Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
J Pharmacol Exp Ther ; 387(2): 214-225, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37643795

ABSTRACT

Hereditary angioedema (HAE) is a rare autosomal dominant disorder caused by a deficiency in functional C1 esterase inhibitor, a serpin family protein that blocks the activity of plasma kallikrein. Insufficient inhibition of plasma kallikrein results in the overproduction of bradykinin, a vasoactive inflammatory mediator that produces both pain and unpredictable swelling during HAE attacks, with potentially life-threatening consequences. We describe the generation of STAR-0215, a humanized IgG1 antibody with a long circulating half-life (t1/2) that potently inhibits plasma kallikrein activity, with a >1000-fold lower affinity for prekallikrein and no measurable inhibitory activity against other serine proteases. The high specificity and inhibitory effect of STAR-0215 is demonstrated through a unique allosteric mechanism involving N-terminal catalytic domain binding, destabilization of the activation domain, and reversion of the active site to the inactive zymogen state. The YTE (M252Y/S254T/T256E) modified fragment crystallizable (Fc) domain of STAR-0215 enhances pH-dependent neonatal Fc receptor binding, resulting in a prolonged t1/2 in vivo (∼34 days in cynomolgus monkeys) compared with antibodies without this modification. A single subcutaneous dose of STAR-0215 (≥100 mg) was predicted to be active in patients for 3 months or longer, based on simulations using a minimal physiologically based pharmacokinetic model. These data indicate that STAR-0215, a highly potent and specific antibody against plasma kallikrein with extended t1/2, is a potential agent for long-term preventative HAE therapy administered every 3 months or less frequently. SIGNIFICANCE STATEMENT: STAR-0215 is a YTE-modified immunoglobulin G1 monoclonal antibody with a novel binding mechanism that specifically and potently inhibits the enzymatic activity of plasma kallikrein and prevents the generation of bradykinin. It has been designed to be a long-lasting prophylactic treatment to prevent attacks of HAE and to decrease the burden of disease and the burden of treatment for people with HAE.

2.
Neuromuscul Disord ; 31(5): 385-396, 2021 05.
Article in English | MEDLINE | ID: mdl-33678513

ABSTRACT

Chronic activation of NF-κB is a key driver of muscle degeneration and suppression of muscle regeneration in Duchenne muscular dystrophy. Edasalonexent (CAT-1004) is an orally-administered novel small molecule that covalently links two bioactive compounds (salicylic acid and docosahexaenoic acid) that inhibit NF-κB. This placebo-controlled, proof-of-concept phase 2 study with open-label extension in boys ≥4-<8 years old with any dystrophin mutation examined the effect of edasalonexent (67 or 100 mg/kg/day) compared to placebo or off-treatment control. Endpoints were safety/tolerability, change from baseline in MRI T2 relaxation time of lower leg muscles and functional assessment, as well as pharmacodynamics and biomarkers. Treatment was well-tolerated and the majority of adverse events were mild, and most commonly of the gastrointestinal system (primarily diarrhea). There were no serious adverse events in the edasalonexent groups. Edasalonexent 100 mg/kg was associated with slowing of disease progression and preservation of muscle function compared to an off-treatment control period, with decrease in levels of NF-κB-regulated genes and improvements in biomarkers of muscle health and inflammation. These results support investigating edasalonexent in future trials and have informed the design of the edasalonexent phase 3 clinical trial in boys with Duchenne.


Subject(s)
Arachidonic Acids/therapeutic use , Muscular Dystrophy, Duchenne/drug therapy , NF-kappa B , Salicylamides/therapeutic use , Child , Child, Preschool , Disease Progression , Double-Blind Method , Dystrophin/genetics , Humans , Male , Muscle, Skeletal , Proof of Concept Study
3.
J Neuromuscul Dis ; 6(1): 43-54, 2019.
Article in English | MEDLINE | ID: mdl-30452422

