Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 61
Filter
2.
PLoS Negl Trop Dis ; 13(6): e0006983, 2019 06.
Article in English | MEDLINE | ID: mdl-31242184

ABSTRACT

BACKGROUND: T cell immunoglobulin mucin domain-1 (TIM-1) is a phosphatidylserine (PS) receptor, mediating filovirus entry into cells through interactions with PS on virions. TIM-1 expression has been implicated in Ebola virus (EBOV) pathogenesis; however, it remains unclear whether this is due to TIM-1 serving as a filovirus receptor in vivo or, as others have suggested, TIM-1 induces a cytokine storm elicited by T cell/virion interactions. Here, we use a BSL2 model virus that expresses EBOV glycoprotein to demonstrate the importance of TIM-1 as a virus receptor late during in vivo infection. METHODOLOGY/PRINCIPAL FINDINGS: Infectious, GFP-expressing recombinant vesicular stomatitis virus encoding either full length EBOV glycoprotein (EBOV GP/rVSV) or mucin domain deleted EBOV glycoprotein (EBOV GPΔO/rVSV) was used to assess the role of TIM-1 during in vivo infection. GFP-expressing rVSV encoding its native glycoprotein G (G/rVSV) served as a control. TIM-1-sufficient or TIM-1-deficient BALB/c interferon α/ß receptor-/- mice were challenged with these viruses. While G/rVSV caused profound morbidity and mortality in both mouse strains, TIM-1-deficient mice had significantly better survival than TIM-1-expressing mice following EBOV GP/rVSV or EBOV GPΔO/rVSV challenge. EBOV GP/rVSV or EBOV GPΔO/rVSV in spleen of infected animals was high and unaffected by expression of TIM-1. However, infectious virus in serum, liver, kidney and adrenal gland was reduced late in infection in the TIM-1-deficient mice, suggesting that virus entry via this receptor contributes to virus load. Consistent with higher virus loads, proinflammatory chemokines trended higher in organs from infected TIM-1-sufficient mice compared to the TIM-1-deficient mice, but proinflammatory cytokines were more modestly affected. To assess the role of T cells in EBOV GP/rVSV pathogenesis, T cells were depleted in TIM-1-sufficient and -deficient mice and the mice were challenged with virus. Depletion of T cells did not alter the pathogenic consequences of virus infection. CONCLUSIONS: Our studies provide evidence that at late times during EBOV GP/rVSV infection, TIM-1 increased virus load and associated mortality, consistent with an important role of this receptor in virus entry. This work suggests that inhibitors which block TIM-1/virus interaction may serve as effective antivirals, reducing virus load at late times during EBOV infection.


Subject(s)
Ebolavirus/physiology , Hemorrhagic Fever, Ebola/virology , Hepatitis A Virus Cellular Receptor 1/metabolism , Receptors, Virus/metabolism , Virus Internalization , Animals , Ebolavirus/genetics , Female , Green Fluorescent Proteins/analysis , Green Fluorescent Proteins/genetics , Hepatitis A Virus Cellular Receptor 1/deficiency , Mice, Inbred BALB C , Mice, Knockout , Receptors, Virus/deficiency , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Reverse Genetics , Vesiculovirus/genetics , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism
3.
J Bacteriol ; 201(11)2019 06 01.
Article in English | MEDLINE | ID: mdl-30858299

ABSTRACT

The Gram-negative outer membrane (OM) is a selectively permeable asymmetric bilayer that allows vital nutrients to diffuse into the cell but prevents toxins and hydrophobic molecules from entering. Functionally and structurally diverse ß-barrel outer membrane proteins (OMPs) build and maintain the permeability barrier, making the assembly of OMPs crucial for cell viability. In this work, we characterize an assembly-defective mutant of the maltoporin LamB, LamBG439D We show that the folding defect of LamBG439D results in an accumulation of unfolded substrate that is toxic to the cell when the periplasmic protease DegP is removed. Selection for suppressors of this toxicity identified the novel mutant degSA323E allele. The mutant DegSA323E protein contains an amino acid substitution at the PDZ/protease domain interface that results in a partially activated conformation of this protein. This activation increases basal levels of downstream σE stress response signaling. Furthermore, the enhanced σE activity of DegSA323E suppresses a number of other assembly-defective conditions without exhibiting the toxicity associated with high levels of σE activity. We propose that the increased basal levels of σE signaling primes the cell to respond to envelope stress before OMP assembly defects threaten cell viability. This finding addresses the importance of envelope stress responses in monitoring the OMP assembly process and underpins the critical balance between envelope defects and stress response activation.IMPORTANCE Gram-negative bacteria, such as Escherichia coli, inhabit a natural environment that is prone to flux. In order to cope with shifting growth conditions and the changing availability of nutrients, cells must be capable of quickly responding to stress. Stress response pathways allow cells to rapidly shift gene expression profiles to ensure survival in this unpredictable environment. Here we describe a mutant that partially activates the σE stress response pathway. The elevated basal level of this stress response allows the cell to quickly respond to overwhelming stress to ensure cell survival.


