1
|
Kwakowsky A, Chawdhary B, de Souza A, Meyer E, Kaye AH, Green CR, Stylli SS, Danesh-Meyer H. Tonabersat Significantly Reduces Disease Progression in an Experimental Mouse Model of Multiple Sclerosis. Int J Mol Sci 2023; 24:17454. [PMID: 38139284 PMCID: PMC10744318 DOI: 10.3390/ijms242417454] [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: 11/30/2023] [Revised: 12/11/2023] [Accepted: 12/11/2023] [Indexed: 12/24/2023] Open
Abstract
Multiple sclerosis (MS) is a neurodegenerative disease marked by chronic neuroinflammation thought to be mediated by the inflammasome pathway. Connexin 43 (Cx43) hemichannels contribute to the activation of the inflammasome through the release of adenosine triphosphate (ATP) inflammasome activation signals. The objective of the study was to evaluate if the Cx43 hemichannel blocker, tonabersat, is effective in modulating the inflammatory response and reducing disability in the myelin oligodendrocyte glycoprotein 35-55-induced experimental autoimmune encephalomyelitis (MOG35-55 EAE) model of MS. Here, we show that the Cx43 hemichannel blocking drug, tonabersat, significantly reduced expression of neuroinflammatory markers for microglial activation (ionized calcium-binding adapter molecule 1 (Iba1)) and astrogliosis (glial fibrillary acidic protein (GFAP)) while preserving myelin basic protein (MBP) expression levels in the corpus callosum, motor cortex, and striatum regions of the brain in MOG35-55 EAE mice. Reduced NOD-like receptor protein 3 (NLRP3) inflammasome complex assembly and Caspase-1 activation confirmed the drug's mode of action. MOG35-55 EAE mice showed clinical signs of MS, but MOG35-55 EAE mice treated with tonabersat retained behavior closer to normal. These data suggest that clinical trial phase IIb-ready tonabersat may merit further investigation as a promising candidate for MS treatment.
Collapse
Affiliation(s)
- Andrea Kwakowsky
- Department of Anatomy and Medical Imaging, Centre for Brain Research, Faculty of Medical and Health Science, University of Auckland, Auckland 1023, New Zealand; (B.C.); (A.d.S.); (E.M.)
- Pharmacology and Therapeutics, Galway Neuroscience Centre, School of Medicine, Ollscoil na Gaillimhe—University of Galway, H91 W5P7 Galway, Ireland
| | - Bhavya Chawdhary
- Department of Anatomy and Medical Imaging, Centre for Brain Research, Faculty of Medical and Health Science, University of Auckland, Auckland 1023, New Zealand; (B.C.); (A.d.S.); (E.M.)
- Department of Opthalmology, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1023, New Zealand; (C.R.G.); (H.D.-M.)
| | - Antonio de Souza
- Department of Anatomy and Medical Imaging, Centre for Brain Research, Faculty of Medical and Health Science, University of Auckland, Auckland 1023, New Zealand; (B.C.); (A.d.S.); (E.M.)
- Department of Opthalmology, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1023, New Zealand; (C.R.G.); (H.D.-M.)
| | - Emily Meyer
- Department of Anatomy and Medical Imaging, Centre for Brain Research, Faculty of Medical and Health Science, University of Auckland, Auckland 1023, New Zealand; (B.C.); (A.d.S.); (E.M.)
- Department of Opthalmology, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1023, New Zealand; (C.R.G.); (H.D.-M.)
| | - Andrew H. Kaye
- Department of Surgery, University of Melbourne, Melbourne, VIC 3010, Australia; (A.H.K.); (S.S.S.)
- Department of Neurosurgery, Hadassah Hebrew University Hospital, Jerusalem 91120, Israel
| | - Colin R. Green
- Department of Opthalmology, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1023, New Zealand; (C.R.G.); (H.D.-M.)
| | - Stanley S. Stylli
- Department of Surgery, University of Melbourne, Melbourne, VIC 3010, Australia; (A.H.K.); (S.S.S.)
- Department of Neurosurgery, The Royal Melbourne Hospital, Parkville, VIC 3052, Australia
| | - Helen Danesh-Meyer
- Department of Opthalmology, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1023, New Zealand; (C.R.G.); (H.D.-M.)
| |
Collapse
|
2
|
Zhang H, Gao J, Tang Y, Jin T, Tao J. Inflammasomes cross-talk with lymphocytes to connect the innate and adaptive immune response. J Adv Res 2023; 54:181-193. [PMID: 36681114 DOI: 10.1016/j.jare.2023.01.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Revised: 10/15/2022] [Accepted: 01/13/2023] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Innate and adaptive immunity are two different parts of the immune system that have different characteristics and work together to provide immune protection. Inflammasomes are a major part of the innate immune system that are expressed widely in myeloid cells and are responsible for inflammatory responses. Recent studies have shown that inflammasomes are also expressed and activated in lymphocytes, especially in T and B cells, to regulate the adaptive immune response. Activation of inflammasomes is also under the control of lymphocytes. Therefore, we propose that inflammasomes act as a bridge and they provide crosstalk between the innate and adaptive immune systems to obtain a fine balance in immune responses. AIM OF REVIEW This review systematially summarizes the interaction between inflammasomes and lymphocytes and describes the crosstalk between the innate and adaptive immune systems induced by inflammasomes, with the aim of providing new directions and important areas for further research. KEY SCIENTIFIC CONCEPTS OF REVIEW When considering the novel function of inflammasomes in various lymphocytes, attention should be given to the activity of specific inflammasomes in studies of lymphocyte function. Moreover, research on the function of various inflammasomes in lymphocytes will help advance knowledge on the mechanisms and treatment of various diseases, including autoimmune diseases and tumors. In addition, when studying inflammatory responses, inflammasomes in both lymphocytes and myeloid cells need to be considered.
Collapse
Affiliation(s)
- Hongliang Zhang
- Department of Rheumatology and Immunology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; College of Medicine and Health, Lishui University, No. 1 Xueyuan Road, Liandu District, Lishui 323000, China
| | - Jie Gao
- Department of Rheumatology and Immunology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Yujie Tang
- Department of Rheumatology and Immunology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Tengchuan Jin
- Laboratory of Structural Immunology, CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
| | - Jinhui Tao
- Department of Rheumatology and Immunology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
| |
Collapse
|
3
|
Reinmuth L, Hsiao CC, Hamann J, Rosenkilde M, Mackrill J. Multiple Targets for Oxysterols in Their Regulation of the Immune System. Cells 2021; 10:cells10082078. [PMID: 34440846 PMCID: PMC8391951 DOI: 10.3390/cells10082078] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 08/05/2021] [Accepted: 08/11/2021] [Indexed: 02/07/2023] Open
Abstract
Oxysterols, or cholesterol oxidation products, are naturally occurring lipids which regulate the physiology of cells, including those of the immune system. In contrast to effects that are mediated through nuclear receptors or by epigenetic mechanism, which take tens of minutes to occur, changes in the activities of cell-surface receptors caused by oxysterols can be extremely rapid, often taking place within subsecond timescales. Such cell-surface receptor effects of oxysterols allow for the regulation of fast cellular processes, such as motility, secretion and endocytosis. These cellular processes play critical roles in both the innate and adaptive immune systems. This review will survey the two broad classes of cell-surface receptors for oxysterols (G-protein coupled receptors (GPCRs) and ion channels), the mechanisms by which cholesterol oxidation products act on them, and their presence and functions in the different cell types of the immune system. Overall, this review will highlight the potential of oxysterols, synthetic derivatives and their receptors for physiological and therapeutic modulation of the immune system.
Collapse
Affiliation(s)
- Lisa Reinmuth
- Laboratory for Molecular Pharmacology, Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark;
| | - Cheng-Chih Hsiao
- Department of Experimental Immunology, Amsterdam Institute for Infection and Immunity, Amsterdam University Medical Centers, Meibergdreef 9, 1105AZ Amsterdam, The Netherlands; (C.-C.H.); (J.H.)
- Neuroimmunology Research Group, The Netherlands Institute for Neuroscience, 1105BA Amsterdam, The Netherlands
| | - Jörg Hamann
- Department of Experimental Immunology, Amsterdam Institute for Infection and Immunity, Amsterdam University Medical Centers, Meibergdreef 9, 1105AZ Amsterdam, The Netherlands; (C.-C.H.); (J.H.)
- Neuroimmunology Research Group, The Netherlands Institute for Neuroscience, 1105BA Amsterdam, The Netherlands
| | - Mette Rosenkilde
- Laboratory for Molecular Pharmacology, Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark;
- Correspondence: (M.R.); (J.M.); Tel.: +353-(0)21-490-1400 (J.M.)
| | - John Mackrill
- Department of Physiology, School of Medicine, BioSciences Institute, University College Cork, College Road, Cork T12 YT20, Ireland
- Correspondence: (M.R.); (J.M.); Tel.: +353-(0)21-490-1400 (J.M.)
| |
Collapse
|
4
|
Shi W, Qiu Q, Feng Z, Tong Z, Guo W, Zou F, Yue N, Huang W, Qian H. Design, synthesis and immunological evaluation of self-assembled antigenic peptides from dual-antigen targets: a broad-spectrum candidate for an effective antibreast cancer therapy. J Immunother Cancer 2021; 9:jitc-2021-002523. [PMID: 34083420 PMCID: PMC8183215 DOI: 10.1136/jitc-2021-002523] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/02/2021] [Indexed: 12/11/2022] Open
Abstract
Background Considering the narrow immune response spectrum of a single epitope, and the nanoparticles (NPs) as a novel adjuvant can achieve efficient delivery of antigenic peptides safely, a nano-system (denoted as DSPE-PEG-Man@EM-NPs) based on cathepsin B-responsive antigenic peptides was designed and synthesized. Methods Highly affinitive antigenic peptides were delivered by self-assembled NPs, and targeted erythrocyte membranes acted as a peptide carrier to improve antigenic peptides presentation and to strengthen cytotoxic T-cells reaction. Cathepsin B coupling could release antigenic peptides rapidly in dendritic cells. Results Evaluations showed that DSPE-PEG-Man@EM-NPs had obvious inhibitory effects towards both MCF-7 and MDA-MB-231 human breast cancer cell lines. Conclusion Overall, this strategy provides a novel strategy for boosting cytotoxic T lymphocytes response, thereby expanding the adaptation range of tumor antigenic peptides and improving the therapeutic effect of tumor immunotherapy with nanomedicine.
