Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Sci Transl Med ; 10(427)2018 02 07.
Article in English | MEDLINE | ID: mdl-29437146

ABSTRACT

There are no approved drugs for the treatment of heart failure with preserved ejection fraction (HFpEF), which is characterized by left ventricular (LV) diastolic dysfunction. We demonstrate that ITF2357 (givinostat), a clinical-stage inhibitor of histone deacetylase (HDAC) catalytic activity, is efficacious in two distinct murine models of diastolic dysfunction with preserved EF. ITF2357 blocked LV diastolic dysfunction due to hypertension in Dahl salt-sensitive (DSS) rats and suppressed aging-induced diastolic dysfunction in normotensive mice. HDAC inhibitor-mediated efficacy was not due to lowering blood pressure or inhibiting cellular and molecular events commonly associated with diastolic dysfunction, including cardiac fibrosis, cardiac hypertrophy, or changes in cardiac titin and myosin isoform expression. Instead, ex vivo studies revealed impairment of cardiac myofibril relaxation as a previously unrecognized, myocyte-autonomous mechanism for diastolic dysfunction, which can be ameliorated by HDAC inhibition. Translating these findings to humans, cardiac myofibrils from patients with diastolic dysfunction and preserved EF also exhibited compromised relaxation. These data suggest that agents such as HDAC inhibitors, which potentiate cardiac myofibril relaxation, hold promise for the treatment of HFpEF in humans.


Subject(s)
Blood Pressure/physiology , Histone Deacetylases/metabolism , Animals , Blood Pressure/genetics , Connectin/metabolism , Female , Heart Failure , Hemodynamics/physiology , Histone Deacetylase Inhibitors/therapeutic use , Humans , Hydroxamic Acids/therapeutic use , Mice , Myocardium/metabolism , Myocytes, Cardiac , Myosins/metabolism , Rats , Rats, Sprague-Dawley , Ventricular Dysfunction, Left/metabolism
2.
Proc Natl Acad Sci U S A ; 111(3): 1055-9, 2014 Jan 21.
Article in English | MEDLINE | ID: mdl-24395784

ABSTRACT

Type 1 diabetes is due to destruction of pancreatic ß-cells. Lysine deacetylase inhibitors (KDACi) protect ß-cells from inflammatory destruction in vitro and are promising immunomodulators. Here we demonstrate that the clinically well-tolerated KDACi vorinostat and givinostat revert diabetes in the nonobese diabetic (NOD) mouse model of type 1 diabetes and counteract inflammatory target cell damage by a mechanism of action consistent with transcription factor--rather than global chromatin--hyperacetylation. Weaning NOD mice received low doses of vorinostat and givinostat in their drinking water until 100-120 d of age. Diabetes incidence was reduced by 38% and 45%, respectively, there was a 15% increase in the percentage of islets without infiltration, and pancreatic insulin content increased by 200%. Vorinostat treatment increased the frequency of functional regulatory T-cell subsets and their transcription factors Gata3 and FoxP3 in parallel to a decrease in inflammatory dendritic cell subsets and their cytokines IL-6, IL-12, and TNF-α. KDACi also inhibited LPS-induced Cox-2 expression in peritoneal macrophages from C57BL/6 and NOD mice. In insulin-producing ß-cells, givinostat did not upregulate expression of the anti-inflammatory genes Socs1-3 or sirtuin-1 but reduced levels of IL-1ß + IFN-γ-induced proinflammatory Il1a, Il1b, Tnfα, Fas, Cxcl2, and reduced cytokine-induced ERK phosphorylation. Further, NF-κB genomic iNos promoter binding was reduced by 50%, and NF-κB-dependent mRNA expression was blocked. These effects were associated with NF-κB subunit p65 hyperacetylation. Taken together, these data provide a rationale for clinical trials of safety and efficacy of KDACi in patients with autoimmune disease such as type 1 diabetes.


