflammation 2020; 17:369. [PMID: 33272299 PMCID: PMC7713670 DOI: 10.1186/s12974-020-02043-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 11/24/2020] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Neuron-glial antigen 2 (NG2) cells are a glial cell type tiled throughout the gray and white matter of the central nervous system (CNS). NG2 cells are known for their ability to differentiate into oligodendrocytes and are commonly referred to as oligodendrocyte precursor cells. However, recent investigations have begun to identify additional functions of NG2 cells in CNS health and pathology. NG2 cells form physical and functional connections with neurons and other glial cell types throughout the CNS, allowing them to monitor and respond to the neural environment. Growing evidence indicates that NG2 cells become reactive under pathological conditions, though their specific roles are only beginning to be elucidated. While reactive microglia and astrocytes are well-established contributors to neuroinflammation and the development of epilepsy following CNS infection, the dynamics of NG2 cells remain unclear. Therefore, we investigated NG2 cell reactivity in a viral-induced mouse model of temporal lobe epilepsy. METHODS C57BL6/J mice were injected intracortically with Theiler's murine encephalomyelitis virus (TMEV) or PBS. Mice were graded twice daily for seizures between 3 and 7 days post-injection (dpi). At 4 and 14 dpi, brains were fixed and stained for NG2, the microglia/macrophage marker IBA1, and the proliferation marker Ki-67. Confocal z stacks were acquired in both the hippocampus and the overlying cortex. Total field areas stained by each cell marker and total field area of colocalized pixels between NG2 and Ki67 were compared between groups. RESULTS Both NG2 cells and microglia/macrophages displayed increased immunoreactivity and reactive morphologies in the hippocampus of TMEV-injected mice. While increased immunoreactivity for IBA1 was also present in the cortex, there was no significant change in NG2 immunoreactivity in the cortex following TMEV infection. Colocalization analysis for NG2 and Ki-67 revealed a significant increase in overlap between NG2 and Ki-67 in the hippocampus of TMEV-injected mice at both time points, but no significant differences in cortex. CONCLUSIONS NG2 cells acquire a reactive phenotype and proliferate in response to TMEV infection. These results suggest that NG2 cells alter their function in response to viral encephalopathy, making them potential targets to prevent the development of epilepsy following viral infection.
Collapse
Affiliation(s)
- Laura A Bell
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, UT, 84112, USA
- Interdepartmental Program in Neuroscience, University of Utah, Salt Lake City, UT, 84112, USA
| | - Glenna J Wallis
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, UT, 84112, USA
| | - Karen S Wilcox
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, UT, 84112, USA.
- Interdepartmental Program in Neuroscience, University of Utah, Salt Lake City, UT, 84112, USA.
| |
Collapse
|
15
|
Bosco DB, Tian DS, Wu LJ. Neuroimmune interaction in seizures and epilepsy: focusing on monocyte infiltration. FEBS J 2020; 287:4822-4837. [PMID: 32473609 DOI: 10.1111/febs.15428] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 05/06/2020] [Accepted: 05/26/2020] [Indexed: 12/30/2022]
Abstract
Epilepsy is a major neurological condition that affects millions of people globally. While a number of interventions have been developed to mitigate this condition, a significant number of patients are refractory to these treatments. Consequently, other avenues of research are needed. One such avenue is modulation of the immune system response to this condition, which has mostly focused on microglia, the resident immune cells of the central nervous system (CNS). However, other immune cells can impact neurological conditions, principally blood-borne monocytes that can infiltrate into brain parenchyma after seizures. As such, this review will first discuss how monocytes can be recruited to the CNS and how they can be distinguished from there immunological cousins, microglia. Then, we will explore what is known about the role monocytes have within seizure pathogenesis and epilepsy. Considering how little is known about monocyte function in seizure- and epilepsy-related pathologies, further studies are warranted that investigate infiltrated blood-borne monocytes as a potential therapeutic target for epilepsy treatment.