Collapse
Affiliation(s)
- Wei Shi
- Center of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, China
| | - Qianqian Qiu
- Center of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, China.,School of Pharmacy, Jiangsu Provincial Key Laboratory of Coastal Wetland Bioresources and Environmental Protection, Yancheng Teachers' University, Yancheng 224002, China
| | - Ziying Feng
- Center of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, China
| | - Zhenzhen Tong
- Center of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, China
| | - Weiwei Guo
- Center of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, China
| | - Feng Zou
- Center of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, China
| | - Na Yue
- Center of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, China
| | - Wenlong Huang
- Center of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, China.,Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, Jiangsu, China
| | - Hai Qian
- Center of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, China .,Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, Jiangsu, China
| |
Collapse
|
5
|
Malhotra S, Costa C, Eixarch H, Keller CW, Amman L, Martínez-Banaclocha H, Midaglia L, Sarró E, Machín-Díaz I, Villar LM, Triviño JC, Oliver-Martos B, Parladé LN, Calvo-Barreiro L, Matesanz F, Vandenbroeck K, Urcelay E, Martínez-Ginés ML, Tejeda-Velarde A, Fissolo N, Castilló J, Sanchez A, Robertson AAB, Clemente D, Prinz M, Pelegrin P, Lünemann JD, Espejo C, Montalban X, Comabella M. NLRP3 inflammasome as prognostic factor and therapeutic target in primary progressive multiple sclerosis patients. Brain 2020; 143:1414-1430. [PMID: 32282893 DOI: 10.1093/brain/awaa084] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2019] [Revised: 12/13/2019] [Accepted: 02/05/2020] [Indexed: 01/24/2023] Open
Abstract
Primary progressive multiple sclerosis is a poorly understood disease entity with no specific prognostic biomarkers and scarce therapeutic options. We aimed to identify disease activity biomarkers in multiple sclerosis by performing an RNA sequencing approach in peripheral blood mononuclear cells from a discovery cohort of 44 untreated patients with multiple sclerosis belonging to different clinical forms and activity phases of the disease, and 12 healthy control subjects. A validation cohort of 58 patients with multiple sclerosis and 26 healthy control subjects was included in the study to replicate the RNA sequencing findings. The RNA sequencing revealed an interleukin 1 beta (IL1B) signature in patients with primary progressive multiple sclerosis. Subsequent immunophenotyping pointed to blood monocytes as responsible for the IL1B signature observed in this group of patients. Functional experiments at baseline measuring apoptosis-associated speck-like protein containing a CARD (ASC) speck formation showed that the NOD-leucine rich repeat and pyrin containing protein 3 (NLRP3) inflammasome was overactive in monocytes from patients with primary progressive multiple sclerosis, and canonical NLRP3 inflammasome activation with a combination of ATP plus lipopolysaccharide was associated with increased IL1B production in this group of patients. Primary progressive multiple sclerosis patients with high IL1B gene expression levels in peripheral blood mononuclear cells progressed significantly faster compared to patients with low IL1B levels based on the time to reach an EDSS of 6.0 and the Multiple Sclerosis Severity Score. In agreement with peripheral blood findings, both NLRP3 and IL1B expression in brain tissue from patients with primary progressive multiple sclerosis was mainly restricted to cells of myeloid lineage. Treatment of mice with a specific NLRP3 inflammasome inhibitor attenuated established experimental autoimmune encephalomyelitis disease severity and improved CNS histopathology. NLRP3 inflammasome-specific inhibition was also effective in reducing axonal damage in a model of lipopolysaccharide-neuroinflammation using organotypic cerebellar cultures. Altogether, these results point to a role of IL1B and the NLRP3 inflammasome as prognostic biomarker and potential therapeutic target, respectively, in patients with primary progressive multiple sclerosis.
Collapse
Affiliation(s)
- Sunny Malhotra
- Servei de Neurologia-Neuroimmunologia, Centre d'Esclerosi Múltiple de Catalunya (Cemcat), Institut de Recerca Vall d'Hebron (VHIR), Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Carme Costa
- Servei de Neurologia-Neuroimmunologia, Centre d'Esclerosi Múltiple de Catalunya (Cemcat), Institut de Recerca Vall d'Hebron (VHIR), Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Herena Eixarch
- Servei de Neurologia-Neuroimmunologia, Centre d'Esclerosi Múltiple de Catalunya (Cemcat), Institut de Recerca Vall d'Hebron (VHIR), Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Christian W Keller
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Münster, Germany.,Institute of Experimental Immunology, Laboratory of Neuroinflammation, University of Zurich, Zurich, Switzerland
| | - Lukas Amman
- Institute of Neuropathology, Medical Faculty, University of Freiburg, Freiburg, Germany.,Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Helios Martínez-Banaclocha
- Biomedical Research Institute of Murcia (IMIB-Arrixaca), University Clinical Hospital Virgen de la Arrixaca, Murcia, Spain
| | - Luciana Midaglia
- Servei de Neurologia-Neuroimmunologia, Centre d'Esclerosi Múltiple de Catalunya (Cemcat), Institut de Recerca Vall d'Hebron (VHIR), Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Eduard Sarró
- Renal Physiopathology Group, Institut de Recerca Vall d'Hebron (VHIR) - CIBBIM Nanomedicine, Barcelona, Spain
| | - Isabel Machín-Díaz
- Grupo de Neuroinmuno-Reparación, Hospital Nacional de Parapléjicos-SESCAM, Toledo, Spain
| | - Luisa M Villar
- Departments of Immunology and Neurology, Multiple Sclerosis Unit, Hospital Ramon y Cajal, (IRYCIS), Madrid, Spain
| | | | - Begoña Oliver-Martos
- Neuroimmunology and Neuroinflammation Group, Instituto de Investigación Biomédica de Málaga-IBIMA, UGC Neurociencias, Hospital Regional Universitario de Málaga, Málaga, Spain
| | - Laura Navarro Parladé
- Servei de Neurologia-Neuroimmunologia, Centre d'Esclerosi Múltiple de Catalunya (Cemcat), Institut de Recerca Vall d'Hebron (VHIR), Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Laura Calvo-Barreiro
- Servei de Neurologia-Neuroimmunologia, Centre d'Esclerosi Múltiple de Catalunya (Cemcat), Institut de Recerca Vall d'Hebron (VHIR), Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Fuencisla Matesanz
- Department of Cell Biology and Immunology, Instituto de Parasitología y Biomedicina "López Neyra", Consejo Superior de Investigaciones Científicas (IPBLN-CSIC), Granada, Spain
| | - Koen Vandenbroeck
- Universidad del País Vasco (UPV/EHU), Leioa, Spain.,IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| | - Elena Urcelay
- Hospital Clínico San Carlos, Instituto de Investigación Sanitaria San Carlos (IdISSC), Madrid, Spain
| | | | - Amalia Tejeda-Velarde
- Departments of Immunology and Neurology, Multiple Sclerosis Unit, Hospital Ramon y Cajal, (IRYCIS), Madrid, Spain
| | - Nicolás Fissolo
- Servei de Neurologia-Neuroimmunologia, Centre d'Esclerosi Múltiple de Catalunya (Cemcat), Institut de Recerca Vall d'Hebron (VHIR), Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Joaquín Castilló
- Servei de Neurologia-Neuroimmunologia, Centre d'Esclerosi Múltiple de Catalunya (Cemcat), Institut de Recerca Vall d'Hebron (VHIR), Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Alex Sanchez
- Department of Genetics, Microbiology and Statistics, Universitat de Barcelona, Barcelona, Spain.,Statistics and Bioinformatics Unit, Vall d'Hebron Institut de Recerca, Barcelona, Spain
| | - Avril A B Robertson
- School of Chemistry and Molecular Biosciences, University of Queensland, Brisbane, Australia
| | - Diego Clemente
- Grupo de Neuroinmuno-Reparación, Hospital Nacional de Parapléjicos-SESCAM, Toledo, Spain
| | - Marco Prinz
- Institute of Neuropathology, Medical Faculty, University of Freiburg, Freiburg, Germany.,Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany.,Center for NeuroModulation (NeuroModul), Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Pablo Pelegrin
- Biomedical Research Institute of Murcia (IMIB-Arrixaca), University Clinical Hospital Virgen de la Arrixaca, Murcia, Spain
| | - Jan D Lünemann
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Münster, Germany.,Institute of Experimental Immunology, Laboratory of Neuroinflammation, University of Zurich, Zurich, Switzerland
| | - Carmen Espejo
- Servei de Neurologia-Neuroimmunologia, Centre d'Esclerosi Múltiple de Catalunya (Cemcat), Institut de Recerca Vall d'Hebron (VHIR), Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Xavier Montalban
- Servei de Neurologia-Neuroimmunologia, Centre d'Esclerosi Múltiple de Catalunya (Cemcat), Institut de Recerca Vall d'Hebron (VHIR), Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain.,Center for Multiple Sclerosis, St, Michael's Hospital, University of Toronto, Toronto, ON, Canada
| | - Manuel Comabella
- Servei de Neurologia-Neuroimmunologia, Centre d'Esclerosi Múltiple de Catalunya (Cemcat), Institut de Recerca Vall d'Hebron (VHIR), Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| |
Collapse
|
6
|
Kadowaki A, Quintana FJ. The NLRP3 inflammasome in progressive multiple sclerosis. Brain 2020; 143:1286-1288. [PMID: 32438410 DOI: 10.1093/brain/awaa135] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
This scientific commentary refers to ‘NLRP3 inflammasome as prognostic factor and therapeutic target in primary progressive multiple sclerosis patients’, by Malhotra et al. (doi:10.1093/brain/awaa084).
