1
|
Tacke C, Landgraf P, Dieterich DC, Kröger A. The fate of neuronal synapse homeostasis in aging, infection, and inflammation. Am J Physiol Cell Physiol 2024; 327:C1546-C1563. [PMID: 39495249 DOI: 10.1152/ajpcell.00466.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 10/18/2024] [Accepted: 10/20/2024] [Indexed: 11/05/2024]
Abstract
Neuroplasticity is the brain's ability to reorganize and modify its neuronal connections in response to environmental stimuli, experiences, learning, and disease processes. This encompasses a variety of mechanisms, including changes in synaptic strength and connectivity, the formation of new synapses, alterations in neuronal structure and function, and the generation of new neurons. Proper functioning of synapses, which facilitate neuron-to-neuron communication, is crucial for brain activity. Neuronal synapse homeostasis, which involves regulating and maintaining synaptic strength and function in the central nervous system (CNS), is vital for this process. Disruptions in synaptic balance, due to factors like inflammation, aging, or infection, can lead to impaired brain function. This review highlights the main aspects and mechanisms underlying synaptic homeostasis, particularly in the context of aging, infection, and inflammation.
Collapse
Affiliation(s)
- Charlotte Tacke
- Institute of Medical Microbiology and Hospital Hygiene, Molecular Microbiology Group, Otto-von-Guericke University, Magdeburg, Germany
| | - Peter Landgraf
- Institute of Pharmacology and Toxicology, Otto-von-Guericke University, Magdeburg, Germany
| | - Daniela C Dieterich
- Institute of Pharmacology and Toxicology, Otto-von-Guericke University, Magdeburg, Germany
- Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke University, Magdeburg, Germany
| | - Andrea Kröger
- Institute of Medical Microbiology and Hospital Hygiene, Molecular Microbiology Group, Otto-von-Guericke University, Magdeburg, Germany
- Helmholtz Center for Infection Research, Innate Immunity and Infection Group, Braunschweig, Germany
- Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke University, Magdeburg, Germany
| |
Collapse
|
2
|
Tastan B, Heneka MT. The impact of neuroinflammation on neuronal integrity. Immunol Rev 2024; 327:8-32. [PMID: 39470038 DOI: 10.1111/imr.13419] [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] [Indexed: 10/30/2024]
Abstract
Neuroinflammation, characterized by a complex interplay among innate and adaptive immune responses within the central nervous system (CNS), is crucial in responding to infections, injuries, and disease pathologies. However, the dysregulation of the neuroinflammatory response could significantly affect neurons in terms of function and structure, leading to profound health implications. Although tremendous progress has been made in understanding the relationship between neuroinflammatory processes and alterations in neuronal integrity, the specific implications concerning both structure and function have not been extensively covered, with the exception of perspectives on glial activation and neurodegeneration. Thus, this review aims to provide a comprehensive overview of the multifaceted interactions among neurons and key inflammatory players, exploring mechanisms through which inflammation influences neuronal functionality and structural integrity in the CNS. Further, it will discuss how these inflammatory mechanisms lead to impairment in neuronal functions and architecture and highlight the consequences caused by dysregulated neuronal functions, such as cognitive dysfunction and mood disorders. By integrating insights from recent research findings, this review will enhance our understanding of the neuroinflammatory landscape and set the stage for future interventions that could transform current approaches to preserve neuronal integrity and function in CNS-related inflammatory conditions.
Collapse
Affiliation(s)
- Bora Tastan
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Michael T Heneka
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg
- Department of Infectious Diseases and Immunology, University of Massachusetts Medical School, North Worcester, Massachusetts, USA
| |
Collapse
|
3
|
Hoffet MS, Tomov NS, Hupp S, Mitchell TJ, Iliev AI. Glucose and Oxygen Levels Modulate the Pore-Forming Effects of Cholesterol-Dependent Cytolysin Pneumolysin from Streptococcus pneumoniae. Toxins (Basel) 2024; 16:232. [PMID: 38922127 PMCID: PMC11209487 DOI: 10.3390/toxins16060232] [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: 03/08/2024] [Revised: 05/08/2024] [Accepted: 05/16/2024] [Indexed: 06/27/2024] Open
Abstract
A major Streptococcus pneumoniae pathogenic factor is the cholesterol-dependent cytolysin pneumolysin, binding membrane cholesterol and producing permanent lytic or transient pores. During brain infections, vascular damage with variable ischemia occurs. The role of ischemia on pneumolysin's pore-forming capacity remains unknown. In acute brain slice cultures and primary cultured glia, we studied acute toxin lysis (via propidium iodide staining and LDH release) and transient pore formation (by analyzing increases in the intracellular calcium). We analyzed normal peripheral tissue glucose conditions (80 mg%), normal brain glucose levels (20 mg%), and brain hypoglycemic conditions (3 mg%), in combinations either with normoxia (8% oxygen) or hypoxia (2% oxygen). At 80 mg% glucose, hypoxia enhanced cytolysis via pneumolysin. At 20 mg% glucose, hypoxia did not affect cell lysis, but impaired calcium restoration after non-lytic pore formation. Only at 3 mg% glucose, during normoxia, did pneumolysin produce stronger lysis. In hypoglycemic (3 mg% glucose) conditions, pneumolysin caused a milder calcium increase, but restoration was missing. Microglia bound more pneumolysin than astrocytes and demonstrated generally stronger calcium elevation. Thus, our work demonstrated that the toxin pore-forming capacity in cells continuously diminishes when oxygen is reduced, overlapping with a continuously reduced ability of cells to maintain homeostasis of the calcium influx once oxygen and glucose are reduced.
Collapse
Affiliation(s)
- Michelle Salomé Hoffet
- Institute of Anatomy, University of Bern, Baltzerstrasse 2, 3012 Bern, Switzerland; (M.S.H.); (N.S.T.); (S.H.)
| | - Nikola S. Tomov
- Institute of Anatomy, University of Bern, Baltzerstrasse 2, 3012 Bern, Switzerland; (M.S.H.); (N.S.T.); (S.H.)
| | - Sabrina Hupp
- Institute of Anatomy, University of Bern, Baltzerstrasse 2, 3012 Bern, Switzerland; (M.S.H.); (N.S.T.); (S.H.)
| | - Timothy J. Mitchell
- School of Immunity and Infection, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK;
| | - Asparouh I. Iliev
- Institute of Anatomy, University of Bern, Baltzerstrasse 2, 3012 Bern, Switzerland; (M.S.H.); (N.S.T.); (S.H.)
| |
Collapse
|
4
|
Jennert F, Schaaf D, Nau R, Kohler TP, Hammerschmidt S, Häusler D, Valentin-Weigand P, Seele J. Hydrogen peroxide is responsible for the cytotoxic effects of Streptococcus pneumoniae on primary microglia in the absence of pneumolysin. J Innate Immun 2024; 16:000536514. [PMID: 38569474 PMCID: PMC11060703 DOI: 10.1159/000536514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 01/25/2024] [Indexed: 04/05/2024] Open
Abstract
INTRODUCTION Streptococcus pneumoniae is the most common cause of bacterial meningitis and meningoencephalitis in humans. The bacterium produces numerous virulence determinants, among them hydrogen peroxide (H2O2) and pneumolysin (Ply), which contribute to bacterial cytotoxicity. Microglia, the resident phagocytes in the brain, are distinct from other macrophages, and we thus compared their susceptibility to pneumococcal toxicity and their ability to phagocytose pneumococci with those of bone marrow-derived macrophages (BMDM). METHODS Microglia and BMDM were co-incubated with S. pneumoniae D39 to analyze survival of phagocytes by fluorescence microscopy, bacterial growth by quantitative plating, and phagocytosis by an antibiotic protection assay. Ply was detected by hemolysis assay and Western blot analysis. RESULTS We found that microglia were killed during pneumococcal infection with a wild-type and an isogenic ply-deficient mutant, whereas viability of BMDM was not affected by pneumococci. Treatment with recombinant Ply showed a dose-dependent cytotoxic effect on microglia and BMDM. However, high concentrations of recombinant Ply were required and under the chosen experimental conditions, Ply was not detectable in the supernatant during infection of microglia. Inactivation of H2O2 by exogenously added catalase abolished its cytotoxic effect. Consequently, infection of microglia with pneumococci deficient for the pyruvate oxidase SpxB, primarily producing H2O2, resulted in reduced killing of microglia. CONCLUSION Taken together, in the absence of Ply, H2O2 caused cell death in primary phagocytes in concentrations produced by pneumococci.
Collapse
Affiliation(s)
- Franziska Jennert
- University of Veterinary Medicine Hannover, Institute for Microbiology, Center for Infection Medicine, Hannover, Germany
| | - Désirée Schaaf
- University of Veterinary Medicine Hannover, Institute for Microbiology, Center for Infection Medicine, Hannover, Germany
| | - Roland Nau
- University Medical Center Göttingen, Department of Neuropathology, Göttingen, Germany
- Evangelisches Krankenhaus Göttingen-Weende, Department of Geriatrics, Göttingen, Germany
| | - Thomas P. Kohler
- Greifswald University, Department of Molecular Genetics and Infection Biology, Interfaculty Institute for Genetics and Functional Genomics, Greifswald, Germany
| | - Sven Hammerschmidt
- Greifswald University, Department of Molecular Genetics and Infection Biology, Interfaculty Institute for Genetics and Functional Genomics, Greifswald, Germany
| | - Darius Häusler
- University Medical Center Göttingen, Department of Neuropathology, Göttingen, Germany
- Fraunhofer-Institute for Translational Medicine and Pharmacology ITMP, Göttingen, Germany
| | - Peter Valentin-Weigand
- University of Veterinary Medicine Hannover, Institute for Microbiology, Center for Infection Medicine, Hannover, Germany
| | - Jana Seele
- University Medical Center Göttingen, Department of Neuropathology, Göttingen, Germany
- Evangelisches Krankenhaus Göttingen-Weende, Department of Geriatrics, Göttingen, Germany
| |
Collapse
|
5
|
Pramitasuri TI, Susilawathi NM, Tarini NMA, Sudewi AAR, Evans MC. Cholesterol dependent cytolysins and the brain: Revealing a potential therapeutic avenue for bacterial meningitis. AIMS Microbiol 2023; 9:647-667. [PMID: 38173970 PMCID: PMC10758573 DOI: 10.3934/microbiol.2023033] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 07/24/2023] [Accepted: 08/14/2023] [Indexed: 01/05/2024] Open
Abstract
Bacterial meningitis is a catastrophic nervous system disorder with high mortality and wide range of morbidities. Some of the meningitis-causing bacteria occupy cholesterol dependent cytolysins (CDCs) to increase their pathogenicity and arrange immune-evasion strategy. Studies have observed that the relationship between CDCs and pathogenicity in these meningitides is complex and involves interactions between CDC, blood-brain barrier (BBB), glial cells and neurons. In BBB, these CDCs acts on capillary endothelium, tight junction (TJ) proteins and neurovascular unit (NVU). CDCs also observed to elicit intriguing effects on brain inflammation which involves microglia and astrocyte activations, along with neuronal damage as the end-point of pathological pathways in bacterial meningitis. As some studies mentioned potential advantage of CDC-targeted therapeutic mechanisms to combat CNS infections, it might be a fruitful avenue to deepen our understanding of CDC as a candidate for adjuvant therapy to combat bacterial meningitis.