Subject(s)
Bacterial Outer Membrane Proteins/genetics , Escherichia coli Proteins/genetics , Escherichia coli/genetics , Gene Expression Regulation, Bacterial , Heat-Shock Proteins/genetics , Periplasmic Proteins/genetics , Porins/genetics , Receptors, Virus/genetics , Serine Endopeptidases/genetics , Sigma Factor/genetics , Adaptation, Physiological/genetics , Amino Acid Substitution , Bacterial Outer Membrane Proteins/chemistry , Escherichia coli/metabolism , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Heat-Shock Proteins/deficiency , Microbial Viability , Models, Molecular , Mutation , Periplasm/genetics , Periplasm/metabolism , Porins/chemistry , Porins/deficiency , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Receptors, Virus/chemistry , Receptors, Virus/deficiency , Serine Endopeptidases/deficiency , Sigma Factor/metabolism , Signal Transduction , Stress, Physiological
4.
J Clin Invest ; 128(6): 2613-2625, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29757192

ABSTRACT

Critical immune-suppressive pathways beyond programmed death 1 (PD-1) and programmed death ligand 1 (PD-L1) require greater attention. Nectins and nectin-like molecules might be promising targets for immunotherapy, since they play critical roles in cell proliferation and migration and exert immunomodulatory functions in pathophysiological conditions. Here, we show CD155 expression in both malignant cells and tumor-infiltrating myeloid cells in humans and mice. Cd155-/- mice displayed reduced tumor growth and metastasis via DNAM-1 upregulation and enhanced effector function of CD8+ T and NK cells, respectively. CD155-deleted tumor cells also displayed slower tumor growth and reduced metastases, demonstrating the importance of a tumor-intrinsic role of CD155. CD155 absence on host and tumor cells exerted an even greater inhibition of tumor growth and metastasis. Blockade of PD-1 or both PD-1 and CTLA4 was more effective in settings in which CD155 was limiting, suggesting the clinical potential of cotargeting PD-L1 and CD155 function.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunity, Cellular , Killer Cells, Natural/immunology , Neoplasm Proteins/deficiency , Neoplasms, Experimental/immunology , Receptors, Virus/deficiency , Animals , B7-H1 Antigen/genetics , B7-H1 Antigen/immunology , CD8-Positive T-Lymphocytes/pathology , CTLA-4 Antigen/genetics , CTLA-4 Antigen/immunology , Cell Line, Tumor , Killer Cells, Natural/pathology , Mice , Mice, Inbred BALB C , Mice, Knockout , Neoplasm Proteins/immunology , Neoplasms, Experimental/genetics , Neoplasms, Experimental/pathology , Receptors, Virus/immunology
5.
Nature ; 557(7706): 570-574, 2018 05.
Article in English | MEDLINE | ID: mdl-29769725

ABSTRACT

Arthritogenic alphaviruses comprise a group of enveloped RNA viruses that are transmitted to humans by mosquitoes and cause debilitating acute and chronic musculoskeletal disease 1 . The host factors required for alphavirus entry remain poorly characterized 2 . Here we use a genome-wide CRISPR-Cas9-based screen to identify the cell adhesion molecule Mxra8 as an entry mediator for multiple emerging arthritogenic alphaviruses, including chikungunya, Ross River, Mayaro and O'nyong nyong viruses. Gene editing of mouse Mxra8 or human MXRA8 resulted in reduced levels of viral infection of cells and, reciprocally, ectopic expression of these genes resulted in increased infection. Mxra8 bound directly to chikungunya virus particles and enhanced virus attachment and internalization into cells. Consistent with these findings, Mxra8-Fc fusion protein or anti-Mxra8 monoclonal antibodies blocked chikungunya virus infection in multiple cell types, including primary human synovial fibroblasts, osteoblasts, chondrocytes and skeletal muscle cells. Mutagenesis experiments suggest that Mxra8 binds to a surface-exposed region across the A and B domains of chikungunya virus E2 protein, which are a speculated site of attachment. Finally, administration of the Mxra8-Fc protein or anti-Mxra8 blocking antibodies to mice reduced chikungunya and O'nyong nyong virus infection as well as associated foot swelling. Pharmacological targeting of Mxra8 could form a strategy for mitigating infection and disease by multiple arthritogenic alphaviruses.


Subject(s)
Chikungunya virus/metabolism , Immunoglobulins/metabolism , Membrane Proteins/metabolism , O'nyong-nyong Virus/metabolism , Receptors, Virus/metabolism , 3T3 Cells , Animals , Antibodies, Blocking/immunology , CRISPR-Cas Systems/genetics , Chikungunya virus/pathogenicity , Chondrocytes/metabolism , Fibroblasts/metabolism , Humans , Immunoglobulins/immunology , Male , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/immunology , Mice , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , O'nyong-nyong Virus/pathogenicity , Osteoblasts/metabolism , Receptors, Fc/metabolism , Receptors, Virus/deficiency , Receptors, Virus/genetics
6.
Cell Death Differ ; 25(3): 573-588, 2018 03.
Article in English | MEDLINE | ID: mdl-29229999

ABSTRACT

Heme is required for cell respiration and survival. Nevertheless, its intracellular levels need to be finely regulated to avoid heme excess, which may catalyze the production of reactive oxygen species (ROS) and promote cell death. Here, we show that alteration of heme homeostasis in endothelial cells due to the loss of the heme exporter FLVCR1a, results in impaired angiogenesis. In vitro, FLVCR1a silencing in endothelial cells causes defective tubulogenesis and poor viability due to intracellular heme accumulation. Consistently, endothelial-specific Flvcr1a knockout mice show aberrant angiogenesis responsible for hemorrhages and embryonic lethality. Importantly, we demonstrate that impaired heme export leads to endothelial cell death by paraptosis and provide evidence that endoplasmic reticulum (ER) stress precedes heme-induced paraptosis. These findings highlight a crucial role for the cytosolic heme pool in the control of endothelial cell survival and in the regulation of the angiogenic process. Interfering with endothelial heme export represents a valuable model for a deeper understanding of the molecular mechanisms underlying heme-triggered paraptosis and, in the future, might provide a novel tool for the modulation of angiogenesis in pathophysiologic conditions.


