Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
J Invest Dermatol ; 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38570030

ABSTRACT

BACE1 is well-known for its role in the development of Alzheimer's disease. Recent publications, including our own, have demonstrated a role for this enzyme in other chronic diseases. The aim of this study was to investigate the role of BACE1 in the autoimmune disease systemic sclerosis (SSc). BACE1 protein levels were elevated in the skin of patients with SSc. Inhibition of BACE1 with small-molecule inhibitors or small interfering RNA blocked SSc and fibrotic stimuli-mediated fibroblast activation. Furthermore, we show that BACE1 regulation of dermal fibroblast activation is dependent on ß-catenin and Notch signaling. The neurotropic factor brain-derived neurotrophic factor negatively regulates BACE1 expression and activity in dermal fibroblasts. Finally, sera from patients with SSc show higher ß-amyloid and lower brain-derived neurotrophic factor levels than healthy controls. The ability of BACE1 to regulate SSc fibroblast activation reveals a therapeutic target in SSc. Several BACE1 inhibitors have been shown to be safe in clinical trials for Alzheimer's disease and could be repurposed to ameliorate fibrosis progression.

2.
3.
Proc Natl Acad Sci U S A ; 119(24): e2114309119, 2022 06 14.
Article in English | MEDLINE | ID: mdl-35675424

ABSTRACT

Viruses transmitted by Aedes mosquitoes are an increasingly important global cause of disease. Defining common determinants of host susceptibility to this large group of heterogenous pathogens is key for informing the rational design of panviral medicines. Infection of the vertebrate host with these viruses is enhanced by mosquito saliva, a complex mixture of salivary-gland-derived factors and microbiota. We show that the enhancement of infection by saliva was dependent on vascular function and was independent of most antisaliva immune responses, including salivary microbiota. Instead, the Aedes gene product sialokinin mediated the enhancement of virus infection through a rapid reduction in endothelial barrier integrity. Sialokinin is unique within the insect world as having a vertebrate-like tachykinin sequence and is absent from Anopheles mosquitoes, which are incompetent for most arthropod-borne viruses, whose saliva was not proviral and did not induce similar vascular permeability. Therapeutic strategies targeting sialokinin have the potential to limit disease severity following infection with Aedes-mosquito-borne viruses.


Subject(s)
Aedes , Arbovirus Infections , Arboviruses , Saliva , Tachykinins , Virus Diseases , Aedes/genetics , Aedes/virology , Animals , Arbovirus Infections/transmission , Arboviruses/genetics , Arboviruses/metabolism , Saliva/virology , Tachykinins/genetics , Tachykinins/metabolism , Virus Diseases/transmission
4.
Ann Rheum Dis ; 80(7): 920-929, 2021 07.
Article in English | MEDLINE | ID: mdl-33542104

ABSTRACT

OBJECTIVES: Plasmacytoid dendritic cells (pDC) have been implicated in the pathogenesis of autoimmune diseases, such as scleroderma (SSc). However, this has been derived from indirect evidence using ex vivo human samples or mouse pDC in vivo. We have developed human-specific pDC models to directly identify their role in inflammation and fibrosis, as well as attenuation of pDC function with BDCA2-targeting to determine its therapeutic application. METHODS: RNAseq of human pDC with TLR9 agonist ODN2216 and humanised monoclonal BDCA2 antibody, CBS004. Organotypic skin rafts consisting of fibroblasts and keratinocytes were stimulated with supernatant from TLR9-stimulated pDC and with CBS004. Human pDC were xenotransplanted into Nonobese diabetic/severe combined immunodeficiency (NOD SCID) mice treated with Aldara (inflammatory model), or bleomycin (fibrotic model) with CBS004 or human IgG control. Skin punch biopsies were used to assess gene and protein expression. RESULTS: RNAseq shows TLR9-induced activation of human pDC goes beyond type I interferon (IFN) secretion, which is functionally inactivated by BDCA2-targeting. Consistent with these findings, we show that BDCA2-targeting of pDC can completely suppress in vitro skin IFN-induced response. Most importantly, xenotransplantation of human pDC significantly increased in vivo skin IFN-induced response to TLR agonist and strongly enhanced fibrotic and immune response to bleomycin compared with controls. In these contexts, BDCA2-targeting suppressed human pDC-specific pathological responses. CONCLUSIONS: Our data indicate that human pDC play a key role in inflammation and immune-driven skin fibrosis, which can be effectively blocked by BDCA2-targeting, providing direct evidence supporting the development of attenuation of pDC function as a therapeutic application for SSc.


