Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Clin Pharmacol Ther ; 110(5): 1250-1260, 2021 11.
Article in English | MEDLINE | ID: mdl-34510420

ABSTRACT

Givosiran (trade name GIVLAARI) is a small interfering ribonucleic acid that targets hepatic delta-aminolevulinic acid synthase 1 (ALAS1) messenger RNA for degradation through RNA interference (RNAi) that has been approved for the treatment of acute hepatic porphyria (AHP). RNAi therapeutics, such as givosiran, have a low liability for drug-drug interactions (DDIs) because they are not metabolized by cytochrome 450 (CYP) enzymes, and do not directly inhibit or induce CYP enzymes in the liver. The pharmacodynamic effect of givosiran (lowering of hepatic ALAS1, the first and rate limiting enzyme in the heme biosynthesis pathway) presents a unique scenario where givosiran could potentially impact heme-dependent activities in the liver, such as CYP enzyme activity. This study assessed the impact of givosiran on the pharmacokinetics of substrates of 5 major CYP450 enzymes in subjects with acute intermittent porphyria (AIP), the most common type of AHP, by using the validated "Inje cocktail," comprised of caffeine (CYP1A2), losartan (CYP2C9), omeprazole (CYP2C19), dextromethorphan (CYP2D6), and midazolam (CYP3A4). We show that givosiran treatment had a differential inhibitory effect on CYP450 enzymes in the liver, resulting in a moderate reduction in activity of CYP1A2 and CYP2D6, a minor effect on CYP3A4 and CYP2C19, and a similar weak effect on CYP2C9. To date, this is the first study evaluating the DDI for an oligonucleotide therapeutic and highlights an atypical drug interaction due to the pharmacological effect of givosiran. The results of this study suggest that givosiran does not have a large effect on heme-dependent CYP enzyme activity in the liver.


Subject(s)
Acetylgalactosamine/analogs & derivatives , Cytochrome P-450 Enzyme System/metabolism , Drug Interactions/physiology , Enzyme Activation/physiology , Liver/metabolism , Pyrrolidines/metabolism , RNA, Small Interfering/metabolism , 5-Aminolevulinate Synthetase/metabolism , Acetylgalactosamine/administration & dosage , Acetylgalactosamine/metabolism , Adult , Caffeine/administration & dosage , Caffeine/metabolism , Cross-Over Studies , Enzyme Activation/drug effects , Female , Humans , Liver/drug effects , Male , Metabolic Clearance Rate/drug effects , Metabolic Clearance Rate/physiology , Midazolam/administration & dosage , Midazolam/metabolism , Middle Aged , Omeprazole/administration & dosage , Omeprazole/metabolism , Porphyrias, Hepatic/drug therapy , Porphyrias, Hepatic/metabolism , Pyrrolidines/administration & dosage
2.
JIMD Rep ; 57(1): 85-93, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33473344

ABSTRACT

Acute hepatic porphyria (AHP) is a family of rare, serious, and potentially life-threatening metabolic disorders caused by mutations in genes encoding enzymes involved in hepatic heme biosynthesis. AHP is characterized by accumulation of neurotoxic heme intermediates, δ-aminolevulinic acid (ALA), and porphobilinogen (PBG), which are thought to be causal for the disease manifestations. Novel therapeutic treatments such as givosiran, an RNA interference therapeutic that was recently approved for treatment of adults with AHP, are focused on reducing the levels of ALA and PBG in patients toward levels observed in a healthy population. While there are two published reports on the distribution of urinary ALA and PBG levels in healthy subjects, these lacked the required details to enable the calculation of reference limits for ALA and PBG. Therefore, urinary ALA and PBG levels were quantified in 150 healthy subjects using a validated liquid chromatography tandem mass spectrometry (LC-MS/MS) method that is highly sensitive, specific, accurate, and reproducible. These data were used to establish the upper limit of normal (ULN) values for ALA and PBG as 1.47 and 0.137 mmol/mol Cr, respectively. Relative to these ULN values, baseline urinary ALA and PBG levels in AHP patients were found to be 9.3- to 12-fold, and 238- to 336-fold higher, respectively. Results from this study can serve as a guide to assess the effectiveness of therapeutic interventions in lowering ALA and PBG.

