Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
J Neurosci ; 37(28): 6761-6777, 2017 07 12.
Article in English | MEDLINE | ID: mdl-28592699

ABSTRACT

Ca2+-influx through L-type Ca2+-channels (LTCCs) is associated with activity-related stressful oscillations of Ca2+ levels within dopaminergic (DA) neurons in the substantia nigra (SN), which may contribute to their selective degeneration in Parkinson's disease (PD). LTCC blockers were neuroprotective in mouse neurotoxin models of PD, and isradipine is currently undergoing testing in a phase III clinical trial in early PD. We report no evidence for neuroprotection by in vivo pretreatment with therapeutically relevant isradipine plasma levels, or Cav1.3 LTCC deficiency in 6-OHDA-treated male mice. To explain this finding, we investigated the pharmacological properties of human LTCCs during SN DA-like and arterial smooth muscle (aSM)-like activity patterns using whole-cell patch-clamp recordings in HEK293 cells (Cav1.2 α1-subunit, long and short Cav1.3 α1-subunit splice variants; ß3/α2δ1). During SN DA-like pacemaking, only Cav1.3 variants conducted Ca2+ current (ICa) at subthreshold potentials between action potentials. SN DA-like burst activity increased integrated ICa during (Cav1.2 plus Cav1.3) and after (Cav1.3) the burst. Isradipine inhibition was splice variant and isoform dependent, with a 5- to 11-fold lower sensitivity to Cav1.3 variants during SN DA-like pacemaking compared with Cav1.2 during aSM-like activity. Supratherapeutic isradipine concentrations reduced the pacemaker precision of adult mouse SN DA neurons but did not affect their somatic Ca2+ oscillations. Our data predict that Cav1.2 and Cav1.3 splice variants contribute differentially to Ca2+ load in SN DA neurons, with prominent Cav1.3-mediated ICa between action potentials and after bursts. The failure of therapeutically relevant isradipine levels to protect SN DA neurons can be explained by weaker state-dependent inhibition of SN DA LTCCs compared with aSM Cav1.2.SIGNIFICANCE STATEMENT The high vulnerability of dopamine (DA) neurons in the substantia nigra (SN) to neurodegenerative stressors causes Parkinson's disease (PD). Ca2+ influx through voltage-gated L-type Ca2+ channels (LTCCs), in particular Cav1.3, appears to contribute to this vulnerability, and the LTCC inhibitor isradipine is currently being tested as a neuroprotective agent for PD in a phase III clinical trial. However, in our study isradipine plasma concentrations approved for therapy were not neuroprotective in a PD mouse model. We provide an explanation for this observation by demonstrating that during SN DA-like neuronal activity LTCCs are less sensitive to isradipine than Cav1.2 LTCCs in resistance blood vessels (mediating dose-limiting vasodilating effects) and even at supratherapeutic concentrations isradipine fails to reduce somatic Ca2+ oscillations of SN DA neurons.


Subject(s)
Calcium Channels, L-Type/metabolism , Calcium Signaling/physiology , Dopamine/metabolism , Dopaminergic Neurons/physiology , Isradipine/metabolism , Substantia Nigra/physiology , Animals , Calcium Channel Blockers/pharmacology , Calcium Signaling/drug effects , Dopaminergic Neurons/drug effects , Ion Channel Gating/drug effects , Ion Channel Gating/physiology , Isradipine/pharmacology , Male , Mice , Mice, Inbred C57BL , Neuroprotective Agents/pharmacology , Parkinson Disease/drug therapy , Parkinson Disease/physiopathology , Substantia Nigra/drug effects
2.
NPJ Parkinsons Dis ; 9(1): 91, 2023 Jun 15.
Article in English | MEDLINE | ID: mdl-37322068