Subject(s)
Dendritic Cells/immunology , Lectins, C-Type/metabolism , Membrane Glycoproteins/metabolism , Receptors, Immunologic/metabolism , Scleroderma, Localized/immunology , Scleroderma, Localized/pathology , Animals , Dendritic Cells/pathology , Disease Models, Animal , Fibrosis , Heterografts , Humans , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Mice , Mice, Inbred NOD , Mice, SCID , Scleroderma, Localized/metabolism , Skin/immunology , Skin/metabolism , Skin/pathology
5.
PLoS Pathog ; 16(8): e1008716, 2020 08.
Article in English | MEDLINE | ID: mdl-32780760

ABSTRACT

Pandemic influenza A virus (IAV) remains a significant threat to global health. Preparedness relies primarily upon a single class of neuraminidase (NA) targeted antivirals, against which resistance is steadily growing. The M2 proton channel is an alternative clinically proven antiviral target, yet a near-ubiquitous S31N polymorphism in M2 evokes resistance to licensed adamantane drugs. Hence, inhibitors capable of targeting N31 containing M2 (M2-N31) are highly desirable. Rational in silico design and in vitro screens delineated compounds favouring either lumenal or peripheral M2 binding, yielding effective M2-N31 inhibitors in both cases. Hits included adamantanes as well as novel compounds, with some showing low micromolar potency versus pandemic "swine" H1N1 influenza (Eng195) in culture. Interestingly, a published adamantane-based M2-N31 inhibitor rapidly selected a resistant V27A polymorphism (M2-A27/N31), whereas this was not the case for non-adamantane compounds. Nevertheless, combinations of adamantanes and novel compounds achieved synergistic antiviral effects, and the latter synergised with the neuraminidase inhibitor (NAi), Zanamivir. Thus, site-directed drug combinations show potential to rejuvenate M2 as an antiviral target whilst reducing the risk of drug resistance.


Subject(s)
Influenza A Virus, H1N1 Subtype/drug effects , Influenza, Human/virology , Rimantadine/pharmacology , Viral Matrix Proteins/antagonists & inhibitors , Zanamivir/pharmacology , Antiviral Agents/pharmacology , Drug Resistance, Viral , Drug Synergism , Drug Therapy, Combination , Humans , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H1N1 Subtype/metabolism , Influenza, Human/drug therapy , Viral Matrix Proteins/genetics , Viral Matrix Proteins/metabolism
6.
Acta Neuropathol Commun ; 8(1): 135, 2020 08 13.
Article in English | MEDLINE | ID: mdl-32792006

ABSTRACT

Progressive multi-focal leukoencephalopathy (PML) is a potentially fatal encephalitis caused by JC polyomavirus (JCV). PML principally affects people with a compromised immune system, such as patients with multiple sclerosis (MS) receiving treatment with natalizumab. However, intrathecal synthesis of lipid-reactive IgM in MS patients is associated with a markedly lower incidence of natalizumab-associated PML compared to those without this antibody repertoire. Here we demonstrate that a subset of lipid-reactive human and murine IgMs induce a functional anti-viral response that inhibits replication of encephalitic Alpha and Orthobunyaviruses in multi-cellular central nervous system cultures. These lipid-specific IgMs trigger microglia to produce IFN-ß in a cGAS-STING-dependent manner, which induces an IFN-α/ß-receptor 1-dependent antiviral response in glia and neurons. These data identify lipid-reactive IgM as a mediator of anti-viral activity in the nervous system and provide a rational explanation why intrathecal synthesis of lipid-reactive IgM correlates with a reduced incidence of iatrogenic PML in MS.


Subject(s)
Autoantibodies/cerebrospinal fluid , Immunoglobulin M/cerebrospinal fluid , Leukoencephalopathy, Progressive Multifocal/immunology , Lipids/immunology , Multiple Sclerosis , Animals , Autoantibodies/immunology , Autoantigens/immunology , Humans , Immunocompromised Host/immunology , Immunoglobulin M/immunology , Immunologic Factors/adverse effects , Mice , Mice, Inbred C57BL , Multiple Sclerosis/drug therapy , Multiple Sclerosis/immunology , Natalizumab/adverse effects , Rats , Rats, Sprague-Dawley
7.
J Gen Virol ; 101(10): 1090-1102, 2020 10.
Article in English | MEDLINE | ID: mdl-32692647

