Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Front Cardiovasc Med ; 8: 651230, 2021.
Article in English | MEDLINE | ID: mdl-34026871

ABSTRACT

Rationale: Decreased expression and activity of endothelial nitric oxide synthase (eNOS) in response to inflammatory and metabolic insults is the hallmark of endothelial cell (EC) dysfunction that preludes the development of atherosclerosis and hypertension. We previously reported the atheroprotective properties of the ubiquitin-editing and anti-inflammatory protein A20, also known as TNFAIP3, in part through interrupting nuclear factor-kappa B (NF-κB) and interferon signaling in EC and protecting these cells from apoptosis. However, A20's effect on eNOS expression and function remains unknown. In this study, we evaluated the impact of A20 overexpression or knockdown on eNOS expression in EC, at baseline and after tumor necrosis factor (TNF) treatment, used to mimic inflammation. Methods and Results: A20 overexpression in human coronary artery EC (HCAEC) significantly increased basal eNOS mRNA (qPCR) and protein (western blot) levels and prevented their downregulation by TNF. Conversely, siRNA-induced A20 knockdown decreased eNOS mRNA levels, identifying A20 as a physiologic regulator of eNOS expression. By reporter assays, using deletion and point mutants of the human eNOS promoter, and knockdown of eNOS transcriptional regulators, we demonstrated that A20-mediated increase of eNOS was transcriptional and relied on increased expression of the transcription factor Krüppel-like factor (KLF2), and upstream of KLF2, on activation of extracellular signal-regulated kinase 5 (ERK5). Accordingly, ERK5 knockdown or inhibition significantly abrogated A20's ability to increase KLF2 and eNOS expression. In addition, A20 overexpression in HCAEC increased eNOS phosphorylation at Ser-1177, which is key for the function of this enzyme. Conclusions: This is the first report demonstrating that overexpression of A20 in EC increases eNOS transcription in an ERK5/KLF2-dependent manner and promotes eNOS activating phosphorylation. This effect withstands eNOS downregulation by TNF, preventing EC dysfunction in the face of inflammation. This novel function of A20 further qualifies its therapeutic promise to prevent/treat atherosclerosis.

2.
Biochim Biophys Acta ; 1833(6): 1553-61, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23499873

ABSTRACT

A1/Bfl-1 is a NF-κB dependent, anti-apoptotic Bcl-2 family member that contains four Bcl-2 homology domains (BH) and an amphipathic C-terminal domain, and is expressed in endothelial cells (EC). Based on NF-κB reporter assays in bovine aortic EC, we have previously demonstrated that A1, like Bcl-2 and Bcl-xL, inhibits NF-κB activation. These results, however, do not fully translate when evaluating the cell's own NF-κB machinery in human EC overexpressing A1 by means of recombinant adenovirus (rAd.) mediated gene transfer. Indeed, overexpression of full-length A1 in human umbilical vein EC (HUVEC), and human dermal microvascular EC (HDMEC) failed to inhibit NF-κB activation. However, overexpression of a mutant lacking the C-terminal domain of A1 (A1ΔC) demonstrated a potent NF-κB inhibitory effect in these cells. Disparate effects of A1 and A1ΔC on NF-κB inhibition in human EC correlated with mitochondrial (A1) versus non-mitochondrial (A1ΔC) localization. In contrast, both full-length A1 and A1ΔC protected EC from staurosporine (STS)-induced cell death, indicating that mitochondrial localization was not necessary for A1's cytoprotective function in human EC. In conclusion, our data uncover a regulatory role for the C-terminal domain of A1 in human EC: anchoring A1 to the mitochondrion, which conserves but is not necessary for its cytoprotective function, or by its absence freeing A1 from the mitochondrion and uncovering an additional anti-inflammatory effect.