Subject(s)
Apoptosis , Endothelial Cells/metabolism , Heme/metabolism , Membrane Transport Proteins/metabolism , Neovascularization, Pathologic/metabolism , Receptors, Virus/metabolism , Animals , Apoptosis/genetics , Cells, Cultured , Endoplasmic Reticulum Stress/genetics , Female , Heme/genetics , Humans , Male , Membrane Transport Proteins/deficiency , Membrane Transport Proteins/genetics , Mice , Mice, Knockout , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Receptors, Virus/deficiency , Receptors, Virus/genetics
7.
FASEB J ; 30(12): 4056-4070, 2016 12.
Article in English | MEDLINE | ID: mdl-27609773

ABSTRACT

Pulmonary fibrosis is a progressive and often fatal condition that is believed to be partially orchestrated by macrophages. Mechanisms that control migration of these cells into and within the lung remain undefined. We evaluated the contributions of the semaphorin receptor, plexin C1 (PLXNC1), and the exocytic calcium sensor, synaptotagmin 7 (Syt7), in these processes. We evaluated the role of PLXNC1 in macrophage migration by using Boyden chambers and scratch tests, characterized its contribution to experimentally induced lung fibrosis in mice, and defined the mechanism for our observations. Our findings reveal that relative to control participants, patients with idiopathic pulmonary fibrosis demonstrate excessive monocyte migration and underexpression of PLXNC1 in the lungs and circulation, a finding that is recapitulated in the setting of scleroderma-related interstitial lung disease. Relative to wild type, PLXNC1-/- mouse macrophages are excessively migratory, and PLXNC1-/- mice show exacerbated collagen accumulation in response to either inhaled bleomycin or inducible lung targeted TGF-ß1 overexpression. These findings are ameliorated by replacement of PLXNC1 on bone marrow-derived cells or by genetic deletion of Syt7. These data demonstrate the previously unrecognized observation that PLXNC1 deficiency permits Syt7-mediated macrophage migration and enhances mammalian lung fibrosis.-Peng, X., Moore, M., Mathur, A., Zhou, Y., Sun, H., Gan, Y., Herazo-Maya, J. D., Kaminski, N., Hu, X., Pan, H., Ryu, C., Osafo-Addo, A., Homer, R. J., Feghali-Bostwick, C., Fares, W. H., Gulati, M., Hu, B., Lee, C.-G., Elias, J. A., Herzog, E. L. Plexin C1 deficiency permits synaptotagmin 7-mediated macrophage migration and enhances mammalian lung fibrosis.


Subject(s)
Macrophages/metabolism , Nerve Tissue Proteins/metabolism , Pulmonary Fibrosis/metabolism , Receptors, Cell Surface/metabolism , Receptors, Virus/metabolism , Synaptotagmins/metabolism , Animals , Disease Models, Animal , Humans , Lung/metabolism , Mice, Knockout , Nerve Tissue Proteins/deficiency , Pulmonary Fibrosis/genetics , Receptors, Cell Surface/deficiency , Receptors, Virus/deficiency , Transforming Growth Factor beta1/metabolism
8.
J Leukoc Biol ; 100(4): 781-789, 2016 10.
Article in English | MEDLINE | ID: mdl-27034402

ABSTRACT

Previous studies have suggested that NK cells may limit T cell responses by their ability to eradicate dendritic cells, as demonstrated by NK cell-mediated killing of dendritic cells generated from mouse bone marrow cells or human monocytes with GM-CSF. In the present study, we demonstrated that conventional dendritic cells, generated in vitro with Flt3 ligand or from spleens, were resistant to NK cell-mediated lysis. However, upon stimulation with GM-CSF, NK cells could mediate lysis of these dendritic cells. GM-CSF-stimulated Flt3 ligand dendritic cells or splenic dendritic cells increased surface expression of costimulatory molecules and known NK cell ligands. Likewise, NK cells could target dendritic cells in vivo, which could be inhibited, in part, by anti-GM-CSF antibodies. The blocking of CD54 or CD226 inhibited NK cell-mediated cytotoxicity of the GM-CSF-stimulated Flt3 ligand conventional dendritic cells. Furthermore, the CD226+NKG2A- subset of NK cells was selectively better at targeting GM-CSF-stimulated Flt3 ligand conventional dendritic cells. However, CD155, a known ligand for CD226, could also act as an inhibitor of NK cell-mediated lysis, as dendritic cells lacking CD155 were more sensitive to NK cell-mediated lysis than wild-type dendritic cells. We hypothesize that by only permitting a subset of NK cells to target activated dendritic cells during inflammation, this would allow the immune system to balance between dendritic cells able to drive adaptive immune responses and dendritic cells targeted for elimination by NK cells to hinder, e.g., spread of infection.


