Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Front Immunol ; 13: 977525, 2022.
Article in English | MEDLINE | ID: mdl-36275731

ABSTRACT

Vaccination, as a public health measure, offers effective protection of populations against infectious diseases. Optimising vaccination efficacy, particularly for higher-risk individuals, like the elderly whose immunocompromised state can prevent the development of robust vaccine responses, is vital. It is now clear that 24-hour circadian rhythms, which govern virtually all aspects of physiology, can generate oscillations in immunological responses. Consequently, vaccine efficacy may depend critically on the time of day of administration(s), including for Covid-19, current vaccines, and any future diseases or pandemics. Published clinical vaccine trials exploring diurnal immune variations suggest this approach could represent a powerful adjunct strategy for optimising immunisation, but important questions remain to be addressed. This review explores the latest insights into diurnal immune variation and the outcomes of circadian timing of vaccination or 'chronovaccination'.


Subject(s)
COVID-19 , Vaccines , Humans , Aged , Circadian Rhythm , COVID-19/prevention & control , Vaccination
2.
Sci Rep ; 7(1): 16057, 2017 11 22.
Article in English | MEDLINE | ID: mdl-29167574

ABSTRACT

Two crystal structures of Japanin, an 18 kDa immune-modulatory lipocalin from the Brown Ear Tick (Rhipicephalus appendiculatus), have been determined at 2.2 and 2.4 Å resolution. In both crystal forms the protein is in complex with cholesterol, which sits in a closed pocket at the centre of the lipocalin barrel. Both crystal forms are dimers, which are also observed in solution. Molecular modelling suggests that previously-described members of a tick protein family bearing high sequence homology to Japanin are also likely to bind cholesterol or cholesterol derivatives.


Subject(s)
Cholesterol/metabolism , Dendritic Cells/metabolism , Rhipicephalus/metabolism , Amino Acid Sequence , Animals , Binding Sites , Crystallography, X-Ray , Dimerization , Lipocalins/chemistry , Lipocalins/metabolism , Protein Structure, Secondary , Structural Homology, Protein
3.
Oncotarget ; 8(40): 67439-67456, 2017 Sep 15.
Article in English | MEDLINE | ID: mdl-28978044

ABSTRACT

Immunotherapy for metastatic melanoma offers great promise but, to date, only a subset of patients have responded. There is an urgent need to identify ways of allocating patients to the most beneficial therapy, to increase survival and decrease therapy-associated morbidity and costs. Blood-based biomarkers are of particular interest because of their straightforward implementation in routine clinical care. We sought to identify markers for dendritic cell (DC) vaccine-based immunotherapy against metastatic melanoma through gene expression analysis of peripheral blood mononuclear cells. A large-scale microarray analysis of 74 samples from two treatment centers, taken directly after the first round of DC vaccination, was performed. We found that phosphatidylethanolamine binding protein 1 (PEBP1)/Raf Kinase inhibitory protein (RKIP) expression can be used to identify a significant proportion of patients who performed poorly after DC vaccination. This result was validated by q-PCR analysis on blood samples from a second cohort of 95 patients treated with DC vaccination in four different centers. We conclude that low PEBP1 expression correlates with poor overall survival after DC vaccination. Intriguingly, this was only the case for expression of PEBP1 after, but not prior to, DC vaccination. Moreover, the change in PEBP1 expression upon vaccination correlated well with survival. Further analyses revealed that PEBP1 expression positively correlated with genes involved in T cell responses but inversely correlated with genes associated with myeloid cells and aberrant inflammation including STAT3, NOTCH1, and MAPK1. Concordantly, PEBP1 inversely correlated with the myeloid/lymphoid-ratio and was suppressed in patients suffering from chronic inflammatory disease.

