Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
J Exp Med ; 217(12)2020 12 07.
Article in English | MEDLINE | ID: mdl-32880630

ABSTRACT

Intestinal barrier leakage constitutes a potential therapeutic target for many inflammatory diseases and represents a disease progression marker during chronic viral infections. However, the causes of altered gut barrier remain mostly unknown. Using murine infection with lymphocytic choriomeningitis virus, we demonstrate that, in contrast to an acute viral strain, a persistent viral isolate leads to long-term viral replication in hematopoietic and mesenchymal cells, but not epithelial cells (IECs), in the intestine. Viral persistence drove sustained intestinal epithelial barrier leakage, which was characterized by increased paracellular flux of small molecules and was associated with enhanced colitis susceptibility. Type I IFN signaling caused tight junction dysregulation in IECs, promoted gut microbiome shifts and enhanced intestinal CD8 T cell responses. Notably, both type I IFN receptor blockade and CD8 T cell depletion prevented infection-induced barrier leakage. Our study demonstrates that infection with a virus that persistently replicates in the intestinal mucosa increases epithelial barrier permeability and reveals type I IFNs and CD8 T cells as causative factors of intestinal leakage during chronic infections.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Interferon Type I/metabolism , Intestinal Mucosa/pathology , Intestinal Mucosa/virology , Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/virology , Lymphocytic choriomeningitis virus/physiology , Animals , Antibodies/pharmacology , Chronic Disease , Clostridiales/physiology , Colitis/complications , Colitis/immunology , Colitis/virology , Epithelial Cells/virology , Female , Firmicutes , Gastrointestinal Microbiome , Gene Expression Regulation , Hematopoietic Stem Cells/virology , Intestinal Mucosa/microbiology , Lymphocytic Choriomeningitis/genetics , Lymphocytic Choriomeningitis/microbiology , Mesoderm/virology , Mice, Inbred C57BL , Permeability , Signal Transduction , Tight Junction Proteins/genetics , Tight Junction Proteins/metabolism
2.
Vet Microbiol ; 248: 108801, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32827929

ABSTRACT

An oral mass was surgically excised from the gingiva of a pig. As the mass had a similar histological appearance to an equine sarcoid, DNA was extracted and consensus PCR primers used to amplify papillomavirus (PV) DNA. DNA sequences from Ovis aries papillomavirus (OaPV) type 2 were amplified both from a section of the entire mass as well as an area deeper within the mass away from the surface of the lesion. No other PV types were detected within the oral lesion. Ovis aries PV2 is a Delta PV that is closely related to the bovine Delta PVs that cause sarcoids in horses and cats. These results suggest that OaPV2 may be able to infect pigs and this virus could have caused the mesenchymal neoplasm in the mouth of this pig. This is the first evidence that a non-bovine PV can infect a non-host species and the first report of a sarcoid-like mass in pigs. These observations add to the range of species in which PV-associated neoplasia has been reported and suggest that cross-species infection by other Delta PV types may be possible.


Subject(s)
Mesoderm/virology , Mouth/pathology , Mouth/virology , Papillomaviridae/genetics , Papillomavirus Infections/veterinary , Skin Neoplasms/veterinary , Animals , Cattle , DNA Primers/genetics , Female , Mesoderm/pathology , New Zealand , Papillomavirus Infections/diagnosis , Pets/virology , Sheep/virology , Skin Neoplasms/diagnosis , Skin Neoplasms/virology , Swine
3.
PLoS One ; 9(3): e93033, 2014.
Article in English | MEDLINE | ID: mdl-24667642

ABSTRACT

Generating cardiomyocytes from embryonic stem cells is an important technique for understanding cardiovascular development, the origins of cardiovascular diseases and also for providing potential reagents for cardiac repair. Numerous methods have been published but often are technically challenging, complex, and are not easily adapted to assessment of specific gene contributions to cardiac myocyte differentiation. Here we report the development of an optimized protocol to induce the differentiation of mouse embryonic stem cells to cardiac myocytes that is simplified and easily adapted for genetic studies. Specifically, we made four critical findings that distinguish our protocol: 1) mouse embryonic stem cells cultured in media containing CHIR99021 and PD0325901 to maintain pluripotency will efficiently form embryoid bodies containing precardiac mesoderm when cultured in these factors at a reduced dosage, 2) low serum conditions promote cardiomyocyte differentiation and can be used in place of commercially prepared StemPro nutrient supplement, 3) the Wnt inhibitor Dkk-1 is dispensable for efficient cardiac differentiation and 4) tracking differentiation efficiency may be done with surface expression of PDGFRα alone. In addition, cardiac mesodermal precursors generated by this system can undergo lentiviral infection to manipulate the expression of specific target molecules to assess effects on cardiac myocyte differentiation and maturation. Using this approach, we assessed the effects of CHF1/Hey2 on cardiac myocyte differentiation, using both gain and loss of function. Overexpression of CHF1/Hey2 at the cardiac mesoderm stage had no apparent effect on cardiac differentiation, while knockdown of CHF1/Hey2 resulted in increased expression of atrial natriuretic factor and connexin 43, suggesting an alteration in the phenotype of the cardiomyocytes. In summary we have generated a detailed and simplified protocol for generating cardiomyocytes from mES cells that is optimized for investigating factors that affect cardiac differentiation.