ABSTRACT

BACKGROUND: Edasalonexent is an orally administered small molecule designed to inhibit NF-κB, which is activated from infancy in Duchenne muscular dystrophy and is central to causing muscle damage and preventing muscle regeneration. OBJECTIVE: Evaluate the safety, tolerability, pharmacokinetics and exploratory pharmacodynamics of three doses of edasalonexent in ambulatory males ≥4 to <8 years of age with genetically confirmed Duchenne muscular dystrophy. METHODS: This was a 1-week, open-label, multiple-dose study with 3 sequential ascending doses (33, 67 and 100 mg/kg/day) of edasalonexent administered under different dietary conditions to 17 males with a mean age of 5.5 years. RESULTS: All doses of edasalonexent were well tolerated, with no serious adverse events, no drug discontinuations and no dose reductions. The majority of adverse events were mild, and the most common adverse events were gastrointestinal (primarily diarrhea). Edasalonexent was rapidly absorbed with peak levels observed 2-6 hours after dosing and exposures appeared to increase nearly proportionally to dose for the 2 lower and all 3 doses under low-fat and high-fat meal conditions, respectively. Only minor plasma accumulation of edasalonexent was observed with 7 days of dosing. After treatment with edasalonexent for 7 days, levels of NF-κB-regulated genes and serum proteins were decreased. CONCLUSIONS: This first report of edasalonexent oral administration for one week in male pediatric patients with Duchenne muscular dystrophy showed that treatment was well tolerated and inhibited NF-kB pathways.


Subject(s)
Arachidonic Acids/therapeutic use , Muscular Dystrophy, Duchenne/drug therapy , Neuromuscular Agents/therapeutic use , Salicylamides/therapeutic use , Administration, Oral , Arachidonic Acids/adverse effects , Arachidonic Acids/pharmacokinetics , Child , Child, Preschool , Humans , Male , Muscular Dystrophy, Duchenne/blood , Muscular Dystrophy, Duchenne/urine , NF-kappa B/antagonists & inhibitors , NF-kappa B/blood , Neuromuscular Agents/adverse effects , Neuromuscular Agents/pharmacokinetics , Salicylamides/adverse effects , Salicylamides/pharmacokinetics
4.
Hepatol Commun ; 1(4): 311-325, 2017 06.
Article in English | MEDLINE | ID: mdl-29404461

ABSTRACT

CAT-2003 is a novel conjugate of eicosapentaenoic acid (EPA) and niacin designed to be hydrolyzed by fatty acid amide hydrolase to release EPA inside cells at the endoplasmic reticulum. In cultured liver cells, CAT-2003 blocked the maturation of sterol regulatory element-binding protein (SREBP)-1 and SREBP-2 proteins and decreased the expression of multiple SREBP target genes, including HMGCR and PCSK9. Consistent with proprotein convertase subtilisin/kexin type 9 (PCSK9) reduction, both low-density lipoprotein receptor protein at the cell surface and low-density lipoprotein particle uptake were increased. In apolipoprotein E*3-Leiden mice fed a cholesterol-containing western diet, CAT-2003 decreased hepatic inflammation and steatosis as evidenced by fewer inflammatory cell aggregates in histopathologic sections, decreased nuclear factor kappa B activity in liver lysates, reduced inflammatory gene expression, reduced intrahepatic cholesteryl ester and triglyceride levels, and decreased liver mass. Plasma PCSK9 was reduced and hepatic low-density lipoprotein receptor protein expression was increased; plasma cholesterol and triglyceride levels were lowered. Aortic root segments showed reduction of several atherosclerotic markers, including lesion size, number, and severity. CAT-2003, when dosed in combination with atorvastatin, further lowered plasma cholesterol levels and decreased hepatic expression of SREBP target genes. Conclusion: SREBP inhibition is a promising new strategy for the prevention and treatment of diseases associated with abnormal lipid metabolism, such as atherosclerosis and nonalcoholic steatohepatitis. (Hepatology Communications 2017;1:311-325).