Collapse
Affiliation(s)
- Atsushi Kadowaki
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Francisco J Quintana
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.,Broad Institute of MIT and Harvard, Cambridge, MA, USA
| |
Collapse
|
7
|
Mehra V, Rhone E, Widya S, Zuckerman M, Potter V, Raj K, Kulasekararaj A, McLornan D, de Lavallade H, Benson-Quarm N, Lim C, Ware S, Sudhanva M, Malik O, Nicholas R, Muraro PA, Marsh J, Mufti GJ, Silber E, Pagliuca A, Kazmi MA. Epstein-Barr Virus and Monoclonal Gammopathy of Clinical Significance in Autologous Stem Cell Transplantation for Multiple Sclerosis. Clin Infect Dis 2020; 69:1757-1763. [PMID: 30980715 DOI: 10.1093/cid/ciz047] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2018] [Accepted: 01/14/2019] [Indexed: 12/24/2022] Open
Abstract
INTRODUCTION Autologous hematopoietic stem cell transplantation (AHSCT) with anti-thymocyte globulin (ATG) conditioning as treatment of active multiple sclerosis (MS) is rapidly increasing across Europe (EBMT registry data 2017). Clinically significant Epstein-Barr virus reactivation (EBV-R) following AHSCT with ATG for severe autoimmune conditions is an underrecognized complication relative to T-cell deplete transplants performed for hematological diseases. This retrospective study reports EBV-R associated significant clinical sequelae in MS patients undergoing AHSCT with rabbit ATG. METHODS Retrospective data were analyzed for 36 consecutive MS-AHSCT patients at Kings College Hospital, London. All patients routinely underwent weekly EBV DNA polymerase chain reaction monitoring and serum electrophoresis for monoclonal gammopathy (MG or M-protein). EBV-R with rising Epstein-Barr viral load, M-protein, and associated clinical sequelae were captured from clinical records. RESULTS All patients had evidence of rising EBV DNA-emia, including 7 who were lost to long-term follow-up, with a number of them developing high EBV viral load and associated lymphoproliferative disorder (LPD). Nearly 72% (n = 18/29) developed de novo MG, some with significant neurological consequences with high M-protein and EBV-R. Six patients required anti-CD20 therapy (rituximab) with complete resolution of EBV related symptoms. Receiver operating characteristics estimated a peak EBV viremia of >500 000 DNA copies/mL correlated with high sensitivity (85.5%) and specificity (82.5%) (area under the curve: 0.87; P = .004) in predicting EBV-R related significant clinical events. CONCLUSION Symptomatic EBV reactivation increases risk of neurological sequelae and LPD in MS-AHSCT. We recommend regular monitoring for EBV and serum electrophoresis for MG in MS patients in the first 3 months post-AHSCT.
Collapse
Affiliation(s)
- Varun Mehra
- Department of Hematology, King's College Hospital NHS Foundation Trust, Denmark Hill
| | - Elijah Rhone
- Department of Neurology, King's College Hospital NHS Foundation Trust, Denmark Hill
| | - Stefani Widya
- GKT School of Medical Education, Kings College London University
| | - Mark Zuckerman
- Department of Virology, King's College Hospital NHS Foundation Trust, Denmark Hill
| | - Victoria Potter
- Department of Hematology, King's College Hospital NHS Foundation Trust, Denmark Hill
| | - Kavita Raj
- Department of Hematology, King's College Hospital NHS Foundation Trust, Denmark Hill.,Department of Hematology, Guy's and St. Thomas' NHS Foundation Trust
| | - Austin Kulasekararaj
- Department of Hematology, King's College Hospital NHS Foundation Trust, Denmark Hill
| | - Donal McLornan
- Department of Hematology, King's College Hospital NHS Foundation Trust, Denmark Hill.,Department of Hematology, Guy's and St. Thomas' NHS Foundation Trust
| | - Hugues de Lavallade
- Department of Hematology, King's College Hospital NHS Foundation Trust, Denmark Hill
| | - Nana Benson-Quarm
- Department of Hematology, King's College Hospital NHS Foundation Trust, Denmark Hill
| | - Christina Lim
- Department of Hematology, King's College Hospital NHS Foundation Trust, Denmark Hill
| | - Sarah Ware
- Department of Hematology, King's College Hospital NHS Foundation Trust, Denmark Hill
| | - Malur Sudhanva
- Department of Virology, King's College Hospital NHS Foundation Trust, Denmark Hill
| | - Omar Malik
- Department of Neurology, Imperial College Healthcare, United Kingdom
| | - Richard Nicholas
- Department of Neurology, Imperial College Healthcare, United Kingdom
| | - Paolo A Muraro
- Department of Neurology, Imperial College Healthcare, United Kingdom.,Department of Neuroimmunology, Imperial College London, United Kingdom
| | - Judith Marsh
- Department of Hematology, King's College Hospital NHS Foundation Trust, Denmark Hill
| | - Ghulam J Mufti
- Department of Hematology, King's College Hospital NHS Foundation Trust, Denmark Hill
| | - Eli Silber
- Department of Neurology, King's College Hospital NHS Foundation Trust, Denmark Hill
| | - Antonio Pagliuca
- Department of Hematology, King's College Hospital NHS Foundation Trust, Denmark Hill
| | - Majid A Kazmi
- Department of Hematology, King's College Hospital NHS Foundation Trust, Denmark Hill.,Department of Hematology, Guy's and St. Thomas' NHS Foundation Trust
| |
Collapse
|
8
|
Sun X, Pang H, Li J, Luo S, Huang G, Li X, Xie Z, Zhou Z. The NLRP3 Inflammasome and Its Role in T1DM. Front Immunol 2020; 11:1595. [PMID: 32973739 PMCID: PMC7481449 DOI: 10.3389/fimmu.2020.01595] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Accepted: 06/16/2020] [Indexed: 12/15/2022] Open
Abstract
The NLRP3 (nucleotide-binding and oligomerization domain-like receptor family pyrin domain-containing 3) inflammasome is a protein complex expressed in cells. It detects danger signals and induces the production of active caspase-1 and the maturation and release of IL (interleukin)-33, IL-18, IL-1β and other cytokines. T1DM (type 1 diabetes mellitus) is defined as a chronic autoimmune disorder characterized by the autoreactive T cell-mediated elimination of insulin-positive pancreatic beta-cells. Although the exact underlying mechanisms are obscure, researchers have proposed that both environmental and genetic factors are involved in the pathogenesis of T1DM. Furthermore, immune responses, including innate and adaptive immunity, play an important role in this process. Recently, the NLRP3 inflammasome, a critical component of innate immunity, was reported to be associated with T1DM. Here, we review the assembly and function of the NLRP3 inflammasome. In addition, the activation and regulatory mechanisms that enhance or attenuate NLRP3 inflammasome activation are discussed. Finally, we focus on the relationship between the NLRP3 inflammasome and T1DM, as well as its potential value for clinical use.
Collapse
Affiliation(s)
- Xiaoxiao Sun
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China.,Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, National Clinical Research Center for Metabolic Diseases, Changsha, China
| | - Haipeng Pang
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China.,Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, National Clinical Research Center for Metabolic Diseases, Changsha, China
| | - Jiaqi Li
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China.,Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, National Clinical Research Center for Metabolic Diseases, Changsha, China
| | - Shuoming Luo
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China.,Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, National Clinical Research Center for Metabolic Diseases, Changsha, China
| | - Gan Huang
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China.,Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, National Clinical Research Center for Metabolic Diseases, Changsha, China
| | - Xia Li
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China.,Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, National Clinical Research Center for Metabolic Diseases, Changsha, China
| | - Zhiguo Xie
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China.,Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, National Clinical Research Center for Metabolic Diseases, Changsha, China
| | - Zhiguang Zhou
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China.,Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, National Clinical Research Center for Metabolic Diseases, Changsha, China
| |
Collapse
|
9
|
Advances in the molecular mechanisms of NLRP3 inflammasome activators and inactivators. Biochem Pharmacol 2020; 175:113863. [PMID: 32081791 DOI: 10.1016/j.bcp.2020.113863] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Accepted: 02/13/2020] [Indexed: 12/15/2022]
Abstract
NLRP3 inflammasome is an intracellular protein complex that initiates cellular injury via assembly of NLRP3, ASC and caspase-1 in response to microbial infection and sterile stressors. The importance of NLRP3 inflammasome in immunity and human diseases has been well documented. Up to now, targeted inhibition of the assembly of NLRP3 inflammasome complex and of its activation was thought to be therapeutic strategy for associated diseases. Recent studies show that a host of molecules such as NIMA-related kinase 7 (Nek7) and DEAD-box helicase 3 X-linked (DDX3X) and a large number of biological mediators including cytokines, microRNAs, nitric oxide, carbon monoxide, nuclear factor erythroid-2 related factor 2 (Nrf2) and cellular autophagy participate in the activation and inactivation of NLRP3 inflammasome. This review summarizes current understanding of the molecular basis of NLRP3 inflammasome activation and inactivation. This knowledge may lead to development of new therapies directed at NLRP3 inflammasome related diseases.
