1
|
Yang Z, Chen J, Zhang C, Peng H. Pathological mechanisms of glial cell activation and neurodegenerative and neuropsychiatric disorders caused by Toxoplasma gondii infection. Front Microbiol 2024; 15:1512233. [PMID: 39723133 PMCID: PMC11668811 DOI: 10.3389/fmicb.2024.1512233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 11/26/2024] [Indexed: 12/28/2024] Open
Abstract
Toxoplasma gondii is an intracellular opportunistic parasite that exists in a latent form within the human central nervous system (CNS), even in immune-competent hosts. During acute infection, T. gondii traverses the blood-brain barrier (BBB). In the subsequent chronic infection phase, the infiltration of immune cells into the brain, driven by T. gondii infection and the formation of parasitic cysts, leads to persistent activation and proliferation of astrocytes and microglia. This process results in neuronal damages that are fatal in some cases. Through inducing systemic immune responses, T. gondii infection can dramatically alter the behavior of rodents and increase the risk of various neuropsychiatric disorders in humans. In this review, we explore some recent research progress on the major events involved in BBB disruption, glial cell activation and neuronal damage following T. gondii infection in hosts. It further discusses potential pathological mechanisms and the feasible treatment approaches for the neurodegenerative and neuropsychiatric disorders caused by T. gondii infection to extend our understanding for pathogenesis and preventive control of toxoplasmosis in humans.
Collapse
Affiliation(s)
| | | | | | - Hongjuan Peng
- Department of Pathogen Biology, Guangdong Provincial Key Laboratory of Tropical Diseases Research, School of Public Health, Key Laboratory of Infectious Diseases Research in South China (Southern Medical University), Ministry of Education, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
2
|
Hashemi S, Saadat P, Gorgani-Firouzjaee T, Ferdosi-Shahandashti E, Jafarzadeh J. Potential genetic polymorphism of matrix metalloproteinase (MMP)-9 in Iranian migraine patients with Toxoplasma gondii infection. Parasitol Res 2024; 123:140. [PMID: 38386175 DOI: 10.1007/s00436-024-08156-7] [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: 11/18/2023] [Accepted: 02/06/2024] [Indexed: 02/23/2024]
Abstract
Toxoplasma gondii is an intracellular protozoan parasite that causes neuroinflammation in the brain and a constant need for peripheral leukocyte migration. Matrix metalloproteinase 9 (MMP-9) can play a major role in this neuroinflammation and be implicated in some neurological disorders, such as migraines. Therefore, the genetic polymorphism evaluation of MMP-9 in migraine patients with T. gondii infection was performed. One hundred fourteen migraine patients and 114 healthy controls were evaluated for the presence of anti-Toxoplasma IgG antibodies. Seventy-two migraine patients and 40 healthy controls were randomly selected for assessment of the MMP 9-1562C/T genetic polymorphism. In the preliminary examination, 61 (53.5%) migraine patients and 43 (37.3%) healthy controls were positive for IgG antibodies, with a significant association between T. gondii seropositivity and migraine (OR = 1.90; 95% CI = 1.21-3.223; P = 0.012). Genetic distribution for the polymorphism was not in Hardy-Weinberg equilibrium in cases but showed no significant variation in control groups (P = 0.03 and P = 0.180, respectively). A significant preponderance of the CT + TT genotype was found in migraine subjects compared to controls (P = 0.042) (OR, 1.77, CI, 1.013-2.229). The homozygote muted allele TT had a higher rate in migraine patients (6.9%). There were significant differences in CT + TT genotype between T. gondii positive and negative migraine patients (P = 0.024), but T allele frequencies had no significant variation (OR 1.7 CI, 1.084-2.44 and 0.42 CI, 0.044-3.97, respectively). In conclusion, the results may provide the first evidence for the involvement of the MMP-9 gene polymorphism in the mechanism of migraine pathology following Toxoplasma infection.
Collapse
Affiliation(s)
- Sepideh Hashemi
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
| | - Payam Saadat
- Mobility Impairment Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Tahmineh Gorgani-Firouzjaee
- Infectious Diseases and Tropical Medicine Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran.
| | | | - Jalal Jafarzadeh
- Department of Mycology and Parasitology, School of Medicine, Babol University of Medical Sciences, Babol, Iran
| |
Collapse
|
3
|
Lee CSM, Kaur A, Montague SJ, Hicks SM, Andrews RK, Gardiner EE. Tissue inhibitors of metalloproteinases (TIMPs) modulate platelet ADAM10 activity. Platelets 2023; 34:2288213. [PMID: 38031964 DOI: 10.1080/09537104.2023.2288213] [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: 08/21/2023] [Accepted: 11/21/2023] [Indexed: 12/01/2023]
Abstract
Platelet-specific collagen receptor glycoprotein (GP)VI is stable on the surface of circulating platelets but undergoes ectodomain cleavage on activated platelets. Activation-dependent GPVI metalloproteolysis is primarily mediated by A Disintegrin And Metalloproteinase (ADAM) 10. Regulation of platelet ADAMs activity is not well-defined however Tissue Inhibitors of Metalloproteinases (TIMPs) may play a role. As levels of TIMPs on platelets and the control of ADAMs-mediated shedding by TIMPs has not been evaluated, we quantified the levels of TIMPs on the surface of resting and activated platelets from healthy donors by flow cytometry and multiplex ELISA. Variable levels of all TIMPs could be detected on platelets. Plasma contained significant quantities of TIMP1 and TIMP2, but only trace amounts of TIMP3 and TIMP4. Recombinant TIMP3 strongly ablated resting and activated platelet ADAM10 activity, when monitored using a quenched fluorogenic peptide substrate with ADAM10 specificity. Whilst ADAM10-specific inhibitor GI254023X or ethylenediamine tetraacetic acid (EDTA) could modulate ligand-initiated shedding of GPVI, only recombinant TIMP2 achieved a modest (~20%) inhibition. We conclude that some platelet TIMPs are able to modulate platelet ADAM10 activity but none strongly regulate ligand-dependent shedding of GPVI. Our findings provide new insights into the regulation of platelet receptor sheddase activity.
Collapse
Affiliation(s)
- Christine Shu Mei Lee
- Division of Genome Science and Cancer, John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| | - Amandeep Kaur
- Division of Genome Science and Cancer, John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| | - Samantha J Montague
- Division of Genome Science and Cancer, John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| | - Sarah M Hicks
- Division of Genome Science and Cancer, John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| | - Robert K Andrews
- Division of Genome Science and Cancer, John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| | - Elizabeth E Gardiner
- Division of Genome Science and Cancer, John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| |
Collapse
|
4
|
Sutter PA, Willis CM, Menoret A, Nicaise AM, Sacino A, Sikkema AH, Jellison E, Win KK, Han DK, Church W, Baron W, Vella AT, Crocker SJ. Astrocytic TIMP-1 regulates production of Anastellin, a novel inhibitor of oligodendrocyte differentiation and FTY720 responses. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.17.529003. [PMID: 36824834 PMCID: PMC9949145 DOI: 10.1101/2023.02.17.529003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/20/2023]
Abstract
Astrocyte activation is associated with neuropathology and the production of tissue inhibitor of metalloproteinase-1 (TIMP1). TIMP1 is a pleiotropic extracellular protein that functions both as a protease inhibitor and as a growth factor. We have previously demonstrated that murine astrocytes that lack expression of Timp1 do not support rat oligodendrocyte progenitor cell (rOPC) differentiation, and adult global Timp1 knockout ( Timp1 KO ) mice do not efficiently remyelinate following a demyelinating injury. To better understand the basis of this, we performed unbiased proteomic analyses and identified a fibronectin-derived peptide called anastellin that is unique to the murine Timp1 KO astrocyte secretome. Anastellin was found to block rOPC differentiation in vitro and enhanced the inhibitory influence of fibronectin on rOPC differentiation. Anastellin is known to act upon the sphingosine-1-phosphate receptor 1 (S1PR1), and we determined that anastellin also blocked the pro-myelinating effect of FTY720 (or fingolimod) on rOPC differentiation in vitro . Further, administration of FTY720 to wild-type C57BL/6 mice during MOG 35-55 -EAE ameliorated clinical disability while FTY720 administered to mice lacking expression of Timp1 in astrocytes ( Timp1 cKO ) had no effect. Analysis of human TIMP1 and fibronectin ( FN1 ) transcripts from healthy and multiple sclerosis (MS) patient brain samples revealed an inverse relationship where lower TIMP1 expression was coincident with elevated FN1 in MS astrocytes. Lastly, we analyzed proteomic databases of MS samples and identified anastellin peptides to be more abundant in the cerebrospinal fluid (CSF) of human MS patients with high versus low disease activity. The prospective role for anastellin generation in association with myelin lesions as a consequence of a lack of astrocytic TIMP-1 production could influence both the efficacy of fingolimod responses and the innate remyelination potential of the the MS brain. Significance Statement Astrocytic production of TIMP-1 prevents the protein catabolism of fibronectin. In the absence of TIMP-1, fibronectin is further digested leading to a higher abundance of anastellin peptides that can bind to sphingosine-1-phosphate receptor 1. The binding of anastellin with the sphingosine-1-phosphate receptor 1 impairs the differentiation of oligodendrocytes progenitor cells into myelinating oligodendrocytes in vitro , and negates the astrocyte-mediated therapeutic effects of FTY720 in the EAE model of chronic CNS inflammation. These data indicate that TIMP-1 production by astrocytes is important in coordinating astrocytic functions during inflammation. In the absence of astrocyte produced TIMP-1, elevated expression of anastellin may represent a prospective biomarker for FTY720 therapeutic responsiveness.