Subject(s)
Antigens, Differentiation, T-Lymphocyte/immunology , Cytotoxicity, Immunologic/immunology , Dendritic Cells/immunology , Intercellular Adhesion Molecule-1/immunology , Killer Cells, Natural/immunology , Animals , Cells, Cultured , Dendritic Cells/transplantation , Genes, RAG-1 , Graft Rejection/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Inflammation , Interleukin-18/pharmacology , Membrane Proteins/pharmacology , Mice , Mice, Knockout , Programmed Cell Death 1 Receptor/deficiency , Receptors, Virus/deficiency , Recombinant Proteins/pharmacology , Spleen/immunology
9.
Nature ; 530(7588): 108-12, 2016 Feb 04.
Article in English | MEDLINE | ID: mdl-26814968

ABSTRACT

Adeno-associated virus (AAV) vectors are currently the leading candidates for virus-based gene therapies because of their broad tissue tropism, non-pathogenic nature and low immunogenicity. They have been successfully used in clinical trials to treat hereditary diseases such as haemophilia B (ref. 2), and have been approved for treatment of lipoprotein lipase deficiency in Europe. Considerable efforts have been made to engineer AAV variants with novel and biomedically valuable cell tropisms to allow efficacious systemic administration, yet basic aspects of AAV cellular entry are still poorly understood. In particular, the protein receptor(s) required for AAV entry after cell attachment remains unknown. Here we use an unbiased genetic screen to identify proteins essential for AAV serotype 2 (AAV2) infection in a haploid human cell line. The most significantly enriched gene of the screen encodes a previously uncharacterized type I transmembrane protein, KIAA0319L (denoted hereafter as AAV receptor (AAVR)). We characterize AAVR as a protein capable of rapid endocytosis from the plasma membrane and trafficking to the trans-Golgi network. We show that AAVR directly binds to AAV2 particles, and that anti-AAVR antibodies efficiently block AAV2 infection. Moreover, genetic ablation of AAVR renders a wide range of mammalian cell types highly resistant to AAV2 infection. Notably, AAVR serves as a critical host factor for all tested AAV serotypes. The importance of AAVR for in vivo gene delivery is further highlighted by the robust resistance of Aavr(-/-) (also known as Au040320(-/-) and Kiaa0319l(-/-)) mice to AAV infection. Collectively, our data indicate that AAVR is a universal receptor involved in AAV infection.


Subject(s)
Dependovirus/physiology , Parvoviridae Infections/metabolism , Parvoviridae Infections/virology , Receptors, Cell Surface/metabolism , Receptors, Virus/metabolism , Viral Tropism , Animals , Antibodies/immunology , Antibodies/pharmacology , Cell Line , Dependovirus/classification , Dependovirus/drug effects , Endocytosis/drug effects , Female , Gene Deletion , Genetic Therapy/methods , Host Specificity , Humans , Male , Mice , Receptors, Cell Surface/antagonists & inhibitors , Receptors, Cell Surface/deficiency , Receptors, Cell Surface/genetics , Receptors, Virus/antagonists & inhibitors , Receptors, Virus/deficiency , Receptors, Virus/genetics , Viral Tropism/drug effects , Virus Internalization/drug effects , trans-Golgi Network/drug effects
10.
J Clin Invest ; 125(5): 2077-89, 2015 May.
Article in English | MEDLINE | ID: mdl-25893601

ABSTRACT

Multiple myeloma (MM) is an age-dependent hematological malignancy. Evaluation of immune interactions that drive MM relies on in vitro experiments that do not reflect the complex cellular stroma involved in MM pathogenesis. Here we used Vk*MYC transgenic mice, which spontaneously develop MM, and demonstrated that the immune system plays a critical role in the control of MM progression and the response to treatment. We monitored Vk*MYC mice that had been crossed with Cd226 mutant mice over a period of 3 years and found that CD226 limits spontaneous MM development. The CD226-dependent anti-myeloma immune response against transplanted Vk*MYC MM cells was mediated both by NK and CD8+ T cells through perforin and IFN-γ pathways. Moreover, CD226 expression was required for optimal antimyeloma efficacy of cyclophosphamide (CTX) and bortezomib (Btz), which are both standardly used to manage MM in patients. Activation of costimulatory receptor CD137 with mAb (4-1BB) exerted strong antimyeloma activity, while inhibition of coinhibitory receptors PD-1 and CTLA-4 had no effect. Taken together, the results of this study provide in vivo evidence that CD226 is important for MM immunosurveillance and indicate that specific immune components should be targeted for optimal MM treatment efficacy. As progressive immunosuppression associates with MM development, strategies aimed to increase immune functions may have important therapeutic implications in MM.


Subject(s)
Antigens, Differentiation, T-Lymphocyte/physiology , Immunologic Surveillance/immunology , Multiple Myeloma/immunology , Neoplasm Proteins/physiology , Animals , Antibodies, Monoclonal/therapeutic use , Antigens, Differentiation, T-Lymphocyte/genetics , Antigens, Differentiation, T-Lymphocyte/immunology , Antineoplastic Agents/therapeutic use , Boronic Acids/therapeutic use , Bortezomib , CD8-Positive T-Lymphocytes/immunology , CTLA-4 Antigen/antagonists & inhibitors , Crosses, Genetic , Cyclophosphamide/therapeutic use , Disease Progression , Genes, myc , Genetic Predisposition to Disease , Immunotherapy , Interferon-gamma/deficiency , Interferon-gamma/genetics , Interferon-gamma/physiology , Killer Cells, Natural/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Multiple Myeloma/drug therapy , Multiple Myeloma/genetics , Neoplasm Proteins/deficiency , Neoplasm Proteins/genetics , Neoplasm Proteins/immunology , Neoplasm Transplantation , Pore Forming Cytotoxic Proteins/deficiency , Pore Forming Cytotoxic Proteins/genetics , Pore Forming Cytotoxic Proteins/physiology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Pyrazines/therapeutic use , Receptors, Virus/deficiency , Receptors, Virus/genetics , Receptors, Virus/physiology , Tumor Burden , Tumor Necrosis Factor Receptor Superfamily, Member 9/antagonists & inhibitors , Tumor Necrosis Factor Receptor Superfamily, Member 9/immunology
11.
Stem Cell Res ; 11(3): 1149-62, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24012543

