Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Eur J Pharmacol ; 600(1-3): 117-22, 2008 Dec 14.
Article in English | MEDLINE | ID: mdl-18926814

ABSTRACT

Ozone is a potent oxidant and causes airway hyperresponsiveness and neutrophilia. To determine the role of p38 mitogen-activated protein kinase (MAPK) activation, we studied the effect of a p38alpha inhibitor SD-282 (Scios Inc, Fremont, CA USA) on ozone-induced airway hyperresponsiveness and neutrophilia. Balb/c mice received SD-282 (30 or 90 mg/kg i.p) or vehicle 1 h before exposure to either ozone (3 ppm, 3 h) or air. Three hours after exposure, lungs were analysed for cytokine levels and bronchoalveolar lavage was performed. Another set of mice were dosed 6 h after exposure and 1 h before assessing airway hyperresponsiveness. SD-282 (90 mg/kg) significantly inhibited ozone-induced airway hyperresponsiveness (-LogPC(150): SD-282: -1.73+/-0.14 vs. vehicle: -0.99+/-0.15, P<0.05). Bronchoalveolar lavage neutrophil numbers were time-dependently increased in vehicle-dosed, ozone-exposed mice, greatest at 20-24 h after exposure. SD-282 (30 and 90 mg/kg) significantly inhibited ozone induced neutrophil numbers at 3 h and 20-24 h after ozone SD-282 significantly inhibited ozone-induced increases in phosphorylated p38 MAPK expression, and in cyclooxygenase-2 (COX-2), interleukin-6 (IL-6) and IL-1beta but not MIP-1alpha gene expression. We conclude that p38 MAPK is involved in ozone-induced airway hyperresponsiveness and lung neutrophilia. Inhibition of p38 MAPK with small molecule kinase inhibitors may be a means of reducing ozone-induced inflammation and airway hyperresponsiveness.


Subject(s)
Bronchial Hyperreactivity/chemically induced , Indoles/pharmacology , Ozone/toxicity , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , Animals , Bronchial Hyperreactivity/physiopathology , Bronchoalveolar Lavage Fluid , Cytokines/drug effects , Cytokines/metabolism , Dose-Response Relationship, Drug , Gene Expression Regulation/drug effects , Indoles/administration & dosage , Inflammation/chemically induced , Inflammation/physiopathology , Lung/pathology , Male , Mice , Mice, Inbred BALB C , Neutrophils/metabolism , Oxidants, Photochemical/toxicity , Time Factors
2.
Respir Res ; 9: 30, 2008 Apr 01.
Article in English | MEDLINE | ID: mdl-18380907

ABSTRACT

BACKGROUND: Bacterial infections are a cause of exacerbation of airway disease. Airway smooth muscle cells (ASMC) are a source of inflammatory cytokines/chemokines that may propagate local airway inflammatory responses. We hypothesize that bacteria and bacterial products could induce cytokine/chemokine release from ASMC. METHODS: Human ASMC were grown in culture and treated with whole bacteria or pathogen associated molecular patterns (PAMPs) for 24 or 48 h. The release of eotaxin-1, CXCL-8 or GMCSF was measured by ELISA. RESULTS: Gram-negative E. coli or gram-positive S. aureus increased the release of CXCL-8, as did IL-1beta, LPS, FSL-1 and Pam3CSK4, whereas FK565, MODLys18 or Poly I:C did not. E. coli inhibited eotaxin-1 release under control conditions and after stimulation with IL-1beta. S. aureus tended to inhibit eotaxin-1 release stimulated with IL-1beta. E. coli or LPS, but not S. aureus, induced the release of GMCSF. CONCLUSION: Gram-positive or gram-negative bacteria activate human ASMC to release CXCL-8. By contrast gram-negative bacteria inhibited the release of eotaxin-1 from human ASMCs. E. coli, but not S. aureus induced GMCSF release from cells. Our findings that ASMC can respond directly to gram-negative and gram-positive bacteria by releasing the neutrophil selective chemokine, CXCL-8, is consistent with what we know about the role of neutrophil recruitment in bacterial infections in the lung. Our findings that bacteria inhibit the release of the eosinophil selective chemokine, eotaxin-1 may help to explain the mechanisms by which bacterial immunotherapy reduces allergic inflammation in the lung.


Subject(s)
Bronchi/microbiology , Chemokine CCL11/metabolism , Escherichia coli/pathogenicity , Interleukin-8/metabolism , Myocytes, Smooth Muscle/microbiology , Staphylococcus aureus/pathogenicity , Acetylmuramyl-Alanyl-Isoglutamine/analogs & derivatives , Acetylmuramyl-Alanyl-Isoglutamine/pharmacology , Bronchi/drug effects , Bronchi/immunology , Bronchi/metabolism , Cells, Cultured , Diglycerides/pharmacology , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , Interleukin-1beta/metabolism , Lipopeptides , Lipopolysaccharides/pharmacology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/immunology , Myocytes, Smooth Muscle/metabolism , Nod1 Signaling Adaptor Protein/metabolism , Nod2 Signaling Adaptor Protein/metabolism , Oligopeptides/pharmacology , Peptides/pharmacology , Poly I-C/pharmacology , Tacrolimus/pharmacology , Time Factors , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 4/metabolism
3.
Pulm Pharmacol Ther ; 21(2): 292-7, 2008.
Article in English | MEDLINE | ID: mdl-17692547

ABSTRACT

Cigarette smoke is the leading risk factor for the development of chronic obstructive pulmonary disease. We have recently shown that cigarette smoke extract synergises with tumour necrosis factor alpha (TNFalpha) in the induction of interleukin-8 (IL-8) from human airway smooth muscle cells. We have investigated the effect of fluticasone propionate, a corticosteroid, and salmeterol, a beta 2-adrenergic receptor agonist, on cigarette smoke extract-induced IL-8 production by human airway smooth muscle cells. Human airway smooth muscle cells in primary culture were exposed to cigarette smoke extract and/or TNFalpha (1 ng ml(-1)) with and without pretreatment with fluticasone (10(-13)-10(-8)M) and/or salmeterol (10(-11)-10(-6)M). IL-8 was analysed by ELISA. Fluticasone dose-dependently inhibited IL-8 release induced by cigarette smoke extract, TNFalpha or combined cigarette smoke extract and TNFalpha. However, while IL-8 release in the presence of cigarette smoke extract alone was completely inhibited by fluticasone, IL-8 production induced by cigarette smoke extract and TNFalpha was only partially reduced. Salmeterol alone had no effect on cigarette smoke extract and/or TNFalpha-induced IL-8 production from human airway smooth muscle cells. Combined fluticasone and salmeterol did not cause further inhibitory effects compared to fluticasone alone. Fluticasone but not salmeterol is effective in reducing cigarette smoke extract-induced IL-8 production in human airway smooth muscle cells. The reduced inhibition of cigarette smoke extract- and TNFalpha-induced IL-8 release by fluticasone may explain why corticosteroids are less effective in chronic obstructive pulmonary disease where increased amounts of TNFalpha are present.