Collapse
Affiliation(s)
- Dale B Bosco
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - Dai-Shi Tian
- Department of Neurology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Long-Jun Wu
- Department of Neurology, Mayo Clinic, Rochester, MN, USA.,Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA.,Department of Immunology, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
16
|
Abstract
PURPOSE OF REVIEW The present review will outline neuroprotective and neurotoxic effects of central nervous system (CNS) infiltrating T cells during viral infections. Evidence demonstrating differential roles for antiviral effector and resident memory T-cell subsets in virologic control and immunopathology in the CNS will be discussed. Potential therapeutic targets emanating from a growing understanding of T-cell-initiated neuropathology that impacts learning and memory will also be delineated. RECENT FINDINGS The critical role for T cells in preventing and clearing CNS infections became incontrovertible during the era of acquired immunodeficiency syndrome. Recent studies have further defined differential roles of T-cell subsets, including resident memory T cells (Trm), in antiviral immunity and, unexpectedly, in postinfectious cognitive dysfunction. Mechanisms of T-cell-mediated effects include differential innate immune signaling within neural cells that are virus-specific. SUMMARY T-cell cytokines that are essential for cell-mediated virologic control during neurotropic viral infections have recently been identified as potential targets to prevent post-infection memory disorders. Further identification of T-cell subsets, their antigen specificity, and postinfection localization of Trm will enhance the efficacy of immunotherapies through minimization of immunopathology.
Collapse
Affiliation(s)
| | - Robyn S. Klein
- Departments of Medicine
- Pathology and Immunology
- Neurosciences Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
17
|
DePaula-Silva AB, Gorbea C, Doty DJ, Libbey JE, Sanchez JMS, Hanak TJ, Cazalla D, Fujinami RS. Differential transcriptional profiles identify microglial- and macrophage-specific gene markers expressed during virus-induced neuroinflammation. J Neuroinflammation 2019; 16:152. [PMID: 31325960 PMCID: PMC6642742 DOI: 10.1186/s12974-019-1545-x] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 07/10/2019] [Indexed: 01/09/2023] Open
Abstract
Background In the healthy central nervous system (CNS), microglia are found in a homeostatic state and peripheral macrophages are absent from the brain. Microglia play key roles in maintaining CNS homeostasis and acting as first responders to infection and inflammation, and peripheral macrophages infiltrate the CNS during neuroinflammation. Due to their distinct origins and functions, discrimination between these cell populations is essential to the comprehension of neuroinflammatory disorders. Studies comparing the gene profiles of microglia and peripheral macrophages, or macrophages in vitro-derived from bone marrow, under non-infectious conditions of the CNS, have revealed valuable microglial-specific genes. However, studies comparing gene profiles between CNS-infiltrating macrophages and microglia, when both are isolated from the CNS during viral-induced neuroinflammation, are lacking. Methods We isolated, via flow cytometry, microglia and infiltrating macrophages from the brains of Theiler’s murine encephalomyelitis virus-infected C57BL/6 J mice and used RNA-Seq, followed by validation with qPCR, to examine the differential transcriptional profiles of these cells. We utilized primary literature defining subcellular localization to determine whether or not particular proteins extracted from the transcriptional profiles were expressed at the cell surface. The surface expression and cellular specificity of triggering receptor expressed on myeloid cells 1 (TREM-1) protein were examined via flow cytometry. We also examined the immune response gene profile within the transcriptional profiles of these isolated microglia and infiltrating macrophages. Results We have identified and validated new microglial- and macrophage-specific genes, encoding cell surface