Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Acta Neuropathol ; 128(5): 691-703, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25149081

ABSTRACT

Multiple sclerosis (MS) lesions are characterized by the presence of activated astrocytes, which are thought to actively take part in propagating lesion progression by secreting pro-inflammatory mediators. Conversely, reactive astrocytes may exert disease-dampening effects through the production of trophic factors and anti-inflammatory mediators. Astrocytic control of the blood-brain barrier (BBB) is crucial for normal brain homeostasis and BBB disruption is a well-established early event in MS lesion development. Here, we set out to unravel potential protective effects of reactive astrocytes on BBB function under neuroinflammatory conditions as seen in MS, where we focus on the role of the brain morphogen retinoic acid (RA). Immunohistochemical analysis revealed that retinaldehyde dehydrogenase 2 (RALDH2), a key enzyme for RA synthesis, is highly expressed by reactive astrocytes throughout white matter lesions compared to control and normal appearing white matter. In vitro modeling of reactive astrocytes resulted in increased expression of RALDH2, enhanced RA synthesis, and a protective role for astrocyte-derived RA on BBB function during inflammation-induced barrier loss. Furthermore, RA induces endothelial immune quiescence and decreases monocyte adhesion under inflammatory conditions. Finally, we demonstrated that RA attenuated oxidative stress in inflamed endothelial cells, through activation of the antioxidant transcription factor nuclear factor E2 related factor 2. In summary, RA synthesis by reactive astrocytes represents an endogenous protective response to neuroinflammation, possibly aimed at protecting the BBB against inflammatory insult. A better understanding of RA signaling in MS pathophysiology may lead to the discovery of novel targets to halt disease progression.


Subject(s)
Astrocytes/drug effects , Blood-Brain Barrier/physiopathology , Brain/pathology , Multiple Sclerosis/pathology , Tretinoin/pharmacology , Adult , Aged , Aged, 80 and over , Aldehyde Dehydrogenase/genetics , Aldehyde Dehydrogenase/metabolism , Aldehyde Dehydrogenase 1 Family , Astrocytes/metabolism , Autopsy , Cells, Cultured , Cytokines/metabolism , Endothelial Cells/drug effects , Endothelial Cells/physiology , Female , Glial Fibrillary Acidic Protein/metabolism , HEK293 Cells , Humans , Male , Middle Aged , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Reactive Oxygen Species/metabolism , Retinal Dehydrogenase/genetics , Retinal Dehydrogenase/metabolism , Time Factors
2.
Acta Neuropathol ; 127(5): 699-711, 2014 May.
Article in English | MEDLINE | ID: mdl-24429546

ABSTRACT

The trafficking of cytotoxic CD8(+) T lymphocytes across the lining of the cerebral vasculature is key to the onset of the chronic neuro-inflammatory disorder multiple sclerosis. However, the mechanisms controlling their final transmigration across the brain endothelium remain unknown. Here, we describe that CD8(+) T lymphocyte trafficking into the brain is dependent on the activity of the brain endothelial adenosine triphosphate-binding cassette transporter P-glycoprotein. Silencing P-glycoprotein activity selectively reduced the trafficking of CD8(+) T cells across the brain endothelium in vitro as well as in vivo. In response to formation of the T cell-endothelial synapse, P-glycoprotein was found to regulate secretion of endothelial (C-C motif) ligand 2 (CCL2), a chemokine that mediates CD8(+) T cell migration in vitro. Notably, CCL2 levels were significantly enhanced in microvessels isolated from human multiple sclerosis lesions in comparison with non-neurological controls. Endothelial cell-specific elimination of CCL2 in mice subjected to experimental autoimmune encephalomyelitis also significantly diminished the accumulation of CD8(+) T cells compared to wild-type animals. Collectively, these results highlight a novel (patho)physiological role for P-glycoprotein in CD8(+) T cell trafficking into the central nervous system during neuro-inflammation and illustrate CCL2 secretion as a potential link in this mechanism.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B/metabolism , Brain/immunology , CD8-Positive T-Lymphocytes/physiology , Encephalomyelitis, Autoimmune, Experimental/immunology , Multiple Sclerosis/immunology , Transendothelial and Transepithelial Migration/physiology , ATP Binding Cassette Transporter, Subfamily B/genetics , Animals , Blood-Brain Barrier/physiology , Brain/blood supply , Brain/pathology , CD4-Positive T-Lymphocytes/physiology , Cell Line , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Humans , Mice, Inbred C57BL , Mice, Knockout , Microvessels/pathology , Microvessels/physiopathology , Multiple Sclerosis/pathology , ATP-Binding Cassette Sub-Family B Member 4
3.
Acta Neuropathol ; 128(2): 267-77, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24356983