Collapse
|
5
|
Hu Y, Yu L, Fan H, Huang G, Wu Q, Nie Y, Liu S, Yan L, Wei F. Genomic Signatures of Coevolution between Nonmodel Mammals and Parasitic Roundworms. Mol Biol Evol 2021; 38:531-544. [PMID: 32960966 PMCID: PMC7826172 DOI: 10.1093/molbev/msaa243] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Antagonistic coevolution between host and parasite drives species evolution. However, most of the studies only focus on parasitism adaptation and do not explore the coevolution mechanisms from the perspective of both host and parasite. Here, through the de novo sequencing and assembly of the genomes of giant panda roundworm, red panda roundworm, and lion roundworm parasitic on tiger, we investigated the genomic mechanisms of coevolution between nonmodel mammals and their parasitic roundworms and those of roundworm parasitism in general. The genome-wide phylogeny revealed that these parasitic roundworms have not phylogenetically coevolved with their hosts. The CTSZ and prolyl 4-hydroxylase subunit beta (P4HB) immunoregulatory proteins played a central role in protein interaction between mammals and parasitic roundworms. The gene tree comparison identified that seven pairs of interactive proteins had consistent phylogenetic topology, suggesting their coevolution during host–parasite interaction. These coevolutionary proteins were particularly relevant to immune response. In addition, we found that the roundworms of both pandas exhibited higher proportions of metallopeptidase genes, and some positively selected genes were highly related to their larvae’s fast development. Our findings provide novel insights into the genetic mechanisms of coevolution between nonmodel mammals and parasites and offer the valuable genomic resources for scientific ascariasis prevention in both pandas.
Collapse
Affiliation(s)
- Yibo Hu
- CAS Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China.,Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, China
| | - Lijun Yu
- CAS Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Huizhong Fan
- CAS Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Guangping Huang
- CAS Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Qi Wu
- CAS Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Yonggang Nie
- CAS Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, China
| | - Shuai Liu
- CAS Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Li Yan
- CAS Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Fuwen Wei
- CAS Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China.,Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, China
| |
Collapse
|
6
|
Elsheikha HM, Marra CM, Zhu XQ. Epidemiology, Pathophysiology, Diagnosis, and Management of Cerebral Toxoplasmosis. Clin Microbiol Rev 2021; 34:e00115-19. [PMID: 33239310 PMCID: PMC7690944 DOI: 10.1128/cmr.00115-19] [Citation(s) in RCA: 93] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Toxoplasma gondii is known to infect a considerable number of mammalian and avian species and a substantial proportion of the world's human population. The parasite has an impressive ability to disseminate within the host's body and employs various tactics to overcome the highly regulatory blood-brain barrier and reside in the brain. In healthy individuals, T. gondii infection is largely tolerated without any obvious ill effects. However, primary infection in immunosuppressed patients can result in acute cerebral or systemic disease, and reactivation of latent tissue cysts can lead to a deadly outcome. It is imperative that treatment of life-threatening toxoplasmic encephalitis is timely and effective. Several therapeutic and prophylactic regimens have been used in clinical practice. Current approaches can control infection caused by the invasive and highly proliferative tachyzoites but cannot eliminate the dormant tissue cysts. Adverse events and other limitations are associated with the standard pyrimethamine-based therapy, and effective vaccines are unavailable. In this review, the epidemiology, economic impact, pathophysiology, diagnosis, and management of cerebral toxoplasmosis are discussed, and critical areas for future research are highlighted.
Collapse
Affiliation(s)
- Hany M Elsheikha
- Faculty of Medicine and Health Sciences, School of Veterinary Medicine and Science, University of Nottingham, Loughborough, United Kingdom
| | - Christina M Marra
- Departments of Neurology and Medicine, University of Washington School of Medicine, Seattle, Washington, USA
| | - Xing-Quan Zhu
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu Province, People's Republic of China
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi Province, People's Republic of China
| |
Collapse
|
7
|
McGovern KE, Nance JP, David CN, Harrison RES, Noor S, Worth D, Landrith TA, Obenaus A, Carson MJ, Morikis D, Wilson EH. SPARC coordinates extracellular matrix remodeling and efficient recruitment to and migration of antigen-specific T cells in the brain following infection. Sci Rep 2021; 11:4549. [PMID: 33633185 PMCID: PMC7907143 DOI: 10.1038/s41598-021-83952-0] [Citation(s) in RCA: 4] [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: 11/18/2020] [Accepted: 02/03/2021] [Indexed: 01/14/2023] Open
Abstract
Central nervous system (CNS) injury and infection can result in profound tissue remodeling in the brain, the mechanism and purpose of which is poorly understood. Infection with the protozoan parasite Toxoplasma gondii causes chronic infection and inflammation in the brain parenchyma. Control of parasite replication requires the continuous presence of IFNγ-producing T cells to keep T. gondii in its slowly replicating cyst form. During infection, a network of extracellular matrix fibers, revealed using multiphoton microscopy, forms in the brain. The origin and composition of these structures are unknown but the fibers have been observed to act as a substrate for migrating T cells. In this study, we show a critical regulator of extracellular matrix (ECM) remodeling, Secreted Protein, Acidic, Rich in Cysteine (SPARC), is upregulated in the brain during the early phases of infection in the frontal cortex. In the absence of SPARC, a reduced and disordered fibrous network, increased parasite burden, and reduced antigen-specific T cell entry into the brain points to a role for SPARC in T cell recruitment to and migration within the brain. We also report SPARC can directly bind to CCR7 ligands CCL19 and CCL21 but not CXCL10, and enhance migration toward a chemokine gradient. Measurement of T cell behavior points to tissue remodeling being important for access of immune cells to the brain and facilitating cellular locomotion. Together, these data identify SPARC as an important regulatory component of immune cell trafficking and access to the inflamed CNS.