proteins, expressed at the peak of neuroinflammation. TREM-1 protein was confirmed to be expressed by infiltrating macrophages, not microglia, at the peak of neuroinflammation. We also identified both unique and redundant immune functions, through examination of the immune response gene profiles, of microglia and infiltrating macrophages during neurotropic viral infection. Conclusions The differential expression of cell surface-specific genes during neuroinflammation can potentially be used to discriminate between microglia and macrophages as well as provide a resource that can be further utilized to target and manipulate specific cell responses during neuroinflammation. Electronic supplementary material The online version of this article (10.1186/s12974-019-1545-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ana Beatriz DePaula-Silva
- Department of Pathology, University of Utah School of Medicine, 15 North Medical Drive East, 2600 EEJMRB, Salt Lake City, UT, 84112, USA
| | - Carlos Gorbea
- Department of Biochemistry, University of Utah, 15 North Medical Drive East, 4100 EEJMRB, Salt Lake City, UT, 84112, USA
| | - Daniel J Doty
- Department of Pathology, University of Utah School of Medicine, 15 North Medical Drive East, 2600 EEJMRB, Salt Lake City, UT, 84112, USA
| | - Jane E Libbey
- Department of Pathology, University of Utah School of Medicine, 15 North Medical Drive East, 2600 EEJMRB, Salt Lake City, UT, 84112, USA
| | - John Michael S Sanchez
- Department of Pathology, University of Utah School of Medicine, 15 North Medical Drive East, 2600 EEJMRB, Salt Lake City, UT, 84112, USA
| | - Tyler J Hanak
- Department of Pathology, University of Utah School of Medicine, 15 North Medical Drive East, 2600 EEJMRB, Salt Lake City, UT, 84112, USA
| | - Demián Cazalla
- Department of Biochemistry, University of Utah, 15 North Medical Drive East, 4100 EEJMRB, Salt Lake City, UT, 84112, USA
| | - Robert S Fujinami
- Department of Pathology, University of Utah School of Medicine, 15 North Medical Drive East, 2600 EEJMRB, Salt Lake City, UT, 84112, USA.
| |
Collapse
|
18
|
Sanchez JMS, DePaula-Silva AB, Doty DJ, Truong A, Libbey JE, Fujinami RS. Microglial cell depletion is fatal with low level picornavirus infection of the central nervous system. J Neurovirol 2019; 25:415-421. [PMID: 30859497 PMCID: PMC6635090 DOI: 10.1007/s13365-019-00740-3] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 02/13/2019] [Accepted: 02/21/2019] [Indexed: 12/21/2022]
Abstract
Microglia are the only resident myeloid cell in the central nervous system (CNS) parenchyma, but the role of microglia in the context of neurotropic viral infection is poorly understood. Using different amounts of Theiler's murine encephalomyelitis virus (TMEV) in a preclinical model of epilepsy and PLX5622, a colony stimulating factor-1 receptor inhibitor that selectively depletes microglia in the CNS, we report that microglia-depleted, TMEV-infected mice develop seizures, manifest paralysis, and uniformly succumb to fatal encephalitis regardless of viral amount. CNS demyelination correlates with viral amount; however, viral amount does not correlate with axon damage and TMEV antigen in the CNS.
Collapse
Affiliation(s)
- John Michael S Sanchez
- Department of Pathology, University of Utah School of Medicine, 15 North Medical Drive East, 2600 EEJMRB, Salt Lake City, UT, 84112, USA
| | - Ana Beatriz DePaula-Silva
- Department of Pathology, University of Utah School of Medicine, 15 North Medical Drive East, 2600 EEJMRB, Salt Lake City, UT, 84112, USA
| | - Daniel J Doty
- Department of Pathology, University of Utah School of Medicine, 15 North Medical Drive East, 2600 EEJMRB, Salt Lake City, UT, 84112, USA
| | - Amanda Truong
- Department of Oncological Sciences, Huntsman Cancer Institute, 2000 Circle of Hope, 2724 HCI-SOUTH, Salt Lake City, UT, 84112, USA
| | - Jane E Libbey
- Department of Pathology, University of Utah School of Medicine, 15 North Medical Drive East, 2600 EEJMRB, Salt Lake City, UT, 84112, USA
| | - Robert S Fujinami
- Department of Pathology, University of Utah School of Medicine, 15 North Medical Drive East, 2600 EEJMRB, Salt Lake City, UT, 84112, USA.