ABSTRACT

Multiple sclerosis (MS) is a chronic neuro-inflammatory disorder, which is marked by the invasion of the central nervous system by monocyte-derived macrophages and autoreactive T cells across the brain vasculature. Data from experimental animal models recently implied that the passage of leukocytes across the brain vasculature is preceded by their traversal across the blood-cerebrospinal fluid barrier (BCSFB) of the choroid plexus. The correlation between the presence of leukocytes in the CSF of patients suffering from MS and the number of inflammatory lesions as detected by magnetic resonance imaging suggests that inflammation at the choroid plexus contributes to the disease, although in a yet unknown fashion. We here provide first insights into the involvement of the choroid plexus in the onset and severity of the disease and in particular address the role of the tight junction protein claudin-3 (CLDN3) in this process. Detailed analysis of human post-mortem brain tissue revealed a selective loss of CLDN3 at the choroid plexus in MS patients compared to control tissues. Importantly, mice that lack CLDN3 have an impaired BCSFB and experience a more rapid onset and exacerbated clinical signs of experimental autoimmune encephalomyelitis, which coincides with enhanced levels of infiltrated leukocytes in their CSF. Together, this study highlights a profound role for the choroid plexus in the pathogenesis of multiple sclerosis, and implies that CLDN3 may be regarded as a crucial and novel determinant of BCSFB integrity.


Subject(s)
Choroid Plexus/physiopathology , Claudin-3/metabolism , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Multiple Sclerosis/physiopathology , Adult , Aged , Aged, 80 and over , Animals , Brain/blood supply , Brain/pathology , Brain/physiopathology , Choroid Plexus/pathology , Claudin-3/genetics , Disease Progression , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Humans , Male , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Microvessels/pathology , Microvessels/physiopathology , Middle Aged , Multiple Sclerosis/pathology , Myelin-Oligodendrocyte Glycoprotein , Peptide Fragments , Severity of Illness Index
4.
Glia ; 61(11): 1890-905, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24038577

ABSTRACT

Early events in multiple sclerosis (MS) lesion formation are loss of blood-brain barrier (BBB) integrity, immune cell trafficking into the central nervous system, and demyelination. To date, the molecular mechanisms underlying these pathogenic events are poorly understood. Heparin-binding epidermal growth factor (HB-EGF) is a trophic factor that is induced by inflammatory stimuli and has previously been shown to interact with tetraspanins (TSPs), a family of transmembrane proteins that are involved in cellular migration and adhesion. Given the known roles of TSPs and HB-EGF, we hypothesized that HB-EGF and TSPs may play a role in the processes that underlie MS lesion formation. We examined the expression of HB-EGF and the TSPs CD9 and CD81 in MS brain and found that HB-EGF was highly induced in reactive astrocytes in active lesions. TSPs were constitutively expressed throughout normal appearing white matter and control white matter. In contrast, CD9 was reduced in demyelinated lesions and increased on blood vessels in lesion areas. In vitro studies revealed that expression of HB-EGF and TSPs is regulated during inflammation. Importantly, blocking either HB-EGF or CD9 significantly reduced the migration of monocytes across brain endothelial cell monolayers. Moreover, blocking CD9 strongly enhanced the barrier function of the BBB in vitro. Together, we demonstrate that these molecules are likely implicated in processes that are highly relevant for MS lesion formation, and therefore, HB-EGF and TSPs are promising therapeutic targets.


Subject(s)
Blood-Brain Barrier/metabolism , Brain/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Multiple Sclerosis/metabolism , Tetraspanin 29/metabolism , Adult , Aged , Aged, 80 and over , Brain/pathology , Cell Movement/physiology , Female , Heparin-binding EGF-like Growth Factor , Humans , Male , Membrane Proteins/metabolism , Middle Aged , Multiple Sclerosis/pathology
5.
J Neurosci ; 33(16): 6857-63, 2013 Apr 17.
Article in English | MEDLINE | ID: mdl-23595744

ABSTRACT

Blood-brain barrier (BBB) dysfunction is a major hallmark of many neurological diseases, including multiple sclerosis (MS). Using a genomics approach, we defined a microRNA signature that is diminished at the BBB of MS patients. In particular, miR-125a-5p is a key regulator of brain endothelial tightness and immune cell efflux. Our findings suggest that repair of a disturbed BBB through microRNAs may represent a novel avenue for effective treatment of MS.