ABSTRACT

Prion-like transmission of pathology in α-synucleinopathies like Parkinson's disease or multiple system atrophy is increasingly recognized as one potential mechanism to address disease progression. Active and passive immunotherapies targeting insoluble, aggregated α-synuclein are already being actively explored in the clinic with mixed outcomes so far. Here, we report the identification of 306C7B3, a highly selective, aggregate-specific α-synuclein antibody with picomolar affinity devoid of binding to the monomeric, physiologic protein. 306C7B3 binding is Ser129-phosphorylation independent and shows high affinity to several different aggregated α-synuclein polymorphs, increasing the likelihood that it can also bind to the pathological seeds assumed to drive disease progression in patients. In support of this, highly selective binding to pathological aggregates in postmortem brains of MSA patients was demonstrated, with no staining in samples from other human neurodegenerative diseases. To achieve CNS exposure of 306C7B3, an adeno-associated virus (AAV) based approach driving expression of the secreted antibody within the brain of (Thy-1)-[A30P]-hα-synuclein mice was used. Widespread central transduction after intrastriatal inoculation was ensured by using the AAV2HBKO serotype, with transduction being spread to areas far away from the inoculation site. Treatment of (Thy-1)-[A30P]-hα-synuclein mice at the age of 12 months demonstrated significantly increased survival, with 306C7B3 concentration reaching 3.9 nM in the cerebrospinal fluid. These results suggest that AAV-mediated expression of 306C7B3, targeting extracellular, presumably disease-propagating aggregates of α-synuclein, has great potential as a disease-modifying therapy for α-synucleinopathies as it ensures CNS exposure of the antibody, thereby mitigating the selective permeability of the blood-brain barrier.

3.
Chembiochem ; 12(18): 2740-4, 2011 Dec 16.
Article in English | MEDLINE | ID: mdl-22162214

ABSTRACT

Gas-phase protein separation by ion mobility: With its ability to separate the Parkinson's disease protein α-synuclein and its autoproteolytic products-despite the small concentrations of the latter-ion-mobility MS has enabled the characterization of intermediate fragments in in vitro oligomerization-aggregation. In particular, a possible key fragment, the highly aggregating C-terminal fragment, αSyn(72-140), has been revealed.


Subject(s)
Biopolymers/metabolism , Mass Spectrometry/methods , Parkinson Disease/metabolism , alpha-Synuclein/metabolism , Chromatography, High Pressure Liquid , Electrophoresis, Polyacrylamide Gel , Humans , Proteolysis , Spectrometry, Mass, Electrospray Ionization , Tandem Mass Spectrometry
4.
Transl Vis Sci Technol ; 10(11): 15, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34520511

ABSTRACT

Purpose: Retinopathies display complex pathologies, including vasculopathies, inflammation, and fibrosis, leading ultimately to visual impairment. However, animal models accurately reflecting these pathologies are lacking. In this study, we evaluate the suitability of using Adeno-associated virus (AAV)-mediated long-term expression of cytokines to establish retinal pathology in the murine retina. Methods: We administered recombinant, Müller-glia targeted AAV-ShH10 into the mouse vitreous to induce retinal expression of either human vascular endothelial growth factor (VEGF)-A165, tumor necrosis factor alpha (TNF-α), or interleukin-6 (IL-6) and evaluated consequent effects by optical coherence tomography, fluorescein angiography, and histology. Results: Intravitreal injection of AAVs resulted in rapid and stable expression of the transgenes within 1 to 6 weeks. Akin to the role of VEGF-A in wet age-related macular degeneration, expression of VEGF-A led to several vasculopathies in mice, including neovascularization and vascular leakage. In contrast, the expression of the proinflammatory cytokines TNF-α or IL-6 induced retinal inflammation, as indicated by microglial activation. Furthermore, the expression of TNF-α, but not of IL-6, induced immune cell infiltration into the vitreous as well as vasculitis, and subsequently induced the development of fibrosis and epiretinal membranes. Conclusions: In summary, the long-term expression of human VEGF-A165, TNF-α, or IL-6 in the mouse eye induced specific pathologies within 6 weeks that mimic different aspects of human retinopathies. Translational Relevance: AAV-mediated expression of human genes in mice is an attractive approach to provide valuable insights into the underlying molecular mechanisms causing retinopathies and is easily adaptable to other genes and preclinical species supporting drug discovery for retinal diseases.


Subject(s)
Tumor Necrosis Factor-alpha , Vascular Endothelial Growth Factor A , Animals , Dependovirus/genetics , Humans , Interleukin-6/genetics , Mice , Retina , Tumor Necrosis Factor-alpha/genetics , Vascular Endothelial Growth Factor A/genetics
5.
Invest Ophthalmol Vis Sci ; 62(9): 39, 2021 07 01.
Article in English | MEDLINE | ID: mdl-34313720

ABSTRACT

Purpose: We aim to characterize the pathways required for autofluorescent granule (AFG) formation by RPE cells using cultured monolayers. Methods: We fed RPE monolayers in culture with a single pulse of photoreceptor outer segments (POS). After 24 hours the cells started accumulating AFGs that were comparable to lipofuscin in vivo. Using this model, we used a variety of light and electron microscopical techniques, flow cytometry and Western blot to analyze the formation of AFGs. We also generated a mutant RPE line lacking cathepsin D by gene editing. Results: AFGs seem to derive from incompletely digested POS-containing phagosomes and after 3 days are surrounded by a single membrane positive for lysosome markers. We show by various methods that lysosome-phagosome fusion is required for AFG formation, and that impairment of lysosomal pH or catalytic activity, particularly cathepsin D activity, enhances AF accumulation. Conclusions: We conclude that lysosomal dysfunction results in incomplete POS degradation and enhanced AFG accumulation.


