Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Biochem J ; 440(3): 327-4, 2011 Dec 15.
Article in English | MEDLINE | ID: mdl-21848513

ABSTRACT

Skeletal muscle responds to exercise by activation of signalling pathways that co-ordinate gene expression to sustain muscle performance. MEF2 (myocyte enhancer factor 2)-dependent transcriptional activation of MHC (myosin heavy chain) genes promotes the transformation from fast-twitch into slow-twitch fibres, with MEF2 activity being tightly regulated by interaction with class IIa HDACs (histone deacetylases). PKD (protein kinase D) is known to directly phosphorylate skeletal muscle class IIa HDACs, mediating their nuclear export and thus derepression of MEF2. In the present study, we report the generation of transgenic mice with inducible conditional expression of a dominant-negative PKD1kd (kinase-dead PKD1) protein in skeletal muscle to assess the role of PKD in muscle function. In control mice, long-term voluntary running experiments resulted in a switch from type IIb+IId/x to type IIa plantaris muscle fibres as measured by indirect immunofluorescence of MHCs isoforms. In mice expressing PKD1kd, this fibre type switch was significantly impaired. These mice exhibited altered muscle fibre composition and decreased running performance compared with control mice. Our findings thus indicate that PKD activity is essential for exercise-induced MEF2-dependent skeletal muscle remodelling in vivo.


Subject(s)
Muscle, Skeletal/physiology , TRPP Cation Channels/metabolism , Actins/metabolism , Amino Acid Substitution , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Enzyme Assays , Enzyme Induction , Female , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , MEF2 Transcription Factors , Mice , Mice, Transgenic , Motor Activity , Muscle Fibers, Fast-Twitch/metabolism , Muscle Fibers, Slow-Twitch/metabolism , Muscle, Skeletal/metabolism , Myogenic Regulatory Factors/metabolism , Myosin Heavy Chains/genetics , Myosin Heavy Chains/metabolism , Protein Engineering , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Running , TRPP Cation Channels/genetics
2.
J Immunol ; 176(4): 2538-45, 2006 Feb 15.
Article in English | MEDLINE | ID: mdl-16456015

ABSTRACT

Neutrophils infiltrate airway walls in patients with allergic airway diseases and in animal models of these illnesses, but their contribution to the pathogenesis of airway allergy is not established. We hypothesized that, in a mouse model of airway allergy to the ubiquitous environmental mold, Aspergillus fumigatus, airway neutrophils contribute to disease severity. Ab-mediated neutrophil depletion resulted in reduced airway hyperresponsiveness and remodeling, whereas conditional transgenic overexpression of the neutrophil chemotactic molecule, CXCL1, in airway walls resulted in worsened allergic responses. This worsened phenotype was associated with a marked increase in the number of airway neutrophils but not other lung leukocytes, including eosinophils and lymphocyte subsets, and depletion of neutrophils in sensitized mice with transgenic overexpression of CXCL1 resulted in attenuated airway responses. The number of lung neutrophils correlated with lung matrix metalloproteinase 9 (MMP-9) activity both in the context of neutrophil depletion and with augmented neutrophil recruitment to the airways. Although wild-type and MMP-9-deficient neutrophils homed to the inflamed airways to a similar extent, transfer of wild-type, but not MMP-9-deficient, neutrophils to MMP-9-deficient animals resulted in augmented allergic airway responses. Taken together, these data implicate neutrophils in the pathogenesis of fungal allergic airway disease.


Subject(s)
Aspergillosis/immunology , Aspergillus fumigatus/immunology , Disease Models, Animal , Hypersensitivity/immunology , Lung Diseases/immunology , Neutrophils/physiology , Animals , Aspergillosis/genetics , Aspergillosis/pathology , Cytokines/metabolism , Hypersensitivity/genetics , Hypersensitivity/pathology , Immunoglobulin E/blood , Immunoglobulin E/immunology , Lung Diseases/genetics , Lung Diseases/pathology , Matrix Metalloproteinase 9/metabolism , Mice , Neutrophils/immunology , T-Lymphocytes, Helper-Inducer/metabolism , Transgenes/genetics
3.
Am J Pathol ; 164(6): 2289-97, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15161661

