Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
J Pharmacol Exp Ther ; 372(1): 107-118, 2020 01.
Article in English | MEDLINE | ID: mdl-31732698

ABSTRACT

Cystic fibrosis (CF) is the most common monogenic autosomal recessive disease in Caucasians caused by pathogenic mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene (CFTR). Significant small molecule therapeutic advances over the past two decades have been made to target the defective CFTR protein and enhance its function. To address the most prevalent defect of the defective CFTR protein (i.e., F508del mutation) in CF, two biomolecular activities are required, namely, correctors to increase the amount of properly folded F508delCFTR levels at the cell surface and potentiators to allow the effective opening, i.e., function of the F508delCFTR channel. Combined, these activities enhance chloride ion transport yielding improved hydration of the lung surface and subsequent restoration of mucociliary clearance. To enhance clinical benefits to CF patients, a complementary triple combination therapy consisting of two corrector molecules, type 1 (C1) and type 2, with additive mechanisms along with a potentiator are being investigated in the clinic for maximum restoration of mutated CFTR function. We report the identification and in vitro biologic characterization of ABBV-2222/GLPG2222 (4-[(2R,4R)-4-({[1-(2,2-difluoro-1,3-benzodioxol-5-yl)cyclopropyl]carbonyl}amino)-7-(difluoromethoxy)-3,4-dihydro-2H-chromen-2-yl]benzoic acid),-a novel, potent, and orally bioavailable C1 corrector developed by AbbVie-Galapagos and currently in clinical trials-which exhibits substantial improvements over the existing C1 correctors. This includes improvements in potency and drug-drug interaction (DDI) compared with 3-(6-(1-(2,2-difluorobenzo[d][1,3]dioxol-5-yl)cyclopropanecarboxamido)-3-methylpyridin-2-yl)benzoic acid (VX-809, Lumacaftor) and improvements in potency and efficacy compared with 1-(2,2-difluoro-1,3-benzodioxol-5-yl)-N-[1-[(2R)-2,3-dihydroxypropyl]-6-fluoro-2-(1-hydroxy-2-methylpropan-2-yl)indol-5-yl]cyclopropane-1-carboxamide (VX-661, Tezacaftor). ABBV-2222/GLPG2222 exhibits potent in vitro functional activity in primary patient cells harboring F508del/F508del CFTR with an EC50 value <10 nM. SIGNIFICANCE STATEMENT: To address the most prevalent defect of the defective CFTR protein (i.e., F508del mutation) in cystic fibrosis, AbbVie-Galapagos has developed ABBV-2222/GLPG2222, a novel, potent, and orally bioavailable C1 corrector of this protein. ABBV-2222/GLPG2222, which is currently in clinical trials, exhibits potent in vitro functional activity in primary patient cells harboring F508del/F508del CFTR and substantial improvements over the existing C1 correctors.


Subject(s)
Benzoates/pharmacology , Benzopyrans/pharmacology , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Protein Folding/drug effects , Animals , Binding Sites , Cell Membrane/metabolism , Cells, Cultured , Chlorides/metabolism , Cricetinae , Cystic Fibrosis Transmembrane Conductance Regulator/chemistry , HEK293 Cells , Humans , Membrane Transport Modulators/pharmacology , Protein Binding , Protein Transport/drug effects , Respiratory Mucosa/drug effects , Respiratory Mucosa/metabolism
2.
Mol Neurodegener ; 7: 8, 2012 Mar 16.
Article in English | MEDLINE | ID: mdl-22423893