ABSTRACT

Some free fatty acids derived from milk and vegetable oils are known to have potent antiviral and antibacterial properties. However, therapeutic applications of short- to medium-chain fatty acids are limited by physical characteristics such as immiscibility in aqueous solutions. We evaluated a novel proprietary formulation based on an emulsion of short-chain caprylic acid, ViroSAL, for its ability to inhibit a range of viral infections in vitro and in vivo. In vitro, ViroSAL inhibited the enveloped viruses Epstein-Barr, measles, herpes simplex, Zika and orf parapoxvirus, together with Ebola, Lassa, vesicular stomatitis and severe acute respiratory syndrome coronavirus 1 (SARS-CoV-1) pseudoviruses, in a concentration- and time-dependent manner. Evaluation of the components of ViroSAL revealed that caprylic acid was the main antiviral component; however, the ViroSAL formulation significantly inhibited viral entry compared with caprylic acid alone. In vivo, ViroSAL significantly inhibited Zika and Semliki Forest virus replication in mice following the inoculation of these viruses into mosquito bite sites. In agreement with studies investigating other free fatty acids, ViroSAL had no effect on norovirus, a non-enveloped virus, indicating that its mechanism of action may be surfactant disruption of the viral envelope. We have identified a novel antiviral formulation that is of great interest for the prevention and/or treatment of a broad range of enveloped viruses, particularly those of the skin and mucosal surfaces.


Subject(s)
Antiviral Agents , Severe acute respiratory syndrome-related coronavirus , Viruses , Zika Virus Infection , Zika Virus , Animals , Antiviral Agents/pharmacology , Lipids , Mice , Virus Internalization
8.
Sci Transl Med ; 12(527)2020 01 22.
Article in English | MEDLINE | ID: mdl-31969486

ABSTRACT

Arthropod-borne viruses (arboviruses) are important human pathogens for which there are no specific antiviral medicines. The abundance of genetically distinct arbovirus species, coupled with the unpredictable nature of their outbreaks, has made the development of virus-specific treatments challenging. Instead, we have defined and targeted a key aspect of the host innate immune response to virus at the arthropod bite that is common to all arbovirus infections, potentially circumventing the need for virus-specific therapies. Using mouse models and human skin explants, we identify innate immune responses by dermal macrophages in the skin as a key determinant of disease severity. Post-exposure treatment of the inoculation site by a topical TLR7 agonist suppressed both the local and subsequent systemic course of infection with a variety of arboviruses from the Alphavirus, Flavivirus, and Orthobunyavirus genera. Clinical outcome was improved in mice after infection with a model alphavirus. In the absence of treatment, antiviral interferon expression to virus in the skin was restricted to dermal dendritic cells. In contrast, stimulating the more populous skin-resident macrophages with a TLR7 agonist elicited protective responses in key cellular targets of virus that otherwise proficiently replicated virus. By defining and targeting a key aspect of the innate immune response to virus at the mosquito bite site, we have identified a putative new strategy for limiting disease after infection with a variety of genetically distinct arboviruses.


Subject(s)
Arbovirus Infections/immunology , Arbovirus Infections/metabolism , Arboviruses/immunology , Arboviruses/pathogenicity , Macrophages/metabolism , Skin/cytology , Alphavirus/immunology , Alphavirus/pathogenicity , Animals , Flavivirus/immunology , Flavivirus/pathogenicity , Humans , Membrane Glycoproteins/metabolism , Mice , Orthobunyavirus/immunology , Orthobunyavirus/pathogenicity , Toll-Like Receptor 7/metabolism
9.
J Biol Chem ; 293(8): 3003-3012, 2018 02 23.
Article in English | MEDLINE | ID: mdl-29279330

ABSTRACT

Chemokines are the principal regulators of leukocyte migration and are essential for initiation and maintenance of inflammation. Atypical chemokine receptor 2 (ACKR2) binds and scavenges proinflammatory CC-chemokines, regulates cutaneous T-cell positioning, and limits the spread of inflammation in vivo Altered ACKR2 function has been implicated in several inflammatory disorders, including psoriasis, a common and debilitating T-cell-driven disorder characterized by thick erythematous skin plaques. ACKR2 expression is abnormal in psoriatic skin, with decreased expression correlating with recruitment of T-cells into the epidermis and increased inflammation. However, the molecular mechanisms that govern ACKR2 expression are not known. Here, we identified specific psoriasis-associated microRNAs (miRs) that bind ACKR2, inhibit its expression, and are active in primary cultures of human cutaneous cells. Using both in silico and in vitro approaches, we show that miR-146b and miR-10b directly bind the ACKR2 3'-UTR and reduce expression of ACKR2 transcripts and protein in keratinocytes and lymphatic endothelial cells, respectively. Moreover, we demonstrate that ACKR2 expression is further down-regulated upon cell trauma, an important trigger for the development of new plaques in many psoriasis patients (the Koebner phenomenon). We found that tensile cell stress leads to rapid ACKR2 down-regulation and concurrent miR-146b up-regulation. Together, we provide, for the first time, evidence for epigenetic regulation of an atypical chemokine receptor. We propose a mechanism by which cell trauma and miRs coordinately exacerbate inflammation via down-regulation of ACKR2 expression and provide a putative mechanistic explanation for the Koebner phenomenon in psoriasis.