Subject(s)
Blood-Brain Barrier/physiopathology , Brain/pathology , Endothelial Cells/physiology , Inflammation/pathology , MicroRNAs/metabolism , Multiple Sclerosis/pathology , Blood-Brain Barrier/drug effects , Cell Line, Transformed , Cytokines/metabolism , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Gene Expression Regulation/drug effects , Genetic Vectors/physiology , Humans , MicroRNAs/genetics , RNA, Small Interfering/pharmacology , Transendothelial and Transepithelial Migration/drug effects , Transfection
6.
J Neurosci ; 33(4): 1660-71, 2013 Jan 23.
Article in English | MEDLINE | ID: mdl-23345238

ABSTRACT

The blood-brain barrier (BBB) is crucial in the maintenance of a controlled environment within the brain to safeguard optimal neuronal function. The endothelial cells (ECs) of the BBB possess specific properties that restrict the entry of cells and metabolites into the CNS. The specialized BBB endothelial phenotype is induced during neurovascular development by surrounding cells of the CNS. However, the molecular differentiation of the BBB endothelium remains poorly understood. Retinoic acid (RA) plays a crucial role in the brain during embryogenesis. Because radial glial cells supply the brain with RA during the developmental cascade and associate closely with the developing vasculature, we hypothesize that RA is important for the induction of BBB properties in brain ECs. Analysis of human postmortem fetal brain tissue shows that the enzyme mainly responsible for RA synthesis, retinaldehyde dehydrogenase, is expressed by radial glial cells. In addition, the most important receptor for RA-driven signaling in the CNS, RA-receptor ß (RARß), is markedly expressed by the developing brain vasculature. Our findings have been further corroborated by in vitro experiments showing RA- and RARß-dependent induction of different aspects of the brain EC barrier. Finally, pharmacologic inhibition of RAR activation during the differentiation of the murine BBB resulted in the leakage of a fluorescent tracer as well as serum proteins into the developing brain and reduced the expression levels of important BBB determinants. Together, our results point to an important role for RA in the induction of the BBB during human and mouse development.


Subject(s)
Blood-Brain Barrier/embryology , Blood-Brain Barrier/metabolism , Neuroglia/metabolism , Tretinoin/metabolism , Animals , Blotting, Western , Cell Differentiation/physiology , Cell Line , Endothelial Cells/cytology , Endothelial Cells/metabolism , Fetus , Gene Expression Regulation, Developmental , Humans , Immunohistochemistry , Mice , Mice, Inbred C57BL , Real-Time Polymerase Chain Reaction , Signal Transduction/physiology
7.
Acta Neuropathol ; 125(2): 231-43, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23073717

ABSTRACT

There is growing evidence that mitochondrial dysfunction and associated reactive oxygen species (ROS) formation contribute to neurodegenerative processes in multiple sclerosis (MS). Here, we investigated whether alterations in transcriptional regulators of key mitochondrial proteins underlie mitochondrial dysfunction in MS cortex and contribute to neuronal loss. Hereto, we analyzed the expression of mitochondrial transcriptional (co-)factors and proteins involved in mitochondrial redox balance regulation in normal-appearing grey matter (NAGM) samples of cingulate gyrus and/or frontal cortex from 15 MS patients and nine controls matched for age, gender and post-mortem interval. PGC-1α, a transcriptional co-activator and master regulator of mitochondrial function, was consistently and significantly decreased in pyramidal neurons in the deeper layers of MS cortex. Reduced PGC-1α levels coincided with reduced expression of oxidative phosphorylation subunits and a decrease in gene and protein expression of various mitochondrial antioxidants and uncoupling proteins (UCPs) 4 and 5. Short-hairpin RNA-mediated silencing of PGC-1α in a neuronal cell line confirmed that reduced levels of PGC-1α resulted in a decrease in transcription of OxPhos subunits, mitochondrial antioxidants and UCPs. Moreover, PGC-1α silencing resulted in a decreased mitochondrial membrane potential, increased ROS formation and enhanced susceptibility to ROS-induced cell death. Importantly, we found extensive neuronal loss in NAGM from cingulate gyrus and frontal cortex of MS patients, which significantly correlated with the extent of PGC-1α decrease. Taken together, our data indicate that reduced neuronal PGC-1α expression in MS cortex partly underlies mitochondrial dysfunction in MS grey matter and thereby contributes to neurodegeneration in MS cortex.


