Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.352
Filter
1.
PLoS One ; 9(11): e113752, 2014.
Article in English | MEDLINE | ID: mdl-25409512

ABSTRACT

Macrophage accumulation in the arterial wall and smooth muscle cell (SMC) proliferation are features of type 2 diabetes mellitus (DM) and its vascular complications. However, the effects of diabetic monocyte-derived macrophages on vascular SMC proliferation are not clearly understood. In the present study, we investigated the pro-proliferative effect of macrophages isolated from DM patients on vascular SMCs. Macrophage-conditioned media (MCM) were prepared from macrophages isolated from DM patients. DM-MCM treatment induced HASMC proliferation, decreased p21(Cip1) and p27(Kip1) expressions, and increased microRNA (miR)-17-5p and miR-221 expressions. Inhibition of either miR-17-5p or miR-221 inhibited DM-MCM-induced cell proliferation. Inhibition of miR-17-5p abolished DM-MCM-induced p21(Cip1) down-regulation; and inhibition of miR-221 attenuated the DM-MCM-induced p27(Kip1) down-regulation. Furthermore, blocking assays demonstrated that PDGF-CC in DM-MCM is the major mediators of cell proliferation in SMCs. In conclusion, our present data support the hypothesis that SMC proliferation stimulated by macrophages may play critical roles in vascular complications in DM patients and suggest a new mechanism by which arterial disease is accelerated in diabetes.


Subject(s)
Aorta/cytology , Macrophages/cytology , Monocytes/cytology , Muscle, Smooth, Vascular/metabolism , Adult , Becaplermin , Cell Proliferation/drug effects , Cells, Cultured , Culture Media, Conditioned/pharmacology , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Down-Regulation/drug effects , Enzyme-Linked Immunosorbent Assay , Humans , Lymphokines/analysis , Macrophages/immunology , Macrophages/metabolism , MicroRNAs/metabolism , Middle Aged , Monocytes/immunology , Monocytes/metabolism , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Osteopontin/genetics , Osteopontin/metabolism , Platelet-Derived Growth Factor/analysis , Proto-Oncogene Proteins c-sis/analysis
2.
Lab Invest ; 94(6): 674-82, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24709779

ABSTRACT

Platelet-derived growth factor (PDGF)-C is a member of the PDGF family and is critical for neuronal survival in the central nervous system. We studied the possible survival and antiapoptotic effects of PDGF-C on focal retinal lesions in Ccl2(-/-)/Cx3cr1(-/-) on C57BL/6N [Crb1(rd8)] (DKO rd8) background mice, a model for progressive and focal retinal degeneration. We found no difference in transcript and protein expression of PDGF-C in the retina between DKO rd8 mice and wild type (WT, C57BL/6N). Recombinant PDGF-CC protein (500 ng/eye) was injected intravitreally into the right eye of DKO rd8 mice with phosphate-buffered saline as controls into the left eye. The retinal effects of PDGF-C were assessed by fundoscopy, ocular histopathology, A2E levels, apoptotic molecule analysis, and direct flat mount retinal vascular labeling. We found that the PDGF-CC-treated eyes showed slower progression or attenuation of the focal retinal lesions, lesser photoreceptor and retinal pigment epithelial degeneration resulting in better-preserved photoreceptor structure. Lower expression of apoptotic molecules was detected in the PDGF-CC-treated eyes than in controls. In addition, no retinal neovascularization was observed after PDGF-CC treatment. Our results demonstrate that PDGF-C potently ameliorates photoreceptor degeneration via the suppression of apoptotic pathways without inducing retinal angiogenesis. The protective effects of PDGF-C suggest a novel alternative approach for potential age-related retinal degeneration treatment.


Subject(s)
Apoptosis/drug effects , Lymphokines/metabolism , Lymphokines/pharmacology , Platelet-Derived Growth Factor/metabolism , Platelet-Derived Growth Factor/pharmacology , Retina/drug effects , Retinal Degeneration/metabolism , Animals , Lymphokines/analysis , Lymphokines/genetics , Mice , Mice, Knockout , Neovascularization, Pathologic , Platelet-Derived Growth Factor/analysis , Platelet-Derived Growth Factor/genetics , Retina/pathology , Retinal Degeneration/genetics , Retinal Degeneration/pathology
3.
Reproduction ; 147(6): 753-64, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24516176