Subject(s)
Cell Culture Techniques/methods , Cell Differentiation , Embryonic Stem Cells/cytology , Myocytes, Cardiac/cytology , Animals , Basic Helix-Loop-Helix Transcription Factors/deficiency , Basic Helix-Loop-Helix Transcription Factors/genetics , Bone Morphogenetic Protein 4/metabolism , Cell Survival , Embryoid Bodies/cytology , Embryonic Stem Cells/metabolism , Gene Expression Regulation , Gene Knockdown Techniques , HEK293 Cells , Homeobox Protein Nkx-2.5 , Homeodomain Proteins/metabolism , Humans , Lentivirus/physiology , Mesoderm/cytology , Mesoderm/virology , Mice , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Repressor Proteins/deficiency , Repressor Proteins/genetics , Serum/metabolism , Transcription Factors/metabolism
4.
Vet Pathol ; 50(4): 715-21, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23051916

ABSTRACT

Opportunistic viral infections are common in simian immunodeficiency virus-infected rhesus macaques and include simian polyomavirus 40 (SV40), which causes interstitial nephritis, pneumonia, meningoencephalitis, and progressive multifocal leukoencephalopathy and rhesus cytomegalovirus (Macacine herpesvirus-3), which is associated with many pathologic manifestations, including the formation of neutrophil-rich gastrointestinal masses. Herein we report the findings of a simian immunodeficiency virus-infected rhesus macaque that presented to necropsy with multiple nodular masses restricted to the proximal jejunum. Histologically, the masses within the lamina propria were composed of abundant, loosely organized, mesenchymal tissue forming broad interlacing whorls and sheets admixed with variable numbers of neutrophils. Cells within the mesenchymoproliferative nodules contained numerous basophilic, intranuclear inclusion bodies with only scattered cytomegalic cells. Immunohistochemistry for rhesus cytomegalovirus and SV40 demonstrated variable numbers of immunopositive cells within the affected nodules. This report is the first description of SV40-associated pathology in the small intestine of a rhesus macaque and highlights the role that opportunistic viral infections can have on gastrointestinal pathology in immunosuppressed rhesus macaques.


Subject(s)
Cytomegalovirus Infections/veterinary , Macaca mulatta , Monkey Diseases/pathology , Polyomavirus Infections/veterinary , Simian virus 40/isolation & purification , Tumor Virus Infections/veterinary , Animals , Cell Proliferation , Cytomegalovirus/isolation & purification , Cytomegalovirus Infections/complications , Cytomegalovirus Infections/pathology , Cytomegalovirus Infections/virology , Immunocompromised Host , Immunohistochemistry , Intestine, Small/pathology , Intestine, Small/virology , Jejunum/pathology , Jejunum/virology , Mesoderm/pathology , Mesoderm/virology , Monkey Diseases/virology , Mucous Membrane/pathology , Mucous Membrane/virology , Opportunistic Infections/complications , Opportunistic Infections/pathology , Opportunistic Infections/veterinary , Opportunistic Infections/virology , Polyomavirus Infections/complications , Polyomavirus Infections/pathology , Polyomavirus Infections/virology , Simian Acquired Immunodeficiency Syndrome/complications , Simian Acquired Immunodeficiency Syndrome/pathology , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/physiology , Tumor Virus Infections/complications , Tumor Virus Infections/pathology , Tumor Virus Infections/virology
5.
Vet Pathol ; 49(6): 941-9, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22411549

ABSTRACT

Recently, several cases of human cowpox virus (CPXV) infections were reported in France and Germany, which had been acquired through close contact with infected pet rats. The animals exhibited respiratory signs or skin lesions and died shortly after purchase. After natural infection of white rats with CPXV in the USSR in 1978, a peracute pulmonary form, a milder dermal form, and a mixed form exhibiting features of both have been described. To the best of the authors' knowledge, 3 experimental cowpox virus infection studies using rats have been performed to date; however, neither results of histomorphological examinations nor immunohistochemical analyses have yet been reported in rats after experimental infections. To investigate the impact of the infection route on the clinical course, the development of lesions, and tropism, rats were infected intradermally, intranasally, or by a combination of both routes. The authors found a correlation between clinical manifestation, pathology, and infection routes. Intradermal and contact exposure yielded a mild dermal form, characterized by the development of vesiculopustular dermatitis. In contrast, intranasally infected animals died peracutely, showing severe dyspnea. Occasionally, a combination of the dermal and the respiratory form occurred after intranasal infection. Immunohistochemically, CPXV antigen was detected in the epithelial and mesenchymal cells of the upper respiratory tract and affected skin lesions and rarely in mesenchymal cells of lymph nodes. This is the first histomorphological and immunohistochemical analysis of CPXV in rats after experimental infection.