4.
Microbiol Spectr ; 4(6)2016 12.
Article in English | MEDLINE | ID: mdl-28087939

ABSTRACT

The aim of this review is to provide a coherent framework for understanding dendritic cells (DCs). It has seven sections. The introduction provides an overview of the immune system and essential concepts, particularly for the nonspecialist reader. Next, the "History" section outlines the early evolution of ideas about DCs and highlights some sources of confusion that still exist today. The "Lineages" section then focuses on five different populations of DCs: two subsets of "classical" DCs, plasmacytoid DCs, monocyte-derived DCs, and Langerhans cells. It highlights some cellular and molecular specializations of each, and also notes other DC subsets that have been proposed. The following "Tissues" section discusses the distribution and behavior of different DC subsets within nonlymphoid and secondary lymphoid tissues that are connected by DC migration pathways between them. In the "Tolerance" section, the role of DCs in central and peripheral tolerance is considered, including their ability to drive the differentiation of different populations of regulatory T cells. In contrast, the "Immunity" section considers the roles of DCs in sensing of infection and tissue damage, the initiation of primary responses, the T-cell effector phase, and the induction of immunological memory. The concluding section provides some speculative ideas about the evolution of DCs. It also revisits earlier concepts of generation of diversity and clonal selection in terms of DCs driving the evolution of T-cell responses. Throughout, this review highlights certain areas of uncertainty and suggests some avenues for future investigation.


Subject(s)
Adaptive Immunity , Dendritic Cells/physiology , Immunity, Innate , Animals , Humans
5.
Oncoimmunology ; 4(8): e1019197, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26405571

ABSTRACT

Autologous dendritic cell (DC) therapy is an experimental cellular immunotherapy that is safe and immunogenic in patients with advanced melanoma. In an attempt to further improve the therapeutic responses, we treated 15 patients with melanoma, with autologous monocyte-derived immature DC electroporated with mRNA encoding CD40 ligand (CD40L), CD70 and a constitutively active TLR4 (caTLR4) together with mRNA encoding a tumor-associated antigen (TAA; respectively gp100 or tyrosinase). In addition, DC were pulsed with keyhole limpet hemocyanin (KLH) that served as a control antigen. Production of this DC vaccine with high cellular viability, high expression of co-stimulatory molecules and MHC class I and II and production of IL-12p70, was feasible in all patients. A vaccination cycle consisting of three vaccinations with up to 15×106 DC per vaccination at a biweekly interval, was repeated after 6 and 12 months in the absence of disease progression. mRNA-optimized DC were injected intranodally, because of low CCR7 expression on the DC, and induced de novo immune responses against control antigen. T cell responses against tyrosinase were detected in the skin-test infiltrating lymphocytes (SKIL) of two patients. One mixed tumor response and two durable tumor stabilizations were observed among 8 patients with evaluable disease at baseline. In conclusion, autologous mRNA-optimized DC can be safely administered intranodally to patients with metastatic melanoma but showed limited immunological responses against tyrosinase and gp100.

6.
J Exp Med ; 211(6): 1019-25, 2014 Jun 02.
Article in English | MEDLINE | ID: mdl-24799501

ABSTRACT

There is currently no paradigm in immunology that enables an accurate prediction of how the immune system will respond to any given agent. Here we show that the immunological responses induced by members of a broad class of inorganic crystalline materials are controlled purely by their physicochemical properties in a highly predictable manner. We show that structurally and chemically homogeneous layered double hydroxides (LDHs) can elicit diverse human dendritic cell responses in vitro. Using a systems vaccinology approach, we find that every measured response can be modeled using a subset of just three physical and chemical properties for all compounds tested. This correlation can be reduced to a simple linear equation that enables the immunological responses stimulated by newly synthesized LDHs to be predicted in advance from these three parameters alone. We also show that mouse antigen-specific antibody responses in vivo and human macrophage responses in vitro are controlled by the same properties, suggesting they may control diverse responses at both individual component and global levels of immunity. This study demonstrates that immunity can be determined purely by chemistry and opens the possibility of rational manipulation of immunity for therapeutic purposes.


