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










Publication year range
1.
Proc Natl Acad Sci U S A ; 112(32): 9914-9, 2015 Aug 11.
Article in English | MEDLINE | ID: mdl-26221020

ABSTRACT

Intracerebral hemorrhagic stroke and vascular dementia are age- and hypertension-associated manifestations of human cerebral small vessel disease (SVD). Cerebral microvessels are formed by endothelial cells (ECs), which are connected through tight junctions, adherens junctions, and stabilizing basement membrane structures. These endothelial connections ensure both vessel stability and blood-brain barrier (BBB) functions, the latter enabling selective exchange of ions, bioactive molecules, and cells between the bloodstream and brain tissue. Srf(iECKO) mice, permitting conditional EC-specific depletion of the transcription factor Serum Response Factor (SRF), suffer from loss of BBB integrity and intracerebral hemorrhaging. Cerebral microbleeds and larger hemorrhages developed upon postnatal and adult depletion of either SRF or its cofactors Myocardin Related Transcription Factor (MRTF-A/-B), revealing essential requirements of ongoing SRF/MRTF activity for maintenance of cerebral small vessel integrity. In vivo magnetic resonance imaging allowed detection, localization, and time-resolved quantification of BBB permeability and hemorrhage formation in Srf(iECKO) brains. At the molecular level, direct and indirect SRF/MRTF target genes, encoding structural components of tight junctions (Claudins and ZO proteins), adherens junctions (VE-cadherin, α-Actinin), and the basement membrane (Collagen IV), were down-regulated upon SRF depletion. These results identify SRF and its MRTF cofactors as major transcriptional regulators of EC junctional stability, guaranteeing physiological functions of the cerebral microvasculature. We hypothesize that impairments in SRF/MRTF activity contribute to human SVD pathology.


Subject(s)
Cerebral Hemorrhage/complications , Endothelial Cells/metabolism , Serum Response Factor/metabolism , Stroke/etiology , Stroke/metabolism , Trans-Activators/metabolism , Transcription Factors/metabolism , Animals , Animals, Newborn , Astrocytes/metabolism , Astrocytes/pathology , Basement Membrane/metabolism , Basement Membrane/pathology , Blood-Brain Barrier/metabolism , Brain/blood supply , Brain/metabolism , Brain/pathology , Brain/physiopathology , Cadherins/metabolism , Cerebral Hemorrhage/metabolism , Cerebral Hemorrhage/pathology , Cerebral Hemorrhage/physiopathology , Collagen Type IV/metabolism , Down-Regulation , Evans Blue/metabolism , Exploratory Behavior , Extravasation of Diagnostic and Therapeutic Materials , Gene Deletion , Magnetic Resonance Imaging , Mice, Knockout , Microvessels/metabolism , Microvessels/pathology , Motor Activity , Permeability , Serum Response Factor/genetics , Stroke/pathology , Stroke/physiopathology , Tight Junctions/metabolism , Time Factors
2.
Growth Factors ; 29(6): 298-309, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22023218

ABSTRACT

This study aimed to investigate the tissue-specific role of the insulin-like growth factor 1 (IGF-I) on glucose homeostasis in the high-fatness selected Berlin Fat Mouse Inbred (BFMI) line. Therefore, the expression of different IGF-I transcripts and IGF-I protein, IGF-binding proteins, insulin as well as glucose tolerance was analyzed in BFMI in comparison with that in lean mice. In addition, dietary effects were investigated. The BFMI line showed normal blood glucose clearance on standard diet, but on high-fat diet the clearance was impaired, indicating the beginning of insulin resistance. Circulating IGF-I and insulin levels were elevated in BFMI than in lean mice on both diets along with a down-regulation of three IGF-I binding proteins in BFMI mice. Serum IGF-I levels corresponded with the expression pattern for both hepatic and one class II splice variants in reproductive adipose tissue, but not in muscle. High insulin and high IGF-I levels likely prevent BFMI mice from diabetes.