Subject(s)
Cowpox virus/physiology , Cowpox/pathology , Respiratory Tract Infections/pathology , Animals , Antigens, Viral/analysis , Cowpox/virology , Cowpox virus/immunology , Cowpox virus/pathogenicity , Disease Models, Animal , Epithelial Cells/pathology , Epithelial Cells/virology , Female , Humans , Immunohistochemistry , Inclusion Bodies, Viral/metabolism , Lung/pathology , Lung/virology , Male , Mesoderm/pathology , Mesoderm/virology , Nasal Cavity/virology , Rats , Rats, Wistar , Respiratory Tract Infections/virology , Skin/virology , Viral Tropism
6.
Am J Physiol Lung Cell Mol Physiol ; 300(2): L274-85, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21036917

ABSTRACT

Although γherpesvirus infections are associated with enhanced lung fibrosis in both clinical and animal studies, there is limited understanding about fibrotic effects of γherpesviruses on cell types present in the lung, particularly during latent infection. Wild-type mice were intranasally infected with a murine γherpesvirus (γHV-68) or mock-infected with saline. Twenty-eight days postinfection (dpi), ∼14 days following clearance of the lytic infection, alveolar macrophages (AMs), mesenchymal cells, and CD19-enriched cell populations from the lung and spleen express M(3) and/or glycoprotein B (gB) viral mRNA and harbor viral genome. AMs from infected mice express more transforming growth factor (TGF)-ß(1), CCL2, CCL12, TNF-α, and IFN-γ than AMs from mock-infected mice. Mesenchymal cells express more total TGF-ß(1), CCL12, and TNF-α than mesenchymal cells from mock-infected mice. Lung and spleen CD19-enriched cells express more total TGF-ß(1) 28 dpi compared with controls. The CD19-negative fraction of the spleen overexpresses TGF-ß(1) and harbors viral genome, but this likely represents infection of monocytes. Purified T cells from the lung harbor almost no viral genome. Purified T cells overexpress IL-10 but not TGF-ß(1). Intracellular cytokine staining demonstrated that lung T cells at 28 dpi produce IFN-γ but not IL-4. Thus infection with a murine γherpesvirus is sufficient to upregulate profibrotic and proinflammatory factors in a variety of lung resident and circulating cell types 28 dpi. Our results provide new information about possible contributions of these cells to fibrogenesis in the lungs of individuals harboring a γherpesvirus infection and may help explain why γHV-68 infection can augment or exacerbate fibrotic responses in mice.


Subject(s)
Cytokines/biosynthesis , Herpesviridae Infections/immunology , Pulmonary Fibrosis/etiology , Rhadinovirus/pathogenicity , Tumor Virus Infections/immunology , Animals , Base Sequence , Chemokine CCL2/biosynthesis , DNA Primers/genetics , DNA, Viral/genetics , Disease Models, Animal , Herpesviridae Infections/genetics , Herpesviridae Infections/virology , Interferon-gamma/biosynthesis , Interleukin-10/biosynthesis , Lung/immunology , Lung/virology , Macrophage Activation , Macrophages, Alveolar/immunology , Macrophages, Alveolar/virology , Male , Mesoderm/immunology , Mesoderm/virology , Mice , Mice, Inbred C57BL , Monocyte Chemoattractant Proteins/biosynthesis , Pulmonary Fibrosis/genetics , Pulmonary Fibrosis/immunology , Pulmonary Fibrosis/virology , Spleen/immunology , Spleen/virology , T-Lymphocytes/immunology , T-Lymphocytes/virology , Transforming Growth Factor beta1/biosynthesis , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Virus Infections/genetics , Tumor Virus Infections/virology , Viral Load
7.
PLoS Pathog ; 6(6): e1000940, 2010 Jun 03.
Article in English | MEDLINE | ID: mdl-20532215