Subject(s)
Anti-Inflammatory Agents/metabolism , Dermis/metabolism , Endothelium, Vascular/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Animals , Apoptosis , Blotting, Western , Cattle , Cell Proliferation , Dermis/cytology , Endothelium, Vascular/cytology , Fluorescent Antibody Technique , Gene Expression Regulation , Human Umbilical Vein Endothelial Cells/cytology , Humans , Intercellular Adhesion Molecule-1/metabolism , Luciferases/metabolism , Minor Histocompatibility Antigens , NF-kappa B/genetics , NF-kappa B/metabolism , Protein Structure, Tertiary , Proto-Oncogene Proteins c-bcl-2/genetics
3.
FASEB J ; 20(9): 1418-30, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16816117

ABSTRACT

A20 is a NF-kappaB-dependent gene that has dual anti-inflammatory and antiapoptotic functions in endothelial cells (EC). The function of A20 in smooth muscle cells (SMC) is unknown. We demonstrate that A20 is induced in SMC in response to inflammatory stimuli and serves an anti-inflammatory function via blockade of NF-kappaB and NF-kappaB-dependent proteins ICAM-1 and MCP-1. A20 inhibits SMC proliferation via increased expression of cyclin-dependent kinase inhibitors p21waf1 and p27kip1. Surprisingly, A20 sensitizes SMC to cytokine- and Fas-mediated apoptosis through a novel NO-dependent mechanism. In vivo, adenoviral delivery of A20 to medial rat carotid artery SMC after balloon angioplasty prevents neointimal hyperplasia by blocking SMC proliferation and accelerating re-endothelialization, without causing apoptosis. However, expression of A20 in established neointimal lesions leads to their regression through increased apoptosis. This is the first demonstration that A20 exerts two levels of control of vascular remodeling and healing. A20 prevents neointimal hyperplasia through combined anti-inflammatory and antiproliferative functions in medial SMC. If SMC evade this first barrier and neointima is formed, A20 has a therapeutic potential by uniquely sensitizing neointimal SMC to apoptosis. A20-based therapies hold promise for the prevention and treatment of neointimal disease.


Subject(s)
Hyperplasia/prevention & control , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/physiology , NF-kappa B/antagonists & inhibitors , Proteins/physiology , Proteins/therapeutic use , Tunica Intima/pathology , Adenoviridae , Animals , Aorta , Apoptosis , Cell Cycle , Cell Division , DNA Primers , DNA-Binding Proteins , Endothelium, Vascular/physiology , Gene Expression Regulation , Genetic Vectors , Humans , Intercellular Adhesion Molecule-1/genetics , Intracellular Signaling Peptides and Proteins , Nuclear Proteins , Reverse Transcriptase Polymerase Chain Reaction , Swine , Tumor Necrosis Factor alpha-Induced Protein 3 , Vascular Diseases/prevention & control
4.
Kidney Int ; 68(4): 1520-32, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16164629

ABSTRACT

BACKGROUND: Apoptotic death of renal proximal tubular epithelial cells (RPTECs) is a feature of acute and chronic renal failure. RPTECs are directly damaged by ischemia, inflammatory, and cytotoxic mediators but also contribute to their own demise by up-regulating proinflammatory nuclear factor-kappaB (NF-kappaB)-dependent proteins. In endothelial cells, the Bcl family member A1 and the zinc finger protein A20 have redundant and dual antiapoptotic and anti-inflammatory effects. We studied the function(s) of A1 and A20 in human RPTECs in vitro. METHODS: Expression of A1 [reverse transcription-polymerase chain reaction (RT-PCR) and A20 (Northern and Western blot analysis)] in RPTECs was evaluated. A1 and A20 were overexpressed in RPTECs by recombinant adenoviral-mediated gene transfer. Their effect upon inhibitor of NFkappaB alpha (IkappaBalpha) degradation (Western blot), NF-kappaB nuclear translocation [electrophoretic mobility shift assay (EMSA)], up-regulation of intercellular adhesion molecule-1 (ICAM-1) [fluorescence-activated cell sorter (FACS)] and monocyte chemoattractant protein-1 (MCP-1) (Northern blot) and apoptosis [terminal deoxynucleotiddyl transferase (TdT)-mediated deoxyuridine triphosphate (dUTP) nick-end labeling (TUNEL)] and FACS analysis of DNA content) was determined. RESULTS: A1 and A20 were induced in RPTECs as part of the physiologic response to tumor necrosis factor (TNF). A20, but not A1, inhibited TNF-induced NF-kappaB activation by preventing IkappaBalpha degradation, hence subsequent up-regulation of the proinflammatory molecules ICAM-1 and MCP-1. Unexpectedly, A20 did not protect RPTECs from TNF and Fas-mediated apoptosis while A1 protected against both stimuli. Coexpression of A1 and A20 in RPTECs achieved additive anti-inflammatory and antiapoptotic cytoprotection. CONCLUSION: A1 and A20 exert differential cytoprotective effects in RPTECs. A1 is antiapoptotic. A20 is anti-inflammatory via blockade of NF-kappaB. We propose that A1 and A20 are both required for optimal protection of RPTECs from apoptosis (A1) and inflammation (A20) in conditions leading to renal damage.