Subject(s)
Antibody Formation/immunology , Dendritic Cells/immunology , Hydroxides/immunology , Macrophages/immunology , Animals , Antibodies/blood , Antibodies/immunology , Cells, Cultured , Crystallization , Cytokines/immunology , Cytokines/metabolism , Dendritic Cells/metabolism , Enzyme-Linked Immunosorbent Assay , Humans , Hydroxides/chemistry , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Microscopy, Electron, Transmission , Multivariate Analysis , Ovalbumin/immunology
7.
PLoS Pathog ; 9(6): e1003450, 2013.
Article in English | MEDLINE | ID: mdl-23825947

ABSTRACT

Hard ticks subvert the immune responses of their vertebrate hosts in order to feed for much longer periods than other blood-feeding ectoparasites; this may be one reason why they transmit perhaps the greatest diversity of pathogens of any arthropod vector. Tick-induced immunomodulation is mediated by salivary components, some of which neutralise elements of innate immunity or inhibit the development of adaptive immunity. As dendritic cells (DC) trigger and help to regulate adaptive immunity, they are an ideal target for immunomodulation. However, previously described immunoactive components of tick saliva are either highly promiscuous in their cellular and molecular targets or have limited effects on DC. Here we address the question of whether the largest and globally most important group of ticks (the ixodid metastriates) produce salivary molecules that specifically modulate DC activity. We used chromatography to isolate a salivary gland protein (Japanin) from Rhipicephalus appendiculatus ticks. Japanin was cloned, and recombinant protein was produced in a baculoviral expression system. We found that Japanin specifically reprogrammes DC responses to a wide variety of stimuli in vitro, radically altering their expression of co-stimulatory and co-inhibitory transmembrane molecules (measured by flow cytometry) and their secretion of pro-inflammatory, anti-inflammatory and T cell polarising cytokines (assessed by Luminex multiplex assays); it also inhibits the differentiation of DC from monocytes. Sequence alignments and enzymatic deglycosylation revealed Japanin to be a 17.7 kDa, N-glycosylated lipocalin. Using molecular cloning and database searches, we have identified a group of homologous proteins in R. appendiculatus and related species, three of which we have expressed and shown to possess DC-modulatory activity. All data were obtained using DC generated from at least four human blood donors, with rigorous statistical analysis. Our results suggest a previously unknown mechanism for parasite-induced subversion of adaptive immunity, one which may also facilitate pathogen transmission.


Subject(s)
Arthropod Proteins/immunology , Dendritic Cells/immunology , Immunologic Factors/immunology , Monocytes/immunology , Rhipicephalus/immunology , Salivary Proteins and Peptides/immunology , Adaptive Immunity/drug effects , Adaptive Immunity/genetics , Adaptive Immunity/immunology , Animals , Arthropod Proteins/genetics , Arthropod Proteins/pharmacology , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Line , Cytokines/genetics , Cytokines/immunology , Dendritic Cells/pathology , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Gene Expression Regulation/immunology , Humans , Immunologic Factors/genetics , Immunologic Factors/pharmacology , Monocytes/pathology , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/pharmacology , Rhipicephalus/genetics , Salivary Proteins and Peptides/genetics , Salivary Proteins and Peptides/pharmacology
8.
PLoS Pathog ; 9(1): e1003100, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23382671