ABSTRACT

It has been recently reported that a side population of cells in nasopharyngeal carcinoma (NPC) displayed characteristics of stem-like cancer cells. However, the molecular mechanisms underlying the modulation of such stem-like cell populations in NPC remain unclear. Epstein-Barr virus was the first identified human tumor virus to be associated with various malignancies, most notably NPC. LMP2A, the Epstein-Barr virus encoded latent protein, has been reported to play roles in oncogenic processes. We report by immunostaining in our current study that LMP2A is overexpressed in 57.6% of the nasopharyngeal carcinoma tumors sampled and is mainly localized at the tumor invasive front. We found also in NPC cells that the exogenous expression of LMP2A greatly increases their invasive/migratory ability, induces epithelial-mesenchymal transition (EMT)-like cellular marker alterations, and stimulates stem cell side populations and the expression of stem cell markers. In addition, LMP2A enhances the transforming ability of cancer cells in both colony formation and soft agar assays, as well as the self-renewal ability of stem-like cancer cells in a spherical culture assay. Additionally, LMP2A increases the number of cancer initiating cells in a xenograft tumor formation assay. More importantly, the endogenous expression of LMP2A positively correlates with the expression of ABCG2 in NPC samples. Finally, we demonstrate that Akt inhibitor (V) greatly decreases the size of the stem cell side populations in LMP2A-expressing cells. Taken together, our data indicate that LMP2A induces EMT and stem-like cell self-renewal in NPC, suggesting a novel mechanism by which Epstein-Barr virus induces the initiation, metastasis and recurrence of NPC.


Subject(s)
Herpesvirus 4, Human/genetics , Mesoderm/pathology , Nasopharyngeal Neoplasms/pathology , Nasopharyngeal Neoplasms/virology , Neoplastic Stem Cells/pathology , Neoplastic Stem Cells/virology , Viral Matrix Proteins/metabolism , Animals , Biomarkers, Tumor , Blotting, Western , Case-Control Studies , Cell Adhesion , Cell Movement , Cell Transformation, Neoplastic , Colony-Forming Units Assay , Epithelial Cells/pathology , Epithelial Cells/virology , Epstein-Barr Virus Infections/metabolism , Epstein-Barr Virus Infections/virology , Flow Cytometry , Fluorescent Antibody Technique , Herpesvirus 4, Human/isolation & purification , Humans , Mesoderm/virology , Mice , Mice, Nude , Nasopharyngeal Neoplasms/metabolism , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplastic Stem Cells/metabolism , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Survival Rate , Viral Matrix Proteins/genetics
8.
J Biol Chem ; 285(27): 20882-90, 2010 Jul 02.
Article in English | MEDLINE | ID: mdl-20435897

ABSTRACT

Measles virus (MV), an enveloped negative-strand RNA virus, remains a major cause of morbidity and mortality in developing countries. MV predominantly infects immune cells by using signaling lymphocyte activation molecule (SLAM; also called CD150) as a receptor, but it also infects polarized epithelial cells, forming tight junctions in a SLAM-independent manner. Although the ability of MV to infect polarized epithelial cells is thought to be important for its transmission, the epithelial cell receptor for MV has not been identified. A transcriptional repressor, Snail, induces epithelial-mesenchymal transition (EMT), in which epithelial cells lose epithelial cell phenotypes, such as adherens and tight junctions. In this study, EMT was induced by expressing Snail in a lung adenocarcinoma cell line, II-18, which is highly susceptible to wild-type MV. Snail-expressing II-18 cells lost adherens and tight junctions. Microarray analysis confirmed the induction of EMT in II-18 cells and suggested a novel function of Snail in protein degradation and distribution. Importantly, wild-type MV no longer entered EMT-induced II-18 cells, suggesting that the epithelial cell receptor is down-regulated by the induction of EMT. Other polarized cell lines, NCI-H358 and HT-29, also lost susceptibility to wild-type MV when EMT was induced. However, the complete formation of tight junctions rather reduced MV entry into HT-29 cells. Taken together, these data suggest that the unidentified epithelial cell receptor for MV is involved in the formation of epithelial intercellular junctions.


Subject(s)
Epithelial Cells/cytology , Measles virus/pathogenicity , Measles/prevention & control , Mesoderm/cytology , Animals , Cell Line , Disease Susceptibility , Epithelial Cells/physiology , Epithelial Cells/virology , Flow Cytometry , Genetic Predisposition to Disease , Genetic Vectors , Genome, Viral , Humans , Lymphocyte Activation , Macaca mulatta/virology , Measles/immunology , Measles/transmission , Measles/veterinary , Measles virus/genetics , Membrane Cofactor Protein/physiology , Mesoderm/physiology , Mesoderm/virology , Oligonucleotide Array Sequence Analysis , Plasmids , Receptors, Virus/physiology , Tight Junctions/physiology , Virus Shedding
9.
Mol Vis ; 15: 2821-8, 2009 Dec 19.
Article in English | MEDLINE | ID: mdl-20029647