Subject(s)
Glucose/metabolism , Insulin-Like Growth Factor Binding Proteins/metabolism , Insulin-Like Growth Factor I/metabolism , Obesity/metabolism , Adipose Tissue , Animals , Blood Glucose/metabolism , Diabetes Mellitus/physiopathology , Diet, High-Fat , Homeostasis , Insulin/blood , Insulin Resistance , Insulin-Like Growth Factor Binding Proteins/genetics , Insulin-Like Growth Factor I/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Obesity/blood , Obesity/genetics
3.
J Immunol ; 186(11): 6106-18, 2011 Jun 01.
Article in English | MEDLINE | ID: mdl-21518973

ABSTRACT

Every person harbors a population of potentially self-reactive lymphocytes controlled by tightly balanced tolerance mechanisms. Failures in this balance evoke immune activation and autoimmunity. In this study, we investigated the contribution of self-reactive CD8(+) T lymphocytes to chronic pulmonary inflammation and a possible role for naturally occurring CD4(+)CD25(+)Foxp3(+) regulatory T cells (nTregs) in counterbalancing this process. Using a transgenic murine model for autoimmune-mediated lung disease, we demonstrated that despite pulmonary inflammation, lung-specific CD8(+) T cells can reside quiescently in close proximity to self-antigen. Whereas self-reactive CD8(+) T cells in the inflamed lung and lung-draining lymph nodes downregulated the expression of effector molecules, those located in the spleen appeared to be partly Ag-experienced and displayed a memory-like phenotype. Because ex vivo-reisolated self-reactive CD8(+) T cells were very well capable of responding to the Ag in vitro, we investigated a possible contribution of nTregs to the immune control over autoaggressive CD8(+) T cells in the lung. Notably, CD8(+) T cell tolerance established in the lung depends only partially on the function of nTregs, because self-reactive CD8(+) T cells underwent only biased activation and did not acquire effector function after nTreg depletion. However, although transient ablation of nTregs did not expand the population of self-reactive CD8(+) T cells or exacerbate the disease, it provoked rapid accumulation of activated CD103(+)CD62L(lo) Tregs in bronchial lymph nodes, a finding suggesting an adaptive phenotypic switch in the nTreg population that acts in concert with other yet-undefined mechanisms to prevent the detrimental activation of self-reactive CD8(+) T cells.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Forkhead Transcription Factors/immunology , Interleukin-2 Receptor alpha Subunit/immunology , Pneumonia/immunology , T-Lymphocytes, Regulatory/immunology , Adoptive Transfer , Animals , Antigens, CD/genetics , Antigens, CD/immunology , Antigens, CD/metabolism , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/transplantation , Flow Cytometry , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Gene Expression Profiling , Immune Tolerance/immunology , Immunologic Memory/immunology , Integrin alpha Chains/genetics , Integrin alpha Chains/immunology , Integrin alpha Chains/metabolism , Interleukin-2 Receptor alpha Subunit/genetics , Interleukin-2 Receptor alpha Subunit/metabolism , L-Selectin/genetics , L-Selectin/immunology , L-Selectin/metabolism , Lung/immunology , Lung/metabolism , Lung/physiopathology , Mice , Mice, Inbred BALB C , Mice, Transgenic , Oligonucleotide Array Sequence Analysis , Pneumonia/genetics , Pneumonia/metabolism , Respiratory Function Tests , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes, Regulatory/metabolism
4.
Lipids Health Dis ; 9: 99, 2010 Sep 10.
Article in English | MEDLINE | ID: mdl-20831792

ABSTRACT

BACKGROUND: The Berlin Fat Mouse Inbred (BFMI) line is a new mouse model for obesity, which was long-term selected for high fatness. Peroxisome proliferator-activated receptors (PPARs) are involved in the control of energy homeostasis, nutrient metabolism and cell proliferation. Here, we studied the expression patterns of the different Ppar genes and the genes in the PPAR pathway in the BFMI line in comparison to physiological changes. RESULTS: At the age of 10 weeks, the BFMI mice exhibited marked obesity with enlarged adipocytes and high serum triglycerides concentrations in comparison to the often used mouse line C57BL/6 (B6). Between these two lines, gene expression analyses revealed differentially expressed genes belonging to the PPAR pathway, in particular genes of the lipogenesis and the fatty acid transport. CONCLUSION: Surprisingly, the Ppar-α gene expression was up-regulated in liver and Ppar-γ gene expression was down-regulated in the white adipose tissue, indicating the activation of a mechanism that counteracts the rise of obesity.


