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










Publication year range
1.
Am J Transplant ; 18(4): 916-926, 2018 04.
Article in English | MEDLINE | ID: mdl-28980446

ABSTRACT

The classical pathway (CP) of complement may contribute to the pathogenesis of antibody-mediated rejection (ABMR). Selective CP blockade may be a promising strategy to counteract rejection. The objective of this first-in-patient phase 1b trial was to evaluate the safety/tolerability and CP-blocking potential of 4 weekly doses (60 mg/kg) of the anti-C1s antibody BIVV009 in complement-mediated disorders. Here we describe the results in a cohort of 10 stable kidney transplant recipients (median of 4.3 years posttransplantation) with late active ABMR and features of CP activation, such as capillary C4d or complement-fixing donor-specific antibodies (DSA). During 7 weeks follow-up, no severe adverse events were reported, and BIVV009 profoundly inhibited overall and DSA-triggered CP activation in serum. Five of 8 C4d-positive recipients turned C4d-negative in 5-week follow-up biopsies, while another 2 recipients showed a substantial decrease in C4d scores. There was, however, no change in microcirculation inflammation, gene expression patterns, DSA levels, or kidney function. In conclusion, we demonstrate that BIVV009 effectively blocks alloantibody-triggered CP activation, even though short-course treatment had no effect on indices of activity in late ABMR. This initial trial provides a valuable basis for future studies designed to clarify the therapeutic value of CP blockade in transplantation. ClinicalTrials.gov NCT#02502903.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Complement C1s/immunology , Graft Rejection/drug therapy , Graft Survival/drug effects , Isoantibodies/adverse effects , Kidney Failure, Chronic/surgery , Kidney Transplantation/adverse effects , Adult , Aged , Allografts , Complement Activation/immunology , Female , Follow-Up Studies , Glomerular Filtration Rate , Graft Rejection/etiology , Graft Rejection/pathology , Graft Survival/immunology , HLA Antigens/immunology , Humans , Kidney Function Tests , Male , Middle Aged , Postoperative Complications , Prognosis , Prospective Studies , Risk Factors , Tissue Donors
2.
Am J Transplant ; 17(9): 2300-2311, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28251805

ABSTRACT

The classic pathway (CP) of complement is believed to significantly contribute to alloantibody-mediated transplant injury, and targeted complement inhibition is currently considered to be a promising approach for preventing rejection. Here, we investigated the mode of action and efficacy of the humanized anti-C1s monoclonal antibody TNT009 and its parental mouse variant, TNT003, in preclinical in vitro models of HLA antibody-triggered CP activation. In flow cytometric assays, we measured the attachment of C1 subcomponents and C4/C3 split products (C4b/d, C3b/d) to HLA antigen-coated flow beads or HLA-mismatched aortic endothelial cells and splenic lymphocytes. Anti-C1s antibodies profoundly inhibited C3 activation at concentrations >20 µg/mL, in both solid phase and cellular assays. While C4 activation was also prevented, this was not the case for C1 subcomponent attachment. Analysis of serum samples obtained from 68 sensitized transplant candidates revealed that the potency of inhibition was related to the extent of baseline CP activation. This study demonstrates that anti-C1s antibodies TNT009 and TNT003 are highly effective in blocking HLA antibody-triggered complement activation downstream of C1. Our results provide the foundation for clinical studies designed to investigate the potential of TNT009 in the treatment or prevention of complement-mediated tissue injury in sensitized transplant recipients.


Subject(s)
Antibodies, Monoclonal/pharmacology , Complement Activation/immunology , Complement C1s/immunology , Graft Rejection/drug therapy , HLA Antigens/immunology , Isoantibodies/adverse effects , Kidney Transplantation/adverse effects , Animals , Follow-Up Studies , Glomerular Filtration Rate , Graft Rejection/etiology , Graft Survival , Humans , Kidney Failure, Chronic/immunology , Kidney Failure, Chronic/surgery , Kidney Function Tests , Mice , Prognosis
3.
Am J Transplant ; 15(8): 2037-49, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25904443

ABSTRACT

Antibody-mediated rejection (AMR) of solid organ transplants (SOT) is characterized by damage triggered by donor-specific antibodies (DSA) binding donor Class I and II HLA (HLA-I and HLA-II) expressed on endothelial cells. While F(ab')2 portions of DSA cause cellular activation and proliferation, Fc regions activate the classical complement cascade, resulting in complement deposition and leukocyte recruitment, both hallmark features of AMR. We characterized the ability of an anti-C1s monoclonal antibody, TNT003, to inhibit HLA antibody (HLA-Ab)-induced complement activation. Complement deposition induced by HLA-Ab was evaluated using novel cell- and bead-based assays. Human aortic endothelial cells (HAEC) were cultured with HLA-Ab and human complement; production of activated complement proteins was measured by flow cytometry. Additionally, C3d deposition was measured on single antigen beads (SAB) mixed with HLA-Ab and human complement. TNT003 inhibited HLA-Ab mediated complement deposition on HAEC in a concentration-dependent manner; C3a, C4a and C5a anaphylatoxin production was also diminished by TNT003. Finally, TNT003 blocked C3d deposition induced by Class I (HLAI-Ab)- and Class II (HLAII-Ab)-specific antibodies on SAB. These data suggest TNT003 may be useful for modulating the effects of DSA, as TNT003 inhibits complement deposition and split product formation generated by HLA-I/II-Ab in vitro.


