1
|
Tsang RSM, Timpson NJ, Khandaker GM. Inflammation proteomic profiling of psychosis in young adults: Findings from the ALSPAC birth cohort. Psychoneuroendocrinology 2025; 171:107188. [PMID: 39442229 DOI: 10.1016/j.psyneuen.2024.107188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 08/12/2024] [Accepted: 09/16/2024] [Indexed: 10/25/2024]
Abstract
Psychotic disorder is associated with altered levels of various inflammatory markers in blood, but existing studies have typically focused on a few selected biomarkers, have not examined specific symptom domains notably negative symptoms, and are based on individuals with established/chronic illness. Based on data from young people aged 24 years from the Avon Longitudinal Study of Parents and Children (ALSPAC), a UK birth cohort, we have examined the associations of 67 plasma immune/inflammatory proteins assayed using the Olink Target 96 Inflammation panel with psychotic disorder, positive (any psychotic experiences and definite psychotic experiences) and negative symptoms, using linear models with empirical Bayes estimation. The analyses included between 2317 and 2854 individuals. After adjustment for age, sex, body mass index and smoking and correction for multiple testing, positive symptoms and psychotic disorder were consistently associated with upregulation of CDCP1 and IL-6, and psychotic disorder was additionally associated with upregulation of MMP-10. Negative symptoms were associated with upregulation of CDCP1 and TRAIL. CDCP1 and MMP-10 are novel markers of psychosis identified in this study, and are involved in immune regulation, immune cell activation/migration, blood-brain barrier disruption, and extracellular matrix abnormalities. Our findings highlight psychosis symptom domains have overlapping and distinct immune associations, and support a role of inflammation and immune dysfunction in the pathogenesis of psychosis.
Collapse
Affiliation(s)
- Ruby S M Tsang
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK; Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK; Centre for Academic Mental Health, Population Health Sciences, University of Bristol, Bristol, UK.
| | - Nicholas J Timpson
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK; Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Golam M Khandaker
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK; Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK; Centre for Academic Mental Health, Population Health Sciences, University of Bristol, Bristol, UK; NIHR Bristol Biomedical Research Centre, University Hospitals Bristol and Weston NHS Foundation Trust, Bristol, UK
| |
Collapse
|
2
|
Vasco C, Rizzo A, Cordiglieri C, Corsini E, Maderna E, Ciusani E, Salmaggi A. The Role of Adhesion Molecules and Extracellular Vesicles in an In Vitro Model of the Blood-Brain Barrier for Metastatic Disease. Cancers (Basel) 2023; 15:cancers15113045. [PMID: 37297006 DOI: 10.3390/cancers15113045] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 05/28/2023] [Accepted: 05/31/2023] [Indexed: 06/12/2023] Open
Abstract
Metastatic brain disease (MBD) has seen major advances in clinical management, focal radiation therapy approaches and knowledge of biological factors leading to improved prognosis. Extracellular vesicles (EVs) have been found to play a role in tumor cross-talk with the target organ, contributing to the formation of a premetastatic niche. Human lung and breast cancer cell lines were characterized for adhesion molecule expression and used to evaluate their migration ability in an in vitro model. Conditioned culture media and isolated EVs, characterized by super resolution and electron microscopy, were tested to evaluate their pro-apoptotic properties on human umbilical vein endothelial cells (HUVECs) and human cerebral microvascular endothelial cells (HCMEC/D3) by annexin V binding assay. Our data showed a direct correlation between expression of ICAM1, ICAM2, β3-integrin and α2-integrin and the ability to firmly adhere to the blood-brain barrier (BBB) model, whereas the same molecules were down-regulated at a later step. Extracellular vesicles released by tumor cell lines were shown to be able to induce apoptosis in HUVEC while brain endothelial cells showed to be more resistant.
Collapse
Affiliation(s)
- Chiara Vasco
- Laboratory of Clinical Chemistry SMeL122, Fondazione IRCCS Istituto Neurologico "Carlo Besta", 20133 Milan, Italy
| | - Ambra Rizzo
- Laboratory of Clinical Chemistry SMeL122, Fondazione IRCCS Istituto Neurologico "Carlo Besta", 20133 Milan, Italy
| | - Chiara Cordiglieri
- Preclinical Neuroimmunology Lab, Neurology IV Fondazione IRCCS Istituto Neurologico "Carlo Besta", 20133 Milan, Italy
- Imaging Facility, National Institute of Molucular Genetics (INGM) "Romeo ed Enrica Invernizzi", c/o Policlinico di Milano Hospital, Padiglione Invernizzi, Via Francesco Sforza 35, 20122 Milan, Italy
| | - Elena Corsini
- Laboratory of Clinical Chemistry SMeL122, Fondazione IRCCS Istituto Neurologico "Carlo Besta", 20133 Milan, Italy
| | - Emanuela Maderna
- Neurology 5/Neuropathology Unit, Fondazione IRCCS Istituto Neurologico C. Besta, 20133 Milan, Italy
| | - Emilio Ciusani
- Laboratory of Clinical Chemistry SMeL122, Fondazione IRCCS Istituto Neurologico "Carlo Besta", 20133 Milan, Italy
| | - Andrea Salmaggi
- Neuroscience Department-Neurology/Stroke Unit, Ospedale A. Manzoni, ASST Lecco, 23900 Lecco, Italy
| |
Collapse
|
3
|
Low-Level Endothelial TRAIL-Receptor Expression Obstructs the CNS-Delivery of Angiopep-2 Functionalised TRAIL-Receptor Agonists for the Treatment of Glioblastoma. Molecules 2021; 26:molecules26247582. [PMID: 34946664 PMCID: PMC8706683 DOI: 10.3390/molecules26247582] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 12/07/2021] [Accepted: 12/10/2021] [Indexed: 12/12/2022] Open
Abstract
Glioblastoma (GBM) is the most malignant and aggressive form of glioma and is associated with a poor survival rate. Latest generation Tumour Necrosis Factor Related Apoptosis-Inducing Ligand (TRAIL)-based therapeutics potently induce apoptosis in cancer cells, including GBM cells, by binding to death receptors. However, the blood-brain barrier (BBB) is a major obstacle for these biologics to enter the central nervous system (CNS). We therefore investigated if antibody-based fusion proteins that combine hexavalent TRAIL and angiopep-2 (ANG2) moieties can be developed, with ANG2 promoting receptor-mediated transcytosis (RMT) across the BBB. We demonstrate that these fusion proteins retain the potent apoptosis induction of hexavalent TRAIL-receptor agonists. Importantly, blood-brain barrier cells instead remained highly resistant to this fusion protein. Binding studies indicated that ANG2 is active in these constructs but that TRAIL-ANG2 fusion proteins bind preferentially to BBB endothelial cells via the TRAIL moiety. Consequently, transport studies indicated that TRAIL-ANG2 fusion proteins can, in principle, be shuttled across BBB endothelial cells, but that low TRAIL receptor expression on BBB endothelial cells interferes with efficient transport. Our work therefore demonstrates that TRAIL-ANG2 fusion proteins remain highly potent in inducing apoptosis, but that therapeutic avenues will require combinatorial strategies, such as TRAIL-R masking, to achieve effective CNS transport.