Collapse
Affiliation(s)
- Kathryn E McGovern
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, 92521, USA
- BIO5 Institute, Department of Immunobiology, University of Arizona, Tucson, AZ, 85724, USA
| | - J Philip Nance
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, 92521, USA
| | - Clément N David
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, 92521, USA
- Nanostring Technologies, Inc, 530 Fairview Ave N, Seattle, WA, 98109, USA
| | - Reed E S Harrison
- Department of Bioengineering, University of California, Riverside, Riverside, CA, 92521-0129, USA
- UCSD Bioengineering and the Institute for Engineering in Medicine, San Diego, CA, 92093, USA
| | - Shahani Noor
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, 92521, USA
- School of Medicine, MSC08, University of New Mexico, Albequerque, NM, 87131, USA
| | - Danielle Worth
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, 92521, USA
| | - Tyler A Landrith
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, 92521, USA
- Ambrey Genetics, Aliso Viejo, CA, 92656, USA
| | - Andre Obenaus
- School of Medicine, University of California, Irvine, Irvine, CA, 92697, USA
| | - Monica J Carson
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, 92521, USA
| | - Dimitrios Morikis
- Department of Bioengineering, University of California, Riverside, Riverside, CA, 92521-0129, USA
| | - Emma H Wilson
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, 92521, USA.
| |
Collapse
|
8
|
Mukhopadhyay D, Arranz-Solís D, Saeij JPJ. Influence of the Host and Parasite Strain on the Immune Response During Toxoplasma Infection. Front Cell Infect Microbiol 2020; 10:580425. [PMID: 33178630 PMCID: PMC7593385 DOI: 10.3389/fcimb.2020.580425] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 09/11/2020] [Indexed: 01/02/2023] Open
Abstract
Toxoplasma gondii is an exceptionally successful parasite that infects a very broad host range, including humans, across the globe. The outcome of infection differs remarkably between hosts, ranging from acute death to sterile infection. These differential disease patterns are strongly influenced by both host- and parasite-specific genetic factors. In this review, we discuss how the clinical outcome of toxoplasmosis varies between hosts and the role of different immune genes and parasite virulence factors, with a special emphasis on Toxoplasma-induced ileitis and encephalitis.
Collapse
Affiliation(s)
| | | | - Jeroen P. J. Saeij
- Department of Pathology, Microbiology & Immunology, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
| |
Collapse
|
9
|
Mitoma H, Manto M. Disruption of the Blood-Brain Barrier During Neuroinflammatory and Neuroinfectious Diseases. NEUROIMMUNE DISEASES 2019. [PMCID: PMC7121618 DOI: 10.1007/978-3-030-19515-1_7] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
As the organ of highest metabolic demand, utilizing over 25% of total body glucose utilization via an enormous vasculature with one capillary every 73 μm, the brain evolves a barrier at the capillary and postcapillary venules to prevent toxicity during serum fluctuations in metabolites and hormones, to limit brain swelling during inflammation, and to prevent pathogen invasion. Understanding of neuroprotective barriers has since evolved to incorporate the neurovascular unit (NVU), the blood-cerebrospinal fluid (CSF) barrier, and the presence of CNS lymphatics that allow leukocyte egress. Identification of the cellular and molecular participants in BBB function at the NVU has allowed detailed analyses of mechanisms that contribute to BBB dysfunction in various disease states, which include both autoimmune and infectious etiologies. This chapter will introduce some of the cellular and molecular components that promote barrier function but may be manipulated by inflammatory mediators or pathogens during neuroinflammation or neuroinfectious diseases.
Collapse
Affiliation(s)
- Hiroshi Mitoma
- Medical Education Promotion Center, Tokyo Medical University, Tokyo, Japan
| | - Mario Manto
- Department of Neurology, CHU-Charleroi, Charleroi, Belgium, Department of Neurosciences, University of Mons, Mons, Belgium
| |
Collapse
|
10
|
Ólafsson EB, Ross EC, Varas-Godoy M, Barragan A. TIMP-1 promotes hypermigration of Toxoplasma-infected primary dendritic cells via CD63-ITGB1-FAK signaling. J Cell Sci 2019; 132:jcs.225193. [PMID: 30635444 DOI: 10.1242/jcs.225193] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 12/27/2018] [Indexed: 01/11/2023] Open
Abstract
Tissue inhibitor of metalloproteinases-1 (TIMP-1) exerts pleiotropic effects on cells including conferring metastatic properties to cancer cells. As for metastatic cells, recent paradigms of leukocyte migration attribute important roles to the amoeboid migration mode of dendritic cells (DCs) for rapid locomotion in tissues. However, the role of TIMP-1 in immune cell migration and in the context of infection has not been addressed. We report that, upon challenge with the obligate intracellular parasite Toxoplasma gondii, primary DCs secrete TIMP-1 with implications for their migratory properties. Using a short hairpin RNA (shRNA) gene silencing approach, we demonstrate that secreted TIMP-1 and its ligand CD63 are required for the onset of hypermotility in DCs challenged with T. gondii Further, gene silencing and antibody blockade of the β1-integrin CD29 (ITGB1) inhibited DC hypermotility, indicating that signal transduction occurred via ITGB1. Finally, gene silencing of the ITGB1-associated focal adhesion kinase (FAK, also known as PTK2), as well as pharmacological antagonism of FAK and associated kinases SRC and PI3K, abrogated hypermotility. The present study identifies a TIMP-1-CD63-ITGB1-FAK signaling axis in primary DCs, which T. gondii hijacks to drive high-speed amoeboid migration of the vehicle cells that facilitate its systemic dissemination.
Collapse
Affiliation(s)
- Einar B Ólafsson
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, 106 09 Stockholm, Sweden
| | - Emily C Ross
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, 106 09 Stockholm, Sweden
| | - Manuel Varas-Godoy
- Centro de Investigación Biomédica, Faculty of Medicine, Universidad de los Andes, 7620001 Santiago, Chile
| | - Antonio Barragan
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, 106 09 Stockholm, Sweden
| |
Collapse
|
11
|
Chuntova P, Downey KM, Hegde B, Almeida ND, Okada H. Genetically Engineered T-Cells for Malignant Glioma: Overcoming the Barriers to Effective Immunotherapy. Front Immunol 2019; 9:3062. [PMID: 30740109 PMCID: PMC6357938 DOI: 10.3389/fimmu.2018.03062] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 12/11/2018] [Indexed: 12/12/2022] Open
Abstract
Malignant gliomas carry a dismal prognosis. Conventional treatment using chemo- and radiotherapy has limited efficacy with adverse events. Therapy with genetically engineered T-cells, such as chimeric antigen receptor (CAR) T-cells, may represent a promising approach to improve patient outcomes owing to their potential ability to attack highly infiltrative tumors in a tumor-specific manner and possible persistence of the adaptive immune response. However, the unique anatomical features of the brain and susceptibility of this organ to irreversible tissue damage have made immunotherapy especially challenging in the setting of glioma. With safety concerns in mind, multiple teams have initiated clinical trials using CAR T-cells in glioma patients. The valuable lessons learnt from those trials highlight critical areas for further improvement: tackling the issues of the antigen presentation and T-cell homing in the brain, immunosuppression in the glioma microenvironment, antigen heterogeneity and off-tumor toxicity, and the adaptation of existing clinical therapies to reflect the intricacies of immune response in the brain. This review summarizes the up-to-date clinical outcomes of CAR T-cell clinical trials in glioma patients and examines the most pressing hurdles limiting the efficacy of these therapies. Furthermore, this review uses these hurdles as a framework upon which to evaluate cutting-edge pre-clinical strategies aiming to overcome those barriers.
Collapse
Affiliation(s)
- Pavlina Chuntova
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, United States
| | - Kira M Downey
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, United States
| | - Bindu Hegde
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, United States
| | - Neil D Almeida
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, United States.,George Washington University School of Medicine and Health Sciences, Washington, DC, United States
| | - Hideho Okada
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, United States.,The Parker Institute for Cancer Immunotherapy, University of California, San Francisco, San Francisco, CA, United States.,Cancer Immunotherapy Program, University of California, San Francisco, San Francisco, CA, United States
| |
Collapse
|
12
|
The Activity of Matrix Metalloproteinases (MMP-2, MMP-9) and Their Tissue Inhibitors (TIMP-1, TIMP-3) in the Cerebral Cortex and Hippocampus in Experimental Acanthamoebiasis. Int J Mol Sci 2018; 19:ijms19124128. [PMID: 30572657 PMCID: PMC6321078 DOI: 10.3390/ijms19124128] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 12/13/2018] [Accepted: 12/14/2018] [Indexed: 12/20/2022] Open
Abstract
The pathological process occurring within the central nervous system (CNS) as a result of the infection by Acanthamoeba spp. is not fully understood. Therefore, the aim of this study was to determine whether Acanthamoeba spp. may affect the levels of matrix metalloproteinases (MMP-2,-9), their tissue inhibitors (TIMP-1,-3) and MMP-9/TIMP-1, MMP-2/TIMP-3 ratios in the cerebral cortex and hippocampus, in relation to the host’s immunological status. Our results showed that Acanthamoeba spp. infection can change the levels of MMP and TIMP in the CNS and may be amenable targets for limiting amoebic encephalitis. The increase in the activity of matrix metalloproteinases during acanthamoebiasis may be primarily the result of inflammation process, probably an increased activity of proteolytic processes, but also (to a lesser extent) a defense mechanism preventing the processes of neurodegeneration.
