Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
J Ayurveda Integr Med ; 8(3): 211-212, 2017.
Article in English | MEDLINE | ID: mdl-28648875
2.
Circulation ; 104(25): 3103-8, 2001 Dec 18.
Article in English | MEDLINE | ID: mdl-11748108

ABSTRACT

BACKGROUND: Inflammation is implicated in atherogenesis and plaque disruption. Toll-like receptor 2 (TLR-2) and TLR-4, a human homologue of drosophila Toll, play an important role in the innate and inflammatory signaling responses to microbial agents. To investigate a potential role of these receptors in atherosclerosis, we assessed the expression of TLR-2 and TLR-4 in murine and human atherosclerotic plaques. METHODS AND RESULTS: Aortic root lesions of high-fat diet-fed apoE-deficient mice (n=5) and human coronary atherosclerotic plaques (n=9) obtained at autopsy were examined for TLR-4 and TLR-2 expression by immunohistochemistry. Aortic atherosclerotic lesions in all apoE-deficient mice expressed TLR-4, whereas aortic tissue obtained from control C57BL/6J mice showed no TLR-4 expression. All 5 lipid-rich human plaques expressed TRL-4, whereas the 4 fibrous plaques and 4 normal human arteries showed no or minimal expression. Serial sections and double immunostaining showed TLR-4 colocalizing with macrophages both in murine atherosclerotic lesions and at the shoulder region of human coronary artery plaques. In contrast to TLR-4, none of the plaques expressed TLR-2. Furthermore, basal TLR-4 mRNA expression by human monocyte-derived macrophages was upregulated by ox-LDL in vitro. CONCLUSIONS: Our study demonstrates that TLR-4 is preferentially expressed by macrophages in murine and human lipid-rich atherosclerotic lesions, where it may play a role to enhance and sustain the innate immune and inflammatory responses. Moreover, upregulation of TLR-4 in macrophages by oxidized LDL suggests that TLR-4 may provide a potential pathophysiological link between lipids and infection/inflammation and atherosclerosis.


Subject(s)
Arteriosclerosis/metabolism , Drosophila Proteins , Lipid Metabolism , Lipoproteins, LDL/pharmacology , Macrophages/drug effects , Membrane Glycoproteins/drug effects , Receptors, Cell Surface/drug effects , Animals , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Arteriosclerosis/pathology , Coronary Vessels/chemistry , Coronary Vessels/pathology , Gene Expression Regulation/drug effects , Humans , Immunohistochemistry , Macrophages/metabolism , Macrophages/pathology , Membrane Glycoproteins/biosynthesis , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA, Messenger/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Cell Surface/biosynthesis , Receptors, Cell Surface/genetics , Reverse Transcriptase Polymerase Chain Reaction , Toll-Like Receptor 2 , Toll-Like Receptor 4 , Toll-Like Receptors
3.
J Am Coll Cardiol ; 35(2): 493-501, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10676700

ABSTRACT

OBJECTIVES: To examine the effect of a polymeric-based periadventitial delivery of a nitric oxide (NO)-releasing diazeniumdiolate, spermine/NO (SPER/NO), on balloon injury-induced neointimal hyperplasia in rat ileofemoral arteries. BACKGROUND: Reduced local bioavailability and adverse side effects limit systemic administration of NO to modulate vascular response to injury. METHODS: A polylactic-polyglycolic acid polymeric matrix containing 2.5% SPER/NO (w/w) was applied around the injured arteries. Quantitative histomorphometry was performed at day 14, proliferating cell nuclear antigen (PCNA) immunohistochemistry at day 3 to assess effects on smooth muscle proliferation and electrophoretic mobility shift assay to evaluate effects on transcription factor, nuclear factor-kappaB (NF-kappaB). RESULTS: Treatment with SPER/NO reduced the intimal area (0.011 +/- 0.009 vs. 0.035 +/- 0.006 mm2 control, p < 0.01) and the intima to media ratio (0.089 +/- 0.062 vs. 0.330 +/- 0.057 control, p < 0.005). Spermine/nitric oxide produced a profound inhibition of PCNA-positive cells (>75%, p < 0.005) and significantly suppressed the injury-induced activation of NF-kappaB. Vascular cyclic guanosine monophosphate (cGMP) levels were elevated after treatment with the SPER/NO (0.28 +/- 0.03 vs. 0.17 +/- 0.02 pmol/mg tissue control, p < 0.01). The inhibitory effects on neointimal proliferation were localized to the site of application of SPER/NO and were not associated with any changes in platelet aggregation or bleeding time. Neither SPER nor polymer alone had any significant effects on any of the variables examined. CONCLUSIONS: Polymeric-based perivascular delivery of a NO donor produces a marked localized inhibition of neointimal proliferation in balloon-injured arteries. This phenomenon is associated with suppression of NF-kappaB activation and elevation of the vascular cGMP at the site of injury.