3.
N Engl J Med ; 382(24): 2289-2301, 2020 06 11.
Article in English | MEDLINE | ID: mdl-32521132

ABSTRACT

BACKGROUND: Up-regulation of hepatic delta-aminolevulinic acid synthase 1 (ALAS1), with resultant accumulation of delta-aminolevulinic acid (ALA) and porphobilinogen, is central to the pathogenesis of acute attacks and chronic symptoms in acute hepatic porphyria. Givosiran, an RNA interference therapy, inhibits ALAS1 expression. METHODS: In this double-blind, placebo-controlled, phase 3 trial, we randomly assigned symptomatic patients with acute hepatic porphyria to receive either subcutaneous givosiran (2.5 mg per kilogram of body weight) or placebo monthly for 6 months. The primary end point was the annualized rate of composite porphyria attacks among patients with acute intermittent porphyria, the most common subtype of acute hepatic porphyria. (Composite porphyria attacks resulted in hospitalization, an urgent health care visit, or intravenous administration of hemin at home.) Key secondary end points were levels of ALA and porphobilinogen and the annualized attack rate among patients with acute hepatic porphyria, along with hemin use and daily worst pain scores in patients with acute intermittent porphyria. RESULTS: A total of 94 patients underwent randomization (48 in the givosiran group and 46 in the placebo group). Among the 89 patients with acute intermittent porphyria, the mean annualized attack rate was 3.2 in the givosiran group and 12.5 in the placebo group, representing a 74% lower rate in the givosiran group (P<0.001); the results were similar among the 94 patients with acute hepatic porphyria. Among the patients with acute intermittent porphyria, givosiran led to lower levels of urinary ALA and porphobilinogen, fewer days of hemin use, and better daily scores for pain than placebo. Key adverse events that were observed more frequently in the givosiran group were elevations in serum aminotransferase levels, changes in serum creatinine levels and the estimated glomerular filtration rate, and injection-site reactions. CONCLUSIONS: Among patients with acute intermittent porphyria, those who received givosiran had a significantly lower rate of porphyria attacks and better results for multiple other disease manifestations than those who received placebo. The increased efficacy was accompanied by a higher frequency of hepatic and renal adverse events. (Funded by Alnylam Pharmaceuticals; ENVISION ClinicalTrials.gov number, NCT03338816.).


Subject(s)
Acetylgalactosamine/analogs & derivatives , Aminolevulinic Acid/urine , Porphobilinogen/urine , Porphyria, Acute Intermittent/drug therapy , Pyrrolidines/therapeutic use , RNAi Therapeutics , Acetylgalactosamine/adverse effects , Acetylgalactosamine/therapeutic use , Adult , Double-Blind Method , Fatigue/etiology , Female , Humans , Injections, Subcutaneous , Least-Squares Analysis , Liver/drug effects , Male , Nausea/etiology , Pain/etiology , Patient Outcome Assessment , Porphyria, Acute Intermittent/complications , Porphyria, Acute Intermittent/urine , Pyrrolidines/adverse effects , Renal Insufficiency, Chronic/chemically induced , Transaminases/blood
4.
Clin Pharmacol Ther ; 108(1): 63-72, 2020 07.
Article in English | MEDLINE | ID: mdl-31994716

ABSTRACT

Givosiran is a small interfering ribonucleic acid agent that was recently approved in the United States for the treatment of acute hepatic porphyria (AHP). This phase I study evaluated the safety, pharmacokinetic, and pharmacodynamic profile of subcutaneously (SC) administered givosiran in patients with acute intermittent porphyria, the most common AHP type. Givosiran was rapidly absorbed from the SC injection site with peak plasma concentrations achieved within 0.5-5 hours followed by elimination with a short half-life of 4-10 hours. Plasma exposures of AS(N-1)3' givosiran, an active metabolite with equal potency as givosiran, was 35%-75%. Givosiran treatment resulted in a rapid and dose-dependent reduction in urinary aminolevulinic acid (ALA) and porphobilinogen (PBG) towards the upper limit of normal (ULN) in AHP patients. Greater and more sustained reductions in ALA and PBG were achieved with once monthly dosing compared with once quarterly dosing. After monthly dosing, trough ALA levels were reduced to below the ULN, approximately 95% reduction from baseline, at both the 2.5 and 5.0 mg/kg doses.