5.
Antioxid Redox Signal ; 24(18): 1058-71, 2016 06 20.
Article in English | MEDLINE | ID: mdl-26980071

ABSTRACT

AIMS: Gastro-resistant dimethyl fumarate (DMF) is an oral therapeutic indicated for the treatment of relapsing multiple sclerosis. Recent data suggest that a primary pharmacodynamic response to DMF treatment is activation of the nuclear factor (erythroid-derived 2)-like 2 (NRF2) pathway; however, the gene targets modulated downstream of NRF2 that contribute to DMF-dependent effects are poorly understood. RESULTS: Using wild-type and NRF2 knockout mice, we characterized DMF transcriptional responses throughout the brain and periphery to understand DMF effects in vivo and to explore the necessity of NRF2 in this process. Our findings identified tissue-specific expression of NRF2 target genes as well as NRF2-dependent and -independent gene regulation after DMF administration. Furthermore, using gene ontology, we identified common biological pathways that may be regulated by DMF and contribute to in vivo functional effects. INNOVATION: Together, these data suggest that DMF modulates transcription through multiple pathways, which has implications for the cytoprotective, immunomodulatory, and clinical properties of DMF. CONCLUSION: These findings provide further understanding of the DMF mechanism of action and propose potential therapeutic targets that warrant further investigation for treating neurodegenerative diseases. Antioxid. Redox Signal. 24, 1058-1071.


Subject(s)
Anti-Inflammatory Agents/pharmacokinetics , Dimethyl Fumarate/pharmacokinetics , NF-E2-Related Factor 2/metabolism , Administration, Oral , Animals , Anti-Inflammatory Agents/administration & dosage , Brain/drug effects , Brain/metabolism , Dimethyl Fumarate/administration & dosage , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Gene Expression Regulation/drug effects , Male , Mice, Inbred C57BL , Mice, Knockout , Organ Specificity , Tissue Distribution , Transcriptome/drug effects
6.
J Med Chem ; 59(3): 1217-31, 2016 Feb 11.
Article in English | MEDLINE | ID: mdl-26784936

ABSTRACT

This report describes the synthesis and preliminary biological characterization of novel fatty acid niacin conjugates and fatty acid salicylate conjugates. These molecular entities were created by covalently linking two bioactive molecules, either niacin or salicylic acid, to an omega-3 fatty acid. This methodology allows the simultaneous intracellular delivery of two bioactives in order to elicit a pharmacological response that could not be replicated by administering the bioactives individually or in combination. The fatty acid niacin conjugate 5 has been shown to be an inhibitor of the sterol regulatory element binding protein (SREBP), a key regulator of cholesterol metabolism proteins such as PCSK9, HMG-CoA reductase, ATP citrate lyase, and NPC1L1. On the other hand, the fatty acid salicylate conjugate 11 has been shown to have a unique anti-inflammatory profile based on its ability to modulate the NF-κB pathway through the intracellular release of the two bioactives.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/chemistry , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Fatty Acids/chemistry , Niacin/chemistry , Niacin/pharmacology , Salicylic Acid/chemistry , Salicylic Acid/pharmacology , Administration, Oral , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Cell Line , Dogs , Dose-Response Relationship, Drug , Hep G2 Cells , Humans , Hydrolysis , Liver/drug effects , Liver/metabolism , Mice , Molecular Structure , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Niacin/administration & dosage , Rats , Rats, Sprague-Dawley , Salicylic Acid/administration & dosage , Sterol Regulatory Element Binding Protein 1/antagonists & inhibitors , Sterol Regulatory Element Binding Protein 1/metabolism , Sterol Regulatory Element Binding Protein 2/antagonists & inhibitors , Sterol Regulatory Element Binding Protein 2/metabolism , Structure-Activity Relationship , Tissue Distribution
7.
J Pharmacol Exp Ther ; 341(1): 274-84, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22267202

ABSTRACT

Oxidative stress is central to the pathology of several neurodegenerative diseases, including multiple sclerosis, and therapeutics designed to enhance antioxidant potential could have clinical value. The objective of this study was to characterize the potential direct neuroprotective effects of dimethyl fumarate (DMF) and its primary metabolite monomethyl fumarate (MMF) on cellular resistance to oxidative damage in primary cultures of central nervous system (CNS) cells and further explore the dependence and function of the nuclear factor (erythroid-derived 2)-like 2 (Nrf2) pathway in this process. Treatment of animals or primary cultures of CNS cells with DMF or MMF resulted in increased nuclear levels of active Nrf2, with subsequent up-regulation of canonical antioxidant target genes. DMF-dependent up-regulation of antioxidant genes in vivo was lost in mice lacking Nrf2 [Nrf2(-/-)]. DMF or MMF treatment increased cellular redox potential, glutathione, ATP levels, and mitochondrial membrane potential in a concentration-dependent manner. Treating astrocytes or neurons with DMF or MMF also significantly improved cell viability after toxic oxidative challenge in a concentration-dependent manner. This effect on viability was lost in cells that had eliminated or reduced Nrf2. These data suggest that DMF and MMF are cytoprotective for neurons and astrocytes against oxidative stress-induced cellular injury and loss, potentially via up-regulation of an Nrf2-dependent antioxidant response. These data also suggest DMF and MMF may function through improving mitochondrial function. The clinical utility of DMF in multiple sclerosis is being explored through phase III trials with BG-12, which is an oral therapeutic containing DMF as the active ingredient.