Subject(s)
Adrenergic beta-Agonists/pharmacology , Albuterol/analogs & derivatives , Androstadienes/pharmacology , Bronchi/drug effects , Glucocorticoids/pharmacology , Interleukin-8/biosynthesis , Muscle, Smooth/drug effects , Smoke , Albuterol/pharmacology , Bronchi/cytology , Bronchi/metabolism , Cells, Cultured , Dose-Response Relationship, Drug , Fluticasone , Humans , Muscle, Smooth/cytology , Muscle, Smooth/metabolism , Salmeterol Xinafoate , Nicotiana , Tumor Necrosis Factor-alpha/pharmacology
4.
J Appl Physiol (1985) ; 103(4): 1189-95, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17626835

ABSTRACT

Exposure to air pollutants such as ozone (O(3)) induces airway hyperresponsiveness (AHR) and airway inflammation. Toll-like receptors (TLR) are first-line effector molecules in innate immunity to infections and signal via adapter proteins, including myeloid differentiation factor-88 (MyD88). We investigated the sensing of ozone by TLR2, TLR4, and MyD88. Ozone induced AHR in wild-type (WT) C57BL/6 mice, but AHR was absent in TLR2(-/-), TLR4(-/-), and MyD88(-/-) mice. Bronchoalveolar lavage neutrophilia induced by ozone was inhibited at 3 h but not at 24 h in TLR2(-/-) and TLR4(-/-) mice, while in MyD88(-/-) mice, this was inhibited at 24 h. We investigated the expression of inflammatory cytokines and TLR2, TLR4, and MyD88 in these mice. Ozone induced time-dependent increases in inflammatory gene expression of keratinocyte chemoattractant (KC) and IL-6 and of TLR2, TLR4, and MyD88 in WT mice. IL-6 and KC expression induced by ozone was inhibited in TLR2(-/-), TLR4(-/-), and MyD88(-/-) mice. Expression of MyD88 was increased in TLR2(-/-) and TLR4(-/-) mice, while induction of TLR2 or TLR4 was reduced in TLR2(-/-) and TLR4(-/-) mice, respectively. TLR2 and TLR4 mediate AHR induced by oxidative stress such as ozone, while the adapter protein MyD88, but not TLR2 or TLR4, is important in mediating ozone-induced neutrophilia. TLR2 and TLR4 may also be important in regulating the speed of neutrophilic response. Therefore, ozone may induce murine AHR and neutrophilic inflammation through the activation of the Toll-like receptor pathway that may sense noninfectious stimuli such as oxidative stress.


Subject(s)
Bronchial Hyperreactivity/metabolism , Myeloid Differentiation Factor 88/metabolism , Neutrophils/metabolism , Ozone/pharmacology , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 4/metabolism , Animals , Bronchial Hyperreactivity/chemically induced , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/cytology , Disease Models, Animal , Gene Expression , Gene Expression Regulation , Gene Silencing , Interleukin-6/analysis , Male , Mice , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , Neutrophil Infiltration/drug effects , Neutrophils/cytology , Neutrophils/drug effects , Oxidative Stress/drug effects , Proteins/analysis , RNA, Messenger/metabolism , Toll-Like Receptor 2/genetics , Toll-Like Receptor 4/genetics
5.
J Immunol ; 178(11): 7366-75, 2007 Jun 01.
Article in English | MEDLINE | ID: mdl-17513787

ABSTRACT

Expression of the inflammatory chemokine, growth-related oncogene protein-alpha (GRO-alpha), from airway smooth muscle cells (ASMC) is regulated by pathways involving NF-kappaB and MAPK activation. We determined the effects of dexamethasone on GRO-alpha induced by IL-1beta or TNF-alpha with respect to the role of MAPK pathways and of MAPK phosphatase-1 (MKP-1). Human ASMC were studied in primary culture at confluence. Dexamethasone (10(-8)-10(-5) M) partially inhibited GRO-alpha expression and release induced by IL-1beta and TNF-alpha; this was associated with an inhibition of JNK, but not of p38 or ERK phosphorylation. Together with IL-1beta or TNF-alpha, dexamethasone rapidly induced mRNA and protein expression of MKP-1, which dephosphorylates MAPKs. Using MKP-1 small interfering RNA (siRNA) to block the expression of IL-1beta- and dexamethasone-induced MKP-1 by 50%, JNK phosphorylation was doubled. The inhibitory effect of dexamethasone on GRO-alpha release was partially reversed in ASMC treated with MKP-1 siRNA compared with those treated with scrambled siRNA. In contrast, overexpression of MKP-1 led to a reduction in IL-1beta-induced release of GRO-alpha, but the inhibitory effects of dexamethasone were preserved. Nuclear translocation of the glucocorticoid receptor was increased in ASMC exposed to dexamethasone and IL-1beta. Using chromatin immunoprecipitation assay, glucocorticoid receptor binding to the MKP-1 promoter was increased by IL-1beta and dexamethasone compared with either alone. Glucocorticoids and IL-1beta or TNF-alpha modulate GRO-alpha release partly through the inhibition of JNK pathway, resulting from an up-regulation of MKP-1 expression.