Subject(s)
Cerebral Cortex/pathology , Heat-Shock Proteins/physiology , Mitochondria/pathology , Multiple Sclerosis/genetics , Multiple Sclerosis/pathology , Neurons/pathology , Transcription Factors/physiology , Adult , Aged , Aged, 80 and over , Blotting, Western , Cell Count , Down-Regulation , Female , Genetic Vectors , Gyrus Cinguli/pathology , Heat-Shock Proteins/biosynthesis , Heat-Shock Proteins/genetics , Humans , Immunohistochemistry , Lentivirus/genetics , Male , Middle Aged , Oxidation-Reduction , Oxidative Phosphorylation , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Pyramidal Cells/pathology , RNA, Small Interfering/genetics , Reactive Oxygen Species/metabolism , Real-Time Polymerase Chain Reaction , Tissue Banks , Transcription Factors/biosynthesis , Transcription Factors/genetics
8.
FASEB J ; 26(6): 2639-47, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22415301

ABSTRACT

In patients with glioblastomas, vascular endothelial growth factor (VEGF) is a key mediator of tumor-associated angiogenesis. Glioblastomas are notorious for their capacity to induce neovascularization, driving continued tumor growth. Here we report that miR-125b is down-regulated in glioblastoma-associated endothelial cells, resulting in increased expression of its target, myc-associated zinc finger protein (MAZ), a transcription factor that regulates VEGF. The down-regulation of miR-125b was also observed on exposure of endothelial cells to glioblastoma-conditioned medium or VEGF, resulting in increased MAZ expression. Further analysis revealed that inhibition of MAZ accumulation by miR-125b, or by MAZ-specific shRNAs, attenuated primary human brain endothelial cell migration and tubule formation in vitro, phenomena considered to mimick angiogenic processes in vitro. Moreover, MAZ expression was elevated in brain blood vessels of glioblastoma patients. Altogether these results demonstrate a functional feed-forward loop in glioblastoma-related angiogenesis, in which VEGF inhibits the expression of miR-125b, resulting in increased expression of MAZ, which in its turn causes transcriptional activation of VEGF. This loop is functionally impeded by the VEGF receptor inhibitor vandetanib, and our results may contribute to the further development of inhibitors of tumor-angiogenesis.


Subject(s)
DNA-Binding Proteins/genetics , Glioblastoma/blood supply , MicroRNAs/physiology , Neovascularization, Pathologic/pathology , Transcription Factors/genetics , Vascular Endothelial Growth Factor A/physiology , Coculture Techniques , DNA-Binding Proteins/biosynthesis , Down-Regulation , Endothelial Cells/metabolism , Glioblastoma/metabolism , HEK293 Cells , Humans , Transcription Factors/biosynthesis , Tumor Cells, Cultured
9.
J Neurochem ; 121(5): 730-7, 2012 Jun.
Article in English | MEDLINE | ID: mdl-21777246

ABSTRACT

Homeostasis of the brain is dependent on the blood-brain barrier (BBB). This barrier tightly regulates the exchange of essential nutrients and limits the free flow of immune cells into the CNS. Perturbations of BBB function and the loss of its immune quiescence are hallmarks of a variety of brain diseases, including multiple sclerosis (MS), vascular dementia, and stroke. In particular, diapedesis of monocytes and subsequent trafficking of monocyte-derived macrophages into the brain are key mediators of demyelination and axonal damage in MS. Endothelin-1 (ET-1) is considered as a potent pro-inflammatory peptide and has been implicated in the development of cardiovascular diseases. Here, we studied the role of different components of the endothelin system, i.e., ET-1, its type B receptor (ET(B)) and endothelin-converting enzyme-1 (ECE-1) in monocyte diapedesis of a human brain endothelial cell barrier. Our pharmacological inhibitory and specific gene knockdown studies point to a regulatory function of these proteins in transendothelial passage of monocytes. Results from this study suggest that the endothelin system is a putative target within the brain for anti-inflammatory treatment in neurological diseases.