Collapse
|
4
|
Zeitelhofer M, Adzemovic MZ, Moessinger C, Stefanitsch C, Strell C, Muhl L, Brundin L, Fredriksson L, Olsson T, Eriksson U, Nilsson I. Blocking PDGF-CC signaling ameliorates multiple sclerosis-like neuroinflammation by inhibiting disruption of the blood-brain barrier. Sci Rep 2020; 10:22383. [PMID: 33361796 PMCID: PMC7759579 DOI: 10.1038/s41598-020-79598-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Accepted: 12/07/2020] [Indexed: 02/07/2023] Open
Abstract
Disruption of blood–brain barrier (BBB) integrity is a feature of various neurological disorders. Here we found that the BBB is differently affected during the preclinical, progression and remission phase of experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis (MS). We have identified an upregulation of pro-inflammatory and pro-angiogenic factors in the BBB transcriptome and down-regulation of endothelial tight junction members coinciding with elevated BBB leakage specifically during the progression phase. These changes were antagonized by blocking PDGFRα signaling with the small tyrosine kinase inhibitor imatinib. Moreover, targeting the PDGFRα ligand PDGF-CC using a neutralizing antibody, facilitated recovery of BBB integrity and improvement of EAE symptoms. Intracerebroventricular injection of PDGF-CC induced upregulation, whereas blocking PDGF-CC during EAE led to downregulation of Tnfa and Il1a at the BBB. Our findings suggest that blocking PDGF-CC counteracts fundamental aspects of endothelial cell activation and disruption of the BBB by decreasing Tnfa and Il1a expression. We also demonstrate that both PDGF-CC and its receptor PDGFRα were upregulated in MS lesions indicating that blocking PDGF-CC may be considered a novel treatment for MS.
Collapse
Affiliation(s)
- Manuel Zeitelhofer
- Division of Vascular Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 77, Stockholm, Sweden.
| | - Milena Z Adzemovic
- Division of Vascular Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 77, Stockholm, Sweden.,Neuroimmunology Unit, Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska University Hospital, 171 76, Stockholm, Sweden
| | - Christine Moessinger
- Division of Vascular Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Christina Stefanitsch
- Division of Vascular Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Carina Strell
- Department of Immunology, Genetics and Pathology, Uppsala University, 75185, Uppsala, Sweden
| | - Lars Muhl
- Division of Vascular Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Lou Brundin
- Neuroimmunology Unit, Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska University Hospital, 171 76, Stockholm, Sweden
| | - Linda Fredriksson
- Division of Vascular Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Tomas Olsson
- Neuroimmunology Unit, Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska University Hospital, 171 76, Stockholm, Sweden
| | - Ulf Eriksson
- Division of Vascular Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Ingrid Nilsson
- Division of Vascular Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 77, Stockholm, Sweden.
| |
Collapse
|
5
|
Iwata K, Wu Q, Ferdousi F, Sasaki K, Tominaga K, Uchida H, Arai Y, Szele FG, Isoda H. Sugarcane ( Saccharum officinarum L.) Top Extract Ameliorates Cognitive Decline in Senescence Model SAMP8 Mice: Modulation of Neural Development and Energy Metabolism. Front Cell Dev Biol 2020; 8:573487. [PMID: 33123536 PMCID: PMC7573230 DOI: 10.3389/fcell.2020.573487] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 09/03/2020] [Indexed: 12/13/2022] Open
Abstract
Age-related biological alterations in brain function increase the risk of mild cognitive impairment and dementia, a global problem exacerbated by aging populations in developed nations. Limited pharmacological therapies have resulted in attention turning to the promising role of medicinal plants and dietary supplements in the treatment and prevention of dementia. Sugarcane (Saccharum officinarum L.) top, largely considered as a by-product because of its low sugar content, in fact contains the most abundant amounts of antioxidant polyphenols relative to the rest of the plant. Given the numerous epidemiological studies on the effects of polyphenols on cognitive function, in this study, we analyzed polyphenolic constituents of sugarcane top and examined the effect of sugarcane top ethanolic extract (STEE) on a range of central nervous system functions in vitro and in vivo. Orally administrated STEE rescued spatial learning and memory deficit in the senescence-accelerated mouse prone 8 (SAMP8) mice, a non-transgenic strain that spontaneously develops a multisystemic aging phenotype including pathological features of Alzheimer's disease. This could be correlated with an increased number of hippocampal newborn neurons and restoration of cortical monoamine levels in STEE-fed SAMP8 mice. Global genomic analysis by microarray in cerebral cortices showed multiple potential mechanisms for the cognitive improvement. Gene set enrichment analysis (GSEA) revealed biological processes such as neurogenesis, neuron differentiation, and neuron development were significantly enriched in STEE-fed mice brain compared to non-treated SAMP8 mice. Furthermore, STEE treatment significantly regulated genes involved in neurotrophin signaling, glucose metabolism, and neural development in mice brain. Our in vitro results suggest that STEE treatment enhances the metabolic activity of neuronal cells promoting glucose metabolism with significant upregulation of genes, namely PGK1, PGAM1, PKM, and PC. STEE also stimulated proliferation of human neural stem cells (hNSCs), regulated bHLH factor expression and induced neuronal differentiation and astrocytic process lengthening. Altogether, our findings suggest the potential of STEE as a dietary intervention, with promising implications as a novel nutraceutical for cognitive health.
Collapse
Affiliation(s)
- Kengo Iwata
- School of Integrative and Global Majors, University of Tsukuba, Tsukuba, Japan.,Nippo Co., Ltd., Daito, Japan
| | - Qingqing Wu
- Alliance for Research on the Mediterranean and North Africa, University of Tsukuba, Tsukuba, Japan.,Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Farhana Ferdousi
- Alliance for Research on the Mediterranean and North Africa, University of Tsukuba, Tsukuba, Japan.,AIST-University of Tsukuba Open Innovation Laboratory for Food and Medicinal Resource Engineering (FoodMed-OIL), AIST, University of Tsukuba, Tsukuba, Japan
| | - Kazunori Sasaki
- Alliance for Research on the Mediterranean and North Africa, University of Tsukuba, Tsukuba, Japan.,AIST-University of Tsukuba Open Innovation Laboratory for Food and Medicinal Resource Engineering (FoodMed-OIL), AIST, University of Tsukuba, Tsukuba, Japan
| | - Kenichi Tominaga
- AIST-University of Tsukuba Open Innovation Laboratory for Food and Medicinal Resource Engineering (FoodMed-OIL), AIST, University of Tsukuba, Tsukuba, Japan
| | | | | | - Francis G Szele
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Hiroko Isoda
- School of Integrative and Global Majors, University of Tsukuba, Tsukuba, Japan.,Alliance for Research on the Mediterranean and North Africa, University of Tsukuba, Tsukuba, Japan.,AIST-University of Tsukuba Open Innovation Laboratory for Food and Medicinal Resource Engineering (FoodMed-OIL), AIST, University of Tsukuba, Tsukuba, Japan.,Faculty of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Japan
| |
Collapse
|
6
|
Taheri M, Nemati S, Movafagh A, Saberi M, Mirfakhraie R, Eftekharian MM, Arsang-Jang S, Rezagholizadeh A, Sayad A. TRAIL gene expression analysis in multiple sclerosis patients. Hum Antibodies 2016; 24:33-38. [PMID: 27472871 DOI: 10.3233/hab-160291] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
BACKGROUND Multiple sclerosis (MS) as an autoimmune disorder in which the insulating covers of neurons in the Central Nervous System are destructed. Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is an immunomodulatory molecule to protect against T cells hyper activation. METHODS In this Case-control study, we compare TRAIL gene expression in peripheral blood between 50 relapse remitting MS patients and 50 healthy controls by TaqMan Real time PCR. All the patients were negative for HLA-DRB1*15 susceptible allele, normal serum vitamin D, responder to Interferon beta. All the health individuals were matched to patients. Also, we tried to find correlation between TRAIL gene expression and clinical characteristics of patients. RESULTS No statistically significant difference was found in TRAIL mRNA expression between MS patients and controls (p> 0.05). There was no correlation in the TRAIL expression and age of onset, disease duration and Expanded Disability Status Scale of Kurtzke (EDSS). As IFN-b may have stimulatory effects on immunoregulatory function of TRAIL and all of our patients were treated with interferon beta and were responder, it lead to no significant change in TRAIL expression. We suggest comparing between responders and non-responders should be investigated.