ABSTRACT

Diet is highly linked to breast cancer risk, yet little is known about its influence on mammary epithelial populations with distinct regenerative and hence, tumorigenic potential. To investigate this, we evaluated the relative frequency of lineage-negative CD29(hi)CD24(+), CD29(lo)CD24(+) and CD29(hi)Thy1(+)CD24(+) epithelial subpopulations in pre-neoplastic mammary tissue of adult virgin MMTV-Wnt1-transgenic mice fed either control (Casein) or soy-based diets. We found that mammary epithelial cells exposed to soy diet exhibited a lower percentage of CD29(hi)CD24(+)Lin(-) population, decreased ability to form mammospheres in culture, lower mammary outgrowth potential when transplanted into cleared fat pads, and reduced appearance of tumor-initiating CD29(hi)Thy1(+)CD24(+) cells, than in those of control diet-fed mice. Diet had no comparable influence on the percentage of the CD29(lo)CD24(+)Lin(-) population. Global gene expression profiling of the CD29(hi)CD24(+)subpopulation revealed markedly altered expression of genes important to inflammation, cytokine and chemokine signaling, and proliferation. Soy-fed relative to casein-fed mice showed lower mammary tumor incidence, shorter tumor latency, and reduced systemic levels of estradiol 17-ß, progesterone and interleukin-6. Our results provide evidence for the functional impact of diet on specific epithelial subpopulations that may relate to breast cancer risk and suggest that diet-regulated cues can be further explored for breast cancer risk assessment and prevention.


Subject(s)
CD24 Antigen/metabolism , Cytokines/genetics , Diet , Epithelial Cells/cytology , Integrin beta1/metabolism , Mammary Neoplasms, Animal/pathology , Animals , Cells, Cultured , Chemokines/genetics , Chemokines/metabolism , Cytokines/metabolism , Down-Regulation , Epithelial Cells/metabolism , Female , Humans , Male , Mammary Glands, Human/metabolism , Mammary Neoplasms, Animal/prevention & control , Mice , Mice, Transgenic , Receptors, Virus/deficiency , Receptors, Virus/genetics , Receptors, Virus/metabolism , Risk Factors , Transcriptome , Wnt1 Protein/deficiency , Wnt1 Protein/genetics , Wnt1 Protein/metabolism
12.
J Immunol ; 190(8): 4185-95, 2013 Apr 15.
Article in English | MEDLINE | ID: mdl-23487419

ABSTRACT

Liver sinusoidal endothelial cell lectin (LSECtin) was recently reported to suppress intrahepatic T cell immunity and to limit immune-mediated liver injury. However, its role in the outcome and pathogenesis of viral infection has not yet been elucidated. Using a mouse model infected with a hepatotropic adenovirus, we found that the absence of LSECtin led to a higher frequency of intrahepatic effector CTLs. These cells produced higher levels of antiviral cytokines and cytotoxic factors and exhibited an increased expression of the transcription factors T-bet and Runx3. This phenotype observed in the LSECtin-knockout cells mediated a more efficient virus-specific cytotoxicity compared with that of wild-type cells. As a consequence, LSECtin deficiency significantly accelerated liver adenovirus clearance. In contrast, LSECtin upregulation in the liver delayed viral clearance; this delayed clearance was accompanied by the downregulation of the antiviral activity of CTLs. We further constructed an immunocompetent mouse model of acute hepatitis B viral infection to demonstrate that LSECtin significantly delayed the clearance of hepatitis B virus from blood and infected hepatocytes by limiting the frequency of hepatitis B virus-specific IFN-γ-producing cells. Consistent with this function, LSECtin was upregulated in the liver of mouse models of viral hepatitis. Taken together, our results suggest that LSECtin may facilitate the reduction of liver inflammation at the cost of delaying virus clearance and that this effect might be hijacked by the virus as an escape mechanism.


Subject(s)
Hepatitis, Viral, Animal/immunology , Lectins, C-Type/physiology , Receptors, Virus/physiology , T-Lymphocytes, Cytotoxic/immunology , Adenoviridae Infections/immunology , Adenoviridae Infections/pathology , Adenoviridae Infections/virology , Animals , Disease Models, Animal , Hepatitis B/immunology , Hepatitis B/pathology , Hepatitis B/virology , Hepatitis, Viral, Animal/pathology , Hepatitis, Viral, Animal/virology , Inflammation/immunology , Inflammation/pathology , Inflammation/virology , Lectins, C-Type/deficiency , Lectins, C-Type/genetics , Mice , Receptors, Virus/deficiency , Receptors, Virus/genetics , T-Lymphocytes, Cytotoxic/pathology , T-Lymphocytes, Cytotoxic/virology
13.
Oncogene ; 32(41): 4941-9, 2013 Oct 10.
Article in English | MEDLINE | ID: mdl-23160370