Subject(s)
Angioplasty, Balloon/adverse effects , Drug Delivery Systems , NF-kappa B/metabolism , Nitric Oxide/metabolism , Spermine/administration & dosage , Tunica Intima/drug effects , Animals , Arteries/drug effects , Arteries/injuries , Arteries/pathology , Bleeding Time , Cell Division/drug effects , Cyclic GMP/metabolism , Hyperplasia/metabolism , Hyperplasia/pathology , Hyperplasia/prevention & control , Lactic Acid , Male , Platelet Aggregation , Polyglycolic Acid , Polylactic Acid-Polyglycolic Acid Copolymer , Polymers , Proliferating Cell Nuclear Antigen/metabolism , Rats , Rats, Sprague-Dawley , Tunica Intima/injuries , Tunica Intima/pathology
4.
Circulation ; 99(24): 3103-9, 1999 Jun 22.
Article in English | MEDLINE | ID: mdl-10377072

ABSTRACT

BACKGROUND: Matrix metalloproteinases (MMPs) are expressed in atherosclerotic plaques, where in their active form, they may contribute to vascular remodeling and plaque disruption. In this study, we tested the hypothesis that membrane type 1 MMP (MT1-MMP), a novel transmembrane MMP that activates pro-MMP-2 (gelatinase A), is expressed in human atherosclerotic plaques and that its expression is regulated by proinflammatory molecules. METHODS AND RESULTS: MT1-MMP expression was examined in normal and atherosclerotic human arteries by immunocytochemistry with specific antibodies. MT1-MMP expression in human saphenous vein-derived smooth muscle cells (SMCs) maintained in tissue culture was determined under basal conditions and in response to proinflammatory molecules (interleukin [IL]-1alpha, tumor necrosis factor [TNF]-alpha, and oxidized LDL [ox-LDL]) by use of Northern blot and ribonuclease protection assays for mRNA, Western blot and immunoprecipitation for protein, and gelatin zymography for catalytic activity. Medial SMCs of normal vessel wall expressed MT1-MMP. In atherosclerotic arteries, MT1-MMP expression was noted within the complex atheroma colocalizing with SMCs and macrophages (Mphi). Cultured SMCs constitutively expressed MT1-MMP mRNA and protein, which increased 2- to 4-fold over control in a time-dependent manner within 4 to 8 hours of exposure to IL-1alpha, TNF-alpha, and ox-LDL (thiobarbituric acid-reactive substances, 13.4 nmol/mg LDL protein), whereas native LDL had no effect. Flow cytometry revealed MT1-MMP expression by human monocyte-derived Mphi, which increased 3.8-fold over baseline within 6 hours after exposure to 10 ng/mL TNF-alpha. CONCLUSIONS: This study demonstrates that MT1-MMP, an activator of pro-MMP-2, is expressed by SMCs and Mphi in human atherosclerotic plaques. Furthermore, proinflammatory molecules upregulate MT1-MMP expression in vascular SMCs and Mphi. Thus, activation of SMCs and Mphi by proinflammatory molecules may influence extracellular matrix remodeling in atherosclerosis by regulating MT1-MMP expression.


Subject(s)
Arteriosclerosis/metabolism , Gene Expression Regulation, Enzymologic/immunology , Inflammation Mediators/metabolism , Metalloendopeptidases/genetics , Muscle, Smooth, Vascular/enzymology , Antibodies, Monoclonal , Arteriosclerosis/pathology , Blotting, Northern , Cells, Cultured , Coronary Vessels/chemistry , Coronary Vessels/enzymology , Enzyme Precursors/metabolism , Flow Cytometry , Gelatinases/analysis , Gelatinases/biosynthesis , Gelatinases/immunology , Gene Expression Regulation, Enzymologic/drug effects , Humans , Interleukin-1/pharmacology , Lipoproteins/metabolism , Lipoproteins, LDL/pharmacology , Macrophages/chemistry , Macrophages/enzymology , Matrix Metalloproteinase 2 , Matrix Metalloproteinases, Membrane-Associated , Metalloendopeptidases/analysis , Metalloendopeptidases/biosynthesis , Metalloendopeptidases/immunology , Metalloendopeptidases/metabolism , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/pathology , RNA, Messenger/analysis , Saphenous Vein/cytology , Tissue Inhibitor of Metalloproteinase-2/analysis , Tissue Inhibitor of Metalloproteinase-2/immunology , Tumor Necrosis Factor-alpha/pharmacology
5.
J Biol Chem ; 274(17): 11924-9, 1999 Apr 23.
Article in English | MEDLINE | ID: mdl-10207013

ABSTRACT

We investigated whether inflammatory cytokines or oxidized low density lipoproteins (Ox-LDL) present in human atheroma modulate extracellular matrix degradation by inducing membrane type 1-matrix metalloproteinase (MT1-MMP) expression. Cultured human endothelial cells (EC) constitutively expressed MT1-MMP mRNA and protein with enzymatic activity. Tumor necrosis factor-alpha (TNF-alpha), interleukin-1alpha, or interleukin-1beta caused a time-dependent increase in the steady-state MT1-MMP mRNA levels within 4 h of exposure, peaking about 4-fold by 6 h, and remaining elevated for 12 h. Increased MT1-MMP mRNA correlated with a 2.5-fold increase in MT1-MMP protein in EC membranes. Ox-LDL also increased MT1-MMP mRNA levels that varied with the duration of exposure and degree of LDL oxidation. The increase in MT1-MMP mRNA occurred within 6 h of exposure to Ox-LDL and peaked over 3-fold by 6 h. Ox-LDL, but not native LDL, increased MT1-MMP protein by 2-fold in EC membranes. A combination of TNF-alpha and Ox-LDL was additive in increasing MT1-MMP expression. Nuclear run-on assays showed that TNF-alpha or Ox-LDL augmented steady-state mRNA levels by increased transcription of the MT1-MMP gene. These findings indicate that activation of EC by inflammatory cytokines and/or Ox-LDL increase MT1-MMP expression. Since MT1-MMP promotes matrix degradation by activating pro-MMP-2, these results suggest a novel mechanism whereby cytokines or Ox-LDL may influence extracellular matrix remodeling.