ABSTRACT

Chemokines have been implicated in the pathogenesis of a wide variety of diseases. This report describes the generation of transgenic mice that conditionally express M3, a herpesvirus protein that binds and inhibits chemokines. In response to doxycycline, M3 expression was induced in a variety of tissues and M3 was detectable in the blood by Western blotting. No gross or histological abnormalities were seen in mice expressing M3. To determine whether M3 expression could modify a significant pathophysiological response, we examined its effect on the development of intimal hyperplasia in response to femoral arterial injury. Intimal hyperplasia is thought to play a critical role in the development of restenosis after percutaneous transluminal coronary angioplasty and in the progression of atherosclerosis. Induction of M3 expression resulted in a 67% reduction in intimal area and a 68% reduction in intimal/medial ratio after femoral artery injury. These data support a role for chemokines in regulating intimal hyperplasia and suggest that M3 may be effective in attenuating this process. This transgenic mouse model should be a valuable tool for investigating the role of chemokines in a variety of pathological states.


Subject(s)
Tunica Intima/pathology , Viral Proteins/genetics , Animals , Blood Chemical Analysis , Doxycycline/pharmacology , Femoral Artery/pathology , Gene Expression Regulation, Viral , Hyperplasia/genetics , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Transgenic , Viral Proteins/physiology , beta-Galactosidase/genetics
4.
J Pharmacol Exp Ther ; 310(1): 291-300, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15028780

ABSTRACT

Two genes with high sequence homology to human CXCR1 (hCXCR1) and CXCR2 (hCXCR2) were cloned from blood of cynomolgus monkey (Macaca fascicularis). Comparison of the expression pattern of these receptors in different species demonstrated that, like in humans, cynomolgus CXCR1 (cCXCR1) and CXCR2 (cCXCR2) are highly expressed in blood. Membranes from transfected BaF3 cells expressing cCXCR1 bind interleukin (IL)-8 with an affinity similar to hCXCR1 (Kd values, 170 +/- 87 and 103 +/- 37 pM, respectively) and show low binding affinity to Gro-alpha. Cynomolgus CXCR2 also binds hIL-8 but with somewhat higher affinity than the hCXCR2 (46 +/- 28 and 220 +/- 14 pM, respectively). Surprisingly, cCXCR2 has a reduced binding affinity to hGro-alpha (3.7 +/- 2.2 nM), a specific ligand of hCXCR2 (540 +/- 140 pM). Furthermore, the CXCR2-specific antagonist SB225002 [N-(2-hydroxy-4-nitrophenyl)-N'-(2-bromophenyl)urea] is 10-fold more potent in inhibiting IL-8 binding to hCXCR2 than to cCXCR2, suggesting that some of the observed differences in the amino acid sequences of the human and monkey receptor affect ligand binding sites or the conformation of the receptor. Both cynomolgus receptors were functionally active in inducing guanosine 5'-O-(3-thio)triphosphate exchange on membranes in response to IL-8 and Gro-alpha and in mediating chemotactic activity of recombinant BA/F3 cells in response to IL-8 and Gro-alpha. These results identify the products of the novel cynomolgus genes as functional homologs of hCXCR1 and hCXCR2.


Subject(s)
Macaca fascicularis/genetics , Receptors, Interleukin-8A/genetics , Receptors, Interleukin-8B/genetics , Amino Acid Sequence , Animals , Gene Expression , Humans , Interleukin-8/pharmacology , Molecular Sequence Data , Receptors, Interleukin-8A/chemistry , Receptors, Interleukin-8A/drug effects , Receptors, Interleukin-8B/chemistry , Receptors, Interleukin-8B/drug effects , Sequence Homology, Amino Acid , Species Specificity
6.
J Immunol ; 170(6): 2843-52, 2003 Mar 15.
Article in English | MEDLINE | ID: mdl-12626534

