Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Epilepsia ; 64(8): 2126-2136, 2023 08.
Article in English | MEDLINE | ID: mdl-37177976

ABSTRACT

OBJECTIVE: Gain of function variants in the sodium-activated potassium channel KCNT1 have been associated with pediatric epilepsy disorders. Here, we systematically examine a spectrum of KCNT1 variants and establish their impact on channel function in multiple cellular systems. METHODS: KCNT1 variants identified from published reports and genetic screening of pediatric epilepsy patients were expressed in Xenopus oocytes and HEK cell lines. Variant impact on current magnitude, current-voltage relationships, and sodium ion modulation were examined. RESULTS: We determined basic properties of KCNT1 in Xenopus oocyte and HEK systems, including the role of extra- and intracellular sodium in regulating KCNT1 activity. The most common six KCNT1 variants demonstrated strong gain of function (GOF) effects on one or more channel properties. Analysis of 36 total variants identified phenotypic heterogeneity but a strong tendency for pathogenic variants to exert GOF effects on channel properties. By controlling intracellular sodium, we demonstrate that multiple pathogenic KCNT1 variants modulate channel voltage dependence by altering the sensitivity to sodium ions. SIGNIFICANCE: This study represents the largest systematic functional examination of KCNT1 variants to date. We both confirm previously reported GOF channel phenotypes and expand the number of variants with in vitro GOF effects. Our data provide further evidence that novel KCNT1 variants identified in epilepsy patients lead to disease through generalizable GOF mechanisms including increases in current magnitude and/or current-voltage relationships.


Subject(s)
Epilepsy , Gain of Function Mutation , Humans , Potassium Channels, Sodium-Activated/genetics , Mutation , Epilepsy/genetics , Potassium Channels/genetics , Potassium Channels/metabolism , Nerve Tissue Proteins/genetics
2.
Nucleic Acids Res ; 49(20): 11560-11574, 2021 11 18.
Article in English | MEDLINE | ID: mdl-34718736

ABSTRACT

Friedreich's ataxia (FRDA) is a severe multisystem disease caused by transcriptional repression induced by expanded GAA repeats located in intron 1 of the Frataxin (FXN) gene encoding frataxin. FRDA results from decreased levels of frataxin; thus, stabilization of the FXN mRNA already present in patient cells represents an attractive and unexplored therapeutic avenue. In this work, we pursued a novel approach based on oligonucleotide-mediated targeting of FXN mRNA ends to extend its half-life and availability as a template for translation. We demonstrated that oligonucleotides designed to bind to FXN 5' or 3' noncoding regions can increase FXN mRNA and protein levels. Simultaneous delivery of oligonucleotides targeting both ends increases efficacy of the treatment. The approach was confirmed in several FRDA fibroblast and induced pluripotent stem cell-derived neuronal progenitor lines. RNA sequencing and single-cell expression analyses confirmed oligonucleotide-mediated FXN mRNA upregulation. Mechanistically, a significant elongation of the FXN mRNA half-life without any changes in chromatin status at the FXN gene was observed upon treatment with end-targeting oligonucleotides, indicating that transcript stabilization is responsible for frataxin upregulation. These results identify a novel approach toward upregulation of steady-state mRNA levels via oligonucleotide-mediated end targeting that may be of significance to any condition resulting from transcription downregulation.