Subject(s)
Antibodies, Monoclonal/immunology , Complement Activation/immunology , Complement C1s/immunology , HLA Antigens/immunology , Cells, Cultured , Endothelium, Vascular/cytology , Endothelium, Vascular/immunology , Heart Transplantation , Humans
4.
Arterioscler Thromb Vasc Biol ; 20(6): 1668-74, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10845887

ABSTRACT

In previous studies of the role of tissue plasminogen activator (tPA) in the lung inflammatory response, we observed that tPA expression was present exclusively in the small arteries and arterioles within the lung and absent from the capillaries, veins, and large pulmonary arteries. To define more completely the expression pattern of tPA, we evaluated the distribution of this protein during prenatal and postnatal development. tPA was first observed in the rat fetus at day 13 in the large arteries of both the thoracic and cranial cavities, including the dorsal aortas and pulmonary arteries in the former and the internal carotid and middle cerebral arteries in the latter. By day 15, tPA was no longer detectable in the aortas but appeared throughout the pulmonary, subclavian, vertebral, and basilar arteries. At day 17, tPA had disappeared from the subclavian artery and the proximal portion of the vertebral artery but was found in the smaller arterial branches of these 2 large vessels. By the end of gestation, tPA had also disappeared from the main pulmonary arteries but remained in the branches at the hilus of the lung. At birth, tPA was concentrated in the endothelia of arteries within the pia mater, the basilar and superficial cerebral arteries, and the lung arterial system. As the animals reached maturity, tPA disappeared from the larger cerebral arteries and their cortical branches but continued to be expressed in the vessels of the pia mater and lung. This study indicates that tPA expression is a dynamic process that responds to a changing arterial environment during vascular development.


Subject(s)
Arteries/embryology , Embryonic and Fetal Development , Tissue Plasminogen Activator/analysis , Animals , Aorta/chemistry , Aorta/embryology , Arteries/chemistry , Endothelium, Vascular/chemistry , Endothelium, Vascular/embryology , Female , Gestational Age , Immunohistochemistry , Lung/blood supply , Lung/embryology , Pia Mater/blood supply , Pia Mater/embryology , Pregnancy , Pulmonary Artery/chemistry , Pulmonary Artery/embryology , Rats , Rats, Wistar , Tissue Plasminogen Activator/metabolism
5.
J Clin Invest ; 105(11): 1547-54, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10841513

ABSTRACT

Recent studies indicate that tissue factor (TF) acts in embryogenesis, metastasis, and angiogenesis. Three independent groups showed that targeted disruption of the murine TF (mTF) gene results in 90% lethality of mTF null embryos at embryonic days 9. 5-10.5. We have demonstrated that expression of wild-type human TF (hTF) from a minigene rescues the embryonic lethality of mTF null embryos. To investigate the role of TF in embryogenesis, we made mutant hTF minigenes whose products either bound FVII/VIIa at a reduced level or lacked the cytoplasmic domain. Two independent transgenic lines expressing the hTF extracellular domain mutant failed to rescue the embryonic lethality of mTF null embryos, suggesting that FVII/VIIa binding by TF, proteolytic activity by the TF/FVIIa complex, or both were required for embryogenesis. In contrast, two transgenic lines expressing the hTF cytoplasmic domain mutant rescued the embryonic lethality of mTF null embryos, indicating that the cytoplasmic domain of TF was not required for embryogenesis. We propose that TF/FVIIa-dependent extracellular protease activity is required for embryogenesis.