| |
Collapse
|
19
|
Gerhauser I, Hansmann F, Ciurkiewicz M, Löscher W, Beineke A. Facets of Theiler's Murine Encephalomyelitis Virus-Induced Diseases: An Update. Int J Mol Sci 2019; 20:ijms20020448. [PMID: 30669615 PMCID: PMC6358740 DOI: 10.3390/ijms20020448] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 01/15/2019] [Accepted: 01/18/2019] [Indexed: 12/31/2022] Open
Abstract
Theiler’s murine encephalomyelitis virus (TMEV), a naturally occurring, enteric pathogen of mice is a Cardiovirus of the Picornaviridae family. Low neurovirulent TMEV strains such as BeAn cause a severe demyelinating disease in susceptible SJL mice following intracerebral infection. Furthermore, TMEV infections of C57BL/6 mice cause acute polioencephalitis initiating a process of epileptogenesis that results in spontaneous recurrent epileptic seizures in approximately 50% of affected mice. Moreover, C3H mice develop cardiac lesions after an intraperitoneal high-dose application of TMEV. Consequently, TMEV-induced diseases are widely used as animal models for multiple sclerosis, epilepsy, and myocarditis. The present review summarizes morphological lesions and pathogenic mechanisms triggered by TMEV with a special focus on the development of hippocampal degeneration and seizures in C57BL/6 mice as well as demyelination in the spinal cord in SJL mice. Furthermore, a detailed description of innate and adaptive immune responses is given. TMEV studies provide novel insights into the complexity of organ- and mouse strain-specific immunopathology and help to identify factors critical for virus persistence.
Collapse
Affiliation(s)
- Ingo Gerhauser
- Department of Pathology, University of Veterinary Medicine, Bünteweg 17, 30559 Hannover, Germany.
| | - Florian Hansmann
- Department of Pathology, University of Veterinary Medicine, Bünteweg 17, 30559 Hannover, Germany.
- Center for System Neuroscience, 30559 Hannover, Germany.
| | - Malgorzata Ciurkiewicz
- Department of Pathology, University of Veterinary Medicine, Bünteweg 17, 30559 Hannover, Germany.
- Center for System Neuroscience, 30559 Hannover, Germany.
| | - Wolfgang Löscher
- Center for System Neuroscience, 30559 Hannover, Germany.
- Department of Pharmacology, University of Veterinary Medicine, Bünteweg 17, 30559 Hannover, Germany.
| | - Andreas Beineke
- Department of Pathology, University of Veterinary Medicine, Bünteweg 17, 30559 Hannover, Germany.
- Center for System Neuroscience, 30559 Hannover, Germany.
| |
Collapse
|
20
|
Waltl I, Käufer C, Gerhauser I, Chhatbar C, Ghita L, Kalinke U, Löscher W. Microglia have a protective role in viral encephalitis-induced seizure development and hippocampal damage. Brain Behav Immun 2018; 74:186-204. [PMID: 30217535 PMCID: PMC7111316 DOI: 10.1016/j.bbi.2018.09.006] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 08/23/2018] [Accepted: 09/06/2018] [Indexed: 12/12/2022] Open
Abstract
In the central nervous system (CNS), innate immune surveillance is mainly coordinated by microglia. These CNS resident myeloid cells are assumed to help orchestrate the immune response against infections of the brain. However, their specific role in this process and their interactions with CNS infiltrating immune cells, such as blood-borne monocytes and T cells are only incompletely understood. The recent development of PLX5622, a specific inhibitor of colony-stimulating factor 1 receptor that depletes microglia, allows studying the role of microglia in conditions of brain injury such as viral encephalitis, the most common form of brain infection. Here we used this inhibitor in a model of viral infection-induced epilepsy, in which C57BL/6 mice are infected by a picornavirus (Theiler's murine encephalomyelitis virus) and display seizures and hippocampal damage. Our results show that microglia are required early after infection to limit virus distribution and persistence, most likely by modulating T cell activation. Microglia depletion accelerated the occurrence of seizures, exacerbated hippocampal damage, and led to neurodegeneration in the spinal cord, which is normally not observed in this mouse strain. This study enhances our understanding of the role of microglia in viral encephalitis and adds to the concept of microglia-T cell crosstalk.