Subject(s)
Friedreich Ataxia/therapy , Genetic Therapy/methods , Iron-Binding Proteins/genetics , RNA Stability , RNA, Messenger/metabolism , 3' Untranslated Regions , 5' Untranslated Regions , Cells, Cultured , Humans , Iron-Binding Proteins/metabolism , Oligonucleotides, Antisense/chemistry , Oligonucleotides, Antisense/metabolism , RNA, Messenger/chemistry , RNA, Messenger/genetics , Frataxin
3.
Bioorg Med Chem Lett ; 27(23): 5144-5148, 2017 12 01.
Article in English | MEDLINE | ID: mdl-29103974

ABSTRACT

Spinal muscular atrophy (SMA) is a neurodegenerative disorder that results from mutations in the SMN1 gene, leading to survival motor neuron (SMN) protein deficiency. One therapeutic strategy for SMA is to identify compounds that enhance the expression of the SMN2 gene, which normally only is a minor contributor to functional SMN protein production, but which is unaffected in SMA. A recent high-throughput screening campaign identified a 3,4-dihydro-4-phenyl-2(1H)-quinolinone derivative (2) that increases the expression of SMN2 by 2-fold with an EC50 = 8.3 µM. A structure-activity relationship (SAR) study revealed that the array of tolerated substituents, on either the benzo portion of the quinolinone or the 4-phenyl, was very narrow. However, the lactam ring of the quinolinone was more amenable to modifications. For example, the quinazolinone (9a) and the benzoxazepin-2(3H)-one (19) demonstrated improved potency and efficacy for increase in SMN2 expression as compared to 2.


Subject(s)
Quinolones/chemistry , Survival of Motor Neuron 2 Protein/metabolism , Animals , Cell Line , Cyclization , Gene Expression/drug effects , Humans , Mice , Microsomes, Liver/metabolism , Muscular Atrophy, Spinal/metabolism , Muscular Atrophy, Spinal/pathology , Quinolones/pharmacology , RNA, Messenger/metabolism , Solubility , Structure-Activity Relationship , Survival of Motor Neuron 2 Protein/genetics
4.
Biochem Biophys Res Commun ; 414(1): 25-30, 2011 Oct 14.
Article in English | MEDLINE | ID: mdl-21925145

ABSTRACT

Spinal muscular atrophy (SMA) is an autosomal recessive neurodegenerative disorder that is the leading genetic cause of infantile death. SMA is caused by homozygous deletion or mutation of the survival of motor neuron 1 gene (SMN1). The SMN2 gene is nearly identical to SMN1, however is alternatively spliced. The close relationship to SMN1 results in SMN2 being a very power genetic modifier of SMA disease severity and a target for therapies. We sought to identify the regulatory role individual HDAC proteins use to control expression of full length protein from the SMN2 genes. We used quantitative PCR to determine the effects shRNA silencing of individual HDACs on the steady state levels of a SMN2-luciferase reporter transcripts. We determined that reduction of individual HDAC proteins was sufficient to increase SMN protein levels in a transgenic reporter system. Knockdown of class I HDAC proteins preferentially activated the reporter by increased promoter transcription. Silencing of class II HDAC proteins maintained transcriptional activity; however silencing of HDAC 5 and 6 also appeared to enhance inclusion of an alternatively spliced exon. This work highlights HDAC proteins 2 and 6 as excellent investigative targets. These data are important to the basic understanding of SMN expression regulation and the refinements of current therapeutic compounds as well as the development of novel SMA therapeutics.


Subject(s)
Gene Expression Regulation , Histone Deacetylases/metabolism , Muscular Atrophy, Spinal/genetics , Gene Knockdown Techniques , Gene Silencing , Genes, Reporter , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/genetics , Histones/metabolism , Humans , Luciferases/genetics , Promoter Regions, Genetic , RNA Polymerase II/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , Survival of Motor Neuron 2 Protein/genetics
5.
J Virol ; 82(1): 71-6, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17942561