Collapse
|
13
|
Gene Profile Expression Related to Type I Interferons in HT-29 Cells Exposed to Cryptosporidium parvum. Jundishapur J Microbiol 2018. [DOI: 10.5812/jjm.63071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
14
|
Sundqvist KG. T Cell Co-Stimulation: Inhibition of Immunosuppression? Front Immunol 2018; 9:974. [PMID: 29774033 PMCID: PMC5943593 DOI: 10.3389/fimmu.2018.00974] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 04/19/2018] [Indexed: 11/18/2022] Open
Affiliation(s)
- Karl-Gösta Sundqvist
- Division of Clinical Immunology, Department of Laboratory Medicine, Karolinska Institutet, Clinical Immunology and Transfusion Medicine at Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
15
|
Brasil TR, Freire-de-Lima CG, Morrot A, Vetö Arnholdt AC. Host- Toxoplasma gondii Coadaptation Leads to Fine Tuning of the Immune Response. Front Immunol 2017; 8:1080. [PMID: 28955329 PMCID: PMC5601305 DOI: 10.3389/fimmu.2017.01080] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Accepted: 08/21/2017] [Indexed: 12/22/2022] Open
Abstract
Toxoplasma gondii has successfully developed strategies to evade host's immune response and reach immune privileged sites, which remains in a controlled environment inside quiescent tissue cysts. In this review, we will approach several known mechanisms used by the parasite to modulate mainly the murine immune system at its favor. In what follows, we review recent findings revealing interference of host's cell autonomous immunity and cell signaling, gene expression, apoptosis, and production of microbicide molecules such as nitric oxide and oxygen reactive species during parasite infection. Modulation of host's metalloproteinases of extracellular matrix is also discussed. These immune evasion strategies are determinant to parasite dissemination throughout the host taking advantage of cells from the immune system to reach brain and retina, crossing crucial hosts' barriers.
Collapse
Affiliation(s)
- Thaís Rigueti Brasil
- Laboratório de Biologia do Reconhecer, Universidade Estadual do Norte Fluminense, Rio de Janeiro, Brazil
| | | | - Alexandre Morrot
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro, Brazil
| | | |
Collapse
|
16
|
Wohlfert EA, Blader IJ, Wilson EH. Brains and Brawn: Toxoplasma Infections of the Central Nervous System and Skeletal Muscle. Trends Parasitol 2017; 33:519-531. [PMID: 28483381 PMCID: PMC5549945 DOI: 10.1016/j.pt.2017.04.001] [Citation(s) in RCA: 93] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Revised: 03/29/2017] [Accepted: 04/08/2017] [Indexed: 02/06/2023]
Abstract
Toxoplasma gondii is a widespread parasitic pathogen that infects over a third of the world's population. Following an acute infection, the parasite can persist within its mammalian host as intraneuronal or intramuscular cysts. Cysts will occasionally reactivate, and - depending on the host's immune status and site of reactivation - encephalitis or myositis can develop. Because these diseases have high levels of morbidity and can be lethal, it is important to understand how Toxoplasma traffics to these tissues, how the immune response controls parasite burden and contributes to tissue damage, and what mechanisms underlie neurological and muscular pathologies that toxoplasmosis patients present with. This review aims to summarize recent important developments addressing these critical topics.
Collapse
Affiliation(s)
- Elizabeth A Wohlfert
- Department of Microbiology and Immunology, University at Buffalo School of Medicine, Buffalo, NY, USA.
| | - Ira J Blader
- Department of Microbiology and Immunology, University at Buffalo School of Medicine, Buffalo, NY, USA.
| | - Emma H Wilson
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, USA.
| |
Collapse
|
17
|
Elsheikha HM, Büsselberg D, Zhu XQ. The known and missing links between Toxoplasma gondii and schizophrenia. Metab Brain Dis 2016; 31:749-59. [PMID: 27041387 DOI: 10.1007/s11011-016-9822-1] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 03/20/2016] [Indexed: 12/14/2022]
Abstract
Toxoplasma gondii, an intracellular protozoan parasite, has a striking predilection for infecting the Central Nervous System and has been linked to an increased incidence of a number of psychiatric diseases. Several in vitro and in vivo studies have shown that T. gondii infection can affect the structure, bioenergetics and function of brain cells, and alters several host cell processes, including dopaminergic, tryptophan-kynurenine, GABAergic, AKT1, Jak/STAT, and vasopressinergic pathways. These mechanisms underlying the neuropathology of latent toxoplasmosis seem to operate also in schizophrenia, supporting the link between the two disorders. Better understanding of the intricate parasite-neuroglial communications holds the key to unlocking the mystery of T. gondii-mediated schizophrenia and offers substantial prospects for the development of disease-modifying therapies.
Collapse
Affiliation(s)
- Hany M Elsheikha
- Faculty of Medicine and Health Sciences, School of Veterinary Medicine and Science, University of Nottingham, Sutton Bonington, Leicestershire, LE12 5RD, UK.
| | - Dietrich Büsselberg
- Weill Cornell Medical College in Qatar, Qatar Foundation - Education City, P.O. Box: 24144, Doha, Qatar
| | - Xing-Quan Zhu
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu Province, 730046, People's Republic of China
| |
Collapse
|
18
|
Möhle L, Israel N, Paarmann K, Krohn M, Pietkiewicz S, Müller A, Lavrik IN, Buguliskis JS, Schott BH, Schlüter D, Gundelfinger ED, Montag D, Seifert U, Pahnke J, Dunay IR. Chronic Toxoplasma gondii infection enhances β-amyloid phagocytosis and clearance by recruited monocytes. Acta Neuropathol Commun 2016; 4:25. [PMID: 26984535 PMCID: PMC4793516 DOI: 10.1186/s40478-016-0293-8] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Accepted: 02/19/2016] [Indexed: 01/02/2023] Open
Abstract
INTRODUCTION Alzheimer's disease (AD) is associated with the accumulation of β-amyloid (Aβ) as senile plaques in the brain, thus leading to neurodegeneration and cognitive impairment. Plaque formation depends not merely on the amount of generated Aβ peptides, but more importantly on their effective removal. Chronic infections with neurotropic pathogens, most prominently the parasite Toxoplasma (T.) gondii, are frequent in the elderly, and it has been suggested that the resulting neuroinflammation may influence the course of AD. In the present study, we investigated how chronic T. gondii infection and resulting neuroinflammation affect plaque deposition and removal in a mouse model of AD. RESULTS Chronic infection with T. gondii was associated with reduced Aβ and plaque load in 5xFAD mice. Upon infection, myeloid-derived CCR2(hi) Ly6C(hi) monocytes, CCR2(+) Ly6C(int), and CCR2(+) Ly6C(low) mononuclear cells were recruited to the brain of mice. Compared to microglia, these recruited mononuclear cells showed highly increased phagocytic capacity of Aβ ex vivo. The F4/80(+) Ly6C(low) macrophages expressed high levels of Triggering Receptor Expressed on Myeloid cells 2 (TREM2), CD36, and Scavenger Receptor A1 (SCARA1), indicating phagocytic activity. Importantly, selective ablation of CCR2(+) Ly6C(hi) monocytes resulted in an increased amount of Aβ in infected mice. Elevated insulin-degrading enzyme (IDE), matrix metalloproteinase 9 (MMP9), as well as immunoproteasome subunits β1i/LMP2, β2i/MECL-1, and β5i/LMP7 mRNA levels in the infected brains indicated increased proteolytic Aβ degradation. Particularly, LMP7 was highly expressed by the recruited mononuclear cells in the brain, suggesting a novel mechanism of Aβ clearance. CONCLUSIONS Our results indicate that chronic Toxoplasma infection ameliorates β-amyloidosis in a murine model of AD by activation of the immune system, specifically by recruitment of Ly6C(hi) monocytes and by enhancement of phagocytosis and degradation of soluble Aβ. Our findings provide evidence for a modulatory role of inflammation-induced Aβ phagocytosis and degradation by newly recruited peripheral immune cells in the pathophysiology of AD.