Collapse
Affiliation(s)
- Mohammad Taheri
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Shirin Nemati
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abolfazl Movafagh
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Saberi
- Department of Medical Genetics, Tehran medical University of Medical Sciences, Tehran, Iran
| | - Reza Mirfakhraie
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Mahdi Eftekharian
- Neurophysiology Research Center, Hamadan University of Medical Sciences and Health Services, Hamadan, Iran
- Molecular Immunology Research Group, Hamadan University of Medical Sciences and Health Services, Hamadan, Iran
| | - Shahram Arsang-Jang
- Department of Epidemiology and Biostatistics, Faculty of Health, Qom University of Medical Sciences, Qom, Iran
| | - Amir Rezagholizadeh
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Arezou Sayad
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
7
|
Luo H, Broux B, Wang X, Hu Y, Ghannam S, Jin W, Larochelle C, Prat A, Wu J. EphrinB1 and EphrinB2 regulate T cell chemotaxis and migration in experimental autoimmune encephalomyelitis and multiple sclerosis. Neurobiol Dis 2016; 91:292-306. [PMID: 27039370 DOI: 10.1016/j.nbd.2016.03.013] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Revised: 03/14/2016] [Accepted: 03/16/2016] [Indexed: 12/27/2022] Open
Abstract
T cells are believed to be key effector cells in multiple sclerosis (MS). In this study, we examined the roles of T cell ephrinB1 (EFNB1) and ephrinB2 (EFNB2) in the pathogenesis of experimental autoimmune encephalomyelitis (EAE) and MS. We provide evidence that animals with T cell specific double deletion of EFNB1 and EFNB2 (dKO) have reduced proliferation in response to MOG35-55, defective Th1 and Th17 differentiations and significantly lower scores of MOG-induced EAE. We further demonstrate that dKO T cells are compromised in their ability to migrate into the CNS of EAE animals in vivo and towards multiple chemokines in vitro. Using deletion mutations, we identified a critical 11-aa EFNB1 intracellular domain segment that controls T cell chemotaxis towards CCL21. In humans, EFNB1 and EFNB2 are highly expressed in Th1 and Th17 cells and EFNB1- and EFNB2-expressing T cells are found among immune cell infiltrates in MS lesions. Reverse signaling through EFNB1 and EFNB2 in human Th17 cells enhances their migration through a monolayer of blood brain barrier endothelial cells. Our study demonstrates that expression of EFNB1 and EFNB2 is implicated in Th cell differentiation and migration to inflammatory sites in both EAE and MS.
Collapse
Affiliation(s)
- Hongyu Luo
- The Research Center of the University of Montreal Hospital Center e (CRCHUM), 900 Rue Saint-Denis, Montréal H2X 0A9, Québec, Canada
| | - Bieke Broux
- The Research Center of the University of Montreal Hospital Center e (CRCHUM), 900 Rue Saint-Denis, Montréal H2X 0A9, Québec, Canada; Department of Neuroscience, Faculty of Medicine, Université de Montréal, 900 Rue Saint-Denis, Montréal H2X 0A9, Québec, Canada; Hasselt University, Biomedical Research Institute and transnationale Universiteit Limburg, School of Life Sciences, Agoralaan building C, 3590 Diepenbeek, Belgium
| | - Xuehai Wang
- The Research Center of the University of Montreal Hospital Center e (CRCHUM), 900 Rue Saint-Denis, Montréal H2X 0A9, Québec, Canada
| | - Yan Hu
- The Research Center of the University of Montreal Hospital Center e (CRCHUM), 900 Rue Saint-Denis, Montréal H2X 0A9, Québec, Canada
| | - Soufiane Ghannam
- The Research Center of the University of Montreal Hospital Center e (CRCHUM), 900 Rue Saint-Denis, Montréal H2X 0A9, Québec, Canada; Department of Neuroscience, Faculty of Medicine, Université de Montréal, 900 Rue Saint-Denis, Montréal H2X 0A9, Québec, Canada
| | - Wei Jin
- The Research Center of the University of Montreal Hospital Center e (CRCHUM), 900 Rue Saint-Denis, Montréal H2X 0A9, Québec, Canada
| | - Catherine Larochelle
- The Research Center of the University of Montreal Hospital Center e (CRCHUM), 900 Rue Saint-Denis, Montréal H2X 0A9, Québec, Canada; Department of Neuroscience, Faculty of Medicine, Université de Montréal, 900 Rue Saint-Denis, Montréal H2X 0A9, Québec, Canada
| | - Alexandre Prat
- The Research Center of the University of Montreal Hospital Center e (CRCHUM), 900 Rue Saint-Denis, Montréal H2X 0A9, Québec, Canada; Department of Neuroscience, Faculty of Medicine, Université de Montréal, 900 Rue Saint-Denis, Montréal H2X 0A9, Québec, Canada.
| | - Jiangping Wu
- The Research Center of the University of Montreal Hospital Center e (CRCHUM), 900 Rue Saint-Denis, Montréal H2X 0A9, Québec, Canada; Division of Nephrology, Department of Medicine, Faculty of Medicine, Université de Montréal, 900 Rue Saint-Denis, Montréal H2X 0A9, Québec, Canada.