Subject(s)
Embryonic and Fetal Development , Thromboplastin/physiology , Animals , CHO Cells , Cricetinae , Cytoplasm/chemistry , Female , Humans , Mice , Mice, Transgenic , Pregnancy , RNA, Messenger/analysis , Thromboplastin/genetics , Transgenes
6.
Proc Natl Acad Sci U S A ; 96(14): 8138-43, 1999 Jul 06.
Article in English | MEDLINE | ID: mdl-10393961

ABSTRACT

We employed a novel mouse line that expresses low levels of human tissue factor (TF) in the absence of murine TF to analyze the role of TF in gestation. Low-TF female mice had a 14-18% incidence of fatal postpartum uterine hemorrhage, suggesting that TF plays an important role in uterine hemostasis. Low-TF female mice mated with low-TF male mice had a 42% incidence of fatal midgestational hemorrhage (n = 41), whereas no fatal midgestational hemorrhages were observed in low-TF female mice mated with wild-type male mice (n = 43). Placentas of low-TF embryos from both low-TF and normal (+/-) TF females were abnormal and contained numerous maternal blood pools in the labyrinth. Placentas of TF null embryos surviving beyond embryonic day 10.5 exhibited similar defects. The mouse maternal-embryonic placental barrier consists of four cellular layers (layers I, II, and III and endothelial cells), where layer I lines the maternal lacunae. Comparison of TF-deficient placentas with control placentas by immunohistochemical and ultrastructural analyses revealed thinning of layer I and a reduction in the number of cellular contacts of layer I trophoblasts spanning the maternal blood space between adjacent trabeculae. These structural changes in low-TF and TF null placentas result in enlarged maternal lacunae, as determined by morphometric analysis, and placental hemorrhage, which leads to midgestational death of low-TF female mice. This study demonstrated that TF is required for uterine hemostasis and revealed an unexpected role of TF in the maintenance of the placental labyrinth.


Subject(s)
Placenta/physiology , Thromboplastin/physiology , Uterine Hemorrhage/physiopathology , Uterus/physiology , Animals , Crosses, Genetic , Female , Hemostasis , Humans , Male , Mice , Mice, Knockout , Placenta/abnormalities , Placenta/blood supply , Pregnancy , Puerperal Disorders/genetics , Puerperal Disorders/pathology , Puerperal Disorders/physiopathology , Thromboplastin/deficiency , Thromboplastin/genetics , Uterine Hemorrhage/genetics , Uterine Hemorrhage/pathology , Uterus/abnormalities , Uterus/blood supply
7.
J Clin Invest ; 102(9): 1645-52, 1998 Nov 01.
Article in English | MEDLINE | ID: mdl-9802878

ABSTRACT

Recognition of bacterial endotoxin (LPS) elicits multiple host responses, including activation of cells of the innate immune system. LPS exposure occurs repeatedly during septicemia, making strict regulation of gene expression necessary. Such regulation might prevent, for example, the continuous production of proinflammatory cytokines such as tumor necrosis factor (TNF), which could lead to severe vascular collapse. Tolerance to LPS is characterized by a diminished production of TNF during prolonged exposure to LPS, and is therefore likely to represent an essential control mechanism during sepsis. In the present study, which uses mice with genetic deletions of the proteins of NF-kappaB complex, we provide data demonstrating that increased expression of the p50 subunit of NF-kappaB directly results in the downregulation of LPS-induced TNF production. This contention is supported by the following observations: (1) tolerance to LPS is not induced in macrophages from p50-/- mice; (2) long-term pretreatment with LPS does not block synthesis of TNF mRNA in p50-/- macrophages (in contrast to wild-type macrophages); (3) ectopic overexpression of p50 reduces transcriptional activation of the murine TNF promoter; and (4) analysis of the four kappaB sites from the murine TNF promoter demonstrates that binding of p50 homodimers to the positively acting kappaB3 element is associated with development of the LPS-tolerant phenotype. Thus, p50 expression plays a key role in the development of LPS tolerance.


Subject(s)
Immunity, Innate/physiology , Macrophages, Peritoneal/immunology , NF-kappa B/immunology , Animals , Cells, Cultured , Down-Regulation , Immune Tolerance , Lipopolysaccharides/immunology , Lipopolysaccharides/pharmacology , Macrophages, Peritoneal/drug effects , Mice , Mice, Knockout , NF-kappa B p50 Subunit , Promoter Regions, Genetic , RNA, Messenger , Tumor Necrosis Factor-alpha/genetics
8.
Arterioscler Thromb Vasc Biol ; 18(6): 934-40, 1998 Jun.
Article in English | MEDLINE | ID: mdl-9633934

ABSTRACT

Human monocyte chemoattractant protein-1 (MCP-1) is expressed by a variety of cell types in response to various stimuli. MCP-1 expressed by the endothelium plays an important role in cell migration and activation. MCP-1 is a major chemoattractant for monocytes, T lymphocytes, and basophils. In the present study, we present evidence that the proteasome complex is involved in mediating the interleukin (IL)-1beta induction of MCP-1 in endothelial cells. We present evidence that a proteasome inhibitor, N-acetyl-leucinyl-leucinyl-norleucinal (norLeu), and the protease inhibitor tosyl-Phe-chloromethylketone (TPCK) block IL-1beta induction of MCP-1 protein expression. norLeu and TPCK also blocked IL-1beta-induced MCP-1 promoter-driven reporter gene expression as well as nuclear factor (NF)-kappaB-mediated reporter gene expression. The effects of norLeu were due to its inhibition of the proteasome rather than calpain, because other calpain inhibitors had no effect on MCP-1 expression. In contrast to TPCK, which blocked NF-kappaB translocation to the nucleus, norLeu had no effect on NF-kappaB nuclear translocation or IL-1beta-induced phosphorylation of p65. This study demonstrates that the proteasome pathway is involved in IL-1beta-induced MCP-1 gene expression in human endothelial cells.