Subject(s)
Gene Expression Regulation/genetics , Obesity/genetics , PPAR alpha/metabolism , PPAR gamma/metabolism , Signal Transduction/genetics , Adipose Tissue, White/metabolism , Adipose Tissue, White/pathology , Animals , Body Constitution , Cell Size , Dietary Fats/administration & dosage , Disease Models, Animal , Gene Expression Profiling , Genetic Predisposition to Disease , Hypertriglyceridemia/blood , Hypertriglyceridemia/genetics , Lipid Metabolism/genetics , Liver/metabolism , Male , Mice , Mice, Inbred Strains , Obesity/blood , Obesity/pathology , Oligonucleotide Array Sequence Analysis , PPAR alpha/genetics , PPAR gamma/genetics , RNA, Messenger/metabolism
5.
J Virol ; 84(20): 10708-18, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20686040

ABSTRACT

We assessed the prediction that access of the viral NS1 protein to cellular PDZ domain protein networks enhances the virulence of highly pathogenic avian influenza A viruses. The NS1 proteins of most avian influenza viruses bear the C-terminal ligand sequence Glu-Ser-Glu-Val (ESEV) for PDZ domains present in multiple host proteins, whereas no such motif is found in the NS1 homologues of seasonal human virus strains. Previous analysis showed that a C-terminal ESEV motif increases viral virulence when introduced into the NS1 protein of mouse-adapted H1N1 influenza virus. To examine the role of the PDZ domain ligand motif in avian influenza virus virulence, we generated three recombinants, derived from the prototypic H5N1 influenza A/Vietnam/1203/04 virus, expressing NS1 proteins that either have the C-terminal ESEV motif or the human influenza virus RSKV consensus or bear a natural truncation of this motif, respectively. Cell biological analyses showed strong control of NS1 nuclear migration in infected mammalian and avian cells, with only minor differences between the three variants. The ESEV sequence attenuated viral replication on cultured human, murine, and duck cells but not on chicken fibroblasts. However, all three viruses caused highly lethal infections in mice and chickens, with little difference in viral titers in organs, mean lethal dose, or intravenous pathogenicity index. These findings demonstrate that a PDZ domain ligand sequence in NS1 contributes little to the virulence of H5N1 viruses in these hosts, and they indicate that this motif modulates viral replication in a strain- and host-dependent manner.


Subject(s)
Influenza A Virus, H5N1 Subtype/genetics , Influenza A Virus, H5N1 Subtype/pathogenicity , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/physiology , Amino Acid Motifs , Animals , Cell Line , Chick Embryo , Chickens , Ducks , Female , Genetic Variation , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Host-Pathogen Interactions/physiology , Humans , Influenza A Virus, H5N1 Subtype/physiology , Influenza in Birds/etiology , Influenza in Birds/virology , Interferon-beta/biosynthesis , Interferon-beta/genetics , Mice , Mice, Inbred BALB C , Orthomyxoviridae Infections/etiology , Orthomyxoviridae Infections/pathology , Orthomyxoviridae Infections/virology , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Species Specificity , Viral Nonstructural Proteins/chemistry , Virulence/genetics , Virulence/physiology
6.
Am J Physiol Lung Cell Mol Physiol ; 299(4): L472-82, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20639353

ABSTRACT

Neutrophils are potent contributors to the lung pathophysiological changes occurring in allergic airway inflammation, which typically involve T helper type 2 (Th2) cytokine overexpression. We have previously reported that equine pulmonary endothelial cells are activated by the Th2 cytokine IL-4 and express chemotactic factors for neutrophils after stimulation. We have further explored the possible mechanisms linking Th2-driven inflammation and neutrophilia by studying the effects of recombinant equine IL-4, a prototypical Th2 cytokine, on peripheral blood neutrophils (PBN) isolated from normal animals and from horses with asthmatic airway inflammation (equine heaves). We found that IL-4 induced morphological changes in PBN, dose- and time-dependent expression of IL-8 mRNA, as well as the release of chemotactic factors for neutrophils in culture supernatants. Also, IL-4 induced a mixed inflammatory response in PBN from control and asthmatic-animals with increased expression of proinflammatory IL-8 and TNF-α but a marked inhibition of IL-1ß. IL-4 type I receptor (IL-4Rα) and CD23 (FcεRII) expression were also upregulated by IL-4. Importantly, disease as well as chronic antigenic exposure modified gene expression by PBN. Finally, we found that activation of equine neutrophils with IL-4 involved STAT6 phosphorylation and p38 MAPK and phosphatidylinositol 3-kinase (PI3K); the pharmacological inhibitors, SB-203580 and LY-294002, respectively, significantly reversed IL-4-induced gene modulation in PBN. Overall, results from this study add to the link between Th2-driven inflammation and neutrophilia in the equine model and further extend the characterization of IL-4 effects on neutrophils.