ABSTRACT

PURPOSE: Congenital eye malformations are a leading cause of blindness in children. Influenza virus infections prevail worldwide and have been implicated in congenital defects. Infections acquired during gestation may disrupt eye morphogenesis. We investigated the effects of influenza B virus infection on eye malformations during early embryogenesis. METHODS: Chick embryos were exposed to influenza B virus at Hamburger-Hamilton stage 9. Maternal infection was conducted by exposing pregnant ICR mice to influenza B virus at the embryonic gestation stage E 5.0. After infection, virus RNA distribution was detected by in situ hybridization at various developmental stages. The distribution of periocular neural crest cells and the extent of apoptosis were examined by immunohistochemical staining, in correlation with eye malformations. RESULTS: Microphthalmos and anophthalmos, together with neural tube defects, were found in the chick and mouse embryos following the infections. The viral RNA was detected in the head neuroepithelium, optic vesicle, periocular mesenchyme, and the forming ventricles of the developing brain. Immunohistochemical staining revealed aberrant neural crest distribution and extensive apoptosis in the head surface ectoderm, periocular mesenchyme, and optic vesicle in the chick embryos. Furthermore, transplacental infection was confirmed by the presence of viral RNA in the mouse fetuses, with eye and neural tube defects similar to those found in the chick embryos after experimental infections. CONCLUSIONS: Influenza B virus may act as a teratogen to cause aberrant periocular neural crest cell contribution to eye development and extensive apoptosis, resulting in congenital eye malformations.


Subject(s)
Apoptosis , Embryonic Development , Eye Abnormalities/embryology , Eye Abnormalities/virology , Influenza B virus/physiology , Neural Crest/pathology , Orthomyxoviridae Infections/virology , Animals , Chick Embryo , Disease Models, Animal , Embryo, Mammalian/abnormalities , Embryo, Mammalian/pathology , Embryo, Mammalian/virology , Eye/embryology , Eye/pathology , Eye/virology , Eye Abnormalities/pathology , Female , Maternal-Fetal Exchange , Mesoderm/pathology , Mesoderm/virology , Mice , Neural Crest/embryology , Neural Crest/virology , Orthomyxoviridae Infections/embryology , Pregnancy , RNA Transport , RNA, Viral/metabolism
10.
J Biomed Sci ; 16: 105, 2009 Nov 24.
Article in English | MEDLINE | ID: mdl-19930697

ABSTRACT

Epithelial-mesenchymal transition (EMT) is an important process in tumor metastasis. The EMT-related events associated with metastasis of NPC in the absence of EBV have not been elucidated. We established an EBV-negative NPC cell line from a bone marrow biopsy of an NPC patient. Using a Matrigel system we isolated an invasive and non-invasive sublines, designated NPC-BM29 and NPC-BM00. NPC-BM29 acquired an invasive-like phenotype characterized by EMT, marked by down-regulation of E-cadherin and beta-catenin with concomitant increased expression of Ets1. NPC-BM29 cells expressed >or= 10-fold higher of MMP-9 than NPC-BM00 cells. NPC-BM29 cells grew better in 2% serum than NPC-BM00 cells, with a population doubling-time of 26.8 h and 30.7 h, respectively. A marked reduction in colony-formation ability of NPC-BM00 cells compared to NPC-BM29 was observed. Wound-healing assay revealed that NPC-BM29 cells displayed higher motility than NPC-BM00 and the motility was further enhanced by cell treatment with TPA, a PKC activator. Cell surface markers and tumor-associated molecules, AE3, MAK6 and sialyl-Tn, were up-regulated in NPC-BM29 cells, whereas the expression of HLA-DR and CD54 was significantly increased in NPC-BM00 cells. NPC-BM29 consistently released higher levels of IL-8 and IL-10 than NPC-BM00, with low levels of IL-1alpha expression in both cell lines. Higher level of VEGF production was detected in NPC-BM00 than NPC-BM29 cells. These data show that EBV is not required for exhibiting multiple metastatic phenotypes associated with EMT. More studies that target right molecules/signalings associated with the EMT may offer new therapeutic intervention options for NPC invasion and metastasis.