Subject(s)
Central Nervous System/cytology , Central Nervous System/metabolism , Cytoprotection/genetics , Fumarates/pharmacology , NF-E2-Related Factor 2/physiology , Neurons/metabolism , Oxidative Stress/genetics , Signal Transduction/genetics , Animals , Cells, Cultured , Central Nervous System/drug effects , Cytoprotection/drug effects , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-E2-Related Factor 2/deficiency , Neurons/drug effects , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Signal Transduction/drug effects
8.
Brain ; 134(Pt 3): 678-92, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21354971

ABSTRACT

Inflammation and oxidative stress are thought to promote tissue damage in multiple sclerosis. Thus, novel therapeutics enhancing cellular resistance to free radicals could prove useful for multiple sclerosis treatment. BG00012 is an oral formulation of dimethylfumarate. In a phase II multiple sclerosis trial, BG00012 demonstrated beneficial effects on relapse rate and magnetic resonance imaging markers indicative of inflammation as well as axonal destruction. First we have studied effects of dimethylfumarate on the disease course, central nervous system, tissue integrity and the molecular mechanism of action in an animal model of chronic multiple sclerosis: myelin oligodendrocyte glycoprotein induced experimental autoimmune encephalomyelitis in C57BL/6 mice. In the chronic phase of experimental autoimmune encephalomyelitis, preventive or therapeutic application of dimethylfumarate ameliorated the disease course and improved preservation of myelin, axons and neurons. In vitro, the application of fumarates increased murine neuronal survival and protected human or rodent astrocytes against oxidative stress. Application of dimethylfumarate led to stabilization of the transcription factor nuclear factor (erythroid-derived 2)-related factor 2, activation of nuclear factor (erythroid-derived 2)-related factor 2-dependent transcriptional activity and accumulation of NADP(H) quinoline oxidoreductase-1 as a prototypical target gene. Furthermore, the immediate metabolite of dimethylfumarate, monomethylfumarate, leads to direct modification of the inhibitor of nuclear factor (erythroid-derived 2)-related factor 2, Kelch-like ECH-associated protein 1, at cysteine residue 151. In turn, increased levels of nuclear factor (erythroid-derived 2)-related factor 2 and reduced protein nitrosylation were detected in the central nervous sytem of dimethylfumarate-treated mice. Nuclear factor (erythroid-derived 2)-related factor 2 was also upregulated in the spinal cord of autopsy specimens from untreated patients with multiple sclerosis. In dimethylfumarate-treated mice suffering from experimental autoimmune encephalomyelitis, increased immunoreactivity for nuclear factor (erythroid-derived 2)-related factor 2 was detected by confocal microscopy in neurons of the motor cortex and the brainstem as well as in oligodendrocytes and astrocytes. In mice deficient for nuclear factor (erythroid-derived 2)-related factor 2 on the same genetic background, the dimethylfumarate mediated beneficial effects on clinical course, axon preservation and astrocyte activation were almost completely abolished thus proving the functional relevance of this transcription factor for the neuroprotective mechanism of action. We conclude that the ability of dimethylfumarate to activate nuclear factor (erythroid-derived 2)-related factor 2 may offer a novel cytoprotective modality that further augments the natural antioxidant responses in multiple sclerosis tissue and is not yet targeted by other multiple sclerosis therapies.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/prevention & control , Fumarates/therapeutic use , NF-E2-Related Factor 2/metabolism , Neuroprotective Agents/therapeutic use , Signal Transduction/drug effects , 2',3'-Cyclic-Nucleotide Phosphodiesterases/metabolism , Aldehyde Reductase/metabolism , Animals , Antigens, Differentiation/metabolism , Antioxidants/metabolism , Astrocytes/drug effects , Astrocytes/immunology , Axons/metabolism , Axons/pathology , CD3 Complex/metabolism , Cell Proliferation/drug effects , Cells, Cultured , Chromatography, High Pressure Liquid/methods , Cytokines/metabolism , Disease Models, Animal , Embryo, Mammalian , Encephalomyelitis, Autoimmune, Experimental/etiology , Female , Fumarates/pharmacology , Gene Expression Regulation/drug effects , Glycoproteins/adverse effects , Green Fluorescent Proteins/genetics , Humans , Hydrogen Peroxide/pharmacology , Mass Spectrometry , Mice , Mice, Inbred C57BL , Motor Neurons/cytology , Motor Neurons/drug effects , Motor Neurons/metabolism , Multiple Sclerosis/metabolism , Multiple Sclerosis/pathology , Myelin Proteins/metabolism , Myelin-Oligodendrocyte Glycoprotein , NAD(P)H Dehydrogenase (Quinone)/metabolism , Neuroprotective Agents/pharmacology , Nogo Proteins , Oxidative Stress/drug effects , Peptide Fragments/adverse effects , RNA, Small Interfering/pharmacology , Sleep/physiology , Spinal Cord/cytology , Statistics, Nonparametric , Tandem Mass Spectrometry/methods , Time Factors , Transfection
9.
J Biol Chem ; 285(17): 12971-8, 2010 Apr 23.
Article in English | MEDLINE | ID: mdl-20185819