Subject(s)
Bronchi/drug effects , Bronchi/metabolism , Cell Cycle Proteins/physiology , Chemokines, CXC/antagonists & inhibitors , Chemokines, CXC/biosynthesis , Dexamethasone/pharmacology , Immediate-Early Proteins/physiology , Muscle, Smooth/drug effects , Muscle, Smooth/metabolism , Phosphoprotein Phosphatases/physiology , Protein Tyrosine Phosphatases/physiology , Active Transport, Cell Nucleus/drug effects , Bronchi/cytology , Bronchi/enzymology , Cell Cycle Proteins/biosynthesis , Cells, Cultured , Chemokine CXCL1 , Chemokines, CXC/genetics , Chemokines, CXC/metabolism , DNA/metabolism , Dexamethasone/antagonists & inhibitors , Dual Specificity Phosphatase 1 , Humans , Immediate-Early Proteins/biosynthesis , Interleukin-1beta/physiology , Muscle, Smooth/cytology , Muscle, Smooth/enzymology , NF-kappa B/metabolism , Phosphoprotein Phosphatases/biosynthesis , Protein Binding/drug effects , Protein Phosphatase 1 , Protein Tyrosine Phosphatases/biosynthesis , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , Receptors, Glucocorticoid/metabolism , Transfection , Up-Regulation/drug effects
6.
Am J Physiol Lung Cell Mol Physiol ; 293(1): L245-53, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17468136

ABSTRACT

Airway smooth muscle (ASM) hyperplasia is a characteristic feature of the asthmatic airway, but the underlying mechanisms that induce ASM hyperplasia remain unknown. Because transforming growth factor (TGF)-beta is a potent regulator of ASM cell proliferation, we determined its expression and mitogenic signaling pathways in ASM cells. We obtained ASM cells by laser capture microdissection of bronchial biopsies and found that ASM cells from asthmatic patients expressed TGF-beta1 mRNA and protein to a greater extent than nonasthmatic individuals using real-time RT-PCR and immunohistochemistry, respectively. TGF-beta1 stimulated the growth of nonconfluent and confluent ASM cells either in the presence or absence of serum in a time- and concentration-dependent manner. The mitogenic activity of TGF-beta1 on ASM cells was inhibited by selective inhibitors of TGF-beta receptor I kinase (SD-208), phosphatidylinositol 3-kinase (PI3K, LY-294002), ERK (PD-98059), JNK (SP-600125), and NF-kappaB (AS-602868). On the other hand, p38 MAPK inhibitor (SB-203580) augmented TGF-beta1-induced proliferation. To study role of the Smads, we transduced ASM cells with an adenovirus vector-expressing Smad4, Smad7, or dominant-negative Smad3 and found no involvement of these Smads in TGF-beta1-induced proliferation. Dexamethasone caused a dose-dependent inhibition in TGF-beta1-induced proliferation. Our findings suggest that TGF-beta1 may act in an autocrine fashion to induce ASM hyperplasia, mediated by its receptor and several kinases including PI3K, ERK, and JNK, whereas p38 MAPK is a negative regulator. NF-kappaB is also involved in the TGF-beta1 mitogenic signaling, but Smad pathway does not appear important.


Subject(s)
Myocytes, Smooth Muscle/pathology , Respiratory System/pathology , Transforming Growth Factor beta1/metabolism , Adult , Asthma/metabolism , Cell Proliferation/drug effects , Culture Media, Serum-Free , Dexamethasone/pharmacology , Down-Regulation/drug effects , Female , Gene Expression Regulation/drug effects , Humans , Hyperplasia , Male , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Myocytes, Smooth Muscle/metabolism , NF-kappa B/metabolism , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor, Transforming Growth Factor-beta Type I , Receptors, Glucocorticoid/agonists , Receptors, Transforming Growth Factor beta , Respiratory System/drug effects , Smad Proteins/metabolism , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/pharmacology
7.
J Pharmacol Exp Ther ; 322(1): 351-9, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17460151

ABSTRACT

Ozone has potent oxidizing properties, and exposure to ozone causes airway hyper-responsiveness (AHR) and lung inflammation. We determined the importance of c-Jun NH(2) terminal kinase (JNK), a member of the mitogen-activated protein kinase pathway, in ozone-induced AHR and inflammation. SP600125 [anthra[1,9-cd] pyrazol-6 (2H)-one], a specific JNK inhibitor (30 mg/kg) or vehicle, was administered by intraperitoneal injection before and after ozone exposure (3 ppm for 3 h). SP600125 significantly reduced total cells, and neutrophils in bronchoalveolar fluid recovered at 20 to 24 h after exposure and inhibited ozone-induced AHR. Ozone exposure induced activation of JNK in the lung as measured by the expression of phosphorylated-c-Jun, an effect abolished by SP600125. Gene-microarray analysis revealed that ozone increased the expression of over 400 genes by more than 2-fold, including interleukin-6 (IL-6), CXCL1 (keratinocyte cytokine), and CCL2 (monocyte chemoattractant protein-1). SP600125 modulated the expression of a subset of 29 ozone-induced genes; IL-6 and CCL2 expression were further increased, whereas the expression of metallothionein 1, hemopexin, and mitogen-activated 3 kinase 6 was decreased in SP600125-treated ozone-exposed mice. Changes in mRNA for IL-6, CXCL1, and CCL2 were confirmed by real-time polymerase chain reaction. Ozone also decreased the expression of over 500 genes, with the most potent effect on angiopoietin-1. SP600125 modulated the expression of 15 of these genes, and in particular, SP600125 reversed ozone-induced decrease in expression of the redox-sensitive transcription factor, hypoxia-induced factor-1alpha. This study highlights an important role for JNK in response to oxidative stress through modulation of specific inflammatory and redox mediators. Inhibition of JNK with small molecule kinase inhibitors may be a means of reducing ozone-induced inflammation and AHR.