Subject(s)
Blood-Brain Barrier/metabolism , Endothelial Cells/metabolism , Endothelins/metabolism , Monocytes/cytology , Transendothelial and Transepithelial Migration/physiology , Aspartic Acid Endopeptidases/metabolism , Blotting, Western , Cell Line , Endothelin-Converting Enzymes , Gene Knockdown Techniques , Humans , Immunohistochemistry , Metalloendopeptidases/metabolism , Receptors, Endothelin/metabolism
10.
J Infect Dis ; 204(6): 837-44, 2011 Sep 15.
Article in English | MEDLINE | ID: mdl-21849281

ABSTRACT

Accumulating evidence indicates that neutralizing antibodies play an important role in protection from chronic hepatitis C virus (HCV) infection. Efforts to elicit such responses by immunization with intact heterodimeric E1E2 envelope proteins have met with limited success. To determine whether antigenic sites, which are not exposed by the combined E1E2 heterodimer structure, are capable of eliciting neutralizing antibody responses, we expressed and purified each as separate recombinant proteins E1 and E2, from which the immunodominant hypervariable region (HVR-1) was deleted. Immunization of chimpanzees with either E1 or E2 alone induced antigen-specific T-helper cytokines of similar magnitude. Unexpectedly, the capacity to neutralize HCV was observed in E1 but not in animals immunized with E2 devoid of HVR-1. Furthermore, in vivo only E1-vaccinated animals exposed to the heterologous HCV-1b inoculum cleared HCV infection.


Subject(s)
Antibodies, Neutralizing/blood , Hepacivirus/immunology , Hepatitis C/therapy , Viral Envelope Proteins/immunology , Viral Hepatitis Vaccines/immunology , Animals , Disease Models, Animal , Genotype , Hepacivirus/genetics , Pan troglodytes , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Viral Envelope Proteins/genetics , Viral Hepatitis Vaccines/administration & dosage , Viral Hepatitis Vaccines/genetics
11.
Brain ; 134(Pt 2): 555-70, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21183485

ABSTRACT

Adenosine triphosphate-binding cassette efflux transporters are highly expressed at the blood-brain barrier and actively hinder passage of harmful compounds, thereby maintaining brain homoeostasis. Since, adenosine triphosphate-binding cassette transporters drive cellular exclusion of potential neurotoxic compounds or inflammatory molecules, alterations in their expression and function at the blood-brain barrier may contribute to the pathogenesis of neuroinflammatory disorders, such as multiple sclerosis. Therefore, we investigated the expression pattern of different adenosine triphosphate-binding cassette efflux transporters, including P-glycoprotein, multidrug resistance-associated proteins-1 and -2 and breast cancer resistance protein in various well-characterized human multiple sclerosis lesions. Cerebrovascular expression of P-glycoprotein was decreased in both active and chronic inactive multiple sclerosis lesions. Interestingly, foamy macrophages in active multiple sclerosis lesions showed enhanced expression of multidrug resistance-associated protein-1 and breast cancer resistance protein, which coincided with their increased function of cultured foamy macrophages. Strikingly, reactive astrocytes display an increased expression of P-glycoprotein and multidrug resistance-associated protein-1 in both active and inactive multiple sclerosis lesions, which correlated with their enhanced in vitro activity on astrocytes derived from multiple sclerosis lesions. To investigate whether adenosine triphosphate-binding cassette transporters on reactive astrocytes can contribute to the inflammatory process, primary cultures of reactive human astrocytes were generated through activation of Toll-like receptor-3 to mimic the astrocytic phenotype as observed in multiple sclerosis lesions. Notably, blocking adenosine triphosphate-binding cassette transporter activity on reactive astrocytes inhibited immune cell migration across a blood-brain barrier model in vitro, which was due to the reduction of astrocytic release of the chemokine (C-C motif) ligand 2. Our data point towards a novel (patho)physiological role for adenosine triphosphate-binding cassette transporters, suggesting that limiting their activity by dampening astrocyte activation may open therapeutic avenues to diminish tissue damage during multiple sclerosis pathogenesis.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Astrocytes/metabolism , Blood-Brain Barrier/metabolism , Chemokine CCL2/metabolism , Multiple Sclerosis/metabolism , ATP-Binding Cassette Transporters/antagonists & inhibitors , Adult , Aged , Aged, 80 and over , Blood-Brain Barrier/physiology , Brain/metabolism , Brain/physiopathology , Cell Culture Techniques , Cell Movement/physiology , Female , Humans , Macrophages/metabolism , Male , Middle Aged , Monocytes/metabolism , Monocytes/physiology , Multiple Sclerosis/physiopathology
12.
Free Radic Biol Med ; 49(8): 1283-9, 2010 Nov 01.
Article in English | MEDLINE | ID: mdl-20673799