| |
Collapse
|
8
|
Miraglia MC, Costa Franco MM, Rodriguez AM, Bellozi PMQ, Ferrari CC, Farias MI, Dennis VA, Barrionuevo P, de Oliveira ACP, Pitossi F, Kim KS, Delpino MV, Oliveira SC, Giambartolomei GH. Glial Cell-Elicited Activation of Brain Microvasculature in Response to Brucella abortus Infection Requires ASC Inflammasome-Dependent IL-1β Production. THE JOURNAL OF IMMUNOLOGY 2016; 196:3794-805. [PMID: 26983788 DOI: 10.4049/jimmunol.1500908] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Accepted: 02/18/2016] [Indexed: 01/18/2023]
Abstract
Blood-brain barrier activation and/or dysfunction are a common feature of human neurobrucellosis, but the underlying pathogenic mechanisms are largely unknown. In this article, we describe an immune mechanism for inflammatory activation of human brain microvascular endothelial cells (HBMEC) in response to infection with Brucella abortus Infection of HBMEC with B. abortus induced the secretion of IL-6, IL-8, and MCP-1, and the upregulation of CD54 (ICAM-1), consistent with a state of activation. Culture supernatants (CS) from glial cells (astrocytes and microglia) infected with B. abortus also induced activation of HBMEC, but to a greater extent. Although B. abortus-infected glial cells secreted IL-1β and TNF-α, activation of HBMEC was dependent on IL-1β because CS from B. abortus-infected astrocytes and microglia deficient in caspase-1 and apoptosis-associated speck-like protein containing a CARD failed to induce HBMEC activation. Consistently, treatment of CS with neutralizing anti-IL-1β inhibited HBMEC activation. Both absent in melanoma 2 and Nod-like receptor containing a pyrin domain 3 are partially required for caspase-1 activation and IL-1β secretion, suggesting that multiple apoptosis-associated speck-like protein containing CARD-dependent inflammasomes contribute to IL-1β-induced activation of the brain microvasculature. Inflammasome-mediated IL-1β secretion in glial cells depends on TLR2 and MyD88 adapter-like/TIRAP. Finally, neutrophil and monocyte migration across HBMEC monolayers was increased by CS from Brucella-infected glial cells in an IL-1β-dependent fashion, and the infiltration of neutrophils into the brain parenchyma upon intracranial injection of B. abortus was diminished in the absence of Nod-like receptor containing a pyrin domain 3 and absent in melanoma 2. Our results indicate that innate immunity of the CNS set in motion by B. abortus contributes to the activation of the blood-brain barrier in neurobrucellosis and IL-1β mediates this phenomenon.
Collapse
Affiliation(s)
- M Cruz Miraglia
- Institute of Immunology, Genetics and Metabolism (National Scientific and Technical Research Council/University of Buenos Aires), Clinical Hospital José de San Martín, Faculty of Medicine, University of Buenos Aires, Buenos Aires C1120AAR, Argentina
| | - Miriam M Costa Franco
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte-Minas Gerais 31270-901, Brazil
| | - Ana M Rodriguez
- Institute of Immunology, Genetics and Metabolism (National Scientific and Technical Research Council/University of Buenos Aires), Clinical Hospital José de San Martín, Faculty of Medicine, University of Buenos Aires, Buenos Aires C1120AAR, Argentina
| | - Paula M Q Bellozi
- Department of Pharmacology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte-Minas Gerais 31270-901, Brazil
| | - Carina C Ferrari
- Leloir Institute Foundation, Biochemical Research Institute of Buenos Aires/National Scientific and Technical Research Council, Buenos Aires C1405BWE, Argentina
| | - Maria I Farias
- Leloir Institute Foundation, Biochemical Research Institute of Buenos Aires/National Scientific and Technical Research Council, Buenos Aires C1405BWE, Argentina
| | - Vida A Dennis
- Center for NanoBiotechnology Research, Alabama State University, Montgomery, AL 36104; and
| | - Paula Barrionuevo
- Institute of Immunology, Genetics and Metabolism (National Scientific and Technical Research Council/University of Buenos Aires), Clinical Hospital José de San Martín, Faculty of Medicine, University of Buenos Aires, Buenos Aires C1120AAR, Argentina
| | - Antonio C P de Oliveira
- Department of Pharmacology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte-Minas Gerais 31270-901, Brazil
| | - Fernando Pitossi
- Leloir Institute Foundation, Biochemical Research Institute of Buenos Aires/National Scientific and Technical Research Council, Buenos Aires C1405BWE, Argentina
| | - Kwang Sik Kim
- Division of Pediatric Infectious Diseases, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21287
| | - M Victoria Delpino
- Institute of Immunology, Genetics and Metabolism (National Scientific and Technical Research Council/University of Buenos Aires), Clinical Hospital José de San Martín, Faculty of Medicine, University of Buenos Aires, Buenos Aires C1120AAR, Argentina
| | - Sergio Costa Oliveira
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte-Minas Gerais 31270-901, Brazil
| | - Guillermo H Giambartolomei
- Institute of Immunology, Genetics and Metabolism (National Scientific and Technical Research Council/University of Buenos Aires), Clinical Hospital José de San Martín, Faculty of Medicine, University of Buenos Aires, Buenos Aires C1120AAR, Argentina;
| |
Collapse
|
9
|
Pennisi M, Russo G, Motta S, Pappalardo F. Agent based modeling of the effects of potential treatments over the blood-brain barrier in multiple sclerosis. J Immunol Methods 2015; 427:6-12. [PMID: 26343337 DOI: 10.1016/j.jim.2015.08.014] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Revised: 06/15/2015] [Accepted: 08/28/2015] [Indexed: 10/23/2022]
Abstract
Multiple sclerosis is a disease of the central nervous system that involves the destruction of the insulating sheath of axons, causing severe disabilities. Since the etiology of the disease is not yet fully understood, the use of novel techniques that may help to understand the disease, to suggest potential therapies and to test the effects of candidate treatments is highly advisable. To this end we developed an agent based model that demonstrated its ability to reproduce the typical oscillatory behavior observed in the most common form of multiple sclerosis, relapsing-remitting multiple sclerosis. The model has then been used to test the potential beneficial effects of vitamin D over the disease. Many scientific studies underlined the importance of the blood-brain barrier and of the mechanisms that influence its permeability on the development of the disease. In the present paper we further extend our previously developed model with a mechanism that mimics the blood-brain barrier behavior. The goal of our work is to suggest the best strategies to follow for developing new potential treatments that intervene in the blood-brain barrier. Results suggest that the best treatments should potentially prevent the opening of the blood-brain barrier, as treatments that help in recovering the blood-brain barrier functionality could be less effective.
Collapse
Affiliation(s)
- Marzio Pennisi
- Department of Mathematics and Computer Science, University of Catania, 95125 Catania, Italy
| | - Giulia Russo
- Department of Drug Science, University of Catania, 95125 Catania, Italy
| | - Santo Motta
- Department of Mathematics and Computer Science, University of Catania, 95125 Catania, Italy
| | | |
Collapse
|
10
|
Characterization of HDAC9 isoforms in brain microvessel bEnd.3 cells upon oxygen and glucose deprivation–reperfusion injury. Genes Genomics 2015. [DOI: 10.1007/s13258-015-0282-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
11
|
Interrelation of oxidative stress and inflammation in neurodegenerative disease: role of TNF. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2015:610813. [PMID: 25834699 PMCID: PMC4365363 DOI: 10.1155/2015/610813] [Citation(s) in RCA: 462] [Impact Index Per Article: 51.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2014] [Accepted: 02/18/2015] [Indexed: 12/22/2022]
Abstract
Neuroinflammation and mitochondrial dysfunction are common features of chronic neurodegenerative diseases of the central nervous system. Both conditions can lead to increased oxidative stress by excessive release of harmful reactive oxygen and nitrogen species (ROS and RNS), which further promote neuronal damage and subsequent inflammation resulting in a feed-forward loop of neurodegeneration. The cytokine tumor necrosis factor (TNF), a master regulator of the immune system, plays an important role in the propagation of inflammation due to the activation and recruitment of immune cells via its receptor TNF receptor 1 (TNFR1). Moreover, TNFR1 can directly induce oxidative stress by the activation of ROS and RNS producing enzymes. Both TNF-induced oxidative stress and inflammation interact and cooperate to promote neurodegeneration. However, TNF plays a dual role in neurodegenerative disease, since stimulation via its second receptor, TNFR2, is neuroprotective and promotes tissue regeneration. Here we review the interrelation of oxidative stress and inflammation in the two major chronic neurodegenerative diseases, Alzheimer's and Parkinson's disease, and discuss the dual role of TNF in promoting neurodegeneration and tissue regeneration via its two receptors.