Collapse
Affiliation(s)
- Tjokorda Istri Pramitasuri
- Doctoral Program in Medical Science, Faculty of Medicine, Universitas Udayana, Bali, Indonesia
- Postgraduate Research Student, Faculty of Medicine, Imperial College London, United Kingdom
| | - Ni Made Susilawathi
- Department of Neurology, Faculty of Medicine, Universitas Udayana, Bali, Indonesia
| | - Ni Made Adi Tarini
- Department of Microbiology, Faculty of Medicine, Universitas Udayana-Rumah Sakit Umum Pusat Prof Dr dr IGNG Ngoerah, Bali, Indonesia
| | - AA Raka Sudewi
- Department of Neurology, Faculty of Medicine, Universitas Udayana, Bali, Indonesia
| | - Matthew C Evans
- Pain Research, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, United Kingdom
- Department of Brain Sciences, Care Research and Technology Centre, UK Dementia Research Institute, London, United Kingdom
| |
Collapse
|
6
|
Pramitasuri TI, Susilawathi NM, Sudewi AAR. Bacterial meningitis-induced demyelination: A logical fallacy or groundbreaking avenue in neuroscience? AIMS Neurosci 2023; 10:172-174. [PMID: 37426778 PMCID: PMC10323260 DOI: 10.3934/neuroscience.2023013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 06/18/2023] [Indexed: 07/11/2023] Open
Affiliation(s)
| | - Ni Made Susilawathi
- Department of Neurology, Faculty of Medicine, Universitas Udayana, Bali, Indonesia 80232
| | - AA Raka Sudewi
- Department of Neurology, Faculty of Medicine, Universitas Udayana, Bali, Indonesia 80232
| |
Collapse
|
7
|
Baronti D, Tomov N, Hupp S, Mitchell TJ, Iliev AI. Dendritic spine loss deep in the neocortex and dendrite distortion with diffusion disturbances occur early in experimental pneumococcal meningitis. Front Neurosci 2023; 16:912445. [PMID: 36704002 PMCID: PMC9871924 DOI: 10.3389/fnins.2022.912445] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 12/20/2022] [Indexed: 01/11/2023] Open
Abstract
Introduction Streptococcus pneumoniae (pneumococcus) meningitis is a serious disease with substantial lethality and long-term disability in survivors. Loss of synaptic staining in the superficial layers of the neocortex in rodent models and in humans, and pneumolysin (a major pneumococcal toxin)-dependent dendritic spine collapse in brain slices have been described. It remains unclear how deep in the neocortex more discrete changes are present, how soon after disease onset these changes occur, and whether other properties of dendrites are also affected. Methods Using a mouse model of pneumococcal meningitis, we studied changes in the neocortex shortly (3-6 h) after the onset of clinical symptoms via modified Golgi-Cox silver staining. Results Dendritic changes were present in areas with otherwise unchanged cell numbers and no signs of necrosis or other apparent neuronal pathology. Mature dendritic spines were reduced in the pyramidal neurons running through layers 1-5. Additionally, spine morphology changes (swelling, spine neck distortion), were also observed in the deeper layers 4 and 5 of the neocortex. Immature spines (filopodia) remained unchanged between groups, as well as the dendritic arborization of the analyzed neurons. In a third of the animals with meningitis, massive mechanical distortion of the primary dendrites of most of the pyramidal neurons through layers 1-5 was observed. This distortion was reproduced in acute brain slices after exposure to pneumolysin-containing bacterial lysates (S. pneumoniae D39 strain), but not to lysates of pneumolysin-deficient bacteria, which we explain by the tissue remodeling effect of the toxin. Experimental mechanical dendrite distortion in primary neural cultures demonstrated diminished FRAP diffusion of neuronally-expressed enhanced green fluorescent protein (eGFP), indicative of disturbed dendritic diffusion. Discussion Our work extends earlier knowledge of synaptic loss in the superficial cortical layers during meningitis to deeper layers. These changes occurred surprisingly early in the course of the disease, substantially limiting the effective therapeutic window. Methodologically, we demonstrate that the dendritic spine collapse readout is a highly reliable and early marker of neural damage in pneumococcal meningitis models, allowing for reduction of the total number of animals used per a group due to much lower variation among animals.
Collapse
Affiliation(s)
- Dario Baronti
- Institute of Anatomy, University of Bern, Bern, Switzerland
| | - Nikola Tomov
- Institute of Anatomy, University of Bern, Bern, Switzerland
| | - Sabrina Hupp
- Institute of Anatomy, University of Bern, Bern, Switzerland
| | - Timothy J. Mitchell
- School of Immunity and Infection, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Asparouh I. Iliev
- Institute of Anatomy, University of Bern, Bern, Switzerland,*Correspondence: Asparouh I. Iliev,
| |
Collapse
|
8
|
Liu CY, Chen JB, Liu YY, Zhou XM, Zhang M, Jiang YM, Ma QY, Xue Z, Zhao ZY, Li XJ, Chen JX. Saikosaponin D exerts antidepressant effect by regulating Homer1-mGluR5 and mTOR signaling in a rat model of chronic unpredictable mild stress. Chin Med 2022; 17:60. [PMID: 35610650 PMCID: PMC9128259 DOI: 10.1186/s13020-022-00621-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 05/08/2022] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND Many studies about depression have focused on the dysfunctional synaptic signaling in the hippocampus that drives the pathophysiology of depression. Radix Bupleuri has been used in China for over 2000 years to regulate liver-qi. Extracted from Radix Bupleuri, Saikosaponin D (SSD) is a pharmacologically active substance that has antidepressant effects. However, its underlying mechanism remains unknown. MATERIALS AND METHODS A chronic unpredictable mild stress (CUMS) paradigm was used as a rat model of depression. SD rats were randomly assigned to a normal control (NC) group or one exposed to a CUMS paradigm. Of the latter group, rats were assigned to four subgroups: no treatment (CUMS), fluoxetine-treated (FLU), high-dose and low-dose SSD-treated (SSDH and SSDL). SSD was orally administrated of 1.50 mg/kg and 0.75 mg/kg/days for three weeks in the SSDH and SSDL groups, respectively. Fluoxetine was administrated at a dose of 2.0 mg/kg/days. SSD's antidepressant effects were assessed using the open field test, forced swim test, and sucrose preference test. Glutamate levels were quantified by ELISA. Western blot and immunochemical analyses were conducted to quantify proteins in the Homer protein homolog 1 (Homer1)-metabotropic glutamate receptor 5 (mGluR5) and mammalian target of rapamycin (mTOR) pathways in the hippocampal CA1 region. To measure related gene expression, RT-qPCR was employed. RESULTS CUMS-exposed rats treated with SSD exhibited increases in food intake, body weight, and improvements in the time spent in the central are and total distance traveled in the OFT, and less pronounced pleasure-deprivation behaviors. SSD also decreased glutamate levels in CA1. In CA1 region of CUMS-exposed rats, SSD treatment increased mGluR5 expression while decreasing Homer1 expression. SSD also increased expressions of postsynaptic density protein 95 (PSD95) and synapsin I (SYP), and the ratios of p-mTOR/mTOR, p-p70S6k/p70S6k, and p-4E-BP1/4E-BP1 in the CA1 region in CUMS-exposed rats. CONCLUSIONS SSD treatment reduces glutamate levels in the CA1 region and promotes the expression of the synaptic proteins PSD-95 and SYP via the regulation of the Homer1-mGluR5 and downstream mTOR signaling pathways. These findings suggest that SSD could act as a natural neuroprotective agent in the prevention of depression.
Collapse
Affiliation(s)
- Chen-Yue Liu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Jian-Bei Chen
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Yue-Yun Liu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Xue-Ming Zhou
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
- School of Basic Medical Sciences, Heilongjiang University of Chinese Medicine, Haerbin, 150040, China
| | - Man Zhang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - You-Ming Jiang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Qing-Yu Ma
- Formula-Pattern Research Center, School of Traditional Chinese Medicine, Jinan University, Guangzhou, 510632, China
| | - Zhe Xue
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Zong-Yao Zhao
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Xiao-Juan Li
- Formula-Pattern Research Center, School of Traditional Chinese Medicine, Jinan University, Guangzhou, 510632, China.
| | - Jia-Xu Chen
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China.