Collapse
|
10
|
Olcum M, Tastan B, Kiser C, Genc S, Genc K. Microglial NLRP3 inflammasome activation in multiple sclerosis. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2019; 119:247-308. [PMID: 31997770 DOI: 10.1016/bs.apcsb.2019.08.007] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Multiple sclerosis (MS) is a chronic, autoimmune and neuroinflammatory disease of the central nervous system (CNS) mediated by autoreactive T cells directed against myelin antigens. Although the crucial role of adaptive immunity is well established in MS, the contribution of innate immunity has only recently been appreciated. Microglia are the main innate immune cells of the CNS. Similar to other myeloid cells, microglia recognize both exogenous and host-derived endogenous danger signals through pattern recognition receptors (PRRs) localized on their cell surface such as Toll Like receptor 4, or in the cytosol such as NLRP3. The second one is the sensor protein of the multi-molecular NLRP3 inflammasome complex in activated microglia that promotes the maturation and secretion of proinflammatory cytokines, interleukin-1β and interleukin-18. Overactivation of microglia and aberrant activation of the NLRP3 inflammasome have been implicated in the pathogenesis of MS. Indeed, experimental data, together with post-mortem and clinical studies have revealed an increased expression of NLRP3 inflammasome complex elements in microglia and other immune cells. In this review, we focus on microglial NLRP3 inflammasome activation in MS. First, we overview the basic knowledge about MS, microglia and the NLRP3 inflammasome. Then, we summarize studies about microglial NLRP3 inflammasome activation in MS and its animal models. We also highlight experimental therapeutic approaches that target different steps of NLRP inflammasome activation. Finally, we discuss future research avenues and new methods in this rapidly evolving area.
Collapse
Affiliation(s)
- Melis Olcum
- Izmir Biomedicine and Genome Center, Dokuz Eylul University Health Campus Balcova, Izmir, Turkey
| | - Bora Tastan
- Izmir Biomedicine and Genome Center, Dokuz Eylul University Health Campus Balcova, Izmir, Turkey; Izmir International Biomedicine and Genome Institute (iBG-Izmir), Dokuz Eylul University Health Campus, Balcova, Izmir, Turkey
| | - Cagla Kiser
- Izmir Biomedicine and Genome Center, Dokuz Eylul University Health Campus Balcova, Izmir, Turkey; Izmir International Biomedicine and Genome Institute (iBG-Izmir), Dokuz Eylul University Health Campus, Balcova, Izmir, Turkey
| | - Sermin Genc
- Izmir Biomedicine and Genome Center, Dokuz Eylul University Health Campus Balcova, Izmir, Turkey; Izmir International Biomedicine and Genome Institute (iBG-Izmir), Dokuz Eylul University Health Campus, Balcova, Izmir, Turkey; Department of Neuroscience, Institute of Health and Science, Dokuz Eylul University Health Campus, Balcova, Izmir, Turkey
| | - Kursad Genc
- Department of Neuroscience, Institute of Health and Science, Dokuz Eylul University Health Campus, Balcova, Izmir, Turkey
| |
Collapse
|
11
|
Vališ M, Sobíšek L, Vyšata O, Klímová B, Andrýs C, Vokurková D, Masopust J, Pavelek Z. CD4+/CD45RO+: A Potential Biomarker of the Clinical Response to Glatiramer Acetate. Cells 2019; 8:cells8050456. [PMID: 31096621 PMCID: PMC6562382 DOI: 10.3390/cells8050456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 05/10/2019] [Accepted: 05/11/2019] [Indexed: 11/26/2022] Open
Abstract
Background: Glatiramer acetate (GA) is an effective treatment for the earliest stages of multiple sclerosis (MS)—clinically isolated syndrome (CIS) or clinically definite MS (CDMS). Objective: This study aims to determine the differences in the lymphocyte population (at baseline and the course of five years) between confirmed sustained progression (CSP) and non-CSP groups and to identify potential biomarkers among these parameters that can predict a positive response to the treatment. Methods: Twelve male and 60 female patients were included in the study. Peripheral blood samples were collected before and five years after treatment with GA. The authors compared lymphocyte parameters between the CSP and non-CSP groups by statistical analyses. Univariate and penalized logistic regression models were fitted to identify the best lymphocyte parameters at baseline and their combination for potential biomarkers. Subsequently, the ROC analysis was used to identify cut-offs for selected parameters. Results: The parameter CD4+/CD45RO+ was identified as the best single potential biomarker, demonstrating the ability to identify patients with CSP. Moreover, a combination of four lymphocyte parameters at baseline, relative lymphocyte counts, CD3+/CD69+, CD4+/CD45RO+, and CD4+/CD45RA+ab, was identified as a potential composite biomarker. This combination explains 23% of the variability in CSP, which is better than the best univariate parameter when compared to CD4+/CD45RO+ at baseline. Conclusions: The results suggest that other biomarkers can help monitor the conditions of patients and predict a favourable outcome.
Collapse
Affiliation(s)
- Martin Vališ
- Department of Neurology, Faculty of Medicine and University Hospital Hradec Králové, Charles University in Prague, Sokolská 581, 500 05 Hradec Králové, Czech Republic.
| | - Lukáš Sobíšek
- Department of Neurology, Faculty of Medicine and University Hospital Hradec Králové, Charles University in Prague, Sokolská 581, 500 05 Hradec Králové, Czech Republic.
| | - Oldřich Vyšata
- Department of Neurology, Faculty of Medicine and University Hospital Hradec Králové, Charles University in Prague, Sokolská 581, 500 05 Hradec Králové, Czech Republic.
| | - Blanka Klímová
- Department of Neurology, Faculty of Medicine and University Hospital Hradec Králové, Charles University in Prague, Sokolská 581, 500 05 Hradec Králové, Czech Republic.
| | - Ctirad Andrýs
- Department of Clinical Immunology and Allergology, University Hospital Hradec Králové, Sokolská 581, 500 05 Hradec Králové, Czech Republic.
| | - Doris Vokurková
- Department of Clinical Immunology and Allergology, University Hospital Hradec Králové, Sokolská 581, 500 05 Hradec Králové, Czech Republic.
| | - Jiří Masopust
- Department of Neurology, Faculty of Medicine and University Hospital Hradec Králové, Charles University in Prague, Sokolská 581, 500 05 Hradec Králové, Czech Republic.
| | - Zbyšek Pavelek
- Department of Neurology, Faculty of Medicine and University Hospital Hradec Králové, Charles University in Prague, Sokolská 581, 500 05 Hradec Králové, Czech Republic.
| |
Collapse
|
12
|
Imani D, Azimi A, Salehi Z, Rezaei N, Emamnejad R, Sadr M, Izad M. Association of nod-like receptor protein-3 single nucleotide gene polymorphisms and expression with the susceptibility to relapsing-remitting multiple sclerosis. Int J Immunogenet 2018; 45:329-336. [DOI: 10.1111/iji.12401] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 05/13/2018] [Accepted: 08/16/2018] [Indexed: 01/18/2023]
Affiliation(s)
- Danyal Imani
- Immunology Department, School of Medicine; Tehran University of Medical Sciences; Tehran Iran
| | - Amirreza Azimi
- Neurology Department, School of Medicine; Tehran University of Medical Sciences; Tehran Iran
- MS Research Center, Neuroscience Institute; Tehran University of Medical Sciences; Tehran Iran
| | - Zahra Salehi
- Immunology Department, School of Medicine; Tehran University of Medical Sciences; Tehran Iran
| | - Nima Rezaei
- Immunology Department, School of Medicine; Tehran University of Medical Sciences; Tehran Iran
- Molecular Immunology Research Center; Tehran University of Medical Sciences; Tehran Iran
| | - Rahimeh Emamnejad
- Immunology Department, School of Medicine; Shahre-Kord University of Medical Sciences, Shahre-Kord; Iran
| | - Maryam Sadr
- Molecular Immunology Research Center; Tehran University of Medical Sciences; Tehran Iran
| | - Maryam Izad
- Immunology Department, School of Medicine; Tehran University of Medical Sciences; Tehran Iran
- MS Research Center, Neuroscience Institute; Tehran University of Medical Sciences; Tehran Iran
| |
Collapse
|
13
|
Role of Inflammasomes in Neuroimmune and Neurodegenerative Diseases: A Systematic Review. Mediators Inflamm 2018; 2018:1549549. [PMID: 29849483 PMCID: PMC5932495 DOI: 10.1155/2018/1549549] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Revised: 12/26/2017] [Accepted: 01/01/2018] [Indexed: 12/31/2022] Open
Abstract
Inflammasomes are multiprotein complexes that can sense pathogen-associated molecular patterns and damage-associated molecular signals. They are involved in the initiation and development of inflammation via activation of IL-1β and IL-18. Many recent studies suggest a strong correlation between inflammasomes and neurological diseases, such as multiple sclerosis (MS), Alzheimer's disease (AD), and Parkinson's disease (PD). Several components of inflammasomes, such as nucleotide-binding oligomerization domain- (NOD-) like receptor, absent in melanoma 2- (AIM2-) like receptors (ALRs), apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC), and caspase-1, as well as the upstream factors and downstream effectors, are associated with the initiation and development of MS and its animal model, experimental autoimmune encephalomyelitis. Additionally, inflammasomes affect the efficacy of interferon-β therapy in patients with MS. Finally, the strong association of inflammasomes with AD and PD needs to be further studied. In this review of latest literatures, we comprehensively tease out diverse roles of different kinds of inflammasomes in neuroimmune and neurodegenerative diseases, especially in the perspective of double roles involved in pathogenesis, and identify future research priorities.