Collapse
|
12
|
Tawdy MH, Abd el Nasser MM, Abd el Shafy SS, Nada MA, El Sirafy MNI, Magd AHA. Role of serum TRAIL level and TRAIL apoptosis gene expression in multiple sclerosis and relation to brain atrophy. J Clin Neurosci 2014; 21:1606-11. [DOI: 10.1016/j.jocn.2013.11.056] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2013] [Revised: 11/28/2013] [Accepted: 11/30/2013] [Indexed: 12/28/2022]
|
13
|
Esser C, Bachmann A, Kuhn D, Schuldt K, Förster B, Thiel M, May J, Koch-Nolte F, Yáñez-Mó M, Sánchez-Madrid F, Schinkel AH, Jalkanen S, Craig AG, Bruchhaus I, Horstmann RD. Evidence of promiscuous endothelial binding by Plasmodium falciparum-infected erythrocytes. Cell Microbiol 2014; 16:701-8. [PMID: 24444337 PMCID: PMC4114535 DOI: 10.1111/cmi.12270] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2013] [Revised: 01/09/2014] [Accepted: 01/14/2014] [Indexed: 12/01/2022]
Abstract
The adhesion of infected red blood cells (iRBCs) to human endothelium is considered a key event in the pathogenesis of cerebral malaria and other life‐threatening complications caused by the most prevalent malaria parasite Plasmodium falciparum. In the past 30 years, 14 endothelial receptors for iRBCs have been identified. Exposing 10 additional surface proteins of endothelial cells to a mixture of P. falciparum isolates from three Ghanaian malaria patients, we identified seven new iRBC receptors, all expressed in brain vessels. This finding strongly suggests that endothelial binding of P. falciparum iRBCs is promiscuous and may use a combination of endothelial surface moieties.
Collapse
Affiliation(s)
- Claudia Esser
- Department of Molecular Medicine, Bernhard Nocht Institute for Tropical Medicine, Bernhard-Nocht-Strasse 74, 20359, Hamburg, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Gurevich M, Achiron A. The switch between relapse and remission in multiple sclerosis: Continuous inflammatory response balanced by Th1 suppression and neurotrophic factors. J Neuroimmunol 2012; 252:83-8. [DOI: 10.1016/j.jneuroim.2012.07.014] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2012] [Revised: 06/13/2012] [Accepted: 07/25/2012] [Indexed: 01/28/2023]
|
15
|
Mochizuki M, Sugita S, Kamoi K. Immunological homeostasis of the eye. Prog Retin Eye Res 2012; 33:10-27. [PMID: 23108335 DOI: 10.1016/j.preteyeres.2012.10.002] [Citation(s) in RCA: 111] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2012] [Revised: 10/05/2012] [Accepted: 10/05/2012] [Indexed: 12/22/2022]
Abstract
Uveitis is a sight-threatening disease caused by autoimmune or infection-related immune responses. Studies in experimental autoimmune uveitis and in human diseases imply that activated CD4(+) T cells, Th1 and Th17 cells, play an effector role in ocular inflammation. The eye has a unique regional immune system to protect vision-related cells and tissues from these effector T cells. The immunological balance between the pathogenic CD4(+) T cells and regional immune system in the eye contributes to the maintenance of ocular homeostasis and good vision. Current studies have demonstrated that ocular parenchymal cells at the inner surface of the blood-ocular barrier, i.e. corneal endothelial (CE) cells, iris pigment epithelial (PE) cells, ciliary body PE cells, and retinal PE cells, contribute to the regional immune system of the eye. Murine ocular resident cells directly suppress activation of bystander T cells and production of inflammatory cytokines. The ocular resident cells possess distinct properties of immunoregulation that are related to disparate anatomical location. CE cells and iris PE cells, which are located at the anterior segment of the eye and face the aqueous humor, suppress activation of T cells via cell-to-cell contact mechanisms, whereas retinal PE cells suppress the activation of T cells via soluble factors. In addition to direct immune suppression, the ocular resident cells have another unique immunosuppressive property, the induction of CD25(+)Foxp3(+) Treg cells that also suppress the activation of bystander T cells. Iris PE cells convert CD8(+) T cells into Treg cells, while retinal PE cells convert CD4(+) T cells greatly and CD8(+) T cells moderately into Treg cells. CE cells also convert both CD4(+) T cells and CD8(+) T cells into Treg cells. The immunomodulation by ocular resident cells is mediated by various soluble or membrane-bound molecules that include TGF-β TSP-1, B7-2 (CD86), CTLA-2α, PD-L1 (B7-H1), galectin 1, pigment epithelial-derived factor PEDF), GIRTL, and retinoic acid. Human retinal PE cells also possess similar immune properties to induce Treg cells. Although there are many issues to be answered, human Treg cells induced by ocular resident cells such as retinal PE cells and related immunosuppressive molecules can be applied as immune therapy for refractive autoimmune uveitis in humans in the future.
Collapse
Affiliation(s)
- Manabu Mochizuki
- Department of Ophthalmology & Visual Science, Tokyo Medical and Dental University, 1-5-45 Yushima, Tokyo 113-8519, Japan.
| | | | | |
Collapse
|
16
|
Lopez-Ramirez MA, Fischer R, Torres-Badillo CC, Davies HA, Logan K, Pfizenmaier K, Male DK, Sharrack B, Romero IA. Role of caspases in cytokine-induced barrier breakdown in human brain endothelial cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2012; 189:3130-9. [PMID: 22896632 DOI: 10.4049/jimmunol.1103460] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
During neuroinflammation, cytokines such as TNF-α and IFN-γ secreted by activated leukocytes and/or CNS resident cells have been shown to alter the phenotype and function of brain endothelial cells (BECs) leading to blood-brain barrier breakdown. In this study, we show that the human BEC line hCMEC/D3 expresses the receptors for TNF-α, TNF receptor 1 and TNF receptor 2, and for IFN-γ. BEC activation with TNF-α alone or in combination with IFN-γ induced endothelial leakage of paracellular tracers. At high cytokine concentrations (10 and 100 ng/ml), this effect was associated with caspase-3/7 activation and apoptotic cell death as evidenced by annexin V staining and DNA fragmentation (TUNEL) assays. In addition, inhibition of JNK and protein kinase C activation at these doses partially prevented activation of caspase-3/7, although only JNK inhibition was partially able to prevent the increase in BEC paracellular permeability induced by cytokines. By contrast, lower cytokine concentrations (1 ng/ml) also led to effector caspase activation, increased paracellular flux, and redistribution of zonula occludens-1 and VE-cadherin but failed to induce apoptosis. Under these conditions, specific caspase-3 and caspase-9, but not caspase-8, inhibitors partially blocked cytokine-induced disruption of tight and adherens junctions and BEC paracellular permeability. Our results suggest that the concentration of cytokines in the CNS endothelial microenvironment determines the extent of caspase-mediated barrier permeability changes, which may be generalized as a result of apoptosis or more subtle as a result of alterations in the organization of junctional complex molecules.