- Formula-Pattern Research Center, School of Traditional Chinese Medicine, Jinan University, Guangzhou, 510632, China.
| |
Collapse
|
9
|
Kivisäkk P, Carlyle BC, Sweeney T, Quinn JP, Ramirez CE, Trombetta BA, Mendes M, Brock M, Rubel C, Czerkowicz J, Graham D, Arnold SE. Increased levels of the synaptic proteins PSD-95, SNAP-25, and neurogranin in the cerebrospinal fluid of patients with Alzheimer's disease. Alzheimers Res Ther 2022; 14:58. [PMID: 35461266 PMCID: PMC9034610 DOI: 10.1186/s13195-022-01002-x] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 04/08/2022] [Indexed: 01/08/2023]
Abstract
Background There is currently a lack of reliable and easily accessible biomarkers predicting cognitive decline in Alzheimer’s disease (AD). Synaptic dysfunction and loss occur early in AD and synaptic loss measured in the brain tissue and by PET are closely linked to cognitive decline, rendering synaptic proteins a promising target for biomarker development. Methods We used novel Simoa assays to measure cerebrospinal fluid (CSF) levels of two synaptic biomarker candidates, postsynaptic density protein 95 (PSD-95/DLG4), and the presynaptically localized synaptosomal-associated protein 25 (SNAP-25), as well as neurogranin (Ng), an established postsynaptic biomarker. CSF samples from two well-characterized cohorts (n=178 and n=156) were selected from banked samples obtained from diagnostic lumbar punctures containing subjects with amyloid-ß (Aß) positive AD, subjects with non-AD neurodegenerative diseases, subjects with other neurological conditions, and healthy controls (HC). Results All subjects had detectable CSF levels of PSD-95, SNAP-25, and Ng. CSF levels of PSD-95, SNAP-25, and Ng were all correlated, with the strongest correlation between the presynaptic SNAP-25 and the postsynaptic neurogranin. AD subjects had on average higher concentrations of all three synaptic markers compared to those with non-AD neurodegenerative diseases, other neurological disorders, and HCs. Increased CSF levels of PSD-95, SNAP-25, and Ng were, however, not specific for AD and were present in sporadic cases with inflammatory or vascular disorders as well. High CSF levels of PSD-95 were also observed in a few subjects with other neurodegenerative disorders. Conclusion The data establishes PSD-95 as a promising CSF marker for neurodegenerative disease synaptic pathology, while SNAP-25 and Ng appear to be somewhat more specific for AD. Together, these synaptic markers hold promise to identify early AD pathology, to correlate with cognitive decline, and to monitor responses to disease-modifying drugs reducing synaptic degeneration. Supplementary Information The online version contains supplementary material available at 10.1186/s13195-022-01002-x.
Collapse
Affiliation(s)
- Pia Kivisäkk
- Department of Neurology, Alzheimer's Clinical and Translational Research Unit, Massachusetts General Hospital, 114 16th Street, Room 2300, Charlestown, MA, 02129, USA.
| | - Becky C Carlyle
- Department of Neurology, Alzheimer's Clinical and Translational Research Unit, Massachusetts General Hospital, 114 16th Street, Room 2300, Charlestown, MA, 02129, USA
| | - Thadryan Sweeney
- Department of Neurology, Alzheimer's Clinical and Translational Research Unit, Massachusetts General Hospital, 114 16th Street, Room 2300, Charlestown, MA, 02129, USA
| | - James P Quinn
- Department of Neurology, Alzheimer's Clinical and Translational Research Unit, Massachusetts General Hospital, 114 16th Street, Room 2300, Charlestown, MA, 02129, USA
| | - Christopher E Ramirez
- Department of Neurology, Alzheimer's Clinical and Translational Research Unit, Massachusetts General Hospital, 114 16th Street, Room 2300, Charlestown, MA, 02129, USA
| | - Bianca A Trombetta
- Department of Neurology, Alzheimer's Clinical and Translational Research Unit, Massachusetts General Hospital, 114 16th Street, Room 2300, Charlestown, MA, 02129, USA
| | | | - Mary Brock
- Quanterix Corporation, Billerica, MA, USA
| | | | | | | | - Steven E Arnold
- Department of Neurology, Alzheimer's Clinical and Translational Research Unit, Massachusetts General Hospital, 114 16th Street, Room 2300, Charlestown, MA, 02129, USA
| |
Collapse
|
10
|
Hupp S, Tomov NS, Bischoff C, Baronti D, Iliev AI. Easy to build cost-effective acute brain slice incubation system for parallel analysis of multiple treatment conditions. J Neurosci Methods 2021; 366:109405. [PMID: 34785269 DOI: 10.1016/j.jneumeth.2021.109405] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 10/25/2021] [Accepted: 11/02/2021] [Indexed: 11/27/2022]
Abstract
BACKGROUND Acute brain slices represent a powerful tool for analysis of brain function in physiology and pathology. Commercial systems and custom-build solutions with carbogen (95% O2/5% CO2) aeration, but they are expensive, have a high working volume requiring large amount of substances, and only limited options for treatment in parallel are possible. NEW METHOD We developed a novel cost-effective incubation system using materials available in every laboratory, allowing parallel incubation of several treatment conditions, thus also reducing the number of experimental animals. Our system incubation parameters were optimized for cortical neuron observation. RESULTS We tested several different options using 6, 12 or 24 standard culture well plates, combining them with cell strainer baskets inside. The system was placed in a pre-warmed incubator at 37 °C. Carbogen was injected through a 22 gauge needle, positioned between the basket and the wall of the well. Best results were achieved in a 6-well plate. In 12 and 24-well plates bubbles accumulated beneath the basket, displacing it upwards, making it unsuitable for our purposes. The gas oxygenized the medium without mechanically disturbing the slices, protected within the strainer basket, but still allowing optimal diffusion through the 100 µm pores. In a 6-well plate, six simultaneous treatments were possible in parallel. LDH/Cytotoxicity tests showed an acute toxicity of less than 7%. The system lost about 2.5% per hour of the fluid through evaporation, which was replenished every 2 h. Up to 6 h after treatment, however, this evaporation was excellently tolerated by the neurons even without fluid replenishment, most probably due to the anti-swelling effect of the mildly hypertonic medium. We performed two staining procedures, working excellently with this experimental setup, namely - a modified DiI staining and a slice silver impregnation method, both confirming the intact neuronal morphology. Preserved CA3 calcium influx and removal response following KCl depolarization confirmed the normal physiology of the pyramidal neurons 6 h after exposure in the system. COMPARISON TO EXISTING METHODS The proposed system is much cheaper than the commercial solutions, can be constructed in any lab, allows up to 6 different treatments in parallel, which none of the existing systems allows. Antibiotic presence in the incubation medium and adequate evaporation control is required if longer incubation (> 6 h) is needed. Lower incubation volumes (3-6 ml) allow sparing expensive reagents. Our procedure was optimized for cortical neurons, further fine tuning to meet other specific requirements is possible. CONCLUSIONS The system we propose allows filling the gap for budget solutions for short to mid-term incubation of acute brain slices.
Collapse
Affiliation(s)
- Sabrina Hupp
- Institute of Anatomy, University of Bern, Baltzerstrasse 2, 3012 Bern, Switzerland.
| | | | - Carolin Bischoff
- Institute of Pharmacology and Toxicology, University of Würzburg, Versbacherstrasse 9, 97073 Würzburg, Germany.
| | - Dario Baronti
- Institute of Anatomy, University of Bern, Baltzerstrasse 2, 3012 Bern, Switzerland.
| | - Asparouh I Iliev
- Institute of Anatomy, University of Bern, Baltzerstrasse 2, 3012 Bern, Switzerland.
| |
Collapse
|
11
|
Abstract
Purpose of review Community-acquired bacterial meningitis is a continually changing disease. This review summarises both dynamic epidemiology and emerging data on pathogenesis. Updated clinical guidelines are discussed, new agents undergoing clinical trials intended to reduce secondary brain damage are presented. Recent findings Conjugate vaccines are effective against serotype/serogroup-specific meningitis but vaccine escape variants are rising in prevalence. Meningitis occurs when bacteria evade mucosal and circulating immune responses and invade the brain: directly, or across the blood–brain barrier. Tissue damage is caused when host genetic susceptibility is exploited by bacterial virulence. The classical clinical triad of fever, neck stiffness and headache has poor diagnostic sensitivity, all guidelines reflect the necessity for a low index of suspicion and early Lumbar puncture. Unnecessary cranial imaging causes diagnostic delays. cerebrospinal fluid (CSF) culture and PCR are diagnostic, direct next-generation sequencing of CSF may revolutionise diagnostics. Administration of early antibiotics is essential to improve survival. Dexamethasone partially mitigates central nervous system inflammation in high-income settings. New agents in clinical trials include C5 inhibitors and daptomycin, data are expected in 2025. Summary Clinicians must remain vigilant for bacterial meningitis. Constantly changing epidemiology and emerging pathogenesis data are increasing the understanding of meningitis. Prospects for better treatments are forthcoming.
Collapse
|
12
|
Role of astroglial Connexin 43 in pneumolysin cytotoxicity and during pneumococcal meningitis. PLoS Pathog 2020; 16:e1009152. [PMID: 33370401 PMCID: PMC7793270 DOI: 10.1371/journal.ppat.1009152] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 01/08/2021] [Accepted: 11/11/2020] [Indexed: 12/19/2022] Open
Abstract
Streptococcus pneumoniae or pneumococcus (PN) is a major causative agent of bacterial meningitis with high mortality in young infants and elderly people worldwide. The mechanism underlying PN crossing of the blood brain barrier (BBB) and specifically, the role of non-endothelial cells of the neurovascular unit that control the BBB function, remains poorly understood. Here, we show that the astroglial connexin 43 (aCx43), a major gap junctional component expressed in astrocytes, plays a predominant role during PN meningitis. Following intravenous PN challenge, mice deficient for aCx43 developed milder symptoms and showed severely reduced bacterial counts in the brain. Immunofluorescence analysis of brain slices indicated that PN induces the aCx43–dependent destruction of the network of glial fibrillary acid protein (GFAP), an intermediate filament protein specifically expressed in astrocytes and up-regulated in response to brain injury. PN also induced nuclear shrinkage in astrocytes associated with the loss of BBB integrity, bacterial translocation across endothelial vessels and replication in the brain cortex. We found that aCx4-dependent astrocyte damages could be recapitulated using in vitro cultured cells upon challenge with wild-type PN but not with a ply mutant deficient for the pore-forming toxin pneumolysin (Ply). Consistently, we showed that purified Ply requires Cx43 to promote host cell plasma membrane permeabilization in a process involving the Cx43-dependent release of extracellular ATP and prolonged increase of cytosolic Ca2+ in host cells. These results point to a critical role for astrocytes during PN meningitis and suggest that the cytolytic activity of the major virulence factor Ply at concentrations relevant to bacterial infection requires co-opting of connexin plasma membrane channels. The role of non-endothelial cells constituting the neurovascular unit during infectious meningitis is poorly appreciated despite their key regulatory functions on the blood-brain barrier integrity. Here, we show that Streptococcus pneumoniae or pneumococcus, a major causative agent of bacterial meningitis, targets astroglial cells to translocate across brain endothelial vessels. We found that astroglial connexin 43, a gap junctional component, played a major role during PN meningitis in mice. PN translocation and replication in the brain cortex were associated with connexin-dependent fragmentation of astrocytic the GFAP network, a process associated with brain injury. These findings were recapitulated and extended in vitro using cultured primary astrocytes and the major PN virulence determinant Pneumolysin. Ply-mediated cytotoxicity was linked to Ca2+ increase and required aCx43, arguing against a direct toxin activity. The results reveal a key role for astroglial signaling during PN crossing of the BBB and shed light on the mechanism of Ply-mediated cytotoxicity during meningitis.