Subject(s)
Endothelium, Vascular/metabolism , Interleukin-1/metabolism , Lipoproteins, LDL/metabolism , Metalloendopeptidases/metabolism , Tumor Necrosis Factor-alpha/metabolism , Base Sequence , Cells, Cultured , DNA Primers , Endothelium, Vascular/cytology , Enzyme Activation , Gelatinases/metabolism , Gene Expression Regulation , Humans , Matrix Metalloproteinase 2 , Matrix Metalloproteinases, Membrane-Associated , Metalloendopeptidases/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcription, Genetic
6.
Circulation ; 99(8): 993-8, 1999 Mar 02.
Article in English | MEDLINE | ID: mdl-10051290

ABSTRACT

BACKGROUND: Macrophages in human atherosclerotic plaques produce a family of matrix metalloproteinases (MMPs), which may influence vascular remodeling and plaque disruption. Because oxidized LDL (ox-LDL) is implicated in many proatherogenic events, we hypothesized that ox-LDL would regulate expression of MMP-9 and tissue inhibitor of metalloproteinase-1 (TIMP-1) in monocyte-derived macrophages. MWRHOSA AND RESULTS: Mononuclear cells were isolated from normal human subjects with Ficoll-Paque density gradient centrifugation, and adherent cells were allowed to differentiate into macrophages during 7 days of culture in plastic dishes. On day 7, by use of serum-free medium, the macrophages were incubated with various concentrations of native LDL (n-LDL) and copper-oxidized LDL. Exposure to ox-LDL (10 to 50 microg/mL) increased MMP-9 mRNA expression as analyzed by Northern blot, protein expression as measured by ELISA and Western blot, and gelatinolytic activity as determined by zymography. The increase in MMP-9 expression was associated with increased nuclear binding of transcription factor NF-kappaB and AP-1 complex on electromobility shift assay. In contrast, ox-LDL (10 to 50 microg/mL) decreased TIMP-1 expression. Ox-LDL-induced increase in MMP-9 expression was abrogated by HDL (100 microg/mL). n-LDL had no significant effect on MMP-9 or TIMP-1 expression. CONCLUSIONS: These data demonstrate that unlike n-LDL, ox-LDL upregulates MMP-9 expression while reducing TIMP-1 expression in monocyte-derived macrophages. Furthermore, HDL abrogates ox-LDL-induced MMP-9 expression. Thus, ox-LDL may contribute to macrophage-mediated matrix breakdown in the atherosclerotic plaques, thereby predisposing them to plaque disruption and/or vascular remodeling.


Subject(s)
Collagenases/genetics , Gene Expression Regulation/drug effects , Lipoproteins, LDL/pharmacology , Macrophages/metabolism , Tissue Inhibitor of Metalloproteinase-1/genetics , Humans , Lipoproteins, HDL/pharmacology , Lipoproteins, LDL/metabolism , Macrophages/drug effects , Matrix Metalloproteinase 9 , NF-kappa B/metabolism , Oxidation-Reduction , RNA, Messenger/analysis , Transcription Factor AP-1/metabolism
7.
DNA Cell Biol ; 17(9): 799-809, 1998 Sep.
Article in English | MEDLINE | ID: mdl-9778039

ABSTRACT

Macrophage colony-stimulating factor (M-CSF; CSF-1) is a member of a complex network of cytokines that regulate monocytic cell development and activity. It is produced in nearly all organs by cell types commonly found in connective tissue, including fibroblasts and monocytes. Whether different cell types share common or have divergent mechanisms for regulating CSF-1 gene expression is not known. To address this question, the identity of cis-acting elements and cognate trans-acting factors was characterized in a region of the CSF-1 promoter known to be more active in monocytes than in fibroblasts. The results of DNase I protection assays performed with fibroblast- or monocyte-derived nuclear extracts revealed a difference in the pattern of DNA-binding proteins. One protected region, common to both fibroblasts and monocytes, spans a putative phorbol ester-responsive element (TRE), and binding to the TRE by AP1 was verified with antibodies directed against c-fos and c-jun family members. Mutational analysis revealed that the TRE is required for CSF-1 gene expression in proliferating fibroblasts and monocytes. Binding of a second putative trans-acting factor, preferentially expressed in fibroblasts, to the region immediately upstream of the TRE was also detected. Screening a mouse expression library with oligonucleotides spanning the putative cis-acting element identified cellular nucleic acid-binding protein (CNBP) as the cognate binding activity, and antiserum to CNBP disrupted the electromobility shift assay complex. Mutational analysis revealed that loss of CNBP binding leads to a decrease in CSF-1 promoter activity in fibroblasts but has no effect on CSF-1 promoter activity in monocytes. Our results demonstrate that control of CSF-1 gene expression in monocytes and fibroblasts is mediated by common and cell type-specific trans-acting factors.


Subject(s)
DNA-Binding Proteins/metabolism , Macrophage Colony-Stimulating Factor/genetics , Promoter Regions, Genetic/genetics , RNA-Binding Proteins , Transcription Factor AP-1/metabolism , Animals , Base Sequence , Binding, Competitive , Cell Extracts , Cell Line , Cell Nucleus , DNA/metabolism , DNA Methylation , Fibroblasts , Mice , Molecular Sequence Data , Monocytes , Mutation , Tetradecanoylphorbol Acetate , Transcriptional Activation/genetics , Transfection
8.
J Clin Invest ; 101(12): 2702-10, 1998 Jun 15.
Article in English | MEDLINE | ID: mdl-9637704