ABSTRACT

The human papillomavirus (HPV) type 16 (HPV16) E6 protein stimulates transcription of the catalytic subunit of telomerase, hTERT, in epithelial cells. It has been reported that binding to the ubiquitin ligase E6AP is required for this E6 activity, with E6 directing E6AP to the hTERT promoter. We previously reported two E6AP binding-defective HPV16 E6 mutations that induced immortalization of human mammary epithelial cells. Because activation of hTERT is proposed to be necessary for epithelial cell immortalization, we sought to further characterize the relationship between E6/E6AP association and telomerase induction. We demonstrate that while these E6 mutants do not bind E6AP, they retain the capability to stimulate the expression of hTERT. Chromatin immunoprecipitation assays confirmed the presence of Myc, wild-type E6, and the E6AP binding-defective E6 mutants, but not E6AP itself, at the endogenous hTERT promoter. Interestingly, an immortalization-defective E6 mutant localized to the hTERT promoter but failed to increase transcription. We conclude that binding to E6AP is not necessary for E6 localization to or activation of the hTERT promoter and that another activity of E6 is involved in hTERT activation.


Subject(s)
Human papillomavirus 16/physiology , Keratinocytes/virology , Oncogene Proteins, Viral/metabolism , Repressor Proteins/metabolism , Telomerase/biosynthesis , Ubiquitin-Protein Ligases/metabolism , Cells, Cultured , Chromatin Immunoprecipitation , Humans , Mutant Proteins/genetics , Mutant Proteins/metabolism , Oncogene Proteins, Viral/genetics , Promoter Regions, Genetic , Protein Binding , Repressor Proteins/genetics , Telomerase/genetics
6.
Stem Cells Dev ; 16(6): 1027-41, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18078382

ABSTRACT

Human embryonic stem (ES) cells are promising resources for developing new treatments for neurodegenerative diseases. Spinal muscular atrophy (SMA) is one of the leading causes of childhood paralysis and infant mortality. SMA is caused by inactivation of the survival motor neuron-1 (SMN1) gene. The nearly identical SMN2 gene contains a silent polymorphism that disrupts splicing and as a result cannot compensate for loss of SMN1. The SMA Project was established by the National Institute of Neurological Disorders and Stroke (NINDS) as a pilot effort to establish a fully transparent coalition between academics, industry, and government to create a centralized network of shared resources and information to identify and test new SMA therapeutics. As one of the funded projects, the work described here tested the feasibility of generating a SMA cell-based assay using neural lineages derived from human ES cells approved for National Institutes of Health (NIH)-funded research. Minigene cassettes were constructed, employing firefly luciferase or green fluorescent protein (GFP) as reporters for splicing efficiency of SMN1 and/or SMN2 under the control of the SMN1, SMN2, or cytomegalovirus (CMV) promoters. Transient transfection of proliferating neuroprogenitors in a 96-well format with plasmid DNA or adenoviral vectors showed differential levels that correlated with the splicing minigene and the promoter used; luciferase activities with SMN1 splicing minigenes were higher than SMN2, and the CMV promoter generated higher levels of activity than the SMN1 and SMN2 promoters. Our results indicate that human ES cell-derived neuroprogenitors provide a promising new primary cell source for assays of new therapeutics for neurodegenerative diseases.


Subject(s)
Embryonic Stem Cells/cytology , Embryonic Stem Cells/physiology , Muscular Atrophy, Spinal/pathology , Muscular Atrophy, Spinal/therapy , Neural Crest/cytology , Stem Cell Transplantation , Aneuploidy , Cell Adhesion , Child , Child, Preschool , Cyclic AMP Response Element-Binding Protein/genetics , Female , Genes, Reporter , Humans , Infant , Luciferases/analysis , Luciferases/genetics , Muscular Atrophy, Spinal/embryology , Muscular Atrophy, Spinal/mortality , Nerve Tissue Proteins/genetics , Neural Crest/physiology , Plasmids , Polymorphism, Genetic , Pregnancy , RNA-Binding Proteins/genetics , SMN Complex Proteins , Survival of Motor Neuron 1 Protein , Survival of Motor Neuron 2 Protein , Transfection
7.
J Med Chem ; 60(11): 4594-4610, 2017 06 08.
Article in English | MEDLINE | ID: mdl-28481536