Collapse
|
13
|
Wall EC, Brownridge P, Laing G, Terra VS, Mlozowa V, Denis B, Nyirenda M, Allain T, Ramos-Sevillano E, Carrol E, Collins A, Gordon SB, Lalloo DG, Wren B, Beynon R, Heyderman RS, Brown JS. CSF Levels of Elongation Factor Tu Is Associated With Increased Mortality in Malawian Adults With Streptococcus pneumoniae Meningitis. Front Cell Infect Microbiol 2020; 10:603623. [PMID: 33363056 PMCID: PMC7759504 DOI: 10.3389/fcimb.2020.603623] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 11/10/2020] [Indexed: 12/03/2022] Open
Abstract
Background Mortality from bacterial meningitis, predominately caused by Streptococcus pneumoniae, exceeds 50% in sub-Saharan African countries with high HIV prevalence. Underlying causes of high mortality are poorly understood. We examined the host and pathogen proteome in the CSF of adults with proven pneumococcal meningitis (PM), testing if there was an association between differentially expressed proteins and outcome. Materials/Methods CSF proteomes were analyzed by quantitative Mass-Spectrometry. Spectra were identified using the Swissprot human and TIGR4 pneumococcal protein libraries. Proteins were quantitated and analyzed against mortality. Unique proteins in PM were identified against published normal CSF proteome. Random-Forest models were used to test for protein signatures discriminating outcome. Proteins of interest were tested for their effects on growth and neutrophil opsonophagocytic killing of S. pneumoniae. Results CSF proteomes were available for 57 Adults with PM (median age 32 years, 60% male, 70% HIV-1 co-infected, mortality 63%). Three hundred sixty individual human and 23 pneumococcal proteins were identified. Of the human protein hits, 30% were not expressed in normal CSF, and these were strongly associated with inflammation and primarily related to neutrophil activity. No human protein signature predicted outcome. However, expression of the essential S. pneumoniae protein Elongation Factor Tu (EF-Tu) was significantly increased in CSF of non-survivors [False Discovery Rate (q) <0.001]. Expression of EF-Tu was negatively co-correlated against expression of Neutrophil defensin (r 0.4 p p < 0.002), but not against complement proteins C3 or Factor H. In vitro, addition of EF-Tu protein impaired S. pneumoniae neutrophil killing in CSF. Conclusions Excessive S. pneumoniae EF-Tu protein in CSF was associated with reduced survival in meningitis in a high HIV prevalence population. We show EF-Tu may inhibit neutrophil mediated killing of S. pneumoniae in CSF. Further mechanistic work is required to better understand how S. pneumoniae avoids essential innate immune responses during PM through production of excess EF-Tu.
Collapse
Affiliation(s)
- Emma C. Wall
- The Francis Crick Institute, London, United Kingdom
- Division of Infection and Immunity, University College London, London, United Kingdom
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, College of Medicine, University of Malawi, Blantyre, Malawi
| | - Philip Brownridge
- Centre for Proteomics, Institute of Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| | - Gavin Laing
- Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Vanessa S. Terra
- London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Veronica Mlozowa
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, College of Medicine, University of Malawi, Blantyre, Malawi
| | - Brigitte Denis
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, College of Medicine, University of Malawi, Blantyre, Malawi
| | - Mulinda Nyirenda
- Adult Emergency Trauma Centre, Queen Elizabeth Central Hospital, Ministry of Health, Blantyre, Malawi
- College of Medicine, University of Malawi, Blantyre, Malawi
| | - Theresa Allain
- College of Medicine, University of Malawi, Blantyre, Malawi
| | - Elisa Ramos-Sevillano
- UCL Respiratory, Division of Medicine, University College London, London, United Kingdom
| | - Enitan Carrol
- Institute of Infection and Global Health, University of Liverpool, Liverpool, United Kingdom
| | - Andrea Collins
- Liverpool School of Tropical Medicine, Liverpool, United Kingdom
- Liverpool University Hospital Foundation Trust, Liverpool, United Kingdom
| | - Stephen B. Gordon
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, College of Medicine, University of Malawi, Blantyre, Malawi
- Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - David G. Lalloo
- Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Brendan Wren
- London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Robert Beynon
- Centre for Proteomics, Institute of Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| | - Robert S. Heyderman
- Division of Infection and Immunity, University College London, London, United Kingdom
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, College of Medicine, University of Malawi, Blantyre, Malawi
| | - Jeremy S. Brown
- UCL Respiratory, Division of Medicine, University College London, London, United Kingdom
| |
Collapse
|
14
|
Interaction of Macrophages and Cholesterol-Dependent Cytolysins: The Impact on Immune Response and Cellular Survival. Toxins (Basel) 2020; 12:toxins12090531. [PMID: 32825096 PMCID: PMC7551085 DOI: 10.3390/toxins12090531] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 08/13/2020] [Accepted: 08/15/2020] [Indexed: 02/07/2023] Open
Abstract
Cholesterol-dependent cytolysins (CDCs) are key virulence factors involved in many lethal bacterial infections, including pneumonia, necrotizing soft tissue infections, bacterial meningitis, and miscarriage. Host responses to these diseases involve myeloid cells, especially macrophages. Macrophages use several systems to detect and respond to cholesterol-dependent cytolysins, including membrane repair, mitogen-activated protein (MAP) kinase signaling, phagocytosis, cytokine production, and activation of the adaptive immune system. However, CDCs also promote immune evasion by silencing and/or destroying myeloid cells. While there are many common themes between the various CDCs, each CDC also possesses specific features to optimally benefit the pathogen producing it. This review highlights host responses to CDC pathogenesis with a focus on macrophages. Due to their robust plasticity, macrophages play key roles in the outcome of bacterial infections. Understanding the unique features and differences within the common theme of CDCs bolsters new tools for research and therapy.
Collapse
|
15
|
Gordon SM, Srinivasan L, Taylor DM, Master SR, Tremoglie MA, Hankeova A, Flannery DD, Abbasi S, Fitzgerald JC, Harris MC. Derivation of a metabolic signature associated with bacterial meningitis in infants. Pediatr Res 2020; 88:184-191. [PMID: 32120377 PMCID: PMC7390682 DOI: 10.1038/s41390-020-0816-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 02/03/2020] [Accepted: 02/09/2020] [Indexed: 02/01/2023]
Abstract
BACKGROUND Diagnosis of bacterial meningitis (BM) is challenging in newborn infants. Presently, biomarkers of BM have limited diagnostic accuracy. Analysis of cerebrospinal fluid (CSF) metabolites may be a useful diagnostic tool in BM. METHODS In a nested case-control study, we examined >400 metabolites in CSF of uninfected infants and infants with culture-confirmed BM using gas and liquid chromatography mass spectrometry. Preterm and full-term infants in a Level III or IV Neonatal Intensive Care Unit were prospectively enrolled when evaluated for serious bacterial infection. RESULTS Over 200 CSF metabolites significantly differed in uninfected infants and infants with BM. Using machine learning, we found that as few as 6 metabolites distinguished infants with BM from uninfected infants in this pilot cohort. Further analysis demonstrated three metabolites associated with Group B Streptococcal meningitis. CONCLUSIONS We report the first comprehensive metabolic analysis of CSF in infants with BM. In our pilot cohort, we derived a metabolic signature that predicted the presence or absence of BM, irrespective of gestational age, postnatal age, sex, race and ethnicity, presence of neurosurgical hardware, white blood cell count in CSF, and red blood cell contamination in CSF. Metabolic analysis may aid diagnosis of BM and facilitate clinical decision-making in infants. IMPACT In a pilot cohort, metabolites in cerebrospinal fluid distinguished infants with bacterial meningitis from uninfected infants.We report the first comprehensive metabolic analysis of cerebrospinal fluid in infants with bacterial meningitis.Our findings may be used to improve diagnosis of bacterial meningitis and to offer mechanistic insights into the pathophysiology of bacterial meningitis in infants.
Collapse
Affiliation(s)
- Scott M Gordon
- Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, PA, USA.
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Lakshmi Srinivasan
- Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Deanne M Taylor
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Biomedical & Health Informatics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Stephen R Master
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Divisions of Laboratory Medicine and Pathology Informatics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Marissa A Tremoglie
- Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Adriana Hankeova
- Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Dustin D Flannery
- Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- CHOP Newborn Care at Pennsylvania Hospital, Philadelphia, PA, USA
| | - Soraya Abbasi
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- CHOP Newborn Care at Pennsylvania Hospital, Philadelphia, PA, USA
| | - Julie C Fitzgerald
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Anesthesiology and Critical Care, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Mary C Harris
- Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
16
|
Shortall SE, Brown AM, Newton-Mann E, Dawe-Lane E, Evans C, Fowler M, King MV. Calbindin Deficits May Underlie Dissociable Effects of 5-HT 6 and mGlu 7 Antagonists on Glutamate and Cognition in a Dual-Hit Neurodevelopmental Model for Schizophrenia. Mol Neurobiol 2020; 57:3439-3457. [PMID: 32533466 PMCID: PMC7340678 DOI: 10.1007/s12035-020-01938-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 05/13/2020] [Indexed: 12/11/2022]
Abstract
Despite several compounds entering clinical trials for the negative and cognitive symptoms of schizophrenia, few have progressed beyond phase III. This is partly attributed to a need for improved preclinical models, to understand disease and enable predictive evaluation of novel therapeutics. To this end, one recent approach incorporates "dual-hit" neurodevelopmental insults like neonatal phencyclidine plus isolation rearing (PCP-Iso). Glutamatergic dysfunction contributes to schizophrenia pathophysiology and may represent a treatment target, so we used enzyme-based microsensors to evaluate basal- and drug-evoked glutamate release in hippocampal slices from rats that received neonatal PCP and/or isolation rearing. 5-HT6 antagonist-evoked glutamate release (thought to be mediated indirectly via GABAergic disinhibition) was reduced in PCP-Iso, as were cognitive effects of a 5-HT6 antagonist in a hippocampal glutamate-dependent novel object discrimination task. Yet mGlu7 antagonist-evoked glutamatergic and cognitive responses were spared. Immunohistochemical analyses suggest these findings (which mirror the apparent lack of clinical response to 5-HT6 antagonists in schizophrenia) are not due to reduced hippocampal 5-HT input in PCP-Iso, but may be explained by reduced calbindin expression. This calcium-binding protein is present in a subset of GABAergic interneurons receiving preferential 5-HT innervation and expressing 5-HT6 receptors. Its loss (in schizophrenia and PCP-Iso) would be expected to reduce interneuron firing and potentially prevent further 5-HT6 antagonist-mediated disinhibition, without impacting on responses of VIP-expressing interneurons to mGlu7 antagonism. This research highlights the importance of improved understanding for selection of appropriate preclinical models, especially where disease neurobiology impacts on cells mediating the effects of potential therapeutics.