ABSTRACT

This study was conducted to characterize the major proteins of the peccary seminal plasma, based on the semen samples collected from nine adult and reproductively sound animals. Our approach included the use of two-dimensional electrophoresis followed by Coomassie blue staining and analysis of polypeptide maps with PDQuest Software (Bio-Rad). Proteins were identified by tandem mass spectrometry (LC-MS/MS). We detected 179 protein spots per gel and 98 spots were identified by mass spectrometry, corresponding to 23 different proteins. The combined intensity of those spots accounted for 56.2±6% of the intensities of all spots and 60.9% of the intensities of spots presented in every protein map. Protein spots identified as clusterin represented 19.7±8.3% of the integrated optical densities of all spots detected in the seminal plasma maps. There was a negative association (r=-0.87; P<0.05) between the intensity of a clusterin spot and the percentage of sperm with functional membrane. Spermadhesin porcine seminal plasma protein 1 and bodhesin 2 comprised 5.4±1.9 and 8.8±3.9% of the total intensity of all spots respectively. Many proteins appeared in a polymorphic pattern, such as clusterin (27 spots), epididymal secretory glutathione peroxidase (ten spots), inter-α-trypsin inhibitor (12 spots), and IgG-binding protein (ten spots), among others. In conclusion, we presently describe the major seminal plasma proteome of the peccary, which exhibits a distinct high expression of clusterin isoforms. Knowledge of wild species reproductive biology is crucial for an understanding of their survival strategies and adaptation in a changing environment.


Subject(s)
Artiodactyla/metabolism , Semen/chemistry , Seminal Plasma Proteins/analysis , Alpha-Globulins/analysis , Animals , Chromatography, Liquid , Clusterin/analysis , Conservation of Natural Resources , Electrophoresis, Gel, Two-Dimensional , Glutathione Peroxidase/analysis , Lymphokines/analysis , Male , Protein Interaction Maps , Protein Isoforms , Proteomics/methods , Tandem Mass Spectrometry
4.
J Immunol ; 190(3): 881-5, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23267024

ABSTRACT

Mice lacking IL-6 are resistant to autoimmune diseases, such as experimental autoimmune encephalomyelitis (EAE), which is driven by CNS-reactive CD4(+) T cells. There are multiple cellular sources of IL-6, but the critical source in EAE has been uncertain. Using cell-specific IL-6 deficiency in models of EAE induced by active immunization, passive transfer, T cell transfer, and dendritic cell transfer, we show that neither the pathogenic T cells nor CNS-resident cells are required to produce IL-6. Instead, the requirement for IL-6 was restricted to the early stages of T cell activation and was entirely controlled by dendritic cell-derived IL-6. This reflected the loss of IL-6R expression by T cells over time. These data explain why blockade of IL-6R only achieves protection against EAE if used at the time of T cell priming. The implications for therapeutic manipulation of IL-6 signaling in human T cell-driven autoimmune conditions are considered.


Subject(s)
Dendritic Cells/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Interleukin-6/immunology , Adoptive Transfer , Animals , Autoantigens/immunology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/transplantation , Crosses, Genetic , Dendritic Cells/metabolism , Dendritic Cells/transplantation , Histocompatibility Antigens Class II/immunology , Immunization, Passive , Interleukin-6/deficiency , Interleukin-6/metabolism , Lymphocyte Activation , Lymphokines/analysis , Mice , Mice, Inbred C57BL , Mice, Knockout , Myelin Basic Protein/immunology , Myelin-Oligodendrocyte Glycoprotein/immunology , Peptide Fragments/immunology , Receptors, Interleukin-6/biosynthesis , Receptors, Interleukin-6/immunology , Specific Pathogen-Free Organisms , T-Cell Antigen Receptor Specificity
5.
Lasers Med Sci ; 25(4): 531-7, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20162318

ABSTRACT

Irradiation by light emitting diode (LED) promotes fibroblast proliferation and wound healing. However, its mechanism is still unknown. The purpose of this study was to clarify the mechanism of fibroblast proliferation by LED irradiation. Cultured NIH3T3 fibroblasts from normal mice were irradiated by LED with a center wavelength of 627 nm. LED irradiation was performed with an energy density of 4 J/cm(2), at subculture and 24 h later. The expression of several growth factors and their receptors was analyzed by reverse transcriptase-polymerase chain reaction (RT-PCR): platelet-derived growth factor (PDGF)-A, PDGF-B, and PDGF-C, transforming growth factor-beta (TGF-beta), basic fibroblast growth factor (bFGF), PDGF-alpha receptor, and TGF-beta receptor. Then, the activation of the extracellular signal-regulated kinase (ERK) pathway was examined by Western blotting with and without the PDGF receptor inhibitor. LED irradiation induced cell growth of NIH3T3 fibroblasts. The expression of PDGF-C had significantly increased in the irradiated group (P < 0.01). Although strong activation of the ERK pathway was observed in the irradiated group, its activation was completely suppressed by the PDGF receptor inhibitor. We concluded that LED irradiation promotes fibroblast proliferation by increasing autocrine production of PDGF-C and activating the ERK pathway through phosphorylation of the PDGF receptor.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/metabolism , Light , Animals , Cell Division/radiation effects , Cells, Cultured , Enzyme Activation/radiation effects , Fibroblast Growth Factor 2/analysis , Fibroblasts/radiation effects , Lymphokines/analysis , Mice , Platelet-Derived Growth Factor/analysis , Proto-Oncogene Proteins c-sis/analysis , Receptors, Platelet-Derived Growth Factor/analysis , Receptors, Transforming Growth Factor beta/analysis , Signal Transduction/radiation effects , Transforming Growth Factor beta/analysis
6.
J Endod ; 35(3): 377-81, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19249599