ABSTRACT

Components of lymphotoxin beta receptor (LTBR)-associated signaling complexes, including TRAF2, TRAF3, NIK, IKK1, and IKK2 have been shown to participate in the coupling of LTBR to NFkappaB. Here, we report that TRAF3 functions as a negative regulator of LTBR signaling via both canonical and non-canonical NFkappaB pathways by two distinct mechanisms. Analysis of NFkappaB signaling in cell lines with functionally intact NFkappaB pathway but lacking LTBR-mediated induction of NFkappaB target genes revealed an inverse association of cellular TRAF3 levels with LTBR-specific defect in canonical NFkappaB activation. Increased expression of TRAF3 correlated with its increased recruitment to LTBR-induced signaling complexes, decreased recruitment of TRAF2, and attenuated phosphorylation of IkappaB alpha and RelA. In contrast, activation of NFkappaB by TNF did not depend on TRAF3 levels. siRNA-mediated depletion of TRAF3 promoted recruitment of TRAF2 and IKK1 to activated LTBR, enabling LTBR-inducible canonical NFkappaB signaling and NFkappaB target gene expression. TRAF3 knock-down also increased mRNA and protein expression of several non-canonical NFkappaB components, including NFkappaB2/p100, RelB, and NIK, accompanied by processing of NFkappaB2/p100 into p52. These effects of TRAF3 depletion did not require LTBR signaling and were consistent with autonomous activation of the non-canonical NFkappaB pathway. Our data illustrate the function of TRAF3 as a dual-mode repressor of LTBR signaling that controls activation of canonical NFkappaB, and de-repression of the intrinsic activity of non-canonical NFkappaB. Modulation of cellular TRAF3 levels may thus contribute to regulation of NFkappaB-dependent gene expression by LTBR by affecting the balance of LTBR-dependent activation of canonical and non-canonical NFkappaB pathways.