Subject(s)
Carcinoma/virology , Genome, Viral , Herpesvirus 4, Human/genetics , Nasopharyngeal Neoplasms/virology , Biopsy , Bone Marrow Cells/pathology , Bone Marrow Cells/virology , Cell Adhesion , Cell Line, Tumor , Cell Proliferation , Epithelium/virology , Humans , Mesoderm/virology , Neoplasm Metastasis , Phenotype , Wound Healing
11.
Exp Cell Res ; 315(11): 1819-31, 2009 Jul 01.
Article in English | MEDLINE | ID: mdl-19361498

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a refractory and lethal interstitial lung disease characterized by alveolar epithelial cells apoptosis, fibroblast proliferation and extra-cellular matrix protein deposition. EBV, localised to alveolar epithelial cells of pulmonary fibrosis patients is associated with a poor prognosis. A strategy based on microarray-differential gene expression analysis to identify molecular drivers of EBV-associated lung fibrosis was utilized. Alveolar epithelial cells were infected with EBV to identify genes whose expression was altered following TGFbeta1-mediated lytic phase. EBV lytic reactivation by TGFbeta1 drives a selective alteration in CUX1 variant (a) (NCBI accession number NM_181552) expression, inducing activation of non-canonical Wnt pathway mediators, implicating it in Epithelial Mesenchymal Transition (EMT), the molecular event underpinning scar production in tissue fibrosis. The role of EBV in EMT can be attenuated by antiviral strategies and inhibition of Wnt signaling by using All-Trans Retinoic Acids (ATRA). Activation of non-canonical Wnt signaling pathway by EBV in epithelial cells suggests a novel mechanism of EMT via CUX1 signaling. These data present a framework for further description of the link between infectious agents and fibrosis, a significant disease burden.


Subject(s)
Herpesvirus 4, Human/pathogenicity , Homeodomain Proteins/metabolism , Nuclear Proteins/metabolism , Pulmonary Alveoli/metabolism , Pulmonary Alveoli/virology , Repressor Proteins/metabolism , Wnt Proteins/metabolism , Adult , Aged , Antiviral Agents/pharmacology , Base Sequence , Cells, Cultured , DNA Primers/genetics , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Epithelial Cells/virology , Epstein-Barr Virus Infections/complications , Female , Gene Expression/drug effects , Herpesvirus 4, Human/drug effects , Homeodomain Proteins/genetics , Host-Pathogen Interactions , Humans , Idiopathic Pulmonary Fibrosis/etiology , Idiopathic Pulmonary Fibrosis/metabolism , Idiopathic Pulmonary Fibrosis/pathology , Idiopathic Pulmonary Fibrosis/virology , Male , Mesoderm/drug effects , Mesoderm/metabolism , Mesoderm/pathology , Mesoderm/virology , Middle Aged , Nuclear Proteins/genetics , Promoter Regions, Genetic , Pulmonary Alveoli/drug effects , Pulmonary Alveoli/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , Repressor Proteins/genetics , Signal Transduction/drug effects , Transcription Factors , Transforming Growth Factor beta1/pharmacology , Wnt Proteins/antagonists & inhibitors , Wnt Proteins/genetics
12.
Biochem Biophys Res Commun ; 382(3): 555-60, 2009 May 08.
Article in English | MEDLINE | ID: mdl-19302982

ABSTRACT

The relationships between epithelial-to-mesenchymal transition (EMT), hepatitis B virus X protein (HBx), and the non-receptor tyrosine kinase c-Src were investigated. The HBx gene transfected SMMC-7721 cells underwent morphological changes from a classic epithelial morphology to a spindle-like shape. The HBx transfection increased the invasive potential of these cells. When the transfected cells were exposed to the c-Src kinase inhibitor PP2, the cells recovered their original epithelial morphology and the mRNA and protein expression of epithelial and mesenchymal markers returned to the parental cell levels. Our data suggest that activated c-Src played a critical role in the HBx-induced EMT of SMMC-7721 cells.


Subject(s)
Carcinoma, Hepatocellular/pathology , Hepatitis B virus/metabolism , Liver Neoplasms/pathology , Trans-Activators/metabolism , Viral Regulatory and Accessory Proteins/metabolism , src-Family Kinases/metabolism , Carcinoma, Hepatocellular/enzymology , Carcinoma, Hepatocellular/virology , Cell Line, Tumor , Epithelium/enzymology , Epithelium/pathology , Epithelium/virology , Humans , Liver Neoplasms/enzymology , Liver Neoplasms/virology , Mesoderm/enzymology , Mesoderm/pathology , Mesoderm/virology , Neoplasm Invasiveness , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Trans-Activators/genetics , Viral Regulatory and Accessory Proteins/genetics
13.
Hum Pathol ; 39(4): 473-83, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18342658

ABSTRACT

Epstein-Barr virus (EBV) is a ubiquitous human pathogen that usually maintains a harmonious relationship with its host. Rarely, this host-virus balance is perturbed, causing a diverse group of malignancies in both immunocompetent and immunosuppressed patients. In addition to its role in hematologic malignancies (Burkitt lymphoma, subsets of Hodgkin and T-cell lymphomas, posttransplant lymphomas), EBV has been implicated in both epithelial (undifferentiated nasopharyngeal carcinoma, a subset of gastric adenocarcinomas) and mesenchymal (EBV-associated smooth muscle tumor, inflammatory pseudotumor-like follicular dendritic cell tumor) neoplasms. This review will focus on EBV-associated epithelial and mesenchymal neoplasms.