Subject(s)
Asthma/metabolism , Interleukin-4/metabolism , Interleukin-8/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Asthma/genetics , Asthma/pathology , Blotting, Western , Chemotaxis, Leukocyte/immunology , Gene Expression Profiling , Horses , Immunoenzyme Techniques , Inflammation/genetics , Inflammation/immunology , Interleukin-4/genetics , Interleukin-8/genetics , Neutrophil Activation , Phosphatidylinositol 3-Kinases/genetics , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , STAT6 Transcription Factor/genetics , STAT6 Transcription Factor/metabolism , Tumor Necrosis Factor-alpha/genetics , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
8.
J Med Primatol ; 38(5): 335-9, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19558457

ABSTRACT

BACKGROUND: A 38-year-old male Bornean orangutan (Pongo pygmaeus pygmaeus) developed progressive hind leg paresis. A computed tomography scan of the vertebral column revealed soft tissue type densities within vertebral bones. METHODS AND RESULTS: At necropsy infiltrating tumor masses were found in the vertebral bodies, protruding into the spinal canal and compressing the spinal cord. Microscopically neoplastic plasma cells infiltrated the vertebral bodies and adjacent soft tissues. Immunohistochemically, tumor cells tested positive for B cell markers (CD38, CD79alpha), kappa, and lambda light chains, while vimentin, GFAP, S100, and CD138 were not expressed. The tumor was classified as multiple myeloma on the basis of radiographic, pathological, and immunohistochemical findings. CONCLUSIONS: This first systematic case description on multiple myeloma in a non-human primate revealed many similarities with the disease in humans and the immunohistochemical tools proved suitable for their use in the orangutan.


Subject(s)
Ape Diseases/etiology , Multiple Myeloma/veterinary , Paraplegia/veterinary , Pongo pygmaeus , Animals , Ape Diseases/pathology , Cervical Vertebrae/pathology , Male , Multiple Myeloma/complications , Multiple Myeloma/pathology , Paraplegia/etiology , Paraplegia/pathology
9.
PLoS One ; 4(3): e4857, 2009.
Article in English | MEDLINE | ID: mdl-19293935

ABSTRACT

The genetic make-up of the host has a major influence on its response to combat pathogens. For influenza A virus, several single gene mutations have been described which contribute to survival, the immune response and clearance of the pathogen by the host organism. Here, we have studied the influence of the genetic background to influenza A H1N1 (PR8) and H7N7 (SC35M) viruses. The seven inbred laboratory strains of mice analyzed exhibited different weight loss kinetics and survival rates after infection with PR8. Two strains in particular, DBA/2J and A/J, showed very high susceptibility to viral infections compared to all other strains. The LD(50) to the influenza virus PR8 in DBA/2J mice was more than 1000-fold lower than in C57BL/6J mice. High susceptibility in DBA/2J mice was also observed after infection with influenza strain SC35M. In addition, infected DBA/2J mice showed a higher viral load in their lungs, elevated expression of cytokines and chemokines, and a more severe and extended lung pathology compared to infected C57BL/6J mice. These findings indicate a major contribution of the genetic background of the host to influenza A virus infections. The overall response in highly susceptible DBA/2J mice resembled the pathology described for infections with the highly virulent influenza H1N1-1918 and newly emerged H5N1 viruses.


Subject(s)
Orthomyxoviridae Infections/genetics , Animals , Bronchi/pathology , Cytokines/metabolism , Epithelial Cells/pathology , Inflammation Mediators/metabolism , Influenza A Virus, H1N1 Subtype , Mice , Mice, Inbred Strains , Species Specificity , Viral Load
10.
Vet Immunol Immunopathol ; 128(1-3): 16-23, 2009 Mar 15.
Article in English | MEDLINE | ID: mdl-19056130