ABSTRACT

Previous studies of osteopetrotic (op) mice lacking macrophage colony-stimulating factor (M-CSF) have revealed an inhibition of atherosclerosis development in the apolipoprotein E (apo E)-deficient model and in a diet-induced model. Using LDL receptor-deficient mice, we now show that atheroma development depends on M-CSF concentration, as not only did homozygous osteopetrotic (op/op) mice have dramatically reduced lesions (approximately 0.3% of control lesion size) but heterozygous (op/+) mice had lesions < 1% of controls. Mice heterozygous for the op mutation (op/+) had plasma levels of M-CSF about half those in controls (+/+). The finding that an approximately 2-fold reduction in M-CSF expression reduced lesion size approximately 100-fold suggests the requirement for a threshold level of M-CSF. The effect of M-CSF on atherosclerosis did not appear to be mediated either by changes in plasma lipoprotein levels or alterations in the number of circulating monocytes, since both op/op and op/+ mice exhibited higher levels of atherogenic lipoprotein particles and (op/+) mice showed a near normal number of circulating monocytes. LDL receptor-null littermates of genotypes from op/op, op/+, to +/+ showed monocyte differentials of approximately 4.5, 8, and 10%, respectively. Taken together, these results suggest that the effects of M-CSF on atherogenesis may not be mediated by expression of M-CSF systemically or by modulation of the number of circulating monocytes. These studies support the conclusion that M-CSF participates critically in fatty streak formation and progression to a complex fibrous lesion.


Subject(s)
Arteriosclerosis/genetics , Macrophage Colony-Stimulating Factor/genetics , Osteopetrosis/genetics , Receptors, LDL/genetics , Animals , Arteriosclerosis/metabolism , Arteriosclerosis/pathology , Cell Differentiation , Heterozygote , Homozygote , Macrophage Colony-Stimulating Factor/deficiency , Macrophages/pathology , Mice , Monocytes/pathology , Mutation , Osteopetrosis/metabolism , Osteopetrosis/pathology , Receptors, LDL/deficiency
9.
Biol Reprod ; 57(5): 1193-201, 1997 Nov.
Article in English | MEDLINE | ID: mdl-9369187

ABSTRACT

The major objectives of the present study were to document the temporal and stage-specific acceleration of germ cell apoptosis in adult rats after selective suppression of pituitary gonadotropins by GnRH antagonist (GnRH-A) treatment, and to examine the possibility that apoptosis is the sole mechanism of germ cell death in response to hormonal deprivation. Groups of adult male rats were given a daily injection of a vehicle for 14 days or GnRH-A (1.25 mg/kg BW) for 2, 5, 7, and 14 days. Analysis of testicular apoptotic DNA fragmentation revealed a detectable increase at Day 5 and a maximal increase at 14 days after treatment. In situ analysis of germ cell apoptosis fully corroborated the observed increase in the degree of DNA fragmentation with time and also revealed a stage-related activation of apoptosis of specific germ cells. A low incidence (0.06-0.09) of germ cell apoptosis (expressed as numbers per Sertoli cell) was detectable at stages I, IX-XI, and XII-XIV in control rats. Mean incidence of apoptotic germ cells specifically at stages VII-VIII increased significantly (0.40 +/- 0.06) by Day 5 and increased another 2.2-fold (over the 5-day treatment values) on Day 7 after GnRH-A treatment as compared to values in controls, where no apoptosis was detected. Significantly increased incidence of apoptosis at stages IX-XI (0.37 +/- 0.05) over control values (0.07 +/- 0.01) was noted by Day 7. Within the study paradigm, the highest number of dying cells occurred by Day 14, at which time a modest but significant (p < 0.05) increase in the incidence of apoptosis was also noted at stages I, II-IV, V-VI, and XII-XIV in comparison with control values. Stages VII-VIII and IX-XI still exhibited the higher number of cells undergoing apoptosis (0.97 +/- 0.22, and 1.03 +/- 0.22, respectively). Comparison between rates of apoptosis and cell degeneration measured at stages VII-VIII demonstrated an intimate association (r = 0.94; p < 0.001) between apoptosis and germ cell loss, strongly supporting the concept that germ cell death (at these stages) after removal of hormonal support in the adult rat occurs almost exclusively via apoptosis.


Subject(s)
Apoptosis/physiology , Germ Cells/physiology , Gonadotropins/deficiency , Testis/cytology , Animals , DNA Fragmentation/drug effects , Follicle Stimulating Hormone/blood , Gonadotropin-Releasing Hormone/antagonists & inhibitors , Luteinizing Hormone/blood , Male , Rats , Rats, Sprague-Dawley , Testis/drug effects , Testis/metabolism , Testosterone/blood , Testosterone/metabolism
10.
Am J Pathol ; 150(5): 1687-99, 1997 May.
Article in English | MEDLINE | ID: mdl-9137093

ABSTRACT

Previous in vitro and in vivo studies have suggested that macrophage colony-stimulating factor (M-CSF) plays a role in atherogenesis. To examine this hypothesis, we have studied atherogenesis in osteopetrotic (op/op) mice, which lack M-CSF due to a structural gene mutation. Atherogenesis was induced either by feeding the mice a high fat, high cholesterol diet or by crossing op mice with apolipoprotein E (apo E) knockout mice to generate mice lacking both M-CSF and apo E. In both the dietary and apo E knockout models, M-CSF deficiency resulted in significantly reduced atherogenesis. For example, in the apo E knockout model, homozygosity for the op mutation totally abolished aortic atherogenesis in male mice and reduced the size of the lesions approximately 97% in female mice. Mice heterozygous for the op mutation also exhibited a significant decrease in lesion size. Among apo E knockout mice, the frequency of atherosclerosis in aortic arch was 0/6 (op/op), 1/15 (op/+), and 12/16 (+/+). The effect of the M-CSF on atherosclerosis did not appear to be mediated by changes in plasma lipoproteins, as the op mice exhibited higher levels of atherogenic lipoprotein particles. The effects of the op mutation on atherogenesis may have resulted from decreased circulating monocytes, reduced tissue macrophages, or diminished arterial M-CSF.