Collapse
Affiliation(s)
- Luisa Möhle
- Institute for Medical Microbiology and Hospital Hygiene, University of Magdeburg, Leipziger Str. 44, 39120, Magdeburg, Germany
| | - Nicole Israel
- Institute for Molecular and Clinical Immunology, University of Magdeburg, Magdeburg, Germany
| | - Kristin Paarmann
- Department of Pathology (PAT), Translational Neurodegeneration Research and Neuropathology Lab, University of Oslo (UiO) and Oslo University Hospital (OUS), Oslo, Norway
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
- Neurogenetics, Leibniz Institute for Neurobiology, Magdeburg, Germany
| | - Markus Krohn
- Department of Pathology (PAT), Translational Neurodegeneration Research and Neuropathology Lab, University of Oslo (UiO) and Oslo University Hospital (OUS), Oslo, Norway
| | - Sabine Pietkiewicz
- Department of Translational Inflammation Research, Institute of Experimental Internal Medicine, University of Magdeburg, Magdeburg, Germany
| | - Andreas Müller
- Institute for Molecular and Clinical Immunology, University of Magdeburg, Magdeburg, Germany
- Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Inna N Lavrik
- Department of Translational Inflammation Research, Institute of Experimental Internal Medicine, University of Magdeburg, Magdeburg, Germany
| | | | - Björn H Schott
- Center for Behavioral Brain Sciences (CBBS), University of Magdeburg, Magdeburg, Germany
- Department of Behavioral Neurology, Leibniz Institute for Neurobiology, Magdeburg, Germany
- Department of Psychiatry and Psychotherapy, Campus Mitte, Charité Universitätsmedizin, Berlin, Germany
| | - Dirk Schlüter
- Institute for Medical Microbiology and Hospital Hygiene, University of Magdeburg, Leipziger Str. 44, 39120, Magdeburg, Germany
- Center for Behavioral Brain Sciences (CBBS), University of Magdeburg, Magdeburg, Germany
| | - Eckart D Gundelfinger
- Center for Behavioral Brain Sciences (CBBS), University of Magdeburg, Magdeburg, Germany
- Department of Neurochemistry and Molecular Biology, Leibniz Institute for Neurobiology, Magdeburg, Germany
- Medical Faculty, University of Magdeburg, Magdeburg, Germany
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
| | - Dirk Montag
- Neurogenetics, Leibniz Institute for Neurobiology, Magdeburg, Germany
| | - Ulrike Seifert
- Institute for Molecular and Clinical Immunology, University of Magdeburg, Magdeburg, Germany
| | - Jens Pahnke
- Department of Pathology (PAT), Translational Neurodegeneration Research and Neuropathology Lab, University of Oslo (UiO) and Oslo University Hospital (OUS), Oslo, Norway
- University of Lübeck (UzL), LIED, Lübeck, Germany
- Leibniz Institute of Plant Biochemistry (IPB), Halle, Germany
| | - Ildiko Rita Dunay
- Institute for Medical Microbiology and Hospital Hygiene, University of Magdeburg, Leipziger Str. 44, 39120, Magdeburg, Germany.
- Center for Behavioral Brain Sciences (CBBS), University of Magdeburg, Magdeburg, Germany.
| |
Collapse
|
19
|
Tomasik J, Schultz TL, Kluge W, Yolken RH, Bahn S, Carruthers VB. Shared Immune and Repair Markers During Experimental Toxoplasma Chronic Brain Infection and Schizophrenia. Schizophr Bull 2016; 42:386-95. [PMID: 26392628 PMCID: PMC4753603 DOI: 10.1093/schbul/sbv134] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Chronic neurologic infection with Toxoplasma gondii is relatively common in humans and is one of the strongest known risk factors for schizophrenia. Nevertheless, the exact neuropathological mechanisms linking T gondii infection and schizophrenia remain unclear. Here we utilize a mouse model of chronic T gondii infection to identify protein biomarkers that are altered in serum and brain samples at 2 time points during chronic infection. Furthermore, we compare the identified biomarkers to those differing between "postmortem" brain samples from 35 schizophrenia patients and 33 healthy controls. Our findings suggest that T gondii infection causes substantial and widespread immune activation indicative of neural damage and reactive tissue repair in the animal model that partly overlaps with changes observed in the brains of schizophrenia patients. The overlapping changes include increases in C-reactive protein (CRP), interleukin-1 beta (IL-1β), interferon gamma (IFNγ), plasminogen activator inhibitor 1 (PAI-1), tissue inhibitor of metalloproteinases 1 (TIMP-1), and vascular cell adhesion molecule 1 (VCAM-1). Potential roles of these factors in the pathogenesis of schizophrenia and toxoplasmosis are discussed. Identifying a defined set of markers shared within the pathophysiological landscape of these diseases could be a key step towards understanding their specific contributions to pathogenesis.
Collapse
Affiliation(s)
- Jakub Tomasik
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, UK;,Department of Neuroscience, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - Tracey L. Schultz
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI
| | - Wolfgang Kluge
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, UK
| | - Robert H. Yolken
- Stanley Laboratory of Developmental Neurovirology, Department of Pediatrics, Johns Hopkins School of Medicine, Baltimore, MD
| | - Sabine Bahn
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, UK;,Department of Neuroscience, Erasmus Medical Centre, Rotterdam, The Netherlands;, Joint last authors/ these authors contributed equally to the study
| | - Vern B. Carruthers
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI;, Joint last authors/ these authors contributed equally to the study
| |
Collapse
|
20
|
Borgmann K, Ghorpade A. HIV-1, methamphetamine and astrocytes at neuroinflammatory Crossroads. Front Microbiol 2015; 6:1143. [PMID: 26579077 PMCID: PMC4621459 DOI: 10.3389/fmicb.2015.01143] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Accepted: 10/05/2015] [Indexed: 12/30/2022] Open
Abstract
As a popular psychostimulant, methamphetamine (METH) use leads to long-lasting, strong euphoric effects. While METH abuse is common in the general population, between 10 and 15% of human immunodeficiency virus-1 (HIV-1) patients report having abused METH. METH exacerbates the severity and onset of HIV-1-associated neurocognitive disorders (HAND) through direct and indirect mechanisms. Repetitive METH use impedes adherence to antiretroviral drug regimens, increasing the likelihood of HIV-1 disease progression toward AIDS. METH exposure also directly affects both innate and adaptive immunity, altering lymphocyte numbers and activity, cytokine signaling, phagocytic function and infiltration through the blood brain barrier. Further, METH triggers the dopamine reward pathway and leads to impaired neuronal activity and direct toxicity. Concurrently, METH and HIV-1 alter the neuroimmune balance and induce neuroinflammation, which modulates a wide range of brain functions including neuronal signaling and activity, glial activation, viral infection, oxidative stress, and excitotoxicity. Pathologically, reactive gliosis is a hallmark of both HIV-1- and METH-associated neuroinflammation. Significant commonality exists in the neurotoxic mechanisms for both METH and HAND; however, the pathways dysregulated in astroglia during METH exposure are less clear. Thus, this review highlights alterations in astrocyte intracellular signaling pathways, gene expression and function during METH and HIV-1 comorbidity, with special emphasis on HAND-associated neuroinflammation. Importantly, this review carefully evaluates interventions targeting astrocytes in HAND and METH as potential novel therapeutic approaches. This comprehensive overview indicates, without a doubt, that during HIV-1 infection and METH abuse, a complex dialog between all neural cells is orchestrated through astrocyte regulated neuroinflammation.