ABSTRACT

The purpose of this study was to examine the expression of platelet-derived growth factor C (PDGF-C), which is a new member of the PDGF family, in experimental periapical lesions. Periapical lesions were induced in Sprague-Dawley rats by occlusal pulp exposure in the mandibular first molars. Animals were sacrificed randomly at 0, 7, 14, 21, and 28 days after pulp exposure. Frontal sections were prepared for histological analysis and enzyme histochemistry. Reverse-transcription polymerase chain reaction and immunohistochemistry were performed to detect PDGF-C expression. From day 0 to day 28, PDGF-C messenger RNA were expressed and increased until day 28. A few PDGF-C-positive cells and osteoclasts could be observed on day 7. Both ascended in number on day 14. In the 21- and 28-day samples, the PDGF-C-positive cells increased, whereas fewer osteoclasts were observed. Many of the PDGF-C-positive cells were inflammatory cells with different morphologies. These findings showed that PDGF-C could be observed and might also be involved in the pathogenesis of periapical lesions.


Subject(s)
Alveolar Bone Loss/metabolism , Dental Pulp/metabolism , Lymphokines/biosynthesis , Periapical Periodontitis/metabolism , Platelet-Derived Growth Factor/biosynthesis , Alveolar Bone Loss/etiology , Animals , Dental Pulp Exposure , Gene Expression , Immunoenzyme Techniques , Lymphokines/analysis , Male , Osteoclasts/physiology , Platelet-Derived Growth Factor/analysis , Protein Isoforms , RNA, Messenger/biosynthesis , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction
7.
Article in English | MEDLINE | ID: mdl-18930835

ABSTRACT

This study examined ontogeny of development for a range of adipokines in neonatal adipose tissue. Pigs (Sus scrofa) were selected across six litters for sampling subcutaneous (SQ) and perirenal (PR) adipose tissues at d1, d4, d7 or d21 of age and total RNA extraction. Reverse transcription and real-time PCR were used to quantify mRNA abundance for: leptin, adiponectin, interleukin 1beta (IL-1beta), IL-6, IL-8, IL-10, IL-15, tumor necrosis factor alpha (TNFalpha), haptoglobin, vascular endothelial growth factor (VEGF), macrophage migration inhibitory factor (MIF), monocyte chemoattractant protein 1 (MCP1) and cyclophilin. Leptin, adiponectin and IL-15 expression increased from d1 to d 21 of age in both SQ and PR. Haptoglobin, VEGF, MIF and IL-8 expression decreased between d1 and d4 of age in SQ. TNFalpha expression was unchanged from d1-7 and then increased at d21. IL-1beta, IL-6 and IL-10 expression were unchanged with age in SQ; whereas IL-1beta and IL-6 mRNA abundance in the PR increased with age. Analysis of the mRNA abundance for these adipokines within adipose tissue from d1 to d21 of age demonstrated that neonatal development of adipokine expression varies among the different adipokines and the internal and external sites of adipose tissue deposition (PR versus SQ).


Subject(s)
Adipokines/genetics , Adipose Tissue/chemistry , Adipokines/analysis , Age Factors , Animals , Animals, Newborn , Haptoglobins/analysis , Haptoglobins/genetics , Interleukins/analysis , Interleukins/genetics , Lymphokines/analysis , Lymphokines/genetics , RNA, Messenger/analysis , Swine , Vascular Endothelial Growth Factor A/analysis , Vascular Endothelial Growth Factor A/genetics
8.
Pediatr Infect Dis J ; 27(10 Suppl): S92-6, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18820587

ABSTRACT

Respiratory syncytial virus (RSV) and influenza virus are common causes of infantile lower respiratory tract infection (LRTI). It is widely believed that both viral replication and inappropriately enhanced immune responses contribute to disease severity. In infants, RSV LRTI is known to be more severe than influenza virus LRTI. We compared cytokines and chemokines in secretions of infants surviving various forms of respiratory illness caused by RSV or influenza viruses, to determine which mediators were associated with more severe illness. We analyzed lung tissue from fatal cases of RSV and influenza LRTI to determine the types of inflammatory cells present. Quantities of lymphocyte-derived cytokines were minimal in secretions from infants with RSV infection. Concentrations of most cytokines were greater in influenza, rather than RSV, infection. Lung tissues from fatal RSV and influenza LRTI cases demonstrated extensive presence of viral antigen and a near absence of CD8-positive lymphocytes and natural killer cells, with marked expression of markers of apoptosis. Severe infantile RSV and influenza virus LRTI is characterized by inadequate (rather than excessive) adaptive immune responses, robust viral replication and apoptotic crisis.