Collapse
Affiliation(s)
- Inken Waltl
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Germany,Center for Systems Neuroscience, Hannover, Germany
| | - Christopher Käufer
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Germany
| | - Ingo Gerhauser
- Department of Pathology, University of Veterinary Medicine Hannover, Germany
| | - Chintan Chhatbar
- Institute for Experimental Infection Research, TWINCORE, Center for Experimental and Clinical Infection Research, a Joint Venture Between the Helmholtz Center for Infection Research, Braunschweig, and the Hannover Medical School, Hannover, Germany
| | - Luca Ghita
- Institute for Experimental Infection Research, TWINCORE, Center for Experimental and Clinical Infection Research, a Joint Venture Between the Helmholtz Center for Infection Research, Braunschweig, and the Hannover Medical School, Hannover, Germany
| | - Ulrich Kalinke
- Center for Systems Neuroscience, Hannover, Germany,Institute for Experimental Infection Research, TWINCORE, Center for Experimental and Clinical Infection Research, a Joint Venture Between the Helmholtz Center for Infection Research, Braunschweig, and the Hannover Medical School, Hannover, Germany
| | - Wolfgang Löscher
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Germany; Center for Systems Neuroscience, Hannover, Germany.
| |
Collapse
|
21
|
Hanak TJ, Libbey JE, Doty DJ, Sim JT, DePaula-Silva AB, Fujinami RS. Positive modulation of mGluR5 attenuates seizures and reduces TNF-α + macrophages and microglia in the brain in a murine model of virus-induced temporal lobe epilepsy. Exp Neurol 2018; 311:194-204. [PMID: 30316834 DOI: 10.1016/j.expneurol.2018.10.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 09/12/2018] [Accepted: 10/10/2018] [Indexed: 12/29/2022]
Abstract
Viral encephalitis markedly increases the risk for the development of epilepsy. The Theiler's murine encephalomyelitis virus (TMEV)-induced model of seizures/epilepsy is a murine model of both viral-induced seizures/epilepsy and human Temporal Lobe Epilepsy. The inflammatory cytokines interleukin (IL)-6 and tumor necrosis factor (TNF)-α have been shown to play a role in seizure development in the TMEV-induced model of seizures/epilepsy, and infiltrating macrophages along with microglia have been shown to be major producers of these cytokines. The metabotropic glutamate receptor 5 (mGluR5) is a G-protein coupled receptor that has been shown to reduce IL-6 and TNF-α and to provide neuroprotection in other disease models. Therefore, we hypothesized that stimulation of mGluR5 would not only reduce seizures but attenuate IL-6 and TNF-α production in microglia and macrophages in the TMEV model. We found that pharmacological stimulation of mGluR5 with the selective positive allosteric modulator VU0360172 not only reduced acute seizure outcomes, but also reduced the percent of microglia and macrophages producing TNF-α 3 days post infection. Furthermore, treatment with VU0360172 did not alter the level of viral antigen, compared to controls, showing that this treatment does not compromise viral clearance. These results establish that mGluR5 may represent a therapeutic target in the TMEV-induced model of seizures/epilepsy.