Subject(s)
Down-Regulation , Gene Expression Regulation , Keratinocytes/metabolism , MicroRNAs/metabolism , RNA Interference , Receptors, Chemokine/antagonists & inhibitors , 3' Untranslated Regions , Cells, Cultured , Computational Biology , Endothelial Cells/cytology , Endothelial Cells/immunology , Endothelial Cells/metabolism , Endothelial Cells/pathology , Epigenesis, Genetic , Expert Systems , Genes, Reporter , HEK293 Cells , Humans , Interferon-gamma/genetics , Interferon-gamma/metabolism , Keratinocytes/cytology , Keratinocytes/immunology , Keratinocytes/pathology , Organ Specificity , Psoriasis/immunology , Psoriasis/metabolism , Psoriasis/pathology , Receptors, Chemokine/genetics , Receptors, Chemokine/metabolism , Recombinant Proteins/metabolism , Skin/immunology , Skin/injuries , Skin/metabolism , Skin/pathology , Tensile Strength
10.
Trends Parasitol ; 33(8): 645-657, 2017 08.
Article in English | MEDLINE | ID: mdl-28495485

ABSTRACT

Mosquito-borne infections are increasing in number and are spreading to new regions at an unprecedented rate. In particular, mosquito-transmitted viruses, such as those that cause Zika, dengue, West Nile encephalitis, and chikungunya, have become endemic or have caused dramatic epidemics in many parts of the world. Aedes and Culex mosquitoes are the main culprits, spreading infection when they bite. Importantly, mosquitoes do not act as simple conduits that passively transfer virus from one individual to another. Instead, host responses to mosquito-derived factors have an important influence on infection and disease, aiding replication and dissemination within the host. Here, we discuss the latest research developments regarding this fascinating interplay between mosquito, virus, and the mammalian host.


Subject(s)
Arbovirus Infections/immunology , Arbovirus Infections/transmission , Arboviruses/immunology , Insect Bites and Stings/immunology , Mosquito Vectors/immunology , Skin/immunology , Animals , Humans , Insect Bites and Stings/virology , Mosquito Vectors/virology , Saliva/immunology , Skin/virology
11.
J Invest Dermatol ; 137(1): 85-94, 2017 01.
Article in English | MEDLINE | ID: mdl-27568525

ABSTRACT

Elucidating the poorly defined mechanisms by which inflammatory lesions are spatially restricted in vivo is of critical importance in understanding skin disease. Chemokines are the principal regulators of leukocyte migration and are essential in the initiation and maintenance of inflammation. The membrane-bound psoriasis-associated atypical chemokine receptor 2 (ACKR2) binds, internalizes and degrades most proinflammatory CC-chemokines. Here we investigate the role of ACKR2 in limiting the spread of cutaneous psoriasiform inflammation to sites that are remote from the primary lesion. Circulating factors capable of regulating ACKR2 function at remote sites were identified and examined using a combination of clinical samples, relevant primary human cell cultures, in vitro migration assays, and the imiquimod-induced model of psoriasiform skin inflammation. Localized inflammation and IFN-γ together up-regulate ACKR2 in remote tissues, protecting them from the spread of inflammation. ACKR2 controls inflammatory T-cell chemotaxis and positioning within the skin, preventing an epidermal influx that is associated with lesion development. Our results have important implications for our understanding of how spatial restriction is imposed on the spread of inflammatory lesions and highlight systemic ACKR2 induction as a therapeutic strategy in the treatment and prevention of psoriasis and potentially a broad range of other immune-mediated diseases.