ABSTRACT

Spinal muscular atrophy (SMA) is the leading genetic cause of infant death. We previously developed a high-throughput assay that employs an SMN2-luciferase reporter allowing identification of compounds that act transcriptionally, enhance exon recognition, or stabilize the SMN protein. We describe optimization and characterization of an analog suitable for in vivo testing. Initially, we identified analog 4m that had good in vitro properties but low plasma and brain exposure in a mouse PK experiment due to short plasma stability; this was overcome by reversing the amide bond and changing the heterocycle. Thiazole 27 showed excellent in vitro properties and a promising mouse PK profile, making it suitable for in vivo testing. This series post-translationally stabilizes the SMN protein, unrelated to global proteasome or autophagy inhibition, revealing a novel therapeutic mechanism that should complement other modalities for treatment of SMA.


Subject(s)
Anilides/pharmacology , Benzamides/pharmacology , Isoxazoles/pharmacology , Molecular Probes , Muscular Atrophy, Spinal/therapy , Protein Processing, Post-Translational , Quinolones/pharmacology , Survival of Motor Neuron 1 Protein/metabolism , Thiazoles/pharmacology , Anilides/pharmacokinetics , Anilides/therapeutic use , Area Under Curve , Benzamides/pharmacokinetics , Benzamides/therapeutic use , Cell Line , Drug Discovery , Half-Life , Humans , Isoxazoles/pharmacokinetics , Isoxazoles/therapeutic use , Protein Stability , Quinolones/pharmacokinetics , Quinolones/therapeutic use , Structure-Activity Relationship , Thiazoles/pharmacokinetics , Thiazoles/therapeutic use
8.
J Med Chem ; 60(7): 3094-3108, 2017 04 13.
Article in English | MEDLINE | ID: mdl-28257199

ABSTRACT

The C-5 substituted 2,4-diaminoquinazoline RG3039 (compound 1), a member of a chemical series that was identified and optimized using an SMN2 promoter screen, prolongs survival and improves motor function in a mouse model of spinal muscular atrophy (SMA). It is a potent inhibitor of the mRNA decapping scavenger enzyme (DcpS), but the mechanism whereby DcpS inhibition leads to therapeutic benefit is unclear. Compound 1 is a dibasic lipophilic molecule that is predicted to accumulate in lysosomes. To understand if the in vivo efficacy is due to DcpS inhibition or other effects resulting from the physicochemical properties of the chemotype, we undertook structure based molecular design to identify DcpS inhibitors with improved physicochemical properties. Herein we describe the design, synthesis, and in vitro pharmacological characterization of these DcpS inhibitors along with the in vivo mouse CNS PK profile of PF-DcpSi (compound 24), one of the analogs found to be efficacious in SMA mouse model.


Subject(s)
Drug Design , Endoribonucleases/antagonists & inhibitors , Muscular Atrophy, Spinal/drug therapy , Quinazolines/chemistry , Quinazolines/therapeutic use , RNA, Messenger/antagonists & inhibitors , Animals , Disease Models, Animal , Endoribonucleases/genetics , Endoribonucleases/metabolism , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacokinetics , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , HEK293 Cells , Humans , Mice , Molecular Docking Simulation , Muscular Atrophy, Spinal/genetics , Muscular Atrophy, Spinal/metabolism , Quinazolines/pharmacokinetics , Quinazolines/pharmacology , RNA, Messenger/genetics , Survival of Motor Neuron 2 Protein
9.
PLoS One ; 12(9): e0185079, 2017.
Article in English | MEDLINE | ID: mdl-28945765

