1
|
Harutyunyan L, Callaerts P, Vermeer S. PHARC syndrome: an overview. Orphanet J Rare Dis 2024; 19:416. [PMID: 39501272 PMCID: PMC11539745 DOI: 10.1186/s13023-024-03418-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Accepted: 10/13/2024] [Indexed: 11/08/2024] Open
Abstract
PHARC, polyneuropathy, hearing loss, cerebellar ataxia, retinitis pigmentosa and cataracts, or PHARC is a very rare progressive neurodegenerative autosomal recessive disease caused by biallelic mutations in the ABHD12 (a/b-hydrolase domain containing 12) gene, which encodes a lyso-phosphatidylserine (lyso-PS) lipase. The Orpha number for PHARC is ORPHA171848. The clinical picture of PHARC syndrome is very heterogeneous with a wide range of age at onset for each symptom, making a clinical diagnosis very challenging. Differential diagnoses of the disease include Refsum disease, Charcot-Marie-Tooth disease, and Usher syndrome. Many aspects of the disease, such as the biochemistry and pathophysiology, are still not fully understood. We generated a clinical overview of all PHARC patients, including their mutations, described in literature so far. Furthermore, we give an outline of the most recent developments in research on the pathophysiology of PHARC syndrome in an attempt to gain more insight into and increase awareness of the heterogeneity of the disease. We included 58 patients with PHARC from 37 different families with 27 known ABHD12 mutations. The age at onset (from early childhood to late thirties) and the severity of each feature of PHARC varied widely among patients. Demyelinating polyneuropathy was reported in 91% of the patients. In 86% of patients, hearing loss was present and 74% had cerebellar ataxia, the most variable symptom of PHARC. Retinitis pigmentosa and cataracts occurred in 82% and 86% of patients, respectively. Due to the rareness of the disease and the variable clinical phenotype, a diagnosis of PHARC is often delayed and mostly only made after an extensive genetic work-up. Therefore, we recommend adding the ABHD12 gene to diagnostic gene panels for polyneuropathy, cerebellar ataxia, hearing loss, retinal dystrophy, and cataracts. In addition, a full clinical work-up, neurological (with EMG and neuroimaging of the brain) and ophthalmological (with ERG) examination and audiological tests are indispensable to obtain a comprehensive overview of the clinical phenotype as some symptoms in PHARC may be very subtle and easily overlooked if not tested for. In conclusion, we strongly recommend that patients with (suspected) PHARC should be evaluated in a multidisciplinary setting involving ophthalmologists, audiologists, neurologists, and geneticists to ensure the best possible care. Furthermore, we discuss whether PHARC is a spectrum with various incomplete phenotypes even later in life, or whether it is a syndrome in which the clinical symptoms are variable in severity and age of onset.
Collapse
Affiliation(s)
- Lusine Harutyunyan
- Laboratory for Behavioral and Developmental Genetics, Department of Human Genetics, KU Leuven, Louvain, Belgium
- Centre of Human Genetics, University Hospitals Leuven, Herestraat 49, 3000, Louvain, Belgium
- Disability Studies, Family Medicine and Population Health, University Antwerp, Antwerp, Belgium
| | - Patrick Callaerts
- Laboratory for Behavioral and Developmental Genetics, Department of Human Genetics, KU Leuven, Louvain, Belgium
| | - Sascha Vermeer
- Centre of Human Genetics, University Hospitals Leuven, Herestraat 49, 3000, Louvain, Belgium.
| |
Collapse
|
2
|
Griesman T, McMillen CM, Negatu SG, Hulahan JJ, Whig K, Dohnalová L, Dittmar M, Thaiss CA, Jurado KA, Schultz DC, Hartman AL, Cherry S. The lipopeptide Pam3CSK4 inhibits Rift Valley fever virus infection and protects from encephalitis. PLoS Pathog 2024; 20:e1012343. [PMID: 38935789 PMCID: PMC11236204 DOI: 10.1371/journal.ppat.1012343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 07/10/2024] [Accepted: 06/12/2024] [Indexed: 06/29/2024] Open
Abstract
Rift Valley fever virus (RVFV) is an encephalitic bunyavirus that can infect neurons in the brain. There are no approved therapeutics that can protect from RVFV encephalitis. Innate immunity, the first line of defense against infection, canonically antagonizes viruses through interferon signaling. We found that interferons did not efficiently protect primary cortical neurons from RVFV, unlike other cell types. To identify alternative neuronal antiviral pathways, we screened innate immune ligands and discovered that the TLR2 ligand Pam3CSK4 inhibited RVFV infection, and other bunyaviruses. Mechanistically, we found that Pam3CSK4 blocks viral fusion, independent of TLR2. In a mouse model of RVFV encephalitis, Pam3CSK4 treatment protected animals from infection and mortality. Overall, Pam3CSK4 is a bunyavirus fusion inhibitor active in primary neurons and the brain, representing a new approach toward the development of treatments for encephalitic bunyavirus infections.
Collapse
Affiliation(s)
- Trevor Griesman
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia Pennsylvania, United States of America
| | - Cynthia M. McMillen
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Infectious Diseases and Microbiology, University of Pittsburgh School of Public Health, Pittsburgh, Pennsylvania, United States of America
| | - Seble Getenet Negatu
- Department of Microbiology, University of Pennsylvania, Philadelphia Pennsylvania, Unites States of America
| | - Jesse J. Hulahan
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia Pennsylvania, United States of America
| | - Kanupriya Whig
- High throughput screening core, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Lenka Dohnalová
- Department of Microbiology, University of Pennsylvania, Philadelphia Pennsylvania, Unites States of America
| | - Mark Dittmar
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia Pennsylvania, United States of America
| | - Christoph A. Thaiss
- Department of Microbiology, University of Pennsylvania, Philadelphia Pennsylvania, Unites States of America
| | - Kellie A. Jurado
- Department of Microbiology, University of Pennsylvania, Philadelphia Pennsylvania, Unites States of America
| | - David C. Schultz
- High throughput screening core, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Amy L. Hartman
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Infectious Diseases and Microbiology, University of Pittsburgh School of Public Health, Pittsburgh, Pennsylvania, United States of America
| | - Sara Cherry
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia Pennsylvania, United States of America
- High throughput screening core, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| |
Collapse
|
3
|
McGurran H, Kumbol V, Krüger C, Wallach T, Lehnardt S. miR-154-5p Is a Novel Endogenous Ligand for TLR7 Inducing Microglial Activation and Neuronal Injury. Cells 2024; 13:407. [PMID: 38474371 PMCID: PMC10930870 DOI: 10.3390/cells13050407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 02/20/2024] [Accepted: 02/22/2024] [Indexed: 03/14/2024] Open
Abstract
Toll-like receptors (TLRs) are a collection of pattern recognition sensors that form a first line of defence by detecting pathogen- or damage-associated molecular patterns and initiating an inflammatory response. TLR activation in microglia, the major immune cells in the brain, can trigger the release of inflammatory molecules, which may contribute to various CNS diseases including Alzheimer's disease. Recently, some microRNAs were shown to serve as signalling molecules for TLRs. Here, we present miR-154-5p as a novel TLR7 ligand. Exposing microglia to miR-154-5p results in cytokine release and alters expression of the TLR signalling pathway dependent on TLR7. Additionally, miR-154-5p causes neuronal injury in enriched cortical neuron cultures and additive toxicity in the presence of microglia. Finally, intrathecal injection of miR-154-5p into mice leads to neuronal injury and accumulation of microglia in the cerebral cortex dependent on TLR7 expression. In conclusion, this study establishes miR-154-5p as a direct activator of TLR7 that can cause neuroinflammation and neuronal injury, which may contribute to CNS disease.
Collapse
Affiliation(s)
- Hugo McGurran
- Charité—Universitätsmedizin Berlin, Einstein Center for Neurosciences Berlin, 10117 Berlin, Germany; (H.M.); (V.K.)
- Institute of Cell Biology and Neurobiology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, 10117 Berlin, Germany; (C.K.); (T.W.)
| | - Victor Kumbol
- Charité—Universitätsmedizin Berlin, Einstein Center for Neurosciences Berlin, 10117 Berlin, Germany; (H.M.); (V.K.)
- Institute of Cell Biology and Neurobiology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, 10117 Berlin, Germany; (C.K.); (T.W.)
| | - Christina Krüger
- Institute of Cell Biology and Neurobiology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, 10117 Berlin, Germany; (C.K.); (T.W.)
| | - Thomas Wallach
- Institute of Cell Biology and Neurobiology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, 10117 Berlin, Germany; (C.K.); (T.W.)
| | - Seija Lehnardt
- Institute of Cell Biology and Neurobiology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, 10117 Berlin, Germany; (C.K.); (T.W.)
- Department of Neurology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, 10117 Berlin, Germany
| |
Collapse
|
4
|
Zheng H, Zhang C, Zhang J, Duan L. "Sentinel or accomplice": gut microbiota and microglia crosstalk in disorders of gut-brain interaction. Protein Cell 2023; 14:726-742. [PMID: 37074139 PMCID: PMC10599645 DOI: 10.1093/procel/pwad020] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 04/06/2023] [Indexed: 04/20/2023] Open
Abstract
Abnormal brain-gut interaction is considered the core pathological mechanism behind the disorders of gut-brain interaction (DGBI), in which the intestinal microbiota plays an important role. Microglia are the "sentinels" of the central nervous system (CNS), which participate in tissue damage caused by traumatic brain injury, resist central infection and participate in neurogenesis, and are involved in the occurrence of various neurological diseases. With in-depth research on DGBI, we could find an interaction between the intestinal microbiota and microglia and that they are jointly involved in the occurrence of DGBI, especially in individuals with comorbidities of mental disorders, such as irritable bowel syndrome (IBS). This bidirectional regulation of microbiota and microglia provides a new direction for the treatment of DGBI. In this review, we focus on the role and underlying mechanism of the interaction between gut microbiota and microglia in DGBI, especially IBS, and the corresponding clinical application prospects and highlight its potential to treat DGBI in individuals with psychiatric comorbidities.
Collapse
Affiliation(s)
- Haonan Zheng
- Department of Gastroenterology, Peking University Third Hospital, Beijing 100191, China
- Beijing Key Laboratory for Helicobacter Pylori Infection and Upper Gastrointestinal Diseases, Beijing 100191, China
| | - Cunzheng Zhang
- Department of Gastroenterology, Peking University Third Hospital, Beijing 100191, China
- Beijing Key Laboratory for Helicobacter Pylori Infection and Upper Gastrointestinal Diseases, Beijing 100191, China
| | - Jindong Zhang
- Department of Gastroenterology, Peking University Third Hospital, Beijing 100191, China
- Beijing Key Laboratory for Helicobacter Pylori Infection and Upper Gastrointestinal Diseases, Beijing 100191, China
| | - Liping Duan
- Department of Gastroenterology, Peking University Third Hospital, Beijing 100191, China
- Beijing Key Laboratory for Helicobacter Pylori Infection and Upper Gastrointestinal Diseases, Beijing 100191, China
| |
Collapse
|
5
|
Jiang W, Cheng Y, Wang Y, Wu J, Rong Z, Sun L, Zhou Y, Zhang K. Involvement of Abnormal p-α-syn Accumulation and TLR2-Mediated Inflammation of Schwann Cells in Enteric Autonomic Nerve Dysfunction of Parkinson's Disease: an Animal Model Study. Mol Neurobiol 2023:10.1007/s12035-023-03345-4. [PMID: 37148524 DOI: 10.1007/s12035-023-03345-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 04/10/2023] [Indexed: 05/08/2023]
Abstract
The study was designed to investigate the pathogenesis of gastrointestinal (GI) impairment in Parkinson's disease (PD). We utilized 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP, 20 mg/kg) and probenecid (250 mg/kg) to prepare a PD mice model. MPTP modeling was first confirmed. GI motility was measured using stool collection test and enteric plexus loss was also detected. Intestinal phosphorylated α-synuclein (p-α-syn), inflammation, and S100 were assessed using western blotting. Association between Toll-like receptor 2(TLR2) and GI function was validated by Pearson's correlations. Immunofluorescence was applied to show co-localizations of intestinal p-α-syn, inflammation, and Schwann cells (SCs). CU-CPT22 (3 mg/kg, a TLR1/TLR2 inhibitor) was adopted then. Success in modeling, damaged GI neuron and function, and activated intestinal p-α-syn, inflammation, and SCs responses were observed in MPTP group, with TLR2 related to GI damage. Increased p-α-syn and inflammatory factors were shown in SCs of myenteron for MPTP mice. Recovered fecal water content and depression of inflammation, p-α-syn deposition, and SCs activity were noticed after TLR2 suppression. The study investigates a novel mechanism of PD GI autonomic dysfunction, demonstrating that p-α-syn accumulation and TLR2 signaling of SCs were involved in disrupted gut homeostasis and treatments targeting TLR2-mediated pathway might be a possible therapy for PD.
Collapse
Affiliation(s)
- Wenwen Jiang
- Department of Neurology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Yue Cheng
- Department of Neurology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Ye Wang
- Department of Neurology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Jing Wu
- Department of Neurology, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, 212000, China
| | - Zhe Rong
- Department of Neurology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Li Sun
- Department of Neurology, Suzhou Ninth People's Hospital, Suzhou, 215200, China
| | - Yan Zhou
- Department of Clinical Pharmacy, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201620, China.
| | - Kezhong Zhang
- Department of Neurology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.
| |
Collapse
|
6
|
Almutairi MM, Nadeem A, Ansari MA, Bakheet SA, Attia SM, Albekairi TH, Alhosaini K, Algahtani M, Alsaad AMS, Al-Mazroua HA, Ahmad SF. Lead (Pb) exposure exacerbates behavioral and immune abnormalities by upregulating Th17 and NF-κB-related signaling in BTBR T + Itpr3 tf/J autistic mouse model. Neurotoxicology 2022; 91:340-348. [PMID: 35760230 DOI: 10.1016/j.neuro.2022.06.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 06/22/2022] [Accepted: 06/22/2022] [Indexed: 11/16/2022]
Abstract
Autism spectrum disorder (ASD) is a highly prevalent neurodevelopmental disorder that are characterized by abnormal social interaction impairments in communication and repetitive and restricted activities or interests. Even though the exact etiology of ASD remains unknown. Lead (Pb) is a toxin known to harm many organs in the body, it is one of the most ubiquitous metal exposures which is associated with neurological deficits. Previous studies have shown that the exposure to Pb may play a role in ASD. BTBR T+ Itpr3tf/J (BTBR) mouse model is commonly used as a preclinical model for ASD. In this study, we investigated the effects of Pb exposure on sociability, self-grooming and marble burying behaviors tests in BTBR mice. We further examined the effects of Pb on IL-17A- RORγT-, STAT3-, NF-κB p65-, iNOS-, TLR-2- and TLR-4-producing CD45+ cells in spleen using flow cytometry. We also explored the effects of Pb on IL-17A, RORγT, STAT3, NF-κB p65, and TLR-2 mRNA expression in the brain tissue using RT-PCR analysis. Our results demonstrated that Pb exposure substantially increased repetitive behavior, marble burying and decrease social interactions in BTBR mice. In addition, in spleen cells, Pb exposure exaggerated CD45+IL-17A+, CD45+RORγT+, CD45+STAT3+, CD45+NF-κB p65+, CD45+iNOS+, CD45+TLR-2+ and CD45+TLR-4+ in BTBR mice. We also found that Pb significantly increased IL-17A, RORγT, STAT3, NF-κB p65, and TLR-2 mRNA in the brain tissue. Therefore, Pb exposure exacerbates behavioral and neuroimmune function in BTBR mice, suggesting a potentially strong role for Pb in ASD.