Subject(s)
Acetylgalactosamine/analogs & derivatives , Aminolevulinic Acid/urine , Porphobilinogen/urine , Porphyria, Acute Intermittent/drug therapy , Pyrrolidines/administration & dosage , Acetylgalactosamine/administration & dosage , Acetylgalactosamine/pharmacokinetics , Acetylgalactosamine/pharmacology , Adult , Dose-Response Relationship, Drug , Drug Administration Schedule , Female , Half-Life , Humans , Injections, Subcutaneous , Male , Middle Aged , Pyrrolidines/pharmacokinetics , Pyrrolidines/pharmacology , Young Adult
5.
N Engl J Med ; 380(6): 549-558, 2019 02 07.
Article in English | MEDLINE | ID: mdl-30726693

ABSTRACT

BACKGROUND: Induction of delta aminolevulinic acid synthase 1 ( ALAS1) gene expression and accumulation of neurotoxic intermediates result in neurovisceral attacks and disease manifestations in patients with acute intermittent porphyria, a rare inherited disease of heme biosynthesis. Givosiran is an investigational RNA interference therapeutic agent that inhibits hepatic ALAS1 synthesis. METHODS: We conducted a phase 1 trial of givosiran in patients with acute intermittent porphyria. In part A of the trial, patients without recent porphyria attacks (i.e., no attacks in the 6 months before baseline) were randomly assigned to receive a single subcutaneous injection of one of five ascending doses of givosiran (0.035, 0.10, 0.35, 1.0, or 2.5 mg per kilogram of body weight) or placebo. In part B, patients without recent attacks were randomly assigned to receive once-monthly injections of one of two doses of givosiran (0.35 or 1.0 mg per kilogram) or placebo (total of two injections 28 days apart). In part C, patients who had recurrent attacks were randomly assigned to receive injections of one of two doses of givosiran (2.5 or 5.0 mg per kilogram) or placebo once monthly (total of four injections) or once quarterly (total of two injections) during a 12-week period, starting on day 0. Safety, pharmacokinetic, pharmacodynamic, and exploratory efficacy outcomes were evaluated. RESULTS: A total of 23 patients in parts A and B and 17 patients in part C underwent randomization. Common adverse events included nasopharyngitis, abdominal pain, and diarrhea. Serious adverse events occurred in 6 patients who received givosiran in parts A through C combined. In part C, all 6 patients who were assigned to receive once-monthly injections of givosiran had sustained reductions in ALAS1 messenger RNA (mRNA), delta aminolevulinic acid, and porphobilinogen levels to near normal. These reductions were associated with a 79% lower mean annualized attack rate than that observed with placebo (exploratory efficacy end point). CONCLUSIONS: Once-monthly injections of givosiran in patients who had recurrent porphyria attacks resulted in mainly low-grade adverse events, reductions in induced ALAS1 mRNA levels, nearly normalized levels of the neurotoxic intermediates delta aminolevulinic acid and porphobilinogen, and a lower attack rate than that observed with placebo. (Funded by Alnylam Pharmaceuticals; ClinicalTrials.gov number, NCT02452372 .).


Subject(s)
5-Aminolevulinate Synthetase/antagonists & inhibitors , Amides/administration & dosage , Porphyria, Acute Intermittent/drug therapy , RNAi Therapeutics , 5-Aminolevulinate Synthetase/genetics , 5-Aminolevulinate Synthetase/metabolism , Acetylgalactosamine/analogs & derivatives , Adult , Amides/adverse effects , Dose-Response Relationship, Drug , Drug Administration Schedule , Female , Humans , Injections, Subcutaneous , Liver/metabolism , Male , Middle Aged , Molecular Targeted Therapy , Porphobilinogen/blood , Pyrrolidines , RNA, Messenger/metabolism , RNA, Messenger/urine
6.
J Heart Lung Transplant ; 35(2): 213-21, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26452996