Subject(s)
Bronchiolitis, Viral/immunology , Cytokines/analysis , Influenza, Human/immunology , Respiratory Syncytial Virus Infections/immunology , Antigens, Viral/isolation & purification , Apoptosis , Bronchiolitis, Viral/pathology , Bronchiolitis, Viral/virology , CD4 Antigens/immunology , CD8 Antigens/immunology , Child, Preschool , Female , Humans , Immunity, Cellular , Infant , Infant, Newborn , Influenza, Human/pathology , Influenza, Human/virology , Lung/immunology , Lung/pathology , Lung/virology , Lymphokines/analysis , Male , Orthomyxoviridae/immunology , Orthomyxoviridae/isolation & purification , Respiratory Syncytial Virus Infections/pathology , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Virus Vaccines/immunology , Respiratory Syncytial Viruses/immunology , Respiratory Syncytial Viruses/isolation & purification
9.
World J Surg ; 31(4): 733-43, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17372666

ABSTRACT

INTRODUCTION: Although both fluid shear stress and mass transport of atherogenic substances into the vascular wall are known to be important factors in atherogenesis, there has been little research on the effect of shear stress on vascular permeability. Therefore, the effects of shear stress on the permeability of arteries and the expression of the endothelial cell tight junction molecule occludin, an important regulator of vascular permeability, were investigated. METHODS: Porcine carotid arteries were perfusion cultured ex vivo with low (1.5 dyne/cm(2)) or physiologic (15 dyne/cm(2)) shear stress and 100 mmHg pressure for 24 hours. Subsequently, 20 nm gold particles in solution were infused into the lumen of vessels at 100 mmHg for 30 minutes. Frozen sections were then cut and stained for gold particles. Image analysis was used to determine the density of the particles in the vessel walls. The expression of endothelial cell occludin mRNA and protein were determined using reverse transcription-polymerase chain reaction (RT-PCR) and Western blotting, respectively. RESULTS: Permeability results showed a 2.8-fold increase in the apparent permeability of vessels cultured with low versus physiologic shear stress. RT-PCR and Western blotting results showed significant decreases in occludin mRNA and protein expression at 12 and 24 hours in vessels cultured with low versus physiologic shear stress. CONCLUSIONS: These results demonstrate that low shear stress increases vascular permeability in porcine carotid arteries, possibly owing to decreased occludin expression. These results may have implications in the preferential formation of atherosclerotic vascular disease adjacent to branches and bifurcations where low mean shear stresses may occur.


Subject(s)
Endothelial Growth Factors/genetics , Endothelium, Vascular/metabolism , Gene Expression , Lymphokines/genetics , Membrane Proteins/genetics , Animals , Blotting, Western , Carotid Arteries , Cells, Cultured , Endothelial Growth Factors/analysis , Endothelium, Vascular/chemistry , Endothelium, Vascular/cytology , Immunohistochemistry , Lymphokines/analysis , Membrane Proteins/analysis , Occludin , Permeability , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction , Rheology , Swine , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
10.
Arthritis Rheum ; 54(3): 788-94, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16508943

ABSTRACT

OBJECTIVE: To investigate the messenger RNA (mRNA) and protein expression of the recently discovered platelet-derived growth factor C (PDGF-C) and PDGF-D in the synovial membrane (SM) of patients with rheumatoid arthritis (RA) and osteoarthritis (OA) and to assess the localization and cellular source of these proteins in the SM and their functional influence on synovial fibroblasts. METHODS: Expression of mRNA for PDGFs A, B, C, and D as well as for PDGF receptor (PDGFR) alpha and beta chains in RA and OA SM samples was assessed by real-time reverse transcription-polymerase chain reaction. Protein levels of PDGF-C and PDGF-D were quantified by immunoblotting. Regional and cellular localization of PDGF-C and PDGF-D in the SM was investigated by double-staining immunohistochemistry. In addition, the influence of PDGF-D on the proliferation of synovial fibroblasts and their matrix metalloproteinase (MMP-1) mRNA expression were determined. RESULTS: The expression of mRNA for PDGFs A, B, and C and for PDGFR alpha and beta chains was comparable in RA and OA SM samples; in contrast, the expression of mRNA for PDGF-D was significantly higher in OA SM. PDGF-C protein was not differentially expressed in OA and RA. The expression of PDGF-D protein was significantly higher in RA SM (full-length and activated form). PDGF-C and PDGF-D were expressed throughout the SM (lining layer, diffuse infiltrates, and stroma) by both synovial fibroblasts and macrophages. In addition, PDGF-D increased the proliferation of synovial fibroblasts and the expression of mRNA for MMP-1. CONCLUSION: PDGF-C and PDGF-D are expressed by synovial fibroblasts and macrophages in RA and OA SMs. The levels of PDGF-D protein were significantly higher in RA SM. In addition, PDGF-D stimulated synovial fibroblast proliferation and expression of MMP-1. These findings may have pathogenetic implications for cellular transformation and matrix remodeling in the RA SM.