ABSTRACT

BACKGROUND: The form(s) of amyloid-ß peptide (Aß) associated with the pathology characteristic of Alzheimer's disease (AD) remains unclear. In particular, the neurotoxicity of intraneuronal Aß accumulation is an issue of considerable controversy; even the existence of Aß deposits within neurons has recently been challenged by Winton and co-workers. These authors purport that it is actually intraneuronal APP that is being detected by antibodies thought to be specific for Aß. To further address this issue, an anti-Aß antibody was developed (MOAB-2) that specifically detects Aß, but not APP. This antibody allows for the further evaluation of the early accumulation of intraneuronal Aß in transgenic mice with increased levels of human Aß in 5xFAD and 3xTg mice. RESULTS: MOAB-2 (mouse IgG2b) is a pan-specific, high-titer antibody to Aß residues 1-4 as demonstrated by biochemical and immunohistochemical analyses (IHC), particularly compared to 6E10 (a commonly used commercial antibody to Aß residues 3-8). MOAB-2 did not detect APP or APP-CTFs in cell culture media/lysates (HEK-APPSwe or HEK-APPSwe/BACE1) or in brain homogenates from transgenic mice expressing 5 familial AD (FAD) mutation (5xFAD mice). Using IHC on 5xFAD brain tissue, MOAB-2 immunoreactivity co-localized with C-terminal antibodies specific for Aß40 and Aß42. MOAB-2 did not co-localize with either N- or C-terminal antibodies to APP. In addition, no MOAB-2-immunoreactivity was observed in the brains of 5xFAD/BACE-/- mice, although significant amounts of APP were detected by N- and C-terminal antibodies to APP, as well as by 6E10. In both 5xFAD and 3xTg mouse brain tissue, MOAB-2 co-localized with cathepsin-D, a marker for acidic organelles, further evidence for intraneuronal Aß, distinct from Aß associated with the cell membrane. MOAB-2 demonstrated strong intraneuronal and extra-cellular immunoreactivity in 5xFAD and 3xTg mouse brain tissues. CONCLUSIONS: Both intraneuronal Aß accumulation and extracellular Aß deposition was demonstrated in 5xFAD mice and 3xTg mice with MOAB-2, an antibody that will help differentiate intracellular Aß from APP. However, further investigation is required to determine whether a molecular mechanism links the presence of intraneuronal Aß with neurotoxicity. As well, understanding the relevance of these observations to human AD patients is critical.


Subject(s)
Amyloid beta-Peptides/analysis , Amyloid beta-Protein Precursor/analysis , Antibodies/analysis , Immunohistochemistry/methods , Alzheimer Disease/pathology , Amyloid beta-Peptides/immunology , Amyloid beta-Protein Precursor/immunology , Animals , Antibodies/immunology , Antibody Specificity , Antigen-Antibody Reactions , Cells, Cultured , Disease Models, Animal , Humans , Mice , Mice, Inbred BALB C , Mice, Transgenic , Plaque, Amyloid/chemistry , Plaque, Amyloid/pathology
4.
Bioorg Med Chem Lett ; 20(11): 3295-300, 2010 Jun 01.
Article in English | MEDLINE | ID: mdl-20457525

ABSTRACT

A series of quinoline containing histamine H(3) antagonists is reported herein. These analogs were synthesized via the Friedlander quinoline synthesis between an aminoaldehyde intermediate and a methyl ketone allowing for a wide diversity of substituents at the 2-position of the quinoline ring.


Subject(s)
Histamine H3 Antagonists/pharmacology , Quinolines/pharmacology , Animals , Humans , In Vitro Techniques , Rats
5.
Bioorg Med Chem Lett ; 20(6): 1900-4, 2010 Mar 15.
Article in English | MEDLINE | ID: mdl-20171098

ABSTRACT

Three novel series of histamine H(4) receptor (H(4)R) antagonists containing the 2-aminopyrimidine motif are reported. The best of these compounds display good in vitro potency in both functional and binding assays. In addition, representative compounds are able to completely block itch responses when dosed ip in a mouse model of H(4)-agonist induced scratching, thus demonstrating their activities as H(4)R antagonists.