Subject(s)
Aminoquinolines/pharmacology , Inflammation/drug therapy , Inflammation/genetics , Psoriasis/genetics , Psoriasis/pathology , Receptors, Chemokine/genetics , Animals , Biopsy, Needle , Cells, Cultured , Disease Models, Animal , Gene Expression Regulation , Humans , Imiquimod , Immunohistochemistry , Inflammation/pathology , Keratinocytes/cytology , Keratinocytes/drug effects , Mice , Mice, Inbred C57BL , Phenotype , Polymerase Chain Reaction/methods , Psoriasis/drug therapy , Random Allocation , Receptors, Chemokine/metabolism , Reference Values , Statistics, Nonparametric , Up-Regulation
12.
Immunity ; 44(6): 1455-69, 2016 06 21.
Article in English | MEDLINE | ID: mdl-27332734

ABSTRACT

Aedes aegypti mosquitoes are responsible for transmitting many medically important viruses such as those that cause Zika and dengue. The inoculation of viruses into mosquito bite sites is an important and common stage of all mosquito-borne virus infections. We show, using Semliki Forest virus and Bunyamwera virus, that these viruses use this inflammatory niche to aid their replication and dissemination in vivo. Mosquito bites were characterized by an edema that retained virus at the inoculation site and an inflammatory influx of neutrophils that coordinated a localized innate immune program that inadvertently facilitated virus infection by encouraging the entry and infection of virus-permissive myeloid cells. Neutrophil depletion and therapeutic blockade of inflammasome activity suppressed inflammation and abrogated the ability of the bite to promote infection. This study identifies facets of mosquito bite inflammation that are important determinants of the subsequent systemic course and clinical outcome of virus infection.


Subject(s)
Arbovirus Infections/immunology , Bunyamwera virus/physiology , Inflammation/immunology , Insect Bites and Stings/immunology , Neutrophils/immunology , Semliki forest virus/physiology , Virus Replication , Animals , Cell Movement , Cells, Cultured , Culicidae/immunology , Humans , Immunity, Innate , Inflammasomes/metabolism , Inflammation/virology , Insect Bites and Stings/virology , Mice , Neutrophils/virology
13.
J Virol ; 88(17): 9553-67, 2014 Sep 01.
Article in English | MEDLINE | ID: mdl-24899190

ABSTRACT

UNLABELLED: The encephalitic response to viral infection requires local chemokine production and the ensuing recruitment of immune and inflammatory leukocytes. Accordingly, chemokine receptors present themselves as plausible therapeutic targets for drugs aimed at limiting encephalitic responses. However, it remains unclear which chemokines are central to this process and whether leukocyte recruitment is important for limiting viral proliferation and survival in the brain or whether it is predominantly a driver of coincident inflammatory pathogenesis. Here we examine chemokine expression and leukocyte recruitment in the context of avirulent and virulent Semliki Forest virus (SFV) as well as West Nile virus infection and demonstrate rapid and robust expression of a variety of inflammatory CC and CXC chemokines in all models. On this basis, we define a chemokine axis involved in leukocyte recruitment to the encephalitic brain during SFV infection. CXCR3 is the most active; CCR2 is also active but less so, and CCR5 plays only a modest role in leukocyte recruitment. Importantly, inhibition of each of these receptors individually and the resulting suppression of leukocyte recruitment to the infected brain have no effect on viral titer or survival following infection with a virulent SFV strain. In contrast, simultaneous blockade of CXCR3 and CCR2 results in significantly reduced mortality in response to virulent SFV infection. In summary, therefore, our data provide an unprecedented level of insight into chemokine orchestration of leukocyte recruitment in viral encephalitis. Our data also highlight CXCR3 and CCR2 as possible therapeutic targets for limiting inflammatory damage in response to viral infection of the brain. IMPORTANCE: Brain inflammation (encephalitis) in response to viral infection can lead to severe illness and even death. This therefore represents an important clinical problem and one that requires the development of new therapeutic approaches. Central to the pathogenesis of encephalitis is the recruitment of inflammatory leukocytes to the infected brain, a process driven by members of the chemokine family. Here we provide an in-depth analysis of the chemokines involved in leukocyte recruitment to the virally infected brain and demonstrate that simultaneous blockade of two of these receptors, namely, CXCR3 and CCR2, does not alter viral titers within the brain but markedly reduces inflammatory leukocyte recruitment and enhances survival in a murine model of lethal viral encephalitis. Our results therefore highlight chemokine receptors as plausible therapeutic targets in treating viral encephalitis.