Subject(s)
Apoptosis/physiology , Epithelial Cells/physiology , Kidney Tubules, Proximal/physiology , Proteins/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , Cells, Cultured , Chemokine CCL2/genetics , DNA-Binding Proteins , Epithelial Cells/cytology , Gene Expression/physiology , Humans , I-kappa B Proteins/metabolism , Intercellular Adhesion Molecule-1/genetics , Intracellular Signaling Peptides and Proteins , Kidney Tubules, Proximal/cytology , Minor Histocompatibility Antigens , NF-KappaB Inhibitor alpha , NF-kappa B/metabolism , Nephritis/pathology , Nephritis/physiopathology , Nuclear Proteins , RNA, Messenger/analysis , Tumor Necrosis Factor alpha-Induced Protein 3 , Tumor Necrosis Factor-alpha/pharmacology , Up-Regulation , fas Receptor/metabolism
5.
Hepatology ; 42(1): 156-64, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15962316

ABSTRACT

The liver has a remarkable regenerative capacity, allowing recovery following injury. Regeneration after injury is contingent on maintenance of healthy residual liver mass, otherwise fulminant hepatic failure (FHF) may arise. Understanding the protective mechanisms safeguarding hepatocytes and promoting their proliferation is critical for devising therapeutic strategies for FHF. We demonstrate that A20 is part of the physiological response of hepatocytes to injury. In particular, A20 is significantly upregulated in the liver following partial hepatectomy. A20 protects hepatocytes from apoptosis and ongoing inflammation by inhibiting NF-kappaB. Hepatic expression of A20 in BALB/c mice dramatically improves survival following extended and radical lethal hepatectomy. A20 expression in the liver limits hepatocellular damage hence maintains bilirubin clearance and the liver synthetic function. In addition, A20 confers a proliferative advantage to hepatocytes via decreased expression of the cyclin-dependent kinase inhibitor p21(waf1). In conclusion, A20 provides a proliferative advantage to hepatocytes. By combining anti-inflammatory, antiapoptotic and pro-proliferative functions, A20-based therapies could be beneficial in prevention and treatment of FHF.


Subject(s)
Hepatectomy/adverse effects , Liver Failure/genetics , Liver Regeneration/genetics , Proteins/genetics , Zinc Fingers/genetics , Animals , Cell Cycle Proteins/physiology , Cell Proliferation , Cyclin-Dependent Kinase Inhibitor p21 , Cysteine Endopeptidases , Hepatocytes/physiology , Intracellular Signaling Peptides and Proteins , Liver/physiology , Liver Failure/etiology , Mice , Models, Animal , Nuclear Proteins , Recovery of Function , Regeneration/physiology , Survival Analysis , Tumor Necrosis Factor alpha-Induced Protein 3
6.
Blood ; 104(8): 2376-84, 2004 Oct 15.
Article in English | MEDLINE | ID: mdl-15251990