Subject(s)
Aminopyridines/pharmacology , Histamine Antagonists/pharmacology , Receptors, G-Protein-Coupled/antagonists & inhibitors , Animals , Humans , Mice , Receptors, Histamine , Receptors, Histamine H4
6.
Brain Res ; 1250: 41-8, 2009 Jan 23.
Article in English | MEDLINE | ID: mdl-19046950

ABSTRACT

Existing data on the expression of H(4) histamine receptor in the CNS are conflicting and inconclusive. In this report, we present the results of experiments that were conducted in order to elucidate H(4) receptor expression and localization in the brain, spinal cord, and dorsal root ganglia (DRG). Here we show that transcripts of H(4) receptor are present in all analyzed regions of the human CNS, including spinal cord, hippocampus, cortex, thalamus and amygdala, with the highest levels of H(4) mRNA detected in the spinal cord. In rat, H(4) mRNA was detected in cortex, cerebellum, brainstem, amygdala, thalamus and striatum. Very low levels of H(4) mRNA were detected in hypothalamus, and no H(4) signal was detected in the rat hippocampus. Fairly low levels of H(4) mRNA were detected in examined peripheral tissues including spleen and liver. Interestingly, strong expression of H(4) mRNA was detected in the rat DRG and spinal cord. Immunohistochemical analysis revealed expression of H(4) receptors on neurons in the rat lumbar DRG and in the lumbar spinal cord. Our observations provide evidence of the H(4) presence in both human and rodent CNS and offer some insight into possible role of H(4) in itch and pain.


Subject(s)
Brain/metabolism , Ganglia, Spinal/metabolism , Receptors, G-Protein-Coupled/metabolism , Receptors, Histamine/metabolism , Spinal Cord/metabolism , Animals , Cells, Cultured , Humans , Immunohistochemistry , Liver/metabolism , Male , Neuroglia/metabolism , Neurons/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptors, G-Protein-Coupled/genetics , Receptors, Histamine/genetics , Receptors, Histamine H4 , Reverse Transcriptase Polymerase Chain Reaction , Spleen/metabolism
7.
J Med Chem ; 51(22): 7094-8, 2008 Nov 27.
Article in English | MEDLINE | ID: mdl-18983139

ABSTRACT

cis-4-(Piperazin-1-yl)-5,6,7a,8,9,10,11,11a-octahydrobenzofuro[2,3-h]quinazolin-2-amine, 4 (A-987306) is a new histamine H(4) antagonist. The compound is potent in H(4) receptor binding assays (rat H(4), K(i) = 3.4 nM, human H(4) K(i) = 5.8 nM) and demonstrated potent functional antagonism in vitro at human, rat, and mouse H(4) receptors in cell-based FLIPR assays. Compound 4 also demonstrated H(4) antagonism in vivo in mice, blocking H(4)-agonist induced scratch responses, and showed anti-inflammatory activity in mice in a peritonitis model. Most interesting was the high potency and efficacy of this compound in blocking pain responses, where it showed an ED(50) of 42 mumol/kg (ip) in a rat post-carrageenan thermal hyperalgesia model of inflammatory pain.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Benzofurans/pharmacology , Hyperalgesia/drug therapy , Pain/prevention & control , Quinazolines/pharmacology , Receptors, G-Protein-Coupled/antagonists & inhibitors , Animals , Anti-Inflammatory Agents, Non-Steroidal/chemical synthesis , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Benzofurans/chemical synthesis , Benzofurans/chemistry , Carrageenan , Disease Models, Animal , Drug Design , Drug Evaluation, Preclinical , Humans , Hyperalgesia/chemically induced , Ligands , Mice , Molecular Structure , Pain/physiopathology , Peritonitis/drug therapy , Quinazolines/chemical synthesis , Quinazolines/chemistry , Rats , Receptors, Histamine , Receptors, Histamine H4 , Stereoisomerism , Structure-Activity Relationship
8.
Eur J Pharmacol ; 601(1-3): 8-15, 2008 Dec 28.
Article in English | MEDLINE | ID: mdl-18977214