Subject(s)
Lipofuscin/metabolism , Lysosomes/metabolism , Retinal Pigment Epithelium/metabolism , Rod Cell Outer Segment/metabolism , Animals , Blotting, Western , Cells, Cultured , Flow Cytometry , Humans , Models, Animal , Phagocytosis/physiology , Retinal Pigment Epithelium/cytology , Swine
6.
ACS Synth Biol ; 9(6): 1292-1305, 2020 06 19.
Article in English | MEDLINE | ID: mdl-32427483

ABSTRACT

Adeno-associated viral (AAV) vector-mediated gene therapy holds great potential for future medical applications. However, to facilitate safer and broader applicability and to enable patient-centric care, therapeutic protein expression should be controllable, ideally by an orally administered drug. The use of protein-based systems is considered rather undesirable, due to potential immunogenicity and the limited coding space of AAV. Ligand-dependent riboswitches, in contrast, are small and characterized by an attractive mode-of-action based on mRNA-self-cleavage, independent of coexpressed foreign protein. While a promising approach, switches available to date have only shown moderate potency in animals. In particular, ON-switches that induce transgene expression upon ligand administration so far have achieved rather disappointing results. Here we present the utilization of the previously described tetracycline-dependent ribozyme K19 for controlling AAV-mediated transgene expression in mice. Using this tool switch, we provide first proof for the feasibility of clinically desired key features, including multiorgan functionality, potent regulation (up to 15-fold induction), reversibility, and the possibility to fine-tune and repeatedly induce expression. The systematic assessment of ligand and reporter protein plasma levels further enabled the characterization of pharmacokinetic-pharmacodynamic relationships. Thus, our results strongly support future efforts to develop engineered riboswitches for applications in clinical gene therapy.


Subject(s)
Anti-Bacterial Agents/pharmacology , Dependovirus/genetics , Gene Expression/drug effects , Genetic Vectors/metabolism , RNA, Catalytic/metabolism , 3' Untranslated Regions , Animals , Aptamers, Nucleotide/genetics , Aptamers, Nucleotide/metabolism , Cell Line , Genetic Vectors/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Liver/metabolism , Lung/metabolism , Mice , RNA, Catalytic/genetics , Tetracycline/pharmacology
7.
Neuron ; 47(1): 57-69, 2005 Jul 07.
Article in English | MEDLINE | ID: mdl-15996548

ABSTRACT

During development of the retinocollicular projection in mouse, retinal axons initially overshoot their future termination zones (TZs) in the superior colliculus (SC). The formation of TZs is initiated by interstitial branching at topographically appropriate positions. Ephrin-As are expressed in a decreasing posterior-to-anterior gradient in the SC, and they suppress branching posterior to future TZs. Here we investigate the role of an EphA7 gradient in the SC, which has the reverse orientation to the ephrin-A gradient. We find that in EphA7 mutant mice the retinocollicular map is disrupted, with nasal and temporal axons forming additional or extended TZs, respectively. In vitro, retinal axons are repelled from growing on EphA7-containing stripes. Our data support the idea that EphA7 is involved in suppressing branching anterior to future TZs. These findings suggest that opposing ephrin-A and EphA gradients are required for the proper development of the retinocollicular projection.


Subject(s)
Brain Mapping , Ephrins/metabolism , Receptor, EphA7/metabolism , Superior Colliculi/metabolism , Superior Colliculi/physiology , Vision, Ocular/physiology , Visual Pathways/physiology , Animals , Axons/physiology , Histocytochemistry , In Situ Hybridization , Mice , Mice, Knockout , RNA/biosynthesis , RNA/genetics , Retina/cytology
8.
Neuron ; 39(3): 453-65, 2003 Jul 31.
Article in English | MEDLINE | ID: mdl-12895420

ABSTRACT

The mechanisms generating precise connections between specific thalamic nuclei and cortical areas remain poorly understood. Using axon tracing analysis of ephrin/Eph mutant mice, we provide in vivo evidence that Eph receptors in the thalamus and ephrins in the cortex control intra-areal topographic mapping of thalamocortical (TC) axons. In addition, we show that the same ephrin/Eph genes unexpectedly control the inter-areal specificity of TC projections through the early topographic sorting of TC axons in an intermediate target, the ventral telencephalon. Our results constitute the first identification of guidance cues involved in inter-areal specificity of TC projections and demonstrate that the same set of mapping labels is used differentially for the generation of topographic specificity of TC projections between and within individual cortical areas.