Collapse
Affiliation(s)
- Sinead E Shortall
- School of Life Sciences, Medical School, Queen's Medical Centre, The University of Nottingham, Nottingham, NG7 2UH, UK
| | - Angus M Brown
- School of Life Sciences, Medical School, Queen's Medical Centre, The University of Nottingham, Nottingham, NG7 2UH, UK
| | - Eliot Newton-Mann
- School of Life Sciences, Medical School, Queen's Medical Centre, The University of Nottingham, Nottingham, NG7 2UH, UK
| | - Erin Dawe-Lane
- School of Life Sciences, Medical School, Queen's Medical Centre, The University of Nottingham, Nottingham, NG7 2UH, UK
| | - Chanelle Evans
- School of Life Sciences, Medical School, Queen's Medical Centre, The University of Nottingham, Nottingham, NG7 2UH, UK
| | - Maxine Fowler
- School of Life Sciences, Medical School, Queen's Medical Centre, The University of Nottingham, Nottingham, NG7 2UH, UK
| | - Madeleine V King
- School of Life Sciences, Medical School, Queen's Medical Centre, The University of Nottingham, Nottingham, NG7 2UH, UK.
| |
Collapse
|
17
|
Pleckaityte M. Cholesterol-Dependent Cytolysins Produced by Vaginal Bacteria: Certainties and Controversies. Front Cell Infect Microbiol 2020; 9:452. [PMID: 31998661 PMCID: PMC6966277 DOI: 10.3389/fcimb.2019.00452] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 12/12/2019] [Indexed: 01/16/2023] Open
Abstract
Bacterial vaginosis (BV) is a vaginal anaerobic dysbiosis that affects women of reproductive age worldwide. BV is microbiologically characterized by the depletion of vaginal lactobacilli and the overgrowth of anaerobic bacterial species. Accumulated evidence suggests that Gardnerella spp. have a pivotal role among BV-associated bacteria in the initiation and development of BV. However, Gardnerella spp. often colonize healthy women. Lactobacillus iners is considered as a prevalent constituent of healthy vaginal microbiota, and is abundant in BV. Gardnerella spp. and L. iners secrete the toxins vaginolysin (VLY) and inerolysin (INY), which have structural and activity features attributed to cholesterol-dependent cytolysins (CDCs). CDCs are produced by many pathogenic bacteria as virulence factors that participate in various stages of disease progression by forming lytic and non-lytic pores in cell membranes or via pore-independent pathways. VLY is expressed in the majority of Gardnerella spp. isolates; less is known about the prevalence of the gene that encodes INY. INY is a classical CDC; membrane cholesterol acts a receptor for INY. VLY uses human CD59 as its receptor, although cholesterol remains indispensable for VLY pore-forming activity. INY-induced damage of artificial membranes is directly dependent on cholesterol concentration in the bilayer, whereas VLY-induced damage occurs with high levels of membrane cholesterol (>40 mol%). VLY primarily forms membrane-embedded complete rings in the synthetic bilayer, whereas INY forms arciform structures with smaller pore sizes. VLY activity is high at elevated pH, which is characteristic of BV, whereas INY activity is high at more acidic pH, which is specific for a healthy vagina. Increased VLY levels in vaginal mucosa in vivo were associated with clinical indicators of BV. However, experimental evidence is lacking for the specific roles of VLY and INY in BV. The interplay between vaginal bacterial species affects the expression of the gene encoding VLY, thereby modulating the virulence of Gardnerella spp. This review discusses the current evidence for VLY and INY cytolysins, including their structures and activities, factors affecting their expression, and their potential impacts on the progression of anaerobic dysbiosis.
Collapse
Affiliation(s)
- Milda Pleckaityte
- Laboratory of Immunology and Cell Biology, Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| |
Collapse
|
18
|
In vivo proteomics identifies the competence regulon and AliB oligopeptide transporter as pathogenic factors in pneumococcal meningitis. PLoS Pathog 2019; 15:e1007987. [PMID: 31356624 PMCID: PMC6687184 DOI: 10.1371/journal.ppat.1007987] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Revised: 08/08/2019] [Accepted: 07/15/2019] [Indexed: 01/09/2023] Open
Abstract
Streptococcus pneumoniae (pneumococci) is a leading cause of severe bacterial meningitis in many countries worldwide. To characterize the repertoire of fitness and virulence factors predominantly expressed during meningitis we performed niche-specific analysis of the in vivo proteome in a mouse meningitis model, in which bacteria are directly inoculated into the cerebrospinal fluid (CSF) cisterna magna. We generated a comprehensive mass spectrometry (MS) spectra library enabling bacterial proteome analysis even in the presence of eukaryotic proteins. We recovered 200,000 pneumococci from CSF obtained from meningitis mice and by MS we identified 685 pneumococci proteins in samples from in vitro filter controls and 249 in CSF isolates. Strikingly, the regulatory two-component system ComDE and substrate-binding protein AliB of the oligopeptide transporter system were exclusively detected in pneumococci recovered from the CSF. In the mouse meningitis model, AliB-, ComDE-, or AliB-ComDE-deficiency resulted in attenuated meningeal inflammation and disease severity when compared to wild-type pneumococci indicating the crucial role of ComDE and AliB in pneumococcal meningitis. In conclusion, we show here mechanisms of pneumococcal adaptation to a defined host compartment by a proteome-based approach. Further, this study provides the basis of a promising strategy for the identification of protein antigens critical for invasive disease caused by pneumococci and other meningeal pathogens. Pneumococci are one of the most common and aggressive meningitis pathogens associated with mortality rates between 10% and 30%. Due to severe complications during therapeutic intervention, prevention strategies to combat pneumococcal meningitis (PM) are preferred. The vaccines available are so far suboptimal and inefficient to prevent serious PM. Hence, deciphering the mechanisms employed by pneumococci to encounter and survive in the cerebrospinal fluid (CSF) will pave the way for the development of new antimicrobial strategies. This work used an in vivo proteome-based approach to identify pneumococcal proteins expressed in the CSF during acute meningitis. This strategy identified a nutrient uptake system and regulatory system to be highly expressed in the CSF and being crucial for PM. Knocking out two of the highly in vivo expressed proteins (AliA and ComDE) in S. pneumoniae yields to a significant increase in survival and decrease in pathogen burden of infected mice. These host compartment specific expressed pneumococcal antigens represent promising candidates for antimicrobials or protein-based vaccines.
Collapse
|
19
|
Hupp S, Grandgirard D, Mitchell TJ, Leib SL, Hathaway LJ, Iliev AI. Pneumolysin and the bacterial capsule of Streptococcus pneumoniae cooperatively inhibit taxis and motility of microglia. J Neuroinflammation 2019; 16:105. [PMID: 31103037 PMCID: PMC6525981 DOI: 10.1186/s12974-019-1491-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Accepted: 04/30/2019] [Indexed: 01/11/2023] Open
Abstract
Background Streptococcus pneumoniae is the cause of a highly lethal form of meningitis in humans. Microglial cells in the brain represent the first line of defense against pathogens, and they participate in the inflammatory response. The cholesterol-dependent cytolysin pneumolysin and the bacterial capsule are key pathogenic factors, known to exacerbate the course of pneumococcal meningitis. Methods We utilized live imaging and immunostaining of glial cells in dissociated and acute brain slice cultures to study the effect of pneumococcal factors, including the cholesterol-dependent cytolysin pneumolysin and the pneumococcal capsule, on microglial motility and taxis. Results In brain tissue, primary microglia cells showed an enhanced response towards lysates from bacteria lacking capsules and pneumolysin as they moved rapidly to areas with an abundance of bacterial factors. The presence of bacterial capsules and pneumolysin cumulatively inhibited microglial taxis. In mixed cultures of astrocytes and microglia, the motility of microglia was inhibited by capsular components within minutes after exposure. The reduced motility was partially reversed by mannan, a mannose receptor inhibitor. The effects on microglia were not mediated by astrocytes because pure microglial cells responded to various pneumococcal lysates similarly with distinct cell shape changes as seen in mixed cultures. Conclusions Our data indicate that microglia possess the capacity for a very agile response towards bacterial pathogens, but key pathogenic factors, such as pneumococcal capsules and pneumolysin, inhibited this response shortly after a bacterial challenge. Furthermore, we demonstrate for the first time that the bacterial capsule affects cellular behaviors such as motility and taxis. Electronic supplementary material The online version of this article (10.1186/s12974-019-1491-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sabrina Hupp
- Institute of Anatomy, University of Bern, Baltzerstrasse 2, 3012, Bern, Switzerland
| | - Denis Grandgirard
- Institute for Infectious Diseases, University of Bern, Friedbühlstrasse 51, 3010, Bern, Switzerland
| | - Timothy J Mitchell
- Institute of Microbiology and Infection, College of Medical and Dental Sciences, Biosciences Building, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Stephen L Leib
- Institute for Infectious Diseases, University of Bern, Friedbühlstrasse 51, 3010, Bern, Switzerland
| | - Lucy J Hathaway
- Institute for Infectious Diseases, University of Bern, Friedbühlstrasse 51, 3010, Bern, Switzerland
| | - Asparouh I Iliev
- Institute of Anatomy, University of Bern, Baltzerstrasse 2, 3012, Bern, Switzerland.