ABSTRACT

We have recently identified three splice isoforms of the histamine H(3) receptor in multiple brain regions of cynomolgus monkey (Macaca fascicularis). Two of the novel isoforms displayed a deletion in the third intracellular loop (H(3)(413) and H(3)(410)), the third isoform H(3)(335) displayed a deletion in the i3 intracellular loop and a complete deletion of the putative fifth transmembrane domain TM5. We have confirmed by RT-PCR the expression of full-length H(3)(445) mRNA as well as H(3)(413), H(3)(410), and H(3)(335) splice isoform mRNA in multiple monkey brain regions including the frontal, parietal and occipital cortex, parahippocampal gyrus, hippocampus, amygdala, caudate nucleus, putamen, thalamus, hypothalamus, and cerebellum. The full-length isoform H(3)(445) was predominant in all of the regions tested, followed by H(3)(335), with the H(3)(413) and H(3)(410) being of low abundance. When expressed in C6 cells, H(3)(445), H(3)(413), and H(3)(410) exhibit high affinity binding to the agonist ligand [(3)H]-(N)-alpha-methylhistamine with respective pK(D) values of 9.7, 9.7, and 9.6. As expected, the H(3)(335) isoform did not display any saturable binding with [(3)H]-(N)-alpha-methylhistamine. The histamine H(3) receptor agonists histamine, (R)-alpha-methylhistamine, imetit and proxyfan were able to activate calcium mobilization responses through H(3)(445), H(3)(413) and H(3)(410) receptors when they were co-expressed with the chimeric G alpha(qi5)-protein in HEK293 cells, while no response was elicited in cells expressing the H(3)(335) isoform. The existence of multiple H(3) receptor splice isoforms across species raises the possibility that isoform specific properties including ligand affinity, signal transduction coupling, and brain localization may differentially contribute to observed in vivo effects of histamine H(3) receptor antagonists.


Subject(s)
Gene Expression , RNA, Messenger/metabolism , Receptors, Histamine H3/metabolism , Animals , Brain/metabolism , Cell Line , Cloning, Molecular , Histamine Agonists/pharmacology , Humans , Ligands , Macaca fascicularis , Male , Protein Binding , Protein Isoforms , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Species Specificity
9.
Assay Drug Dev Technol ; 6(3): 339-49, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18593375

ABSTRACT

Guanosine 5'-O-(3-[(35)S]thio)triphosphate ([(35)S]GTPgammaS) binding assays were established and utilized as a reliable and high-capacity functional assay for determining antagonist and inverse agonist pharmacological parameters of novel histamine H(3) ligands, at the recombinant human H(3) receptor. [(35)S]GTPgammaS binding assays were performed with membranes prepared from human embryonic kidney 293 cells stably expressing the full-length (445 amino acids) human H(3) receptor isoform, at approximately 1 pmol/mg of protein. Utilizing robotic liquid handling, assay filtration, and scintillation counting in a 96-well format, concentration-response curves were determined for up to 40 compounds per assay. The imidazole-containing H(3) receptor antagonist ciproxifan and the non-imidazole antagonist ABT-239 inhibited (R)-alpha-methylhistamine (RAMH)-stimulated [(35)S]GTPgammaS binding in a competitive manner, and negative logarithm of the dissociation equilibrium constant (pK(b)) values determined for nearly 200 structurally diverse H(3) antagonists were very similar to the respective negative logarithm of the equilibrium inhibition constant values from N-alpha-[(3)H]methylhistamine competition binding assays. H(3) antagonists also concentration-dependently decreased basal [(35)S]GTPgammaS binding, thereby displaying inverse agonism at the constitutively active H(3) receptor. At maximally effective concentrations, non-imidazole H(3) antagonists inhibited basal [(35)S]GTPgammaS binding by approximately 20%. For over 100 of these antagonists, negative logarithm of the 50% effective concentration values for inverse agonism were very similar to the respective pK(b) values. Both H(3) receptor agonist-dependent and -independent (constitutive) [(35)S]GTPgammaS binding were sensitive to changes in assay concentrations of sodium, magnesium, and the guanine nucleotide GDP; however, the potency of ABT-239 for inhibition of RAMH-stimulated [(35)S]GTPgammaS binding was not significantly affected. These robust and reliable [(35)S]GTPgammaS binding assays have become one of the important tools in our pharmacological analysis and development of novel histamine H(3) receptor antagonists/inverse agonists.