Collapse
MESH Headings
- Blood-Brain Barrier/enzymology
- Blood-Brain Barrier/immunology
- Blood-Brain Barrier/pathology
- Brain/enzymology
- Brain/immunology
- Brain/pathology
- Cell Line
- Cytokines/physiology
- Endothelium, Vascular/enzymology
- Endothelium, Vascular/immunology
- Endothelium, Vascular/pathology
- Humans
- Inflammation Mediators/metabolism
- Inflammation Mediators/physiology
- Interferon-gamma/metabolism
- Microcirculation/immunology
- Receptors, Interferon/biosynthesis
- Receptors, Tumor Necrosis Factor, Type I/biosynthesis
- Receptors, Tumor Necrosis Factor, Type I/metabolism
- Receptors, Tumor Necrosis Factor, Type II/biosynthesis
- Receptors, Tumor Necrosis Factor, Type II/metabolism
- Subcellular Fractions/enzymology
- Subcellular Fractions/immunology
- Subcellular Fractions/pathology
- Interferon gamma Receptor
Collapse
|
17
|
Johnson HL, Chen Y, Suidan GL, McDole JR, Lohrey AK, Hanson LM, Jin F, Pirko I, Johnson AJ. A hematopoietic contribution to microhemorrhage formation during antiviral CD8 T cell-initiated blood-brain barrier disruption. J Neuroinflammation 2012; 9:60. [PMID: 22452799 PMCID: PMC3350446 DOI: 10.1186/1742-2094-9-60] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2011] [Accepted: 03/27/2012] [Indexed: 01/09/2023] Open
Abstract
Background The extent to which susceptibility to brain hemorrhage is derived from blood-derived factors or stromal tissue remains largely unknown. We have developed an inducible model of CD8 T cell-initiated blood-brain barrier (BBB) disruption using a variation of the Theiler's murine encephalomyelitis virus (TMEV) model of multiple sclerosis. This peptide-induced fatal syndrome (PIFS) model results in severe central nervous system (CNS) vascular permeability and death in the C57BL/6 mouse strain, but not in the 129 SvIm mouse strain, despite the two strains' having indistinguishable CD8 T-cell responses. Therefore, we hypothesize that hematopoietic factors contribute to susceptibility to brain hemorrhage, CNS vascular permeability and death following induction of PIFS. Methods PIFS was induced by intravenous injection of VP2121-130 peptide at 7 days post-TMEV infection. We then investigated brain inflammation, astrocyte activation, vascular permeability, functional deficit and microhemorrhage formation using T2*-weighted magnetic resonance imaging (MRI) in C57BL/6 and 129 SvIm mice. To investigate the contribution of hematopoietic cells in this model, hemorrhage-resistant 129 SvIm mice were reconstituted with C57BL/6 or autologous 129 SvIm bone marrow. Gadolinium-enhanced, T1-weighted MRI was used to visualize the extent of CNS vascular permeability after bone marrow transfer. Results C57BL/6 and 129 SvIm mice had similar inflammation in the CNS during acute infection. After administration of VP2121-130 peptide, however, C57BL/6 mice had increased astrocyte activation, CNS vascular permeability, microhemorrhage formation and functional deficits compared to 129 SvIm mice. The 129 SvIm mice reconstituted with C57BL/6 but not autologous bone marrow had increased microhemorrhage formation as measured by T2*-weighted MRI, exhibited a profound increase in CNS vascular permeability as measured by three-dimensional volumetric analysis of gadolinium-enhanced, T1-weighted MRI, and became moribund in this model system. Conclusion C57BL/6 mice are highly susceptible to microhemorrhage formation, severe CNS vascular permeability and morbidity compared to the 129 SvIm mouse. This susceptibility is transferable with the bone marrow compartment, demonstrating that hematopoietic factors are responsible for the onset of brain microhemorrhage and vascular permeability in immune-mediated fatal BBB disruption.
Collapse
|
18
|
A shear stress responsive gene product PP1201 protects against Fas-mediated apoptosis by reducing Fas expression on the cell surface. Apoptosis 2011; 16:162-73. [PMID: 21107705 DOI: 10.1007/s10495-010-0556-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Cells that form vascular system employ different mechanisms to offset deleterious consequences of exposure to cytokines and cells present in blood. Vascular homeostasis is sustained in part by genes, whose expression increases in response to hemodynamic forces in these cells. PP1201 (also known as RECS1) is one such gene whose expression level increases in response to laminar shear stress. Aged mice deficient in PP1201 are prone to develop cystic medial degeneration (CMD), a form of aortic aneurism manifested with loss of smooth muscle cells and accumulation of basophilic substances. Here we found that higher levels of PP1201 can protect against Fas ligand (FasL)-induced apoptosis. PP1201 interacted with the Fas receptor (CD95/Apo1) and colocalized with it in the Golgi compartment. Unlike its homolog lifeguard (LFG), PP1201 overexpression in several types of cells including primary human aortic smooth muscle cells (AoSMC) decreased the expression of Fas on the plasma membrane without changing the total Fas levels. Only high but not constitutive level of PP1201 controls Fas signaling. Our data suggest that PP1201 functions as an anti-apoptotic protein and its increased expression in vascular cells can contribute to homeostasis by reducing Fas trafficking to the cell membrane.
Collapse
|
19
|
Perrot-Applanat M, Vacher S, Toullec A, Pelaez I, Velasco G, Cormier F, Saad HES, Lidereau R, Baud V, Bièche I. Similar NF-κB gene signatures in TNF-α treated human endothelial cells and breast tumor biopsies. PLoS One 2011; 6:e21589. [PMID: 21754991 PMCID: PMC3130773 DOI: 10.1371/journal.pone.0021589] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2010] [Accepted: 06/06/2011] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Endothelial dysfunction has been implicated in the pathogenesis of diverse pathologies ranging from vascular and immune diseases to cancer. TNF-α is one of the mediators of endothelial dysfunction through the activation of transcription factors, including NF-κB. While HUVEC (macrovascular cells) have been largely used in the past, here, we documented an NF-κB gene signature in TNFα-stimulated microvascular endothelial cells HMEC often used in tumor angiogenesis studies. METHODOLOGY/PRINCIPAL FINDINGS We measured mRNA expression of 55 NF-κB related genes using quantitative RT-PCR in HUVEC and HMEC. Our study identified twenty genes markedly up-regulated in response to TNFα, including adhesion molecules, cytokines, chemokines, and apoptosis regulators, some of them being identified as TNF-α-inducible genes for the first time in endothelial cells (two apoptosis regulators, TNFAIP3 and TNFRSF10B/Trail R2 (DR5), the chemokines GM-CSF/CSF2 and MCF/CSF1, and CD40 and TNF-α itself, as well as NF-κB components (RELB, NFKB1 or 50/p105 and NFKB2 or p52/p100). For eight genes, the fold induction was much higher in HMEC, as compared to HUVEC. Most importantly, our study described for the first time a connection between NF-κB activation and the induction of most, if not all, of these genes in HMEC as evaluated by pharmacological inhibition and RelA expression knock-down by RNA interference. Moreover, since TNF-α is highly expressed in tumors, we further applied the NF-κB gene signature documented in TNFα-stimulated endothelial cells to human breast tumors. We found a significant positive correlation between TNF and the majority (85 %) of the identified endothelial TNF-induced genes in a well-defined series of 96 (48 ERα positive and 48 ERα negative) breast tumors. CONCLUSION/SIGNIFICANCE Taken together these data suggest the potential use of this NF-κB gene signature in analyzing the role of TNF-α in the endothelial dysfunction, as well as in breast tumors independently of the presence of ERα.