Subject(s)
Arteriosclerosis/genetics , Macrophage Colony-Stimulating Factor/physiology , Osteopetrosis/genetics , Animals , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Arteriosclerosis/etiology , Arteriosclerosis/pathology , Calcinosis/etiology , Calcinosis/genetics , Calcinosis/pathology , Diet, Atherogenic , Female , Hypercholesterolemia/etiology , Hypercholesterolemia/genetics , Hypercholesterolemia/pathology , Macrophage Colony-Stimulating Factor/deficiency , Macrophage Colony-Stimulating Factor/genetics , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout
11.
Proc Natl Acad Sci U S A ; 94(10): 5296-301, 1997 May 13.
Article in English | MEDLINE | ID: mdl-9144231

ABSTRACT

In Alzheimer disease (AD), neurons are thought to be subjected to the deleterious cytotoxic effects of activated microglia. We demonstrate that binding of amyloid-beta peptide (Abeta) to neuronal Receptor for Advanced Glycation Endproduct (RAGE), a cell surface receptor for Abeta, induces macrophage-colony stimulating factor (M-CSF) by an oxidant sensitive, nuclear factor kappaB-dependent pathway. AD brain shows increased neuronal expression of M-CSF in proximity to Abeta deposits, and in cerebrospinal fluid from AD patients there was approximately 5-fold increased M-CSF antigen (P < 0.01), compared with age-matched controls. M-CSF released by Abeta-stimulated neurons interacts with its cognate receptor, c-fms, on microglia, thereby triggering chemotaxis, cell proliferation, increased expression of the macrophage scavenger receptor and apolipoprotein E, and enhanced survival of microglia exposed to Abeta, consistent with pathologic findings in AD. These data delineate an inflammatory pathway triggered by engagement of Abeta on neuronal RAGE. We suggest that M-CSF, thus generated, contributes to the pathogenesis of AD, and that M-CSF in cerebrospinal fluid might provide a means for monitoring neuronal perturbation at an early stage in AD.


Subject(s)
Alzheimer Disease/physiopathology , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/pharmacology , Glycation End Products, Advanced/metabolism , Macrophage Colony-Stimulating Factor/biosynthesis , Neurons/physiology , Receptors, Immunologic/physiology , Alzheimer Disease/cerebrospinal fluid , Animals , Cells, Cultured , Humans , Inflammation , Macrophage Colony-Stimulating Factor/cerebrospinal fluid , Mice , NF-kappa B/metabolism , Neuroblastoma , Neurons/drug effects , Oxidative Stress , Receptor for Advanced Glycation End Products , Recombinant Fusion Proteins/biosynthesis , Reference Values , Transfection , Tumor Cells, Cultured , Vascular Cell Adhesion Molecule-1/biosynthesis
12.
Biol Reprod ; 56(4): 954-63, 1997 Apr.
Article in English | MEDLINE | ID: mdl-9096878

ABSTRACT

Erectile dysfunction is mainly due to the inability of the cavernosal smooth muscle of the penis to undergo complete relaxation. In the aging rat model, erectile dysfunction is accompanied by a reduction of penile smooth muscle compliance and, in very old animals, by a decrease in penile nitric oxide synthase (NOS), which is responsible for the synthesis of the mediator of penile erection, nitric oxide (NO). We have investigated whether the stimulation of penile NOS expression by local induction or gene therapy can mitigate erectile dysfunction in the aged rat. A mix of iNOS (inducible NOS) inducers was continuously delivered to the penises of 5- ("adult"), 20- ("old"), and 30- ("very old") mo-old rats for 3-6 days, and the erectile response to electrical field stimulation of the cavernosal nerve was measured. The erectile dysfunction observed in old and very old rats as compared to adult animals was ameliorated by treatment with iNOS inducers. Penile iNOS was detectable in the penis of these rats by Western blot, NADPH diaphorase, and NOS activity assays. Inducible NOS was inducible in vitro in both rat and human corpora cavernosal tissue and in rat penile smooth muscle cells (RPSMC), as shown by Western blots. However, NO synthesis in cavernosal tissue upon iNOS protein induction remained low, indicating that the increased NOS levels were under physiological control. The iNOS cDNA was cloned from induced RPSMC mRNA and generated by reverse transcriptase polymerase chain reaction (RT-PCR) from induced human penile smooth muscle cells and corporal tissue. The coding regions from both the rat (RPiNOS) and human (HPiNOS) penile iNOS showed several amino acid differences from their analogous isoform in nonpenile tissues. RPiNOS cDNA injected into the penis mitigated the aging-associated erectile dysfunction. The iNOS construct was detected in cavernosal tissue by PCR, and its expression by RT-PCR and Western blots. These results open the way for the possible use of NOS isoforms in the management of erectile dysfunction.


Subject(s)
Aging/metabolism , Muscle, Smooth/enzymology , Nitric Oxide Synthase/biosynthesis , Penile Erection/physiology , Penis/enzymology , Amino Acid Sequence , Animals , Base Sequence , Cells, Cultured , Cloning, Molecular , Codon , Enzyme Induction , Erectile Dysfunction/therapy , Genetic Therapy , Humans , Male , NADPH Dehydrogenase/analysis , Nitric Oxide Synthase/analysis , Penis/growth & development , Penis/physiology , Polymerase Chain Reaction , Rats , Recombinant Proteins/biosynthesis
13.
Endocrinology ; 137(12): 5407-14, 1996 Dec.
Article in English | MEDLINE | ID: mdl-8940364