ABSTRACT

During disease progression to AIDS, HIV-1 infected individuals become increasingly immunosuppressed and susceptible to opportunistic infections. It has also been demonstrated that multiple subsets of dendritic cells (DC), including DC-SIGN⁺ cells, become significantly depleted in the blood and lymphoid tissues of AIDS patients, which may contribute to the failure in initiating effective host immune responses. The mechanism for DC depletion, however, is unclear. It is also known that vast quantities of viral envelope protein gp120 are shed from maturing HIV-1 virions and form circulating immune complexes in the serum of HIV-1-infected individuals, but the pathological role of gp120 in HIV-1 pathogenesis remains elusive. Here we describe a previously unrecognized mechanism of DC death in chronic HIV-1 infection, in which ligation of DC-SIGN by gp120 sensitizes DC to undergo accelerated apoptosis in response to a variety of activation stimuli. The cultured monocyte-derived DC and also freshly-isolated DC-SIGN⁺ blood DC that were exposed to either cross-linked recombinant gp120 or immune-complex gp120 in HIV⁺ serum underwent considerable apoptosis after CD40 ligation or exposure to bacterial lipopolysaccharide (LPS) or pro-inflammatory cytokines such as TNFα and IL-1ß. Furthermore, circulating DC-SIGN⁺ DC that were isolated directly from HIV-1⁺ individuals had actually been pre-sensitized by serum gp120 for activation-induced exorbitant apoptosis. In all cases the DC apoptosis was substantially inhibited by DC-SIGN blockade. Finally, we showed that accelerated DC apoptosis was a direct consequence of excessive activation of the pro-apoptotic molecule ASK-1 and transfection of siRNA against ASK-1 significantly prevented the activation-induced excessive DC death. Our study discloses a previously unknown mechanism of immune modulation by envelope protein gp120, provides new insights into HIV immunopathogenesis, and suggests potential therapeutic approaches to prevent DC depletion in chronic HIV infection.


Subject(s)
Apoptosis/physiology , Cell Adhesion Molecules/metabolism , Dendritic Cells/metabolism , HIV Envelope Protein gp120/metabolism , Lectins, C-Type/metabolism , MAP Kinase Kinase Kinase 5/metabolism , Receptors, Cell Surface/metabolism , Apoptosis/drug effects , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD40 Antigens/immunology , Cell Adhesion Molecules/immunology , Cells, Cultured , Dendritic Cells/immunology , Dendritic Cells/pathology , Gene Silencing , HIV Envelope Protein gp120/immunology , HIV Infections/blood , HIV Infections/immunology , Host-Pathogen Interactions , Humans , Lectins, C-Type/immunology , Lipopolysaccharides/pharmacology , MAP Kinase Kinase Kinase 5/immunology , Protein Binding , RNA, Small Interfering/genetics , Receptors, Cell Surface/immunology , Transfection
9.
Brief Bioinform ; 12(6): 562-75, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21969471

ABSTRACT

Biomedical research relies increasingly on large collections of data sets and knowledge whose generation, representation and analysis often require large collaborative and interdisciplinary efforts. This dimension of 'big data' research calls for the development of computational tools to manage such a vast amount of data, as well as tools that can improve communication and access to information from collaborating researchers and from the wider community. Whenever research projects have a defined temporal scope, an additional issue of data management arises, namely how the knowledge generated within the project can be made available beyond its boundaries and life-time. DC-THERA is a European 'Network of Excellence' (NoE) that spawned a very large collaborative and interdisciplinary research community, focusing on the development of novel immunotherapies derived from fundamental research in dendritic cell immunobiology. In this article we introduce the DC-THERA Directory, which is an information system designed to support knowledge management for this research community and beyond. We present how the use of metadata and Semantic Web technologies can effectively help to organize the knowledge generated by modern collaborative research, how these technologies can enable effective data management solutions during and beyond the project lifecycle, and how resources such as the DC-THERA Directory fit into the larger context of e-science.