ABSTRACT

BACKGROUND: Respiratory syncytial virus (RSV) infection in lung transplant (LTx) patients is associated with an increased incidence of bronchiolitis obliterans syndrome (BOS). ALN-RSV01 is a small interfering RNA targeting RSV replication that was shown in an earlier Phase 2a trial to be safe and to reduce the incidence of BOS when compared with placebo. METHODS: We performed a Phase 2b randomized, double-blind, placebo-controlled trial in RSV-infected LTx patients to examine the impact of ALN-RSV01 on the incidence of new or progressive BOS. Subjects were randomized (1:1) to receive aerosolized ALN-RSV01 or placebo daily for 5 days. RESULTS: Of 3,985 symptomatic patients screened, 218 were RSV-positive locally, of whom 87 were randomized to receive ALN-RSV01 or placebo (modified intention-to-treat [mITT] cohort). RSV infection was confirmed by central laboratory in 77 patients (ALN-RSV01, n = 44; placebo, n = 33), which comprised the primary analysis cohort (central mITT [mITTc]). ALN-RSV01 was found to be safe and well-tolerated. At Day 180, in ALN-RSV01-treated patients, compared with placebo, in the mITTc cohort there was a trend toward a decrease in new or progressive BOS (13.6% vs 30.3%, p = 0.058), which was significant in the per-protocol cohort (p = 0.025). Treatment effect was enhanced when ALN-RSV01 was started <5 days from symptom onset, and was observed even without ribavirin treatment. There was no significant impact on viral parameters or symptom scores. CONCLUSIONS: These results confirm findings of the earlier Phase 2a trial and provide further support that ALN-RSV01 reduces the risk of BOS after RSV in LTx recipients.


Subject(s)
Antiviral Agents/therapeutic use , Bronchiolitis Obliterans/prevention & control , Lung Transplantation , RNA, Small Interfering/therapeutic use , Respiratory Syncytial Virus Infections/complications , Adult , Bronchiolitis Obliterans/etiology , Double-Blind Method , Female , Humans , Male , Middle Aged , Postoperative Complications , Syndrome , Treatment Outcome
7.
Nat Med ; 21(5): 492-7, 2015 May.
Article in English | MEDLINE | ID: mdl-25849132

ABSTRACT

Hemophilia A and B are inherited bleeding disorders characterized by deficiencies in procoagulant factor VIII (FVIII) or factor IX (FIX), respectively. There remains a substantial unmet medical need in hemophilia, especially in patients with inhibitory antibodies against replacement factor therapy, for novel and improved therapeutic agents that can be used prophylactically to provide effective hemostasis. Guided by reports suggesting that co-inheritance of prothrombotic mutations may ameliorate the clinical phenotype in hemophilia, we developed an RNA interference (RNAi) therapeutic (ALN-AT3) targeting antithrombin (AT) as a means to promote hemostasis in hemophilia. When administered subcutaneously, ALN-AT3 showed potent, dose-dependent, and durable reduction of AT levels in wild-type mice, mice with hemophilia A, and nonhuman primates (NHPs). In NHPs, a 50% reduction in AT levels was achieved with weekly dosing at approximately 0.125 mg/kg, and a near-complete reduction in AT levels was achieved with weekly dosing at 1.5 mg/kg. Treatment with ALN-AT3 promoted hemostasis in mouse models of hemophilia and led to improved thrombin generation in an NHP model of hemophilia A with anti-factor VIII inhibitors. This investigational compound is currently in phase 1 clinical testing in subjects with hemophilia A or B.


Subject(s)
Antithrombins/chemistry , Blood Coagulation/drug effects , Factor IX/chemistry , Factor VIII/chemistry , Hemophilia A/drug therapy , RNA Interference , Animals , Dose-Response Relationship, Drug , Female , Hemophilia A/genetics , Hemostasis/drug effects , Homozygote , Humans , Male , Mice , Mutation
9.
Proc Natl Acad Sci U S A ; 106(11): 4372-7, 2009 Mar 17.
Article in English | MEDLINE | ID: mdl-19246396

ABSTRACT

Systemic inflammation arising from the organismal distribution of pathogen-associated molecular patterns is a major cause of clinical morbidity and mortality. Herein we report a critical and previously unrecognized in vivo role for germinal center kinase (GCK, genome nomenclature: map4k2), a mammalian Sterile 20 (STE20) orthologue, in PAMP signaling, and systemic inflammation. We find that disruption of gck in mice strongly impairs PAMP-stimulated macrophage cytokine and chemokine release and renders mice resistant to endotoxin-mediated lethality. Bone marrow transplantation studies show that hematopoietic cell GCK signaling is essential to systemic inflammation. Disruption of gck substantially reduces PAMP activation of macrophage Jun-N-terminal kinase (JNK) and p38 mitogen-activated protein kinases (MAPKs) via reduced activation of the MAPK-kinase-kinases (MAP3Ks) mixed lineage kinases (MLKs)-2 and -3. Extracellular signal-regulated kinase (ERK) and nuclear factor-kappaB (NF-kappaB) activation are largely unaffected. Thus, GCK is an essential PAMP effector coupling JNK and p38, but not ERK or NF-kappaB to systemic inflammation.