Collapse
Affiliation(s)
- Mashal M Almutairi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh-11451, Saudi Arabia
| | - Ahmed Nadeem
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh-11451, Saudi Arabia
| | - Mushtaq A Ansari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh-11451, Saudi Arabia
| | - Saleh A Bakheet
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh-11451, Saudi Arabia
| | - Sabry M Attia
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh-11451, Saudi Arabia
| | - Thamer H Albekairi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh-11451, Saudi Arabia
| | - Khaled Alhosaini
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh-11451, Saudi Arabia
| | - Mohammad Algahtani
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh-11451, Saudi Arabia
| | - Abdulaziz M S Alsaad
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh-11451, Saudi Arabia
| | - Haneen A Al-Mazroua
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh-11451, Saudi Arabia
| | - Sheikh F Ahmad
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh-11451, Saudi Arabia.
| |
Collapse
|
7
|
Phosphorylated α-synuclein aggregated in Schwann cells exacerbates peripheral neuroinflammation and nerve dysfunction in Parkinson's disease through TLR2/NF-κB pathway. Cell Death Discov 2021; 7:289. [PMID: 34642321 PMCID: PMC8511120 DOI: 10.1038/s41420-021-00676-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 09/11/2021] [Accepted: 09/24/2021] [Indexed: 12/12/2022] Open
Abstract
To investigate the mechanism of peripheral neuropathy in Parkinson's disease (PD), we prepared a PD mice model by long-term exposure of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) to mimic PD pathology in humans and the sciatic nerves were taken for further research. It turned out that phosphorylated α-synuclein (p-α-syn) was significantly deposited in Schwann cells (SCs) of sciatic nerves possibly contributing to degenerated myelin SCs and atrophied axons in MPTP group. Further analysis confirmed that toll-like receptors (TLRs) were implicated with PD peripheral neuropathy, in which TLR2 exhibits the predominant expression. Increased expression of inflammatory factors about TLR2/nuclear factor kappa-B (NF-κB) pathway was noted in MPTP group compared to saline group, with proteins on other pathways showing no changes. Moreover, MPTP-challenged mice exhibited worse motor ability and damaged nerve conduction, implicating that p-α-syn neurotoxicity might be relevant to impairments of motor and sensory nerves. After the treatment of CU-CPT22, a TLR2 antagonist, p-α-syn accumulation, motor and sensory function were ameliorated in CU-CPT22 combined with MPTP group. Thus, we demonstrated that pathological p-α-syn might combine TLR2 to affect SCs activation, inflammatory response as well as motor and sensory function through TLR2/nuclear factor kappa-B (NF-κB) signaling pathway. This study firstly demonstrates a novel mechanism of p-α-syn accumulated in SCs of peripheral nerves, which extends our understanding on SCs-mediated peripheral neuroinflammation related to TLR2/NF-κB signaling pathway and sheds light on potential new therapeutic avenues for PD.
Collapse
|
8
|
Klammer MG, Dzaye O, Wallach T, Krüger C, Gaessler D, Buonfiglioli A, Derkow K, Kettenmann H, Brinkmann MM, Lehnardt S. UNC93B1 Is Widely Expressed in the Murine CNS and Is Required for Neuroinflammation and Neuronal Injury Induced by MicroRNA let-7b. Front Immunol 2021; 12:715774. [PMID: 34589086 PMCID: PMC8475950 DOI: 10.3389/fimmu.2021.715774] [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: 05/28/2021] [Accepted: 08/19/2021] [Indexed: 12/12/2022] Open
Abstract
The chaperone protein Unc-93 homolog B1 (UNC93B1) regulates internalization, trafficking, and stabilization of nucleic acid-sensing Toll-like receptors (TLR) in peripheral immune cells. We sought to determine UNC93B1 expression and its functional relevance in inflammatory and injurious processes in the central nervous system (CNS). We found that UNC93B1 is expressed in various CNS cells including microglia, astrocytes, oligodendrocytes, and neurons, as assessed by PCR, immunocyto-/histochemistry, and flow cytometry. UNC93B1 expression in the murine brain increased during development. Exposure to the microRNA let-7b, a recently discovered endogenous TLR7 activator, but also to TLR3 and TLR4 agonists, led to increased UNC93B1 expression in microglia and neurons. Microglial activation by extracellular let-7b required functional UNC93B1, as assessed by TNF ELISA. Neuronal injury induced by extracellular let-7b was dependent on UNC93B1, as UNC93B1-deficient neurons were unaffected by the microRNA's neurotoxicity in vitro. Intrathecal application of let-7b triggered neurodegeneration in wild-type mice, whereas mice deficient for UNC93B1 were protected against injurious effects on neurons and axons. In summary, our data demonstrate broad UNC93B1 expression in the murine brain and establish this chaperone as a modulator of neuroinflammation and neuronal injury triggered by extracellular microRNA and subsequent induction of TLR signaling.
Collapse
Affiliation(s)
- Markus G Klammer
- Institute of Cell Biology and Neurobiology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Department of Neurology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Omar Dzaye
- Department of Radiology and Neuroradiology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Thomas Wallach
- Institute of Cell Biology and Neurobiology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Christina Krüger
- Institute of Cell Biology and Neurobiology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Dorothea Gaessler
- Institute of Cell Biology and Neurobiology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Alice Buonfiglioli
- Institute of Cell Biology and Neurobiology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Katja Derkow
- Institute of Cell Biology and Neurobiology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Helmut Kettenmann
- Cellular Neuroscience, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Melanie M Brinkmann
- Viral Immune Modulation Research Group, Helmholtz Centre for Infection Research, Braunschweig, Germany.,Institute of Genetics, Technische Universität Braunschweig, Braunschweig, Germany
| | - Seija Lehnardt
- Institute of Cell Biology and Neurobiology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Department of Neurology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| |
Collapse
|
9
|
Seele J, Ballüer M, Tauber SC, Bunkowski S, Schulz K, Stadelmann C, Beineke A, Pägelow D, Fulde M, Nau R. Neural Injury and Repair in a Novel Neonatal Mouse Model of Listeria Monocytogenes Meningoencephalitis. J Neuropathol Exp Neurol 2021; 80:861-867. [PMID: 34486672 DOI: 10.1093/jnen/nlab079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
To improve the therapy of neonatal central nervous system infections, well-characterized animal models are urgently needed. The present study analyzes neuropathological alterations with particular focus on neural injury and repair in brains of neonatal mice with Listeria monocytogenes (LM) meningitis/meningoencephalitis using a novel nasal infection model. The hippocampal formation and frontal cortex of 14 neonatal mice with LM meningitis/meningoencephalitis and 14 uninfected controls were analyzed by histology, immunohistochemistry, and in situ tailing for morphological alterations. In the dentate gyrus of the hippocampal formation of mice with LM meningitis/meningoencephalitis, an increased density of apoptotic neurons visualized by in situ tailing (p = 0.04) and in situ tailing plus immunohistochemistry for activated Caspase-3 (p < 0.0001) was found. A decreased density of dividing cells stained with an anti-PCNA-antibody (p < 0.0001) and less neurogenesis visualized by anti-calretinin (p < 0.0001) and anti-calbindin (p = 0.01) antibodies were detected compared to uninfected controls. The density of microglia was higher in LM meningitis (p < 0.0001), while the density of astrocytes remained unchanged. Infiltrating monocytes and neutrophilic granulocytes likely contributed to tissue damage. In conclusion, in the brains of LM-infected mice a strong immune response was observed which led to neuronal apoptosis and an impaired neural regeneration. This model appears very suitable to study therapies against long-term sequelae of neonatal LM meningitis.
Collapse
Affiliation(s)
- Jana Seele
- Department of Neuropathology, University Medical Center Göttingen, Georg-August-University Göttingen, Göttingen, Germany.,Department of Geriatrics, Evangelisches Krankenhaus Göttingen-Weende, Göttingen, Germany
| | - Melissa Ballüer
- Department of Neuropathology, University Medical Center Göttingen, Georg-August-University Göttingen, Göttingen, Germany.,Department of Geriatrics, Evangelisches Krankenhaus Göttingen-Weende, Göttingen, Germany
| | - Simone C Tauber
- Department of Neurology, RWTH University Hospital, Aachen, Germany
| | - Stephanie Bunkowski
- Department of Neuropathology, University Medical Center Göttingen, Georg-August-University Göttingen, Göttingen, Germany
| | - Katja Schulz
- Department of Neuropathology, University Medical Center Göttingen, Georg-August-University Göttingen, Göttingen, Germany
| | - Christine Stadelmann
- Department of Neuropathology, University Medical Center Göttingen, Georg-August-University Göttingen, Göttingen, Germany
| | - Andreas Beineke
- Department of Pathology, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Dennis Pägelow
- Institute of Microbiology and Epizootics, Centre of Infection Medicine, Freie Universität Berlin, Berlin, Germany
| | - Marcus Fulde
- Institute of Microbiology and Epizootics, Centre of Infection Medicine, Freie Universität Berlin, Berlin, Germany
| | - Roland Nau
- Department of Neuropathology, University Medical Center Göttingen, Georg-August-University Göttingen, Göttingen, Germany.,Department of Geriatrics, Evangelisches Krankenhaus Göttingen-Weende, Göttingen, Germany
| |
Collapse
|
10
|
Assis MA, Carranza PG, Ambrosio E. A "Drug-Dependent" Immune System Can Compromise Protection against Infection: The Relationships between Psychostimulants and HIV. Viruses 2021; 13:v13050722. [PMID: 33919273 PMCID: PMC8143316 DOI: 10.3390/v13050722] [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: 03/01/2021] [Revised: 04/14/2021] [Accepted: 04/19/2021] [Indexed: 01/31/2023] Open
Abstract
Psychostimulant use is a major comorbidity in people living with HIV, which was initially explained by them adopting risky behaviors that facilitate HIV transmission. However, the effects of drug use on the immune system might also influence this phenomenon. Psychostimulants act on peripheral immune cells even before they reach the central nervous system (CNS) and their effects on immunity are likely to influence HIV infection. Beyond their canonical activities, classic neurotransmitters and neuromodulators are expressed by peripheral immune cells (e.g., dopamine and enkephalins), which display immunomodulatory properties and could be influenced by psychostimulants. Immune receptors, like Toll-like receptors (TLRs) on microglia, are modulated by cocaine and amphetamine exposure. Since peripheral immunocytes also express TLRs, they may be similarly affected by psychostimulants. In this review, we will summarize how psychostimulants are currently thought to influence peripheral immunity, mainly focusing on catecholamines, enkephalins and TLR4, and shed light on how these drugs might affect HIV infection. We will try to shift from the classic CNS perspective and adopt a more holistic view, addressing the potential impact of psychostimulants on the peripheral immune system and how their systemic effects could influence HIV infection.
Collapse
Affiliation(s)
- María Amparo Assis
- Facultad de Ciencias Médicas, Universidad Nacional de Santiago del Estero (UNSE), Santiago del Estero G4200, Argentina;
- Laboratorio de Biología Molecular, Inmunología y Microbiología, Instituto Multidisciplinario de Salud, Tecnología y Desarrollo (IMSaTeD), CONICET-UNSE, Santiago del Estero G4206, Argentina
- Departamento de Psicobiología, Facultad de Psicología, Universidad Nacional de Educación a Distancia (UNED), 28040 Madrid, Spain;
- Correspondence:
| | - Pedro Gabriel Carranza
- Facultad de Ciencias Médicas, Universidad Nacional de Santiago del Estero (UNSE), Santiago del Estero G4200, Argentina;
- Laboratorio de Biología Molecular, Inmunología y Microbiología, Instituto Multidisciplinario de Salud, Tecnología y Desarrollo (IMSaTeD), CONICET-UNSE, Santiago del Estero G4206, Argentina
- Facultad de Agronomía y Agroindustrias, Universidad Nacional de Santiago del Estero, Santiago del Estero G4206, Argentina
| | - Emilio Ambrosio
- Departamento de Psicobiología, Facultad de Psicología, Universidad Nacional de Educación a Distancia (UNED), 28040 Madrid, Spain;
| |
Collapse
|
11
|
Inflammatory Response, a Key Pathophysiological Mechanism of Obesity-Induced Depression. Mediators Inflamm 2020; 2020:8893892. [PMID: 33299381 PMCID: PMC7707993 DOI: 10.1155/2020/8893892] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 11/17/2020] [Accepted: 11/18/2020] [Indexed: 12/12/2022] Open
Abstract
In recent years, with the acceleration of life rhythm and the increase of social competition, the incidence of obesity and depression has been increasing, which has seriously affected the quality of life and health of people. Obesity and depression, two seemingly unrelated physical and psychological diseases, in fact, are closely related: obese people are more likely to have depression than nonobese ones. We have reviewed and analyzed the relevant research literature and found that the inflammatory response plays a key role in obesity-induced depression. This article will discuss in detail the inflammatory mechanisms by which obesity induces depression.
Collapse
|
12
|
Ifuku M, Hinkelmann L, Kuhrt LD, Efe IE, Kumbol V, Buonfiglioli A, Krüger C, Jordan P, Fulde M, Noda M, Kettenmann H, Lehnardt S. Activation of Toll-like receptor 5 in microglia modulates their function and triggers neuronal injury. Acta Neuropathol Commun 2020; 8:159. [PMID: 32912327 PMCID: PMC7488138 DOI: 10.1186/s40478-020-01031-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 08/29/2020] [Indexed: 12/12/2022] Open
Abstract
Microglia are the primary immune-competent cells of the central nervous system (CNS) and sense both pathogen- and host-derived factors through several receptor systems including the Toll-like receptor (TLR) family. Although TLR5 has previously been implicated in different CNS disorders including neurodegenerative diseases, its mode of action in the brain remained largely unexplored. We sought to determine the expression and functional consequences of TLR5 activation in the CNS. Quantitative real-time PCR and immunocytochemical analysis revealed that microglia is the major CNS cell type that constitutively expresses TLR5. Using Tlr5−/− mice and inhibitory TLR5 antibody we found that activation of TLR5 in microglial cells by its agonist flagellin, a principal protein component of bacterial flagella, triggers their release of distinct inflammatory molecules, regulates chemotaxis, and increases their phagocytic activity. Furthermore, while TLR5 activation does not affect tumor growth in an ex vivo GL261 glioma mouse model, it triggers microglial accumulation and neuronal apoptosis in the cerebral cortex in vivo. TLR5-mediated microglial function involves the PI3K/Akt/mammalian target of rapamycin complex 1 (mTORC1) pathway, as specific inhibitors of this signaling pathway abolish microglial activation. Taken together, our findings establish TLR5 as a modulator of microglial function and indicate its contribution to inflammatory and injurious processes in the CNS.