ABSTRACT

The analysis of the development and function of distinct subsets of murine dendritic cells (DC) has been hampered by the limited number of these cells in vivo. To circumvent this limitation we have developed a conditional transgenic mouse model for producing large numbers of DC. We used the tetracycline-inducible system to conditionally express murine Flt3 ligand (FL), a potent hemopoietic growth factor that promotes the differentiation and mobilization of DC. Acute treatment (96 h) of the transgenic animals with the tetracycline analog doxycycline (DOX) promoted an approximately 200-fold increase in serum levels of FL without affecting the number of circulating DC. However, within 1 wk of DOX treatment, the relative number of DC in peripheral blood increased from approximately 8 to approximately 40%. Interestingly, both the levels of FL and the number of DC remained elevated for at least 9 mo with continual DOX treatment. Chronic treatment of the mice with DOX led to dramatic increases in the number of DC in multiple tissues without any apparent pathological consequences. Most DC populations were expanded, including immature and mature DC, myeloid (CD11c(+)CD11b(+)CD8a(-)), lymphoid (CD11c(+)CD11b(-)CD8a(+)), and the recently defined plasmacytoid (pDC) subsets. Finally, transplantation of BM from green fluorescent protein-expressing mice into lethally irradiated transgenic mice followed by subsequent DOX treatment led to expansion of green fluorescent protein-labeled DC. The transgenic mice described here should thus provide a readily available source of multiple DC subsets and should facilitate the analysis of their role in homeostasis and disease.


Subject(s)
Dendritic Cells/cytology , Dendritic Cells/immunology , Gene Expression Regulation/immunology , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Animals , CD11c Antigen/biosynthesis , CD11c Antigen/blood , Cell Division/drug effects , Cell Division/genetics , Cell Division/immunology , Cell Movement/drug effects , Cell Movement/genetics , Cell Movement/immunology , Crosses, Genetic , Dendritic Cells/metabolism , Doxycycline/pharmacology , Gene Expression Regulation/drug effects , Green Fluorescent Proteins , Immunophenotyping , Ligands , Luminescent Proteins/biosynthesis , Luminescent Proteins/genetics , Lymphocyte Activation/genetics , Lymphocyte Subsets/cytology , Lymphocyte Subsets/immunology , Membrane Proteins/blood , Membrane Proteins/physiology , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Transgenic , Myeloid Cells/cytology , Myeloid Cells/immunology , Organ Specificity/drug effects , Organ Specificity/genetics , Organ Specificity/immunology , Plasma Cells/cytology , Plasma Cells/immunology , Plasma Cells/metabolism , Transgenes/immunology
7.
J Virol ; 77(1): 624-30, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12477865

ABSTRACT

Chemokine-binding proteins represent a novel class of antichemokine agents encoded by poxviruses and herpesviruses. One such protein is encoded by the M3 gene present in the murine gammaherpesvirus 68 (MHV-68) genome. The M3 gene encodes a secreted 44-kDa protein that binds with high affinity to certain murine and human chemokines and has been shown to block chemokine signaling in vitro. However, there has been no direct evidence that M3 blocks chemokine activity in vivo, nor has the nature of M3-chemokine interaction been defined. To better understand the ability of M3 to block chemokine activity in vivo, we examined its interaction with a specific subset of chemokines expressed in lymphoid tissues, areas where gammaherpesviruses characteristically establish latency. Here we show that M3 blocks in vitro chemotaxis induced by CCL19 and CCL21, chemokines expressed constitutively in secondary lymphoid tissues. Moreover, we provide evidence that chemokine M3 binding exhibits positive cooperativity. In vivo, the expression of M3 in the pancreas of transgenic mice inhibits recruitment of lymphocytes induced by transgenic expression of CCL21 in this organ. The ability of M3 to block the biological activity of chemokines may represent an important strategy used by MHV-68 to evade immune detection and favor viral replication in the infected host.


Subject(s)
Chemokines, CC/antagonists & inhibitors , Chemotaxis/drug effects , Viral Proteins/pharmacology , Viral Proteins/physiology , Animals , Cell Movement , Chemokine CCL19 , Chemokine CCL21 , Gammaherpesvirinae/physiology , Islets of Langerhans/pathology , Lymphocytes/physiology , Mice , Mice, Transgenic
8.
J Immunol ; 169(12): 7054-62, 2002 Dec 15.
Article in English | MEDLINE | ID: mdl-12471141