Subject(s)
Information Dissemination/methods , Information Storage and Retrieval/methods , Semantics , Translational Research, Biomedical , Database Management Systems , Internet
10.
Immunome Res ; 6: 10, 2010 Nov 19.
Article in English | MEDLINE | ID: mdl-21092113

ABSTRACT

BACKGROUND: The advent of Systems Biology has been accompanied by the blooming of pathway databases. Currently pathways are defined generically with respect to the organ or cell type where a reaction takes place. The cell type specificity of the reactions is the foundation of immunological research, and capturing this specificity is of paramount importance when using pathway-based analyses to decipher complex immunological datasets. Here, we present DC-ATLAS, a novel and versatile resource for the interpretation of high-throughput data generated perturbing the signaling network of dendritic cells (DCs). RESULTS: Pathways are annotated using a novel data model, the Biological Connection Markup Language (BCML), a SBGN-compliant data format developed to store the large amount of information collected. The application of DC-ATLAS to pathway-based analysis of the transcriptional program of DCs stimulated with agonists of the toll-like receptor family allows an integrated description of the flow of information from the cellular sensors to the functional outcome, capturing the temporal series of activation events by grouping sets of reactions that occur at different time points in well-defined functional modules. CONCLUSIONS: The initiative significantly improves our understanding of DC biology and regulatory networks. Developing a systems biology approach for immune system holds the promise of translating knowledge on the immune system into more successful immunotherapy strategies.

11.
Blood ; 116(16): e74-80, 2010 Oct 21.
Article in English | MEDLINE | ID: mdl-20628149

ABSTRACT

Monocytes and cells of the dendritic cell lineage circulate in blood and eventually migrate into tissue where they further mature and serve various functions, most notably in immune defense. Over recent years these cells have been characterized in detail with the use of cell surface markers and flow cytometry, and subpopulations have been described. The present document proposes a nomenclature for these cells and defines 3 types of monocytes (classical, intermediate, and nonclassical monocytes) and 3 types of dendritic cells (plasmacytoid and 2 types of myeloid dendritic cells) in human and in mouse blood. This classification has been approved by the Nomenclature Committee of the International Union of Immunological Societies, and we are convinced that it will facilitate communication among experts and in the wider scientific community.


Subject(s)
Blood Cells/classification , Dendritic Cells/classification , Monocytes/classification , Terminology as Topic , Animals , Humans , Mice
12.
Blood ; 112(9): 3723-34, 2008 Nov 01.
Article in English | MEDLINE | ID: mdl-18694997

ABSTRACT

Dendritic cells (DCs) are professional antigen-presenting cells (APCs) that patrol tissues to sense danger signals and activate specific immune responses. In addition, they also play a role in inflammation and tissue repair. Here, we show that oxygen availability is necessary to promote full monocyte-derived DC differentiation and maturation. Low oxygen tension (hypoxia) inhibits expression of several differentiation and maturation markers (CD1a, CD40, CD80, CD83, CD86, and MHC class II molecules) in response to lipopolysaccharide (LPS), as well as their stimulatory capacity for T-cell functions. These events are paralleled by impaired up-regulation of the chemokine receptor CCR7, an otherwise necessary event for the homing of mature DCs to lymph nodes. In contrast, hypoxia strongly up-regulates production of proinflammatory cytokines, particularly TNFalpha and IL-1beta, as well as the inflammatory chemokine receptor CCR5. Subcutaneous injection of hypoxic DCs into the footpads of mice results in defective DC homing to draining lymph nodes, but enhanced leukocyte recruitment at the site of injection. Thus, hypoxia uncouples the promotion of inflammatory and tissue repair from sentinel functions in DCs, which we suggest is a safeguard mechanism against immune reactivity to damaged tissues.


Subject(s)
Cell Hypoxia/immunology , Cell Hypoxia/physiology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Animals , Cell Differentiation , Cells, Cultured , Chemokines/metabolism , Chemotaxis , Cytokines/metabolism , Dendritic Cells/cytology , Dendritic Cells/drug effects , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Ligands , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred C57BL , Monocytes/cytology , Monocytes/immunology , Monocytes/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Toll-Like Receptors/agonists , Toll-Like Receptors/genetics , Toll-Like Receptors/metabolism
13.
Blood ; 111(6): 3090-6, 2008 Mar 15.
Article in English | MEDLINE | ID: mdl-18094328