ABSTRACT

A20 is a stress response gene in endothelial cells (ECs). A20 serves a dual cytoprotective function, protecting from tumor necrosis factor (TNF)-mediated apoptosis and inhibiting inflammation via blockade of the transcription factor nuclear factor-kappaB (NF-kappaB). In this study, we evaluated the molecular basis of the cytoprotective function of A20 in EC cultures and questioned whether its protective effect extends beyond TNF to other apoptotic and necrotic stimuli. Our data demonstrate that A20 targets the TNF apoptotic pathway by inhibiting proteolytic cleavage of apical caspases 8 and 2, executioner caspases 3 and 6, Bid cleavage, and release of cytochrome c, thus preserving mitochondrion integrity. A20 also protects from Fas/CD95 and significantly blunts natural killer cell-mediated EC apoptosis by inhibiting caspase 8 activation. In addition to protecting ECs from apoptotic stimuli, A20 safeguards ECs from complement-mediated necrosis. These data demonstrate, for the first time, that the cytoprotective effect of A20 in ECs is not limited to TNF-triggered apoptosis. Rather, A20 affords broad EC protective functions by effectively shutting down cell death pathways initiated by inflammatory and immune offenders.


Subject(s)
Apoptosis/drug effects , Caspase Inhibitors , Endothelial Cells/drug effects , Killer Cells, Natural/immunology , Proteins/metabolism , Tumor Necrosis Factor-alpha/pharmacology , fas Receptor/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Caspase 3 , Caspase 6 , Caspase 8 , Caspases/metabolism , Cattle , Cells, Cultured , Complement System Proteins/immunology , Cycloheximide/pharmacology , DNA-Binding Proteins , Endothelial Cells/cytology , Endothelial Cells/metabolism , Enzyme Activation/drug effects , Fas-Associated Death Domain Protein , Gene Expression , Hot Temperature , Humans , Intracellular Signaling Peptides and Proteins , Killer Cells, Natural/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , NF-kappa B/metabolism , Necrosis , Nuclear Proteins , Proteins/genetics , Signal Transduction/drug effects , Swine , Tumor Necrosis Factor alpha-Induced Protein 3 , fas Receptor/genetics
7.
Transplantation ; 77(7): 990-7, 2004 Apr 15.
Article in English | MEDLINE | ID: mdl-15087759

ABSTRACT

BACKGROUND: The prevention of recurrent autoimmunity is a prerequisite for successful islet transplantation in patients with type I diabetes. Therapies effective in preserving pancreatic beta-cell mass in patients with newly diagnosed diabetes are good candidates for achieving this goal. Anti-CD3 monoclonal antibody (mAb) and antilymphocyte antisera are the only therapies to date that have cured early diabetic disease in the nonobese diabetic (NOD) mouse. We investigated whether other immunosuppressive therapies, including short-term depleting anti-CD4 mAb or costimulation blockade, would affect the disease progression in recently diabetic NOD mice. We also evaluated the effect of the anti-CD4 mAb on syngeneic and allogeneic graft survival in diabetic NOD recipients. METHODS AND RESULTS: We demonstrate that a short course of anti-CD4 mAb early after hyperglycemia onset cured diabetes. Normal islets and islets with CD4+ and CD8+ T-cell peri-insulitic infiltrate were found in the pancreata of cured NOD mice. A similar regimen prevented the recurrence of autoimmune diabetes in NOD/severe combined immunodeficient disease (SCID) islet isografts and delayed the rejection of allogeneic C57BL/6 islet allografts in diabetic female NOD mice. The co-transfer of diabetogenic splenocytes with splenocytes from anti-CD4 mAb-treated and cured NOD mice into 7-week-old, irradiated, NOD male mice was not able to protect from diabetes occurrence. This indicates that an anti-CD4-mediated cure of diabetes is independent of the induction of immunoregulatory T cells. Anti-CD154 mAb and cytotoxic T-lymphocyte antigen 4 immunoglobulin were ineffective in early-onset diabetes. CONCLUSION: Our results provide the first evidence that newly established autoimmune islet destruction in NOD mice responds to a short course of anti-CD4 mAb. In contrast, costimulation blockade is ineffective in this clinically relevant model.