Subject(s)
Anthracenes/pharmacology , Bronchial Hyperreactivity/prevention & control , Inflammation/prevention & control , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Lung/drug effects , Ozone/toxicity , Protein Kinase Inhibitors/pharmacology , Animals , Bronchoalveolar Lavage Fluid/cytology , Gene Expression Regulation/drug effects , Interleukin-6/genetics , Lung/metabolism , Male , Mice , Mice, Inbred BALB C , Oligonucleotide Array Sequence Analysis , Phosphorylation , Polymerase Chain Reaction , Proto-Oncogene Proteins c-jun/metabolism
8.
J Allergy Clin Immunol ; 118(3): 641-8, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16950283

ABSTRACT

BACKGROUND: Host defense against microbial pathogens is elicited through the innate immune system by means of Toll-like receptors (TLRs). Airway smooth muscle cells (ASMCs) display proinflammatory and immunomodulatory functions. ASMCs might participate in airway inflammatory responses associated with innate immune activation. OBJECTIVES: We determined the effects of cytokines, TLR ligands, and corticosteroids on TLR expression and function in human ASMCs. METHODS: Real-time PCR and flow cytometry were used to assess TLR mRNA and protein expression, respectively. ASMCs were stimulated with TLR ligands, and chemokine release was measured by means of ELISA. RESULTS: ASMCs expressed TLR1 to TLR10 mRNA, and TLR2 and TLR3 protein expression was demonstrated. TNF-alpha and double-stranded RNA (dsRNA; TLR3 ligand) were potent inducers of TLR2 and TLR3 mRNA expression, and both stimuli had additive or synergistic effects with IFN-gamma on TLR2 and TLR4, but not TLR3, mRNA expression. Peptidoglycan (TLR2 ligand) and LPS (TLR4 ligand) weakly enhanced TLR2 mRNA expression. Peptidoglycan, dsRNA, and LPS induced IL-8 and eotaxin release, with dsRNA being most potent. dsRNA also modulated cytokine-induced chemokine release in a differential manner. Dexamethasone inhibited cytokine- and ligand-induced TLR2, TLR3, and TLR4 expression and chemokine release. However, dexamethasone potentiated TLR2 expression induced by combined IFN-gamma and TNF-alpha stimulation. CONCLUSION: Expression of TLR2, TLR3, and TLR4 is regulated by cytokines and TLR ligands, and their activation mediates chemokine release in ASMCs. CLINICAL IMPLICATIONS: Proinflammatory responses mediated by activation of pathogen-recognition receptors in ASMCs might contribute to infectious exacerbations of airway inflammatory conditions, such as asthma and chronic obstructive pulmonary disease.


Subject(s)
Lung/immunology , Lung/metabolism , Myocytes, Smooth Muscle/immunology , Myocytes, Smooth Muscle/metabolism , Toll-Like Receptors/physiology , Bronchi/cytology , Bronchi/immunology , Bronchi/metabolism , Cell Separation , Cells, Cultured , Chemokines/metabolism , Humans , Ligands , Lung/cytology , Myocytes, Smooth Muscle/cytology , RNA, Messenger/metabolism , Toll-Like Receptor 2/biosynthesis , Toll-Like Receptor 2/genetics , Toll-Like Receptor 2/physiology , Toll-Like Receptor 3/biosynthesis , Toll-Like Receptor 3/genetics , Toll-Like Receptor 3/physiology , Toll-Like Receptor 4/biosynthesis , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/physiology , Toll-Like Receptors/biosynthesis , Toll-Like Receptors/genetics
9.
Am J Physiol Lung Cell Mol Physiol ; 291(1): L66-74, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16617094

ABSTRACT

Airway smooth muscle cells (ASMC) are a source of inflammatory chemokines that may propagate airway inflammatory responses. We investigated the production of the CXC chemokine growth-related oncogene protein-alpha (GRO-alpha) from ASMC induced by cytokines and the role of MAPK and NF-kappaB pathways. ASMC were cultured from human airways, grown to confluence, and exposed to cytokines IL-1beta and TNF-alpha after growth arrest. GRO-alpha release, measured by ELISA, was increased by >50-fold after IL-1beta (0.1 ng/ml) or 5-fold after TNF-alpha (1 ng/ml) in a dose- and time-dependent manner. GRO-alpha release was not affected by the T helper type 2 cytokines IL-4, IL-10, and IL-13. IL-1beta and TNF-alpha also induced GRO-alpha mRNA expression. Supernatants from IL-1beta-stimulated ASMC were chemotactic for neutrophils; this effect was inhibited by anti-GRO-alpha blocking antibody. AS-602868, an inhibitor of IKK-2, and PD-98059, an inhibitor of ERK, inhibited GRO-alpha release and mRNA expression, whereas SP-600125, an inhibitor of JNK, reduced GRO-alpha release without effect on mRNA expression. SB-203580, an inhibitor of p38 MAPK, had no effect. AS-602868 but not PD-98059 or SP-600125 inhibited p65 DNA-binding induced by IL-1beta and TNF-alpha. By chromatin immunoprecipitation assay, IL-1beta and TNF-alpha enhanced p65 binding to the GRO-alpha promoter, which was inhibited by AS-602868. IL-1beta- and TNF-alpha-stimulated expression of GRO-alpha from ASMC is regulated by independent pathways involving NF-kappaB activation and ERK and JNK pathways. GRO-alpha released from ASMC participates in neutrophil chemotaxis.


Subject(s)
Bronchi/cytology , Chemokines, CXC/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Interleukin-1/metabolism , MAP Kinase Signaling System/immunology , Myocytes, Smooth Muscle/metabolism , Transcription Factor RelA/metabolism , Tumor Necrosis Factor-alpha/metabolism , Bronchi/immunology , Chemokine CXCL1 , Chemokines, CXC/genetics , Chemotaxis, Leukocyte/drug effects , Enzyme Inhibitors/pharmacology , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression/drug effects , Humans , I-kappa B Kinase/antagonists & inhibitors , I-kappa B Kinase/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Interleukin-1/pharmacology , Interleukin-10/pharmacology , Interleukin-13/pharmacology , Interleukin-4/pharmacology , JNK Mitogen-Activated Protein Kinases/metabolism , MAP Kinase Signaling System/drug effects , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/drug effects , Neutrophils/cytology , Neutrophils/drug effects , Promoter Regions, Genetic , Tumor Necrosis Factor-alpha/pharmacology
10.
Respir Res ; 6: 148, 2005 Dec 16.
Article in English | MEDLINE | ID: mdl-16359550