| |
Collapse
|
20
|
Zhang J, Wang Y, Zheng Z, Sun X, Chen T, Li C, Zhang X, Guo J. Intracellular ion and protein nanoparticle-induced osmotic pressure modify astrocyte swelling and brain edema in response to glutamate stimuli. Redox Biol 2019; 21:101112. [PMID: 30685709 PMCID: PMC6351271 DOI: 10.1016/j.redox.2019.101112] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 01/02/2019] [Accepted: 01/14/2019] [Indexed: 12/28/2022] Open
Abstract
Intracellular tension activity plays a crucial role in cytotoxic brain edema and astrocyte swelling. Here, a few genetically encoded FRET-based tension probes were designed to detect cytoskeletal structural tension optically, including their magnitude and vectors. The astrocyte swelling resulted in GFAP tension increment, which is associated with the antagonistic effect of inward microfilaments (MFs) and microtubules (MTs) forces. In glutamate-induced astrocyte swelling, GFAP tension rise resulted from outward ion and protein nanoparticle-induced osmotic pressure (PN-OP) increases, where PN-OP could be elicited by MF and MT depolymerization, protein nanoparticle production, and activation of cofilin and stathmin-1. Attenuation of both ion osmotic pressure and PN-OP by drug combinations, together with free-radical scavenger, relieved cerebral edema in vivo. The study suggests that intracellular osmotic pressure (especially PN-OP) has a pivotal role in glutamate-induced astrocyte swelling and brain edema. Recovery of cytoplasmic potential is a promising target to develop new drugs and cure brain edema.
Collapse
Affiliation(s)
- JiaRui Zhang
- State Key Laboratory Cultivation Base For TCM Quality and Efficacy, School of Medicine and Life Science, Nanjing University of Chinese Medicine, Nanjing 210023, PR China; Key Laboratory of Drug Target and Drug for Degenerative Disease, Nanjing University of Chinese Medicine, Nanjing, PR China
| | - YuXuan Wang
- State Key Laboratory Cultivation Base For TCM Quality and Efficacy, School of Medicine and Life Science, Nanjing University of Chinese Medicine, Nanjing 210023, PR China; Key Laboratory of Drug Target and Drug for Degenerative Disease, Nanjing University of Chinese Medicine, Nanjing, PR China
| | - ZiHui Zheng
- State Key Laboratory Cultivation Base For TCM Quality and Efficacy, School of Medicine and Life Science, Nanjing University of Chinese Medicine, Nanjing 210023, PR China; Key Laboratory of Drug Target and Drug for Degenerative Disease, Nanjing University of Chinese Medicine, Nanjing, PR China
| | - XiaoHe Sun
- State Key Laboratory Cultivation Base For TCM Quality and Efficacy, School of Medicine and Life Science, Nanjing University of Chinese Medicine, Nanjing 210023, PR China; Key Laboratory of Drug Target and Drug for Degenerative Disease, Nanjing University of Chinese Medicine, Nanjing, PR China
| | - TingTing Chen
- State Key Laboratory Cultivation Base For TCM Quality and Efficacy, School of Medicine and Life Science, Nanjing University of Chinese Medicine, Nanjing 210023, PR China; Key Laboratory of Drug Target and Drug for Degenerative Disease, Nanjing University of Chinese Medicine, Nanjing, PR China
| | - Chen Li
- State Key Laboratory Cultivation Base For TCM Quality and Efficacy, School of Medicine and Life Science, Nanjing University of Chinese Medicine, Nanjing 210023, PR China; Key Laboratory of Drug Target and Drug for Degenerative Disease, Nanjing University of Chinese Medicine, Nanjing, PR China
| | - XiaoLong Zhang
- State Key Laboratory Cultivation Base For TCM Quality and Efficacy, School of Medicine and Life Science, Nanjing University of Chinese Medicine, Nanjing 210023, PR China; Key Laboratory of Drug Target and Drug for Degenerative Disease, Nanjing University of Chinese Medicine, Nanjing, PR China
| | - Jun Guo
- State Key Laboratory Cultivation Base For TCM Quality and Efficacy, School of Medicine and Life Science, Nanjing University of Chinese Medicine, Nanjing 210023, PR China; Key Laboratory of Drug Target and Drug for Degenerative Disease, Nanjing University of Chinese Medicine, Nanjing, PR China; Jiangsu Key Laboratory of Pediatric Respiratory Disease, Institute of Pediatrics, Nanjing University of Chinese Medicine, Nanjing, PR China.
| |
Collapse
|
21
|
Characterization of Brain Dysfunction Induced by Bacterial Lipopeptides That Alter Neuronal Activity and Network in Rodent Brains. J Neurosci 2018; 38:10672-10691. [PMID: 30381406 DOI: 10.1523/jneurosci.0825-17.2018] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Revised: 08/24/2018] [Accepted: 09/15/2018] [Indexed: 12/11/2022] Open
Abstract
The immunopathological states of the brain induced by bacterial lipoproteins have been well characterized by using biochemical and histological assays. However, these studies have limitations in determining functional states of damaged brains involving aberrant synaptic activity and network, which makes it difficult to diagnose brain disorders during bacterial infection. To address this, we investigated the effect of Pam3CSK4 (PAM), a synthetic bacterial lipopeptide, on synaptic dysfunction of female mice brains and cultured neurons in parallel. Our functional brain imaging using PET with [18F]fluorodeoxyglucose and [18F] flumazenil revealed that the brain dysfunction induced by PAM is closely aligned to disruption of neurotransmitter-related neuronal activity and functional correlation in the region of the limbic system rather than to decrease of metabolic activity of neurons in the injection area. This finding was verified by in vivo tissue experiments that analyzed synaptic and dendritic alterations in the regions where PET imaging showed abnormal neuronal activity and network. Recording of synaptic activity also revealed that PAM reorganized synaptic distribution and decreased synaptic plasticity in hippocampus. Further study using in vitro neuron cultures demonstrated that PAM decreased the number of presynapses and the frequency of miniature EPSCs, which suggests PAM disrupts neuronal function by damaging presynapses exclusively. We also showed that PAM caused aggregation of synapses around dendrites, which may have caused no significant change in expression level of synaptic proteins, whereas synaptic number and function were impaired by PAM. Our findings could provide a useful guide for diagnosis and treatment of brain disorders specific to bacterial infection.SIGNIFICANCE STATEMENT It is challenging to diagnose brain disorders caused by bacterial infection because neural damage induced by bacterial products involves nonspecific neurological symptoms, which is rarely detected by laboratory tests with low spatiotemporal resolution. To better understand brain pathology, it is essential to detect functional abnormalities of brain over time. To this end, we investigated characteristic patterns of altered neuronal integrity and functional correlation between various regions in mice brains injected with bacterial lipopeptides using PET with a goal to apply new findings to diagnosis of brain disorder specific to bacterial infection. In addition, we analyzed altered synaptic density and function using both in vivo and in vitro experimental models to understand how bacterial lipopeptides impair brain function and network.
Collapse
|
22
|
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
|
23
|
Hupp S, Ribes S, Seele J, Bischoff C, Förtsch C, Maier E, Benz R, Mitchell TJ, Nau R, Iliev AI. Magnesium therapy improves outcome in Streptococcus pneumoniae meningitis by altering pneumolysin pore formation. Br J Pharmacol 2017; 174:4295-4307. [PMID: 28888095 DOI: 10.1111/bph.14027] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Revised: 08/26/2017] [Accepted: 08/29/2017] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND AND PURPOSE Streptococcus pneumoniae is the most common cause of bacterial meningitis in adults and is characterized by high lethality and substantial cognitive disabilities in survivors. Here, we have studied the capacity of an established therapeutic agent, magnesium, to improve survival in pneumococcal meningitis by modulating the neurological effects of the major pneumococcal pathogenic factor, pneumolysin. EXPERIMENTAL APPROACH We used mixed primary glial and acute brain slice cultures, pneumolysin injection in infant rats, a mouse meningitis model and complementary approaches such as Western blot, a black lipid bilayer conductance assay and live imaging of primary glial cells. KEY RESULTS Treatment with therapeutic concentrations of magnesium chloride (500 mg·kg-1 in animals and 2 mM in cultures) prevented pneumolysin-induced brain swelling and tissue remodelling both in brain slices and in animal models. In contrast to other divalent ions, which diminish the membrane binding of pneumolysin in non-therapeutic concentrations, magnesium delayed toxin-driven pore formation without affecting its membrane binding or the conductance profile of its pores. Finally, magnesium prolonged the survival and improved clinical condition of mice with pneumococcal meningitis, in the absence of antibiotic treatment. CONCLUSIONS AND IMPLICATIONS Magnesium is a well-established and safe therapeutic agent that has demonstrated capacity for attenuating pneumolysin-triggered pathogenic effects on the brain. The improved animal survival and clinical condition in the meningitis model identifies magnesium as a promising candidate for adjunctive treatment of pneumococcal meningitis, together with antibiotic therapy.