Subject(s)
Antibodies, Monoclonal/therapeutic use , CD4-Positive T-Lymphocytes/immunology , Diabetes Mellitus, Type 1/prevention & control , Graft Rejection/prevention & control , Islets of Langerhans Transplantation/immunology , Lymphocyte Depletion , Adoptive Transfer , Animals , Female , Mice , Mice, Inbred NOD , Pancreas/immunology , Pancreas/pathology , Recurrence , Transplantation, Homologous
8.
Circulation ; 108(9): 1113-8, 2003 Sep 02.
Article in English | MEDLINE | ID: mdl-12885753

ABSTRACT

BACKGROUND: CD40/CD40 ligand (CD40L) signaling is a potent activator of endothelial cells (ECs) and promoter of atherosclerosis. In this study, we investigate whether A20 (a gene we have shown to be antiinflammatory and antiapoptotic in ECs) can protect from CD40/CD40L-mediated EC activation. METHODS AND RESULTS: Overexpression of CD40, in a transient transfection system, activates the transcription factor NF-kappaB and upregulates IkappaBalpha, E-selectin, and tissue factor (TF) reporter activity. Coexpression of A20 inhibits NF-kappaB and upregulation of IkappaBalpha and E-Selectin but not TF, suggesting that CD40 induces TF in a non-NF-kappaB-dependent manner. In human coronary artery ECs (HCAECs), adenovirus-mediated overexpression of A20 blocks physiological, CD40-induced activation of NF-kappaB, upstream of IkappaBalpha degradation (Western blot) and subsequently upregulation of ICAM-1, VCAM-1, and E-selectin (flow cytometry). Although A20 does not block TF transcription its expression in HCAECs inhibits TF induction (colorimetric assay and RT-PCR) by blunting CD40 upregulation. We demonstrate that CD40 signaling induces apoptosis in a proinflammatory microenvironment. A20 overexpression protects from CD40-mediated EC apoptosis (DNA content analysis and trypan blue exclusion). We also demonstrate that signaling through CD40L activates NF-kappaB and induces apoptosis in ECs, both of which are inhibited by A20 overexpression. CONCLUSIONS: A20 works at multiple levels to protect ECs from CD40/CD40L mediated activation and apoptosis. A20-based therapy could be beneficial for the treatment of vascular diseases such as atherosclerosis and transplant-associated vasculopathy.


Subject(s)
Apoptosis , CD40 Antigens/metabolism , CD40 Ligand/pharmacology , Endothelium, Vascular/metabolism , Proteins/physiology , Animals , CD40 Antigens/genetics , Cattle , Cells, Cultured , Cytokines/antagonists & inhibitors , Cytoprotection , DNA-Binding Proteins , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Humans , Intracellular Signaling Peptides and Proteins , NF-kappa B/metabolism , Nuclear Proteins , Proteins/genetics , Signal Transduction , Thromboplastin/biosynthesis , Thromboplastin/genetics , Transcriptional Activation , Transfection , Tumor Necrosis Factor alpha-Induced Protein 3 , Tumor Necrosis Factor-alpha/pharmacology , U937 Cells , Up-Regulation
9.
J Immunol ; 170(12): 6250-6, 2003 Jun 15.
Article in English | MEDLINE | ID: mdl-12794157