Collapse
|
14
|
Pavelek Z, Vyšata O, Klímová B, Andrýs C, Vokurková D, Vališ M. Lymphocytes in the treatment with interferon beta-1 b. Mult Scler Relat Disord 2017; 18:29-32. [PMID: 29141817 DOI: 10.1016/j.msard.2017.08.020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 08/18/2017] [Accepted: 08/31/2017] [Indexed: 11/28/2022]
Abstract
BACKGROUND Multiple sclerosis (MS) is a chronic inflammatory demyelinating and neurodegenerative disease affecting the central nervous system. One of the basic medications for the treatment of a clinically isolated syndrome (CIS) or relapsing-remitting MS is interferon beta (INFβ). Although the exact mechanism of its effects is unknown, the medication has an anti-inflammatory and immunomodulatory effect. The goal of this study was to determine the characters which are affected in patients treated with INFβ. METHODS A total of 97 patients (25 males and 72 females) were included into the study. Patients were treated by INFβ 1-b (subcutaneous injection, 250µg, each other day). Clinical evaluations were performed by an attending neurologist. Peripheral blood samples were obtained just prior to treatment and 5 years after INFβ 1-b. Statistical analysis and processing of the obtained data were performed by using a comprehensive statistical software package MATLAB®. RESULTS A significant decrease of the observed parameters after 5 years' of treatment (significant at the 1% significance level) was found in the absolute and relative CD69 count, absolute cytotoxic/suppressor T lymphocyte count, absolute total leukocyte count, absolute natural killer cells count. A significant decrease of the observed parameters after 5 years' of treatment (significant at the 5% significance level) was found in the absolute lymphocyte count, relative cytotoxic/suppressor T lymphocyte count, relative CD3+CD69+ count and absolute CD8+CD38+ count. CONCLUSION The treatment with interferon beta reduces clinical exacerbations in multiple sclerosis (MS) through several known immunomodulatory mechanisms. However, the exact mechanism of effect of this medication is not known. This study presents some parameters that were affected by the long-term INFβ treatment.
Collapse
Affiliation(s)
- Zbyšek Pavelek
- Department of Neurology, Faculty of Medicine and University Hospital Hradec Králové, Charles University in Prague, Hradec Králové, Sokolská 581, 500 05, Czech Republic.
| | - Oldřich Vyšata
- Department of Neurology, Faculty of Medicine and University Hospital Hradec Králové, Charles University in Prague, Hradec Králové, Sokolská 581, 500 05, Czech Republic
| | - Blanka Klímová
- Department of Neurology, Faculty of Medicine and University Hospital Hradec Králové, Charles University in Prague, Hradec Králové, Sokolská 581, 500 05, Czech Republic
| | - Ctirad Andrýs
- Department of Clinical Immunology and Allergology, University Hospital Hradec Králové, Hradec Králové, Sokolská 581, 500 05, Czech Republic
| | - Doris Vokurková
- Department of Clinical Immunology and Allergology, University Hospital Hradec Králové, Hradec Králové, Sokolská 581, 500 05, Czech Republic
| | - Martin Vališ
- Department of Neurology, Faculty of Medicine and University Hospital Hradec Králové, Charles University in Prague, Hradec Králové, Sokolská 581, 500 05, Czech Republic
| |
Collapse
|
15
|
Pedraza-Alva G, Pérez-Martínez L, Valdez-Hernández L, Meza-Sosa KF, Ando-Kuri M. Negative regulation of the inflammasome: keeping inflammation under control. Immunol Rev 2016; 265:231-57. [PMID: 25879297 DOI: 10.1111/imr.12294] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
In addition to its roles in controlling infection and tissue repair, inflammation plays a critical role in diverse and distinct chronic diseases, such as cancer, metabolic syndrome, and neurodegenerative disorders, underscoring the harmful effect of an uncontrolled inflammatory response. Regardless of the nature of the stimulus, initiation of the inflammatory response is mediated by assembly of a multimolecular protein complex called the inflammasome, which is responsible for the production of inflammatory cytokines, such as interleukin-1β (IL-1β) and IL-18. The different stimuli and mechanisms that control inflammasome activation are fairly well understood, but the mechanisms underlying the control of undesired inflammasome activation and its inactivation remain largely unknown. Here, we review recent advances in our understanding of the molecular mechanisms that negatively regulate inflammasome activation to prevent unwanted activation in the resting state, as well as those involved in terminating the inflammatory response after a specific insult to maintain homeostasis.
Collapse
Affiliation(s)
- Gustavo Pedraza-Alva
- Laboratorio de Neuroinmunobiología, Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, México
| | | | | | | | | |
Collapse
|
16
|
Chen J, Qian C, Duan H, Cao S, Yu X, Li J, Gu C, Yan F, Wang L, Chen G. Melatonin attenuates neurogenic pulmonary edema via the regulation of inflammation and apoptosis after subarachnoid hemorrhage in rats. J Pineal Res 2015; 59:469-77. [PMID: 26383078 DOI: 10.1111/jpi.12278] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 09/11/2015] [Indexed: 01/09/2023]
Abstract
Neurogenic pulmonary edema (NPE) is a serious non-neurological complication that can occur after a subarachnoid hemorrhage (SAH) and is associated with decreased survival and a poor neurological outcome. Melatonin is a strong antioxidant that has beneficial effects against SAH in rats, including reduced mortality and reduced neurological deficits. The molecular mechanisms underlying these clinical effects in the SAH model, however, have not been clearly identified. This study was undertaken to determine the influence of melatonin on SAH-induced NPE and the potential mechanism of these effects using the filament perforation model of SAH in male Sprague Dawley rats. Either melatonin (150 mg/kg) or a vehicle was given via an intraperitoneal injection 2 hr after an SAH induction. Lung samples were extracted 24 hr after SAH. The results show that the melatonin treatment attenuated SAH-induced NPE by preventing alveolar-capillary barrier dysfunctions via inhibiting the disruption of tight junction proteins (ZO-1 and occludin). Moreover, the treatment downregulated the levels of mature interleukin (IL) -1β, myeloperoxidase (MPO), and matrix metallopeptidase (MMP) 9 expression/activation, which were increased in the lung; also, melatonin treatment improved neurological deficits. Furthermore, the melatonin treatment markedly reduced caspase-3 activity and the number of TUNEL-positive cells in the lung. Taken together, these findings show that administration of melatonin attenuates NPE by preventing alveolar-capillary barrier dysfunctions via repressing the inflammatory response and by anti-apoptosis effects after SAH.
Collapse
Affiliation(s)
- Jingyin Chen
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Cong Qian
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Hongyu Duan
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- Department of Neurosurgery, The First People's Hospital of Wenling, Taizhou, China
| | - Shenglong Cao
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaobo Yu
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Jianru Li
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Chi Gu
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Feng Yan
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Lin Wang
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Gao Chen
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
17
|
Macchi B, Marino-Merlo F, Nocentini U, Pisani V, Cuzzocrea S, Grelli S, Mastino A. Role of inflammation and apoptosis in multiple sclerosis: Comparative analysis between the periphery and the central nervous system. J Neuroimmunol 2015; 287:80-7. [PMID: 26439966 DOI: 10.1016/j.jneuroim.2015.08.016] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Revised: 08/19/2015] [Accepted: 08/24/2015] [Indexed: 01/23/2023]
Abstract
Multiple sclerosis (MS) is a complex, multifactorial disease associated with damage to the axonal myelin sheaths and neuronal degeneration. The pathognomonic event in MS is oligodendrocyte loss accompanied by axonal damage, blood-brain barrier leakage, inflammation and infiltration of immune cells. The etiopathogenesis of MS is far from being elucidated. However, increasing evidence suggests that the inflammatory and apoptotic responses, occurring in patients either at the peripheral level or the central nervous system (CNS), can play a role. In this review, we give a comprehensive picture of general aspects of inflammation and apoptosis in MS, with special emphasis on the until now not well highlighted possible links between phenomena relevant to these aspects occurring in either the periphery or in the CNS during MS.