Subject(s)
Arthritis, Rheumatoid/metabolism , Lymphokines/analysis , Osteoarthritis/metabolism , Platelet-Derived Growth Factor/analysis , Synovial Membrane/chemistry , Fibroblasts/chemistry , Fibroblasts/cytology , Humans , Immunohistochemistry , Macrophages/chemistry , Macrophages/cytology , Matrix Metalloproteinases/analysis , Proto-Oncogene Proteins c-sis/analysis , RNA, Messenger/analysis , Receptors, Platelet-Derived Growth Factor/analysis , Reverse Transcriptase Polymerase Chain Reaction , Synovial Membrane/cytology
11.
J Med Microbiol ; 55(Pt 2): 143-147, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16434705

ABSTRACT

Following withdrawal of immunosuppression, surfactant protein A (SP-A)-deficient and wild-type mice cleared Pneumocystis murina infection in a similar manner, but exhibited significant differences in lymphocyte populations, interleukin (IL)-6 levels and chemokine expression levels. A higher percentage of lymphocytes were detected in lung lavage fluid from SP-A-deficient mice, but more CD4+ T cells were isolated from lung tissue of wild-type mice. Higher concentrations of IL-6 were detected in lavage fluid and enhanced expression of lymphotactin and RANTES were detected in the lungs of wild-type mice. Equal levels of surfactant protein D were detected in SP-A-deficient and wild-type mice and no differences were detected in markers of lung injury between the two strains of mice. Thus, SP-A does not enhance organism clearance, but does modulate the host immune response during resolution of P. murina infection.


Subject(s)
Immunocompromised Host , Pneumocystis , Pneumonia, Pneumocystis/immunology , Pulmonary Surfactant-Associated Protein A/deficiency , Pulmonary Surfactant-Associated Protein A/genetics , Animals , Bronchoalveolar Lavage Fluid/immunology , CD4-Positive T-Lymphocytes/immunology , Cell Count , Chemokine CCL5/analysis , Dexamethasone/administration & dosage , Interleukin-6/analysis , Lung/immunology , Lymphocytes/immunology , Lymphokines/analysis , Mice , Mice, Inbred C3H , Mice, Knockout , Sialoglycoproteins/analysis
12.
Indian J Pediatr ; 72(10): 849-52, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16272656

ABSTRACT

Colostrum is the first milk produced by mammals for their young ones. This transfers the passive immunity gained by the mother to the baby. The bovine colostrum (BC) can be obtained in large quantity and has properties similar to human colostrum. It has been used for various disorders of the body. It has properties to stimulate immune system, contains growth factors and many bioactive substances needed for the body to combat with wear and tear. The BC has been used for various gastrointestinal disorders, respiratory tract infection, rheumatoid arthritis, healing injured tissues of body etc. There are not much double blind placebo-controlled trials to prove its efficacy, though a lot of experience about its good effects in various disorders is available in the literature. The dosage and duration of therapy need to be worked up. The BC has potential to treat as well to prevent certain diseases in the body. In future this will prove to be a very useful product to treat and control diseases in a natural way.


Subject(s)
Colostrum , Dietary Supplements , Abdomen/surgery , Adolescent , Animals , Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Antioxidants/analysis , Bacterial Infections/therapy , Campylobacter Infections/therapy , Cattle , Child , Child, Preschool , Colostrum/chemistry , Colostrum/immunology , Diarrhea/therapy , Double-Blind Method , Female , Gastrointestinal Diseases/chemically induced , Gastrointestinal Diseases/microbiology , Gastrointestinal Diseases/therapy , Growth Substances/analysis , Helicobacter pylori , Humans , Infant , Lymphokines/analysis , Pregnancy , Preoperative Care , Randomized Controlled Trials as Topic
13.
Circ Res ; 97(10): 1036-45, 2005 Nov 11.
Article in English | MEDLINE | ID: mdl-16224065

ABSTRACT

Platelet-derived growth factor (PDGF)-D is a member of the PDGF/vascular endothelial growth factor family that activates PDGF receptor beta (PDGFR-beta). We show that PDGF-D is highly expressed in the myocardium throughout development and adulthood, as well as by arterial vascular smooth muscle cells (vSMCs). To obtain further knowledge regarding the in vivo response to PDGF-D, we generated transgenic mice overexpressing the active core domain of PDGF-D in the heart. Transgenic PDGF-D stimulates proliferation of cardiac interstitial fibroblasts and arterial vSMCs. This results in cardiac fibrosis followed by dilated cardiomyopathy and subsequent cardiac failure. Transgenic mice also display vascular remodeling, including dilation of vessels, increased density of SMC-coated vessels, and proliferation of vSMCs, leading to a thickening of tunica media. The thickening of arterial walls is a unique feature of PDGF-D, because this is not seen when PDGF-C is overexpressed in the heart. These results show that PDGF-D, via PDGFR-beta signaling, is a potent modulator of both vascular and connective tissue growth and may provide both paracrine and autocrine stimulation of PDGFR-beta. Our data raise the possibility that this growth factor may be involved in cardiac fibrosis and atherosclerosis.