Subject(s)
Carcinoma/pathology , Carcinoma/virology , Epstein-Barr Virus Infections/pathology , Mesoderm/pathology , Mesoderm/virology , Epithelial Cells/virology , Gene Expression , Herpesvirus 4, Human/genetics , Herpesvirus 4, Human/physiology , Humans
14.
BMC Dev Biol ; 6: 42, 2006 Sep 07.
Article in English | MEDLINE | ID: mdl-16959038

ABSTRACT

BACKGROUND: Human studies suggest, and mouse models clearly demonstrate, that cytomegalovirus (CMV) is dysmorphic to early organ and tissue development. CMV has a particular tropism for embryonic salivary gland and other head mesenchyme. CMV has evolved to co-opt cell signaling networks so to optimize replication and survival, to the detriment of infected tissues. It has been postulated that mesenchymal infection is the critical step in disrupting organogenesis. If so, organogenesis dependent on epithelial-mesenchymal interactions would be particularly vulnerable. In this study, we chose to model the vulnerability by investigating the cell and molecular pathogenesis of CMV infected mouse embryonic submandibular salivary glands (SMGs). RESULTS: We infected E15 SMG explants with mouse CMV (mCMV). Active infection for up to 12 days in vitro results in a remarkable cell and molecular pathology characterized by atypical ductal epithelial hyperplasia, apparent epitheliomesenchymal transformation, oncocytic-like stromal metaplasia, beta-catenin nuclear localization, and upregulation of Nfkb2, Relb, Il6, Stat3, and Cox2. Rescue with an antiviral nucleoside analogue indicates that mCMV replication is necessary to initiate and maintain SMG dysmorphogenesis. CONCLUSION: mCMV infection of embryonic mouse explants results in dysplasia, metaplasia, and, possibly, anaplasia. The molecular pathogenesis appears to center around the activation of canonical and, perhaps more importantly, noncanonical NFkappaB. Further, COX-2 and IL-6 are important downstream effectors of embryopathology. At the cellular level, there appears to be a consequential interplay between the transformed SMG cells and the surrounding extracellular matrix, resulting in the nuclear translocation of beta-catenin. From these studies, a tentative framework has emerged within which additional studies may be planned and performed.


Subject(s)
Cytomegalovirus Infections/pathology , Cytomegalovirus , Disease Models, Animal , Mesoderm/pathology , Submandibular Gland/embryology , Submandibular Gland/pathology , Animals , Cells, Cultured , Cytomegalovirus Infections/virology , Female , Mesoderm/virology , Mice , Pregnancy , Salivary Glands/embryology , Salivary Glands/pathology , Salivary Glands/virology , Submandibular Gland/virology , Time Factors
15.
Eur Respir J ; 28(6): 1106-16, 2006 Dec.
Article in English | MEDLINE | ID: mdl-16899483

ABSTRACT

Epithelial-mesenchymal transformation is now recognised as an important feature of tissue remodelling. The present report concerns the role of adenovirus infection in inducing this transformation in an animal model of chronic obstructive pulmonary disease. Guinea pig primary peripheral lung epithelial cells (PLECs) transfected with adenovirus E1A (E1A-PLECs) were compared to guinea pig normal lung fibroblasts (NLFs) transfected with E1A (E1A-NLFs). These cells were characterised by PCR, immunocytochemistry, electron microscopy, and Western and Northern blot analyses. Electrophoretic mobility shift assays were performed in order to examine nuclear factor (NF)-kappaB and activator protein (AP)-1 binding activities. E1A-PLECs and E1A-NLFs positive for E1A DNA, mRNA and protein expressed cytokeratin and vimentin but not smooth muscle alpha-actin. Both exhibited cuboidal morphology and junctional complexes, but did not contain lamellar bodies or express surfactant protein A, B or C mRNAs. These two cell types differed, however, in their NF-kappaB and AP-1 binding after lipopolysaccharide stimulation, possibly due to differences in the expression of the subunits that comprise these transcriptional complexes. E1A transfection results in the transformation of peripheral lung epithelial cells and normal lung fibroblasts to a phenotype intermediate between that of the two primary cells. It is postulated that this intermediate phenotype may play a major role in the remodelling of the airways in chronic obstructive pulmonary disease associated with persistence of adenovirus E1A DNA.