Subject(s)
Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Histamine Agonists/pharmacology , Histamine H3 Antagonists/pharmacology , Receptors, Histamine H3/drug effects , Sulfur Radioisotopes , Benzofurans/pharmacology , Cell Line , Drug Inverse Agonism , Humans , Ligands , Methylhistamines/pharmacology , Pyrrolidines/pharmacology
10.
Neurobiol Aging ; 28(8): 1139-47, 2007 Aug.
Article in English | MEDLINE | ID: mdl-16837105

ABSTRACT

Autosomal dominant mutations that increase amyloid-beta(1-42) (Abeta42) cause familial Alzheimer's disease (AD), and the most common genetic risk factor for AD is the presence of the epsilon4 allele of apolipoprotein E (apoE). Previously, we characterized stable preparations of Abeta42 oligomers and fibrils and reported that oligomers induced a 10-fold greater increase in neurotoxicity than fibrils in Neuro-2A cells. To determine the effects of apoE genotype on Abeta42 oligomer- and fibril-induced neurotoxicity in vitro, we co-cultured wild type (WT) neurons with glia from WT, apoE-knockout (apoE-KO), and human apoE2-, E3-, and E4-targeted replacement (TR) mice. Dose-dependent neurotoxicity was induced by oligomeric Abeta42 with a ranking order of apoE4-TR>KO=apoE2-TR=apoE3-TR>WT. Neurotoxicity induced by staurosporine or glutamate were not affected by apoE genotype, indicating specificity for oligomeric Abeta42-induced neurotoxicity. These in vitro data demonstrate a gain of negative function for apoE4, synergistic with oligomeric Abeta42, in mediating neurotoxicity.


Subject(s)
Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/toxicity , Apolipoproteins E/physiology , Neurons/drug effects , Peptide Fragments/metabolism , Peptide Fragments/toxicity , Adenosine Triphosphate/metabolism , Animals , Animals, Newborn , Apolipoproteins E/genetics , Cell Survival/drug effects , Cells, Cultured , Cerebral Cortex/cytology , Coculture Techniques/methods , Dose-Response Relationship, Drug , Embryo, Mammalian , Female , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neuroglia/chemistry , Neuroglia/physiology , Pregnancy , Protein Conformation , Protein Isoforms/genetics , Protein Isoforms/physiology , Time Factors
11.
Neurobiol Dis ; 18(3): 459-65, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15755672

ABSTRACT

Activated glia, as a result of chronic inflammation, are associated with amyloid-beta peptide (Abeta) deposits in the brain of Alzheimer's disease (AD) patients. In vitro, glia are activated by Abeta inducing secretion of pro-inflammatory molecules. Recent studies have focused on soluble oligomers (or protofibrils) of Abeta as the toxic species in AD. In the present study, using rat astrocyte cultures, oligomeric Abeta induced initial high levels of IL-1beta decreasing over time and, in contrast, fibrillar Abeta increased IL-1beta levels over time. In addition, oligomeric Abeta, but not fibrillar Abeta, induced high levels of iNOS, NO, and TNF-alpha. Our results suggest that oligomers induced a profound, early inflammatory response, whereas fibrillar Abeta showed less increase of pro-inflammatory molecules, consistent with a more chronic form of inflammation.


Subject(s)
Amyloid beta-Peptides/metabolism , Amyloid/metabolism , Astrocytes/metabolism , Astrocytes/pathology , Peptide Fragments/metabolism , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/pharmacology , Animals , Astrocytes/drug effects , Cells, Cultured , Chronic Disease , Humans , Inflammation/metabolism , Inflammation/pathology , Interleukin-1/metabolism , Peptide Fragments/pharmacology , Rats , Rats, Sprague-Dawley
12.
J Mol Neurosci ; 23(3): 235-46, 2004.
Article in English | MEDLINE | ID: mdl-15181252