Collapse
Affiliation(s)
- Sabrina Hupp
- Institute of Anatomy, University of Bern, Bern, Switzerland.,DFG Membrane/Cytoskeleton Interaction Group, Institute of Pharmacology and Toxicology & Rudolf Virchow Center for Experimental Medicine, University of Würzburg, Würzburg, Germany
| | - Sandra Ribes
- Department of Neuropathology, University Medical Center Göttingen, Göttingen, Germany.,Department of Geriatrics, Evangelisches Krankenhaus Göttingen-Weende, Göttingen, Germany
| | - Jana Seele
- Department of Neuropathology, University Medical Center Göttingen, Göttingen, Germany.,Department of Geriatrics, Evangelisches Krankenhaus Göttingen-Weende, Göttingen, Germany
| | - Carolin Bischoff
- DFG Membrane/Cytoskeleton Interaction Group, Institute of Pharmacology and Toxicology & Rudolf Virchow Center for Experimental Medicine, University of Würzburg, Würzburg, Germany
| | - Christina Förtsch
- DFG Membrane/Cytoskeleton Interaction Group, Institute of Pharmacology and Toxicology & Rudolf Virchow Center for Experimental Medicine, University of Würzburg, Würzburg, Germany
| | - Elke Maier
- Rudolf Virchow Center for Experimental Medicine, University of Würzburg, Würzburg, Germany
| | - Roland Benz
- Rudolf Virchow Center for Experimental Medicine, University of Würzburg, Würzburg, Germany
| | - Timothy J Mitchell
- Institute of Microbiology and Infection, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Roland Nau
- Department of Neuropathology, University Medical Center Göttingen, Göttingen, Germany.,Department of Geriatrics, Evangelisches Krankenhaus Göttingen-Weende, Göttingen, Germany
| | - Asparouh I Iliev
- Institute of Anatomy, University of Bern, Bern, Switzerland.,DFG Membrane/Cytoskeleton Interaction Group, Institute of Pharmacology and Toxicology & Rudolf Virchow Center for Experimental Medicine, University of Würzburg, Würzburg, Germany
| |
Collapse
|
24
|
Tankovic J, Timinskas A, Janulaitiene M, Zilnyte M, Baudel JL, Maury E, Zvirbliene A, Pleckaityte M. Gardnerella vaginalis bacteremia associated with severe acute encephalopathy in a young female patient. Anaerobe 2017; 47:132-134. [PMID: 28546029 DOI: 10.1016/j.anaerobe.2017.05.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 05/18/2017] [Accepted: 05/20/2017] [Indexed: 10/19/2022]
Abstract
Gardnerella vaginalis is a facultative anaerobic bacterium that inhabits the genitourinary tract of both healthy women and those with bacterial vaginosis. We report a case of G. vaginalis bacteremia associated with severe toxic encephalopathy in a young woman. Anaerobic blood cultures yielded pure growth of small gram-variable rods later identified as G. vaginalis by both rapid biochemical tests and 16S rRNA gene sequencing. The patient recovered after treatment with amoxicillin-clavulanate according to the in vitro susceptibility testing. The complete genome of G. vaginalis isolate from blood cultures was determined. In vitro G. vaginalis isolate produced elevated amounts of a pore-forming toxin vaginolysin compared to control G. vaginalis isolates. We hypothesize that this toxin, if produced in high amounts in blood, is able to disrupt the blood-brain barrier and exert a toxic activity on brain cells.
Collapse
Affiliation(s)
- Jacques Tankovic
- Service de Bactériologie, Assistance Publique - Hôpitaux de Paris, Hôpital Saint-Antoine, 184 rue du Faubourg Saint-Antoine, Paris, France; Université Pierre et Marie Curie, 4 Place Jussieu, Paris, France.
| | - Albertas Timinskas
- Institute of Biotechnology, Vilnius University, Sauletekio 7, Vilnius LT-10257, Lithuania.
| | - Migle Janulaitiene
- Institute of Biotechnology, Vilnius University, Sauletekio 7, Vilnius LT-10257, Lithuania.
| | - Milda Zilnyte
- Institute of Biotechnology, Vilnius University, Sauletekio 7, Vilnius LT-10257, Lithuania.
| | - Jean-Luc Baudel
- Université Pierre et Marie Curie, 4 Place Jussieu, Paris, France; Service de Réanimation Médicale, Assistance Publique - Hôpitaux de Paris, Hôpital Saint-Antoine, 184 rue du Faubourg Saint-Antoine, Paris, France.
| | - Eric Maury
- Université Pierre et Marie Curie, 4 Place Jussieu, Paris, France; Service de Réanimation Médicale, Assistance Publique - Hôpitaux de Paris, Hôpital Saint-Antoine, 184 rue du Faubourg Saint-Antoine, Paris, France.
| | - Aurelija Zvirbliene
- Institute of Biotechnology, Vilnius University, Sauletekio 7, Vilnius LT-10257, Lithuania.
| | - Milda Pleckaityte
- Institute of Biotechnology, Vilnius University, Sauletekio 7, Vilnius LT-10257, Lithuania.
| |
Collapse
|
25
|
Klein RS, Garber C, Howard N. Infectious immunity in the central nervous system and brain function. Nat Immunol 2017; 18:132-141. [PMID: 28092376 DOI: 10.1038/ni.3656] [Citation(s) in RCA: 154] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Accepted: 12/02/2016] [Indexed: 11/09/2022]
Abstract
Inflammation is emerging as a critical mechanism underlying neurological disorders of various etiologies, yet its role in altering brain function as a consequence of neuroinfectious disease remains unclear. Although acute alterations in mental status due to inflammation are a hallmark of central nervous system (CNS) infections with neurotropic pathogens, post-infectious neurologic dysfunction has traditionally been attributed to irreversible damage caused by the pathogens themselves. More recently, studies indicate that pathogen eradication within the CNS may require immune responses that interfere with neural cell function and communication without affecting their survival. In this Review we explore inflammatory processes underlying neurological impairments caused by CNS infection and discuss their potential links to established mechanisms of psychiatric and neurodegenerative diseases.
Collapse
Affiliation(s)
- Robyn S Klein
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA.,Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA.,Department of Neuroscience, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Charise Garber
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Nicole Howard
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA.,Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
26
|
Maurer J, Hupp S, Bischoff C, Foertsch C, Mitchell TJ, Chakraborty T, Iliev AI. Distinct Neurotoxicity Profile of Listeriolysin O from Listeria monocytogenes. Toxins (Basel) 2017; 9:toxins9010034. [PMID: 28098781 PMCID: PMC5308266 DOI: 10.3390/toxins9010034] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 01/09/2017] [Accepted: 01/10/2017] [Indexed: 11/16/2022] Open
Abstract
Cholesterol-dependent cytolysins (CDCs) are protein toxins that originate from Gram-positive bacteria and contribute substantially to their pathogenicity. CDCs bind membrane cholesterol and build prepores and lytic pores. Some effects of the toxins are observed in non-lytic concentrations. Two pathogens, Streptococcus pneumoniae and Listeria monocytogenes, cause fatal bacterial meningitis, and both produce toxins of the CDC family-pneumolysin and listeriolysin O, respectively. It has been demonstrated that pneumolysin produces dendritic varicosities (dendrite swellings) and dendritic spine collapse in the mouse neocortex, followed by synaptic loss and astrocyte cell shape remodeling without elevated cell death. We utilized primary glial cultures and acute mouse brain slices to examine the neuropathological effects of listeriolysin O and to compare it to pneumolysin with identical hemolytic activity. In cultures, listeriolysin O permeabilized cells slower than pneumolysin did but still initiated non-lytic astrocytic cell shape changes, just as pneumolysin did. In an acute brain slice culture system, listeriolysin O produced dendritic varicosities in an NMDA-dependent manner but failed to cause dendritic spine collapse and cortical astrocyte reorganization. Thus, listeriolysin O demonstrated slower cell permeabilization and milder glial cell remodeling ability than did pneumolysin and lacked dendritic spine collapse capacity but exhibited equivalent dendritic pathology.
Collapse
Affiliation(s)
- Jana Maurer
- DFG Membrane/Cytoskeleton Interaction Group, Institute of Pharmacology and Toxicology & Rudolf Virchow Center for Experimental Biomedical Science, University of Würzburg, Versbacherstr. 9, 97078 Würzburg, Germany.
- Institute of Physiology and Pathophysiology, Heidelberg University, Im Neuenheimer Feld 326, 69120 Heidelberg, Germany.
| | - Sabrina Hupp
- DFG Membrane/Cytoskeleton Interaction Group, Institute of Pharmacology and Toxicology & Rudolf Virchow Center for Experimental Biomedical Science, University of Würzburg, Versbacherstr. 9, 97078 Würzburg, Germany.
- Institute of Anatomy, University of Bern, Baltzerstrasse 2, 3012 Bern, Switzerland.
| | - Carolin Bischoff
- DFG Membrane/Cytoskeleton Interaction Group, Institute of Pharmacology and Toxicology & Rudolf Virchow Center for Experimental Biomedical Science, University of Würzburg, Versbacherstr. 9, 97078 Würzburg, Germany.
| | - Christina Foertsch
- DFG Membrane/Cytoskeleton Interaction Group, Institute of Pharmacology and Toxicology & Rudolf Virchow Center for Experimental Biomedical Science, University of Würzburg, Versbacherstr. 9, 97078 Würzburg, Germany.
| | - Timothy J Mitchell
- Chair of Microbial Infection and Immunity, Institute of Microbiology and Infection, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK.
| | - Trinad Chakraborty
- Institute for Medical Microbiology, University of Giessen, Schubertstr. 81, 35392 Giessen, Germany.
| | - Asparouh I Iliev
- DFG Membrane/Cytoskeleton Interaction Group, Institute of Pharmacology and Toxicology & Rudolf Virchow Center for Experimental Biomedical Science, University of Würzburg, Versbacherstr. 9, 97078 Würzburg, Germany.
- Institute of Anatomy, University of Bern, Baltzerstrasse 2, 3012 Bern, Switzerland.
| |
Collapse
|
27
|
Valenzuela-Miranda D, Gallardo-Escárate C. Novel insights into the response of Atlantic salmon (Salmo salar) to Piscirickettsia salmonis: Interplay of coding genes and lncRNAs during bacterial infection. FISH & SHELLFISH IMMUNOLOGY 2016; 59:427-438. [PMID: 27818337 DOI: 10.1016/j.fsi.2016.11.001] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2016] [Revised: 10/11/2016] [Accepted: 11/02/2016] [Indexed: 06/06/2023]
Abstract
Despite the high prevalence and impact to Chilean salmon aquaculture of the intracellular bacterium Piscirickettsia salmonis, the molecular underpinnings of host-pathogen interactions remain unclear. Herein, the interplay of coding and non-coding transcripts has been proposed as a key mechanism involved in immune response. Therefore, the aim of this study was to evidence how coding and non-coding transcripts are modulated during the infection process of Atlantic salmon with P. salmonis. For this, RNA-seq was conducted in brain, spleen, and head kidney samples, revealing different transcriptional profiles according to bacterial load. Additionally, while most of the regulated genes annotated for diverse biological processes during infection, a common response associated with clathrin-mediated endocytosis and iron homeostasis was present in all tissues. Interestingly, while endocytosis-promoting factors and clathrin inductions were upregulated, endocytic receptors were mainly downregulated. Furthermore, the regulation of genes related to iron homeostasis suggested an intracellular accumulation of iron, a process in which heme biosynthesis/degradation pathways might play an important role. Regarding the non-coding response, 918 putative long non-coding RNAs were identified, where 425 were newly characterized for S. salar. Finally, co-localization and co-expression analyses revealed a strong correlation between the modulations of long non-coding RNAs and genes associated with endocytosis and iron homeostasis. These results represent the first comprehensive study of putative interplaying mechanisms of coding and non-coding RNAs during bacterial infection in salmonids.