ABSTRACT

Plexin C1 is a type I transmembrane receptor with intrinsic R-Ras GTPase activity, which regulates cytoskeletal remodeling and adhesion in normal human melanocytes. Melanocytes are pigment-producing cells of the epidermis, precursors for melanoma, and express high levels of Plexin C1, which is lost in melanoma in vitro and in vivo. To determine if Plexin C1 is a tumor suppressor for melanoma, we introduced Plexin C1 into a primary human melanoma cell line, and phenotypes including migration, apoptosis, proliferation and tumor growth in mice were analyzed. Complimentary studies in which Plexin C1 was silenced in human melanocytes were performed. Plexin C1 significantly inhibited migration and proliferation in melanoma, whereas in melanocytes, loss of Plexin C1 increased migration and proliferation. In mouse xenografts, Plexin C1 delayed tumor growth of melanoma at early time points, but tumors eventually escaped the suppressive effects of Plexin C1, due to Plexin C1-dependent activation of the pro-survival protein Akt. R-Ras activation stimulates melanoma migration. Plexin C1 lowered R-Ras activity in melanoma and melanocytes, consistent with inhibitory effects of Plexin C1 on migration of melanocytes and melanoma. To determine if R-Ras is expressed in melanocytic lesions in vivo, staining of tissue microarrays of nevi and melanoma were performed. R-Ras expression was highly limited in melanocytic lesions, being essentially confined to primary melanoma, and almost completely absent in nevi and metastatic melanoma. These data suggest that loss of Plexin C1 in melanoma may promote early steps in melanoma progression through suppression of migration and proliferation, but pro-survival effects of Plexin C1 ultimately abrogate the tumor suppressive effects of Plexin C1. In primary melanoma, loss of Plexin C1 may function in early steps of melanoma progression by releasing inhibition of R-Ras activation, and stimulating migration.


Subject(s)
Disease Progression , Melanoma/metabolism , Melanoma/pathology , Receptors, Virus/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Apoptosis , Carcinogenesis , Cell Line, Tumor , Cell Movement , Cell Proliferation , Enzyme Activation , Humans , Melanocytes/metabolism , Melanocytes/pathology , Mice , Receptors, Virus/deficiency , Shelterin Complex , Telomere-Binding Proteins/metabolism , Tumor Suppressor Proteins/deficiency , ras Proteins/metabolism
14.
Gut ; 62(8): 1169-78, 2013 Aug.
Article in English | MEDLINE | ID: mdl-22637699

ABSTRACT

OBJECTIVE: Adhesion molecules play an important role in tumour metastasis. The liver is a frequent target for the metastasis of several tumour types. However, virtually no liver-specific adhesion molecules have been described in terms of organ-specific metastasis. This study aimed to determine the role of liver sinusoidal endothelial cell lectin (LSECtin) in colon carcinoma metastasis to the liver. DESIGN: The role of LSECtin in colon carcinoma metastasis to the liver was determined by LSECtin knockout nude mice and anti-LSECtin antibody. LSECtin promoting the migration of LS174T and LoVo cells was determined by transwell experiment. The serum levels of soluble LSECtin in patients were elevated by ELISA. RESULTS: LSECtin was found to adhere to LS174T and LoVo colon cancer cells in vitro and in vivo. Deficiency or blocking of LSECtin significantly decreased hepatic metastases of LS174T and LoVo cells. Primary colon cancer cells from patients also exhibited remarkably low rates of hepatic metastasis in LSECtin knockout mice. LSECtin promoted the migration of LS174T and LoVo cells and increased the expression of c-Met in these cells. Serum soluble LSECtin was detected at significantly higher levels in colon cancer patients with or without hepatic metastases compared with healthy controls and was also increased in colon cancer patients with metastases compared with those without metastases. CONCLUSION: The results indicate that LSECtin plays an important role in colorectal carcinoma liver metastasis and may be a promising new target for intervention in metastasis formation.


Subject(s)
Colonic Neoplasms/metabolism , Lectins, C-Type/physiology , Liver Neoplasms/secondary , Receptors, Virus/physiology , Adult , Aged , Animals , Cell Adhesion/physiology , Cell Movement/physiology , Colonic Neoplasms/blood , Colonic Neoplasms/pathology , Female , Humans , Lectins, C-Type/blood , Lectins, C-Type/deficiency , Liver/metabolism , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Knockout , Middle Aged , Neoplasm Proteins/blood , Neoplasm Proteins/metabolism , Neoplasm Proteins/physiology , Neoplasm Transplantation , Proto-Oncogene Proteins c-met/metabolism , Receptors, Virus/deficiency , Recombinant Proteins/metabolism , Transplantation, Heterologous
15.
J Virol ; 87(1): 474-81, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23097453

ABSTRACT

Herpes simplex virus (HSV) pathogenesis in mice differs based on availability of the principal entry receptors herpesvirus entry mediator (HVEM) and nectin-1 in a manner dependent upon route of inoculation. After intravaginal or intracranial inoculation of adult mice, nectin-1 is a major mediator of neurologic disease, while the absence of either receptor attenuates disease after ocular infection. We tested the importance of receptor availability and route of infection on disease in mouse models of neonatal HSV. We infected 7-day-old mice lacking neither or one principal HSV receptor or both principal HSV receptors with HSV-2 via a peripheral route (intranasal), via a systemic route (intraperitoneal), or by inoculation directly into the central nervous system (intracranial). Mortality, neurologic disease, and visceral dissemination of virus were significantly attenuated in nectin-1 knockout mice compared with HVEM knockout or wild-type mice after intranasal inoculation. Mice lacking both entry receptors (double-knockout mice) showed no evidence of disease after inoculation by any route. Nectin-1 knockout mice had delayed mortality after intraperitoneal inoculation relative to wild-type and HVEM knockout mice, but virus was able to spread to the brain and viscera in all genotypes except double-knockout mice. Unlike in adult mice, HVEM was sufficient to mediate disease in neonatal mice after direct intracranial inoculation, and the absence of HVEM delayed time to mortality relative to that of wild-type mice. Additionally, in wild-type neonatal mice inoculated intracranially, HSV antigen did not primarily colocalize with NeuN-positive neurons. Our results suggest that differences in receptor expression between adults and newborns may partially explain differences in susceptibility to HSV-2.