Collapse
Affiliation(s)
- Tyler J Hanak
- Department of Pathology, University of Utah School of Medicine, 15 North Medical Drive East, 2600 EEJMRB, Salt Lake City, UT 84112, USA; Interdepartmental Program in Neuroscience, University of Utah, 20 South 2030 East, Salt Lake City, UT 84112, USA
| | - Jane E Libbey
- Department of Pathology, University of Utah School of Medicine, 15 North Medical Drive East, 2600 EEJMRB, Salt Lake City, UT 84112, USA
| | - Daniel J Doty
- Department of Pathology, University of Utah School of Medicine, 15 North Medical Drive East, 2600 EEJMRB, Salt Lake City, UT 84112, USA
| | - Jordan T Sim
- Department of Pathology, University of Utah School of Medicine, 15 North Medical Drive East, 2600 EEJMRB, Salt Lake City, UT 84112, USA
| | - Ana Beatriz DePaula-Silva
- Department of Pathology, University of Utah School of Medicine, 15 North Medical Drive East, 2600 EEJMRB, Salt Lake City, UT 84112, USA
| | - Robert S Fujinami
- Department of Pathology, University of Utah School of Medicine, 15 North Medical Drive East, 2600 EEJMRB, Salt Lake City, UT 84112, USA.
| |
Collapse
|
22
|
Bartolini L, Libbey JE, Ravizza T, Fujinami RS, Jacobson S, Gaillard WD. Viral Triggers and Inflammatory Mechanisms in Pediatric Epilepsy. Mol Neurobiol 2018; 56:1897-1907. [PMID: 29978423 DOI: 10.1007/s12035-018-1215-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 06/27/2018] [Indexed: 12/21/2022]
Abstract
Experimental and clinical findings suggest a crucial role for inflammation in the onset of pediatric seizures; this mechanism is not targeted by conventional antiepileptic drugs and may contribute to refractory epilepsy. Several triggers, including infection with neurotropic viruses such as human herpesvirus 6 (HHV-6), other herpesviruses, and picornaviruses, appear to induce activation of the innate and adaptive immune systems, which results in several neuroinflammatory responses, leading to enhanced neuronal excitability, and ultimately contributing to epileptogenesis. This review discusses the proposed mechanisms by which infection with herpesviruses, and particularly with HHV-6, and ensuing inflammation may lead to seizure generation, and later development of epilepsy. We also examine the evidence that links herpesvirus and picornavirus infections with acute seizures and chronic forms of epilepsy. Understanding the mechanisms by which specific viruses may trigger a cascade of alterations in the CNS ultimately leading to epilepsy appears critical for the development of therapeutic agents that may target the virus or inflammatory mechanisms early and prevent progression of epileptogenesis.
Collapse
Affiliation(s)
- Luca Bartolini
- Clinical Epilepsy Section, National Institute of Neurological Disorders and Stroke, NIH, Building 10, room 7-5680, 10 Center Drive, Bethesda, MD, 20814, USA. .,Division of Neuroimmunology and Neurovirology, National Institute of Neurological Disorders and Stroke, NIH, 10 Center Drive, Bethesda, MD, 20892, USA. .,Center for Neuroscience, Children's National Medical Center, George Washington University, 111 Michigan Ave NW, Washington, DC, 20010, USA.
| | - Jane E Libbey
- Department of Pathology, University of Utah School of Medicine, 15 North Medical Drive East, 2600 EEJMRB, Salt Lake City, UT, 84112, USA
| | - Teresa Ravizza
- Neuroscience Department, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Via La Masa 19, 20156, Milan, Italy
| | - Robert S Fujinami
- Department of Pathology, University of Utah School of Medicine, 15 North Medical Drive East, 2600 EEJMRB, Salt Lake City, UT, 84112, USA
| | - Steven Jacobson
- Division of Neuroimmunology and Neurovirology, National Institute of Neurological Disorders and Stroke, NIH, 10 Center Drive, Bethesda, MD, 20892, USA
| | - William D Gaillard
- Center for Neuroscience, Children's National Medical Center, George Washington University, 111 Michigan Ave NW, Washington, DC, 20010, USA
| |
Collapse
|