Subject(s)
Cerebral Cortex/metabolism , Ephrin-A4/genetics , Ephrin-A5/genetics , Receptor, EphA4/genetics , Receptor, EphA5/genetics , Thalamus/metabolism , Animals , Brain Mapping/methods , Cerebral Cortex/embryology , Cerebral Cortex/enzymology , Ephrin-A4/biosynthesis , Ephrin-A4/physiology , Ephrin-A5/biosynthesis , Ephrin-A5/physiology , Female , Ligands , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neural Pathways/embryology , Neural Pathways/enzymology , Neural Pathways/metabolism , Neural Pathways/physiology , Receptor, EphA4/biosynthesis , Receptor, EphA4/physiology , Receptor, EphA5/biosynthesis , Receptor, EphA5/physiology , Thalamus/embryology , Thalamus/enzymology
9.
J Neuropathol Exp Neurol ; 77(6): 443-453, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29718367

ABSTRACT

Increasing evidence suggests a relationship between oxidative stress and α-synuclein aggregation, the primary pathological hallmark of Parkinson disease (PD). However, a direct causal relationship has not yet been established in vivo in mouse models of PD. Superoxide dismutase 2 (SOD2) is rate limiting in the antioxidant machinery of the mitochondria and even its partial deficiency elevates oxidative stress in mice. Therefore, in order to investigate a possible interaction between oxidative stress and α-synuclein aggregation in vivo, a transgenic model of PD with haplodeficiency for SOD2 was generated on the basis of the well-characterized murine (Thy-1)-h[A30P]-α-synuclein transgenic line. In comparison with littermate controls with full SOD2 capacity, α-synuclein transgenic mice with partial SOD2 deficiency exhibited a significantly more advanced stage of synucleinopathy at 16 months, as demonstrated by higher median PK-PET blot scores (p < 0.01) and a greater amount of truncated α-synuclein in the insoluble fraction of homogenized brains (p < 0.05). These results show that compromising the capacity to scavenge free radicals can exacerbate α-synuclein aggregation, indicating that elevated levels of oxidative stress could modulate the progression of PD.


Subject(s)
Oxidative Stress , alpha-Synuclein/metabolism , Animals , Brain/pathology , Brain Chemistry/genetics , Free Radical Scavengers , Mice , Mice, Transgenic , Parkinson Disease, Secondary/genetics , Parkinson Disease, Secondary/pathology , Superoxide Dismutase/biosynthesis , Superoxide Dismutase/genetics , alpha-Synuclein/genetics
10.
Neurobiol Aging ; 66: 97-111, 2018 06.
Article in English | MEDLINE | ID: mdl-29550548

ABSTRACT

Mutations in leucine-rich repeat kinase 2 (LRRK2) age-dependently cause Parkinson's disease and are associated with several inflammatory diseases. So far, the potential role of LRRK2 expression in glial cells as mediators of neuroinflammation and the influence of aging have not been investigated in viral vector-based LRRK2 animal models. In this study, we compared the effect of striatal injection of high-capacity adenoviral vectors expressing either a kinase-overactive LRRK2 with the familial G2019S mutation or a kinase-inactive LRRK2 variant in young and old C57BL/6J mice. The intrinsic adenovirus tropism guided preferentially glial transduction, and the vector design led to stable expression for at least 6 months. In histopathological analysis, young mice expressing either LRRK2 variant presented with transient vacuolization of striatal white fiber tracts accompanied by accumulation of microglial cells and astrogliosis, but inflammation resolved without permanent damage. Old mice had a stronger and prolonged inflammatory reaction and experienced permanent damage in form of partial neuron loss after 3 months exclusively in case of LRRK2_G2019S expression. The autophagic receptor p62 accumulated in cells with high levels of either LRRK2 variant, even more so in old mice. We conclude that the aging mouse brain is more susceptible to LRRK2-associated pathology, and in this model, glial LRRK2 expression significantly contributed to neuroinflammation, ultimately causing neurodegeneration.