Collapse
|
13
|
Kim M, Lee S, Lim H, Lee J, Park JY, Kwon HJ, Lee IC, Ryu YB, Kim J, Shin T, Ahn G, Rho MC, Jung K. Oleanolic Acid Acetate Alleviates Symptoms of Experimental Autoimmune Encephalomyelitis in Mice by Regulating Toll-Like Receptor 2 Signaling. Front Pharmacol 2020; 11:556391. [PMID: 33013394 PMCID: PMC7494849 DOI: 10.3389/fphar.2020.556391] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 08/18/2020] [Indexed: 02/06/2023] Open
Abstract
Toll-like receptor 2 (TLR2) is expressed by several immune cells in the central nervous system and plays an important role in neuroinflammation. TLR2 upregulation has been reported in multiple sclerosis patients and in experimental autoimmune encephalomyelitis (EAE), a mouse model for multiple sclerosis. Therefore, modulating TLR2 signaling can be an effective treatment strategy against MS. Oleanolic acid acetate (OAA) has antiinflammatory and immunomodulatory effects. Hence, this study aimed to examine the effects of OAA on TLR2 signaling and neuroinflammation in EAE. EAE was induced in C57/BL6 mice using synthesized myelin oligodendrocyte glycoprotein (MOG)35-55 peptide, and OAA was administered daily. Hind limb paralysis and inflammatory cell infiltration were observed in the spinal cords of EAE mice. Moreover, T-cell proliferation was significantly stimulated in splenic cells from EAE mice. The expression of proinflammatory cytokines in the spinal cord was upregulated, and their serum protein levels were increased in EAE mice. Furthermore, upregulation of TLR2 and downstream signaling molecules was observed in the spinal cord. These pathological changes were reversed by OAA treatment. Our results suggest that OAA might have promising therapeutic properties and that the TLR signaling pathway is an effective therapeutic target against multiple sclerosis.
Collapse
Affiliation(s)
- Minju Kim
- Department of Marine Bio-Food Sciences, Chonnam National University, Yeosu, South Korea
| | - Soyoung Lee
- Immunoregulatory Materials Research Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup-si, South Korea
| | - Hyungjin Lim
- Immunoregulatory Materials Research Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup-si, South Korea
| | - Jihye Lee
- Functional Biomaterials Research Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup-si, South Korea
| | - Ji-Young Park
- Functional Biomaterials Research Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup-si, South Korea
| | - Hyung-Jun Kwon
- Functional Biomaterials Research Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup-si, South Korea
| | - In-Chul Lee
- Functional Biomaterials Research Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup-si, South Korea
| | - Young-Bae Ryu
- Functional Biomaterials Research Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup-si, South Korea
| | - Jeongtae Kim
- Department of Anatomy, Kosin University College of Medicine, Busan, South Korea
| | - Taekyun Shin
- Department of Veterinary Anatomy, College of Veterinary Medicine and Veterinary Medical Research Institute, Jeju National University, Jeju, South Korea
| | - Ginnae Ahn
- Department of Marine Bio-Food Sciences, Chonnam National University, Yeosu, South Korea
| | - Mun-Chual Rho
- Immunoregulatory Materials Research Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup-si, South Korea
| | - Kyungsook Jung
- Functional Biomaterials Research Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup-si, South Korea
| |
Collapse
|
14
|
Rodríguez-Gómez JA, Kavanagh E, Engskog-Vlachos P, Engskog MK, Herrera AJ, Espinosa-Oliva AM, Joseph B, Hajji N, Venero JL, Burguillos MA. Microglia: Agents of the CNS Pro-Inflammatory Response. Cells 2020; 9:E1717. [PMID: 32709045 PMCID: PMC7407646 DOI: 10.3390/cells9071717] [Citation(s) in RCA: 222] [Impact Index Per Article: 44.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Revised: 07/14/2020] [Accepted: 07/15/2020] [Indexed: 12/21/2022] Open
Abstract
The pro-inflammatory immune response driven by microglia is a key contributor to the pathogenesis of several neurodegenerative diseases. Though the research of microglia spans over a century, the last two decades have increased our understanding exponentially. Here, we discuss the phenotypic transformation from homeostatic microglia towards reactive microglia, initiated by specific ligand binding to pattern recognition receptors including toll-like receptor-4 (TLR4) or triggering receptors expressed on myeloid cells-2 (TREM2), as well as pro-inflammatory signaling pathways triggered such as the caspase-mediated immune response. Additionally, new research disciplines such as epigenetics and immunometabolism have provided us with a more holistic view of how changes in DNA methylation, microRNAs, and the metabolome may influence the pro-inflammatory response. This review aimed to discuss our current knowledge of pro-inflammatory microglia from different angles, including recent research highlights such as the role of exosomes in spreading neuroinflammation and emerging techniques in microglia research including positron emission tomography (PET) scanning and the use of human microglia generated from induced pluripotent stem cells (iPSCs). Finally, we also discuss current thoughts on the impact of pro-inflammatory microglia in neurodegenerative diseases.
Collapse
Affiliation(s)
- José A. Rodríguez-Gómez
- Institute of Biomedicine of Seville (IBIS)-Hospital Universitario Virgen del Rocío/CSIC/University of Seville, 41012 Seville, Spain; (J.A.R.-G.); (A.J.H.); (A.M.E.-O.); (J.L.V.)
- Department of Medical Physiology and Biophysics, Faculty of Medicine, University of Seville, 41009 Sevilla, Spain
| | - Edel Kavanagh
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, University of Seville, 41012 Seville, Spain;
| | - Pinelopi Engskog-Vlachos
- Institute of Environmental Medicine, Toxicology Unit, Karolinska Institute, 17177 Stockholm, Sweden; (P.E.-V.); (B.J.)
| | - Mikael K.R. Engskog
- Department of Medicinal Chemistry, Analytical Pharmaceutical Chemistry, Uppsala University, 751 23 Uppsala, Sweden;
| | - Antonio J. Herrera
- Institute of Biomedicine of Seville (IBIS)-Hospital Universitario Virgen del Rocío/CSIC/University of Seville, 41012 Seville, Spain; (J.A.R.-G.); (A.J.H.); (A.M.E.-O.); (J.L.V.)
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, University of Seville, 41012 Seville, Spain;
| | - Ana M. Espinosa-Oliva
- Institute of Biomedicine of Seville (IBIS)-Hospital Universitario Virgen del Rocío/CSIC/University of Seville, 41012 Seville, Spain; (J.A.R.-G.); (A.J.H.); (A.M.E.-O.); (J.L.V.)
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, University of Seville, 41012 Seville, Spain;
| | - Bertrand Joseph
- Institute of Environmental Medicine, Toxicology Unit, Karolinska Institute, 17177 Stockholm, Sweden; (P.E.-V.); (B.J.)
| | - Nabil Hajji
- Division of Brain Sciences, The John Fulcher Molecular Neuro-Oncology Laboratory, Imperial College London, London W12 ONN, UK;
| | - José L. Venero
- Institute of Biomedicine of Seville (IBIS)-Hospital Universitario Virgen del Rocío/CSIC/University of Seville, 41012 Seville, Spain; (J.A.R.-G.); (A.J.H.); (A.M.E.-O.); (J.L.V.)
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, University of Seville, 41012 Seville, Spain;
| | - Miguel A. Burguillos
- Institute of Biomedicine of Seville (IBIS)-Hospital Universitario Virgen del Rocío/CSIC/University of Seville, 41012 Seville, Spain; (J.A.R.-G.); (A.J.H.); (A.M.E.-O.); (J.L.V.)
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, University of Seville, 41012 Seville, Spain;
| |
Collapse
|
15
|
Zhao S, Zhang Z, Xu D, Wang Y, Li L. Selective Loss of Brain-Derived Neurotrophic Factor Exacerbates Brain Injury by Enhancing Neuroinflammation in Experimental Streptococcus pneumoniae Meningitis. Front Immunol 2020; 11:1357. [PMID: 32676082 PMCID: PMC7333737 DOI: 10.3389/fimmu.2020.01357] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 05/27/2020] [Indexed: 11/13/2022] Open
Abstract
Streptococcus pneumoniae meningitis is a life-threatening bacterial infection of the central nervous system (CNS), and its unfavorable prognosis usually results from an intense inflammatory response. Recent studies have shown that brain-derived neurotrophic factor (BDNF) mediates anti-inflammatory and neuroprotective effects in CNS diseases; however, the distinct contribution of BDNF to pneumococcal meningitis (PM) remains unknown. In this study, we sought to investigate the effects of endogenous BDNF on the inflammatory response and brain damage in experimental PM. We used Camk2a-CreERT2 mice to delete Bdnf from the cerebral cortex and hippocampus, and meningitis was induced by intracisternal infection with S. pneumoniae. Clinical parameters were assessed during acute meningitis. At 24 h post-infection, histopathology, neutrophil granulocytes infiltration, and microglia/macrophage proliferation of brain tissues were evaluated. Additionally, cortical damage and hippocampal apoptosis were assessed using Nissl staining and terminal deoxynucleotidyl transferase dUTP-nick-end labeling (TUNEL), respectively. Pro-inflammatory cytokine levels were determined using real-time polymerase chain reaction (RT-PCR). Key molecules associated with the related signaling pathways were analyzed by RT-PCR and western blot. To investigate the role of microglia/macrophage in infected BDNF conditional knockout mice, GW2580 was used for microglia/macrophage depletion. Here, we, for the first time, found that BDNF conditional knockouts exhibited more profound clinical impairment, pathological severity, and neuron injury and enhanced microglia/macrophage proliferation than were observed in their littermate controls. Furthermore, the BDNF conditional knockouts showed an obviously increase in the expression of pro-inflammatory factors (Tnf-α, Il-1β, and Il-6). Mechanistically, loss of BDNF activated TLR2- and NOD2-mediated downstream nuclear factor kappa B (NF-κB) p65 and p38 mitogen-activated protein kinase (MAPK) pathways associated with S. pneumoniae infection. Furthermore, targeted depletion of microglia/macrophage population decreased the resistance of mice to PM with diminishing neuroinflammation in BDNF conditional knockouts. Our findings suggest that loss of BDNF may enhance the inflammatory response and contribute to brain injury during PM at least partially by modulating TLR2- and NOD2-mediated signaling pathways, thereby providing a potential therapeutic target for future interventions in bacterial meningitis pathologies.
Collapse
Affiliation(s)
- Shengnan Zhao
- Department of Pediatric Neurology, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Zhijie Zhang
- Department of Pediatric Neurology, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Danfeng Xu
- Department of Pediatric Neurology, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yanfei Wang
- Department of Pediatric Neurology, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Ling Li
- Department of Pediatric Neurology, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
16
|
Pek JH, Yap BJ, Gan MY, Seethor STT, Greenberg R, Hornik CPV, Tan B, Lee JH, Chong SL. Neurocognitive impairment after neonatal sepsis: protocol for a systematic review and meta-analysis. BMJ Open 2020; 10:e038816. [PMID: 32532785 PMCID: PMC7295426 DOI: 10.1136/bmjopen-2020-038816] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 05/11/2020] [Accepted: 05/18/2020] [Indexed: 12/13/2022] Open
Abstract
INTRODUCTION The effect of neonatal sepsis on the developing brain is not well documented. We aim to perform evidence synthesis to determine the outcome of neurodevelopmental impairment and intellectual disability among survivors of neonatal sepsis. The data gathered will inform on the long-term neurocognitive outcomes of neonates with sepsis and the measures used to document their developmental disability. METHODS AND ANALYSIS We will perform a search based on the following parameters: neonates and infants less than 90 days old diagnosed with sepsis who had neurocognitive outcomes or measures of developmental disability reported. We will search PubMed, Cochrane Central, Embase and Web of Science for articles in English language published between January 2010 and December 2019. Clinical trials and observational studies will be included. Two independent reviewers will screen studies for eligibility. Data extraction will then be performed using a standardised form. The quality of evidence and risk of bias will be assessed using Cochrane Collaboration's tool and Risk of Bias in Non-randomised Studies of Intervention (ROBINS-I). The results will be synthesised qualitatively and pooled for meta-analysis. ETHICS AND DISSEMINATION No formal ethical approval is required as there is no collection of primary data. This systematic review and meta-analysis will be disseminated through conference meetings and peer-reviewed publications. PROSPERO REGISTRATION NUMBER Registration submitted CRD42020164334.
Collapse
Affiliation(s)
- Jen Heng Pek
- Department of Emergency Medicine, Sengkang General Hospital, Singapore
| | - Bei Jun Yap
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Ming Ying Gan
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | | | - Rachel Greenberg
- Department of Pediatrics, Duke University School of Medicine, Durham, North Carolina, USA
| | - Christoph Paul Vincent Hornik
- Division of Quantitative Sciences, Department of Pediatrics, Duke University School of Medicine, Durham, North Carolina, USA
| | - Bobby Tan
- Department of Emergency Medicine, KK Women's and Children's Hospital, Singapore
| | - Jan Hau Lee
- Children's Intensive Care Unit, KK Women's and Children's Hospital, Duke-NUS Medical School, Singapore
| | - Shu-Ling Chong
- Department of Emergency Medicine, KK Women's and Children's Hospital, Singapore
| |
Collapse
|
17
|
Characterising lipoteichoic acid as an in vitro model of acute neuroinflammation. Int Immunopharmacol 2020; 85:106619. [PMID: 32485352 DOI: 10.1016/j.intimp.2020.106619] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 04/29/2020] [Accepted: 05/17/2020] [Indexed: 01/12/2023]
Abstract
Toll-like receptor 2 (TLR2) is a primary sensor for pathogens, including those derived from gram-positive bacteria. It can also mediate the effects of endogenous inflammatory signals such as β-amyloid peptide (Aβ), thus promoting the microglial activation and subsequent neuronal dysfunction, characteristic of chronic neuroinflammatory conditions. More recently, a role for TLR2 has been proposed in the pathogenesis of disorders associated with acute inflammation, including anxiety and depression. The current study aims to characterise the acute effects of the TLR2 agonist lipoteichoic acid (LTA) on microglial activation and neuronal integrity, and to evaluate the influence of LTA exposure on sensitivity to the inflammation and neuronal dysfunction associated with Aβ. Using BV2 and N2a cells as an in vitro model, we highlight that acute exposure to LTA robustly promotes inflammatory cytokine and nitric oxide (NO) production in microglia but also in neurons, similar to that reported under longer-term and chronic inflammatory conditions. Moreover, we find that exposure to LTA can enhance sensitivity to subthreshold Aβ, promoting an 'M1'-like phenotype in microglia and provoking dysregulation of neuronal activity in acute hippocampal slices. Anti-inflammatory agents, including mimetics of brain-derived neurotrophic factor (BDNF), have proven effective at alleviating chronic neuroinflammatory complications. We further examined the effects of 7,8,3-trihydroxyflavone (7,8,3-THF), a small-molecule TrkB agonist, on LTA-induced microglial activation. We report that 7,8,3-THF can significantly ameliorate interleukin (IL)-6 and NO production in LTA-stimulated BV2 cells. Taken together, our findings offer support for exploration of TLR2 as a potential target for therapeutic intervention into acute neuroinflammatory conditions. Moreover we propose that exposure to gram-positive bacterial pathogens may promote sensitivity to the inflammatory changes characteristic of the aged brain.
Collapse
|
18
|
Dembny P, Newman AG, Singh M, Hinz M, Szczepek M, Krüger C, Adalbert R, Dzaye O, Trimbuch T, Wallach T, Kleinau G, Derkow K, Richard BC, Schipke C, Scheidereit C, Stachelscheid H, Golenbock D, Peters O, Coleman M, Heppner FL, Scheerer P, Tarabykin V, Ruprecht K, Izsvák Z, Mayer J, Lehnardt S. Human endogenous retrovirus HERV-K(HML-2) RNA causes neurodegeneration through Toll-like receptors. JCI Insight 2020; 5:131093. [PMID: 32271161 DOI: 10.1172/jci.insight.131093] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 03/04/2020] [Indexed: 01/27/2023] Open
Abstract
Although human endogenous retroviruses (HERVs) represent a substantial proportion of the human genome and some HERVs, such as HERV-K(HML-2), are reported to be involved in neurological disorders, little is known about their biological function. We report that RNA from an HERV-K(HML-2) envelope gene region binds to and activates human Toll-like receptor (TLR) 8, as well as murine Tlr7, expressed in neurons and microglia, thereby causing neurodegeneration. HERV-K(HML-2) RNA introduced into the cerebrospinal fluid (CSF) of either C57BL/6 wild-type mice or APPPS1 mice, a mouse model for Alzheimer's disease (AD), resulted in neurodegeneration and microglia accumulation. Tlr7-deficient mice were protected against neurodegenerative effects but were resensitized toward HERV-K(HML-2) RNA when neurons ectopically expressed murine Tlr7 or human TLR8. Transcriptome data sets of human AD brain samples revealed a distinct correlation of upregulated HERV-K(HML-2) and TLR8 RNA expression. HERV-K(HML-2) RNA was detectable more frequently in CSF from individuals with AD compared with controls. Our data establish HERV-K(HML-2) RNA as an endogenous ligand for species-specific TLRs 7/8 and imply a functional contribution of human endogenous retroviral transcripts to neurodegenerative processes, such as AD.