Subject(s)
Inflammation/etiology , JNK Mitogen-Activated Protein Kinases/metabolism , Protein Serine-Threonine Kinases/physiology , Adrenomedullin/pharmacology , Animals , Bone Marrow Transplantation , Cytokines/metabolism , Extracellular Signal-Regulated MAP Kinases , Germinal Center Kinases , Hematopoietic Stem Cells/metabolism , Macrophages/metabolism , Mice , Mice, Knockout , NF-kappa B , Protein Serine-Threonine Kinases/deficiency , Receptors, Pattern Recognition/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
10.
Am J Physiol Lung Cell Mol Physiol ; 294(4): L698-704, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18310224

ABSTRACT

The signal transducers and activators of transcription (STAT) family of transcription factors regulates a variety of biological functions including cellular proliferation, transformation, apoptosis, and differentiation. We have previously determined that PDGF activates the STAT pathway in human airway smooth muscle cells (HASMC) and that the Jak and Src kinases are required for both PDGF-induced STAT activation and HASMC proliferation. As increased airway smooth muscle (ASM) volume is associated with airflow obstruction and disease severity in patients with asthma, it is important to elucidate the cellular and molecular pathways that regulate ASM accumulation. In this paper, we investigated the requirement of STAT3 for PDGF-induced HASMC proliferation. We demonstrate that knockdown of STAT3 expression in HASMC resulted in a significant decrease in mitogen-induced cellular proliferation. Additionally, PDGF-induced activation of STAT3 required the small GTP-binding protein Rac1, and Rac1 was also required for PDGF-induced HASMC proliferation. Furthermore, PDGF treatment induced STAT3 and Rac1 to physically associate and translocate to the nucleus, identifying one mechanism by which STAT3 is regulated in response to PDGF in HASMC. Finally, we determined that STAT3 expression is required for PDGF-mediated regulation of cell cycle targets cyclin D3 and p27. These data define a novel mitogenic signaling pathway in airway smooth muscle cells leading from PDGF to Rac1 and STAT3 and subsequent cell cycle gene regulation. Thus, targeting STAT3 may prove to be a novel therapeutic approach for patients with severe asthma and significant airway wall remodeling, as manifested by ASM accumulation.


Subject(s)
Muscle, Smooth/physiology , Platelet-Derived Growth Factor/pharmacology , Respiratory System/drug effects , STAT3 Transcription Factor/metabolism , rac1 GTP-Binding Protein/metabolism , Cell Division/drug effects , Cells, Cultured , DNA Primers , Humans , Muscle, Smooth/cytology , Muscle, Smooth/drug effects , Polymerase Chain Reaction , RNA, Small Interfering/genetics , Respiratory System/cytology , STAT3 Transcription Factor/genetics , Signal Transduction , Thymidine/metabolism , Trachea , Transfection , rac1 GTP-Binding Protein/genetics
11.
J Immunol ; 178(10): 6191-9, 2007 May 15.
Article in English | MEDLINE | ID: mdl-17475846

ABSTRACT

The STAT3 transcription factor is critical for cytokine signaling and the acute phase response, but its role in allergic asthma is largely undefined. To investigate the role of STAT3 in mediating allergic inflammation, we used chemical and genetic approaches to inactivate STAT3 in the airway epithelium of mice. In a murine model of chronic asthma, we demonstrate that the administration of house dust mite (HDM) leads to robust STAT3 activation in the airway epithelium, smooth muscle, and immune cells in the lungs of C57BL/6 mice. To investigate the role of STAT3 in HDM-induced airway inflammation, a conditional knockout of STAT3 in the airway epithelium was generated, e-STAT3-/-. We determined that e-STAT3-/- mice had a significant decrease in HDM-induced airway eosinophilia, lung Th2 accumulation, and chemokines compared with wild-type animals. Importantly, the e-STAT3-/- mice had a significant decrease in airway hyperresponsiveness to methacholine. The administration of two STAT kinase inhibitors diminished STAT3 activation and markedly abrogated the HDM-induced lung inflammation. These findings suggest that STAT3 acts as a novel epithelial regulator of the allergic response by altering Th2 cell recruitment and effector function, and thus, targeting this molecule may provide the basis for a novel asthma therapy.