Collapse
|
20
|
Kim H, Folks KD, Guo L, Stockard CR, Fineberg NS, Grizzle WE, George JF, Buchsbaum DJ, Morgan DE, Zinn KR. DCE-MRI detects early vascular response in breast tumor xenografts following anti-DR5 therapy. Mol Imaging Biol 2011; 13:94-103. [PMID: 20383593 DOI: 10.1007/s11307-010-0320-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
PURPOSE Dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) measured the early vascular changes after administration of TRA-8, bevacizumab, or TRA-8 combined with bevacizumab in breast tumor xenografts. PROCEDURES Groups 1-4 of nude mice bearing human breast carcinoma were injected with phosphate-buffered saline, TRA-8, bevacizumab, and TRA-8 + bevacizumab on day 0, respectively. DCE-MRI was performed on days 0, 1, 2, and 3, and thereafter tumors were collected for terminal deoxynucleotidyl transferase-mediated dUT nick end labeling and CD31 staining. RESULTS DCE-MRI measured a significant K (trans) change within 3 days after TRA-8 therapy that correlated with tumor growth arrest, which was not shown with statistical significance by histopathology at these early time points posttreatment. The K (trans) changes followed quadratic polynomial curves. CONCLUSION DCE-MRI detected significantly lower K (trans) levels in breast tumor xenografts following TRA-8 monotherapy or combined therapy with bevacizumab.
Collapse
Affiliation(s)
- Hyunki Kim
- The Department of Radiology, University of Alabama at Birmingham, AL 35294, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Larochelle C, Alvarez JI, Prat A. How do immune cells overcome the blood-brain barrier in multiple sclerosis? FEBS Lett 2011; 585:3770-80. [PMID: 21550344 DOI: 10.1016/j.febslet.2011.04.066] [Citation(s) in RCA: 271] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2011] [Revised: 04/26/2011] [Accepted: 04/27/2011] [Indexed: 11/17/2022]
Abstract
The presence of the blood-brain barrier (BBB) restricts the movement of soluble mediators and leukocytes from the periphery to the central nervous system (CNS). Leukocyte entry into the CNS is nonetheless an early event in multiple sclerosis (MS), an inflammatory disorder of the CNS. Whether BBB dysfunction precedes immune cell infiltration or is the consequence of perivascular leukocyte accumulation remains enigmatic, but leukocyte migration modifies BBB permeability. Immune cells of MS subjects express inflammatory cytokines, reactive oxygen species (ROS) and enzymes that can facilitate their migration to the CNS by influencing BBB function, either directly or indirectly. In this review, we describe how immune cells from the peripheral blood overcome the BBB and promote CNS inflammation in MS through BBB disruption.
Collapse
Affiliation(s)
- Catherine Larochelle
- Neuroimmunology Research Laboratory, Center of Excellence in Neuromics, CRCHUM, Notre-Dame Hospital, Faculty of Medicine, Université de Montréal, Montréal, Québec, Canada
| | | | | |
Collapse
|
22
|
Rizzo MT, Leaver HA. Brain Endothelial Cell Death: Modes, Signaling Pathways, and Relevance to Neural Development, Homeostasis, and Disease. Mol Neurobiol 2010; 42:52-63. [DOI: 10.1007/s12035-010-8132-6] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2010] [Accepted: 04/05/2010] [Indexed: 12/31/2022]
|
23
|
Knight JC, Scharf EL, Mao-Draayer Y. Fas activation increases neural progenitor cell survival. J Neurosci Res 2010; 88:746-57. [PMID: 19830835 DOI: 10.1002/jnr.22253] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Although there is a sizable amount of research focusing on adult neural progenitor cells (NPCs) as a therapeutic approach for many neurodegenerative diseases, including multiple sclerosis, little is known about the pathways that govern NPC survival and apoptosis. Fas, a member of the death receptor superfamily, plays a well-characterized role in the immune system, but its function in neural stem cells remains uncertain. Our study focuses on the effects of Fas on NPC survival in vitro. Activation of Fas by recombinant Fas ligand (FasL) did not induce apoptosis in murine NPCs in culture. In fact, both an increase in the amount of viable cells and a decrease in apoptotic and dying cells were observed with FasL treatment. Our data indicate that FasL-mediated adult NPC neuroprotection is characterized by a reduction in apoptosis, but not increased proliferation. Further investigation of this effect revealed that the antiapoptotic effects of FasL are mediated by the up-regulation of Birc3, an inhibitor of apoptosis protein (IAP). Conversely, the observed effect is not the result of altered caspase activation or FLIP (Fas-associated death domain-like interleukin-1beta-converting enzyme inhibitory protein) up-regulation, which is known to inhibit caspase-8-mediated cell death in T cells. Our data indicate that murine adult NPCs are resistant to FasL-induced cell death. Activation of Fas increased cell survival by decreasing apoptosis through Birc3 up-regulation. These results describe a novel pathway involved in NPC survival.
Collapse
Affiliation(s)
- Julia C Knight
- Neuroscience Graduate Program, University of Vermont, Burlington, Vermont, USA
| | | | | |
Collapse
|
24
|
Evidence that tumor necrosis factor-related apoptosis inducing ligand (TRAIL) inhibits angiogenesis by inducing vascular endothelial cell apoptosis. Biochem Biophys Res Commun 2009; 391:936-41. [PMID: 19962958 DOI: 10.1016/j.bbrc.2009.11.168] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2009] [Accepted: 11/26/2009] [Indexed: 01/18/2023]
Abstract
Tumor necrosis factor (TNF) and its related ligands TNF-related apoptosis inducing ligand (TRAIL) and Fas ligand (FasL) play roles in the regulation of vascular responses, but their effect on the formation of new blood vessels (angiogenesis) is unclear. Therefore, we have examined the effects of these ligands on angiogenesis modeled with primary cultures of human umbilical vein endothelial cells (HUVEC). To examine angiogenesis in the context of the central nervous system, we have also modeled cerebral angiogenesis with the human brain endothelial cell line hCMEC/D3. Parameters studied were bromodeoxyuridine (BrdU) incorporation and cell number (MTT) assay (to assess endothelial proliferation), scratch assay (migration) and networks on Matrigel (tube formation). In our hands, neither TRAIL nor FasL (1, 10, and 100 ng/ml) had an effect on parameters of angiogenesis in the HUVEC model. In hCMEC/D3 cells by contrast, TRAIL inhibited all parameters (10-100 ng/ml, 24h). This was due to apoptosis, since its action was blocked by the pan-caspase inhibitor zVADfmk (5 x 10(-5) mol/l) and TRAIL increased caspase-3 activity 1h after application. However FasL (100 ng/ml) increased BrdU uptake without other effects. We conclude that TRAIL has different effects on in vitro angiogenesis depending on which model is used, but that FasL is generally ineffective when applied in vitro. The data suggest that TRAIL primarily influences angiogenesis by the induction of vascular endothelial apoptosis, leading to vessel regression.