ABSTRACT

BACKGROUND: The elastolytic enzyme matrix metalloproteinase (MMP)-12 has been implicated in the development of airway inflammation and remodeling. We investigated whether human airway smooth muscle cells could express and secrete MMP-12, thereby participating in the pathogenesis of airway inflammatory diseases. METHODS: Laser capture microdissection was used to collect smooth muscle cells from human bronchial biopsy sections. MMP-12 mRNA expression was analysed by quantitative real-time RT-PCR. MMP-12 protein expression and secretion from cultured primary airway smooth muscle cells was further analysed by Western blot. MMP-12 protein localization in bronchial tissue sections was detected by immunohistochemistry. MMP-12 activity was determined by zymography. The TransAM AP-1 family kit was used to measure c-Jun activation and nuclear binding. Analysis of variance was used to determine statistical significance. RESULTS: We provide evidence that MMP-12 mRNA and protein are expressed by in-situ human airway smooth muscle cells obtained from bronchial biopsies of normal volunteers, and of patients with asthma, COPD and chronic cough. The pro-inflammatory cytokine, interleukin (IL)-1beta, induced a >100-fold increase in MMP-12 gene expression and a >10-fold enhancement in MMP-12 activity of primary airway smooth muscle cell cultures. Selective inhibitors of extracellular signal-regulated kinase, c-Jun N-terminal kinase and phosphatidylinositol 3-kinase reduced the activity of IL-1beta on MMP-12, indicating a role for these kinases in IL-1beta-induced induction and release of MMP-12. IL-1beta-induced MMP-12 activity and gene expression was down-regulated by the corticosteroid dexamethasone but up-regulated by the inflammatory cytokine tumour necrosis factor (TNF)-alpha through enhancing activator protein-1 activation by IL-1beta. Transforming growth factor-beta had no significant effect on MMP-12 induction. CONCLUSION: Our findings indicate that human airway smooth muscle cells express and secrete MMP-12 that is up-regulated by IL-1beta and TNF-alpha. Bronchial smooth muscle cells may be an important source of elastolytic activity, thereby participating in remodeling in airway diseases such as COPD and chronic asthma.


Subject(s)
Bronchi/cytology , Bronchi/metabolism , Interleukin-1/administration & dosage , Metalloendopeptidases/metabolism , Muscle, Smooth/metabolism , Myocytes, Smooth Muscle/metabolism , Tumor Necrosis Factor-alpha/administration & dosage , Bronchi/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Enzymologic/physiology , Humans , Matrix Metalloproteinase 12 , Muscle, Smooth/drug effects , Myocytes, Smooth Muscle/drug effects
11.
Angiogenesis ; 8(1): 53-62, 2005.
Article in English | MEDLINE | ID: mdl-16132618

ABSTRACT

Our previous work has demonstrated that angiogenesis occurs in the damaged brain tissue of patients surviving acute ischaemic stroke and increased microvessel density in the penumbra is associated with longer patient survival. The brain is one of the richest sources of FGF-2 and several studies have noted its angiogenic and neuroprotective effects in the nervous system. These findings led us to investigate the expression and localisation of both FGF-2 mRNA and protein in brain tissue collected within 12 h of death from 10 patients who survived for between 24 h and 43 days after acute stroke caused by thrombosis or embolus. Western blot analysis demonstrated increased FGF-2 protein expression in both grey and white matter in the infarcted core and the penumbra region compared to the normal contralateral hemisphere of all 10 patients studied. Using indirect immunoperoxidase staining of paraffin embedded sections, we observed the presence of FGF-2 in neurones, astrocytes, macrophages and endothelial cells. In situ hybridisation was used to localise and quantify mRNA expression in ischaemic brain tissue of the same 10 patients. The expression of FGF-2 in the penumbra of all patients was significantly raised compared with infarcted tissue and normal-looking contralateral hemisphere. In addition, serum FGF-2 was significantly increased between 1 and 14 days (P<0.001) in many patients with both ischaemic stroke (n=28) and intra-cerebral haemorrhage (n=16) compared with age-matched control subjects undergoing routine medical examinations (n=20). We suggest that up-regulation of FGF-2 is one of the mechanisms that leads to angiogenesis and neuro-protection in the penumbra region after acute stroke in man.


Subject(s)
Brain/metabolism , Fibroblast Growth Factor 2/biosynthesis , RNA, Messenger/biosynthesis , Stroke/metabolism , Blotting, Western , Case-Control Studies , Cerebral Hemorrhage/metabolism , Cerebral Infarction/metabolism , Fibroblast Growth Factor 2/genetics , Humans , Immunohistochemistry , In Situ Hybridization , Male , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Neovascularization, Pathologic/metabolism , Up-Regulation
12.
Am J Physiol Lung Cell Mol Physiol ; 288(1): L68-76, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15377500

ABSTRACT

Transforming growth factor (TGF)-beta may play an important role in airway remodeling, and the fibrogenic effect of TGF-beta may be mediated through connective tissue growth factor (CTGF) release. We investigated the role of MAPKs and phosphatidylinositol 3-kinase (PI3K) and the effects of inflammatory cytokines on TGF-beta-induced CTGF expression in human airway smooth muscle cells (ASMC). We examined whether Smad signal was involved in the regulatory mechanisms. TGF-beta 1 induced a time- and concentration-dependent expression of CTGF gene and protein as analyzed by real-time RT-PCR and Western blot. Inhibition of ERK and c-jun NH(2)-terminal kinase (JNK), but not of p38 MAPK and PI3K, blocked the effect of TGF-beta 1 on CTGF mRNA and protein expression and on Smad2/3 phosphorylation. T helper lymphocyte 2-derived cytokines, IL-4 and IL-13, attenuated TGF-beta 1-stimulated mRNA and protein expression of CTGF and inhibited TGF-beta 1-stimulated ERK1/2 and Smad2/3 activation in ASMC. The proinflammatory cytokines tumor necrosis factor-alpha and IL-1 beta reduced TGF-beta 1-stimulated mRNA expression of CTGF but did not inhibit TGF-beta-induced Smad2/3 phosphorylation. TGF-beta 1-stimulated CTGF expression is mediated by mechanisms involving ERK and JNK pathways and is downregulated by IL-4 and IL-13 through modulation of Smad and ERK signals.