ABSTRACT

Dendritic cells (DCs) link innate and adaptive immunity, initiating and regulating effector cell responses. They ubiquitously express members of the LILR (ILT, LIR, CD85) family of molecules, some of which recognize self-HLA molecules, but little is known of their possible functions in DC biology. We demonstrate that the inhibitory receptor LILRB1 (ILT2, LIR1, CD85j) is selectively up-regulated during DC differentiation from monocyte precursors in culture. Continuous ligation of LILRB1 modulated cellular differentiation, conferred a unique phenotype upon the resultant cells, induced a profound resistance to CD95-mediated cell death, and inhibited secretion of cytokines IL-10, IL-12p70, and TGF-beta. These features remained stable even after exposure of the cells to bacterial LPS. Ligated DCs exhibited poor stimulatory activity for primary and memory T-cell proliferative responses, but this was substantially reversed by blockade of CD80 or its preferred ligand CTLA-4, or by depleting CD4(+) CD25(+) CD127(lo) regulatory T cells. Our findings suggest that ligation of LILRB1 on DCs by self-HLA molecules may play a key role in controlling the balance between the induction and suppression of adaptive immune responses.


Subject(s)
Antigens, CD/metabolism , Cell Differentiation/drug effects , Cell Differentiation/immunology , Dendritic Cells/cytology , Dendritic Cells/metabolism , Receptors, Immunologic/antagonists & inhibitors , Receptors, Immunologic/metabolism , Cells, Cultured , Cytokines/metabolism , Dendritic Cells/drug effects , Dendritic Cells/immunology , Humans , Immunologic Memory/immunology , Leukocyte Immunoglobulin-like Receptor B1 , Lipopolysaccharides/pharmacology , Phenotype , Phosphotyrosine/metabolism , fas Receptor/immunology
14.
J Invest Dermatol ; 126(7): 1541-8, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16645596

ABSTRACT

Langerhans cells (LCs) can be targeted with DNA-coated gold micro-projectiles ("Gene Gun") to induce potent cellular and humoral immune responses. It is likely that the relative volumetric distribution of LCs and keratinocytes within the epidermis impacts on the efficacy of Gene Gun immunization protocols. This study quantified the three-dimensional (3D) distribution of LCs and keratinocytes in the mouse skin model with a near-infrared multiphoton laser-scanning microscope (NIR-MPLSM). Stratum corneum (SC) and viable epidermal thickness measured with MPLSM was found in close agreement with conventional histology. LCs were located in the vertical plane at a mean depth of 14.9 microm, less than 3 mum above the dermo-epidermal boundary and with a normal histogram distribution. This likely corresponds to the fact that LCs reside in the suprabasal layer (stratum germinativum). The nuclear volume of keratinocytes was found to be approximately 1.4 times larger than that of resident LCs (88.6 microm3). Importantly, the ratio of LCs to keratinocytes in mouse ear skin (1:15) is more than three times higher than that reported for human breast skin (1:53). Accordingly, cross-presentation may be more significant in clinical Gene Gun applications than in pre-clinical mouse studies. These interspecies differences should be considered in pre-clinical trials using mouse models.


Subject(s)
Immunity, Cellular , Keratinocytes/immunology , Langerhans Cells/immunology , Models, Animal , Skin/immunology , Vaccination/methods , Animals , Biolistics/methods , Breast/cytology , Epidermal Cells , Humans , Imaging, Three-Dimensional , Immunity, Cellular/immunology , Immunohistochemistry , Keratinocytes/cytology , Langerhans Cells/cytology , Mice , Mice, Inbred BALB C , Microscopy, Fluorescence, Multiphoton , Skin/cytology
15.
Nat Genet ; 38(1): 38-46, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16369534