Subject(s)
Gene Expression Regulation/physiology , Lymphotoxin beta Receptor/metabolism , NF-kappa B p52 Subunit/metabolism , Signal Transduction/physiology , TNF Receptor-Associated Factor 3/metabolism , Transcription Factor RelA/metabolism , Transcription Factor RelB/metabolism , Cell Line, Tumor , Gene Knockdown Techniques , Humans , I-kappa B Kinase/genetics , I-kappa B Kinase/metabolism , I-kappa B Proteins/genetics , I-kappa B Proteins/metabolism , Lymphotoxin beta Receptor/genetics , NF-kappa B p52 Subunit/genetics , Phosphorylation/physiology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , RNA, Small Interfering/genetics , TNF Receptor-Associated Factor 2/genetics , TNF Receptor-Associated Factor 2/metabolism , TNF Receptor-Associated Factor 3/genetics , Transcription Factor RelA/genetics , Transcription Factor RelB/genetics , NF-kappaB-Inducing Kinase
10.
Cell Cycle ; 8(15): 2425-34, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19556882

ABSTRACT

Most cells in the body are in a resting state and undergo cell cycle progression only upon growth factor stimulation or activation. while much research on proliferation and activation has been performed, very little about signals that maintain quiescent cells in G(0) is known, preventing cell cycle entry or apoptosis. In this study, the pathways of apoptosis induction in quiescent peripheral blood cells and fibroblasts mediated by inhibition or downregulation of Dipeptidyl Peptidase 2 (DPP2) have been explored. A decrease in DPP2 activity was found to cause resting cells to exit from G(0), accompanied by a decrease in p130, p27(Kip1) and p21(Cip1) protein levels. In addition, DPP2-inhibited or downregulated cells exhibit an increase in early G(1)/S progressors, with increases in the levels of retinoblastoma (pRb), p107 and cyclin D proteins. Furthermore, decrease of DPP2 activity leads to an increase in c-Myc and a decrease in Bcl-2, two events that have been associated with apoptosis induction. This apoptosis by DPP2 downregulation is prevented in p53(-/-) cells or by ectopic expression of proteins that suppress p53 or c-Myc activity. Thus, DPP2 is essential for maintaining lymphocytes and fibroblasts in G(0), and its inhibition results in apoptosis mediated by induction of c-Myc and p53.


Subject(s)
Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/metabolism , Leukocytes, Mononuclear/metabolism , Retinoblastoma Protein/metabolism , Apoptosis/drug effects , Apoptosis/physiology , Butadienes/pharmacology , Cyclin D , Cyclins/metabolism , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Extracellular Signal-Regulated MAP Kinases/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Flavonoids/pharmacology , Humans , Leukocytes, Mononuclear/drug effects , MAP Kinase Kinase Kinases/antagonists & inhibitors , MAP Kinase Kinase Kinases/metabolism , Nitriles/pharmacology , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Resting Phase, Cell Cycle/drug effects , Resting Phase, Cell Cycle/physiology , Retinoblastoma-Like Protein p107/metabolism , Tumor Suppressor Protein p53/metabolism
11.
Mol Immunol ; 45(13): 3618-23, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18555530

ABSTRACT

We have shown previously that dipeptidyl peptidase 2 (DPP2) activity is essential for the survival of quiescent, but not activated, lymphocytes. The specific requirement of DPP2 activity for non-dividing cells is indicative of cell cycle specific regulation of this gene product. In the present study, we tested this hypothesis by looking at contact and serum dependence of Dpp2 transcription. We found that transfected promoter-reporter activity, as well as endogenous Dpp2 transcripts, were enhanced in NIH-3T3 cells upon contact-inhibition or serum starvation. Since lung Kruppel-like factor (KLF2), a transcription factor, and TOB1, a transcriptional co-activator, have been shown to be important in maintaining T-lymphocyte quiescence and are both downregulated upon cellular activation, we also looked at the contributions of these factors to Dpp2 transcription. Using a Dpp2 promoter-reporter system, we demonstrate that KLF2 and TOB1 activate the mouse Dpp2 promoter. Finally, we show that in human PBMC, there is a decrease in levels of endogenous DPP2 transcripts upon T cell receptor activation when compared to resting cells. These results demonstrate that Dpp2 transcription is serum and contact-dependent and link two quiescence-specific transcriptional elements to the quiescence-specific requirement of DPP2 enzymatic activity.


Subject(s)
Carrier Proteins/physiology , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/genetics , Kruppel-Like Transcription Factors/physiology , Lymphocyte Activation/genetics , Transcriptional Activation , Animals , Base Sequence , Intracellular Signaling Peptides and Proteins , Mice , Mice, Inbred BALB C , Molecular Sequence Data , NIH 3T3 Cells , Promoter Regions, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...