Subject(s)
Bronchi/metabolism , Immediate-Early Proteins/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Myocytes, Smooth Muscle/metabolism , Transforming Growth Factor beta/pharmacology , Bronchi/cytology , Cells, Cultured , Connective Tissue Growth Factor , DNA-Binding Proteins/metabolism , Drug Interactions , Enzyme Activation , Enzyme Inhibitors/pharmacology , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Immediate-Early Proteins/genetics , Intercellular Signaling Peptides and Proteins/genetics , Interleukin-1/pharmacology , Interleukin-13/pharmacology , Interleukin-4/pharmacology , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/metabolism , Myocytes, Smooth Muscle/cytology , Phosphatidylinositol 3-Kinases/metabolism , RNA, Messenger/metabolism , Recombinant Proteins/pharmacology , Signal Transduction/drug effects , Smad Proteins , Trans-Activators/metabolism , Transforming Growth Factor beta1 , Tumor Necrosis Factor-alpha/pharmacology
13.
Am J Physiol Lung Cell Mol Physiol ; 287(6): L1230-40, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15321787

ABSTRACT

Chemokine synthesis by airway smooth muscle cells (ASMC) may be an important process underlying inflammatory cell recruitment in airway inflammatory diseases such as asthma and chronic obstructive pulmonary disease (COPD). Fractalkine (FKN) is a recently described CX(3)C chemokine that has dual functions, serving as both a cell adhesion molecule and a chemoattractant for monocytes and T cells, expressing its unique receptor, CX(3)CR1. We investigated FKN expression by human ASMC in response to the proinflammatory cytokines IL-1beta, TNF-alpha, and IFN-gamma, the T helper 2-type cytokines IL-4, IL-10, and IL-13, and the fibrogenic cytokine transforming growth factor (TGF)-beta. Neither of these cytokines alone had any significant effect on ASMC FKN production. Combined stimulation with IFN-gamma and TNF-alpha induced FKN mRNA and protein expression in a time- and concentration-dependent manner. TGF-beta had a significant inhibitory effect on cytokine-induced FKN mRNA and protein expression. Dexamethasone (10(-8)-10(-6) M) significantly upregulated cytokine-induced FKN mRNA and protein expression. Finally, we used selective inhibitors of the mitogen-activated protein kinases c-Jun NH(2)-terminal kinase (JNK) (SP-610025), p38 (SB-203580), and extracellular signal-regulated kinase (PD-98095) to investigate their role in FKN production. SP-610025 (25 microM) and SB-203580 (20 microM), but not PD-98095, significantly attenuated cytokine-induced FKN protein synthesis. IFN-gamma- and TNF-alpha-induced JNK phosphorylation remained unaltered in the presence of TGF-beta but was inhibited by dexamethasone, indicating that JNK is not involved in TGF-beta- or dexamethasone-mediated regulation of FKN production. In summary, FKN production by human ASMC in vitro is regulated by inflammatory and anti-inflammatory factors.


Subject(s)
Bronchi/physiology , Chemokines, CX3C/genetics , Gene Expression Regulation/drug effects , Interferon-gamma/pharmacology , Membrane Proteins/genetics , Muscle, Smooth/physiology , Transforming Growth Factor beta/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , Adrenal Cortex Hormones/pharmacology , Base Sequence , Bronchi/cytology , Bronchi/drug effects , Chemokine CX3CL1 , DNA Primers , Dactinomycin/pharmacology , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Kinetics , MAP Kinase Kinase 4 , Mitogen-Activated Protein Kinase Kinases/metabolism , Muscle, Smooth/cytology , Muscle, Smooth/drug effects , Polymerase Chain Reaction , p38 Mitogen-Activated Protein Kinases/metabolism
14.
Gastroenterology ; 126(7): 1795-808, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15188175

ABSTRACT

BACKGROUND & AIMS: Liver fibrosis and cirrhosis result from the excessive secretion of matrix proteins by hepatic stellate cells (HSCs). Previously considered irreversible, we have studied a model of cirrhosis to determine the mechanisms mediating and limiting spontaneous recovery. METHODS: A micronodular cirrhosis was induced in rats after 12 weeks of CCl(4) intoxication. Livers were analyzed for evidence of matrix degradation, matrix metalloproteinase (MMP) and tissue inhibitor of metalloproteinase (TIMP) expression, stellate cell apoptosis, tissue transglutaminase (tTg) expression, and matrix cross-linking during spontaneous recovery of up to 366 days. RESULTS: Over 366 days of recovery, micronodular cirrhosis underwent significant remodeling to a macronodular cirrhosis. Expression of collagen-1 and TIMP messenger RNA (mRNA) decreased significantly and active MMPs were shown in livers during remodeling of fibrosis. Resolution also was characterized by apoptosis of HSCs, predominantly at the margins of fibrotic septa. Residual septa, not remodeled at 366 days, were characterized by tTg-mediated cross-linking and relative hypocellularity. CONCLUSION: Recovery from comparatively advanced cirrhosis is possible and results in remodeling from a micronodular cirrhosis to a macronodular cirrhosis. We suggest resolution is limited by tTg-mediated matrix cross-linking and a failure of HSC apoptosis.


Subject(s)
Extracellular Matrix/metabolism , Liver Cirrhosis/pathology , Liver Cirrhosis/physiopathology , Matrix Metalloproteinases/genetics , Actins/metabolism , Animals , Apoptosis , Carbon Tetrachloride , Collagen Type I/genetics , Cross-Linking Reagents/metabolism , Dipeptides/metabolism , Disease Models, Animal , Liver/metabolism , Liver/pathology , Liver Cirrhosis/chemically induced , Male , Matrix Metalloproteinases/metabolism , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley , Remission, Spontaneous , Tissue Inhibitor of Metalloproteinases/genetics
15.
Matrix Biol ; 22(3): 229-39, 2003 May.
Article in English | MEDLINE | ID: mdl-12853033