Collapse
|
25
|
Hubert KE, Davies MH, Stempel AJ, Griffith TS, Powers MR. TRAIL-deficient mice exhibit delayed regression of retinal neovascularization. THE AMERICAN JOURNAL OF PATHOLOGY 2009; 175:2697-708. [PMID: 19893042 DOI: 10.2353/ajpath.2009.090099] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
While it is well established that tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) induces apoptosis in various cell types, the role of TRAIL in regulation of retinal neovascularization (NV) has not been described. Here we determined the role of TRAIL in retinal NV during oxygen-induced retinopathy using TRAIL deficient ((-/-)) mice. TRAIL and its receptor, DR5, were expressed in wild-type retinas at all time points evaluated (postnatal days 12, 17, 21, 24) during oxygen-induced retinopathy and in age-matched room air control animals. Localization of TRAIL(+) cells within the neovascular tufts of hyperoxia- exposed wild-type mice suggested TRAIL plays a role in oxygen-induced retinopathy. Retinal vascular development appeared normal in the TRAIL(-/-) mice, except for a small but significant difference in the capillary-free zone surrounding major arteries. A minimal difference in avascularity was observed at postnatal day 12 in the retinas of TRAIL(-/-) mice after hyperoxia-exposure compared with wild-type mice, suggesting that TRAIL does not play a major role in the vaso-obliterative phase of oxygen-induced retinopathy. However, at the peak of NV, TRAIL(-/-) mice had a significant increase in retinal neovascularization. In addition, when NV naturally regresses in wild-type mice, TRAIL(-/-) mice continued to display significantly high levels of NV. This was attributed to a significant decrease in neovascular tuft cells undergoing apoptosis in TRAIL(-/-) mice. Together, these data strongly suggest that TRAIL plays a role in the control of retinal NV.
Collapse
Affiliation(s)
- Kristin E Hubert
- Department of Pediatrics, Casey Eye Institute, Oregon Health & Science University, Portland, Oregon 97239-4197, USA
| | | | | | | | | |
Collapse
|
26
|
|
27
|
Hoffmann O, Zipp F, Weber JR. Tumour necrosis factor-related apoptosis-inducing ligand (TRAIL) in central nervous system inflammation. J Mol Med (Berl) 2009; 87:753-63. [DOI: 10.1007/s00109-009-0484-x] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2009] [Revised: 05/05/2009] [Accepted: 05/08/2009] [Indexed: 12/17/2022]
|
28
|
Rinta S, Kuusisto H, Raunio M, Paalavuo R, Levula M, Lehtimäki T, Elovaara I. Apoptosis-related molecules in blood in multiple sclerosis. J Neuroimmunol 2008; 205:135-41. [PMID: 18963025 DOI: 10.1016/j.jneuroim.2008.09.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2008] [Revised: 09/02/2008] [Accepted: 09/05/2008] [Indexed: 12/23/2022]
Abstract
A failure in apoptosis of lymphocytes may lead to harmful immunoreactivity in MS. We analyzed apoptosis-related molecules including TRAIL, sFas, sFasL and MIF in the blood of 117 MS patients and controls to answer whether these molecules may be used in the evaluation of disease activity and immunomodulatory effect of IFN-beta. Increased levels of sTRAIL, sFasL and MIF were found in sera of untreated patients with MS relapse indicating their association with MS disease activity. IFN-beta treated patients in remission had increased TRAIL mRNA, sTRAIL, sFaL and MIF that most likely reflect bioactivity of IFN-beta.
Collapse
Affiliation(s)
- Sanna Rinta
- Neuroimmunology Unit, Department of Neurology, Finn-Medi 3, Biokatu 10, 33014 University of Tampere, Finland
| | | | | | | | | | | | | |
Collapse
|
29
|
Forrester JV, Xu H, Lambe T, Cornall R. Immune privilege or privileged immunity? Mucosal Immunol 2008; 1:372-81. [PMID: 19079201 DOI: 10.1038/mi.2008.27] [Citation(s) in RCA: 92] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Immune privilege is a concept that has come of age. Where previously it was considered to be a passive phenomenon restricted to certain specialized tissues, it is now viewed as comprising several mechanisms, both active and passive, shared in many aspects with emerging notions of the mechanisms of peripheral tolerance. The relative degrees of immune privilege vary from tissue to tissue depending on the number and strength of each of the mechanisms contained in that tissue. Immune privilege can be generated in non-privileged sites such as the skin and allografts, and is a property of the tissue itself. We therefore propose that, in addition to canonical central and peripheral tolerance mechanisms, there is a third route whereby the organism promotes self-antigen non-reactivity centered on the specific properties of each tissue and varying accordingly (relative degrees of immune privilege). This third mechanism of inducing immunological tolerance, as it is a local tissue phenomenon, might have particular therapeutic significance, for instance in devising strategies for induction of immunity to tumors by disrupting immune privilege or in preventing graft rejection by promoting immune privilege.
Collapse
Affiliation(s)
- J V Forrester
- Department of Ophthalmology, University of Aberdeen, Aberdeen, Scotland.
| | | | | | | |
Collapse
|
30
|
Sandoval KE, Witt KA. Blood-brain barrier tight junction permeability and ischemic stroke. Neurobiol Dis 2008; 32:200-19. [PMID: 18790057 DOI: 10.1016/j.nbd.2008.08.005] [Citation(s) in RCA: 745] [Impact Index Per Article: 46.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2008] [Revised: 07/29/2008] [Accepted: 08/10/2008] [Indexed: 01/01/2023] Open
Abstract
The blood-brain barrier (BBB) is formed by the endothelial cells of cerebral microvessels, providing a dynamic interface between the peripheral circulation and the central nervous system. The tight junctions (TJs) between the endothelial cells serve to restrict blood-borne substances from entering the brain. Under ischemic stroke conditions decreased BBB TJ integrity results in increased paracellular permeability, directly contributing to cerebral vasogenic edema, hemorrhagic transformation, and increased mortality. This loss of TJ integrity occurs in a phasic manner, which is contingent on several interdependent mechanisms (ionic dysregulation, inflammation, oxidative and nitrosative stress, enzymatic activity, and angiogenesis). Understanding the inter-relation of these mechanisms is critical for the development of new therapies. This review focuses on those aspects of ischemic stroke impacting BBB TJ integrity and the principle regulatory pathways, respective to the phases of paracellular permeability.
Collapse
Affiliation(s)
- Karin E Sandoval
- Department of Pharmaceutical Sciences, School of Pharmacy, Southern Illinois University Edwardsville, Edwardsville, IL 62026, USA
| | | |
Collapse
|