Subject(s)
Allergens/immunology , Asthma/metabolism , Asthma/pathology , Respiratory Mucosa/metabolism , Respiratory Mucosa/pathology , STAT3 Transcription Factor/physiology , Animals , Asthma/immunology , Cell Movement/immunology , Chronic Disease , Disease Models, Animal , Dust/immunology , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Mites/immunology , Respiratory Mucosa/immunology , STAT3 Transcription Factor/deficiency , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Th2 Cells/immunology , Th2 Cells/metabolism , Th2 Cells/pathology
12.
Cell Biochem Biophys ; 42(3): 263-76, 2005.
Article in English | MEDLINE | ID: mdl-15976459

ABSTRACT

Serotonin (5-HT) stimulates superoxide release, phosphorylation of p42/p44 mitogen-activated protein kinase (MAPK), and DNA synthesis in bovine pulmonary artery smooth muscle cells. Both p42/p44 MAPK and reactive oxygen species (ROS) generation are required for 5-HT-induced growth in SMC. Agents that block the production of ROS, or ROS scavengers, block MAPK activation by 5-HT. However, specific signal transduction by 5-HT leading to proteins that control entrance into the cell cycle are not well defined in smooth muscle cells. Here, we show by Western blot that 5-HT upregulates c-Fos, an immediate early gene product known to regulate the entrance of quiescent cells into the cell cycle. Northern blots showed that c-fos mRNA is induced by 5-HT in 30 min. This induction is blocked by PD98059, indicating that activation of MAPK is required. 5-HT-induced expression of a 350 bp c-fos promoter in a luciferase reporter is blocked by PD98059 and diphenyliodonium (DPI). The GTPases Rac1 and Ras have been implicated in growth factor-induced generation of ROS. Overexpression of either dominant negative (DN) Rac1 or DN Ras inhibited 5-HT-mediated c-fos promoter activation. 5-HT also induced expression from a truncated c-fos promoter containing an isolated serum response element. This activation was blocked by DPI and PD98059. Overexpression of activated Ras and Rac1 were additive for activation of the serum response element promoter. Regulation of cyclin D1, a protein shown to be regulated by c-fos and required for entry into the cell cycle, is upregulated by 5-HT and is blocked by DPI and PD98059. Nuclear factor-kappaB, which can also regulate cyclin D1, was not activated. We conclude that 5-HT stimulates c-fos and cyclin D1 expression through a ROS-dependent mechanism that requires Ras, Rac1, and MAPK.


Subject(s)
Gene Expression Regulation/physiology , Myocytes, Smooth Muscle/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Reactive Oxygen Species/metabolism , Serotonin/metabolism , rac1 GTP-Binding Protein/metabolism , ras GTPase-Activating Proteins/metabolism , Animals , Cattle , Cells, Cultured , Signal Transduction/physiology
13.
Am J Respir Crit Care Med ; 171(8): 858-67, 2005 Apr 15.
Article in English | MEDLINE | ID: mdl-15665321

ABSTRACT

The cascade of cellular and molecular pathways mediating acute lung injury is complex and incompletely defined. Although the Src and Jak family of kinases is upregulated in LPS-induced murine lung injury, their role in the development of lung injury is unknown. Here we report that systemic inhibition of these kinases using specific small molecule inhibitors (PP2, SU6656, tyrphostin A1) significantly attenuated LPS-induced lung injury, as determined by histologic and capillary permeability assays. These inhibitors blocked LPS-dependent cytokine and chemokine production in the lung and in the serum. In contrast, lung-targeted inhibition of these kinases in the airway epithelium via adenoviral-mediated gene transfer of dominant negative Src or of suppressor of cytokine signaling (SOCS-1) disrupted lung cytokine production but had no effect on systemic cytokine production or lung vascular permeability. Mice were significantly protected from lethal LPS challenge by the small molecule inhibitors of Jak and Src kinase. Importantly, this protection was still evident even when the inhibitors were administered 6 hours after LPS challenge. Taken together, these observations suggest that Jak and Src kinases participate in acute lung injury and verify the potential of this class of selective tyrosine kinase inhibitors to serve as novel therapeutic agents for this disease.