Subject(s)
Adenoviridae/genetics , Aging/genetics , Aging/pathology , Corpus Striatum/metabolism , Corpus Striatum/pathology , Gene Expression , Genetic Vectors/genetics , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/genetics , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/metabolism , Parkinson Disease/etiology , Parkinson Disease/genetics , Animals , Disease Models, Animal , Genetic Predisposition to Disease/genetics , Inflammation/etiology , Inflammation/genetics , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/administration & dosage , Male , Mice, Inbred C57BL , Mutation , Neuroglia/metabolism
11.
J Med Chem ; 50(18): 4405-18, 2007 Sep 06.
Article in English | MEDLINE | ID: mdl-17691763

ABSTRACT

Histone deacetylase (HDAC) inhibitors are considered to be drugs for targeted cancer therapy and second-generation HDIs are currently being tested in clinical trials. Here, we report on the synthesis and biological evaluation of a novel HDAC inhibitor scaffold with the hydroxamate Zn(2+) complexing headgroup, selected from the 2-aroylindol motif. Inhibition of nuclear extract HDAC and recombinant HDAC 1 as well as induction of histone H3K(9+14) hyperacetylation mediated by E-N-hydroxy-(2-aroylindole)acrylamides or E-N-hydroxy-(2-aroylbenzofuran)acrylamides were studied. Moreover, the cytotoxic activity, the effects on the cell cycle, and histone H3S(10) phosphorylation of selected compounds were determined. By use of a panel of 24 different human tumor cell lines, mean IC(50) values of the most potent analogues 6c and 7b were 0.75 and 0.65 microM, respectively. The novel compounds were shown to be no substrates of the P-glycoprotein drug transporter. Comparable to N(1)-hydroxy-N(8)-phenyloctanediamide "2 (SAHA)", cells in the S phase of the cell cycle are depleted, with partial arrest in G1 and G2/M and finally induction of massive apoptosis.


Subject(s)
Acrylamides/chemical synthesis , Antineoplastic Agents/chemical synthesis , Benzofurans/chemical synthesis , Histone Deacetylase Inhibitors , Hydroxamic Acids/chemical synthesis , Indoles/chemical synthesis , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Acetylation , Acrylamides/chemistry , Acrylamides/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Apoptosis , Benzofurans/chemistry , Benzofurans/pharmacology , Cell Cycle/drug effects , Cell Extracts , Cell Line, Tumor , Drug Design , Drug Resistance, Neoplasm , Drug Screening Assays, Antitumor , Histone Deacetylase 1 , Histone Deacetylases/chemistry , Histones/metabolism , Humans , Hydroxamic Acids/chemistry , Hydroxamic Acids/pharmacology , Indoles/chemistry , Indoles/pharmacology , Phosphorylation , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/chemistry , Stereoisomerism , Structure-Activity Relationship , Vorinostat
12.
J Am Soc Mass Spectrom ; 25(8): 1472-81, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24845351

ABSTRACT

Bioaffinity analysis using a variety of biosensors has become an established tool for detection and quantification of biomolecular interactions. Biosensors, however, are generally limited by the lack of chemical structure information of affinity-bound ligands. On-line bioaffinity-mass spectrometry using a surface-acoustic wave biosensor (SAW-MS) is a new combination providing the simultaneous affinity detection, quantification, and mass spectrometric structural characterization of ligands. We describe here an on-line SAW-MS combination for direct identification and affinity determination, using a new interface for MS of the affinity-isolated ligand eluate. Key element of the SAW-MS combination is a microfluidic interface that integrates affinity-isolation on a gold chip, in-situ sample concentration, and desalting with a microcolumn for MS of the ligand eluate from the biosensor. Suitable MS-acquisition software has been developed that provides coupling of the SAW-MS interface to a Bruker Daltonics ion trap-MS, FTICR-MS, and Waters Synapt-QTOF- MS systems. Applications are presented for mass spectrometric identifications and affinity (K(D)) determinations of the neurodegenerative polypeptides, ß-amyloid (Aß), and pathophysiological and physiological synucleins (α- and ß-synucleins), two key polypeptide systems for Alzheimer's disease and Parkinson's disease, respectively. Moreover, first in vivo applications of αSyn polypeptides from brain homogenate show the feasibility of on-line affinity-MS to the direct analysis of biological material. These results demonstrate on-line SAW-bioaffinity-MS as a powerful tool for structural and quantitative analysis of biopolymer interactions.