Subject(s)
Adenovirus E1A Proteins/physiology , Fibroblasts/virology , Lung/virology , Mesoderm/metabolism , Actins/metabolism , Animals , Binding Sites , Blotting, Northern , Blotting, Western , Cell Transformation, Viral , Cells, Cultured , DNA, Viral , Electrophoretic Mobility Shift Assay , Epithelial Cells/metabolism , Epithelial Cells/virology , Female , Fibroblasts/cytology , Fibroblasts/metabolism , Guinea Pigs , Immunoenzyme Techniques , Keratins/metabolism , Lung/cytology , Lung/metabolism , Mesoderm/cytology , Mesoderm/virology , Microscopy, Electron , Muscle, Smooth/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , Phenotype , Polymerase Chain Reaction , Pulmonary Surfactant-Associated Protein A , RNA, Messenger , RNA, Viral , Transcription Factor AP-1 , Transfection , Vimentin/metabolism
16.
Hum Gene Ther ; 8(12): 1417-27, 1997 Aug 10.
Article in English | MEDLINE | ID: mdl-9287142

ABSTRACT

Human marrow-derived mesenchymal progenitor cells (hMPCs), which have the capacity for osteogenic and marrow stromal differentiation, were transduced with the myeloproliferative sarcoma virus (MPSV)-based retrovirus, vM5LacZ, that contains the LacZ and neo genes. Stable transduction and gene expression occurred in 18% of cells. After culture expansion and selection in G418, approximately 70% of neo(r) hMPCs co-expressed LacZ. G418-selected hMPC retain their osteogenic potential and form bone in vivo when seeded into porous calcium phosphate ceramic cubes implanted subcutaneously into SCID mice. LacZ expression was evident within osteoblasts and osteocytes in bone developing within the ceramics 6 and 9 weeks after implantation. Likewise, hMPCs transduced with human interleukin-3 (hIL-3) cDNA, adhered to ceramic cubes and implanted into SCID mice, formed bone and secreted detectable levels of hIL-3 into the systemic circulation for at least 12 weeks. These data indicate that genetically transduced, culture-expanded bone marrow-derived hMPCs retain a precursor phenotype and maintain similar levels of transgene expression during osteogenic lineage commitment and differentiation in vivo. Because MPCs have been shown to differentiate into bone, cartilage, and tendon, these cells may be a useful target for gene therapy.


Subject(s)
Bone Marrow Cells , Bone Marrow/virology , Cell Transplantation/methods , Interleukin-3/genetics , Retroviridae/genetics , beta-Galactosidase/genetics , Adult , Animals , Bone and Bones/cytology , Bone and Bones/physiology , Cell Differentiation , Cells, Cultured , Ceramics , Gene Expression Regulation , Humans , Interleukin-3/metabolism , Interleukin-3/pharmacology , Mesoderm/cytology , Mesoderm/virology , Mice , Mice, SCID , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Stem Cells/virology , Transduction, Genetic , beta-Galactosidase/metabolism
17.
Article in English | MEDLINE | ID: mdl-8850485

ABSTRACT

Four HIV-positive patients with herpes simplex virus and cytomegalovirus coinfected oral ulcers are presented. All patients had persistent oral pain associated with nonhealing mucosal ulcers. Lesions occurred on the palate, retromolar pad, tongue, and lip, and the clinical appearance of the ulcers was nonspecific. Histologic and immunohistochemical stains showed herpes simples virus alterations in keratinocyte nuclei and cytomegalovirus alterations in mesenchymal/endothelial cell nuclei and cytoplasm. Lesions in one patient responded to ganciclovir therapy. One patient improved with acyclovir, and another healed normally after excisional biopsy. Each virus alone has been described as causing oral ulcerations; their appearance together in the same lesion would suggest a synergistic relationship.


Subject(s)
AIDS-Related Opportunistic Infections/pathology , Cytomegalovirus Infections/pathology , HIV Seropositivity/pathology , Mouth Diseases/virology , Stomatitis, Herpetic/pathology , Acyclovir/therapeutic use , Adult , Antiviral Agents/therapeutic use , Cell Nucleus/virology , Cytomegalovirus/ultrastructure , Cytoplasm/virology , Endothelium, Vascular/virology , Follow-Up Studies , Ganciclovir/therapeutic use , Gingival Diseases/pathology , Gingival Diseases/virology , Humans , Immunohistochemistry , Keratinocytes/virology , Lip Diseases/pathology , Lip Diseases/virology , Male , Mesoderm/virology , Middle Aged , Mouth Diseases/pathology , Palate , Simplexvirus/ultrastructure , Tongue Diseases/pathology , Tongue Diseases/virology , Ulcer/pathology , Ulcer/virology
SELECTION OF CITATIONS
SEARCH DETAIL
...