ABSTRACT

Dendritic cells (DCs) are antigen-presenting cells, which are well known for their capacity to stimulate immunity. The ex vivo generation of myeloid DC from monocytes has facilitated the development of DC-vaccination protocols which have been extensively evaluated in tumour immunology and are regarded by some as a gold mine for clinical research. However, there is a considerable amount of work required to overcome the potential risks associated with such therapy. It is therefore mandatory to characterize the system to be applied and to study the reactions, particularly at the level of T cell responses. The first objective of the current study was to test if tumour lysates loaded autologous DC or recombinant human IL-2 are well tolerated in horses and performed an exploratory phase I study on equine sarcoids and squamous cell carcinomas. We consequently intended to establish a robust protocol for the magnetic separation of monocytes such as in use in human clinical studies. Finally we intended to address the limits in the reagents to study equine T cell based immune reactions, and analysed markers for CD25 and FoxP3. The data showed that local application of DC or IL-2 did not cause side effects. Additionally our data show that a polyclonal approach to detect antigens such as CD25 might be successful, where mAbs are not available. Our data also demonstrate that the mAb FJK16s, which has been used successfully in rodents, humans, and dogs, can also be applied in horses. We finally wish to share our concerns regarding quality control for clinical studies and encourage multi-central studies such as in human medicine to ensure that progress along established standards is made for the benefit of veterinary medicine.


Subject(s)
Dendritic Cells/metabolism , Dendritic Cells/physiology , Horse Diseases/immunology , Interleukin-2/pharmacology , T-Lymphocytes/physiology , Animals , Biomarkers , Forkhead Transcription Factors/metabolism , Horse Diseases/therapy , Horses , Humans , Interleukin-2 Receptor alpha Subunit/metabolism , Lymphocyte Activation/physiology , Quality Control
11.
APMIS ; 115(6): 687-700, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17550376

ABSTRACT

Transforming growth factor beta (TGF-ss) is able to inhibit proliferation of epithelial cells and is involved in the carcinogenesis of human mammary tumours. Three latent transforming growth factor-beta binding proteins (LTBP-1, -3 and -4) are involved in TGF-beta function. The aim of the study was to analyze the expression profiles of TGF-beta 1 and 2 and LTBP-4 in human mammary carcinoma cell lines as well as in human mammary tumours. Expression analysis was performed at the transcription and protein level under in vivo and in vitro conditions. LTBP-4 expression was quantitatively analysed in human carcinomas of the mammary gland and in healthy mammary tissues of the same patients. Downregulation of LTBP-4 in all investigated human mammary tumours compared to normal tissues could be demonstrated. Results also revealed that protein levels of TGF-beta 1 are downregulated and of TGF-beta 2 are upregulated in human mammary carcinoma cell lines compared to primary (normal) human mammary epithelial cells. LTBP-4 reduction in neoplasms leads to a possible decrease of TGF-beta 1 extracellular deposition with reduced TGF-beta 1 bioavailability. TGF-beta 2 was upregulated, which indicates a possible compensatory mechanism. This study demonstrated a possible functional role of LTBP-4 for TGF-beta bioavailability with respect to carcinogenesis of human mammary tumours in vivo and in vitro.


Subject(s)
Adenocarcinoma/metabolism , Breast Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Latent TGF-beta Binding Proteins/metabolism , Transforming Growth Factor beta/physiology , Adenocarcinoma/genetics , Breast Neoplasms/genetics , Down-Regulation , Humans , Immunohistochemistry , Latent TGF-beta Binding Proteins/immunology , RNA, Messenger/analysis , RNA, Messenger/biosynthesis , RNA, Neoplasm/chemistry , Transforming Growth Factor beta/immunology , Tumor Cells, Cultured
12.
Immunology ; 117(4): 463-73, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16556260

ABSTRACT

Dendritic cells (DC) are the initiators of immune responses and are present in most tissues in vivo. To generate myeloid DC from monocytes (MoDC) in vitro the necessary cytokines are granulocyte-macrophage colony-stimulating factor (GM-CSF) and interleukin-4 (IL-4). Using degenerated primers delineated from other species and rapid amplification of cDNA ends reverse transcription-polymerase chain reaction (RACE RT-PCR), the cDNA of equine (eq.) GM-CSF was cloned and found to have a point deletion at the 3'-end of eq.GM-CSF, resulting in a 24-nucleotide extended open reading frame not described in any species thus far. For differentiating eq.MoDC, monocytes were stimulated with eq.GM-CSF and eq.IL-4. The eq.MoDC was analysed by both light and electron microscopy and by flow cytometry and mixed lymphocyte reaction. The eq.MoDC obtained had the typical morphology and function of DC, including the ability to stimulate allogeneic T cells in a mixed lymphocyte reaction. In contrast to the human system, however, monocytes had to be differentiated for 6-7 days before immature DC were obtained. Our data also indicate that lipopolysaccharide or poly(I:C) alone are not sufficient to confer the full phenotypic transition into mature DC. Thus our study contributes to understanding the heterogeneity of immunity and adds important information on the equine immune system, which is clearly distinct from those of mice or man.