Collapse
Affiliation(s)
- Beatrice Macchi
- Department of Systems Medicine, University of Rome "Tor Vergata", Via Montpellier 1, 00133 Rome, Italy.
| | - Francesca Marino-Merlo
- Department of Biological and Environmental Sciences, University of Messina, Via F. Stagno d'Alcontres 31, 98166 Messina, Italy.
| | - Ugo Nocentini
- Department of Systems Medicine, University of Rome "Tor Vergata", Via Montpellier 1, 00133 Rome, Italy; I.R.C.C.S. "Santa Lucia" Foundation, Via Ardeatina 306, 00179 Rome, Italy.
| | - Valerio Pisani
- I.R.C.C.S. "Santa Lucia" Foundation, Via Ardeatina 306, 00179 Rome, Italy.
| | - Salvatore Cuzzocrea
- Department of Biological and Environmental Sciences, University of Messina, Via F. Stagno d'Alcontres 31, 98166 Messina, Italy.
| | - Sandro Grelli
- Department of Experimental Medicine and Surgery, University of Rome "Tor Vergata", Via Montpellier 1, 00133 Rome, Italy.
| | - Antonio Mastino
- Department of Biological and Environmental Sciences, University of Messina, Via F. Stagno d'Alcontres 31, 98166 Messina, Italy; The Institute of Translational Pharmacology, CNR, Via Fosso del Cavaliere 100, 00133 Rome, Italy.
| |
Collapse
|
18
|
Zhang CX, Wen PH, Sun YL. Withdrawal of immunosuppression in liver transplantation and the mechanism of tolerance. Hepatobiliary Pancreat Dis Int 2015; 14:470-6. [PMID: 26459722 DOI: 10.1016/s1499-3872(15)60411-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND Immunosuppression reagents have side effects and cause considerable long-term morbidity and mortality in patients after liver transplantation. Sufficient evidences showed that minimization or withdrawal of immunosuppression reagents does not deteriorate the recipient's immune response and physiological function and therefore, is feasible in some recipients of liver transplantation. However, the mechanisms are not clear. The present review was to update the current status of immunosuppression in liver transplantation and the mechanism of minimization or withdrawal of immunosuppression in liver recipients. DATA SOURCES We searched articles in English on minimization or withdrawal of immunosuppression in liver transplantation in PubMed. We focused on the basic mechanisms of immune tolerance in liver transplantation. Studies on immunosuppression minimization or withdrawal protocols and biomarker in tolerant recipients were also analyzed. RESULTS Minimization or withdrawal of immunosuppression can be achieved by the induction of immune tolerance, which may not be permanent and can be affected by various factors. However, accurately evaluating immune status post-transplant is a prerequisite to achieve individualized immunosuppression. Numerous mechanisms for immune tolerance have been found, including immunophenotypic shift of memory CD8+ T cells and CD4+ T cell subsets. Activation of the inflammasome through apoptosis-associated speck-like protein containing a C-terminal caspase recruitment domain (ASC) in dendritic cells is associated with rejection after liver transplantation. CONCLUSIONS Minimization or withdrawal of immunosuppression can be achieved by the induction of immune tolerance via different mechanisms. This process could be affected by immunophenotypic shift of memory CD8+ T cells and CD4+ T cell subsets, which may be correlated with activation of the inflammasome through ASC in dendritic cells.
Collapse
Affiliation(s)
- Chi-Xian Zhang
- Institute of Hepatobiliary and Pancreatic Diseases, Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, Zhengzhou University, School of Medicine, Zhengzhou 450052, China.
| | | | | |
Collapse
|
19
|
Inflammasomes and human autoimmunity: A comprehensive review. J Autoimmun 2015; 61:1-8. [PMID: 26005048 DOI: 10.1016/j.jaut.2015.05.001] [Citation(s) in RCA: 119] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2015] [Accepted: 05/03/2015] [Indexed: 12/20/2022]
Abstract
Inflammasomes are multi-protein complexes composed of a NOD-like receptor (NLR)/an AIM-like receptor (ALR), the adapter molecule apoptosis-associated speck-like protein that contains a CARD (ASC), and caspase-1. Active caspase-1 cleaves pro-IL-1β and pro-IL-18 to IL-1β and IL-18, resulting in inflammation. Genetic mutations in inflammasomes were first recognized to result in autoinflammatory diseases, which are characterized by the absence of both autoantibodies and autoreactive-T/B cells. However, there is increasing attention being placed on genetic polymorphisms that are involved in the components of inflammasomes, and these have implications for innate immunity and the natural history of autoimmune diseases. For example, while the NOD-like receptor family, pyrin domain containing 1 (NLRP1) haplotypes contributes to susceptibility to developing vitiligo; there are other single nucleotide polymorphisms (SNPs) that alters the susceptibility and severity of rheumatoid arthritis (RA) and juvenile idiopathic arthritis. Indeed, there are multiple factors that contribute to lowering the threshold of immunity and inflammasomes play a key role in this threshold. For example, IL-1β and IL-18 further perpetuate Th17 responses and endothelial cell damage, which potentiate a number of autoimmune diseases, including synovitis in RA, cardiovascular disease, and systemic lupus erythematosus (SLE). There is also increasing data on the role of innate immunity in experimental autoimmune encephalomyelitis (EAE), in lupus nephritis, and in a variety of autoimmune pathologies in which activation of the innate immune system is the driver for the adaptive system. Indeed, it is likely that the chronic pathology of autoimmunity is mediated in part by otherwise innocent bystander cells, augmented by inflammasomes.
Collapse
|
20
|
Zheng M, Liu C, Fu R, Wang H, Wu Y, Li L, Liu H, Ding S, Shao Z. Abnormal immunomodulatory ability on memory T cells in humans with severe aplastic anemia. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2015; 8:3659-3669. [PMID: 26097547 PMCID: PMC4466934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Accepted: 03/30/2015] [Indexed: 06/04/2023]
Abstract
Severe aplastic anemia (SAA) is a bone marrow failure disease induced by hyperfunctional autoimmunic Th1 lymphocytes. Memory T cells (TM) are a component of the adaptive immune system. They ensure the host of more aggressive and faster immune response to efficiently eliminate the specific antigens after re-exposure and thus play a key role in T-cell functions. In this study we investigate the quantities and functions of memory T cells in SAA patients before and after immunosuppressive therapy (IST) to further clarify the mechanism of SAA apoptosis of bone marrow hematopoietic cells. Results showed that the percentage of CD4+ effector T cells in peripheral blood and bone marrow lymphocytes was decreased in SAA patients. The ratio of CD4+ memory T lymphocytes to CD8+ memory T subsets (CD4+/CD8+TM) in SAA patients was also lower. The percentage of CD8+ effector T cells in peripheral blood and CD8+ central memory T cells in the bone marrow lymphocytes was significantly higher in newly diagnosed patients. Furthermore, the median expressions of perforin and granzyme B on memory T cells were higher in SAA patients compared to those in normal controls. After IST, the quantities and functions of memory T cells return to normal level. Therefore, we concluded that the abnormal immunomodulatory ability on memory T cells may contribute to the imbalance of Th1/Th2 subsets and thus lead to over-function of T lymphocytes and hematopoiesis failure in SAA.
Collapse
Affiliation(s)
- Mengying Zheng
- Department of Hematology, The General Hospital of Tianjin Medical University Tianjin, China
| | - Chunyan Liu
- Department of Hematology, The General Hospital of Tianjin Medical University Tianjin, China
| | - Rong Fu
- Department of Hematology, The General Hospital of Tianjin Medical University Tianjin, China
| | - Huaquan Wang
- Department of Hematology, The General Hospital of Tianjin Medical University Tianjin, China
| | - Yuhong Wu
- Department of Hematology, The General Hospital of Tianjin Medical University Tianjin, China
| | - Lijuan Li
- Department of Hematology, The General Hospital of Tianjin Medical University Tianjin, China
| | - Hui Liu
- Department of Hematology, The General Hospital of Tianjin Medical University Tianjin, China
| | - Shaoxue Ding
- Department of Hematology, The General Hospital of Tianjin Medical University Tianjin, China
| | - Zonghong Shao
- Department of Hematology, The General Hospital of Tianjin Medical University Tianjin, China
| |
Collapse
|
21
|
Malhotra S, Río J, Urcelay E, Nurtdinov R, Bustamante MF, Fernández O, Oliver B, Zettl U, Brassat D, Killestein J, Lechner-Scott J, Drulovic J, Chan A, Martinelli-Boneschi F, García-Merino A, Montalban X, Comabella M. NLRP3 inflammasome is associated with the response to IFN-β in patients with multiple sclerosis. ACTA ACUST UNITED AC 2015; 138:644-52. [PMID: 25586466 DOI: 10.1093/brain/awu388] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Evidence exists for a potential modulation of inflammasome activity by interferon beta. Here, we investigated the roles of inflammasomes [absent in melanoma 2 (AIM2); NLR family, CARD domain containing 4 (NLRC4); NLR family, pyrin domain containing 1 and 3 (NLRP1 and NLRP3)] and related cytokines (IL1B, IL10, IL18) in the response to interferon beta in patients with relapsing-remitting multiple sclerosis. Ninety-seven patients treated with interferon beta were classified into responders and non-responders according to clinical criteria after 24 months and clinical-radiological criteria after 12 months of treatment. Messenger RNA expression levels of inflammasomes and cytokines were determined by real-time polymerase chain reaction in peripheral blood mononuclear cells collected before treatment with interferon beta. In a subgroup of patients, NLRP3 and IL1B expression was also determined after 3 months (n = 32) and 12 months (n = 20) of interferon beta treatment. A polymorphism located in the NLRP3 gene, rs35829419, was genotyped in 789 multiple sclerosis patients treated with interferon beta. Baseline mRNA expression levels for NLRP3 and IL1B were increased in peripheral blood mononuclear cells from non-responders compared to responders classified according to clinical criteria after 24 months (P = 0.02 and P = 0.001, respectively). No significant differences were observed for other inflammasomes and related cytokines. Differences in NLRP3 and IL1B expression remained significant following a clinical-radiological classification after 12 months (P = 0.007 and P = 0.02, respectively). After treatment with interferon beta, NLRP3 and IL1B expression was increased in responders but unchanged in non-responders. A trend for association was observed between rs35829419 and interferon beta response (pM-H = 0.08). These results point to a role of the NLRP3 inflammasome and its related cytokine IL1B in the response to interferon beta in patients with relapsing-remitting multiple sclerosis.