Subject(s)
Lymphokines/physiology , Muscle, Smooth, Vascular/pathology , Myocardium/pathology , Platelet-Derived Growth Factor/physiology , Receptor, Platelet-Derived Growth Factor beta/physiology , Animals , Apolipoproteins E/physiology , Atherosclerosis/etiology , Cell Proliferation , Fibroblasts/physiology , Fibrosis , Humans , Lymphokines/analysis , Lymphokines/genetics , Mice , Mice, Transgenic , Myocardium/chemistry , Platelet-Derived Growth Factor/analysis , Platelet-Derived Growth Factor/genetics , RNA, Messenger/analysis , Receptor, Platelet-Derived Growth Factor beta/agonists , Receptor, Platelet-Derived Growth Factor beta/analysis , Signal Transduction
14.
J Virol ; 79(17): 10864-74, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16103139

ABSTRACT

We report here the generation of transgenic chickens using a retroviral vector for the production of recombinant proteins. It was found that the transgene expression was suppressed when a Moloney murine leukemia virus-based retroviral vector was injected into chicken embryos at the blastodermal stage. When a concentrated viral solution was injected into the heart of developing embryos after 50 to 60 h of incubation, transgene expression was observed throughout the embryo, including the gonads. For practical production, a retroviral vector encoding an expression cassette of antiprion single-chain Fv fused with the Fc region of human immunoglobulin G1 (scFv-Fc) was injected into chicken embryos. The birds that hatched stably produced scFv-Fc in their serum and eggs at high levels (approximately 5.6 mg/ml). We obtained transgenic progeny from a transgenic chicken generated with this procedure. The transgene was stably integrated into the chromosomes of transgenic progeny. The transgenic progeny also expressed scFv-Fc in the serum and eggs.


Subject(s)
Antibodies/metabolism , Chickens/genetics , Genetic Vectors , Immunoglobulin Fc Fragments/metabolism , Immunoglobulin Fragments/metabolism , Lymphokines/metabolism , Moloney murine leukemia virus/genetics , Ovum/metabolism , Retroviridae/genetics , Sialoglycoproteins/metabolism , Animals , Animals, Genetically Modified , Antibodies/blood , Blastoderm , Chick Embryo , Chickens/metabolism , Egg White/analysis , Female , Heart , Immunoglobulin Fc Fragments/analysis , Immunoglobulin Fragments/analysis , Lymphokines/analysis , Male , Prions/immunology , Recombinant Proteins/analysis , Recombinant Proteins/biosynthesis , Sialoglycoproteins/analysis
15.
Mol Cell Biol ; 25(14): 6279-88, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15988036

ABSTRACT

Platelet-derived growth factor (PDGF) protein family members are potent mitogens and chemoattractants for mesenchymal cells. The classic PDGF ligands A and B are single-domain protein chains which are secreted as active dimers capable of activating their cognate PDGF receptors (PDGFRs). In contrast to PDGFs A and B, PDGF D contains an N-terminal complement subcomponent C1r/C1s, Uegf, and Bmp1 (CUB) domain and a C-terminal PDGF domain. PDGF D must undergo extracellular proteolytic processing, separating the CUB domain from the PDGF domain, before the PDGF domain can stimulate beta-PDGFR-mediated cell signal transduction. Here, we report that prostate carcinoma cells LNCaP and PC3 autoactivate latent full-length PDGF D into its active form under serum-independent conditions and that this autoactivation is inhibited by PAI-1, a urokinase plasminogen activator (uPA)/tissue plasminogen activator (tPA) inhibitor. Interestingly, uPA, but not the closely related protease tPA, is capable of processing recombinant latent PDGF DD into the active form. We identify the uPA cleavage site between the CUB and PDGF domains of the full-length PDGF D by mutational analysis and show that PDGF D and uPA colocalize in human prostate carcinoma. This evidence provides a direct link between uPA- and PDGF D-mediated cell signaling, which may contribute to the progression of prostate cancer.