ABSTRACT

Stem cell factor (SCF) gene expression is regulated by FSH in testicular Sertoli cells. Many functions of FSH are mediated through the second messenger cAMP. We show that cAMP activates transcription of the human SCF promoter in a Sertoli cell line. The human SCF promoter was cloned in cosmid vector pWE15, and its DNA sequence was determined for the promoter region extending 2.3 kilobase pairs upstream from the translation start site at +184 bp. The in vivo messenger RNA (mRNA) start site, by primer-extension studies, was located in exon 1 at +109 bp in human testis mRNA, and at +99 bp in mouse SF7 Sertoli cell line or GC1 germ cell line mRNA. To test which regions of the SCF promoter are necessary for regulation by cAMP, a series of 5'-end deletions of this region were cloned onto the luciferase reporter gene in plasmid pXP1. The SCF promoter region was fused to luciferase downstream (at +120) from its +109 mRNA start site, extending upstream a variable distance to BstXI (-162), BamHI (-313), Bgl2 (-853), or XbaI (-2185). The shortest of these fragments extending only to -162 bp, contains possible SP1 and AP-2 elements. When mouse Sertoli SF7 or human JEG.3 cell lines were transfected with these plasmids, all of the mutants were regulated by 8Br-cAMP or forskolin, as expected for the SCF gene, whereas FSH and TPA had no effect. In the shortest promoter deletion -162, luciferase expression from SF7 cells in serum-free media was at a moderate basal level, but it was induced in six h about 2-fold by 8Br-cAMP, and over 7-fold by forskolin (an adenylate cyclase activator) to high levels, similar to the SV40 positive control promoter. In SCF-luc plasmids extending to -853 or -2185, luciferase expression was still inducible by 8Br-cAMP and forskolin to high levels, but basal promoter activity was repressed to levels over 15-fold lower, in both the absence or presence of testosterone in the media for SF7 cells. The distal portion of the human SCF promoter (between -313 and -853, and also -853 and -2185) inhibits the basal level of transcription, while the proximal region (5' of -162) can mediate activation by cAMP.


Subject(s)
Cyclic AMP/pharmacology , DNA/genetics , Gene Expression Regulation , Promoter Regions, Genetic , Sertoli Cells/physiology , Stem Cell Factor/genetics , 8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Animals , Base Sequence , Cell Line , Colforsin , Gene Expression , Humans , Male , Mice , Molecular Sequence Data , Promoter Regions, Genetic/drug effects , RNA, Messenger/genetics , Sertoli Cells/drug effects , Testosterone/pharmacology , Tetradecanoylphorbol Acetate/pharmacology
14.
Biochem Biophys Res Commun ; 226(1): 145-51, 1996 Sep 04.
Article in English | MEDLINE | ID: mdl-8806605

ABSTRACT

The neuronal nitric oxide synthase (nNOS) is the main NOS isoform in the urogenital tract catalyzing the synthesis of nitric oxide, the mediator of penile erection and presumably an important factor in the control of urinary voiding. We have cloned from the rat penile corpora cavernosa a cDNA coding for a novel nNOS differing from the cerebellar nNOS by the presence of a 102 nucleotides stretch and other features. This new species is the only nNOS mRNA expressed in the rat penis, urethra, prostate, and skeletal muscle, coexists with the cerebellar nNOS in the pelvic plexus and bladder, and is detectable in the cerebellum. The novel insert is present in human penile RNA and is transcribed from intron 16. The features and distribution of the penile nNOS suggest that it is may be regulated differentially from the cerebellar nNOS.


Subject(s)
Neurons/enzymology , Nitric Oxide Synthase/genetics , Penis/enzymology , Urinary Tract/enzymology , Amino Acid Sequence , Animals , Base Sequence , Cloning, Molecular , DNA, Complementary , Male , Molecular Sequence Data , Polymerase Chain Reaction , RNA, Messenger/genetics , Rats
15.
J Androl ; 16(6): 469-81, 1995.
Article in English | MEDLINE | ID: mdl-14959705

ABSTRACT

Nitric oxide (NO), the main mediator of penile erection, is assumed to be synthesized in the penis by the neuronal constitutive nitric oxide synthase (nNOS). However, nNOS has not been identified in the penile smooth muscle, the target of NO action. The other NOS isozymes, the inducible NOS (iNOS) and the endothelial NOS (eNOS) have not been reported in any penile tissue. The smooth muscle vascular and trabecular tissue from rat corpora cavernosa is represented in vitro by cell cultures designated RPSMC. To determine whether iNOS can be expressed in penile smooth muscle, RPSMC were treated with different lymphokines and/or bacterial lipopolysaccharide (LPS). The selected inducer, LPS/interferon, elicited at 48 hours up to a 50-fold increase in nitrites in the medium; the nitroarginine methyl ester (L-NAME), aminoguanidine, actinomycin D, cycloheximide, transforming growth factor-beta1 (TGF-beta1), and dexamethasone, but was resistant to nifedipine and platelet-derived growth factor AB (PDGF-AB). iNOS induction increased with cell passage. The [3H]L-arginine/citrulline measurement of NO synthesis with intact cells confirmed these results. Incubations of soluble and particulate fractions showed that the cytosol contained most of the activity (Km = 43 microM), which was partially inhibited by ethyleneglycal-bis-tetraacetic acid (EGTA). The 4.4-kb iNOS mRNA peaked at a late period (24-30 hours) and remained high for up to 72 hours. iNOS mRNA induction was strongly inhibited by actinomycin D and dexamethasone, partially inhibited by TGF-beta1, inhibited slightly by PDGF-AB, and unaffected by nifedipine. These results show that iNOS can be expressed in RPSMC in a cell passage-dependent fashion that has so far not been reported for other cell lines, and that the induction reaches much higher levels than in rat or human vascular smooth muscle cells. The expression pattern is also distinctive for the penile cells in time course of induction, Ca2+ dependence, response to certain agents, and mRNA stability.