Subject(s)
Amyloid beta-Peptides/analysis , alpha-Synuclein/analysis , Amino Acid Sequence , Amino Acid Substitution , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/radiation effects , Animals , Antibodies, Monoclonal/metabolism , Antibody Affinity , Biosensing Techniques , Brain/metabolism , Cyclotrons , Epitopes , Feasibility Studies , Fourier Analysis , Humans , Mass Spectrometry , Mice, Transgenic , Microfluidic Analytical Techniques , Molecular Weight , Mutant Proteins/analysis , Mutant Proteins/chemistry , Mutant Proteins/radiation effects , Neurons/chemistry , Recombinant Proteins/analysis , Recombinant Proteins/chemistry , Recombinant Proteins/radiation effects , Sound , alpha-Synuclein/chemistry , alpha-Synuclein/genetics , alpha-Synuclein/radiation effects
13.
J Med Chem ; 53(24): 8546-55, 2010 Dec 23.
Article in English | MEDLINE | ID: mdl-21080629

ABSTRACT

Reversible lysine-specific acetylation has been described as an important posttranslational modification, regulating chromatin structure and transcriptional activity in the case of core histone proteins. Histone deacetylases (HDAC) are considered as a promising target for anticancer drug development, with 2a as pan-HDAC inhibitor approved for cutanous T-cell lymphoma therapy and several other HDAC inhibitors currently in preclinical and clinical development. Protein kinases are a well-established target for cancer therapy with the EGFR/HER2 inhibitor 5 approved for treatment of advanced, HER2 positive breast cancer as a prominent example. In the present report, we present a novel strategy for cancer drug development by combination of EGFR/HER2 kinase and HDAC inhibitory activity in one molecule. By combining the structural features of 5 with an (E)-3-(aryl)-N-hydroxyacrylamide motif known from HDAC inhibitors like 1 or 3, we obtained selective inhibitors for both targets with potent cellular activity (target inhibition and cytotoxicity) of selected compounds 6a and 6c. By combining two distinct pharmacologically properties in one molecule, we postulate a broader activity spectrum and less likelihood of drug resistance in cancer patients.


Subject(s)
Acrylamides/chemical synthesis , Antineoplastic Agents/chemical synthesis , ErbB Receptors/antagonists & inhibitors , Histone Deacetylase Inhibitors/chemical synthesis , Quinazolines/chemical synthesis , Receptor, ErbB-2/antagonists & inhibitors , Acetylation , Acrylamides/chemistry , Acrylamides/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Drug Resistance, Neoplasm , Drug Screening Assays, Antitumor , ErbB Receptors/biosynthesis , Histone Deacetylase Inhibitors/chemistry , Histone Deacetylase Inhibitors/pharmacology , Histones/metabolism , Humans , Isoenzymes/antagonists & inhibitors , Lapatinib , Quinazolines/chemistry , Quinazolines/pharmacology , Receptor, ErbB-2/biosynthesis , Stereoisomerism , Structure-Activity Relationship
14.
J Med Chem ; 52(8): 2265-79, 2009 Apr 23.
Article in English | MEDLINE | ID: mdl-19301902

ABSTRACT

Inhibitors of histone deacetylases are a new class of cancer therapeutics with possibly broad applicability. Combinations of HDAC inhibitors with the kinase inhibitor 1 (Imatinib) in recent studies showed additive and synergistic effects. Here we present a new concept by combining inhibition of protein kinases and HDACs, two independent pharmacological activities, in one synthetic small molecule. In general, the HDAC inhibition profile, the potencies, and the probable binding modes to HDAC1 and HDAC6 were similar as for 6 (SAHA). Inhibition of Abl kinase in biochemical assays was maintained for most compounds, but in general the kinase selectivity profile differed from that of 1 with nearly equipotent inhibition of the wild-type and the Imatinib resistant Abl T(315)I mutant. A potent cellular inhibition of PDGFR and cytotoxicity toward EOL-1 cells, a model for idiopathic hypereosinophilic syndrome (HES), are restored or enhanced for selected analogues (12b, 14b, and 18b). Cytotoxicity was evaluated by using a broad panel of tumor cell lines, with selected analogues displaying mean IC(50) values between 3.6 and 7.1 muM.


Subject(s)
Benzamides/chemical synthesis , Histone Deacetylase Inhibitors , Phthalic Acids/chemical synthesis , Piperazines/chemical synthesis , Protein-Tyrosine Kinases/antagonists & inhibitors , Pyrimidines/chemical synthesis , Receptor, Platelet-Derived Growth Factor beta/antagonists & inhibitors , Thiazoles/chemical synthesis , Thiophenes/chemical synthesis , Acetylation , Benzamides/chemistry , Benzamides/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , Fusion Proteins, bcr-abl , Histones/metabolism , Humans , Imatinib Mesylate , Models, Molecular , Mutation , Phthalic Acids/chemistry , Phthalic Acids/pharmacology , Piperazines/chemistry , Piperazines/pharmacology , Protein-Tyrosine Kinases/genetics , Pyrimidines/chemistry , Pyrimidines/pharmacology , Receptor, Platelet-Derived Growth Factor beta/genetics , Stereoisomerism , Structure-Activity Relationship , Thiazoles/chemistry , Thiazoles/pharmacology , Thiophenes/chemistry , Thiophenes/pharmacology
15.
Anal Biochem ; 372(1): 72-81, 2008 Jan 01.
Article in English | MEDLINE | ID: mdl-17868634