Subject(s)
Chemokine CCL2/genetics , Cysteine Endopeptidases/metabolism , Endothelium, Vascular/metabolism , Gene Expression Regulation , Interleukin-1/physiology , Multienzyme Complexes/metabolism , Serine Proteinase Inhibitors/pharmacology , Cells, Cultured , Cysteine Proteinase Inhibitors/pharmacology , Humans , Leupeptins/pharmacology , NF-kappa B/metabolism , Proteasome Endopeptidase Complex , Tosylphenylalanyl Chloromethyl Ketone/pharmacology
9.
EMBO J ; 17(12): 3372-84, 1998 Jun 15.
Article in English | MEDLINE | ID: mdl-9628874

ABSTRACT

We have identified and cloned a novel serine/ threonine kinase, p38-regulated/activated protein kinase (PRAK). PRAK is a 471 amino acid protein with 20-30% sequence identity to the known MAP kinase-regulated protein kinases RSK1/2/3, MNK1/2 and MAPKAP-K2/3. PRAK was found to be expressed in all human tissues and cell lines examined. In HeLa cells, PRAK was activated in response to cellular stress and proinflammatory cytokines. PRAK activity was regulated by p38alpha and p38beta both in vitro and in vivo and Thr182 was shown to be the regulatory phosphorylation site. Activated PRAK in turn phosphorylated small heat shock protein 27 (HSP27) at the physiologically relevant sites. An in-gel kinase assay demonstrated that PRAK is a major stress-activated kinase that can phosphorylate small heat shock protein, suggesting a potential role for PRAK in mediating stress-induced HSP27 phosphorylation in vivo.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Mitogen-Activated Protein Kinases , Protein Serine-Threonine Kinases/genetics , Amino Acid Sequence , Base Sequence , Cytokines/metabolism , Heat-Shock Proteins/metabolism , Humans , Intracellular Signaling Peptides and Proteins , Molecular Sequence Data , Phosphorylation , Phylogeny , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/physiology , Sequence Alignment , Sequence Homology, Amino Acid , Stress, Physiological , Substrate Specificity , p38 Mitogen-Activated Protein Kinases
10.
J Clin Invest ; 101(3): 560-9, 1998 Feb 01.
Article in English | MEDLINE | ID: mdl-9449688

ABSTRACT

Tissue factor (TF) expression is associated with life-threatening thrombosis in a variety of human diseases, including sepsis, cancer, and atherosclerosis. Recently, it was shown that inactivation of the murine TF (mTF) gene results in embryonic lethality. To date, despite extensive studies on the regulation of the TF promoter in vitro, no studies have examined the cis-acting regulatory elements that control TF gene expression in vivo. Here we report that a human TF (hTF) minigene containing the human TF promoter and human TF cDNA directed a low level (approximately 1% relative to mouse TF) of both constitutive and LPS-inducible human TF expression in transgenic mice. Importantly, the human TF minigene rescued the embryonic lethality of murine TF null embryos, suggesting that human TF substituted for murine TF during embryogenesis. Rescued mice (mTF-/-, hTF+), which expressed low levels (approximately 1%) of TF activity, developed normally with no signs of a bleeding diathesis, suggesting that low TF expression can maintain hemostasis compatible with normal survival. These studies establish a novel mouse model system that can be used to examine the regulation of the human TF gene in vivo and the impact of low TF levels on the hemostatic balance in various thrombotic diseases.


Subject(s)
Hemostatics/metabolism , Thromboplastin/metabolism , Animals , Female , Gene Expression , Hemostasis , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Thromboplastin/biosynthesis , Thromboplastin/genetics , Tissue Distribution
11.
Trends Cardiovasc Med ; 8(3): 138-42, 1998 Apr.
Article in English | MEDLINE | ID: mdl-21235924

ABSTRACT

Monocytes and endothelial cells become activated at sites of inflammation and contribute to the pathology of many diseases, including septic shock and atherosclerosis. In these cells, induction of genes expressing various inflammatory mediators, such as adhesion molecules, cytokines, and growth factors, is regulated by NF-κB/Rel transcription factors. Recent studies have identified components of the signal transduction pathways leading to the activation of NF-κB/Rel proteins. Inhibition of these signaling pathways provides a novel therapeutic approach to prevent inducible gene expression in both monocytes and endothelial cells. (Trends Cardiovasc Med 1998;8:138-142). © 1998, Elsevier Science Inc.