Subject(s)
Chromatin/metabolism , Diabetes Mellitus, Type 1/metabolism , Histone Deacetylase Inhibitors/pharmacology , Insulin-Secreting Cells/cytology , Animals , Cell Line , Cytokines/metabolism , Disease Models, Animal , Epigenesis, Genetic , Female , GATA3 Transcription Factor/metabolism , Histone Deacetylases/metabolism , Humans , Hydroxamic Acids/pharmacology , Inflammation , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Phosphorylation , Promoter Regions, Genetic , Protein Binding , Protein Processing, Post-Translational , Rats , Time Factors , Vorinostat
3.
Mol Med ; 19: 79-87, 2013 May 20.
Article in English | MEDLINE | ID: mdl-23552722

ABSTRACT

Previous work has established the existence of dystrophin-nitric oxide (NO) signaling to histone deacetylases (HDACs) that is deregulated in dystrophic muscles. As such, pharmacological interventions that target HDACs (that is, HDAC inhibitors) are of potential therapeutic interest for the treatment of muscular dystrophies. In this study, we explored the effectiveness of long-term treatment with different doses of the HDAC inhibitor givinostat in mdx mice--the mouse model of Duchenne muscular dystrophy (DMD). This study identified an efficacy for recovering functional and histological parameters within a window between 5 and 10 mg/kg/d of givinostat, with evident reduction of the beneficial effects with 1 mg/kg/d dosage. The long-term (3.5 months) exposure of 1.5-month-old mdx mice to optimal concentrations of givinostat promoted the formation of muscles with increased cross-sectional area and reduced fibrotic scars and fatty infiltration, leading to an overall improvement of endurance performance in treadmill tests and increased membrane stability. Interestingly, a reduced inflammatory infiltrate was observed in muscles of mdx mice exposed to 5 and 10 mg/kg/d of givinostat. A parallel pharmacokinetic/pharmacodynamic analysis confirmed the relationship between the effective doses of givinostat and the drug distribution in muscles and blood of treated mice. These findings provide the preclinical basis for an immediate translation of givinostat into clinical studies with DMD patients.


Subject(s)
Carbamates/therapeutic use , Histone Deacetylase Inhibitors/therapeutic use , Muscular Dystrophy, Duchenne/drug therapy , Animals , Carbamates/pharmacology , Cells, Cultured , Exercise Test , Fibrosis/drug therapy , Fibrosis/pathology , Histone Deacetylase Inhibitors/pharmacology , Humans , Mice , Mice, Inbred mdx , Muscle, Skeletal/drug effects , Muscle, Skeletal/pathology , Muscle, Skeletal/physiopathology , Muscular Dystrophy, Duchenne/pathology , Muscular Dystrophy, Duchenne/physiopathology , Myoblasts/cytology , Myoblasts/drug effects , Running
4.
Mol Med ; 17(5-6): 353-62, 2011.
Article in English | MEDLINE | ID: mdl-21365126

ABSTRACT

ITF2357 (givinostat) is a histone deacetylase inhibitor with antiinflammatory properties at low nanomolar concentrations. We report here a phase I safety and pharmacokinetics trial in healthy males administered 50, 100, 200, 400 or 600 mg orally. After 50 mg, mean maximal plasma concentrations reached 104 nmol/L 2 h after dosing, with a half-life of 6.9 h. After 100 mg, maximal concentration reached 199 nmol/L at 2.1 h with a half-life of 6.0 h. Repeat doses for 7 consecutive days of 50, 100 or 200 mg resulted in nearly the same kinetics. There were no serious adverse effects (AEs) and no organ toxicities. However, there was a dose-dependent but transient fall in platelets. After 7 daily doses of 50 or 100 mg, the mean decrease in platelets of 17 and 25% was not statistically significant and returned to baseline within 14 d. Blood removed from the subjects after oral dosing was cultured ex vivo with endotoxin, and the release of tumor necrosis factor (TNF)-α, interleukin (IL)-1ß, IL-6, IL-1Ra, interferon (IFN)-γ and IL-10 was determined. Maximal reduction in IL-1ß, TNFα, IL-6 and IFNγ was observed 4 h after dosing but returned to baseline at 12 h. There was no significant reduction in IL-1Ra or IL-10. With daily dosing, the fall in cytokine production in blood cultures observed on day 7 was nearly the same as that of the first day. We conclude that dosing of 50 or 100 mg ITF2357 is safe in healthy humans and transiently but repeatedly reduces the production of proinflammatory cytokines without affecting production of antiinflammatory cytokines.


Subject(s)
Anti-Inflammatory Agents/adverse effects , Anti-Inflammatory Agents/pharmacokinetics , Cytokines/blood , Hydroxamic Acids/adverse effects , Hydroxamic Acids/pharmacokinetics , Adult , Anti-Inflammatory Agents/administration & dosage , Female , Humans , Hydroxamic Acids/administration & dosage , Interferon-gamma/blood , Interleukin 1 Receptor Antagonist Protein/blood , Interleukin-10/blood , Interleukin-1beta/blood , Interleukin-6/blood , Male , Tumor Necrosis Factor-alpha/blood , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...