ABSTRACT

During liver fibrosis hepatic stellate cells become activated, transforming into proliferative myofibroblastic cells expressing type I collagen and alpha-smooth muscle actin. They become the major producers of the fibrotic neomatrix in injured liver. This study examines if activated stellate cells are a committed phenotype, or whether they can become deactivated by extracellular matrix. Stellate cells isolated from normal rat liver proliferated and expressed mRNA for activation markers, alpha-smooth muscle actin, type I procollagen and tissue inhibitor of metalloproteinases-1 following 5-7 day culture on plastic, but culture on Matrigel suppressed proliferation and mRNA expression. Activated stellate cells were recovered from plastic by trypsinisation and replated onto plastic, type I collagen films or Matrigel. Cells replated on plastic and type I collagen films proliferated and remained morphologically myofibroblastic, expressing alpha-smooth muscle actin and type I procollagen. However, activated cells replated on Matrigel showed <30% of the proliferative rate of these cells, and this was associated with reduced cellular expression of proliferating cell nuclear antigen and phosphorylation of mitogen-activated protein kinase in response to serum. Activated HSC replated on Matrigel for 3-7 days progressively reduced their expression of mRNA for type I procollagen and alpha-smooth muscle actin and both became undetectable after 7 days. We conclude that basement membrane-like matrix induces deactivation of stellate cells. Deactivation represents an important potential mechanism mediating recovery from liver fibrosis in vivo where type I collagen is removed from the liver and stellate cells might re-acquire contact with their normal basement membrane-like pericellular matrix.


Subject(s)
Collagen/biosynthesis , Extracellular Matrix/metabolism , Liver/cytology , Liver/metabolism , Actins/genetics , Animals , Basement Membrane/metabolism , Cell Adhesion , Cell Division , Cells, Cultured , Collagen Type I/genetics , In Vitro Techniques , Integrins/antagonists & inhibitors , Mitogen-Activated Protein Kinases/metabolism , Phenotype , Phosphorylation , Proliferating Cell Nuclear Antigen/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Tissue Inhibitor of Metalloproteinase-1/genetics , Vitamin A/metabolism
16.
Br J Pharmacol ; 139(6): 1228-34, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12871843

ABSTRACT

1. Human airway smooth muscle cells (HASMC) contribute to airway inflammation in asthma by virtue of their capacity to produce several inflammatory mediators including IL-8, GM-CSF and RANTES. The intracellular signal pathway underlying the production of these cytokines in HASMC is not entirely elucidated. 2. We examined the role of the mitogen-activated protein kinase (MAPK) c-jun N-terminal kinase (JNK) in TNFalpha- and IL-1beta-induced GM-CSF, RANTES and IL-8 production in HASMC by using a novel specific inhibitor for JNK (SP600125). 3. Confluent HASMC were treated with TNFalpha or IL-1beta (10 ng ml(-1)) for 24 h in the presence or absence of SP600125 (1-100 micro M). JNK activity was determined by a kinase assay. Phosphorylation of JNK, p38 MAPK and ERK was examined by Western blotting. Culture supernatants were assayed for GM-CSF, RANTES and IL-8 content by ELISA. 4. Maximum TNFalpha- or IL-1beta-induced phosphorylation of JNK in HASMC occurred after 15 min and returned to baseline levels after 4 h. SP600125 inhibited TNFalpha- and IL-1beta-induced JNK activity in HASMC as shown by the reduced phosphorylation of its substrate c-jun. Furthermore, GM-CSF, RANTES and to a lesser extent IL-8 release from HASMC treated with TNFalpha and IL-1beta was inhibited dosedependently by SP600125. 5. JNK activation is involved in TNFalpha- and IL-1beta-induced GM-CSF, RANTES and IL-8 production from HASMC. JNK may therefore represent a critical pathway for cytokine production in HASMC.


Subject(s)
Chemokine CCL5/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Interleukin-8/metabolism , Lung/cytology , Lung/metabolism , Mitogen-Activated Protein Kinases/physiology , Myocytes, Smooth Muscle/metabolism , Cells, Cultured , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Humans , Interleukin-1/pharmacology , JNK Mitogen-Activated Protein Kinases , Lung/drug effects , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Myocytes, Smooth Muscle/drug effects , Tumor Necrosis Factor-alpha/pharmacology
17.
J Cell Sci ; 116(Pt 13): 2677-85, 2003 Jul 01.
Article in English | MEDLINE | ID: mdl-12746487

ABSTRACT

CD105, a marker of endothelial cells, is abundantly expressed in tissues undergoing angiogenesis and is a receptor for transforming growth factorbeta. The pivotal role of CD105 in the vascular system was demonstrated by the severe vascular defects that occur in CD105-knockout mice, but the exact mechanisms for CD105 regulation of vascular development have not been fully elucidated. In light of the function of CD105 and the importance of hypoxia in neovascularisation, we speculated that CD105 is involved in hypoxia-initiated angiogenesis. Using tissue-cultured human microvascular endothelial cells, we have investigated the effects of hypoxic stress on CD105 gene expression. Hypoxia induced a significant increase in membrane-bound and secreted CD105 protein levels. CD105 mRNA and promoter activity were also markedly elevated, the latter returning to the basal level after 16 hours of hypoxic stress. Hypoxia induced cell cycle arrest at the G0/G1 phases and massive cell apoptosis after 24 hours through a reduction in the Bcl-2 to Bax ratio, downregulation of Bcl-XL and Mcl-1, and upregulation of caspase-3 and caspase-8. The consequence of CD105 upregulation was revealed using an antisense approach and a TUNEL assay. Suppression of CD105 increased cell apoptosis under hypoxic stress in the absence of TGFbeta1. Furthermore, hypoxia and TGFbeta1 synergistically induced apoptosis in the CD105-deficient cells but not in the control cells. We conclude that hypoxia is a potent stimulus for CD105 gene expression in vascular endothelial cells, which in turn attenuates cell apoptosis and thus contributes to angiogenesis.