12.
Eur J Immunol ; 27(5): 1091-7, 1997 May.
Article in English | MEDLINE | ID: mdl-9174597

ABSTRACT

Chemokines are potent mediators of cell migration and activation and therefore play an essential role in early events of inflammation. In conjunction with cell adhesion molecules, chemokines help to localize cells to a specific site and enhance the inflammatory reaction at the site. Clinically, elevated levels of chemokines have been found in a variety of inflammatory diseases. The prototype C-C chemokine is monocyte chemoattractant protein-1 (MCP-1) which is synthesized by variety of cell types including endothelial cells in response to a variety of stimuli. MCP-1 is a major chemoattractant for monocytes, T lymphocytes, and basophils. In the present study, we investigated the factors involved in cytokine-induced MCP-1 gene expression in human endothelial cells. We present evidence that the nuclear factor (NF)-kappa B-like binding site and the AP-1 binding site located 90 and 68 base pairs upstream of the transcriptional start site, respectively, are required for maximal induction of the human MCP-1 promoter by interleukin-(IL)-1 beta. Site-directed mutagenesis or deletion of the NF-kappa B-like site decreased the cytokine-induced activity of the promoter. Site-directed mutagenesis of the AP-1 binding site also decreased the cytokine-induced activity of the promoter. We show that the NF-kappa B-like site located at-90 in the MCP-1 promoter binds to the p50/p65 heterodimer of the NF-kappa B/Rel family in IL-1 beta-stimulated human endothelial cells. Overexpression of p65 results in the transactivation of the MCP-1 promoter as well. The data presented in this study suggest that cytokine-induced MCP-1 gene expression in human endothelial cells depends on the cooperative action of NF-kappa B and AP-1.


Subject(s)
Chemokine CCL2/biosynthesis , Chemokine CCL2/genetics , Cytokines/physiology , Endothelium, Vascular/metabolism , Gene Expression Regulation/immunology , NF-kappa B/physiology , Transcription Factor AP-1/physiology , Base Sequence , Cells, Cultured , Dimerization , Humans , Interleukin-1/physiology , Molecular Sequence Data , NF-kappa B/biosynthesis , NF-kappa B/metabolism , NF-kappa B p50 Subunit , Promoter Regions, Genetic/physiology , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins c-rel , Transcription Factor RelA , Transcription Factors/biosynthesis , Tumor Necrosis Factor-alpha/physiology , Umbilical Veins/cytology
13.
J Immunol ; 158(7): 3401-7, 1997 Apr 01.
Article in English | MEDLINE | ID: mdl-9120300

ABSTRACT

Inflammation is characterized by the recruitment of leukocytes and their subsequent migration from the vasculature into the tissue, where they often cause severe damage. Endothelial cells play a major role in this cascade by expressing cell surface adhesion molecules, such as VCAM-1 and ICAM-1, and chemokines, in response to cytokines. Many of these genes are under the control of inflammatory response transcription factors such as nuclear factor (NF)-kappa B. In this study, we examined the effects of 5-lipoxygenase inhibitors (nordihydroguaiaretic acid and AA861) on IL-1 beta-induced VCAM-1 gene expression in HUVECs. We demonstrated that 5-lipoxygenase inhibitors, but not cyclooxygenase inhibitors, block IL-1 beta-induced VCAM-1 cell surface expression and promoter activity. In transiently transfected HUVECs, NF-kappa B-dependent gene expression was inhibited by 5-lipoxygenase inhibitors. These inhibitors did not block IL-1 beta-induced nuclear translocation of NF-kappa B, inhibitor of kappa B-alpha proteolytic degradation, or significantly reduce phosphorylation of p65. These studies indicate that inhibition of 5-lipoxygenase blocks cytokine-induced VCAM-1 gene expression by reducing the functional activity of NF-kappa B/Rel proteins in HUVECs.


Subject(s)
Endothelium, Vascular/immunology , Gene Expression Regulation/drug effects , Interleukin-1/pharmacology , Lipoxygenase Inhibitors , Vascular Cell Adhesion Molecule-1/biosynthesis , Vascular Cell Adhesion Molecule-1/genetics , Benzoquinones/pharmacology , Biological Transport/drug effects , Cell Nucleus/drug effects , Cell Nucleus/genetics , Cells, Cultured , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Humans , Lipoxygenase Inhibitors/pharmacology , Masoprocol/pharmacology , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Phosphorylation/drug effects , Promoter Regions, Genetic/drug effects , Transcription, Genetic/drug effects , Vascular Cell Adhesion Molecule-1/drug effects
14.
J Clin Invest ; 99(4): 737-44, 1997 Feb 15.
Article in English | MEDLINE | ID: mdl-9045878