Collapse
Affiliation(s)
- Paul Dembny
- Institute of Cell Biology and Neurobiology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Andrew G Newman
- Institute of Cell Biology and Neurobiology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Manvendra Singh
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Michael Hinz
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Michal Szczepek
- Institute for Medical Physics and Biophysics, Group Protein X-ray Crystallography and Signal Transduction, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and BIH, Berlin, Germany
| | - Christina Krüger
- Institute of Cell Biology and Neurobiology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | | | - Omar Dzaye
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany.,Department of Radiology.,Department of Neuroradiology
| | | | - Thomas Wallach
- Institute of Cell Biology and Neurobiology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Gunnar Kleinau
- Institute for Medical Physics and Biophysics, Group Protein X-ray Crystallography and Signal Transduction, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and BIH, Berlin, Germany
| | - Katja Derkow
- Institute of Cell Biology and Neurobiology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | | | - Carola Schipke
- Department of Psychiatry and Psychotherapy, and.,Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and BIH.,German Center for Neurodegenerative Diseases, Berlin, Germany
| | - Claus Scheidereit
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Harald Stachelscheid
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, BIH, Berlin-Brandenburg Center for Regenerative Therapies, Berlin, Germany
| | - Douglas Golenbock
- Division of Infectious Diseases and Immunology, Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Oliver Peters
- Department of Psychiatry and Psychotherapy, and.,Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and BIH.,German Center for Neurodegenerative Diseases, Berlin, Germany
| | - Michael Coleman
- Babraham Institute and John van Geest Centre for Brain Repair, University of Cambridge, Cambridge, United Kingdom
| | - Frank L Heppner
- NeuroCure Cluster of Excellence.,Department of Neuropathology.,German Center for Neurodegenerative Diseases, Berlin, Germany
| | - Patrick Scheerer
- Institute for Medical Physics and Biophysics, Group Protein X-ray Crystallography and Signal Transduction, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and BIH, Berlin, Germany.,German Centre for Cardiovascular Research, partner site Berlin, Berlin, Germany
| | - Victor Tarabykin
- Institute of Cell Biology and Neurobiology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Klemens Ruprecht
- Department of Neurology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and BIH, Berlin, Germany
| | - Zsuzsanna Izsvák
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Jens Mayer
- Institute of Human Genetics, Universität des Saarlandes, Hamburg, Germany
| | - Seija Lehnardt
- Institute of Cell Biology and Neurobiology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany.,Department of Neurology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and BIH, Berlin, Germany
| |
Collapse
|
19
|
Li W, Yu Y, Li D, Xu N, Fang J, Sun Y, Xu M, Wang X, Han X, Zhang X, Lv C, Han F. TLR2 deficiency attenuated chronic intermittent hypoxia-induced neurocognitive deficits. Int Immunopharmacol 2020; 81:106284. [PMID: 32058931 DOI: 10.1016/j.intimp.2020.106284] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 01/22/2020] [Accepted: 02/02/2020] [Indexed: 12/17/2022]
Abstract
Chronic intermittent hypoxia (CIH) is the main symptom of obstructive sleep apnea syndrome (OSAS) and causes neural damage and cognitive deficits via neuroinflammation. Toll-like receptors (TLRs), especially TLR2, play an important role in neuroinflammation. However, the mechanisms by which TLR2 participates in CIH-induced cognitive deficits remain unclear. In this study, wild-type (WT) and TLR2 knock out (KO) mice were exposed to CIH for 8 weeks, and their social novelty discrimination, spatial learning and memory were severely compromised. Additionally, seriously damaged neurons and abnormally activated glia were observed in the CA1 and dentate gyrus (DG) areas of the hippocampus. Mechanistically, knocking out the TLR2 gene significantly alleviated these pathological changes and improved the behavioral performance. Together, these findings demonstrate that the TLR2-MyD88 signaling pathway might play an important role in CIH-induced cognitive deficits.
Collapse
Affiliation(s)
- Wanting Li
- Binzhou Medical University, 346 Guanhai Road, YanTai 264003, China
| | - Yan Yu
- Binzhou Medical University, 346 Guanhai Road, YanTai 264003, China.
| | - Dongze Li
- Binzhou Medical University, 346 Guanhai Road, YanTai 264003, China
| | - Na Xu
- Binzhou Medical University, 346 Guanhai Road, YanTai 264003, China
| | - Jidong Fang
- Binzhou Medical University, 346 Guanhai Road, YanTai 264003, China
| | - Yeying Sun
- Binzhou Medical University, 346 Guanhai Road, YanTai 264003, China
| | - Maolei Xu
- Binzhou Medical University, 346 Guanhai Road, YanTai 264003, China
| | - Xi Wang
- Yantai Affiliated Hospital of Binzhou Medical University, 717 Jinbu Street, YanTai 264199, China
| | - Xin Han
- Binzhou Medical University, 346 Guanhai Road, YanTai 264003, China
| | - Xiuli Zhang
- Binzhou Medical University, 346 Guanhai Road, YanTai 264003, China
| | - Changjun Lv
- Binzhou Medical University, 346 Guanhai Road, YanTai 264003, China.
| | - Fang Han
- Binzhou Medical University, 346 Guanhai Road, YanTai 264003, China.
| |
Collapse
|
20
|
Kietzman C, Tuomanen E. Acute Bacterial Meningitis: Challenges to Better Antibiotic Therapy. ACS Infect Dis 2019; 5:1987-1995. [PMID: 31268283 DOI: 10.1021/acsinfecdis.9b00122] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Bacterial meningitis is a medical emergency requiring highly bactericidal antibiotics to achieve cure. Many challenges exist to achieving optimal patient outcome. First, antibiotics must pass the blood brain barrier. Once in the subarachnoid space, achieving bactericidal therapy involves circumventing antibiotic resistance and, more commonly, antibiotic tolerance arising from the slow growth of bacteria in the nutrient poor cerebrospinal fluid. Finally, bactericidal therapy is most often bacteriolytic, and debris from lysis is highly inflammatory. Controlling damage from lytic products may require adjunctive therapy to prevent neuronal death. These challenges are an extreme example of the different requirements for treating infections in different body sites.
Collapse
Affiliation(s)
- Colin Kietzman
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, Tennessee 38105, United States
| | - Elaine Tuomanen
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, Tennessee 38105, United States
| |
Collapse
|
21
|
Hu W, Yang S, Shimada Y, Münch M, Marín-Juez R, Meijer AH, Spaink HP. Infection and RNA-seq analysis of a zebrafish tlr2 mutant shows a broad function of this toll-like receptor in transcriptional and metabolic control and defense to Mycobacterium marinum infection. BMC Genomics 2019; 20:878. [PMID: 31747871 PMCID: PMC6869251 DOI: 10.1186/s12864-019-6265-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 11/05/2019] [Indexed: 02/07/2023] Open
Abstract
Background The function of Toll-like receptor 2 (TLR2) in host defense against pathogens, especially Mycobacterium tuberculosis (Mtb) is poorly understood. To investigate the role of TLR2 during mycobacterial infection, we analyzed the response of tlr2 zebrafish mutant larvae to infection with Mycobacterium marinum (Mm), a close relative to Mtb, as a model for tuberculosis. We measured infection phenotypes and transcriptome responses using RNA deep sequencing in mutant and control larvae. Results tlr2 mutant embryos at 2 dpf do not show differences in numbers of macrophages and neutrophils compared to control embryos. However, we found substantial changes in gene expression in these mutants, particularly in metabolic pathways, when compared with the heterozygote tlr2+/− control. At 4 days after Mm infection, the total bacterial burden and the presence of extracellular bacteria were higher in tlr2−/− larvae than in tlr2+/−, or tlr2+/+ larvae, whereas granuloma numbers were reduced, showing a function of Tlr2 in zebrafish host defense. RNAseq analysis of infected tlr2−/− versus tlr2+/− shows that the number of up-regulated and down-regulated genes in response to infection was greatly diminished in tlr2 mutants by at least 2 fold and 10 fold, respectively. Analysis of the transcriptome data and qPCR validation shows that Mm infection of tlr2 mutants leads to decreased mRNA levels of genes involved in inflammation and immune responses, including il1b, tnfb, cxcl11aa/ac, fosl1a, and cebpb. Furthermore, RNAseq analyses revealed that the expression of genes for Maf family transcription factors, vitamin D receptors, and Dicps proteins is altered in tlr2 mutants with or without infection. In addition, the data indicate a function of Tlr2 in the control of induction of cytokines and chemokines, such as the CXCR3-CXCL11 signaling axis. Conclusion The transcriptome and infection burden analyses show a function of Tlr2 as a protective factor against mycobacteria. Transcriptome analysis revealed tlr2-specific pathways involved in Mm infection, which are related to responses to Mtb infection in human macrophages. Considering its dominant function in control of transcriptional processes that govern defense responses and metabolism, the TLR2 protein can be expected to be also of importance for other infectious diseases and interactions with the microbiome.
Collapse
Affiliation(s)
- Wanbin Hu
- Institute of Biology, Leiden University, P.O. Box 9505, 2300 RA, Leiden, the Netherlands
| | - Shuxin Yang
- Institute of Biology, Leiden University, P.O. Box 9505, 2300 RA, Leiden, the Netherlands.,Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Yasuhito Shimada
- Institute of Biology, Leiden University, P.O. Box 9505, 2300 RA, Leiden, the Netherlands.,Department of Integrative Pharmacology, Mie University Graduate School of Medicine, Tsu, Japan
| | - Magnus Münch
- Mathematical Institute, Leiden University, Leiden, the Netherlands.,Department of Epidemiology & Biostatistics, Amsterdam Public Health Research Institute, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Rubén Marín-Juez
- Institute of Biology, Leiden University, P.O. Box 9505, 2300 RA, Leiden, the Netherlands.,Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Ludwigstrasse 43, 61231, Bad Nauheim, Germany
| | - Annemarie H Meijer
- Institute of Biology, Leiden University, P.O. Box 9505, 2300 RA, Leiden, the Netherlands
| | - Herman P Spaink
- Institute of Biology, Leiden University, P.O. Box 9505, 2300 RA, Leiden, the Netherlands.
| |
Collapse
|
22
|
Baranger K, van Gijsel-Bonnello M, Stephan D, Carpentier W, Rivera S, Khrestchatisky M, Gharib B, De Reggi M, Benech P. Long-Term Pantethine Treatment Counteracts Pathologic Gene Dysregulation and Decreases Alzheimer's Disease Pathogenesis in a Transgenic Mouse Model. Neurotherapeutics 2019; 16:1237-1254. [PMID: 31267473 PMCID: PMC6985318 DOI: 10.1007/s13311-019-00754-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Abstract
The low-molecular weight thiol pantethine, known as a hypolipidemic and hypocholesterolemic agent, is the major precursor of co-enzyme A. We have previously shown that pantethine treatment reduces amyloid-β (Aβ)-induced IL-1β release and alleviates pathological metabolic changes in primary astrocyte cultures. These properties of pantethine prompted us to investigate its potential benefits in vivo in the 5XFAD (Tg) mouse model of Alzheimer's disease (AD).1.5-month-old Tg and wild-type (WT) male mice were submitted to intraperitoneal administration of pantethine or saline control solution for 5.5 months. The effects of such treatments were investigated by performing behavioral tests and evaluating astrogliosis, microgliosis, Αβ deposition, and whole genome expression arrays, using RNAs extracted from the mice hippocampi. We observed that long-term pantethine treatment significantly reduced glial reactivity and Αβ deposition, and abrogated behavioral alteration in Tg mice. Moreover, the transcriptomic profiles revealed that after pantethine treatment, the expression of genes differentially expressed in Tg mice, and in particular those known to be related to AD, were significantly alleviated. Most of the genes overexpressed in Tg compared to WT were involved in inflammation, complement activation, and phagocytosis and were found repressed upon pantethine treatment. In contrast, pantethine restored the expression of a significant number of genes involved in the regulation of Αβ processing and synaptic activities, which were downregulated in Tg mice. Altogether, our data support a beneficial role for long-term pantethine treatment in preserving CNS crucial functions altered by Aβ pathogenesis in Tg mice and highlight the potential efficiency of pantethine to alleviate AD pathology.
Collapse
Affiliation(s)
- Kevin Baranger
- CNRS, INP, Inst Neurophysiopathol, Aix-Marseille Univ, Marseille, France
| | - Manuel van Gijsel-Bonnello
- CNRS, INP, Inst Neurophysiopathol, Aix-Marseille Univ, Marseille, France
- Present Address: MRC Protein Phosphorylation & Ubiquitylation Unit, Sir James Black Centre and School of Life Science - Division of Cell Signalling and Immunology, Welcome Trust Building, University of Dundee, Dundee, DD1 5EH UK
| | - Delphine Stephan
- CNRS, INP, Inst Neurophysiopathol, Aix-Marseille Univ, Marseille, France
| | - Wassila Carpentier
- Sorbonne Universités, UPMC Univ Paris 06, Inserm, UMS Omique, Plateforme Post-génomique de la Pitié-Salpêtrière (P3S), F-75013 Paris, France
| | - Santiago Rivera
- CNRS, INP, Inst Neurophysiopathol, Aix-Marseille Univ, Marseille, France
| | | | - Bouchra Gharib
- CNRS, INP, Inst Neurophysiopathol, Aix-Marseille Univ, Marseille, France
| | - Max De Reggi
- CNRS, INP, Inst Neurophysiopathol, Aix-Marseille Univ, Marseille, France
| | - Philippe Benech
- CNRS, INP, Inst Neurophysiopathol, Aix-Marseille Univ, Marseille, France
| |
Collapse
|
23
|
Myeloid-Related Protein 8/14 Participates in the Progression of Experimental Pneumococcal Meningitis by Augmentation of Inflammation. J Mol Neurosci 2019; 68:631-639. [PMID: 31049785 DOI: 10.1007/s12031-019-01314-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Accepted: 03/25/2019] [Indexed: 10/26/2022]
Abstract
It has been reported that myeloid-related protein 8/14 (MRP8/14) participates in the progression of inflammation after release from neutrophils and monocytes. This study aimed to clarify the mechanism(s) of the MRP8/14-augmented inflammatory response in mice with pneumococcal meningitis. Streptococcus pneumoniae (SP) meningitis was established by intracerebral injection of SP suspension. Balb/c mice were randomly divided into four groups and received the following injections: phosphate-buffer saline (PBS), MRP8/14 alone, SP alone, and SP plus MRP8/14. At 6 h, 24 h and 48 h postinfection, the clinical disease status was measured by the modified neurological severity score test, body weight loss and degree of cerebral edema; mice were anaesthetized, blood samples and brain samples were collected and brain inflammation was detected by haematoxylin and eosin (HE) staining; tumour necrosis factor-alpha (TNF-α), interleukin-6 (IL-6), C-reactive protein (CRP) and monocyte chemoattractant protein-1 (MCP-1) levels in serum and brain homogenates were assessed by an enzyme-linked immunosorbent assay (ELISA), and the mRNA levels of the above cytokines in brain homogenates were measured by polymerase chain reaction (PCR); and the expression of nuclear factor-kappa B (NF-κB) p65 in brain tissues was determined by immunohistochemical assay. In this study, we identified that MRP8/14 substantially augmented the SP-stimulated inflammatory response, aggravated clinical disease status and exacerbated SP-induced brain edema in a murine model of pneumococcal meningitis. Exogenous administration of MRP8/14 significantly enhanced mRNA and protein expression of the proinflammatory cytokines and chemokines TNF-α, CRP, IL-6 and MCP-1 in brain homogenates and serum from mice with pneumococcal meningitis, which may be related to the NF-κB signalling pathway. We further found that MRP8/14 strongly augmented SP-induced phosphorylation of NF-κB p65 in brain tissue slices from the same model. In conclusion, our results indicated that MRP8/14 augmented the inflammatory response in mice with pneumococcal meningitis and contributed to the development of disease, which was probably through NF-κB signalling pathway activation.