ABSTRACT

Abnormalities in the processing of amyloid precursor protein to amyloid-beta (Abeta) are causal factors, and the presence of the epsilon4 allele of apolipoprotein E (apoE) is the primary risk factor for Alzheimer's disease (AD). Based, at least in part, on these genetics, the potential structural and functional interactions between these two proteins are the focus of our research. To understand the nature of the physical interactions between apoE and Abeta, we initially utilized gel-shift assays to demonstrate that native apoE2 and E3 (associated with lipid particles) form an SDS-stable complex with Abeta that is more abundant than the apoE4:Abeta complex. We further demonstrated that exogenous apoE3 but not E4 prevents Abeta-induced neurotoxicity by a process that requires apoE receptors. In addition, both exogenous apoE3 and E4 prevent Abeta-induced, glial-mediated inflammation, also via a process that requires apoE receptors. These functional effects all occur at a molar ratio of apoE to Abeta of 1:30. Because the biological activities for both apoE and Abeta are profoundly influenced by their isoform and conformation, respectively, we further investigated the idea that apoE3 and E4 differentially interact with particular aggregation species of Abeta1-42. Our overall hypothesis is that apoE has two general functions in relation to Abeta. First, apoE interacts with oligomeric Abeta via an apoE receptor-mediated process to inhibit neurotoxicity and neuroinflammation (apoE3 > apoE4) a process possibly related to binding and clearance of apoE3:oligomer complexes. Second, apoE facilitates the deposition of Abeta as amyloid (apoE4 > apoE3). We will continue to investigate the effect of apoE isoform and Abeta conformation on the structural and functional interactions between these two proteins in relation to the pathogenesis of AD.


Subject(s)
Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/metabolism , Apolipoproteins E/genetics , Apolipoproteins E/metabolism , Peptide Fragments/genetics , Peptide Fragments/metabolism , Alzheimer Disease/epidemiology , Alzheimer Disease/genetics , Amyloid beta-Peptides/chemistry , Animals , Humans , Models, Animal , Neurofibrils/pathology , Neurofibrils/physiology , Neurons/physiology , Peptide Fragments/chemistry , Protein Conformation , Risk Factors
13.
J Biol Chem ; 277(35): 32046-53, 2002 Aug 30.
Article in English | MEDLINE | ID: mdl-12058030

ABSTRACT

Genetic evidence predicts a causative role for amyloid-beta (A beta) in Alzheimer's disease. Recent debate has focused on whether fibrils (amyloid) or soluble oligomers of A beta are the active species that contribute to neurodegeneration and dementia. We developed two aggregation protocols for the consistent production of stable oligomeric or fibrillar preparations of A beta-(1-42). Here we report that oligomers inhibit neuronal viability 10-fold more than fibrils and approximately 40-fold more than unaggregated peptide, with oligomeric A beta-(1-42)-induced inhibition significant at 10 nm. Under A beta-(1-42) oligomer- and fibril-forming conditions, A beta-(1-40) remains predominantly as unassembled monomer and had significantly less effect on neuronal viability than preparations of A beta-(1-42). We applied the aggregation protocols developed for wild type A beta-(1-42) to A beta-(1-42) with the Dutch (E22Q) or Arctic (E22G) mutations. Oligomeric preparations of the mutations exhibited extensive protofibril and fibril formation, respectively, but were not consistently different from wild type A beta-(1-42) in terms of inhibition of neuronal viability. However, fibrillar preparations of the mutants appeared larger and induced significantly more inhibition of neuronal viability than wild type A beta-(1-42) fibril preparations. These data demonstrate that protocols developed to produce oligomeric and fibrillar A beta-(1-42) are useful in distinguishing the structural and functional differences between A beta-(1-42) and A beta-(1-40) and genetic mutations of A beta-(1-42).


Subject(s)
Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/physiology , Cell Survival/physiology , Neurons/cytology , Peptide Fragments/chemistry , Alzheimer Disease , Humans , Microscopy, Atomic Force , Neuroblastoma , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...