Subject(s)
Enzyme Inhibitors/pharmacology , Lipopolysaccharides/immunology , Protein-Tyrosine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins/antagonists & inhibitors , Respiratory Distress Syndrome/immunology , src-Family Kinases/antagonists & inhibitors , Adenoviridae/genetics , Animals , Capillary Leak Syndrome/genetics , Capillary Leak Syndrome/immunology , Enzyme Activation/drug effects , Enzyme Activation/genetics , Enzyme Activation/immunology , Escherichia coli , Gene Expression Regulation/drug effects , Gene Transfer Techniques , Indoles/pharmacology , Janus Kinase 2 , Lung/drug effects , Lung/immunology , Lung/pathology , Mice , Mice, Inbred BALB C , Protein-Tyrosine Kinases/genetics , Proto-Oncogene Proteins/genetics , Respiratory Distress Syndrome/pathology , Signal Transduction/drug effects , Signal Transduction/genetics , Sulfonamides/pharmacology , Transcriptional Activation/immunology , Tyrphostins/pharmacology , src-Family Kinases/genetics
14.
Am J Physiol Lung Cell Mol Physiol ; 286(6): L1282-92, 2004 Jun.
Article in English | MEDLINE | ID: mdl-14729509

ABSTRACT

Acute lung injury (ALI) is a devastating clinical problem with a mortality as high as 60%. It is now appreciated that ALI represents a cytokine excess state that involves the microvasculature of multiple organs. The signal transducers and activators of transcription (STAT) family of transcription factors activate critical mediators of cytokine responses, but there is limited knowledge about their role in mediating ALI. In the present study, we demonstrate that the STAT transcription factors are activated rapidly in the lungs after intraperitoneal and intranasal LPS administration in mice. We also demonstrated that LPS activates both the STAT kinases, Src and JAK, in the lung with kinetics that are consistent with STAT activation. LPS treatment resulted in STAT3 activation throughout the resident lung cells, as well as in the recruited inflammatory cells. Whereas direct LPS treatment did not lead to STAT activation in cultured epithelial or endothelial cells, IL-6 activated STAT3 in both of these cell types. Furthermore, IL-6 was induced by LPS in serum and in the lung with kinetics consistent with STAT3 activation, suggesting that IL-6 may be one mechanism of STAT activation by LPS. In addition, STAT activation required reactive oxygen species, as the overexpression of catalase in mice prevented LPS-mediated STAT activation in the lung. STATs may be a common pathway for mediating ALI, regardless of the inciting factor, as STAT activation also occurred in both a gastric acid aspiration and acute pancreatitis model of ALI. Finally, STATs are activated in the lung long before signs of ALI are present, suggesting that the STAT transcription factors may play a role in initiating the inflammatory response seen in the lung.


Subject(s)
DNA-Binding Proteins/metabolism , Proto-Oncogene Proteins , Respiratory Distress Syndrome/metabolism , Trans-Activators/metabolism , Acute Disease , Animals , Cells, Cultured , Disease Models, Animal , Hydrochloric Acid , Interleukin-6/blood , Janus Kinase 2 , Kinetics , Lipopolysaccharides , Liver/metabolism , Lung/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mitogen-Activated Protein Kinases/metabolism , Oxidation-Reduction , Pancreatitis/complications , Pancreatitis/metabolism , Protein-Tyrosine Kinases/metabolism , Respiratory Distress Syndrome/chemically induced , Respiratory Mucosa/cytology , Respiratory Mucosa/metabolism , STAT3 Transcription Factor , Tumor Necrosis Factor-alpha/metabolism , src-Family Kinases/metabolism
15.
Mol Pharmacol ; 63(2): 368-77, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12527808