ABSTRACT

Oxidative stress plays a major role in multiple sclerosis (MS), a chronic inflammatory central nervous system (CNS) disease. Invading leukocytes contribute to cell damage and demyelination by producing excessive amounts of cytotoxic mediators, including reactive oxygen species (ROS). To counteract the damaging effects of ROS the CNS is endowed with a repertoire of endogenous antioxidant enzymes, which are regulated by the transcription factor NF-E2-related factor 2 (Nrf2). Upon exposure to ROS, Nrf2 translocates to the nucleus allowing transcriptional activation of various antioxidant enzymes. DJ1 is a protein that is involved in the stabilization of Nrf2 and hence acts as a positive regulator of Nrf2-driven antioxidant protection. Here, we investigate the (sub)cellular localization of Nrf2 and DJ1 in various MS lesion stages and show that Nrf2 is strikingly upregulated in active MS lesions, in both the nucleus and the cytoplasm of infiltrating macrophages and to a lesser extent in reactive astrocytes. Simultaneously, DJ1 protein expression is predominantly increased in astrocytes in both active and chronic inactive MS lesions compared to control brain tissue and normal-appearing white matter. Together, our findings suggest that persistent Nrf2-mediated transcription occurs in active MS lesions, but that this endogenous response is insufficient to prevent ROS-induced cellular damage, which is abundant in inflammatory MS lesions.


Subject(s)
Astrocytes/metabolism , Brain/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Macrophages/metabolism , Multiple Sclerosis/metabolism , NF-E2-Related Factor 2/metabolism , Oncogene Proteins/metabolism , Aged , Aged, 80 and over , Antioxidants/metabolism , Astrocytes/immunology , Astrocytes/pathology , Autopsy , Brain/immunology , Brain/pathology , Disease Progression , Encephalitis , Female , Humans , Immunohistochemistry , Intracellular Signaling Peptides and Proteins/genetics , Macrophages/immunology , Macrophages/pathology , Male , Middle Aged , Multiple Sclerosis/genetics , Multiple Sclerosis/immunology , Multiple Sclerosis/pathology , Multiple Sclerosis/physiopathology , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/immunology , Oncogene Proteins/genetics , Oxidative Stress , Protein Deglycase DJ-1 , Protein Transport , Up-Regulation/immunology
13.
Hepatology ; 45(3): 602-13, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17326154

ABSTRACT

UNLABELLED: Broad T cell and B cell responses to multiple HCV antigens are observed early in individuals who control or clear HCV infection. The prevailing hypothesis has been that similar immune responses induced by prophylactic immunization would reduce acute virus replication and protect exposed individuals from chronic infection. Here, we demonstrate that immunization of naïve chimpanzees with a multicomponent HCV vaccine induced robust HCV-specific immune responses, and that all vaccinees exposed to heterologous chimpanzee-adapted HCV 1b J4 significantly reduced viral RNA in serum by 84%, and in liver by 99% as compared to controls (P=0.024 and 0.028, respectively). However, despite control of HCV in plasma and liver in the acute period, in the chronic phase, 3 of 4 vaccinated animals developed persistent infection. Analysis of expression levels of proinflammatory cytokines in serial hepatic biopsies failed to reveal an association with vaccine outcome. However, expression of IDO, CTLA-4 [corrected] and PD-1 levels in liver correlated with clearance or chronicity. CONCLUSION: Despite early control of virus load, a virus-associated tolerogenic-like state can develop in certain individuals independent of vaccination history.