ABSTRACT

Using flow channel, we report that the application of a laminar shear stress induced a transient increase of tissue factor (TF) procoagulant activity in human umbilical vein endothelial cells (HUVEC), which was accompanied by a rapid and transient induction of the TF mRNA in the HUVEC. Functional analysis of the 2.2 kb TF 5' promoter indicated that a GC-rich region containing three copies each of the EGR-1 and Sp1 sites was required for induction. Mutation of the Sp1 sites, but not the EGR-1 sites, attenuated the response of TF promoter to shear stress. Thus, Sp1 is a newly defined shear stress responsive element. Electrophoretic mobility shift assays showed there was no increase in binding of nuclear extracts from sheared cells to an Sp1 consensus site. In contrast, immunoblotting of these nuclear extracts with antibody against transcription factor Sp1 demonstrated that shear stress increased the phosphorylation of Sp1. We also showed that shear stress, like the phosphatase inhibitor okadaic acid, increased the transcriptional activity of Sp1. These findings suggest that the shear stress induction of TF gene expression is mediated through an increased Sp1 transcriptional activity with a concomitant hyperphosphorylation of Sp1.


Subject(s)
Gene Expression Regulation , Thromboplastin/genetics , Endothelium, Vascular/metabolism , Hemorheology , Humans , Phosphorylation , Promoter Regions, Genetic/physiology , Protein Binding/genetics , RNA, Messenger/metabolism , Sp1 Transcription Factor/metabolism , Thromboplastin/metabolism , Transcription, Genetic , Umbilical Veins
15.
Arterioscler Thromb Vasc Biol ; 17(2): 365-74, 1997 Feb.
Article in English | MEDLINE | ID: mdl-9081693

ABSTRACT

Tissue factor (TF) expression by peripheral blood monocytes during sepsis initiates intravascular thrombosis. Bacterial lipopolysaccharide (LPS) rapidly induces TF gene transcription in monocytes. The human TF promoter contains binding sites for the transcription factors AP-1, c-Rel/p65, Egr-1, and Sp1. NF-kappa B/Rel proteins have been shown to physically interact with both AP-1 and Sp1 proteins. In this study, we investigated the role of these transcription factors in uninduced and LPS-induced TF gene expression in human monocytic THP-1 cells. Deletional analysis indicated that five Sp1 sites mediated basal expression in uninduced cells. The two AP-1 sites bound c-Fos/c-Jun heterodimers in both unstimulated and LPS-stimulated cells. Maximal LPS induction of the TF promoter required the two AP-1 sites and the kappa B site within the LPS response element. Disruption of the conserved spacing between the proximal AP-1 site and the kappa B site abolished LPS induction. Replacement of the two AP-1 sites with intrinsically bent DNA partially restored LPS induction, suggesting an additional structural role for the AP-1 sites. Synergistic transactivation of the LPS response element in Drosophila Schneider cells by coexpression of c-Fos, c-Jun, c-Rel, and p65 or c-Jun and p65 required the transactivation domains of c-Jun and p65. These data indicated that c-Fos/c-Jun, c-Rel/p65, and Sp1 regulate TF gene expression in human monocytic cells.


Subject(s)
Gene Expression Regulation , Immediate-Early Proteins , Monocytes/physiology , Thromboplastin/genetics , Animals , Cell Line , DNA/genetics , DNA-Binding Proteins/physiology , Drosophila , Early Growth Response Protein 1 , Humans , Lipopolysaccharides/pharmacology , NF-kappa B/physiology , Nuclear Proteins/metabolism , Plasmids/metabolism , Promoter Regions, Genetic , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Proto-Oncogene Proteins c-rel , Sp1 Transcription Factor/metabolism , Transcription Factor AP-1/physiology , Transcription Factors/physiology
16.
J Immunol ; 159(11): 5450-6, 1997 Dec 01.
Article in English | MEDLINE | ID: mdl-9548485

ABSTRACT

The NF-kappaB family of transcription factors regulates the inducible expression of a variety of genes. Recently, we showed that elevation of intracellular cyclic AMP inhibits NF-kappaB-mediated transcription in human monocytes and endothelial cells without preventing nuclear translocation of NF-kappaB complexes. The present study examined the molecular mechanism of this inhibition. We hypothesized that activation of the protein kinase A signaling pathway may inhibit NF-kappaB-mediated transcription by phosphorylating proteins, such as cAMP response element-binding protein (CREB), which compete for limiting amounts of the coactivator CBP. Here, we show that the amino-terminal region (amino acids 1-450) of CBP specifically interacts with the carboxyl-terminal region (amino acids 286-551) of NF-kappaB p65 (RelA) both in vitro and in vivo. Functional studies using human endothelial cells demonstrated that overexpression of CBP rescued cAMP inhibition of NF-kappaB-mediated transcription and transcription mediated by a chimeric protein, GAL4-p65(286-551), which contained the GAL4 DNA binding domain fused to the carboxyl-terminal region of p65 (amino acids 286-551). In contrast, overexpression of CREB inhibited GAL4-p65(286-551)-mediated transcription. These results suggest that activation of the protein kinase A pathway inhibits NF-kappaB transcription by phosphorylating CREB, which competes with p65 for limiting amounts of CBP.