Subject(s)
Muscle, Smooth/enzymology , Nitric Oxide Synthase/biosynthesis , Penis/enzymology , Animals , Cycloheximide/pharmacology , Dactinomycin/pharmacology , Dexamethasone/pharmacology , Egtazic Acid , Enzyme Induction/drug effects , Guanidines/pharmacology , Male , Muscle, Smooth/drug effects , Muscle, Smooth/physiology , NG-Nitroarginine Methyl Ester/pharmacology , Nifedipine/pharmacology , Nitric Oxide/metabolism , Nitric Oxide/physiology , Nitric Oxide Synthase/drug effects , Nitric Oxide Synthase/metabolism , Penis/drug effects , Penis/physiology , Platelet-Derived Growth Factor/pharmacology , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Transforming Growth Factor beta/pharmacology
16.
Arterioscler Thromb Vasc Biol ; 15(10): 1591-8, 1995 Oct.
Article in English | MEDLINE | ID: mdl-7583532

ABSTRACT

Minimally modified LDL (MM-LDL), obtained by mild iron oxidation or prolonged storage at 4 degrees C, has been shown to induce the expression of macrophage-colony stimulating factor (M-CSF) in cultured aortic endothelial cells. To examine whether other cell types also respond to MM-LDL, we investigated its effect on the expression of M-CSF mRNA in mouse L-cells and human aortic smooth muscle cells. Both L-cells and human aortic smooth muscle cells showed increased levels of M-CSF mRNA in response to 10 to 200 micrograms/mL MM-LDL in a dose-dependent manner. This allowed us to use mouse L-cells as a model to study the mechanism involved in MM-LDL-mediated increase in M-CSF mRNA. Nuclear runon assays showed that M-CSF gene transcription was activated by MM-LDL. In the present study, we identified specific elements that conferred MM-LDL-mediated transcriptional activation of the human M-CSF gene. Chimeric constructs containing sequential deletions in the 5'-promoter region of the M-CSF gene linked to a reporter chloramphenicol acetyltransferase (CAT) gene were transfected into mouse L-cells. The human M-CSF promoter region extending upstream from the transcription start site to nucleotide -406 showed maximum induction of CAT activity by MM-LDL. Induction of CAT activity was drastically reduced, with a deletion plasmid lacking the promoter region -406 to -344. A functional nuclear factor (NF)-kappa B binding site present in this critical region was required for MM-LDL-mediated induction of CAT activity since an internal deletion construct lacking this element showed significant loss of transcriptional activation.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Lipoproteins, LDL/pharmacology , Macrophage Colony-Stimulating Factor/biosynthesis , Muscle, Smooth, Vascular/metabolism , NF-kappa B/metabolism , Animals , Base Sequence , Humans , L Cells , Lipid Peroxidation , Lipoproteins, LDL/metabolism , Macrophage Colony-Stimulating Factor/genetics , Mice , Molecular Sequence Data , Promoter Regions, Genetic/genetics , RNA, Messenger/analysis , Transcriptional Activation
17.
J Clin Invest ; 96(1): 60-8, 1995 Jul.
Article in English | MEDLINE | ID: mdl-7542286

ABSTRACT

To test the hypothesis that nitric oxide (NO) limits endothelial activation, we treated cytokine-stimulated human saphenous vein endothelial cells with several NO donors and assessed their effects on the inducible expression of vascular cell adhesion molecule-1 (VCAM-1). In a concentration-dependent manner, NO inhibited interleukin (IL)-1 alpha-stimulated VCAM-1 expression by 35-55% as determined by cell surface enzyme immunoassays and flow cytometry. This inhibition was paralleled by reduced monocyte adhesion to endothelial monolayers in nonstatic assays, was unaffected by cGMP analogues, and was quantitatively similar after stimulation by either IL-1 alpha, IL-1 beta, IL-4, tumor necrosis factor (TNF alpha), or bacterial lipopolysaccharide. NO also decreased the endothelial expression of other leukocyte adhesion molecules (E-selectin and to a lesser extent, intercellular adhesion molecule-1) and secretable cytokines (IL-6 and IL-8). Inhibition of endogenous NO production by L-N-monomethyl-arginine also induced the expression of VCAM-1, but did not augment cytokine-induced VCAM-1 expression. Nuclear run-on assays, transfection studies using various VCAM-1 promoter reporter gene constructs, and electrophoretic mobility shift assays indicated that NO represses VCAM-1 gene transcription, in part, by inhibiting NF-kappa B. We propose that NO's ability to limit endothelial activation and inhibit monocyte adhesion may contribute to some of its antiatherogenic and antiinflammatory properties within the vessel wall.


Subject(s)
Cell Adhesion Molecules/biosynthesis , Cytokines/pharmacology , Endothelium, Vascular/metabolism , Nitric Oxide/physiology , Base Sequence , Cell Adhesion Molecules/genetics , Cells, Cultured , Cytokines/biosynthesis , Glutathione/analogs & derivatives , Glutathione/pharmacology , Humans , Molecular Sequence Data , Molsidomine/analogs & derivatives , Molsidomine/pharmacology , Monocytes/drug effects , Monocytes/physiology , NF-kappa B/metabolism , Nitroprusside/pharmacology , Nitroso Compounds/pharmacology , RNA, Messenger/analysis , S-Nitrosoglutathione , Vascular Cell Adhesion Molecule-1
18.
J Biol Chem ; 270(28): 17050-5, 1995 Jul 14.
Article in English | MEDLINE | ID: mdl-7622526