Collapse
Affiliation(s)
- Kathleen Borgmann
- Department of Cell Biology and Immunology, University of North Texas Health Science Center Fort Worth, TX, USA
| | - Anuja Ghorpade
- Department of Cell Biology and Immunology, University of North Texas Health Science Center Fort Worth, TX, USA
| |
Collapse
|
21
|
Campos TM, Passos ST, Novais FO, Beiting DP, Costa RS, Queiroz A, Mosser D, Scott P, Carvalho EM, Carvalho LP. Matrix metalloproteinase 9 production by monocytes is enhanced by TNF and participates in the pathology of human cutaneous Leishmaniasis. PLoS Negl Trop Dis 2014; 8:e3282. [PMID: 25393535 PMCID: PMC4230914 DOI: 10.1371/journal.pntd.0003282] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Accepted: 09/19/2014] [Indexed: 12/18/2022] Open
Abstract
Introduction Cutaneous leishmaniasis (CL) due to L.braziliensis infection is characterized by a strong inflammatory response with high levels of TNF and ulcer development. Less attention has been given to the role of mononuclear phagocytes to this process. Monocytes constitute a heterogeneous population subdivided into classical, intermediate and non-classical, and are known to migrate to inflammatory sites and secrete inflammatory mediators. TNF participates in the induction of matrix metalloproteinases (MMPs). MMP-9 is an enzyme that degrades basal membrane and its activity is controlled by the tissue inhibitor of metalloproteinase. Methods Mononuclear cells were obtained from ex-vivo labeling sub-populations of monocytes and MMP-9, and the frequency was determined by flow cytometry. Culture was performed during 72 hours, stimulating the cells with SLA, levels of MMP-9 and TIMP-1 in the supernatants were determined by ELISA. Results We observed that cells from CL lesions secrete high amounts of MMP-9 when compared to healthy subjects. Although MMP-9 was produced by monocytes, non-classical ones were the main source of this enzyme. We also observed that TNF produced in high level during CL contributes to MMP-9 production. Conclusions These observations emphasize the role of monocytes, TNF and MMP-9 in the pathogenesis of L. braziliensis infection. To examine the participation of MMP-9 in the pathogenesis of L. braziliensis infection, we realized a cross-sectional study with CL patients in an early phase of the disease or with a classical ulcer, and healthy controls. We evaluated the frequency of MMP-9 in monocyte subsets and its mechanism of production. Our results showed that monocytes were the major cells producing MMP-9. The MMP-9 production by CL patients was presented in higher levels when compared with healthy subjects and early cutaneous leishmaniasis (ECL) patients, and the levels of MMP-9 inhibitor, TIMP-1, were lower in CL patients when compared to healthy subjects. The production of MMP-9 was enhanced by TNF, a cytokine associated with tissue damage in CL patients. Thus, therapeutic modulation of MMP-9 may be a useful approach for improving disease outcome in L. braziliensis patients.
Collapse
Affiliation(s)
- Taís M. Campos
- Serviço de Imunologia, Universidade Federal da Bahia, Salvador, Bahia, Brazil
| | - Sara T. Passos
- Serviço de Imunologia, Universidade Federal da Bahia, Salvador, Bahia, Brazil
- Instituto Nacional de Ciências e Tecnologia – Doenças Tropicais, Salvador, Bahia, Brazil
| | - Fernanda O. Novais
- Department of Pathobiology, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Daniel P. Beiting
- Department of Pathobiology, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Rúbia S. Costa
- Serviço de Imunologia, Universidade Federal da Bahia, Salvador, Bahia, Brazil
| | - Adriano Queiroz
- Serviço de Imunologia, Universidade Federal da Bahia, Salvador, Bahia, Brazil
| | - David Mosser
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, Maryland, United States of America
| | - Phillip Scott
- Department of Pathobiology, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Edgar M. Carvalho
- Serviço de Imunologia, Universidade Federal da Bahia, Salvador, Bahia, Brazil
- Instituto Nacional de Ciências e Tecnologia – Doenças Tropicais, Salvador, Bahia, Brazil
| | - Lucas P. Carvalho
- Serviço de Imunologia, Universidade Federal da Bahia, Salvador, Bahia, Brazil
- Instituto Nacional de Ciências e Tecnologia – Doenças Tropicais, Salvador, Bahia, Brazil
- Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador, Bahia, Brazil
- * E-mail:
| |
Collapse
|
22
|
Anikina AG, Potapova OV, Shkurupii VA, Shestopalov AM. Role of matrix metalloproteinases and their inhibitor in the development of early pulmonary fibrosis in mice infected with influenza A/H5N1 A/goose/Krasnoozerskoye/627/05 virus. Bull Exp Biol Med 2013; 156:11-4. [PMID: 24319704 DOI: 10.1007/s10517-013-2265-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
High levels of macrophages and fibroblasts expressing MMP-2, MMP-9, and MMP-10 against the background of progressing early fibrosis of the lungs (manifesting in an increase in volume density of type I, III, IV, and VI collagens) were found in C57Bl/6 mice infected with influenza A/H5N1 A/goose/Krasnoozerskoye/627/05 virus. Progressing fibrosis of the lungs in infected mice was associated with imbalance of collagen synthesis and degradation processes conjugated with high levels of macrophages and fibroblasts expressing TIMP-2.
Collapse
Affiliation(s)
- A G Anikina
- Research Center of Clinical and Experimental Medicine, Siberian Division of the Russian Academy of Medical Sciences, Novosibirsk; Vector State Research Center of Virology and Biotechnology, Novosibirsk Region, Koltsovo; Novosibirsk State Medical University, Ministry of Health of the Russian Federation, Russia.
| | | | | | | |
Collapse
|
23
|
Abstract
Infection of the CNS (central nervous system) with a sublethal neurotropic coronavirus (JHMV) induces a vigorous inflammatory response. CD4+ and CD8+ T cells are essential to control infectious virus but at the cost of tissue damage. An enigma in understanding the contribution of T cell subsets in pathogenesis resides in their distinct migration pattern across the BBB (blood brain barrier). CD4+ T cells transiently accumulate within the perivascular space, whereas CD8+ T cells migrate directly into the CNS parenchyma. As MMPs (matrix metalloproteinases) facilitate migration across the glia limitans, specific expression of the TIMP (tissue inhibitor of MMPs)-1 by CD4+ T cells present in the perivascular cuffs suggested that TIMP-1 is responsible for stalling CD4+ T cell migration into the CNS parenchyma. Using TIMP-1 deficient mice, the present data demonstrate an increase rather than a decrease in CD4+ T cell accumulation within the perivascular space during JHMV infection. Whereas virus control was not affected by perivascular retention of CD4+ T cells, disease severity was decreased and associated with reduced IFNγ (interferon γ) production. Moreover, decreased CD4+ T cell recruitment into the CNS parenchyma of TIMP-1 deficient mice was not associated with impaired T cell recruiting chemokines or MMP expression, and no compensation by other TIMP molecules was identified. These data suggest an MMP-independent role of TIMP-1 in regulating CD4+ T cell access into the CNS parenchyma during acute JHMV encephalitis.
Collapse
|
24
|
Hsieh JL, Shiau AL, Lee CH, Yang SJ, Lee BO, Jou IM, Wu CL, Chen SH, Shen PC. CD8+ T cell-induced expression of tissue inhibitor of metalloproteinses-1 exacerbated osteoarthritis. Int J Mol Sci 2013; 14:19951-70. [PMID: 24108368 PMCID: PMC3821596 DOI: 10.3390/ijms141019951] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2013] [Revised: 09/11/2013] [Accepted: 09/22/2013] [Indexed: 11/16/2022] Open
Abstract
Despites the fact that T cells are involved in the pathogenesis of osteoarthritis (OA) little is known about the roles of CD8+ T cells in this disease. We investigated the effects of CD8+ T cells and the expression of tissue inhibitor of metalloproteinases 1 (TIMP-1) on joint pathology. Using anterior cruciate ligament-transection (ACLT), OA was induced in mice. The knee joints were histologically assessed for manifestations of OA. The CD8+ T cells from splenocytes and synovium were flow-cytometrically and immunochemically evaluated, respectively. Local expression of TIMP-1, matrix metalloproteinase (MMP)-13, and VEGF were examined. Cartilage degeneration was slower in CD8+ T cell knockout mice than in control mice. CD8+ T cells were activated once OA was initiated and expanded during OA progression. More CD8+ T cells from splenocytes expressed TIMP-1 in ACLT-group mice than in Sham-group mice. The number of TIMP-1-expressing CD8+ T cells in OA mice correlated with the disease severity. TIMP-1 expression in cartilage was co-localized with that of MMP-13 and VEGF. TIMP-1 protein was detected in synovium in which angiogenesis occurred. During the pathogenesis of OA, the expression of TIMP-1, VEGF and MMP-13 accompanying with CD8+ T cells activation were increased. Furthermore, inhibiting the expression of TIMP-1 in joints could retard the progression of OA.