Subject(s)
Dendritic Cells/immunology , Horses/immunology , Monocytes/immunology , Amino Acid Sequence , Animals , Cell Differentiation/immunology , Cells, Cultured , Dendritic Cells/ultrastructure , Flow Cytometry , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Humans , Interleukin-4/immunology , Lymphocyte Activation/immunology , Lymphocyte Culture Test, Mixed , Mice , Molecular Sequence Data , Monocytes/ultrastructure , Reverse Transcriptase Polymerase Chain Reaction/methods , Sequence Alignment , Species Specificity
13.
Vet Immunol Immunopathol ; 108(1-2): 227-36, 2005 Oct 18.
Article in English | MEDLINE | ID: mdl-16112744

ABSTRACT

The myeloid cell system comprises of monocytes, macrophages (MPhi), dendritic cells (DC), Kupffer cells, osteoclasts or microglia and is also known as the mononuclear phagocytic system (MPS). Essential cytokines to differentiate or activate these cells include GM-CSF or IL-4. Important markers for characterization include CD1, CD14, CD68, CD163 and CD206. All these markers, however, were not cloned or further characterized in equids by use of monoclonal antibodies earlier. To overcome this problem with the present study, two approaches were used. First, we cloned equine cytokines and markers, and second we analyzed cross-reactivity of human homologues or anti-human monoclonal antibodies. For cloning of equine cytokines and markers, we used degenerate primers delineated from other species, or equine-specific primers based on previous information in Genbank. Flow cytometry was used to determine the expression of markers on myeloid cells. Cross-reactivity could be shown for anti-human CD14, CD163 and mannose receptor (CD206) mAbs. Surface markers such as CD1 and CD68 that distinguish MPhi and DC were cloned and sequenced. According to blast homology, equine CD1a and CD1b could be identified and distinguished. With the resulting information, dendritic cells and macrophages of horses may be characterized.


Subject(s)
Cytokines/genetics , Horses/genetics , Horses/immunology , Myeloid Cells/immunology , Animals , Antibodies, Monoclonal , Antigens, CD/genetics , Antigens, CD1/genetics , Antigens, Differentiation, Myelomonocytic/genetics , Base Sequence , Cloning, Molecular , Cross Reactions , DNA/genetics , Gene Expression , Genetic Markers , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Humans , Recombinant Proteins/genetics , Recombinant Proteins/immunology
15.
Vet Immunol Immunopathol ; 84(1-2): 83-95, 2002 Jan 01.
Article in English | MEDLINE | ID: mdl-11825600

ABSTRACT

Interferons (IFNs) are important mediators of the immune system. Their antiviral activity is an integral part of the innate immune defence, but all IFNs have immune regulatory functions also. Besides rec.eq.IFN-beta detailed descriptions on other rec.IFNs were lacking and none of the proteins was available. To compare the equine IFNs and allow detailed studies on proteins and bioactivity, we performed the expression cloning of rec.eq.IFN-alpha, -beta and -gamma. To achieve maximal expression, a bacterial expression system was chosen. Additionally, rec.eq.IFN-beta and -gamma were expressed in mouse B-cells. The antiviral activity was characterised using different cell lines and equine viruses. The results demonstrate a broad antiviral activity of rec.eq.IFN-alpha being active against all viruses tested, including the equine herpesviruses EHV-1 and -4, while rec.eq.IFN-beta was only active using primary horse cells. Protection depended on viruses, cell lines, infectious doses, amount and time of IFN action prior to infection. While rec.eq.IFN-gamma did not act antivirally, it was effective as an immune modulator of monocytes in vitro. The implications of our findings on clinical immunology and virology, including therapeutic applications of equine IFNs will be discussed.


Subject(s)
Interferons/biosynthesis , Recombinant Proteins/biosynthesis , Amino Acid Sequence , Animals , Antiviral Agents/pharmacology , Cloning, Molecular , Horses , Interferons/chemistry , Interferons/pharmacology , Molecular Sequence Data , Recombinant Proteins/chemistry , Recombinant Proteins/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...