ABSTRACT

Most cellular assays that quantify the efficacy of histone deacetylase (HDAC) inhibitors measure hyperacetylation of core histone proteins H3 and H4. Here we describe a new approach, directly measuring cellular HDAC enzymatic activity using the substrate Boc-K(Ac)-7-amino-4-methylcoumarin (AMC). After penetration into HeLa cervical carcinoma or K562 chronic myeloid leukemia cells, the deacetylated product Boc-K-AMC is formed which, after cell lysis, is cleaved by trypsin, finally releasing the fluorophor AMC. The cellular potency of suberoylanilide hydroxamic acid, LBH589, trichostatin A, and MS275 as well-known HDAC inhibitors was determined using this assay. IC(50) values derived from concentration-effect curves correlated well with EC(50) values derived from a cellomics array scan histone H3 hyperacetylation assay. The cellular HDAC activity assay was adapted to a homogeneous format, fully compatible with robotic screening. Concentration-effect curves generated on a Tecan Genesis Freedom workstation were highly reproducible with a signal-to-noise ratio of 5.7 and a Z' factor of 0.88, indicating a very robust assay. Finally, a HDAC-inhibitor focused library was profiled in a medium-throughput screening campaign. Inhibition of cellular HDAC activity correlated well with cytotoxicity and histone H3 hyperacetylation in HeLa cells and with inhibition of human recombinant HDAC1 in a biochemical assay. Thus, by using Boc-K(Ac)-AMC as a cell-permeable HDAC substrate, the activity of various protein lysine-specific deacetylases including HDAC1-containing complexes is measurable in intact cells in a simple and homogeneous manner.


Subject(s)
Histone Deacetylases/metabolism , Robotics , Acetylation , Automation , Enzyme Inhibitors/pharmacology , HeLa Cells , Histone Deacetylase Inhibitors , Humans , Kinetics , Substrate Specificity
16.
J Med Chem ; 51(13): 3985-4001, 2008 Jul 10.
Article in English | MEDLINE | ID: mdl-18558669

ABSTRACT

Inhibitors of histone deacetylases (HDAC) are currently developed for the treatment of cancer. These include compounds with a sulfur containing head group like depsipeptide, alkylthiols, thiocarboxylates, and trithiocarbonates with a carbonyl group in the alpha-position. In the present investigation, we report on the synthesis and comprehensive SAR analysis of HDAC inhibitors bearing a tri- or dithiocarbonate motif. Such trithiocarbonates are readily accessible from either preformed or in situ prepared alpha-halogenated methylaryl ketones. A HDAC isotype selectivity and a substrate competitive mode-of-action is shown for defined analogues. Exploration of the head group showed the necessity of the dithio-alpha-carbonyl motif for potent HDAC inhibition. Highly potent, substrate competitive HDAC6 selective inhibitors were identified (12ac:IC 50 = 65 nM and K i = 110 nM). Trithiocarbonate analogues with an aminoquinoline-substituted pyridinyl-thienoacetyl cap demonstrate a cytotoxicity profile and potency comparable to that of suberoylanilide hydroxamic acid (SAHA) as an approved cancer drug.


Subject(s)
Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Histone Deacetylase Inhibitors , Thiones/chemical synthesis , Thiones/pharmacology , Acetylation , Cell Cycle/drug effects , Cell Survival/drug effects , Enzyme Inhibitors/chemistry , Histone Deacetylases/metabolism , Histones/metabolism , Humans , Isoenzymes/antagonists & inhibitors , Isoenzymes/metabolism , Kinetics , Molecular Structure , Structure-Activity Relationship , Thiones/chemistry
17.
Bioorg Med Chem Lett ; 17(17): 4746-52, 2007 Sep 01.
Article in English | MEDLINE | ID: mdl-17606370

ABSTRACT

Inhibition of histone deacetylases class I/II enzymes is a new, promising approach for cancer therapy. In the present study, we disclose a new structural class of HDAC inhibitors with the trithiocarbonate motif. A clear structure-activity-relationship was obtained for the cap-linker motif and the putative Zn(2+) complexing head group. Selected analogs display potent inhibition of HDAC enzymatic activity and a cellular potency comparable to that of suberoylanilide hydroxamic acid (SAHA), recently approved for treatment of patients with advanced cutaneous T-cell lymphoma.