Collapse
|
24
|
Williams PhD MV, Cox B, Lafuse PhD WP, Ariza ME. Epstein-Barr Virus dUTPase Induces Neuroinflammatory Mediators: Implications for Myalgic Encephalomyelitis/Chronic Fatigue Syndrome. Clin Ther 2019; 41:848-863. [PMID: 31040055 PMCID: PMC6525645 DOI: 10.1016/j.clinthera.2019.04.009] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 04/01/2019] [Accepted: 04/01/2019] [Indexed: 01/04/2023]
Abstract
PURPOSE Neuroinflammation is a common feature in myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS), affecting 85%-90% of all patients, yet the underlying mechanism or mechanisms responsible for the initiation and/or promotion of this process is largely unknown. Multiple reports, however, have suggested a role for Epstein-Barr virus (EBV), in particular, in ME/CFS, but its potential role, if any, in the neuroinflammatory process has not been addressed. In support of this premise, studies by our group have found that the EBV protein deoxyuridine triphosphate nucleotidohydrolase (dUTPase) induces anxiety and sickness behaviors in female mice. We also found that a small subset of patients with ME/CFS exhibited prolonged and significantly elevated neutralizing antibodies against EBV dUTPase protein in serum, which inversely correlated with ME/CFS symptoms. A larger ME/CFS case-control cohort study further confirmed that a significant percentage of patients with ME/CFS (30.91%-52.7%) were simultaneously producing antibodies against multiple human herpesviruses-encoded dUTPases and/or human dUTPase. Altogether, these findings suggest that EBV dUTPase protein may be involved in the neuroinflammatory process observed in ME/CFS. Thus, the aim of the present study was to determine whether the EBV dUTPase protein could contribute to neuroinflammation by altering the expression of genes involved with maintaining blood-brain barrier (BBB) integrity and/or modulating synaptic plasticity. METHODS With the use of human immortalized astrocytes, microglia, and cerebral microvascular endothelial cells, we conducted time-course (0-24 h) experiments with EBV dUTPase protein (10 μg/mL) to determine what effect(s) it may have on the expression of genes involved with BBB permeability, astrocytes and microglia cell function, tryptophan metabolism, and synaptic plasticity by quantitative reverse transcription polymerase chain reaction (qRT-PCR). In parallel, in vivo studies were conducted in female C57Bl/6 mice. Mice were injected by the intraperitoneal route with EBV dUTPase protein (10 μg) or vehicle daily for 5 days, and the brains were collected and processed for further qRT-PCR analysis of the in vivo effect of the dUTPase on the dopamine/serotonin and γ-aminobutyric acid/glutamate pathways, which are important for brain function, using RT2 Profiler PCR Arrays. FINDINGS EBV dUTPase protein altered the expression in vitro (12 of 15 genes and 32 of 1000 proteins examined) and in vivo (34 of 84 genes examined) of targets with central roles in BBB integrity/function, fatigue, pain synapse structure, and function, as well as tryptophan, dopamine, and serotonin metabolism. IMPLICATIONS The data suggest that in a subset of patients with ME/CFS, the EBV dUTPase could initiate a neuroinflammatory reaction, which contributes to the fatigue, excessive pain, and cognitive impairments observed in these patients.
Collapse
Affiliation(s)
- Marshall V Williams PhD
- Department of Cancer Biology and Genetics, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Institute for Behavioral Medicine Research, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Brandon Cox
- Department of Cancer Biology and Genetics, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - William P Lafuse PhD
- Department of Microbial Infection and Immunity, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Maria Eugenia Ariza
- Department of Cancer Biology and Genetics, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Institute for Behavioral Medicine Research, The Ohio State University Wexner Medical Center, Columbus, OH, USA.
| |
Collapse
|
25
|
Gorup D, Škokić S, Kriz J, Gajović S. Tlr2 Deficiency is Associated with Enhanced Elements of Neuronal Repair and Caspase 3 Activation Following Brain Ischemia. Sci Rep 2019; 9:2821. [PMID: 30808918 PMCID: PMC6391535 DOI: 10.1038/s41598-019-39541-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 01/11/2019] [Indexed: 01/08/2023] Open
Abstract
The aim of this study was to apply multimodal in vivo imaging to assess the influence of altered innate immunity on brain repair after ischemic lesion. Tlr2-deficient mice were compared to wild type controls, as they lack Tlr2-mediated pro-inflammatory signaling triggered by postischemic necrosis. The ischemic lesion was induced by transient middle cerebral artery occlusion for 60 min, followed by brain imaging and analysis at four time points until 28 days after ischemia. Multimodal in vivo imaging involved a combination of 3 modalities: (1) magnetic resonance imaging by T2-weighted scans to assess brain lesion size, (2) bioluminescence imaging of Gap43-luc/gfp transgenic mice to visualize the axonal remodeling, and (3) caged-luciferin bioluminescence imaging of DEVD-luciferin allowing for visualization of caspase 3 and 7 activity in Gap43-luc/gfp mice. This enabled innovative correlation of the MRI-determined lesion size to photon fluxes obtained by bioluminescence imaging. Our data revealed that following ischemia, Tlr2-deficient mice had higher Gap43 expression and higher levels of caspases 3 and 7 activity, which was accompanied by enhanced levels of synaptic plasticity markers DLG4 and synaptophysin when compared to wild type controls. Altered inflammation in Tlr2-deficient mice was accompanied by enhanced elements of post-stroke repair, in particular during the chronic phase of recovery, but also with delayed final consolidation of the brain lesion.
Collapse
Affiliation(s)
- Dunja Gorup
- Croatian Institute for Brain Research, University of Zagreb School of Medicine, Šalata 12, Zagreb, HR-10000, Croatia
| | - Siniša Škokić
- Croatian Institute for Brain Research, University of Zagreb School of Medicine, Šalata 12, Zagreb, HR-10000, Croatia
| | - Jasna Kriz
- Department of Psychiatry and Neuroscience, Faculty of Medicine Laval University, CERVO Brain Research Center, 2601, de la Canardière, Québec (QC), G1J 2G3, Canada
| | - Srećko Gajović
- Croatian Institute for Brain Research, University of Zagreb School of Medicine, Šalata 12, Zagreb, HR-10000, Croatia.
| |
Collapse
|
26
|
Murugan S, Jakka P, Namani S, Mujumdar V, Radhakrishnan G. The neurosteroid pregnenolone promotes degradation of key proteins in the innate immune signaling to suppress inflammation. J Biol Chem 2019; 294:4596-4607. [PMID: 30647133 DOI: 10.1074/jbc.ra118.005543] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 01/07/2019] [Indexed: 11/06/2022] Open
Abstract
Pregnenolone is a steroid hormone precursor that is synthesized in various steroidogenic tissues, in the brain, and in lymphocytes. In addition to serving as the precursor for other steroid hormones, pregnenolone exerts its own effect as an anti-inflammatory molecule to maintain immune homeostasis in various inflammatory conditions. Pregnenolone and its metabolic derivatives have been shown to have beneficial effects in the brain, including enhancing memory and learning, reversing depressive disorders, and modulating cognitive functions. A decreased level of pregnenolone has been observed in neuroinflammatory diseases, which emphasizes its role in neuroprotection and neuroregeneration. Although the anti-inflammatory property of pregnenolone was recognized several decades ago, its mechanism of action remains unknown. Here we report that pregnenolone promotes ubiquitination and degradation of the TLR2/4 adaptor protein TIRAP and TLR2 in macrophages and microglial cells. Pregnenolone and its metabolites suppressed the secretion of tumor necrosis factor α and interleukin-6 mediated through TLR2 and TLR4 signaling. Pregnenolone has been reported to induce activation of cytoplasmic linker protein 170, and this protein has recently been shown to promote targeted degradation of TIRAP. We observed enhanced degradation of TIRAP and TLR4 suppression by cytoplasmic linker protein 170 in the presence of pregnenolone. Our experimental data reveal novel nongenomic targets of pregnenolone and provide important leads to understand its role in restoring immune homeostasis in various inflammatory conditions.
Collapse
Affiliation(s)
- Subathra Murugan
- From the Laboratory of Immunology and Microbial Pathogenesis, National Institute of Animal Biotechnology, Hyderabad, Telangana 500032, India and
| | - Padmaja Jakka
- From the Laboratory of Immunology and Microbial Pathogenesis, National Institute of Animal Biotechnology, Hyderabad, Telangana 500032, India and.,Graduate Studies, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Swapna Namani
- From the Laboratory of Immunology and Microbial Pathogenesis, National Institute of Animal Biotechnology, Hyderabad, Telangana 500032, India and.,Graduate Studies, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Varadendra Mujumdar
- From the Laboratory of Immunology and Microbial Pathogenesis, National Institute of Animal Biotechnology, Hyderabad, Telangana 500032, India and
| | - Girish Radhakrishnan
- From the Laboratory of Immunology and Microbial Pathogenesis, National Institute of Animal Biotechnology, Hyderabad, Telangana 500032, India and
| |
Collapse
|
27
|
Ogasawara D, Ichu TA, Vartabedian VF, Benthuysen J, Jing H, Reed A, Ulanovskaya OA, Hulce JJ, Roberts A, Brown S, Rosen H, Teijaro JR, Cravatt BF. Selective blockade of the lyso-PS lipase ABHD12 stimulates immune responses in vivo. Nat Chem Biol 2018; 14:1099-1108. [PMID: 30420694 PMCID: PMC6263940 DOI: 10.1038/s41589-018-0155-8] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 09/19/2018] [Indexed: 11/09/2022]
Abstract
ABHD12 metabolizes bioactive lysophospholipids, including lysophosphatidylserine (lyso-PS). Deleterious mutations in human ABHD12 cause the neurological disease PHARC, and ABHD12-/- mice display PHARC-like phenotypes, including hearing loss, along with elevated brain lyso-PS and features of stimulated innate immune cell function. Here, we develop a selective and in vivo-active inhibitor of ABHD12 termed DO264 and show that this compound elevates lyso-PS in mouse brain and primary human macrophages. Unlike ABHD12-/- mice, adult mice treated with DO264 exhibited minimal perturbations in auditory function. On the other hand, both DO264-treated and ABHD12-/- mice displayed heightened immunological responses to lymphocytic choriomeningitis virus (LCMV) clone 13 infection that manifested as severe lung pathology with elevated proinflammatory chemokines. These results reveal similarities and differences in the phenotypic impact of pharmacological versus genetic blockade of ABHD12 and point to a key role for this enzyme in regulating immunostimulatory lipid pathways in vivo.
Collapse
Affiliation(s)
- Daisuke Ogasawara
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, USA
| | - Taka-Aki Ichu
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, USA
| | - Vincent F Vartabedian
- Department of Immunology and Infectious Disease, The Scripps Research Institute, La Jolla, CA, USA
| | | | - Hui Jing
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, USA
| | - Alex Reed
- Abide Therapeutics, San Diego, CA, USA
| | | | - Jonathan J Hulce
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, USA
| | - Amanda Roberts
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA, USA
| | - Steven Brown
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, USA
| | - Hugh Rosen
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, USA
| | - John R Teijaro
- Department of Immunology and Infectious Disease, The Scripps Research Institute, La Jolla, CA, USA.
| | - Benjamin F Cravatt
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, USA.
| |
Collapse
|
28
|
Seele J, Tauber SC, Bunkowski S, Baums CG, Valentin-Weigand P, de Buhr N, Beineke A, Iliev AI, Brück W, Nau R. The inflammatory response and neuronal injury in Streptococcus suis meningitis. BMC Infect Dis 2018; 18:297. [PMID: 29970011 PMCID: PMC6029386 DOI: 10.1186/s12879-018-3206-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 06/22/2018] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Many of the currently used models of bacterial meningitis have limitations due to direct inoculation of pathogens into the cerebrospinal fluid or brain and a relatively insensitive assessment of long-term sequelae. The present study evaluates the utility of a Streptococcus (S.) suis intranasal infection model for the investigation of experimental therapies in meningitis. METHODS We examined the brains of 10 piglets with S. suis meningitis as well as 14 control piglets by histology, immunohistochemistry and in-situ tailing for morphological alterations in the hippocampal dentate gyrus and microglial activation in the neocortex. RESULTS In piglets with meningitis, the density of apoptotic neurons was significantly higher than in control piglets. Moreover, scoring of microglial morphology revealed a significant activation of these cells during meningitis. The slight increase in the density of dividing cells, young neurons and microglia observed in piglets suffering from meningitis was not statistically significant, probably because of the short time frame between onset of clinical signs and organ sampling. CONCLUSIONS The morphological changes found during S. suis meningitis are in accordance with abnormalities in other animal models and human autopsy cases. Therefore, the pig should be considered as a model for evaluating effects of experimental therapeutic approaches on neurological function in bacterial meningitis.