ABSTRACT

Severe acute respiratory syndrome (SARS) is caused by infection of a previously undescribed coronavirus (CoV). L-SIGN, encoded by CLEC4M (also known as CD209L), is a SARS-CoV binding receptor that has polymorphism in its extracellular neck region encoded by the tandem repeat domain in exon 4. Our genetic risk association study shows that individuals homozygous for CLEC4M tandem repeats are less susceptible to SARS infection. L-SIGN is expressed in both non-SARS and SARS-CoV-infected lung. Compared with cells heterozygous for L-SIGN, cells homozygous for L-SIGN show higher binding capacity for SARS-CoV, higher proteasome-dependent viral degradation and a lower capacity for trans infection. Thus, homozygosity for L-SIGN plays a protective role during SARS infection.


Subject(s)
Cell Adhesion Molecules/genetics , Lectins, C-Type/genetics , Receptors, Cell Surface/genetics , Severe Acute Respiratory Syndrome/genetics , Severe acute respiratory syndrome-related coronavirus/pathogenicity , Animals , CHO Cells/virology , Cell Adhesion Molecules/metabolism , Chlorocebus aethiops , Cohort Studies , Cricetinae , Cricetulus , Genetic Predisposition to Disease , Homozygote , Hong Kong/epidemiology , Humans , Intestine, Small/physiology , Lectins, C-Type/metabolism , Lung/physiology , Lung/virology , Molecular Sequence Data , Proteasome Endopeptidase Complex/metabolism , Receptors, Cell Surface/metabolism , Severe acute respiratory syndrome-related coronavirus/metabolism , Severe Acute Respiratory Syndrome/epidemiology , Tandem Repeat Sequences , Vero Cells/virology
16.
Eur J Immunol ; 33(9): 2520-30, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12938228

ABSTRACT

Indinavir (IDV) is a protease inhibitor that successfully suppresses HIV-1 replication as part of anti-retroviral therapy. There is evidence to suggest that IDV may also act non-specifically upon host proteases. In this study we investigated whether IDV could modulate protease-dependent molecules involved in dendritic cell (DC) migration - a pivotal process in immunoregulation. Human monocyte-derived DC were exposed to IDV (IDV-DC) and transendothelial migration (TEM) to inflammatory chemokines was determined. TEM of IDV-DC was significantly impaired compared to non-treated DC (p<0.01). Phenotypic analysis revealed that IDV-DC had reduced DC-SIGN expression, correlating with reduced adhesion to immobilized ICAM-2. Nevertheless, the reduction in migration following exposure to IDV could not be fully attributable to DC-SIGN interactions alone. Investigation of IDV-DC interactions with the underlying matrix protein, fibronectin, demonstrated that IDV significantly impaired DC binding to immobilized fibronectin (p<0.01). IDV appeared to act upon VLA-4 and VLA-5 since addition of antagonist monoclonal antibodies (mAb) similarly reduced adhesion of non-treated DC to fibronectin. Combined blockade of DC using anti-VLA-4, VLA-5 and anti-DC-SIGN mAb inhibited TEM to a similar extent as IDV. Our results strongly suggest that IDV inhibits host proteases necessary for DC migration and may, therefore, affect DC immunoregulation in HIV-1-infected patients.


Subject(s)
Cell Movement/drug effects , Dendritic Cells/drug effects , HIV Protease Inhibitors/pharmacology , Indinavir/pharmacology , Antigens, CD/metabolism , Cell Adhesion Molecules/metabolism , Dendritic Cells/metabolism , Endothelium/drug effects , HIV Infections/metabolism , HIV-1/metabolism , Humans , Immunoglobulins/biosynthesis , Immunoglobulins/genetics , Integrin alpha4beta1/metabolism , Integrin alpha5beta1/metabolism , Lectins, C-Type/metabolism , Membrane Glycoproteins/biosynthesis , Membrane Glycoproteins/genetics , Receptors, Cell Surface/metabolism , CD83 Antigen
SELECTION OF CITATIONS
SEARCH DETAIL
...