Subject(s)
Cell Adhesion Molecules/metabolism , Herpes Simplex/pathology , Herpesvirus 2, Human/pathogenicity , Pregnancy Complications, Infectious/pathology , Receptors, Tumor Necrosis Factor, Member 14/metabolism , Receptors, Virus/metabolism , Animals , Cell Adhesion Molecules/deficiency , Disease Models, Animal , Female , Herpes Simplex/mortality , Herpes Simplex/virology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nectins , Pregnancy Complications, Infectious/mortality , Pregnancy Complications, Infectious/virology , Receptors, Virus/deficiency , Survival Analysis
16.
J Biol Chem ; 287(53): 44083-96, 2012 Dec 28.
Article in English | MEDLINE | ID: mdl-23144462

ABSTRACT

Aortic aneurysm is dilation of the aorta primarily due to degradation of the aortic wall extracellular matrix (ECM). Tissue inhibitors of metalloproteinases (TIMPs) inhibit matrix metalloproteinases (MMPs), the proteases that degrade the ECM. Timp3 is the only ECM-bound Timp, and its levels are altered in the aorta from patients with abdominal aortic aneurysm (AAA). We investigated the causal role of Timp3 in AAA formation. Infusion of angiotensin II (Ang II) using micro-osmotic (Alzet) pumps in Timp3(-/-) male mice, but not in wild type control mice, led to adverse remodeling of the abdominal aorta, reduced collagen and elastin proteins but not mRNA, and elevated proteolytic activities, suggesting excess protein degradation within 2 weeks that led to formation of AAA by 4 weeks. Intriguingly, despite early up-regulation of MMP2 in Timp3(-/-)Ang II aortas, additional deletion of Mmp2 in these mice (Timp3(-/-)/Mmp2(-/-)) resulted in exacerbated AAA, compromised survival due to aortic rupture, and inflammation in the abdominal aorta. Reconstitution of WT bone marrow in Timp3(-/-)/Mmp2(-/-) mice reduced inflammation and prevented AAA in these animals following Ang II infusion. Treatment with a broad spectrum MMP inhibitor (PD166793) prevented the Ang II-induced AAA in Timp3(-/-) and Timp3(-/-)/Mmp2(-/-) mice. Our study demonstrates that the regulatory function of TIMP3 is critical in preventing adverse vascular remodeling and AAA. Hence, replenishing TIMP3, a physiological inhibitor of a number of metalloproteinases, could serve as a therapeutic approach in limiting AAA development or expansion.


Subject(s)
Angiotensin II/metabolism , Aortic Aneurysm, Abdominal/metabolism , Receptors, Virus/deficiency , Angiotensin II/genetics , Animals , Aorta, Abdominal/metabolism , Aortic Aneurysm, Abdominal/genetics , Female , Hepatitis A Virus Cellular Receptor 2 , Humans , Male , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Virus/genetics
17.
Virology ; 432(1): 20-8, 2012 Oct 10.
Article in English | MEDLINE | ID: mdl-22726751

ABSTRACT

Ebolavirus causes severe hemorrhagic fever in humans and non-human primates. Entry of ebolavirus is mediated by the viral glycoprotein, GP; however, the required host factors have not been fully elucidated. A screen utilizing a recombinant Vesicular Stomatitis Virus (VSV) encoding Zaire ebolavirus GP identified four Chinese Hamster Ovary (CHO) cell lines resistant to GP-mediated viral entry. Susceptibility to vectors carrying SARS coronavirus S or VSV-G glycoproteins suggests that endocytic and processing pathways utilized by other viruses are intact in these cells. A cathepsin-activated form of the ebolaviral glycoprotein did not overcome the entry restriction, nor did expression of several host factors previously described as important for ebolavirus entry. Conversely, expression of the recently described ebolavirus host entry factor Niemann-Pick Type C1 (NPC1) restored infection. Resistant cells encode distinct mutations in the NPC1 gene, resulting in loss of protein expression. These studies reinforce the importance of NPC1 for ebolavirus entry.


Subject(s)
Ebolavirus/pathogenicity , Host-Pathogen Interactions , Membrane Glycoproteins/biosynthesis , Membrane Glycoproteins/deficiency , Receptors, Virus/biosynthesis , Receptors, Virus/deficiency , Virus Internalization , Animals , CHO Cells , Cricetinae , Cricetulus , Ebolavirus/physiology , Genetic Complementation Test , Mutation
18.
Cancer Gene Ther ; 19(2): 118-25, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22076042

ABSTRACT

Carrier cells delivering a conditionally replicating adenovirus (CRAd), which selectively replicates in tumor cells and induces tumor cell lysis, have promising potential for treatment of cancer because CRAd-loaded carrier cells evade inhibition by neutralizing anti-adenovirus (Ad) antibodies and because the carrier cells are locally retained at the injection point after local injection. A previous study by Hamada et al. demonstrated that carrier cells (CRAd-containing cell fragments derived from the carrier cells) are engulfed into the target cells, probably through a pathway independent of the primary receptor for Ad, the coxsackievirus and Ad receptor (CAR) (Mol Ther, 15: 1121-1128; 2007); however, it remains to be elucidated whether carrier cells infected with a conventional CRAd, which is composed of subgroup-C Ad serotype-5 (Ad5), mediate antitumor effects on CAR-negative cells. In order to examine whether carrier cells delivering a conventional CRAd (Carrier-F5) induce lysis of CAR-negative tumor cells, CAR-positive and CAR-negative tumor cells were incubated with Carrier-F5. Carrier-F5 mediated efficient killing of CAR-positive tumor cells; however, CAR-negative tumor cells were almost refractory to Carrier-F5. On the other hand, carrier cells loaded with a fiber-substituted CRAd containing fiber proteins of Ad serotype-35 (Ad35) (CRAd-F35), which binds to human CD46 for infection, showed efficient killing of both CAR-positive and CAR-negative tumor cells. Intra-tumoral injection of carrier cells loaded with CRAd-F35 (Carrier-F35) also resulted in efficient regression of both CAR-positive and CAR-negative tumors. These results demonstrated that the expression levels of receptors for Ad are an important factor for CRAd-loaded carrier cell-mediated cancer therapy, and that Carrier-F35 would have potential as a cancer treatment for not only CAR-positive tumors but also CAR-negative tumors.