Collapse
Affiliation(s)
- Jana Seele
- Department of Neuropathology, University Medical Center Göttingen, Georg-August-University Göttingen, Göttingen, Germany. .,Department of Geriatrics, Evangelisches Krankenhaus Göttingen-Weende, Göttingen, Germany.
| | - Simone C Tauber
- Department of Neurology, RWTH University Hospital, Aachen, Germany
| | - Stephanie Bunkowski
- Department of Neuropathology, University Medical Center Göttingen, Georg-August-University Göttingen, Göttingen, Germany
| | - Christoph G Baums
- Institute for Bacteriology and Mycology, Center for Infectious Diseases, Faculty of Veterinary Medicine, University Leipzig, Leipzig, Germany
| | - Peter Valentin-Weigand
- Institute for Microbiology, Center for Infection Medicine, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Nicole de Buhr
- Department of Physiological Chemistry, Department of Infectious Diseases, University of Veterinary Medicine Hannover, Hannover, Germany.,Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine Hannover, Hannover, Germany
| | - Andreas Beineke
- Department of Pathology, University of Veterinary Medicine Hannover, Hannover, Germany
| | | | - Wolfgang Brück
- Department of Neuropathology, University Medical Center Göttingen, Georg-August-University Göttingen, Göttingen, Germany
| | - Roland Nau
- Department of Neuropathology, University Medical Center Göttingen, Georg-August-University Göttingen, Göttingen, Germany.,Department of Geriatrics, Evangelisches Krankenhaus Göttingen-Weende, Göttingen, Germany
| |
Collapse
|
29
|
Xing W, Huang P, Lu Y, Zeng W, Zuo Z. Amantadine attenuates sepsis-induced cognitive dysfunction possibly not through inhibiting toll-like receptor 2. J Mol Med (Berl) 2018; 96:391-402. [PMID: 29502203 PMCID: PMC5902799 DOI: 10.1007/s00109-018-1631-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 02/09/2018] [Accepted: 02/20/2018] [Indexed: 01/24/2023]
Abstract
Amantadine has been shown to reduce anesthesia and surgery-induced neuroinflammation and cognitive dysfunction. It is known that sepsis can impair brain function. We determined whether amantadine-attenuated sepsis-induced neuroinflammation and dysfunction of learning and memory and whether toll-like receptors (TLRs) play a role in the effects. Six- to eight-week-old mice were subjected to cecal ligation and puncture (CLP). Amantadine at 30 mg/kg/day was injected intraperitoneally for 3 days. CU-CPT22, a TLR1/TLR2 inhibitor, at 3 mg/kg/day was injected intraperitoneally for 2 days. Mice were subjected to Barnes maze and fear conditioning tests from 1 week after CLP. CLP induced neuroinflammation and cognitive dysfunction. CLP also increased the expression of toll-like receptor 2 (TLR2), TLR4, and TLR9, three major TLRs in the brain, in CD-1 male mice. Amantadine attenuated CLP-induced neuroinflammation and dysfunction of learning and memory but did not have significant effects on the expression of TLRs. CU-CPT22 also attenuated sepsis-induced neuroinflammation and cognitive dysfunction. Similarly, sepsis induced neuroinflammation and cognitive dysfunction in the C57BL/6J mice. Interestingly, sepsis also induced neuroinflammation and cognitive dysfunction in the TLR2 knockout mice. The effects of amantadine on the neuroinflammation and cognitive dysfunction were still apparent in these knockout mice. TLR2 contributes to sepsis-induced neuroinflammation and cognitive dysfunction. However, inhibiting TLR2 may not be a major mechanism for amantadine to inhibit sepsis-induced neuroinflammation and cognitive dysfunction. KEY MESSAGES Sepsis induces neuroinflammation and cognitive impairment, which were attenuated by amantadine. Toll-like receptors 2 mediates these sepsis effects but may not be the major target for amantadine to reduce these effects.
Collapse
Affiliation(s)
- Wei Xing
- Department of Anesthesiology, University of Virginia Health System, 1 Hospital Drive, PO Box 800710, Charlottesville, VA, 22908-0710, USA
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Department of Anesthesiology, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Pinjie Huang
- Department of Anesthesiology, University of Virginia Health System, 1 Hospital Drive, PO Box 800710, Charlottesville, VA, 22908-0710, USA
- Department of Anesthesiology, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510630, People's Republic of China
| | - Yang Lu
- Department of Anesthesiology, University of Virginia Health System, 1 Hospital Drive, PO Box 800710, Charlottesville, VA, 22908-0710, USA
- Department of Anesthesiology, Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Weian Zeng
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Department of Anesthesiology, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Zhiyi Zuo
- Department of Anesthesiology, University of Virginia Health System, 1 Hospital Drive, PO Box 800710, Charlottesville, VA, 22908-0710, USA.
- Department of Anesthesiology and Laboratory of RNA and Major Diseases of Brain and Heart, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510120, China.
| |
Collapse
|
30
|
Ahmad SF, Ansari MA, Nadeem A, Alzahrani MZ, Bakheet SA, Attia SM. Resveratrol Improves Neuroimmune Dysregulation Through the Inhibition of Neuronal Toll-Like Receptors and COX-2 Signaling in BTBR T + Itpr3 tf/J Mice. Neuromolecular Med 2018; 20:133-146. [PMID: 29468499 DOI: 10.1007/s12017-018-8483-0] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Accepted: 02/16/2018] [Indexed: 12/29/2022]
Abstract
Autism is a neurodevelopmental disorder characterized by deficits in qualitative impairments in communication, repetitive and social interaction, restricted, and stereotyped patterns of behavior. Resveratrol has been extensively studied pharmacologically and biologically and has anti-inflammatory, antioxidant, and neuroprotective effects on neuronal damage in neurodegenerative disorders. The BTBR T+ Itpr3tf/J (BTBR) autistic mouse model has been explored for treatment of autism, which shows low reciprocal social interactions, impaired juvenile play, and decreased social approach. Here, we explored whether resveratrol treatment decreases neuroimmune dysregulation mediated through toll-like receptor (TLR4) and nuclear factor-κB (NF-κB) signaling pathway in BTBR mice. We investigated the effect of resveratrol treatment on TLR2, TLR3, TLR4, NF-κB, and inducible nitric oxide synthase (iNOS or NOS2) levels in CD4 spleen cells. We also assessed the effect of resveratrol treatment on TLR2, TLR3, TLR4, NF-κB, iNOS, and cyclooxygenase (COX-2) mRNA expression levels in the brain tissue. We further explored TLR2, TLR4, NF-κB, iNOS, and COX-2 protein expression levels in the brain tissue. Resveratrol treatment on BTBR mice significantly decreased CD4+TLR2+, CD4+TLR3+, CD4+TLR4+ CD4+NF-κB+, and CD4+iNOS+ levels in spleen cells. Resveratrol treatment on BTBR mice decreased TLR2, TLR3, TLR4, NF-κB, iNOS, and COX-2 mRNA expression levels in brain tissue. Moreover, resveratrol treatment resulted in decreased protein expression of TLR2, TLR3, TLR4, NF-κB, iNOS, and COX-2 in brain tissue. Taken together, these results indicate that resveratrol treatment improves neuroimmune dysregulation through the inhibition of proinflammatory mediators and TLRs/NF-κB transcription factor signaling, which might be help devise future therapies for neuroimmune disorders.
Collapse
Affiliation(s)
- Sheikh F Ahmad
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Kingdom of Saudi Arabia.
| | - Mushtaq A Ansari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Kingdom of Saudi Arabia
| | - Ahmed Nadeem
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Kingdom of Saudi Arabia
| | - Mohammad Z Alzahrani
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Kingdom of Saudi Arabia
| | - Saleh A Bakheet
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Kingdom of Saudi Arabia
| | - Sabry M Attia
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Kingdom of Saudi Arabia.,Department of Pharmacology and Toxicology, College of Pharmacy, Al-Azhar University, Cairo, Egypt
| |
Collapse
|
31
|
Syk and Src-targeted anti-inflammatory activity of aripiprazole, an atypical antipsychotic. Biochem Pharmacol 2018; 148:1-12. [DOI: 10.1016/j.bcp.2017.12.006] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 12/07/2017] [Indexed: 12/20/2022]
|
32
|
Abstract
PURPOSE OF REVIEW To summarize recent clinical and preclinical studies on extracranial pathophysiologies in migraine. It challenges the opinion-based notion that the headache phase of migraine occurs without input from peripheral nociceptors or is caused solely by activation of intracranial nociceptors supplying dural and cerebral vasculature. RECENT FINDINGS Data that support a scenario by which migraine can originate extracranially include the perception of imploding headache that hurts outside the cranium, the existence of a network of sensory fibers that bifurcate from parent axons of intracranial meningeal nociceptors and reach extracranial tissues such as periosteum and pericranial muscles by crossing the calvarial bones through the sutures, the discovery of proinflammatory genes that are upregulated and anti-inflammatory genes that are down regulated in extracranial tissue of chronic migraine patients, and evidence that administration of OnabotulinumtoxinA to peripheral tissues outside the calvaria reduces frequency of migraine headache. SUMMARY These findings seeks to shift clinical practice from prophylactically treating chronic migraine solely with medications that reduce neuronal excitability to treating irritated nociceptors or affected tissues. The findings also seeks to shift current research from focusing solely on central nervous system alterations and activation of meningeal nociceptors as a prerequisite for studying migraine.
Collapse
|
33
|
Toll-like receptors, NF-κB, and IL-27 mediate adenosine A2A receptor signaling in BTBR T + Itpr3 tf/J mice. Prog Neuropsychopharmacol Biol Psychiatry 2017; 79:184-191. [PMID: 28668513 DOI: 10.1016/j.pnpbp.2017.06.034] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Revised: 06/28/2017] [Accepted: 06/28/2017] [Indexed: 12/18/2022]
Abstract
Autism is a predominant neurodevelopmental disorder characterized by impaired communication, social deficits, and repetitive behaviors. Recent research has proposed that the impairment of innate immunity may play an important role in autism. Toll-like receptors (TLRs) are potential therapeutic targets against neuroinflammation. The BTBR T+ Itpr3tf/J (BTBR) mouse is a well-known model of autism, showing repetitive behaviors such as cognitive inflexibility and increased grooming as compared to C57BL/6 (B6) mice. Adenosine A2A receptor (A2AR) signaling is involved in inflammation, brain injury, and lymphocyte infiltration into the CNS, but the role of A2AR in autism remains unknown. We investigated the effect of A2AR antagonist SCH 5826 (SCH) and agonist CGS 21680 (CGS) on the expression levels of TLRs, IL-27, NF-κB p65, and IκBα in BTBR mice. Treatment of BTBR mice with SCH increased the percentage of splenic CD14+TLR2+ cells, CD14+TLR3+ cells, CD14+TLR4+ cells, and decreased the percentage of CD14+IL-27+ cells, as compared to the untreated BTBR mice. Our results reveal that BTBR mice treated with CGS had reversal of SCH-induced immunological responses. Moreover, mRNA and protein expression analyses confirmed increased expression of TLR2, TLR3, TLR4, and NF-κB p65 in brain tissue, and decreased IL-27 and IκBα expression following SCH treatment, as compared to the untreated-BTBR and CGS-treated BTBR mice. Together, these results suggest that the A2AR agonist corrects neuroimmune dysfunction observed in BTBR mice, and thus has the potential as a therapeutic approach in autism.
Collapse
|
34
|
Sun W, Ding Z, Xu S, Su Z, Li H. Crosstalk between TLR2 and Sphk1 in microglia in the cerebral ischemia/reperfusion-induced inflammatory response. Int J Mol Med 2017; 40:1750-1758. [PMID: 29039449 PMCID: PMC5716455 DOI: 10.3892/ijmm.2017.3165] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Accepted: 09/06/2017] [Indexed: 01/06/2023] Open
Abstract
Stroke is associated with high morbidity and mortality, and much remains unknown about the injury-related mechanisms that occur following reperfusion. This study aimed to explore the roles of Toll-like receptor 2 (TLR2) and sphingosine kinase 1 (Sphk1) in microglial cells in inflammatory responses induced by cerebral ischemia/reperfusion (I/R). For this purpose, C57BL/6 mice were randomly divided into 4 groups as follows: the sham-operated group, the I/R group, the I/R group treated with TLR2 antibody, and the I/R group treated with N,N-dimethylsphingosine. Focal cerebral I/R was induced by middle cerebral artery occlusion. Double-labeling immunofluorescence was used to observe the protein expression of TLR2 and Sphk1 in the ischemic brain tissue. Quantitative polymerase chain reaction was performed to determine the mRNA levels of TLR2 and Sphkl in ischemic brain tissue. Enzyme-linked immunosorbent assay was carried out to detect the protein contents of interleukin (IL)-1β, tumor necrosis factor-α (TNF-α), IL-17 and IL-23 in ischemic brain tissue. The results revealed that I/R upregulated TLR2 and Sphk1 expression in microglial cells, and the inhibition of either TLR2 or Sphk1 inhibited the expression of the pro-inflammatory cytokines, IL-1β, TNF-α, IL-17 and IL-23. Notably, the inhibition of TLR2 activity also decreased Sphk1 expression. These results thus indicate that the activation of microglial cells, via a TLR2→Sphk1→pro-inflammatory cytokine (IL-1β, TNF-α, IL-17 and IL-23) pathway, may participate in I/R injury.
Collapse
Affiliation(s)
- Wei Sun
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Zhaoming Ding
- Department of Thyroid Surgery, The Third Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Shengjie Xu
- Department of Pathology, Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Zhiqiang Su
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Hulun Li
- Department of Neurobiology, Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| |
Collapse
|
35
|
Protective Role of Apocynin via Suppression of Neuronal Autophagy and TLR4/NF-κB Signaling Pathway in a Rat Model of Traumatic Brain Injury. Neurochem Res 2017; 42:3296-3309. [DOI: 10.1007/s11064-017-2372-z] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2016] [Revised: 06/14/2017] [Accepted: 08/01/2017] [Indexed: 10/19/2022]
|
36
|
Rodríguez AM, Delpino MV, Miraglia MC, Costa Franco MM, Barrionuevo P, Dennis VA, Oliveira SC, Giambartolomei GH. Brucella abortus-activated microglia induce neuronal death through primary phagocytosis. Glia 2017; 65:1137-1151. [PMID: 28398652 DOI: 10.1002/glia.23149] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 02/13/2017] [Accepted: 03/22/2017] [Indexed: 01/18/2023]
Abstract
Inflammation has long been implicated as a contributor to pathogenesis in neurobrucellosis. Many of the associated neurocognitive symptoms of neurobrucellosis may be the result of neuronal dysfunction resulting from the inflammatory response induced by Brucella abortus infection in the central nervous system. In this manuscript, we describe an immune mechanism for inflammatory activation of microglia that leads to neuronal death upon B. abortus infection. B. abortus was unable to infect or harm primary cultures of mouse neurons. However, when neurons were co-cultured with microglia and infected with B. abortus significant neuronal loss occurred. This phenomenon was dependent on TLR2 activation by Brucella lipoproteins. Neuronal death was not due to apoptosis, but it was dependent on the microglial release of nitric oxide (NO). B. abortus infection stimulated microglial proliferation, phagocytic activity and engulfment of neurons. NO secreted by B. abortus-activated microglia induced neuronal exposure of the "eat-me" signal phosphatidylserine (PS). Blocking of PS-binding to protein milk fat globule epidermal growth factor-8 (MFG-E8) or microglial vitronectin receptor-MFG-E8 interaction was sufficient to prevent neuronal loss by inhibiting microglial phagocytosis without affecting their activation. Taken together, our results indicate that B. abortus is not directly toxic to neurons; rather, these cells become distressed and are killed by phagocytosis in the inflammatory surroundings generated by infected microglia. Neuronal loss induced by B. abortus-activated microglia may explain, in part, the neurological deficits observed during neurobrucellosis.
Collapse
Affiliation(s)
- Ana M Rodríguez
- Instituto de Inmunología, Genética y Metabolismo (INIGEM), CONICET, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - M Victoria Delpino
- Instituto de Inmunología, Genética y Metabolismo (INIGEM), CONICET, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - M Cruz Miraglia
- Instituto de Inmunología, Genética y Metabolismo (INIGEM), CONICET, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Miriam M Costa Franco
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte-Minas Gerais, Brazil
| | - Paula Barrionuevo
- Instituto de Medicina Experimental (CONICET-Academia Nacional de Medicina), Buenos Aires, Argentina
| | - Vida A Dennis
- Center for Nano Biotechnology Research and Department of Biological Sciences, Alabama State University, Montgomery, AL
| | - Sergio C Oliveira
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte-Minas Gerais, Brazil
| | - Guillermo H Giambartolomei
- Instituto de Inmunología, Genética y Metabolismo (INIGEM), CONICET, Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
37
|
Hossain MJ, Tanasescu R, Gran B. Innate immune regulation of autoimmunity in multiple sclerosis: Focus on the role of Toll-like receptor 2. J Neuroimmunol 2016; 304:11-20. [PMID: 28007303 DOI: 10.1016/j.jneuroim.2016.12.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Accepted: 12/11/2016] [Indexed: 02/06/2023]
Abstract
Innate immunity relies on a set of germline-encoded receptors including Toll-like receptors (TLRs) that enable the host to discriminate between self and non-self. Multiple sclerosis (MS) is an autoimmune inflammatory demyelinating disease of the central nervous system (CNS). Infections are thought to play an important role in disease susceptibility. The role of innate immunity in MS has been recently appreciated. TLR2, a member of the TLR family, forms heterodimers with either TLR1 or TLR6 and detects a wide range of microbial as well as self-derived molecular structures. It may thus be important both in fighting infection and in activating autoimmunity. In this review, we discuss innate regulation of autoimmunity in MS with a focus on the role of TLR2 signaling.