Subject(s)
Carcinoma/enzymology , Lymphokines/metabolism , Platelet-Derived Growth Factor/metabolism , Prostatic Neoplasms/enzymology , Urokinase-Type Plasminogen Activator/metabolism , Amino Acid Sequence , Aprotinin/pharmacology , Carcinoma/chemistry , Cell Line, Tumor , DNA Mutational Analysis , Humans , Lymphokines/analysis , Lymphokines/genetics , Male , Molecular Sequence Data , Plasminogen Activator Inhibitor 1/pharmacology , Platelet-Derived Growth Factor/analysis , Platelet-Derived Growth Factor/genetics , Prostatic Neoplasms/chemistry , Urokinase-Type Plasminogen Activator/analysis , Urokinase-Type Plasminogen Activator/antagonists & inhibitors
16.
Vaccine ; 23(19): 2439-53, 2005 Mar 31.
Article in English | MEDLINE | ID: mdl-15752830

ABSTRACT

The aim was to develop T cell costimulatory molecules that are broadly applicable to augment anti-tumor immune responses upon application of a virus-modified tumor vaccine to cancer patients. We generated recombinant bispecific single-chain antibodies with one specificity directed against the CD3 or the CD28 antigen on human T cells and the other against the viral target molecule hemagglutinin-neuraminidase (HN) of Newcastle Disease Virus (NDV). By re-directing unstimulated primary human T cells against HN-expressing NDV-infected tumor cells, the bispecific molecule bsHN-CD3 cross-linked effector and target cells and rapidly induced cytotoxicity at nanomolar concentrations. The bsHN-CD28 molecule exerted T cell co-stimulatory function. Maximal T cell activation was achieved with tumor cells infected by NDV and modified with both new stimulatory molecules. This was revealed by T cell proliferation, upregulation of CD69 and CD25 and by release of cytokines, interferons and chemokines. The new molecules combine high-effectivity with specificity and safety.


Subject(s)
Antibodies/immunology , CD28 Antigens/immunology , CD3 Complex/immunology , Cancer Vaccines/immunology , Newcastle disease virus/immunology , T-Lymphocytes/immunology , Animals , Antibodies/genetics , Antigens, CD/analysis , Antigens, Differentiation, T-Lymphocyte/analysis , Antigens, Viral/genetics , Antigens, Viral/immunology , Cancer Vaccines/genetics , Cell Line, Tumor , Cytokines/analysis , Cytotoxicity, Immunologic , HN Protein/immunology , Humans , Immunoglobulin Variable Region/genetics , Immunoglobulin Variable Region/immunology , Lectins, C-Type , Lymphocyte Activation , Lymphokines/analysis , Mice , Receptors, Interleukin-2/analysis
18.
Transpl Immunol ; 12(1): 63-72, 2003.
Article in English | MEDLINE | ID: mdl-14551033

ABSTRACT

To elucidate the possible immunoregulatory role of nitric oxide (NO) in cellular xenograft rejection we performed rat-to-mouse skin xenotransplantation. The rat skin engrafted mice were treated with the inducible NO synthase (iNOS) inhibitors, aminoguanidine (AMG, 200 mg/kg) and NG-nitro-L-arginine methyl ester (L-NAME, 60 mg/kg) every other day until rejection. Skin xenograft survival was monitored and immune cell infiltration and intragraft cytokine and chemokine mRNA expressions were analyzed 7 days after grafting. Compared with the control mice, the AMG- and L-NAME treated mice showed delayed xenograft rejection by approximately 3 days (8.9 +/- 0.7 days vs. 11.7 +/- 1.2 and 12.0 +/- 0.9 days, respectively). Infiltrations of CD11b+, MOMA-2+ cells and neutrophils were significantly reduced in both AMG- and L-NAME treated graft but CD4+ and CD8+ cells were not. The expression of cytokines such as IL-1beta, IL-2, IL-6, IL-12 and IFN-gamma in AMG- and L-NAME treated grafts were significantly decreased (P<0.01), whereas IL-10, TNF-alpha and TGF-beta1 were unchanged or enhanced. Additionally, the expressions of CC-chemokines, such as RANTES and MIP-1alpha, were significantly reduced (P<0.01) whereas the expressions of CXC-chemokines, such as IP-10 and MIG, were unchanged. These results imply that prolonged rat-to-mouse skin xenograft survival by iNOS inhibitors may be due to the selective inhibition of pro-inflammatory cytokines and chemokines and suggest the possible regulatory role of NO in cytokine and chemokine expressions during xenotransplant rejection.