Subject(s)
Calcium-Binding Proteins , Cyclic AMP Response Element-Binding Protein/physiology , Cyclic AMP/pharmacology , NF-kappa B/pharmacology , Promoter Regions, Genetic , Transcription, Genetic/drug effects , Cells, Cultured , Endothelium/cytology , Humans , Membrane Glycoproteins/metabolism , Monocytes/cytology , Nerve Tissue Proteins/metabolism , Synaptotagmin I , Synaptotagmins
17.
J Biol Chem ; 271(46): 29393-9, 1996 Nov 15.
Article in English | MEDLINE | ID: mdl-8910604

ABSTRACT

The urokinase plasminogen activator (uPA) interacts with its cell surface receptor (uPAR), providing an inducible, localized cell surface proteolytic activity, thereby promoting cellular invasion. Evidence is provided for a novel function of cell surface-associated uPA.uPAR. Specifically, induction of cell surface expression of uPA. uPAR by growth factors or phorbol ester was necessary for vitronectin-dependent carcinoma cell migration, an event mediated by integrin alphavbeta5. Cell migration on vitronectin was blocked with either a soluble form of uPAR, an antibody that disrupts uPA binding to uPAR, or a monoclonal antibody to alphavbeta5. Moreover, plasminogen activator inhibitor type 2 blocked this migration event but did not affect adhesion, suggesting a direct role for uPA enzyme activity in this process and that migration but not adhesion of these cells is regulated by uPA.uPAR. Growth factor-mediated induction of uPA.uPAR on the carcinoma cell surface promotes a specific motility event mediated by integrin alphavbeta5, since cells transfected with the beta3 integrin subunit expressed alphavbeta3 and migrated on vitronectin independently of growth factors or uPA.uPAR expression. This relationship between alphavbeta5 and the uPA.uPAR system has significant implications for regulation of motility events associated with development, angiogenesis, and tumor metastasis.


Subject(s)
Integrins/metabolism , Receptors, Cell Surface/metabolism , Receptors, Vitronectin , Urokinase-Type Plasminogen Activator/metabolism , Cell Movement , Collagen/metabolism , Gene Expression Regulation/drug effects , Humans , Receptors, Cell Surface/genetics , Receptors, Urokinase Plasminogen Activator , Tetradecanoylphorbol Acetate/pharmacology , Transforming Growth Factor alpha/pharmacology , Tumor Cells, Cultured , Urokinase-Type Plasminogen Activator/genetics , Vitronectin/metabolism
18.
J Biol Chem ; 271(34): 20828-35, 1996 Aug 23.
Article in English | MEDLINE | ID: mdl-8702838

ABSTRACT

The NF-kappaB/Rel family of transcription factors regulates the inducible expression of many genes in activated human monocytes and endothelial cells. In this study, we examined the molecular mechanism by which agents that elevate intracellular cAMP inhibit the expression of the tumor necrosis factor alpha (TNFalpha), tissue factor, endothelial leukocyte adhesion molecule-1, and vascular cell adhesion molecule-1 genes. Both forskolin and dibutyryl cAMP, which elevate intracellular cAMP by independent mechanisms, inhibited TNFalpha and tissue factor expression at the level of transcription. Induction of NF-kappaB-dependent gene expression in transiently transfected human monocytic THP-1 cells and human umbilical vein endothelial cells was inhibited by elevated cAMP and by overexpression of the catalytic subunit of protein kinase A (PKA). Elevated cAMP did not prevent nuclear translocation of p50/p65 and c-Rel/p65 heterodimers, decrease nuclear translocation of p65, or significantly modify TNFalpha-induced phosphorylation of p65. Functional studies demonstrated that transcriptional activation of a plasmid containing multimerized kappaB sites by p65 was inhibited by agents that elevate cAMP and by overexpression of the catalytic subunit of PKA. This study indicates that activation of PKA reduces the induction of a distinct set of genes in monocytes and endothelial cells by inhibiting NF-kappaB-mediated transcription.