Collapse
Affiliation(s)
- Diego Valenzuela-Miranda
- Laboratory of Biotechnology and Aquatic Genomics, Interdisciplinary Center for Aquaculture Research (INCAR), University of Concepción, P. O. Box 160-C, Concepción, Chile
| | - Cristian Gallardo-Escárate
- Laboratory of Biotechnology and Aquatic Genomics, Interdisciplinary Center for Aquaculture Research (INCAR), University of Concepción, P. O. Box 160-C, Concepción, Chile.
| |
Collapse
|
28
|
van de Beek D, Brouwer M, Hasbun R, Koedel U, Whitney CG, Wijdicks E. Community-acquired bacterial meningitis. Nat Rev Dis Primers 2016; 2:16074. [PMID: 27808261 DOI: 10.1038/nrdp.2016.74] [Citation(s) in RCA: 183] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Meningitis is an inflammation of the meninges and subarachnoid space that can also involve the brain cortex and parenchyma. It can be acquired spontaneously in the community - community-acquired bacterial meningitis - or in the hospital as a complication of invasive procedures or head trauma (nosocomial bacterial meningitis). Despite advances in treatment and vaccinations, community-acquired bacterial meningitis remains one of the most important infectious diseases worldwide. Streptococcus pneumoniae and Neisseria meningitidis are the most common causative bacteria and are associated with high mortality and morbidity; vaccines targeting these organisms, which have designs similar to the successful vaccine that targets Haemophilus influenzae type b meningitis, are now being used in many routine vaccination programmes. Experimental and genetic association studies have increased our knowledge about the pathogenesis of bacterial meningitis. Early antibiotic treatment improves the outcome, but the growing emergence of drug resistance as well as shifts in the distribution of serotypes and groups are fuelling further development of new vaccines and treatment strategies. Corticosteroids were found to be beneficial in high-income countries depending on the bacterial species. Further improvements in the outcome are likely to come from dampening the host inflammatory response and implementing preventive measures, especially the development of new vaccines.
Collapse
Affiliation(s)
- Diederik van de Beek
- Department of Neurology, Center of Infection and Immunity Amsterdam (CINIMA), Academic Medical Center, P.O. BOX 22660, 1100DD Amsterdam, The Netherlands
| | - Matthijs Brouwer
- Department of Neurology, Center of Infection and Immunity Amsterdam (CINIMA), Academic Medical Center, P.O. BOX 22660, 1100DD Amsterdam, The Netherlands
| | - Rodrigo Hasbun
- Department of Internal Medicine, UT Health McGovern Medical School, Houston, Texas, USA
| | - Uwe Koedel
- Department of Neurology, Clinic Grosshadern of the Ludwig-Maximilians University of Munich, Munich, Germany
| | - Cynthia G Whitney
- Respiratory Diseases Branch, Division of Bacterial Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Eelco Wijdicks
- Division of Critical Care Neurology, Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
29
|
Selvan LDN, Sreenivasamurthy SK, Kumar S, Yelamanchi SD, Madugundu AK, Anil AK, Renuse S, Nair BG, Gowda H, Mathur PP, Satishchandra P, Shankar SK, Mahadevan A, Keshava Prasad TS. Characterization of host response to Cryptococcus neoformans through quantitative proteomic analysis of cryptococcal meningitis co-infected with HIV. MOLECULAR BIOSYSTEMS 2016; 11:2529-40. [PMID: 26181685 DOI: 10.1039/c5mb00187k] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Cryptococcal meningitis is the most common opportunistic fungal infection causing morbidity and mortality (>60%) in HIV-associated immunocompromised individuals caused by Cryptococcus neoformans. Molecular mechanisms of cryptococcal infection in brain have been studied using experimental animal models and cell lines. There are limited studies for the molecular understanding of cryptococcal meningitis in human brain. The proteins involved in the process of invasion and infection in human brain still remains obscure. To this end we carried out mass spectrometry-based quantitative proteomics of frontal lobe brain tissues from cryptococcal meningitis patients and controls to identify host proteins that are associated with the pathogenesis of cryptococcal meningitis. We identified 317 proteins to be differentially expressed (≥2-fold) from a total of 3423 human proteins. We found proteins involved in immune response and signal transduction to be differentially expressed in response to cryptococcal infection in human brain. Immune response proteins including complement factors, major histocompatibility proteins, proteins previously known to be involved in fungal invasion to brain such as caveolin 1 and actin were identified to be differentially expressed in cryptococcal meningitis brain tissues co-infected with HIV. We also validated the expression status of 5 proteins using immunohistochemistry. Overexpression of major histocompatibility complexes, class I, B (HLA-B), actin alpha 2 smooth muscle aorta (ACTA2) and caveolin 1 (CAV1) and downregulation of peripheral myelin protein 2 (PMP2) and alpha crystallin B chain (CRYAB) in cryptococcal meningitis were confirmed by IHC-based validation experiments. This study provides the brain proteome profile of cryptococcal meningitis co-infected with HIV for a better understanding of the host response associated with the disease.
Collapse
|
30
|
Doran KS, Fulde M, Gratz N, Kim BJ, Nau R, Prasadarao N, Schubert-Unkmeir A, Tuomanen EI, Valentin-Weigand P. Host-pathogen interactions in bacterial meningitis. Acta Neuropathol 2016; 131:185-209. [PMID: 26744349 PMCID: PMC4713723 DOI: 10.1007/s00401-015-1531-z] [Citation(s) in RCA: 136] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Revised: 12/21/2015] [Accepted: 12/22/2015] [Indexed: 12/26/2022]
Abstract
Bacterial meningitis is a devastating disease occurring worldwide with up to half of the survivors left with permanent neurological sequelae. Due to intrinsic properties of the meningeal pathogens and the host responses they induce, infection can cause relatively specific lesions and clinical syndromes that result from interference with the function of the affected nervous system tissue. Pathogenesis is based on complex host–pathogen interactions, some of which are specific for certain bacteria, whereas others are shared among different pathogens. In this review, we summarize the recent progress made in understanding the molecular and cellular events involved in these interactions. We focus on selected major pathogens, Streptococcus pneumonia, S. agalactiae (Group B Streptococcus), Neisseria meningitidis, and Escherichia coli K1, and also include a neglected zoonotic pathogen, Streptococcus suis. These neuroinvasive pathogens represent common themes of host–pathogen interactions, such as colonization and invasion of mucosal barriers, survival in the blood stream, entry into the central nervous system by translocation of the blood–brain and blood–cerebrospinal fluid barrier, and induction of meningeal inflammation, affecting pia mater, the arachnoid and subarachnoid spaces.
Collapse
|
31
|
Almirón MA, Goldschmidt E, Bertelli AM, Gomez MI, Argibay P, Sanjuan NA. In Vitroinfection of human dura-mater fibroblasts withStaphylococcus aureus: colonization and reactive production of IL-1beta. Neurol Res 2015; 37:867-73. [DOI: 10.1179/1743132815y.0000000065] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
|
32
|
Wall EC, Gritzfeld JF, Scarborough M, Ajdukiewicz KMB, Mukaka M, Corless C, Lalloo DG, Gordon SB. Genomic pneumococcal load and CSF cytokines are not related to outcome in Malawian adults with meningitis. J Infect 2014; 69:440-6. [PMID: 24975177 PMCID: PMC4209731 DOI: 10.1016/j.jinf.2014.06.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2013] [Revised: 06/09/2014] [Accepted: 06/14/2014] [Indexed: 01/02/2023]
Abstract
Objective Bacterial meningitis in sub-Saharan Africa is predominantly caused by Streptococcus pneumoniae, is often associated with HIV co-infection and mortality rates are double those seen in better resourced settings. Methods To investigate the cause of this excessive mortality we quantified the pneumococcal DNA load and six common pro-inflammatory cytokines in the cerebrospinal fluid (CSF) of Malawian adults with culture proven pneumococcal meningitis and correlated the results to clinical parameters and outcome. There are currently no published data relating bacterial load to outcome in adults with pneumococcal meningitis. Results The mean age of patients was 32 years, 82% were HIV infected and 49% had died by day 40. CSF bacterial loads were high (median 6.5 × 105 copies/ml CSF) and there was no significant variation in bacterial load between survivors and non-survivors. All pro-inflammatory CSF cytokines were elevated in the CSF, with no clinically important differences between survivors and non-survivors. HIV status did not affect the CSF bacterial load or cytokine response. Conclusion Mortality from pneumococcal meningitis in adults in sub-Saharan Africa is not related to pneumococcal bacterial load. More research is needed to understand the very high mortality from meningitis in this region. Pneumococcal meningitis in Malawian adults has a very high mortality rate. Bacterial load and cytokine response may predict outcome in children. Pneumococcal load and cytokine response were not associated with outcome in adults. Low CSF white cell count and Glasgow coma score were associated with mortality. Anti-cytokine therapies are unlikely to be effective in bacterial meningitis.
Collapse
Affiliation(s)
- Emma C Wall
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, Blantyre, Malawi; Liverpool School of Tropical Medicine, Liverpool, UK; College of Medicine, Department of Medicine, University of Malawi, Malawi.
| | | | | | | | - Mavuto Mukaka
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, Blantyre, Malawi
| | | | - David G Lalloo
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, Blantyre, Malawi; Liverpool School of Tropical Medicine, Liverpool, UK
| | - Stephen B Gordon
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, Blantyre, Malawi; Liverpool School of Tropical Medicine, Liverpool, UK
| |
Collapse
|
33
|
Abstract
Abstract
Collapse
|