ABSTRACT

Transplantation of an excessive number of islets of Langerhans (two to four pancreata per recipient) into patients with type I diabetes is required to restore euglycemia. Hypoxia, nutrient deprivation, local inflammation, and the beta cell inflammatory response (up-regulation of NF-kappaB-dependent genes such as inos) result in beta cell destruction in the early post-transplantation period. Genetic engineering of islets with anti-inflammatory and antiapoptotic genes may prevent beta cell loss and primary nonfunction. We have shown in vitro that A20 inhibits NF-kappaB activation in islets and protects from cytokine- and death receptor-mediated apoptosis. In vivo, protection of newly transplanted islets would reduce the number of islets required for successful transplantation. Transplantation of 500 B6/AF(1) mouse islets into syngeneic, diabetic recipients resulted in a cure rate of 100% within 5 days. Transplantation of 250 islets resulted in a cure rate of only 20%. Transplantation of 250 islets overexpressing A20 resulted in a cure rate of 75% with a mean time to cure of 5.2 days, comparable to that achieved with 500 islets. A20-expressing islets preserve functional beta cell mass and are protected from cell death. These data demonstrate that A20 is an ideal cytoprotective gene therapy candidate for islet transplantation.


Subject(s)
Graft Survival/genetics , Graft Survival/immunology , Islets of Langerhans Transplantation/methods , Islets of Langerhans/immunology , Protein Engineering/methods , Proteins/genetics , Proteins/therapeutic use , Adenoviridae/genetics , Animals , Apoptosis/genetics , Apoptosis/immunology , Cysteine Endopeptidases , Cytoprotection/genetics , Cytoprotection/immunology , DNA-Binding Proteins , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Experimental/therapy , Gene Transfer Techniques , Genetic Vectors , Humans , Insulin/biosynthesis , Intracellular Signaling Peptides and Proteins , Islets of Langerhans/metabolism , Islets of Langerhans/pathology , Islets of Langerhans Transplantation/immunology , Islets of Langerhans Transplantation/pathology , Male , Mice , Mice, Inbred A , Mice, Inbred C57BL , Nuclear Proteins , Postoperative Period , Protective Agents/metabolism , Protective Agents/therapeutic use , Protective Agents/toxicity , Protein Biosynthesis , Proteins/toxicity , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Recombinant Proteins/therapeutic use , Recombinant Proteins/toxicity , Tumor Necrosis Factor alpha-Induced Protein 3
10.
Hepatology ; 35(3): 535-43, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11870365

ABSTRACT

Apoptosis of hepatocytes is a seminal feature of fulminant hepatic failure. We show that the anti-apoptotic protein A20 is upregulated in hepatocytes by pro-inflammatory stimuli and functions to protect from apoptosis and limit inflammation by inhibiting NF-kappaB. Adenoviral mediated hepatic expression of A20 in BALB/c mice yields an 85% survival rate in the D-galactosamine (D-gal)/lipolysaccharide (LPS) model of acute toxic hepatitis compared with 15% to 20 % in control mice. Expression of A20 preserves normal liver function as assessed by prothrombin time. The protective effect of A20 is independent of tumor necrosis factor (TNF) inhibition. Maintaining high circulating TNF levels may be advantageous for liver regeneration. Our data supports this hypothesis as evidenced by increased proliferating cell nuclear antigen (PCNA) expression in the livers of mice expressing A20 compared with a dominant negative mutant of the TNF receptor (TNF-R), 6 hours following D-gal/LPS administration. In conclusion, these results qualify A20 as part of a physiologic, protective response of hepatocytes to injury and a promising gene therapy candidate for clinical applications aimed at preventing and treating viral and toxic fulminant hepatic failure.


Subject(s)
Chemical and Drug Induced Liver Injury/therapy , Genetic Therapy , Liver Failure/therapy , Proteins/physiology , Adenoviridae/genetics , Animals , Apoptosis , Cysteine Endopeptidases , Cytokines/biosynthesis , Cytoprotection , DNA-Binding Proteins , Galactosamine/toxicity , Hepatocytes/metabolism , Humans , Inflammation/prevention & control , Intracellular Signaling Peptides and Proteins , Lipopolysaccharides/toxicity , Male , Mice , Mice, Inbred BALB C , NF-kappa B/physiology , Nuclear Proteins , Proteins/genetics , Receptors, Tumor Necrosis Factor/genetics , Tumor Cells, Cultured , Tumor Necrosis Factor alpha-Induced Protein 3
SELECTION OF CITATIONS
SEARCH DETAIL
...