Subject(s)
Alphavirus Infections/immunology , Chemokines/metabolism , Encephalitis, Viral/immunology , Leukocytes/immunology , West Nile Fever/immunology , Animals , Brain/pathology , Brain/virology , Disease Models, Animal , Female , Mice, Inbred C57BL , Receptors, Chemokine/immunology , Semliki forest virus/immunology , Survival Analysis , Viral Load , West Nile virus/immunology
14.
J Immunol ; 192(12): 6120-6130, 2014 Jun 15.
Article in English | MEDLINE | ID: mdl-24850722

ABSTRACT

Cell therapy regimens are frequently compromised by low-efficiency cell homing to therapeutic niches. Improvements in this regard would enhance effectiveness of clinically applicable cell therapy. The major regulators of tissue-specific cellular migration are chemokines, and therefore selection of therapeutic cellular populations for appropriate chemokine receptor expression would enhance tissue-homing competence. A number of practical considerations preclude the use of Abs in this context, and alternative approaches are required. In this study, we demonstrate that appropriately labeled chemokines are at least as effective in detecting their cognate receptors as commercially available Abs. We also demonstrate the utility of biotinylated chemokines as cell-sorting reagents. Specifically, we demonstrate, in the context of CCR7 (essential for lymph node homing of leukocytes), the ability of biotinylated CCL19 with magnetic bead sorting to enrich for CCR7-expressing cells. The sorted cells demonstrate improved CCR7 responsiveness and lymph node-homing capability, and the sorting is effective for both T cells and dendritic cells. Importantly, the ability of chemokines to detect CCR7, and sort for CCR7 positivity, crosses species being effective on murine and human cells. This novel approach to cell sorting is therefore inexpensive, versatile, and applicable to numerous cell therapy contexts. We propose that this represents a significant technological advance with important therapeutic implications.


Subject(s)
Chemokine CCL19/chemistry , Flow Cytometry/methods , Receptors, CCR7/chemistry , Animals , Chemokine CCL19/immunology , Female , Humans , Male , Mice , Receptors, CCR7/immunology
15.
J Biol Chem ; 288(51): 36473-83, 2013 Dec 20.
Article in English | MEDLINE | ID: mdl-24194523

ABSTRACT

The inflammatory response is normally limited by mechanisms regulating its resolution. In the absence of resolution, inflammatory pathologies can emerge, resulting in substantial morbidity and mortality. We have been studying the D6 chemokine scavenging receptor, which played an indispensable role in the resolution phase of inflammatory responses and does so by facilitating removal of inflammatory CC chemokines. In D6-deficient mice, otherwise innocuous cutaneous inflammatory stimuli induce a grossly exaggerated inflammatory response that bears many similarities to human psoriasis. In the present study, we have used transcriptomic approaches to define the molecular make up of this response. The data presented highlight potential roles for a number of cytokines in initiating and maintaining the psoriasis-like pathology. Most compellingly, we provide data indicating a key role for the type I interferon pathway in the emergence of this pathology. Neutralizing antibodies to type I interferons are able to ameliorate the psoriasis-like pathology, confirming a role in its development. Comparison of transcriptional data generated from this mouse model with equivalent data obtained from human psoriasis further demonstrates the strong similarities between the experimental and clinical systems. As such, the transcriptional data obtained in this preclinical model provide insights into the cytokine network active in exaggerated inflammatory responses and offer an excellent tool to evaluate the efficacy of compounds designed to therapeutically interfere with inflammatory processes.


Subject(s)
Interferon Type I/metabolism , Psoriasis/immunology , Receptors, CCR10/genetics , Animals , Female , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Interferon Type I/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Oligonucleotide Array Sequence Analysis , Phorbol Esters/toxicity , Psoriasis/chemically induced , Psoriasis/genetics , Psoriasis/pathology , Transcription, Genetic , Chemokine Receptor D6
16.
Blood ; 121(18): 3768-77, 2013 May 02.
Article in English | MEDLINE | ID: mdl-23479571