Subject(s)
Endothelium, Vascular/metabolism , Monocytes/metabolism , NF-kappa B/metabolism , Tumor Necrosis Factor-alpha/physiology , Biological Transport , Cell Compartmentation , Cell Nucleus/metabolism , Cells, Cultured , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/physiology , E-Selectin/genetics , Gene Expression/drug effects , Humans , Lipopolysaccharides/pharmacology , Promoter Regions, Genetic , RNA, Messenger/genetics , Thromboplastin/genetics , Transcription, Genetic , Vascular Cell Adhesion Molecule-1/genetics
19.
Mol Pharmacol ; 49(6): 998-1004, 1996 Jun.
Article in English | MEDLINE | ID: mdl-8649360

ABSTRACT

At sites of inflammation, endothelial cells play a major role in defining the types of leukocytes that are recruited to a specific area. This is accomplished, at least in part, through the cytokine induction of cell surface adhesion molecules, including vascular cell adhesion molecule 1 (VCAM-1). We investigated the role of phosphatidylcholine-specific phospholipase C in the induction of VCAM-1 gene expression by interleukin-1 beta. D609, a phosphatidylcholine-specific phospholipase C inhibitor, reduced VCAM-1 cell surface expression and VCAM-1 promoter activity in human endothelial cells in a dose-dependent manner. D609 did not affect nuclear translocation of nuclear factor-kappa B but inhibited nuclear factor-kappa B-mediated transcription. The results of this study indicate that phosphatidylcholine-specific phospholipase C is required for activation of nuclear factor-kappa B and cytokine induction of VCAM-1 gene expression in endothelial cells.


Subject(s)
Bridged-Ring Compounds/pharmacology , Endothelium, Vascular/drug effects , Gene Expression Regulation/drug effects , Interleukin-1/pharmacology , Thiones/pharmacology , Type C Phospholipases/antagonists & inhibitors , Vascular Cell Adhesion Molecule-1/genetics , Base Sequence , Cells, Cultured , Endothelium, Vascular/metabolism , Humans , Molecular Sequence Data , NF-kappa B/metabolism , Norbornanes , Promoter Regions, Genetic , Thiocarbamates , Type C Phospholipases/physiology
20.
Eur J Immunol ; 26(4): 839-45, 1996 Apr.
Article in English | MEDLINE | ID: mdl-8625976

ABSTRACT

Endothelial cells play a major role in recruiting leukocytes to sites of inflammation. This is accomplished, at least in part, by up-regulation of cell surface adhesion molecules, including VCAM-1 and ICAM-1, in response to cytokines. In this report, we investigated the role of the proteasome complex in mediating the interleukin (IL)- 1 beta induction of VCAM-1 and ICAM-1 gene expression in human endothelial cells. We present evidence that a proteasome inhibitor, n-acetyl-leucinyl-leucinyl-norleucinal (norLEU), as well as specific protease inhibitors, n-tosyl-Lys-chloromethylketone and N-tosyl-Phe-chloromethylketone, blocked IL-1 beta induction of VCAM-1 and ICAM-1 promoter-driven reporter gene expression in stably transfected endothelial cells. These inhibitors also blocked cytokine induced cell surface expression of VCAM-1 and ICAM-1 by human umbilical vein endothelial cells. As expected, the protease inhibitors blocked the activation of nuclear factor (NF)-kappa B in response to IL-1 beta stimulation. In contrast, norLEU did not prevent IL-1 beta-induced nuclear translocation of NF-kappa B. The effects of norLEU were specific because it did not inhibit the IL-1 beta induction of plasminogen activator inhibitor type 1 gene expression. This study demonstrates that inhibition of the proteolytic activity of the proteasome blocks IL-1 beta induction of VCAM-1 and ICAM-1 gene expression in human endothelial cells.


Subject(s)
Calpain/physiology , Cell Nucleus/metabolism , Cysteine Endopeptidases/physiology , Endothelium, Vascular/metabolism , Gene Expression Regulation/drug effects , Intercellular Adhesion Molecule-1/biosynthesis , Leupeptins/pharmacology , Multienzyme Complexes/physiology , NF-kappa B/metabolism , Tosyllysine Chloromethyl Ketone/pharmacology , Tosylphenylalanyl Chloromethyl Ketone/pharmacology , Vascular Cell Adhesion Molecule-1/biosynthesis , Base Sequence , Biological Transport/drug effects , Calpain/antagonists & inhibitors , Cells, Cultured , Endothelium, Vascular/drug effects , Humans , Intercellular Adhesion Molecule-1/genetics , Interleukin-1/pharmacology , Molecular Sequence Data , Plasminogen Activator Inhibitor 1/biosynthesis , Plasminogen Activator Inhibitor 1/genetics , Protease Inhibitors/pharmacology , Proteasome Endopeptidase Complex , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Recombinant Fusion Proteins/metabolism , Transfection , Vascular Cell Adhesion Molecule-1/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...