Collapse
Affiliation(s)
- Jeng-Long Hsieh
- Department of Nursing, Chung Hwa University of Medical Technology, Tainan 717, Taiwan; E-Mail:
| | - Ai-Li Shiau
- Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan; E-Mails: (A.-L.S.); (S.-H.C.)
| | - Che-Hsin Lee
- Department of Microbiology, School of Medicine, China Medical University, Taichung 404, Taiwan; E-Mail:
| | - Shiu-Ju Yang
- Institute of Basic Medical Science, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan; E-Mail:
| | - Bih-O Lee
- Department of Nursing, Chang Gung University of Technology, Puzih, Chiayi County 613, Taiwan; E-Mail:
| | - I-Ming Jou
- Department of Orthopedics, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan; E-Mail:
| | - Chao-Liang Wu
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan; E-Mail:
| | - Shun-Hua Chen
- Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan; E-Mails: (A.-L.S.); (S.-H.C.)
| | - Po-Chuan Shen
- Department of Orthopedic Surgery, Tainan Hospital, Department of Health, Executive Yuan, Tainan 700, Taiwan
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +886-6-220-0055 (ext. 3165); Fax: +886-6-267-8119
| |
Collapse
|
25
|
Matrix metalloproteinases 2 and 9 are differentially expressed in patients with indeterminate and cardiac clinical forms of Chagas disease. Infect Immun 2013; 81:3600-8. [PMID: 23856618 DOI: 10.1128/iai.00153-13] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Dilated chronic cardiomyopathy (DCC) from Chagas disease is associated with myocardial remodeling and interstitial fibrosis, resulting in extracellular matrix (ECM) changes. In this study, we characterized for the first time the serum matrix metalloproteinase 2 (MMP-2) and MMP-9 levels, as well as their main cell sources in peripheral blood mononuclear cells from patients presenting with the indeterminate (IND) or cardiac (CARD) clinical form of Chagas disease. Our results showed that serum levels of MMP-9 are associated with the severity of Chagas disease. The analysis of MMP production by T lymphocytes showed that CD8(+) T cells are the main mononuclear leukocyte source of both MMP-2 and MMP-9 molecules. Using a new 3-dimensional model of fibrosis, we observed that sera from patients with Chagas disease induced an increase in the extracellular matrix components in cardiac spheroids. Furthermore, MMP-2 and MMP-9 showed different correlations with matrix proteins and inflammatory cytokines in patients with Chagas disease. Our results suggest that MMP-2 and MMP-9 show distinct activities in Chagas disease pathogenesis. While MMP-9 seems to be involved in the inflammation and cardiac remodeling of Chagas disease, MMP-2 does not correlate with inflammatory molecules.
Collapse
|
26
|
Lu CY, Lai SC. Matrix metalloproteinase-2 and -9 lead to fibronectin degradation in astroglia infected with Toxoplasma gondii. Acta Trop 2013. [PMID: 23201304 DOI: 10.1016/j.actatropica.2012.11.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Toxoplasma gondii is a zoonotic parasite and its infection in human can induce toxoplasmic encephalitis in immune disorders. In this study, astroglia were infected with the TS-4 strain of T. gondii tachyzoite in vitro to investigate the changes of matrix metalloproteinase (MMP)-2, MMP-9 and their substrate fibronectin. MMP-2 and MMP-9 were significantly increased at 1 h, 6 h and 12 h post-infection (PI) in the cell homogenates, and increased at 6 h, 12 h, 24 h and 48 h PI in the cell-cultured supernatants. Fibronectin degradation also occurred at the same time points. In addition, immunocytochemistry showed that MMP-2 and MMP-9 localized in the cytoplasm, and confocal scanning laser microscopy revealed co-labeled patterns of MMP-2 and MMP-9 with fibronectin. MMP-2 and MMP-9 interacted with fibronectin, respectively. These results suggest that MMP-2 and MMP-9 induction from astroglia may contribute to extracellular matrix (ECM) degradation occurring in toxoplasmosis. Thus, we hypothesize that MMP-2 and MMP-9 cleave fibronectin and may contribute to the astroglia reaction and leukocyte migration to the sites of T. gondii replication during toxoplasmic encephalitis.
Collapse
|
27
|
Bruschi F, Pinto B. The significance of matrix metalloproteinases in parasitic infections involving the central nervous system. Pathogens 2013; 2:105-29. [PMID: 25436884 PMCID: PMC4235708 DOI: 10.3390/pathogens2010105] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2012] [Revised: 01/30/2013] [Accepted: 02/11/2013] [Indexed: 12/29/2022] Open
Abstract
Matrix metalloproteinases (MMPs) represent a large family of over twenty different secreted or membrane-bound endopeptidases, involved in many physiological (embryogenesis, precursor or stem cell mobilization, tissue remodeling during wound healing, etc.), as well as pathological (inflammation, tumor progression and metastasis in cancer, vascular pathology, etc.) conditions. For a long time, MMPs were considered only for the ability to degrade extracellular matrix (ECM) molecules (e.g., collagen, laminin, fibronectin) and to release hidden epitopes from the ECM. In the last few years, it has been fully elucidated that these molecules have many other functions, mainly related to the immune response, in consideration of their effects on cytokines, hormones and chemokines. Among others, MMP-2 and MMP-9 are endopeptidases of the MMP family produced by neutrophils, macrophages and monocytes. When infection is associated with leukocyte influx into specific organs, immunopathology and collateral tissue damage may occur. In this review, the involvement of MMPs and, in particular, of gelatinases in both protozoan and helminth infections will be described. In cerebral malaria, for example, MMPs play a role in the pathogenesis of such diseases. Also, trypanosomosis and toxoplasmosis will be considered for protozoan infections, as well as neurocysticercosis and angiostrongyloidosis, as regards helminthiases. All these situations have in common the proteolytic action on the blood brain barrier, mediated by MMPs.
Collapse
Affiliation(s)
- Fabrizio Bruschi
- Department of Translational Research, N.T.M.S., University of Pisa, School of Medicine, Via Roma, 55, 56126, Italy.
| | - Barbara Pinto
- Department of Translational Research, N.T.M.S., University of Pisa, School of Medicine, Via Roma, 55, 56126, Italy.
| |
Collapse
|
28
|
Castillo C, López-Muñoz R, Duaso J, Galanti N, Jaña F, Ferreira J, Cabrera G, Maya JD, Kemmerling U. Role of matrix metalloproteinases 2 and 9 in ex vivo Trypanosoma cruzi infection of human placental chorionic villi. Placenta 2012; 33:991-7. [PMID: 23107342 DOI: 10.1016/j.placenta.2012.10.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2012] [Revised: 10/02/2012] [Accepted: 10/06/2012] [Indexed: 12/11/2022]
Abstract
BACKGROUND Chagas' disease is caused by the haemophlagelated protozoan Trypanosoma cruzi (T. cruzi). During congenital transmission the parasite breaks down the placental barrier. In the present study we analyzed the participation of matrix metalloproteases (MMPs) in the extracellular matrix (ECM) remodeling during T. cruzi ex vivo infection of human placental chorionic villi explants. METHODS Chorionic villi from healthy woman placentas were incubated in the presence or absence of 10⁵ or 10⁶ T. cruzi trypomastigotes (Y strain) with or without the MMPs inhibitor doxycycline. Effective infection was tested measuring parasite DNA by real time PCR (qPCR). MMP-2 and MMP-9 expression were determined by western blotting and immunohistochemistry and their activities were measured by zymography. The effect of MMPs on ECM structure was analyzed histochemically. RESULTS T. cruzi induces the expression and activity of MMP-2 and MMP-9 in chorionic villi. Inhibition of the MMPs prevents the tissue damage induced by T. cruzi and partially decreases the ex vivo infection of the chorionic villi. CONCLUSION MMPs are partially responsible for the ECM changes observed in human chorionic villi during T. cruzi infection and participate in tissue invasion. On the other hand, MMPs may be part of a local placental antiparasitic mechanism.