Subject(s)
Adenocarcinoma/therapy , Adenocarcinoma/virology , Adenoviridae/physiology , Lung Neoplasms/therapy , Lung Neoplasms/virology , Oncolytic Virotherapy/methods , Receptors, Virus/deficiency , Urinary Bladder Neoplasms/therapy , Urinary Bladder Neoplasms/virology , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Adenocarcinoma of Lung , Adenoviridae/genetics , Animals , Cell Line, Tumor , Coxsackie and Adenovirus Receptor-Like Membrane Protein , Genetic Vectors , HEK293 Cells , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Membrane Cofactor Protein/biosynthesis , Mice , Mice, Inbred BALB C , Receptors, Virus/biosynthesis , Transduction, Genetic , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/metabolism , Virus Replication , Xenograft Model Antitumor Assays
19.
PLoS One ; 6(6): e20203, 2011.
Article in English | MEDLINE | ID: mdl-21674029

ABSTRACT

To determine the normal function of the Coxsackievirus and Adenovirus Receptor (CAR), a protein found in tight junctions and other intercellular complexes, we constructed a mouse line in which the CAR gene could be disrupted at any chosen time point in a broad spectrum of cell types and tissues. All knockouts examined displayed a dilated intestinal tract and atrophy of the exocrine pancreas with appearance of tubular complexes characteristic of acinar-to-ductal metaplasia. The mice also exhibited a complete atrio-ventricular block and abnormal thymopoiesis. These results demonstrate that CAR exerts important functions in the physiology of several organs in vivo.


Subject(s)
Gene Silencing , Phenotype , Receptors, Virus/deficiency , Receptors, Virus/genetics , Animals , Atrioventricular Block/genetics , Atrophy/genetics , Behavior, Animal/drug effects , Coxsackie and Adenovirus Receptor-Like Membrane Protein , Female , Fertility/drug effects , Fertility/genetics , Gene Knockout Techniques , Gene Silencing/drug effects , Intestinal Mucosa/metabolism , Intestines/drug effects , Intestines/pathology , Male , Mice , Motor Activity/drug effects , Motor Activity/genetics , Pancreas, Exocrine/drug effects , Pancreas, Exocrine/metabolism , Pancreas, Exocrine/pathology , Receptors, Virus/metabolism , Tamoxifen/pharmacology , Thymus Gland/cytology , Thymus Gland/drug effects , Thymus Gland/metabolism
20.
Hum Gene Ther ; 22(9): 1061-9, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21615297

ABSTRACT

Adenovirus (Ad)-based antiangiogenesis gene therapy is a promising approach for cancer treatment. Downregulation or loss of coxsackievirus and adenovirus receptor (CAR) is often detected in various human cancers, which hampers adenoviral gene therapy approaches. Cationic liposome-complexed adenoviral vectors have been proven useful in CAR-deficient cells to enhance therapeutic gene transfer in vivo. Here, we investigated the antitumor effects of recombinant adenovirus encoding endostatin (Ad-hE) encapsulated in cationic liposome (Ad-hE/Lipo) on CAR-deficient CT26 colon carcinoma murine models. In vitro, Ad-hE/Lipo enhanced adenovirus transfection in CAR-deficient cells (CT26), and endostatin gene expression was measured by both qualitative and quantitative detection. In addition, an antibody neutralizing assay indicated that neutralizing serum inhibited naked adenovirus 5 (Ad5) at rather higher dilution than the complexes of Ad5 and cationic liposomes (Ad5-CL), which demonstrated that Ad5-CL was more capable of protecting Ad5 from neutralization. In vivo, Ad-hE/Lipo treatment in the murine CT26 tumor model by intratumoral injection resulted in marked suppression of tumor growth and prolonged survival time, which was associated with a decreased number of microvessels and increased apoptosis of tumor cells. In conclusion, recombinant endostatin adenovirus encapsulated with cationic liposome effectively inhibited CAR-deficient tumor growth through an antiangiogenic mechanism in murine models without marked toxicity, thus showing a feasible strategy for clinical applications.


Subject(s)
Adenocarcinoma/therapy , Adenoviridae/genetics , Colonic Neoplasms/therapy , Endostatins/genetics , Genetic Therapy , Genetic Vectors/administration & dosage , Adenocarcinoma/pathology , Animals , Apoptosis/drug effects , Apoptosis/genetics , Cell Line , Colonic Neoplasms/pathology , Coxsackie and Adenovirus Receptor-Like Membrane Protein , Disease Models, Animal , Endostatins/metabolism , Female , Gene Expression Regulation , Genetic Vectors/toxicity , HEK293 Cells , Humans , Liposomes , Mice , Mice, Inbred BALB C , Neovascularization, Pathologic/genetics , Receptors, Virus/deficiency , Transduction, Genetic , Tumor Burden/drug effects , Tumor Burden/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...