Subject(s)
Apoptosis/physiology , Endothelial Cells/metabolism , Neovascularization, Pathologic/metabolism , Vascular Cell Adhesion Molecule-1/metabolism , Antigens, CD , Apoptosis/drug effects , Caspases/metabolism , Cell Hypoxia/drug effects , Cell Hypoxia/physiology , Cells, Cultured , Cyclin D1/metabolism , Endoglin , Endothelial Cells/cytology , Endothelial Cells/drug effects , G1 Phase/physiology , Genes, cdc/physiology , Humans , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/physiopathology , Oligonucleotides, Antisense/pharmacology , Promoter Regions, Genetic/genetics , RNA, Messenger/metabolism , Receptors, Cell Surface , Stress, Physiological/genetics , Stress, Physiological/metabolism , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/pharmacology , Up-Regulation/drug effects , Up-Regulation/genetics , Vascular Cell Adhesion Molecule-1/genetics
18.
FASEB J ; 17(1): 47-9, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12475903

ABSTRACT

Collagen-I, which predominates in the neomatrix of fibrotic liver, regulates hepatocyte and hepatic stellate cell (HSC) phenotypes. Recovery from liver fibrosis is accompanied by hepatocyte regeneration, matrix degradation, and HSC apoptosis. Using mice bearing a mutated collagen-I gene (r/r mice), which confers resistance to collagenase degradation, we have investigated the hypothesis that collagen-I degradation is critical to HSC apoptosis and hepatocyte regeneration during recovery from liver fibrosis. During a 28-day recovery period after 8 wk of CCl4 treatment, wild-type (WT) livers had significantly (43%) decreased hydroxyproline (OHP) content. In r/r livers, however, OHP content remained elevated at peak fibrosis levels. Expressed markers of activated HSC (alpha-smooth muscle actin, collagen-I), elevated at peak fibrosis, dropped to control levels in WT livers after 28 days but remained raised in the r/r livers. Moreover, relative to WT livers, r/r livers had significantly reduced stellate cell apoptosis and hepatocyte regeneration during the recovery period. Using extracted collagen-I from each genotype as culture substrata, relative to r/r, we show that WT collagen-I promotes hepatocyte proliferation via stimulation of integrin alpha(v)beta3. Failure to degrade collagen-I critically impairs HSC apoptosis and may prevent the effective restoration of hepatocyte mass in liver fibrosis.


Subject(s)
Collagen Type I/metabolism , Liver Cirrhosis, Experimental/metabolism , Liver Regeneration , Liver/cytology , Animals , Apoptosis , Carbon Tetrachloride , Collagen Type I/genetics , Collagen Type I/physiology , Collagenases/metabolism , Hepatocytes/physiology , Kinetics , Liver/metabolism , Liver Cirrhosis, Experimental/chemically induced , Liver Cirrhosis, Experimental/pathology , Mice , Models, Biological , Mutation , RNA, Messenger/biosynthesis
19.
J Biol Chem ; 277(13): 11069-76, 2002 Mar 29.
Article in English | MEDLINE | ID: mdl-11796725

ABSTRACT

The activated hepatic stellate cell (HSC) is central to liver fibrosis as the major source of collagens I and III and the tissue inhibitors of metalloproteinase-1 (TIMP-1). During spontaneous recovery from liver fibrosis, there is a decrease of TIMP expression, an increase in collagenase activity, and increased apoptosis of HSC, highlighting a potential role for TIMP-1 in HSC survival. In this report, we use tissue culture and in vivo models to demonstrate that TIMP-1 directly inhibits HSC apoptosis. TIMP-1 demonstrated a consistent, significant, and dose-dependent antiapoptotic effect for HSC activated in tissue culture and stimulated to undergo apoptosis by serum deprivation, cycloheximide exposure, and nerve growth factor stimulation. A nonfunctional mutated TIMP-1 (T2G mutant) in which all other domains are conserved did not inhibit apoptosis, indicating that inhibition of apoptosis was mediated through MMP inhibition. Synthetic MMP inhibitors also inhibited HSC apoptosis. Studies of experimental liver cirrhosis demonstrated that persistent expression of TIMP-1 mRNA determined by PCR correlated with persistence of activated HSC quantified by alpha smooth muscle actin staining, while in fibrosis, loss of activated HSC correlated with a reduction in TIMP-1 mRNA. We conclude that TIMP-1 inhibits apoptosis of activated HSC via MMP inhibition.


Subject(s)
Apoptosis/physiology , Liver Cirrhosis/therapy , Liver/cytology , Matrix Metalloproteinase Inhibitors , Tissue Inhibitor of Metalloproteinase-1/metabolism , Animals , Apoptosis/drug effects , Caspase 3 , Caspases/metabolism , Cell Division , Cells, Cultured , Cycloheximide/pharmacology , Humans , In Situ Nick-End Labeling , Liver/metabolism , Liver Cirrhosis/pathology , Rats , Tissue Inhibitor of Metalloproteinase-1/genetics
20.
Neurobiol Aging ; 23(2): 237-43, 2002.
Article in English | MEDLINE | ID: mdl-11804709

ABSTRACT

The aim of the present study was to investigate, in patients with Alzheimer's disease (AD), and vascular dementia (VAD), patterns of local release of vascular endothelial growth factor (VEGF) and transforming growth factor-beta (TGF-beta), two cytokines having a pivotal role in hypoxia-induced angiogenesis. The intrathecal levels of these molecules were related to the clinical severity of these diseases and to the intrathecal levels of beta-amyloid protein. Significantly increased cerebrospinal fluid (CSF) levels of both VEGF and TGF-beta were observed in 20 patients with AD and in 26 patients with VAD compared to healthy controls. Interestingly, there was significant correlation between the CSF levels of TGF-beta and VEGF in all the individuals studied. Our study demonstrates, both in patients with AD and in patients with VAD, an intrathecal production of VEGF, a cytokine which plays a pivotal role in angiogenesis. These results suggest that vascular factors might not only play a role in the pathogenesis of VAD but also in the pathogenesis of AD. In addition, we show in AD and VAD an intrathecal production of TGF-beta, a cytokine exerting on one hand anti-inflammatory and angiogenic properties, but on the other promoting amyloidogenesis.


Subject(s)
Alzheimer Disease/cerebrospinal fluid , Dementia, Vascular/cerebrospinal fluid , Endothelial Growth Factors/cerebrospinal fluid , Lymphokines/cerebrospinal fluid , Transforming Growth Factor beta/cerebrospinal fluid , Aged , Albumins/cerebrospinal fluid , Alzheimer Disease/psychology , Amyloid beta-Peptides/cerebrospinal fluid , Dementia, Vascular/psychology , Enzyme-Linked Immunosorbent Assay , Female , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Tomography, X-Ray Computed , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...