Collapse
Affiliation(s)
- Sunny Malhotra
- 1 Servei de Neurologia-Neuroimmunologia, Centre d'Esclerosi Múltiple de Catalunya (Cemcat), Institut de Recerca Vall d'Hebron (VHIR), Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Jordi Río
- 1 Servei de Neurologia-Neuroimmunologia, Centre d'Esclerosi Múltiple de Catalunya (Cemcat), Institut de Recerca Vall d'Hebron (VHIR), Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Elena Urcelay
- 2 Servicio de Neurología, Hospital Clínico San Carlos, Madrid, Spain
| | - Ramil Nurtdinov
- 1 Servei de Neurologia-Neuroimmunologia, Centre d'Esclerosi Múltiple de Catalunya (Cemcat), Institut de Recerca Vall d'Hebron (VHIR), Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Marta F Bustamante
- 1 Servei de Neurologia-Neuroimmunologia, Centre d'Esclerosi Múltiple de Catalunya (Cemcat), Institut de Recerca Vall d'Hebron (VHIR), Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Oscar Fernández
- 3 Unidad de Gestión Clínica de Neurociencias. Instituto de Biomedicina de Málaga (IBIMA), Hospital Universitario Regional de Málaga, Universidad de Málaga, Spain
| | - Begoña Oliver
- 3 Unidad de Gestión Clínica de Neurociencias. Instituto de Biomedicina de Málaga (IBIMA), Hospital Universitario Regional de Málaga, Universidad de Málaga, Spain
| | - Uwe Zettl
- 4 University of Rostock, Department of Neurology, Rostock, Germany
| | - David Brassat
- 5 Pole des Neurosciences et INSERM U1043, Université de Toulouse III, Hopital Purpan, Toulouse, France
| | | | - Jeannette Lechner-Scott
- 7 Department of Neurology, John Hunter Hospital, Newcastle, Australia, Hunter Medical Research Institute, University of Newcastle, Australia
| | - Jelena Drulovic
- 8 Clinic of Neurology, Clinical Centre of Serbia (CCS), Faculty of Medicine, University of Belgrade, Serbia
| | - Andrew Chan
- 9 Department of Neurology, St. Josef-Hospital, Ruhr-University Bochum, Germany
| | - Filippo Martinelli-Boneschi
- 10 Laboratory of Genetics of Neurological Complex Disorders and Department of Neurology, Institute of Experimental Neurology (INSPE), Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | | | - Xavier Montalban
- 1 Servei de Neurologia-Neuroimmunologia, Centre d'Esclerosi Múltiple de Catalunya (Cemcat), Institut de Recerca Vall d'Hebron (VHIR), Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Manuel Comabella
- 1 Servei de Neurologia-Neuroimmunologia, Centre d'Esclerosi Múltiple de Catalunya (Cemcat), Institut de Recerca Vall d'Hebron (VHIR), Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| |
Collapse
|
22
|
Gasparoto TH, de Oliveira CE, de Freitas LT, Pinheiro CR, Hori JI, Garlet GP, Cavassani KA, Schillaci R, da Silva JS, Zamboni DS, Campanelli AP. Inflammasome activation is critical to the protective immune response during chemically induced squamous cell carcinoma. PLoS One 2014; 9:e107170. [PMID: 25268644 PMCID: PMC4182037 DOI: 10.1371/journal.pone.0107170] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Accepted: 08/07/2014] [Indexed: 12/11/2022] Open
Abstract
Chronic inflammation affects most stages of tumorigenesis, including initiation, promotion, malignant differentiation, invasion and metastasis. Inflammasomes have been described as involved with persistent inflammation and are known to exert both pro and antitumour effects. We evaluated the influence of apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC) and caspase (CASP)-1 in the antitumor immune response using a multistage model of squamous cell carcinoma (SCC) development. Absence of ASC and CASP-1 resulted in an earlier incidence and increased number of papilloma. Loss of inflammassome function in mice resulted in decreased presence of natural killer (NK), dendritic (DC), CD4+, CD8+ and CD45RB+ T cells in the tumor lesions as well as in lymph nodes (LN) compared with WT mice. Increased percentage of CD4+CD25+Foxp3+ T cells was associated with association with inflammasome loss of function. Moreover, significant differences were also found with neutrophils and macrophage infiltrating the lesions. Myeloperoxidase (MPO), but not elastase (ELA), activity oscillated among the groups during the SCC development. Levels of proinflammatory cytokines IL-1β, IL-18, Tumor Necrosis Factor (TNF)-α and Interferon (IFN)-γ were decreased in the tumor microenvironment in the absence of inflammasome proteins. These observations suggest a link between inflammasome function and SCC tumorigenesis, indicating an important role for inflammasome activation in the control of SCC development.
Collapse
Affiliation(s)
- Thais Helena Gasparoto
- Department of Biological Sciences - Microbiology and Immunology, Bauru School of Dentistry, University of São Paulo, Bauru, SP, Brazil
| | - Carine Ervolino de Oliveira
- Department of Biological Sciences - Microbiology and Immunology, Bauru School of Dentistry, University of São Paulo, Bauru, SP, Brazil
- Department of Stomatology - Oral Pathology, Bauru School of Dentistry, University of São Paulo, Bauru, SP, Brazil
| | - Luisa Thomazini de Freitas
- Department of Biological Sciences - Microbiology and Immunology, Bauru School of Dentistry, University of São Paulo, Bauru, SP, Brazil
| | - Claudia Ramos Pinheiro
- Department of Biological Sciences - Microbiology and Immunology, Bauru School of Dentistry, University of São Paulo, Bauru, SP, Brazil
| | - Juliana Issa Hori
- Department of Cell Biology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Gustavo Pompermaier Garlet
- Department of Biological Sciences - Microbiology and Immunology, Bauru School of Dentistry, University of São Paulo, Bauru, SP, Brazil
| | - Karen Angélica Cavassani
- Departament of Pathology, Medical School, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Roxana Schillaci
- Laboratorio de Mecanismos Moleculares de Carcinogénesis, Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas de Argentina, Buenos Aires, Argentina
| | - João Santana da Silva
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Dario Simões Zamboni
- Department of Cell Biology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Ana Paula Campanelli
- Department of Biological Sciences - Microbiology and Immunology, Bauru School of Dentistry, University of São Paulo, Bauru, SP, Brazil
- * E-mail:
| |
Collapse
|
23
|
Kwon JH, Nam ES, Shin HS, Cho SJ, Park HR, Kwon MJ. P2X7 Receptor Expression in Coexistence of Papillary Thyroid Carcinoma with Hashimoto's Thyroiditis. KOREAN JOURNAL OF PATHOLOGY 2014; 48:30-5. [PMID: 24627692 PMCID: PMC3950232 DOI: 10.4132/koreanjpathol.2014.48.1.30] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Revised: 01/25/2014] [Accepted: 01/27/2014] [Indexed: 11/17/2022]
Abstract
Background This study was aimed at investigating the relation of P2X7 receptor (P2X7R) expression with the clinicopathological features of papillary thyroid carcinoma (PTC) coexisting with Hashimoto's thyroiditis (HT). Methods We examined 170 patients (84, PTC with HT; 86, PTC without HT). P2X7R expression was examined by immunohistochemical methods. The staining intensity and patterns were evaluated and scored using a semi-quantitative method. Results The PTC with HT group was more likely to contain women and had less extrathyroid extension, lymph node (LN) metastasis, lymphovascular invasion, and recurrence than the PTC without HT group. Patients positive for P2X7R had significantly higher frequencies of lymphovascular invasion, extrathyroid extension, LN metastasis, and absence of HT. As shown by multivariate analysis, the expression of P2X7R was significantly higher if HT was absent and extrathyroid extension was present. In the PTC with HT group, the expression of P2X7R was significantly higher in patients with tumor multifocality, lymphovascular invasion, and extrathyroid extension. In the PTC without HT group, the expression of P2X7R was significantly higher in women and those having tumor multifocality. Conclusions Coexistence of PTC with HT is associated with good prognostic factors, and P2X7R expression in PTC was correlated with poor prognostic factors and the absence of HT.