Subject(s)
Antigens, CD/metabolism , Hepatitis C/immunology , Viral Hepatitis Vaccines/therapeutic use , Animals , Antigens, Viral/immunology , Apoptosis Regulatory Proteins/metabolism , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Chronic Disease/prevention & control , Cytokines/metabolism , DNA, Viral/genetics , Hepacivirus/genetics , Hepacivirus/immunology , Hepatitis C/prevention & control , Pan troglodytes , Programmed Cell Death 1 Receptor , Viral Load
14.
J Infect Dis ; 192(5): 920-9, 2005 Sep 01.
Article in English | MEDLINE | ID: mdl-16088843

ABSTRACT

BACKGROUND: Preventive and therapeutic vaccine strategies aimed at controlling hepatitis C virus (HCV) infection should mimic the immune responses observed in patients who control or clear HCV, specifically T helper (Th) type 1 and CD8+ cell responses to multiple antigens, including nonstructural protein (NS) 3. Given the experience with human immunodeficiency virus, the best candidates for this are based on DNA prime, pox, or adenovirus boost regimens. METHODS: In rhesus macaques, we compared NS3-expressing DNA prime and adenovirus boost strategy with 2 alternative priming approaches aimed at modifying Th1 and CD8+ responses: DNA adjuvanted with interleukin (IL)-2- and -12-encoding plasmids or Semliki Forest virus (SFV). RESULTS: All prime-boost regimens elicited NS3-specific B and T cell responses in rhesus macaques, including CD8+ responses. SFV priming induced higher lymphoproliferation and longer Th1 memory responses. The use of IL-2- and IL-12-expressing vectors resulted in reduced Th2 and antibody responses, which led to increased Th1 skewing but not to an increase in the magnitude of the IFN- gamma and CD8+ responses. CONCLUSIONS: All strategies induced Th1 cellular responses to HCV NS3, with fine modulations depending on the different priming approaches. When they are developed for more HCV antigens, these strategies could be beneficial in therapeutic vaccine approaches.


Subject(s)
Hepacivirus/immunology , Hepatitis C/immunology , Immunization/methods , Viral Hepatitis Vaccines/immunology , Viral Nonstructural Proteins/immunology , Adenoviridae/immunology , Animals , Antibodies, Viral/blood , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Cell Proliferation , Genetic Vectors/immunology , Hepatitis C/genetics , Hepatitis C/prevention & control , Interferon-gamma/immunology , Interleukin-2/immunology , Interleukin-4/immunology , Macaca mulatta , Peptide Fragments/immunology , Viral Nonstructural Proteins/genetics
15.
Hepatology ; 38(4): 851-8, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14512872

ABSTRACT

Chronic hepatitis C virus (HCV) infection in humans is associated with an impairment of dendritic cells (DC). It has been hypothesized that impairment of DC function may be a central mechanism facilitating the establishment of a chronic carrier state. However, the majority of patients studied with DC impairment to date have been identified and, thus, inadvertently selected because of clinical manifestations leading to their diagnosis, which may have been many years following actual infection. We set out to determine whether impaired DC function occurred in the earlier asymptomatic phase of infection and turned to a well-defined cohort of HCV-infected chimpanzees in which the specific date of infection and the nature of the inoculum were well characterized. Results revealed that, in contrast to the observations in human subjects with advanced clinical hepatitis, there was neither impairment of the allostimulatory capacity of monocyte-derived DC from HCV chronic carriers nor impairment of the maturation process. Decreased allostimulatory capacity was only detected in 2 animals and, interestingly, in those that possessed the highest viral loads. Nevertheless, HCV sequences were undetectable in any of the DC derived from HCV-infected chimpanzees. In conclusion, these findings suggest that the mechanisms of establishing persistent HCV infection are separate and independent from those responsible for impaired DC function. Indeed, the maturation and allostimulatory impairment, as described in patient studies, are not necessary prerequisites but rather possible consequences of persistent and active HCV infection associated with disease progression.


Subject(s)
Ape Diseases/immunology , Dendritic Cells/physiology , Hepatitis C, Chronic/veterinary , Pan troglodytes/virology , Animals , Cells, Cultured , Dendritic Cells/virology , Endocytosis , Female , Hepacivirus/isolation & purification , Hepatitis C, Chronic/immunology , Hepatitis C, Chronic/virology , Humans , Immunophenotyping , Male
SELECTION OF CITATIONS
SEARCH DETAIL
...