Collapse
Affiliation(s)
- Md Jakir Hossain
- Division of Clinical Neuroscience, University of Nottingham, School of Medicine, Queen's Medical Centre, Nottingham NG7 2UH, United Kingdom
| | - Radu Tanasescu
- Division of Clinical Neuroscience, University of Nottingham, School of Medicine, Queen's Medical Centre, Nottingham NG7 2UH, United Kingdom; Department of Neurology, Neurosurgery and Psychiatry, University of Medicine and Pharmacy Carol Davila, Colentina Hospital, Bucharest, Romania
| | - Bruno Gran
- Division of Clinical Neuroscience, University of Nottingham, School of Medicine, Queen's Medical Centre, Nottingham NG7 2UH, United Kingdom; Department of Neurology, Nottingham University Hospitals NHS Trust, Nottingham NG7 2UH, United Kingdom.
| |
Collapse
|
38
|
Detection of Haemophilus influenzae type b, Streptococcus agalactiae, Streptococcus pneumoniae and Neisseria meningitidis in CSF specimens of children suspicious of Meningitis in Ahvaz, Iran. Kaohsiung J Med Sci 2016; 32:501-506. [PMID: 27742033 DOI: 10.1016/j.kjms.2016.08.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Revised: 08/19/2016] [Accepted: 08/19/2016] [Indexed: 11/23/2022] Open
Abstract
Meningitis is a life-threatening infection associated with a high mortality and morbidity worldwide. Neisseria meningitidis, Haemophilus influenzae, and Streptococcus pneumoniae are the most prevalent infectious agents that cause bacterial meningitis (BM). The objective of this study was to determine the frequencies of these three bacteria using bacterial cultures and polymerase chain reaction (PCR). In our cross-sectional study, cerebrospinal fluid (CSF) specimens were obtained from 196 patients who were suspected of having BM and referred to the pediatric ward of Abuzar Hospital (Ahvaz, Iran). The samples were monitored by gram stain, cultures, and the PCR method. The patients' age mean was 23 ± 0.56 months. The 196 patients comprised 92 (46.9%) boys and 104 (53.06%) girls. Based on bacterial cultures, just three isolates of H. influenzae were detected. However, PCR detected this bacterium in eight patients. Streptococcus pneumoniae was detected in five (2.5%) patients by the amplification of the lytA gene and in one (0.5%) patient by ply. In this study, no N. meningitidis isolate was in the CSF samples, based on the bacterial culture or PCR results. Streptococcus agalactiae was detected only in one patient, based on PCR. In conclusion, in the present study, the PCR method was more sensitive and rapid than culture for detecting the infectious agents in BM. For this reason, this diagnosis method is recommended for BM.
Collapse
|
39
|
Woo YR, Lim JH, Cho DH, Park HJ. Rosacea: Molecular Mechanisms and Management of a Chronic Cutaneous Inflammatory Condition. Int J Mol Sci 2016; 17:ijms17091562. [PMID: 27649161 PMCID: PMC5037831 DOI: 10.3390/ijms17091562] [Citation(s) in RCA: 119] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Revised: 08/23/2016] [Accepted: 09/07/2016] [Indexed: 01/21/2023] Open
Abstract
Rosacea is a chronic cutaneous inflammatory disease that affects the facial skin. Clinically, rosacea can be categorized into papulopustular, erythematotelangiectatic, ocular, and phymatous rosacea. However, the phenotypic presentations of rosacea are more heterogeneous. Although the pathophysiology of rosacea remains to be elucidated, immunologic alterations and neurovascular dysregulation are thought to have important roles in initiating and strengthening the clinical manifestations of rosacea. In this article, we present the possible molecular mechanisms of rosacea based on recent laboratory and clinical studies. We describe the genetic predisposition for rosacea along with its associated diseases, triggering factors, and suggested management options in detail based on the underlying molecular biology. Understanding the molecular pathomechanisms of rosacea will likely aid toward better comprehending its complex pathogenesis.
Collapse
Affiliation(s)
- Yu Ri Woo
- Department of Dermatology, Yeouido St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul 07345, Korea.
| | - Ji Hong Lim
- Department of Dermatology, Yeouido St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul 07345, Korea.
| | - Dae Ho Cho
- Department of Life Science, Sookmyung Women's University, Seoul 04310, Korea.
| | - Hyun Jeong Park
- Department of Dermatology, Yeouido St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul 07345, Korea.
| |
Collapse
|
40
|
Böhland M, Kress E, Stope MB, Pufe T, Tauber SC, Brandenburg LO. Lack of Toll-like receptor 2 results in higher mortality of bacterial meningitis by impaired host resistance. J Neuroimmunol 2016; 299:90-97. [PMID: 27725130 DOI: 10.1016/j.jneuroim.2016.09.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Revised: 09/01/2016] [Accepted: 09/04/2016] [Indexed: 12/17/2022]
Abstract
Bacterial meningitis is - despite therapeutical progress during the last decades - still characterized by high mortality and severe permanent neurogical sequelae. The brain is protected from penetrating pathogens by both the blood-brain barrier and the innate immune system. Invading pathogens are recognized by so-called pattern recognition receptors including the Toll-like receptors (TLR) which are expressed by glial immune cells in the central nervous system. Among these, TLR2 is responsible for the detection of Gram-positive bacteria such as the meningitis-causing pathogen Streptococcus pneumoniae. Here, we used TLR2-deficient mice to investigate the effects on mortality, bacterial growth and inflammation in a mouse model of pneumococcal meningitis. Our results revealed a significantly increased mortality rate and higher bacterial burden in TLR2-deficient mice with pneumococcal meningitis. Furthermore, infected TLR2-deficient mice suffered from a significantly increased pro-inflammatory cytokine tumor necrosis factor-α (TNF-α) and Chemokine (C-C motif) ligand 2 (CCL2) or CCL3 chemokine expression and decreased expression of anti-inflammatory cytokines and antimicrobial peptides. In contrast, glial cell activation assessed by glial cell marker expression was comparable to wildtype mice. Taken together, the results suggest that TLR2 is essential for an efficient immune response against Streptococcus pneumoniae meningitis since lack of the receptor led to a worse outcome by higher mortality due to increased bacterial burden, weakened innate immune response and reduced expression of antimicrobial peptides.
Collapse
Affiliation(s)
- Martin Böhland
- Department of Anatomy and Cell Biology, RWTH Aachen University, Germany
| | - Eugenia Kress
- Department of Anatomy and Cell Biology, RWTH Aachen University, Germany
| | - Matthias B Stope
- Department of Urology, University Medicine Greifswald, Greifswald, Germany
| | - Thomas Pufe
- Department of Anatomy and Cell Biology, RWTH Aachen University, Germany
| | - Simone C Tauber
- Department of Neurology, RWTH University Hospital Aachen, Germany
| | | |
Collapse
|
41
|
Perry CJ, Blake P, Buettner C, Papavassiliou E, Schain AJ, Bhasin MK, Burstein R. Upregulation of inflammatory gene transcripts in periosteum of chronic migraineurs: Implications for extracranial origin of headache. Ann Neurol 2016; 79:1000-13. [PMID: 27091721 DOI: 10.1002/ana.24665] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Revised: 03/29/2016] [Accepted: 04/07/2016] [Indexed: 02/06/2023]
Abstract
OBJECTIVE Chronic migraine (CM) is often associated with chronic tenderness of pericranial muscles. A distinct increase in muscle tenderness prior to onset of occipital headache that eventually progresses into a full-blown migraine attack is common. This experience raises the possibility that some CM attacks originate outside the cranium. The objective of this study was to determine whether there are extracranial pathophysiologies in these headaches. METHODS We biopsied and measured the expression of gene transcripts (mRNA) encoding proteins that play roles in immune and inflammatory responses in affected (ie, where the head hurts) calvarial periosteum of (1) patients whose CMs are associated with muscle tenderness and (2) patients with no history of headache. RESULTS Expression of proinflammatory genes (eg, CCL8, TLR2) in the calvarial periosteum significantly increased in CM patients attesting to muscle tenderness, whereas expression of genes that suppress inflammation and immune cell differentiation (eg, IL10RA, CSF1R) decreased. INTERPRETATION Because the upregulated genes were linked to activation of white blood cells, production of cytokines, and inhibition of NF-κB, and the downregulated genes were linked to prevention of macrophage activation and cell lysis, we suggest that the molecular environment surrounding periosteal pain fibers is inflamed and in turn activates trigeminovascular nociceptors that reach the affected periosteum through suture branches of intracranial meningeal nociceptors and/or somatic branches of the occipital nerve. This study provides the first set of evidence for localized extracranial pathophysiology in CM. Ann Neurol 2016;79:1000-1013.
Collapse
Affiliation(s)
| | - Pamela Blake
- Headache Center of Greater Heights, Memorial Hermann Greater Heights Hospital, Houston, TX
| | - Catherine Buettner
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA.,Harvard Medical School, Boston, MA
| | - Efstathios Papavassiliou
- Harvard Medical School, Boston, MA.,Department of Surgery, Beth Israel Deaconess Medical Center, Boston, MA
| | - Aaron J Schain
- Harvard Medical School, Boston, MA.,Department of Anesthesia and Critical Care, Beth Israel Deaconess Medical Center, Boston, MA
| | - Manoj K Bhasin
- Harvard Medical School, Boston, MA.,Division of Genomics, Proteomics, Bioinformatics, and Systems Biology, Beth Israel Deaconess Medical Center, Boston, MA
| | - Rami Burstein
- Harvard Medical School, Boston, MA.,Department of Anesthesia and Critical Care, Beth Israel Deaconess Medical Center, Boston, MA
| |
Collapse
|
42
|
Fitzpatrick JMK, Downer EJ. Toll-like receptor signalling as a cannabinoid target in Multiple Sclerosis. Neuropharmacology 2016; 113:618-626. [PMID: 27079840 DOI: 10.1016/j.neuropharm.2016.04.009] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Revised: 01/20/2016] [Accepted: 04/08/2016] [Indexed: 02/06/2023]
Abstract
Toll-like receptors (TLRs) are the sensors of pathogen-associated molecules that trigger tailored innate immune intracellular signalling responses to initiate innate immune reactions. Data from the experimental autoimmune encephalomyelitis (EAE) model indicates that TLR signalling machinery is a pivotal player in the development of murine EAE. To compound this, data from human studies indicate that complex interplay exists between TLR signalling and Multiple Sclerosis (MS) pathogenesis. Cannabis-based therapies are in clinical development for the management of a variety of medical conditions, including MS. In particular Sativex®, a combination of plant-derived cannabinoids, is an oromucosal spray with efficacy in MS patients, particularly those with neuropathic pain and spasticity. Despite this, the precise cellular and molecular mechanisms of action of Sativex® in MS patients remains unclear. This review will highlight evidence that novel interplay exists between the TLR and cannabinoid systems, both centrally and peripherally, with relevance to the pathogenesis of MS. This article is part of the Special Issue entitled 'Lipid Sensing G Protein-Coupled Receptors in the CNS'.
Collapse
Affiliation(s)
- John-Mark K Fitzpatrick
- Department of Physiology, School of Medicine, Trinity Biomedical Sciences Institute, University of Dublin, Trinity College, Dublin 2, Ireland
| | - Eric J Downer
- Department of Physiology, School of Medicine, Trinity Biomedical Sciences Institute, University of Dublin, Trinity College, Dublin 2, Ireland.
| |
Collapse
|
43
|
Rogers CE, Smyser T, Smyser CD, Shimony J, Inder TE, Neil JJ. Regional white matter development in very preterm infants: perinatal predictors and early developmental outcomes. Pediatr Res 2016; 79:87-95. [PMID: 26372513 PMCID: PMC4724306 DOI: 10.1038/pr.2015.172] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2015] [Accepted: 06/29/2015] [Indexed: 11/24/2022]
Abstract
BACKGROUND Preterm infants are at risk for white matter (WM) injury and adverse neurodevelopmental outcomes. METHODS Serial diffusion tensor magnetic resonance imaging data were obtained from very preterm infants (N = 78) born <30 wk gestation imaged up to four times from 26-42 wk postmenstrual age. Slopes were calculated for fractional anisotropy (FA) and mean diffusivity (MD) within regions of interest for infants with ≥2 scans (N = 50). Sixty-five children underwent neurodevelopmental testing at 2 y of age. RESULTS FA slope for the posterior limb of the internal capsule was greater than other regions. The anterior limb of the internal capsule (ALIC), corpus callosum, and optic radiations demonstrated greater FA slope with increasing gestational age. Infants with patent ductus arteriosus had lower FA slope in the ALIC. MD slope was lower with prolonged ventilation or lack of antenatal steroids. At 2 y of age, lower motor scores were associated with lower FA in the left but higher FA in the right inferior temporal lobe at term-equivalent age. Better social-emotional competence was related to lower FA in the left cingulum bundle. CONCLUSION This study demonstrates regional variability in the susceptibility/sensitivity of WM maturation to perinatal factors and relationships between altered diffusion measures and developmental outcomes in preterm neonates.
Collapse
Affiliation(s)
- Cynthia E. Rogers
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA,Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
| | - Tara Smyser
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| | - Christopher D. Smyser
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA,Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Joshua Shimony
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | | | - Jeffrey J. Neil
- Department of Neurology, Boston Children's Hospital, Boston, MA, USA
| |
Collapse
|
44
|
Yau B, Mitchell AJ, Too LK, Ball HJ, Hunt NH. Interferon-γ-Induced Nitric Oxide Synthase-2 Contributes to Blood/Brain Barrier Dysfunction and Acute Mortality in Experimental Streptococcus pneumoniae Meningitis. J Interferon Cytokine Res 2015; 36:86-99. [PMID: 26418460 DOI: 10.1089/jir.2015.0078] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The proinflammatory cytokine interferon-gamma (IFNγ) recently was shown to play a crucial role in experimental pneumococcal meningitis (PM) pathogenesis, and we aimed in this study to investigate IFNγ-driven nitric oxide synthase-2 (NOS2)-mediated pathogenesis of murine PM. We demonstrate that costimulation of toll-like receptors and IFNγ receptors was synergistic for NOS2 expression in cultured murine microglia. Using an experimental PM model, wild-type mice treated with anti-IFNγ antibody, as well as IFNγ and NOS2 gene knockout (GKO) mice, were inoculated intracerebroventricularly with 10(3) colony-forming units of Streptococcus pneumoniae (WU2 strain). Mice were monitored daily during a 200-h disease course to assess survival rate and blood-brain barrier (BBB) permeability measured at 48 h. IFNγ deficiency was protective in PM, with an approximate 3-fold increase in survival rates in both antibody-treated and IFNγ GKO mice compared to controls (P < 0.01). At 48 h postinoculation, brain NOS2 mRNA expression was significantly increased in an IFNγ-dependent manner. Mortality was significantly delayed in NOS2 GKO mice compared to controls (P < 0.01), and BBB dysfunction was reduced by 54% in IFNγ GKO mice and abolished in NOS2 GKO. These data suggest that IFNγ-dependent expression of NOS2 in the brain contributes to BBB breakdown and early mortality in murine PM.