Collapse
Affiliation(s)
- Ji Hyun Kwon
- Department of Pathology, Kangdong Sacred Heart Hospital, Hallym University College of Medicine, Seoul, Korea
| | - Eun Sook Nam
- Department of Pathology, Kangdong Sacred Heart Hospital, Hallym University College of Medicine, Seoul, Korea
| | - Hyung Sik Shin
- Department of Pathology, Kangdong Sacred Heart Hospital, Hallym University College of Medicine, Seoul, Korea
| | - Seong Jin Cho
- Department of Pathology, Kangdong Sacred Heart Hospital, Hallym University College of Medicine, Seoul, Korea
| | - Hye Rim Park
- Department of Pathology, Hallym University Sacred Heart Hospital, Hallym University College of Medicine, Anyang, Korea
| | - Mi Jung Kwon
- Department of Pathology, Hallym University Sacred Heart Hospital, Hallym University College of Medicine, Anyang, Korea
| |
Collapse
|
24
|
Chen S, Zhu Z, Klebe D, Bian H, Krafft PR, Tang J, Zhang J, Zhang JH. Role of P2X purinoceptor 7 in neurogenic pulmonary edema after subarachnoid hemorrhage in rats. PLoS One 2014; 9:e89042. [PMID: 24533168 PMCID: PMC3923073 DOI: 10.1371/journal.pone.0089042] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2013] [Accepted: 01/13/2014] [Indexed: 12/20/2022] Open
Abstract
Introduction Neurogenic pulmonary edema (NPE) is an acute and serious complication after subarachnoid hemorrhage (SAH) with high mortality. The present study aimed to test the therapeutic potential of brilliant blue G (BBG), a selective P2X purinoceptor 7 (P2X7R) antagonist, on NPE in a rat SAH model. Methods SAH was induced by endovascular perforation. 86 Sprague-Dawley rats were randomly divided into sham, vehicle-, or BBG-treatment groups. Mortality, body weight, SAH grading, neurological deficits, NPE clinical symptoms, and pulmonary index were measured at 24 hours following SAH. Western blot, gelatin zymography, lung histopathology, and immunofluorescence staining were performed in the left lung lobe to explore the underlying mechanisms at 24 hours post-surgery. Results The incidence of clinical symptoms was correlated with pulmonary index. P2X7R and the marker of alveolar type I epithelial cells (the mucin-type glycoprotein T1-α) immunoreactivities were generally co-localized. BBG administration decreased mature interleukin-1β, myeloperoxidase, and matrix metallopeptidase-9 activation, but increased tight junction proteins, such as ZO-1 and occludin, which ameliorated pulmonary edema via anti-inflammation and improved neurological deficits. Conclusion P2X7R inhibition prevented NPE after SAH by attenuating inflammation. Thus, BBG is a potential therapeutic application for NPE after SAH and warrants further research.
Collapse
Affiliation(s)
- Sheng Chen
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang, China
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, California, United States of America
| | - Zhigang Zhu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang, China
| | - Damon Klebe
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, California, United States of America
| | - Hetao Bian
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, California, United States of America
| | - Paul R. Krafft
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, California, United States of America
| | - Jiping Tang
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, California, United States of America
| | - Jianmin Zhang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang, China
- * E-mail: (Jianmin Zhang); (John Zhang)
| | - John H. Zhang
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, California, United States of America
- * E-mail: (Jianmin Zhang); (John Zhang)
| |
Collapse
|
25
|
Mahla RS, Reddy MC, Prasad DVR, Kumar H. Sweeten PAMPs: Role of Sugar Complexed PAMPs in Innate Immunity and Vaccine Biology. Front Immunol 2013; 4:248. [PMID: 24032031 PMCID: PMC3759294 DOI: 10.3389/fimmu.2013.00248] [Citation(s) in RCA: 156] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2013] [Accepted: 08/09/2013] [Indexed: 12/12/2022] Open
Abstract
Innate sensors play a critical role in the early innate immune responses to invading pathogens through sensing of diverse biochemical signatures also known as pathogen associated molecular patterns (PAMPs). These biochemical signatures primarily consist of a major family of biomolecules such as proteins, lipids, nitrogen bases, and sugar and its complexes, which are distinct from host molecules and exclusively expressed in pathogens and essential to their survival. The family of sensors known as pattern recognition receptors (PRRs) are germ-line encoded, evolutionarily conserved molecules, and consist of Toll-like receptors (TLRs), RIG-I-like receptors (RLRs), NOD-like receptors (NLRs), C-type lectin-like receptors (CLRs), and DNA sensors. Sensing of PAMP by PRR initiates the cascade of signaling leading to the activation of transcription factors, such as NF-κB and interferon regulatory factors (IRFs), resulting in a variety of cellular responses, including the production of interferons (IFNs) and pro-inflammatory cytokines. In this review, we discuss sensing of different types of glycosylated PAMPs such as β-glucan (a polymeric sugar) or lipopolysaccharides, nucleic acid, and so on (sugar complex PAMPs) by different families of sensors, its role in pathogenesis, and its application in development of potential vaccine and vaccine adjuvants.
Collapse
Affiliation(s)
- Ranjeet Singh Mahla
- Laboratory of Immunology, Department of Biological Sciences, Indian Institute of Science Education and Research (IISER) , Bhopal , India
| | | | | | | |
Collapse
|
26
|
Chen S, Ma Q, Krafft PR, Hu Q, Rolland W, Sherchan P, Zhang J, Tang J, Zhang JH. P2X7R/cryopyrin inflammasome axis inhibition reduces neuroinflammation after SAH. Neurobiol Dis 2013; 58:296-307. [PMID: 23816751 DOI: 10.1016/j.nbd.2013.06.011] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2013] [Revised: 06/06/2013] [Accepted: 06/15/2013] [Indexed: 12/21/2022] Open
Abstract
Neuroinflammation contributes to the pathogenesis of early brain injury (EBI) after subarachnoid hemorrhage (SAH). Cytotoxic events following SAH, such as extracellular accumulation of adenosine triphosphate (ATP), may activate the P2X purinoceptor 7 (P2X7R)/cryopyrin inflammasome axis, thus inducing the proinflammatory cytokine IL-1β/IL-18 secretion. We therefore hypothesized that inhibition of P2X7R/cryopyrin inflammasome axis would ameliorate neuroinflammation after SAH. In the present study, SAH was induced by the endovascular perforation in rats. Small interfering RNAs (siRNAs) of P2X7R or cryopyrin were administered intracerebroventricularly 24h before SAH. Brilliant blue G (BBG), a non-competitive antagonist of P2X7R, was administered intraperitoneally 30min following SAH. Post-assessments including SAH severity score, neurobehavioral test, brain water content, Western blot and immunofluorescence, were performed. Administration of P2X7R and cryopyrin siRNA as well as pharmacologic blockade of P2X7R by BBG ameliorated neurological deficits and brain edema at 24h following SAH. Inhibition of P2X7R/cryopyrin inflammasome axis suppressed caspase-1 activation, which subsequently decreased maturation of IL-1β/IL-18. To investigate the link between P2X7R and cryopyrin inflammasome in vivo, Benzoylbenzoyl-ATP (BzATP), a P2X7R agonist, was given to lipopolysaccharide (LPS) primed naive rats with scramble or cryopyrin siRNAs. In LPS-primed naive rats, BzATP induced caspase-1 activation and mature IL-1β release were neutralized by cryopyrin siRNA. Thus, the P2X7R/cryopyrin inflammasome axis may contribute to neuroinflammation via activation of caspase-1 and thereafter mature IL-1β/IL-18 production following SAH. Therapeutic interventions targeting P2X7R/cryopyrin pathway may be a novel approach to ameliorate EBI following SAH.
Collapse
Affiliation(s)
- Sheng Chen
- Department of Neurosurgery, Zhejiang University, Hangzhou, Zhejiang, China
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Hogan EL, Podbielska M, O'Keeffe J. Implications of Lymphocyte Anergy to Glycolipids in Multiple Sclerosis (MS): iNKT Cells May Mediate the MS Infectious Trigger. ACTA ACUST UNITED AC 2013; 4. [PMID: 26347308 PMCID: PMC4557814 DOI: 10.4172/2155-9899.1000144] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Immunogenic lipids may play key roles in host defenses against infection and in generating autoimmune inflammation and organ-specific damage. In multiple sclerosis (MS) there are unequivocal autoimmune features and vulnerability to aggravation or induction by microbial or viral infection. We have found glycolipid-driven anergy of circulating lymphocytes in MS indicating that this immune response is affected in MS and the robust effects of iNKT activation with potent cellular and cytokine activities emphasizes its potential importance. Diverse glycolipids including the endogenous myelin acetylated-galactosylceramides (AcGalCer) can drive activation that could be critical to the inflammatory demyelination in the central nervous system and clinical consequences. The iNKT cells and their invariant or iTCR (Vα24Jα18Vβ11) receptor an innate defense–a discrete immune arm that is separate from peptide-driven acquired immune responses. This offers new possibilities for insight including a likelihood that the pattern recognition of exogenous microbial and myelin immunogens can overlap and cross-react especially in an inflammatory milieu.
Collapse
Affiliation(s)
- Edward L Hogan
- Georgia Regents University, Institute of Molecular Medicine and Genetics, Department of Neurology, 1120 15 Street, Augusta, 30912-2620 GA, USA ; National University of Ireland Galway, Department of Microbiology, University Road, Galway, Ireland ; Medical University of South Carolina, Department of Neurosciences, 173 Ashley Avenue, Charleston, SC 29401, USA
| | - Maria Podbielska
- Georgia Regents University, Institute of Molecular Medicine and Genetics, Department of Neurology, 1120 15 Street, Augusta, 30912-2620 GA, USA ; Ludwik Hirszfeld Institute of Immunology & Experimental Therapy, Polish Academy of Sciences, Laboratory of Signaling Proteins, R. Weigla Street 12, 53-114 Wrocław, Poland
| | - Joan O'Keeffe
- Department of Life and Physical Sciences, School of Science, Galway-Mayo Institute of Technology, Galway, Ireland
| |
Collapse
|