ABSTRACT

The mechanisms by which CC chemokine receptor (CCR)7 ligands are selectively presented on lymphatic endothelium in the presence of inflammatory chemokines are poorly understood. The chemokine-scavenging receptor D6 is expressed on lymphatic endothelial cells (LEC) and contributes to selective presentation of CCR7 ligands by suppressing inflammatory chemokine binding to LEC surfaces. As well as preventing inappropriate inflammatory cell attachment to LECs, D6 is specifically involved in regulating the ability of LEC to discriminate between mature and immature dendritic cells (DCs). D6 overexpression reduces immature DC (iDC) adhesion to LECs, whereas D6 knockdown increases adhesion of iDCs that displace mature DCs. LEC D6 expression is regulated by growth factors, cytokines, and tumor microenvironments. In particular, interleukin-6 and interferon-γ are potent inducers, indicating a preferential role for D6 in inflamed contexts. Expression of the viral interleukin-6 homolog from Kaposi sarcoma-associated herpesvirus is also sufficient to induce significant D6 upregulation both in vitro and in vivo, and Kaposi sarcoma and primary effusion lymphoma cells demonstrate high levels of D6 expression. We therefore propose that D6, which is upregulated in both inflammatory and tumor contexts, is an essential regulator of inflammatory leukocyte interactions with LECs and is required for immature/mature DC discrimination by LECs.


Subject(s)
Endothelial Cells/metabolism , Receptors, CCR10/genetics , Receptors, CCR10/physiology , Animals , CHO Cells , Cell Communication/genetics , Cell Communication/immunology , Cell Differentiation/genetics , Cell Differentiation/immunology , Cells, Cultured , Cricetinae , Cricetulus , Dendritic Cells/immunology , Dendritic Cells/metabolism , Dendritic Cells/physiology , Endothelial Cells/immunology , HEK293 Cells , Humans , Inflammation/genetics , Inflammation/immunology , Inflammation/metabolism , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/metabolism , Receptors, CCR10/analysis , Receptors, CCR10/metabolism , Transfection , Chemokine Receptor D6
17.
Blood ; 118(23): 6220-9, 2011 Dec 01.
Article in English | MEDLINE | ID: mdl-21979941

ABSTRACT

Lymphatic endothelial cells are important for efficient flow of antigen-bearing fluid and antigen-presenting cells (APCs) from peripheral sites to lymph nodes (LNs). APC movement to LNs is dependent on the constitutive chemokine receptor CCR7, although how conflicting inflammatory and constitutive chemokine cues are integrated at lymphatic surfaces during this process is not understood. Here we reveal a previously unrecognized aspect of the regulation of this process. The D6 chemokine-scavenging receptor, which is expressed on lymphatic endothelial cells (LECs), maintains lymphatic surfaces free of inflammatory CC-chemokines and minimizes interaction of inflammatory leukocytes with these surfaces. D6 does not alter the level of CCR7 ligands on LECs, thus ensuring selective presentation of homeostatic chemokines for interaction with CCR7(+) APCs. Accordingly, in D6-deficient mice, inflammatory CC-chemokine adherence to LECs results in inappropriate perilymphatic accumulation of inflammatory leukocytes at peripheral inflamed sites and draining LNs. This results in lymphatic congestion and impaired movement of APCs, and fluid, from inflamed sites to LNs. We propose that D6, by suppressing inflammatory chemokine binding to lymphatic surfaces, and thereby preventing inappropriate inflammatory leukocyte adherence, is a key regulator of lymphatic function and a novel, and indispensable, contributor to the integration of innate and adaptive immune responses.


Subject(s)
Body Fluids/immunology , Cell Movement/immunology , Endothelial Cells/immunology , Lymph Nodes/immunology , Receptors, Chemokine/immunology , Adaptive Immunity/immunology , Animals , Antigen-Presenting Cells/cytology , Antigen-Presenting Cells/immunology , Chemokine CCL2/immunology , Chemokine CCL2/metabolism , Chemokine CCL5/immunology , Chemokine CCL5/metabolism , Endothelial Cells/cytology , Endothelial Cells/metabolism , Immunity, Innate/immunology , Leukocytes/cytology , Leukocytes/immunology , Lymph/immunology , Lymph/metabolism , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Receptors, CCR2/immunology , Receptors, CCR2/metabolism , Receptors, CCR7/immunology , Receptors, CCR7/metabolism , Receptors, Chemokine/genetics
18.
Biochem Biophys Res Commun ; 394(4): 1006-11, 2010 Apr 16.
Article in English | MEDLINE | ID: mdl-20331977