Subject(s)
Chemokines, CC/genetics , Cytokines/genetics , Graft Survival/drug effects , Nitric Oxide Synthase/antagonists & inhibitors , Skin Transplantation , Animals , CD11b Antigen/analysis , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Cell Count , Chemokines, CC/metabolism , Cytokines/metabolism , Down-Regulation/drug effects , Enzyme Inhibitors/pharmacology , Female , Gene Expression Regulation/drug effects , Guanidines/pharmacology , Immunohistochemistry , Lymphokines/analysis , Lymphokines/drug effects , Lymphokines/genetics , Mice , Mice, Inbred BALB C , NG-Nitroarginine Methyl Ester/pharmacology , Neutrophils/cytology , Neutrophils/drug effects , Neutrophils/immunology , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type II , RNA, Messenger/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Inbred Lew , Skin/drug effects , Skin/metabolism , Transplantation, Heterologous
19.
Cancer ; 98(8): 1658-63, 2003 Oct 15.
Article in English | MEDLINE | ID: mdl-14534882

ABSTRACT

BACKGROUND: Multiple epidemiologic and histologic studies have suggested that ovarian endometriosis can give rise to malignant ovarian tumors, primarily those of epithelial origin. The progression of endometriosis to endometriosis-associated ovarian carcinoma (EAOC) has not been investigated thoroughly and is poorly understood at best. Using immunohistochemical methods, we compared the differential expression patterns of various cytokines and growth factors in atypical endometriosis (AE) and EAOC. METHODS: Using the Johns Hopkins Pathology Data Bank, tissue blocks from patients diagnosed with EAOC or AE were identified. Tissue blocks were stained for 4 markers: vascular endothelial growth factor (VEGF), Ki-67, estrogen receptor (ER), and progesterone receptor (PR). RESULTS: Seventeen cases of EAOC and 8 cases of AE were identified. Staining for VEGF was documented in 16 of 17 (94%) EAOC tissue blocks and in only 1 of 8 (12.5%) AE tissue blocks (P < 0.0001). Only 4 of the 17 (23%) EAOC tissue blocks exhibited positive staining for ER, compared with 8 of 8 (100%) AE tissue blocks (P = 0.0005). Positive staining for PR was noted in only 6 of 17 (35%) EAOC samples but was present in 8 of 8 (100%) AE samples (P = 0.003). Seventy percent of EAOC samples exhibited positive staining for Ki-67, compared with 37.5% of AE samples (P = 0.19). CONCLUSIONS: EAOC appears to be associated with overexpression of VEGF and reduced expression of both ER and PR. Variations in VEGF expression may be associated with the malignant transformation of endometriosis and may present both diagnostic and therapeutic options for the treatment of ovarian malignancies.


Subject(s)
Endometriosis/complications , Endothelial Growth Factors/analysis , Intercellular Signaling Peptides and Proteins/analysis , Lymphokines/analysis , Ovarian Neoplasms/chemistry , Receptors, Estrogen/analysis , Receptors, Progesterone/analysis , Female , Humans , Immunohistochemistry , Ki-67 Antigen/analysis , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
20.
Br J Plast Surg ; 56(7): 653-9, 2003 Oct.
Article in English | MEDLINE | ID: mdl-12969663

ABSTRACT

The induction of endogenous vascular endothelial growth factor (VEGF) production in the skin flap with ischemic injury and the effect of exogenous VEGF on survival of the ischemic skin flap were studied in rats. A dorsal flap model (3x10 cm(2)) was used in this study. In Part I, biopsies were taken from the flap at 2.5, 5.5, and 8.5 cm distances from the distal edge at 0, 6, 12, and 24 h after the flaps were sutured. Malonyldialdehyde (MDA) and VEGF(165) protein level were measured. In Part II, exogenous VEGF (1 microg/ml) was injected subdermally into the flaps in 14 rats before the flaps were replaced. Flaps that received a saline injection were used as the controls. The skin paddle survival was measured on postoperative day five. The results showed that the MDA level in the distal part of the flap significantly increased at 24 h postoperatively when compared to MDA in other parts of the flap. However, VEGF levels in the distal part of the flap significantly decreased when compared to the middle part of the flap. Subdermal injection of exogenous VEGF to the distal area of the flap could significantly improve survival of the distal flap (89% of total skin paddle) when compared to the control, which had a 64% mean percent survival. We conclude that production of endogenous VEGF protein is significantly increased in the skin flap with mild ischemia, but decreased in the flap with severe ischemia. Administration of exogenous VEGF could significantly enhance survival of ischemic flaps.


Subject(s)
Endothelial Growth Factors/analysis , Graft Survival/drug effects , Intercellular Signaling Peptides and Proteins/analysis , Lymphokines/analysis , Surgical Flaps/physiology , Animals , Endothelial Growth Factors/therapeutic use , Enzyme-Linked Immunosorbent Assay/methods , Graft Survival/physiology , Intercellular Signaling Peptides and Proteins/therapeutic use , Ischemia/drug therapy , Lipid Peroxidation , Lymphokines/therapeutic use , Male , Malondialdehyde/analysis , Rats , Rats, Sprague-Dawley , Recombinant Proteins/therapeutic use , Skin/blood supply , Skin/metabolism , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
SELECTION OF CITATIONS
SEARCH DETAIL