ABSTRACT

Macrophage-colony stimulating factor (M-CSF) contributes to atherogenesis by regulating macrophage-derived foam cells in atherosclerotic lesions. Here we report that nitric oxide (NO) inhibits the expression of M-CSF in human vascular endothelial cells independent of guanylyl cyclase activation. The induction of M-CSF mRNA expression by either oxidized low density lipoprotein (ox-LDL) or tumor necrosis factor-alpha (TNF alpha) was attenuated by NO donors, S-nitrosoglutathione (GSNO), sodium nitroprusside (SNP), and 3-morpholinosydnonimine, but not by cGMP analogues, glutathione, or nitrite. Inhibition of endogenous NO production by N-monomethyl-L-arginine (L-NMA) also increased M-CSF expression in control and TNF alpha-stimulated cells. Nuclear run-on assays and transfection studies using M-CSF promoter constructs linked to chloramphenicol acetyltransferase reporter gene indicated that NO repressed M-CSF gene transcription through nuclear factor-kappa B (NF-kappa B). Electrophoretic mobility shift assays demonstrated that activation of NF-kappa B by L-NMA, ox-LDL, and TNF alpha was attenuated by GSNO and SNP, but not by glutathione or cGMP analogues. Since the induction of M-CSF expression depends upon NF-kappa B activation, the ability of NO to inhibit NF-kappa B activation and M-CSF expression may contribute to some of NO's antiatherogenic properties.


Subject(s)
Endothelium, Vascular/metabolism , Gene Expression Regulation , Macrophage Colony-Stimulating Factor/genetics , Nitric Oxide/physiology , Base Sequence , Cells, Cultured , Glutathione/analogs & derivatives , Glutathione/pharmacology , Humans , Lipoproteins, LDL/pharmacology , Molecular Sequence Data , NF-kappa B/metabolism , Nitroso Compounds/pharmacology , S-Nitrosoglutathione , Transcription, Genetic/drug effects , Tumor Necrosis Factor-alpha/pharmacology
19.
Mol Biol Cell ; 3(7): 721-34, 1992 Jul.
Article in English | MEDLINE | ID: mdl-1515675

ABSTRACT

Byr3 was selected as a multicopy suppressor of the sporulation defects of diploid Schizosaccharomyces pombe cells that lack ras1. Like cells mutant at byr1 and byr2, two genes that encode putative protein kinases and that in multiple copies are also suppressors of the sporulation defects of ras1 null diploid cells, cells mutant at byr3 are viable but defective in conjugation. Nucleic acid sequence indicates byr3 has the capacity to encode a protein with seven zinc finger binding domains, similar in structure to the cellular nucleic acid binding protein (CNBP), a human protein that was identified on the basis of its ability to bind DNA. Expression of CNBP in yeast can partially suppress conjugation defects of cells lacking byr3.


Subject(s)
DNA-Binding Proteins/genetics , Fungal Proteins/genetics , Genes, Fungal , Schizosaccharomyces pombe Proteins , Schizosaccharomyces/genetics , Zinc Fingers , Amino Acid Sequence , Base Sequence , Cloning, Molecular , Conjugation, Genetic , DNA, Fungal/genetics , Genes, Suppressor , Genes, ras , Humans , In Vitro Techniques , Molecular Sequence Data , Oligodeoxyribonucleotides/chemistry , Precipitin Tests , Recombinant Fusion Proteins/genetics , Restriction Mapping , Sex Differentiation , Spores, Fungal
20.
J Lipid Res ; 33(6): 831-42, 1992 Jun.
Article in English | MEDLINE | ID: mdl-1512510

ABSTRACT

In mammals, the apolipoprotein (apo) A-I gene is expressed predominantly in liver and intestine, while in avian species it is expressed in all tissues. Although liver and intestine are the major sites of chicken apoA-I mRNA synthesis, there are appreciable amounts of apoA-I mRNA in kidney, ovary/testes, brain, lung, skeletal, and heart muscle. In this study, the nucleotide sequences of the chicken apoA-I gene and its 5' flanking region, as well as the sequences involved in the expression of this gene, have been determined. The gene spans 1.5 kilobases and contains 4 exons and 3 introns, closely resembling the mammalian apoA-I gene. To determine the sequences involved in the expression of the chicken apoA-I gene, plasmid constructs containing serial deletions of the 5' flanking region of the chicken apoA-I gene fused to the bacterial chloramphenicol acetyltransferase (CAT) gene were transfected in human hepatoma (HepG2), colon carcinoma (Caco2), epithelial (Hela), mouse embryonal fibroblast (NIH3T3) cells, and quail myoblasts (QMLA29). The shortest deletion construct, containing 60 bp of the 5' upstream region, was sufficient for maximal transcriptional activity in all cell lines tested. This region contains a short sequence (nucleotides -60 to -54) that is highly conserved in birds and mammals, and an Sp1 binding site. Although the sequence between nucleotides -232 and -101 of the 5' region of the chicken apoA-I gene is partially homologous to the hepatic cell-specific enhancer of the mammalian apoA-I gene (located between nucleotides -222 and -110 upstream of the human apoA-I gene transcription start site), this chicken sequence is transcriptionally inactive in HepG2 cells. These results suggest that differences in the cis-acting regulatory elements of the apoA-I gene play a fundamental role in determining the differences in the tissue-specific expression of this gene in avian and mammalian species.


Subject(s)
Apolipoprotein A-I/genetics , Genes , Regulatory Sequences, Nucleic Acid , Transcription, Genetic , Amino Acid Sequence , Animals , Base Sequence , Biological Evolution , Chickens , Cloning, Molecular , Female , Gene Expression Regulation , Humans , Male , Mice , Molecular Sequence Data , Quail , RNA, Messenger/analysis , Tissue Distribution
SELECTION OF CITATIONS
SEARCH DETAIL
...