ABSTRACT

Immune responses in the central nervous system (CNS) are carefully regulated. Despite the absence of most immune processes and a substantive blood brain barrier, potent immune responses form during infection and autoimmunity. Astrocytes are innate immune sentinels that ensheath parenchymal blood vessels and sit at the gateway to the CNS parenchyma. Viral and bacterial infections trigger the influx of distinct leukocyte subsets. We show that astrocytes alone are sufficient for distinguishing between these two main types of infection and triggers release of relevant chemokines that relate to the microbe recognised. Bacterial-associated molecules induced the preferential expression of CCL2, CXCL1, CCL20 and CCL3 whilst a virus-associated dsRNA analogue preferentially up-regulated CXCL10 and CCL5. Thus, astrocytes can respond to infection in a distinct and appropriate manner suggesting they have the capacity to attract appropriate sets of leukocytes into the brain parenchyma. Astrocytes themselves are unable to respond to these chemokines since they were devoid of most chemokine receptors but expressed CXCR4, CXCR7 and CXCR6 at rest. Stimulation with TGF-beta specifically up-regulated CXCR6 expression and may explain how TGF-beta/CXCL16-expressing gliomas are so effective at attracting astroglial cells.


Subject(s)
Astrocytes/immunology , Astrocytes/microbiology , Chemokines/metabolism , Immunity, Innate , Metabolic Networks and Pathways/immunology , Animals , Astrocytes/virology , Cells, Cultured , Mice , Receptors, Chemokine/metabolism
19.
Biochem Biophys Res Commun ; 390(3): 716-21, 2009 Dec 18.
Article in English | MEDLINE | ID: mdl-19833095

ABSTRACT

We present the first report utilizing, and showing the functional relevance of, self-assembling polyvalent ligands specific for integrin alpha5beta1 in murine embryonic stem (mES) cell adhesion. Di, tri and tetrameric 9th-10th type III fibronectin domains (FIII9'10) were used to generate clustered integrin alpha5beta1 ligand surfaces for mES cell culture. Compared to gelatin, FIII9'10 (monomer), FIII9'10-trimer and -tetramer, the FIII9'10-dimer supported the highest number of mES cell colonies. No evidence of domain unfolding upon surface adsorption was found. Colonies appeared disperse with a spread cell morphology unless subdued back to a tight morphology with increasing concentrations of leukemia inhibitory factor (LIF). In the presence of LIF, mES cells adherent to the FIII9'10-dimer showed transient upregulation of Oct-4, the mesodermal transcription factor, Brachyury, and the ectodermal marker, Nestin. However, dual upregulation of Nanog maintained the mES cells in a pluripotent state, confirmed by alkaline phosphatase staining. Therefore, the behavior of mES cells adherent to dimeric integrin alpha5beta1 ligands is a largely morphological phenomenon conferring pro-differentiation signals towards mesodermal and ectodermal lineages. This work will be of interest to cell and tissue engineering groups aiming to control ES cell behavior through integrin ligand presentation and synthetic substrates.


Subject(s)
Cell Differentiation , Embryonic Stem Cells/physiology , Fibronectins/metabolism , Integrin alpha5beta1/metabolism , Tissue Engineering , Animals , Cell Adhesion , Cells, Cultured , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Leukemia Inhibitory Factor/metabolism , Ligands , Mice , Protein Multimerization , Transcription, Genetic
20.
Blood ; 113(18): 4224-31, 2009 Apr 30.
Article in English | MEDLINE | ID: mdl-19202130

ABSTRACT

Toll-like receptors orchestrate rapid local protective innate-immune responses to invading pathogens and optimize leukocyte priming of subsequent adaptive responses. Paradoxically, systemic excess of the TLR2 ligand, bacterial lipoprotein (BLP), suppresses peripheral inflammatory responses. Here, we demonstrate that this phenomenon is regulated via the TLR2-dependent, cell-autonomous down-regulation of inflammatory chemokine receptor expression on a variety of leukocyte subsets. Remarkably, BLP mediated no effect on constitutive chemokine receptor expression. By tracking adoptively transferred wild-type and TLR2(-/-) leukocytes in vivo, we observed that BLP mediated chemokine receptor switching directed leukocytes away from inflamed sites toward secondary lymphoid organs. These data highlight a novel role for TLR ligands, such as BLP, in regulating leukocyte retention and migration away from innate immune lesions via discrete constitutive and inflammatory chemokine receptor regulation.


Subject(s)
Cell Movement/physiology , Inflammation/prevention & control , Leukocytes/physiology , Lipoproteins/administration & dosage , Receptors, Chemokine/metabolism , Skin/immunology , Toll-Like Receptor 2/physiology , Animals , Cells, Cultured , Chemokines/metabolism , Female , Flow Cytometry , Inflammation/chemically induced , Inflammation/metabolism , Interferon-gamma/pharmacology , Ligands , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/genetics , NF-kappa B/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Skin/metabolism , Tetradecanoylphorbol Acetate/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...