Collapse
Affiliation(s)
- Belinda Yau
- 1 Molecular Immunopathology Unit, School of Medical Sciences, Sydney Medical School, University of Sydney , Sydney, New South Wales, Australia
| | - Andrew J Mitchell
- 1 Molecular Immunopathology Unit, School of Medical Sciences, Sydney Medical School, University of Sydney , Sydney, New South Wales, Australia .,2 Centenary Institute for Cancer Medicine and Cell Biology , Newtown, New South Wales, Australia
| | - Lay Khoon Too
- 1 Molecular Immunopathology Unit, School of Medical Sciences, Sydney Medical School, University of Sydney , Sydney, New South Wales, Australia
| | - Helen J Ball
- 1 Molecular Immunopathology Unit, School of Medical Sciences, Sydney Medical School, University of Sydney , Sydney, New South Wales, Australia
| | - Nicholas H Hunt
- 1 Molecular Immunopathology Unit, School of Medical Sciences, Sydney Medical School, University of Sydney , Sydney, New South Wales, Australia
| |
Collapse
|
45
|
α-TLR2 antibody attenuates the Aβ-mediated inflammatory response in microglia through enhanced expression of SIGIRR. Brain Behav Immun 2015; 46:70-9. [PMID: 25620736 DOI: 10.1016/j.bbi.2015.01.005] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Revised: 01/06/2015] [Accepted: 01/07/2015] [Indexed: 01/11/2023] Open
Abstract
The immunoregulatory function of single-Ig-interleukin-1 related receptor (SIGIRR) is derived from its ability to constrain the inflammatory consequences of interleukin (IL)-1R and toll-like receptor (TLR)4 activation. This role extends to the brain, where SIGIRR deficiency increases the synaptic and cognitive dysfunction associated with IL-1R- and TLR4-mediated signalling. The current study set out to investigate the interaction between SIGIRR and TLR2 in brain tissue and the data demonstrate that the response to the TLR2 agonist, Pam3CysSK4 (Pam3Cys4), is enhanced in glial cells from SIGIRR(-/-) animals. Consistent with the view that β-amyloid peptide (Aβ) signals through activation of TLR2, the data also show that Aβ-induced changes are exaggerated in glia from SIGIRR(-/-) animals. We report that microglia, rather than astrocytes, are the primary glial cell expressing both TLR2 and SIGIRR. While Aβ increased TLR2 expression, it decreased SIGIRR expression in microglia. This was mimicked by direct activation of TLR2 with Pam3Cys4. We investigated the effect of an anti-TLR2 antibody (αTLR2) on the Aβ-induced inflammatory responses and demonstrate that it prevented the expression and release of the pro-inflammatory cytokines TNFα and IL-6 from microglia. In addition, application of αTLR2 alleviated the Aβ-mediated impairment in long-term potentiation (LTP) of hippocampal synaptic activity. The protective effects of αTLR2 were accompanied by an up-regulation in SIGIRR expression. We propose that a mechanism involving activation of PI3 kinase/Akt and the transcription factor peroxisome proliferator-activated receptor (PPAR)γ may facilitate this increase in SIGIRR. These findings highlight a novel role of SIGIRR as a negative regulator of TLR2-mediated inflammation in the brain.
Collapse
|
46
|
Bi D, Qiao L, Bergelson I, Ek CJ, Duan L, Zhang X, Albertsson AM, Pettengill M, Kronforst K, Ninkovic J, Goldmann D, Janzon A, Hagberg H, Wang X, Mallard C, Levy O. Staphylococcus epidermidis Bacteremia Induces Brain Injury in Neonatal Mice via Toll-like Receptor 2-Dependent and -Independent Pathways. J Infect Dis 2015; 212:1480-90. [PMID: 25883383 DOI: 10.1093/infdis/jiv231] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Accepted: 04/07/2015] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Staphylococcus epidermidis causes late-onset sepsis in preterm infants. Staphylococcus epidermidis activates host responses in part via Toll-like receptor 2 (TLR2). Epidemiologic studies link bacteremia and neonatal brain injury, but direct evidence is lacking. METHODS Wild-type and TLR2-deficient (TLR2-/-) mice were injected intravenously with S. epidermidis at postnatal day 1 prior to measuring plasma and brain cytokine and chemokine levels, bacterial clearance, brain caspase-3 activation, white/gray matter volume, and innate transcriptome. RESULTS Staphylococcus epidermidis bacteremia spontaneously resolved over 24 hours without detectable bacteria in the cerebrospinal fluid (CSF). TLR2-/- mice demonstrated delayed S. epidermidis clearance from blood, spleen, and liver. Staphylococcus epidermidis increased the white blood cell count in the CSF, increased interleukin 6, interleukin 12p40, CCL2, and CXCL1 concentrations in plasma; increased the CCL2 concentration in the brain; and caused rapid (within 6 hours) TLR2-dependent brain activation of caspase-3 and TLR2-independent white matter injury. CONCLUSIONS Staphylococcus epidermidis bacteremia, in the absence of bacterial entry into the CSF, impairs neonatal brain development. Staphylococcus epidermidis bacteremia induced both TLR2-dependent and -independent brain injury, with the latter occurring in the absence of TLR2, a condition associated with an increased bacterial burden. Our study indicates that the consequences of transient bacteremia in early life may be more severe than commonly appreciated, and our findings may inform novel approaches to reduce bacteremia-associated brain injury.
Collapse
Affiliation(s)
- Dan Bi
- Department of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Sweden Department of Pediatrics, Third Affiliated Hospital, Zhengzhou University
| | - Lili Qiao
- Department of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Sweden Department of Pediatrics, Song Jiang Central Hospital, Shanghai, China
| | - Ilana Bergelson
- Division of Infectious Diseases, Department of Medicine, Boston Children's Hospital Harvard Medical School, Boston, Massachusetts
| | - C Joakim Ek
- Department of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Luqi Duan
- Department of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Sweden Department of Pediatrics, Third Affiliated Hospital, Zhengzhou University
| | - Xiaoli Zhang
- Department of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Sweden Department of Pediatrics, Third Affiliated Hospital, Zhengzhou University
| | - Anna-Maj Albertsson
- Department of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Matthew Pettengill
- Division of Infectious Diseases, Department of Medicine, Boston Children's Hospital Harvard Medical School, Boston, Massachusetts
| | - Kenny Kronforst
- Department of Pediatrics, Division of Neonatology, Lurie Children's Hospital of Chicago and Prentice Women's Hospital, Feinberg School of Medicine, Northwestern University, Illinois
| | - Jana Ninkovic
- 3M Corporate Research Materials Laboratory, St. Paul, Minnesota
| | - Donald Goldmann
- Division of Infectious Diseases, Department of Medicine, Boston Children's Hospital Harvard Medical School, Boston, Massachusetts
| | - Anders Janzon
- Department of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Henrik Hagberg
- Department of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Sweden Department of Obstetrics and Gynecology, Sahlgrenska Academy, University of Gothenburg, Sweden Centre for the Developing Brain, Department of Perinatal Imaging and Health, King's College London, United Kingdom
| | - Xiaoyang Wang
- Department of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Sweden Department of Pediatrics, Third Affiliated Hospital, Zhengzhou University
| | - Carina Mallard
- Department of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Sweden Centre for the Developing Brain, Department of Perinatal Imaging and Health, King's College London, United Kingdom
| | - Ofer Levy
- Division of Infectious Diseases, Department of Medicine, Boston Children's Hospital Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
47
|
Intrathecal heat shock protein 60 mediates neurodegeneration and demyelination in the CNS through a TLR4- and MyD88-dependent pathway. Mol Neurodegener 2015; 10:5. [PMID: 25887709 PMCID: PMC4365525 DOI: 10.1186/s13024-015-0003-1] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Accepted: 02/03/2015] [Indexed: 01/11/2023] Open
Abstract
Background Toll-like receptors (TLR) constitute a highly conserved class of receptors through which the innate immune system responds to both pathogen- and host-derived factors. Although TLRs are involved in a wide range of central nervous system (CNS) disorders including neurodegenerative diseases, the molecular events leading from CNS injury to activation of these innate immune receptors remain elusive. The stress protein heat shock protein 60 (HSP60) released from injured cells is considered an endogenous danger signal of the immune system. In this context, the main objective of the present study was to investigate the impact of extracellular HSP60 on the brain in vivo. Results We show here that HSP60 injected intrathecally causes neuronal and oligodendrocyte injury in the CNS in vivo through TLR4-dependent signaling. Intrathecal HSP60 results in neuronal cell death, axonal injury, loss of oligodendrocytes, and demyelination in the cerebral cortex of wild-type mice. In contrast both mice lacking TLR4 and the TLR adaptor molecule MyD88 are protected against deleterious effects induced by HSP60. In contrast to the exogenous TLR4 ligand, lipopolysaccharide, intrathecal HSP60 does not induce such a considerable inflammatory response in the brain. In the CNS, endogenous HSP60 is predominantly expressed in neurons and released during brain injury, since the cerebrospinal fluid (CSF) from animals of a mouse stroke model contains elevated levels of this stress protein compared to the CSF of sham-operated mice. Conclusions Our data show a direct toxic effect of HSP60 towards neurons and oligodendrocytes in the CNS. The fact that these harmful effects involve TLR4 and MyD88 confirms a molecular pathway mediated by the release of endogenous TLR ligands from injured CNS cells common to many forms of brain diseases that bi-directionally links CNS injury and activation of the innate immune system to neurodegeneration and demyelination in vivo. Electronic supplementary material The online version of this article (doi:10.1186/s13024-015-0003-1) contains supplementary material, which is available to authorized users.
Collapse
|
48
|
Lymphocytes modulate innate immune responses and neuronal damage in experimental meningitis. Infect Immun 2014; 83:259-67. [PMID: 25348636 DOI: 10.1128/iai.02682-14] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
In bacterial meningitis, excessive immune responses carry significant potential for damage to brain tissue even after successful antibiotic therapy. Bacterial meningitis is regarded primarily as the domain of innate immunity, and the role of lymphocytes remains unclear. We studied the contribution of lymphocytes to acute inflammation and neurodegeneration in experimental Toll-like receptor 2-driven meningitis, comparing wild-type mice with RAG-1-deficient mice that have no mature T and B lymphocytes. At 24 h after intrathecal challenge with the synthetic bacterial lipopeptide Pam(3)CysSK(4), RAG-1-deficient mice displayed more pronounced clinical impairment and an increased concentration of neutrophils, reduced expression of interleukin-10 (IL-10) mRNA, and increased expression of CXCL1 mRNA in the cerebrospinal fluid. Conversely, neuronal loss in the dentate gyrus was reduced in RAG-1-deficient mice, and expression of IL-10, transforming growth factor β and CCL2 mRNA by microglia was increased compared to wild-type mice. Adoptive transfer of wild-type lymphocytes reversed the enhanced meningeal inflammation and functional impairment observed in RAG-1-deficient mice. Our findings suggest compartment-specific effects of lymphocytes during acute bacterial meningitis, including attenuation of meningeal inflammation and shifting of microglial activation toward a more neurotoxic phenotype.
Collapse
|
49
|
Rosenberger K, Derkow K, Dembny P, Krüger C, Schott E, Lehnardt S. The impact of single and pairwise Toll-like receptor activation on neuroinflammation and neurodegeneration. J Neuroinflammation 2014; 11:166. [PMID: 25239168 PMCID: PMC4182775 DOI: 10.1186/s12974-014-0166-7] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Accepted: 09/09/2014] [Indexed: 01/17/2023] Open
Abstract
Background Toll-like receptors (TLRs) enable innate immune cells to respond to pathogen- and host-derived molecules. The central nervous system (CNS) exhibits most of the TLRs identified with predominant expression in microglia, the major immune cells of the brain. Although individual TLRs have been shown to contribute to CNS disorders, the consequences of multiple activated TLRs on the brain are unclear. We therefore systematically investigated and compared the impact of sole and pairwise TLR activation on CNS inflammation and injury. Methods Selected TLRs expressed in microglia and neurons were stimulated with their specific TLR ligands in varying combinations. Cell cultures were then analyzed by immunocytochemistry, FlowCytomix, and ELISA. To determine neuronal injury and neuroinflammation in vivo, C57BL/6J mice were injected intrathecally with TLR agonists. Subsequently, brain sections were analyzed by quantitative real-time PCR and immunohistochemistry. Results Simultaneous stimulation of TLR4 plus TLR2, TLR4 plus TLR9, and TLR2 plus TLR9 in microglia by their respective specific ligands results in an increased inflammatory response compared to activation of the respective single TLR in vitro. In contrast, additional activation of TLR7 suppresses the inflammatory response mediated by the respective ligands for TLR2, TLR4, or TLR9 up to 24 h, indicating that specific combinations of activated TLRs individually modulate the inflammatory response. Accordingly, the composition of the inflammatory response pattern generated by microglia varies depending on the identity and combination of the activated TLRs engaged. Likewise, neuronal injury occurs in response to activation of only selected TLRs and TLR combinations in vitro. Activation of TLR2, TLR4, TLR7, and TLR9 in the brain by intrathecal injection of the respective TLR ligand into C57BL/6J mice leads to specific expression patterns of distinct TLR mRNAs in the brain and causes influx of leukocytes and inflammatory mediators into the cerebrospinal fluid to a variable extent. Also, the intensity of the inflammatory response and neurodegenerative effects differs according to the respective activated TLR and TLR combinations used in vivo. Conclusions Sole and pairwise activation of TLRs modifies the pattern and extent of inflammation and neurodegeneration in the CNS, thereby enabling innate immunity to take account of the CNS diseases’ diversity. Electronic supplementary material The online version of this article (doi:10.1186/s12974-014-0166-7) contains supplementary material, which is available to authorized users.
Collapse
|
50
|
Izadpanah K, Freyer D, Weber JR, Braun JS. Brain parenchymal TNF-α and IL-1β induction in experimental pneumococcal meningitis. J Neuroimmunol 2014; 276:104-11. [PMID: 25218213 DOI: 10.1016/j.jneuroim.2014.08.625] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2014] [Revised: 08/23/2014] [Accepted: 08/26/2014] [Indexed: 10/24/2022]
Abstract
Triggers of brain inflammation in pneumococcal meningitis are unknown. TNF-α and IL-1β were upregulated (real time PCR and in situ hybridization) in neurons and astrocytes time-dependently and maximally in the hippocampus during murine pneumococcal meningitis. Upregulation of TNF-α and IL-1β mRNA in the brain parenchyma was independent of cerebrospinal fluid leukocytosis, pneumococcal pneumolysin and H2O2, but it was potently induced by pneumococcal cell wall (PCW) fragments. Brain TNF-α mRNA was downregulated by a matrix metalloproteinases inhibitor. PCW fragments were located in the brain parenchyma. In conclusion, PCW fragments and matrix metalloproteinases trigger cytokine induction in the brain parenchyma during pneumococcal meningitis.
Collapse
Affiliation(s)
- Kaywan Izadpanah
- Department of Neurology, Charité Universitätsmedizin Berlin, Germany; Department of Orthopedics and Traumatology, University Freiburg, Germany
| | - Dorette Freyer
- Department of Neurology, Charité Universitätsmedizin Berlin, Germany
| | - Joerg R Weber
- Department of Neurology, Charité Universitätsmedizin Berlin, Germany; Department of Neurology, Klinikum Klagenfurt, Austria
| | - Johann S Braun
- Department of Neurology, Charité Universitätsmedizin Berlin, Germany; Department of Internal Medicine, Division Neurology, College of Medicine and Health Sciences, United Arab Emirates University, United Arab Emirates.
| |
Collapse
|