Subject(s)
Carbonates/chemistry , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/chemical synthesis , Histone Deacetylase Inhibitors , Skin Neoplasms/drug therapy , Carbon/chemistry , Chemistry, Pharmaceutical/methods , Drug Design , Humans , Hydroxamic Acids/pharmacology , Inhibitory Concentration 50 , Lymphoma, T-Cell/drug therapy , Models, Chemical , Molecular Conformation , Vorinostat , Zinc/chemistry
18.
Int J Cancer ; 121(5): 1138-48, 2007 Sep 01.
Article in English | MEDLINE | ID: mdl-17455259

ABSTRACT

Advanced second generation inhibitors of histone deacetylases (HDAC) are currently used in clinical development. This study aimed at comparing the pharmacological properties of selected second generation HDAC inhibitors with the hydroxamate and benzamide head group, namely SAHA, LAQ824/LBH589, CI994, MS275 and MGCD0103. In biochemical assays using recombinant HDAC1, 3, 6 and 8 isoenzymes, SAHA and LAQ824/LBH589 behave as quite unselective HDAC inhibitors. In contrast, the benzamides CI994, MS275 and MGCD0103 are more selective, potent inhibitors of at least HDAC1 and HDAC3. All HDAC inhibitors induce histone H3 hyperacetylation, correlating with inhibition of proliferation, induction of cell differentiation and apoptosis. A broad cytotoxicity is seen across cell lines from different tumor entities with LAQ824/LBH589 being the most potent agents. The apoptosis inducing activity is evident in arrested and proliferating RKO colon cancer cells with inducible, heterologous p21(waf1) expression, indicative for a cell-cycle independent mode-of-action. Differentiation of MDA-MB468 breast cancer cells is induced by benzamide and hydroxamate analogs. The reversibility of drug action was evaluated by pulse treatment of A549 lung cancer cells. Whereas paclitaxel induced irreversible cell cycle alterations already after 6 hr treatment, HDAC inhibitor action was retarded and irreversible after >16 hr treatment. Interestingly, pulse treatment was equally effective as continous treatment. Finally, the efficacy of LAQ824, SAHA and MS275 in A549 nude mice xenografts was comparable to that of paclitaxel at well tolerated doses. We conclude that despite a different HDAC isoenzyme inhibition profile, hydroxamate and benzamide analogs as studied display similar cellular profiles.


Subject(s)
Enzyme Inhibitors/pharmacology , Histone Deacetylase Inhibitors , Isoenzymes/antagonists & inhibitors , Acetylation , Apoptosis/drug effects , Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Carcinoma, Non-Small-Cell Lung/enzymology , Carcinoma, Non-Small-Cell Lung/pathology , Cell Differentiation/drug effects , Cell Line , Cell Proliferation/drug effects , Genes, Reporter , Histone Deacetylases/metabolism , Humans , Isoenzymes/metabolism , Luciferases/genetics , Lung Neoplasms/enzymology , Lung Neoplasms/pathology
19.
EMBO Rep ; 4(5): 511-6, 2003 May.
Article in English | MEDLINE | ID: mdl-12717455

ABSTRACT

Obesity is a metabolic disorder related to improper control of energy uptake and expenditure, which results in excessive accumulation of body fat. Initial insights into the genetic pathways that regulate energy metabolism have been provided by a discrete number of obesity-related genes that have been identified in mammals. Here, we report the identification of the adipose (adp) gene, the mutation of which causes obesity in Drosophila. Loss of adp activity promotes increased fat storage, which extends the lifespan of mutant flies under starvation conditions. By contrast, adp gain-of-function causes a specific reduction of the fat body in Drosophila. adp encodes an evolutionarily conserved WD40/tetratricopeptide-repeat-domain protein that is likely to represent an intermediate in a novel signalling pathway.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Fat Body/metabolism , Mutation , Obesity/genetics , Proteins/genetics , Triglycerides/metabolism , Amino Acid Sequence , Animals , Cloning, Molecular , DNA, Complementary/chemistry , Drosophila Proteins/biosynthesis , Drosophila Proteins/genetics , Energy Metabolism/genetics , Evolution, Molecular , Larva/genetics , Larva/metabolism , Molecular Sequence Data , Obesity/metabolism , Phenotype , Proteins/metabolism , Sequence Alignment , Triglycerides/genetics
SELECTION OF CITATIONS
SEARCH DETAIL