ABSTRACT

Anthracyclines are effective cancer chemotherapeutic agents but can induce serious cardiotoxicity. Understanding the mechanism of cardiac damage by these agents will help in development of better therapeutic strategies against cancer. The GATA-4 transcription factor is an important regulator of cardiac muscle cells. The present study demonstrates that anthracyclines can down-regulate GATA-4 activity. Treatment of HL-1 cardiac muscle cells or isolated adult rat ventricular myocytes with anthracyclines such as daunorubicin and doxorubicin decreased the level of GATA-4 DNA-binding activity. The mechanism of decreased GATA-4 activity acts at the level of the GATA-4 gene, because anthracyclines caused significantly decreased levels of GATA-4 protein and mRNA. The rate of decline in GATA-4 transcript levels in the presence of actinomycin D was unaltered by anthracyclines, indicating that these agents may affect directly GATA-4 gene transcription. To determine whether decreased GATA-4 levels are functionally related to cardiac muscle cell death that can be induced by anthracyclines, the ability of ectopic GATA factors to rescue anthracycline-induced apoptosis was tested. Adenovirus-mediated expression of either GATA-4 or GATA-6 was sufficient to attenuate the incidence of apoptosis. Furthermore, suppression of GATA-4 DNA-binding activity by a dominant negative mutant of GATA-4 induced the apoptosis. These results suggest that the mechanism of anthracycline-induced cardiotoxicity may involve the down-regulation of GATA-4 and the induction of apoptosis.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Apoptosis , DNA-Binding Proteins/metabolism , Gene Expression/drug effects , Heart/drug effects , Myocardium/cytology , Transcription Factors/metabolism , Animals , DNA/drug effects , DNA/metabolism , DNA-Binding Proteins/drug effects , DNA-Binding Proteins/genetics , Down-Regulation , GATA4 Transcription Factor , Mice , Myocardium/metabolism , Oxidants/pharmacology , Transcription Factors/drug effects , Transcription Factors/genetics
16.
Am J Physiol Lung Cell Mol Physiol ; 282(6): L1296-304, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12003786

ABSTRACT

Airway remodeling, as manifested by an increase in airway smooth muscle mass, mucous gland hyperplasia, and subepithelial fibrosis, contributes to the airway hyperresponsiveness and fixed obstruction seen in some asthmatic patients. Here we investigated whether the Janus kinase-signal transducer and activator of transcription (JAK-STAT) pathway contributes to platelet-derived growth factor (PDGF)-stimulated mitogenesis of human airway smooth muscle cells (HASMC). PDGF treatment of quiescent HASMC resulted in the rapid tyrosine phosphorylation and DNA binding of STAT1 and STAT3. This phosphorylation was blocked by inhibition of Src and JAK2 kinases. In addition, STAT activation by PDGF was found to be redox dependent. Moreover, PDGF-induced thymidine uptake was completely blocked by pretreatment of HASMC with the STAT kinase inhibitors AG-490, SU-6656, and PP2. Interestingly, the JAK pathway was required for HASMC mitogenesis independently of mitogen-activated protein kinase activation. Inhibition of the Src and JAK kinases blocked PDGF-stimulated gene expression of the STAT target genes cyclin D1 and c-myc. These results indicate that the JAK-STAT pathway contributes to PDGF-induced mitogenesis, and thus this pathway may be important in the airway remodeling seen in some asthmatic patients.


Subject(s)
DNA-Binding Proteins/metabolism , Intracellular Signaling Peptides and Proteins , Muscle, Smooth/metabolism , Platelet-Derived Growth Factor/pharmacology , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins , Repressor Proteins , Signal Transduction/physiology , Trans-Activators/metabolism , Blotting, Northern , Carrier Proteins/metabolism , Cells, Cultured , Cyclin D1/biosynthesis , Cyclin D1/genetics , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Humans , Janus Kinase 2 , Muscle, Smooth/cytology , Muscle, Smooth/drug effects , Proto-Oncogene Proteins c-myc/biosynthesis , Proto-Oncogene Proteins c-myc/genetics , RNA, Messenger/metabolism , Reactive Oxygen Species/metabolism , STAT3 Transcription Factor , Signal Transduction/drug effects , Suppressor of Cytokine Signaling 1 Protein , Suppressor of Cytokine Signaling Proteins , Trachea/cytology , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...