ABSTRACT

Tumor necrosis factor is a proinflammatory cytokine that induces directly many of the components required for inflammation to proceed rapidly. We show in this study that the interplay between TNF and chemokines, now recognized to be essential for normal secondary lymphoid tissue development, is also a feature of CNS inflammation, and that the two apparently dissimilar biological processes share many properties. Thus, induction of seven chemokines, including T cell activation gene 3 (TCA3), monocyte chemoattractant protein-1, and IFN-gamma-inducible protein-10 within the CNS during experimental autoimmune encephalomyelitis fails to occur early in the inflammatory process in TNF-deficient mice, despite local expression of monokines and IFN-gamma. The critical source of TNF in CNS inflammation is the infiltrating hemopoietic cell, and, in its absence, chemokine expression by irradiation-resistant CNS-resident cells fails. The CCR8 ligand, TCA3, is shown to be produced predominantly by resident microglia of the CNS in response to TNF. Using CCR8(-/-) mice, evidence is provided that TCA3-CCR8 interactions contribute to rapid-onset CNS inflammation. Thus, through TNF production, the hemopoietic compartment initiates the signals for its own movement into tissues, although the tissue ultimately defines the nature of that movement. Chemokines are a major, although not exclusive, mechanism by which tissues regulate leukocyte movement in response to TNF.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Hematopoietic Stem Cells/immunology , Hematopoietic Stem Cells/metabolism , Neuroglia/immunology , Neuroglia/metabolism , Spinal Cord/immunology , Spinal Cord/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Cell Movement/genetics , Cell Movement/immunology , Chemokine CCL1 , Chemokines/biosynthesis , Chemokines, CC , Cytokines/biosynthesis , Cytokines/metabolism , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Glycoproteins/administration & dosage , Glycoproteins/immunology , Hematopoietic Stem Cells/pathology , Linear Models , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/immunology , Microglia/metabolism , Models, Immunological , Myelin-Oligodendrocyte Glycoprotein , Peptide Fragments/administration & dosage , Peptide Fragments/immunology , Receptors, CCR8 , Receptors, Chemokine/metabolism , Spinal Cord/pathology , Tumor Necrosis Factor-alpha/deficiency , Tumor Necrosis Factor-alpha/genetics
9.
J Immunol ; 168(3): 1001-8, 2002 Feb 01.
Article in English | MEDLINE | ID: mdl-11801632

ABSTRACT

The CC chemokine CCL21 is a potent chemoattractant for lymphocytes and dendritic cells in vitro. In the murine genome there are multiple copies of CCL21 encoding two CCL21 proteins that differ from each other by one amino acid at position 65 (either a serine or leucine residue). In this report, we examine the expression pattern and biological activities of both forms of CCL21. We found that although both serine and leucine forms are expressed in most tissues examined, the former was the predominant form in lymphoid organs while the latter was predominantly expressed in nonlymphoid organs. When expressed in transgenic pancreas, both forms of CCL21 were capable of inducing the formation of lymph node-like structures composed primarily of T and B cells and a few dendritic cells. Induction of lymph node-like structures by these CCL21 proteins, however, could not be reproduced in every tissue. For instance, no lymphocyte recruitment or accumulation was observed when CCL21 was overexpressed in the skin. We conclude that both forms of CCL21 protein are biologically equivalent in promoting lymphocyte recruitment to the pancreas, and that their ability to induce the formation of lymph node-like structures is dependent on the tissues in which they are expressed.


Subject(s)
Chemokines, CC/biosynthesis , Choristoma/immunology , Lymph Nodes/immunology , Lymphatic Diseases/immunology , Mice, Transgenic/immunology , Pancreas , Skin , Animals , Cell Movement/immunology , Chemokine CCL21 , Chemokines, CC/genetics , Chemokines, CC/physiology , Choristoma/genetics , Choristoma/pathology , Dendritic Cells/cytology , Dendritic Cells/immunology , Gene Expression Regulation/immunology , Islets of Langerhans/cytology , Islets of Langerhans/immunology , Islets of Langerhans/metabolism , Langerhans Cells/cytology , Langerhans Cells/immunology , Lymph Nodes/cytology , Lymphatic Diseases/genetics , Lymphatic Diseases/pathology , Lymphocytes/cytology , Lymphocytes/immunology , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Inbred ICR , Organ Culture Techniques , Organ Specificity/genetics , Organ Specificity/immunology , Protein Isoforms/biosynthesis , Protein Isoforms/genetics , Protein Isoforms/physiology , Skin/cytology , Skin/metabolism , Transgenes/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...