Collapse
Affiliation(s)
- C Castillo
- Programa de Anatomía y Biología del Desarrollo, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Independencia 1027, Region Metropolitana, Santiago de Chile 8380453, Chile
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Schuindt SHS, Oliveira BCDL, Pimentel PMDO, Resende TL, Retamal CA, DaMatta RA, Seipel D, Arnholdt ACV. Secretion of multi-protein migratory complex induced by Toxoplasma gondii infection in macrophages involves the uPA/uPAR activation system. Vet Parasitol 2012; 186:207-15. [DOI: 10.1016/j.vetpar.2011.11.035] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2010] [Revised: 11/10/2011] [Accepted: 11/14/2011] [Indexed: 12/12/2022]
|
30
|
Toxoplasma on the brain: understanding host-pathogen interactions in chronic CNS infection. J Parasitol Res 2012; 2012:589295. [PMID: 22545203 PMCID: PMC3321570 DOI: 10.1155/2012/589295] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2011] [Accepted: 01/04/2012] [Indexed: 11/18/2022] Open
Abstract
Toxoplasma gondii is a prevalent obligate intracellular parasite which chronically infects more than a third of the world's population. Key to parasite prevalence is its ability to form chronic and nonimmunogenic bradyzoite cysts, which typically form in the brain and muscle cells of infected mammals, including humans. While acute clinical infection typically involves neurological and/or ocular damage, chronic infection has been more recently linked to behavioral changes. Establishment and maintenance of chronic infection involves a balance between the host immunity and parasite evasion of the immune response. Here, we outline the known cellular interplay between Toxoplasma gondii and cells of the central nervous system and review the reported effects of Toxoplasma gondii on behavior and neurological disease. Finally, we review new technologies which will allow us to more fully understand host-pathogen interactions.
Collapse
|
31
|
Geurts N, Opdenakker G, Van den Steen PE. Matrix metalloproteinases as therapeutic targets in protozoan parasitic infections. Pharmacol Ther 2011; 133:257-79. [PMID: 22138604 DOI: 10.1016/j.pharmthera.2011.11.008] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2011] [Accepted: 10/28/2011] [Indexed: 12/11/2022]
Abstract
Matrix metalloproteinases (MMPs) are associated with processes of tissue remodeling and are expressed in all infections with protozoan parasites. We here report the status of MMP research in malaria, trypanosomiasis, leishmaniasis and toxoplasmosis. In all these infections, the balances between MMPs and endogenous MMP inhibitors are disturbed, mostly in favor of active proteolysis. When the infection is associated with leukocyte influx into specific organs, immunopathology and collateral tissue damage may occur. These pathologies include cerebral malaria, sleeping sickness (human African trypanosomiasis), Chagas disease (human American trypanosomiasis), leishmaniasis and toxoplasmic encephalitis in immunocompromised hosts. Destruction of the integrity of the blood-brain barrier (BBB) is a common denominator that may be executed by leukocytic MMPs under the control of host cytokines and chemokines as well as influenced by parasite products. Mechanisms by which parasite-derived products alter host expression of MMP and endogenous MMP inhibitors, have only been described for hemozoin (Hz) in malaria. Hence, understanding these interactions in other parasitic infections remains an important challenge. Furthermore, the involved parasites are also known to produce their own metalloproteinases, and this forms an extra stimulus to investigate MMP inhibitory drugs as therapeutics. MMP inhibitors (MMPIs) may dampen collateral tissue damage, as is anecdotically reported for tetracyclines as MMP regulators in parasite infections.
Collapse
Affiliation(s)
- Nathalie Geurts
- Laboratory of Immunobiology, Rega Institute for Medical Research, University of Leuven, Leuven, Minderbroedersstraat 10, B3000 Leuven, Belgium
| | | | | |
Collapse
|
32
|
Interferon-gamma- and perforin-mediated immune responses for resistance against Toxoplasma gondii in the brain. Expert Rev Mol Med 2011; 13:e31. [PMID: 22005272 DOI: 10.1017/s1462399411002018] [Citation(s) in RCA: 94] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Toxoplasma gondii is an obligate intracellular protozoan parasite that causes various diseases, including lymphadenitis, congenital infection of fetuses and life-threatening toxoplasmic encephalitis in immunocompromised individuals. Interferon-gamma (IFN-γ)-mediated immune responses are essential for controlling tachyzoite proliferation during both acute acquired infection and reactivation of infection in the brain. Both CD4+ and CD8+ T cells produce this cytokine in response to infection, although the latter has more potent protective activity. IFN-γ can activate microglia, astrocytes and macrophages, and these activated cells control the proliferation of tachyzoites using different molecules, depending on cell type and host species. IFN-γ also has a crucial role in the recruitment of T cells into the brain after infection by inducing expression of the adhesion molecule VCAM-1 on cerebrovascular endothelial cells, and chemokines such as CXCL9, CXCL10 and CCL5. A recent study showed that CD8+ T cells are able to remove T. gondii cysts, which represent the stage of the parasite in chronic infection, from the brain through their perforin-mediated activity. Thus, the resistance to cerebral infection with T. gondii requires a coordinated network using both IFN-γ- and perforin-mediated immune responses. Elucidating how these two protective mechanisms function and collaborate in the brain against T. gondii will be crucial in developing a new method to prevent and eradicate this parasitic infection.
Collapse
|
33
|
Welser-Alves JV, Crocker SJ, Milner R. A dual role for microglia in promoting tissue inhibitor of metalloproteinase (TIMP) expression in glial cells in response to neuroinflammatory stimuli. J Neuroinflammation 2011; 8:61. [PMID: 21631912 PMCID: PMC3120696 DOI: 10.1186/1742-2094-8-61] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2011] [Accepted: 06/01/2011] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND By neutralizing the effect of the matrix metalloproteinases (MMPs), the tissue inhibitors of matrix metalloproteinases (TIMPs) play a critical role in maintaining tissue proteolysis in balance. As the major reactive glial cell types in the central nervous system (CNS), microglia and astrocytes play fundamental roles in mediating tissue breakdown and repair. As such, it is important to define the TIMP expression profile in these cells, as well as the mechanisms of regulation by neuroinflammatory stimuli. METHODS Primary mixed glial cultures (MGC), pure microglia, and pure astrocytes were used in this study. To study astrocytes, we employed a recently described pure astrocyte culture system, which has the major advantage of totally lacking microglia. The three different types of culture were treated with lipopolysaccharide (LPS) or individual cytokines, and cell culture supernatants assayed for TIMP-1 or TIMP-2 protein expression by western blot. RESULTS LPS induced TIMP-1 expression in MGC, but not in pure astrocyte or microglial cultures. When pure astrocytes were treated with the cytokines IL-1β, IFN-γ, TNF or TGF-β1, only IL-1β induced TIMP-1 expression. Significantly, astrocyte TIMP-1 expression was restored in LPS-treated astrocyte cultures after the addition of microglia, or conditioned medium taken from LPS-activated microglia (MG-CM). Furthermore, this effect was lost after depletion of IL-1β from MG-CM. By contrast, TIMP-2 was constitutively expressed by astrocytes, whereas microglia expressed TIMP-2 only after exposure to serum. CONCLUSIONS Taken together, these results demonstrate an important concept in glial interactions, by showing that microglia play a central role in regulating glial cell expression of TIMPs, and identify microglial IL-1β as playing a key role in mediating microglial-astrocyte communication.
Collapse
Affiliation(s)
- Jennifer V Welser-Alves
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA
| | | | | |
Collapse
|