ABSTRACT

C5-substituted 2,4-diaminoquinazoline inhibitors of the decapping scavenger enzyme DcpS (DAQ-DcpSi) have been developed for the treatment of spinal muscular atrophy (SMA), which is caused by genetic deficiency in the Survival Motor Neuron (SMN) protein. These compounds are claimed to act as SMN2 transcriptional activators but data underlying that claim are equivocal. In addition it is unclear whether the claimed effects on SMN2 are a direct consequence of DcpS inhibitor or might be a consequence of lysosomotropism, which is known to be neuroprotective. DAQ-DcpSi effects were characterized in cells in vitro utilizing DcpS knockdown and 7-methyl analogues as probes for DcpS vs non-DcpS-mediated effects. We also performed analysis of Smn transcript levels, RNA-Seq analysis of the transcriptome and SMN protein in order to identify affected pathways underlying the therapeutic effect, and studied lysosomotropic and non-lysosomotropic DAQ-DCpSi effects in 2B/- SMA mice. Treatment of cells caused modest and transient SMN2 mRNA increases with either no change or a decrease in SMNΔ7 and no change in SMN1 transcripts or SMN protein. RNA-Seq analysis of DAQ-DcpSi-treated N2a cells revealed significant changes in expression (both up and down) of approximately 2,000 genes across a broad range of pathways. Treatment of 2B/- SMA mice with both lysomotropic and non-lysosomotropic DAQ-DcpSi compounds had similar effects on disease phenotype indicating that the therapeutic mechanism of action is not a consequence of lysosomotropism. In striking contrast to the findings in vitro, Smn transcripts were robustly changed in tissues but there was no increase in SMN protein levels in spinal cord. We conclude that DAQ-DcpSi have reproducible benefit in SMA mice and a broad spectrum of biological effects in vitro and in vivo, but these are complex, context specific, and not the result of simple SMN2 transcriptional activation.


Subject(s)
Endoribonucleases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Muscular Atrophy, Spinal/drug therapy , Muscular Atrophy, Spinal/enzymology , Quinazolines/pharmacology , Animals , Cell Line , Disease Models, Animal , Enzyme Inhibitors/chemistry , Female , Gene Knockdown Techniques , HEK293 Cells , Humans , Male , Mice , Mice, Knockout , Muscular Atrophy, Spinal/genetics , Promoter Regions, Genetic , Quinazolines/chemistry , RNA, Messenger/genetics , RNA, Messenger/metabolism , Survival of Motor Neuron 2 Protein/deficiency , Survival of Motor Neuron 2 Protein/genetics , Survival of Motor Neuron 2 Protein/metabolism
10.
Antiviral Res ; 72(1): 49-59, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16690141

ABSTRACT

Human papillomaviruses (HPV) cause cutaneous and genital warts. A subset of HPV types is associated with a high-risk for progression to malignancy. The E6 protein from the high-risk HPV types represents an attractive target for intervention because of its roles in viral propagation and cellular transformation. E6 functions in part by interaction with human cellular proteins, several of which possess a helical E6-binding motif. The role for each amino acid in this motif for binding E6 has been tested through structure determination and site-directed mutagenesis. These structural and molecular biological approaches defined the spatial geometry of functional groups necessary for binding to E6. This E6-binding information (the E6-binding pharmacophore) was transferred into a three-dimensional query format suitable for computational screening of large chemical databases. Compounds were identified and tested using in vitro and cell culture-based assays. Several compounds selectively inhibited E6 interaction with the E6-binding protein E6AP and interfered with the ability of E6 to promote p53 degradation. Such compounds provide leads for the development of new pharmacologic agents to treat papillomavirus infections and their associated cancers.


Subject(s)
Drug Design , Oncogene Proteins, Viral/chemistry , Repressor Proteins/chemistry , Antiviral Agents/chemistry , Antiviral Agents/metabolism , Binding Sites , Oncogene Proteins, Viral/metabolism , Papillomaviridae , Protein Binding , Repressor Proteins/metabolism , Zinc Fingers
11.
JCI Insight ; 1(19): e88427, 2016 11 17.
Article in English | MEDLINE | ID: mdl-27882347

ABSTRACT

Spinal muscular atrophy (SMA) is an autosomal recessive neuromuscular disease and one of the leading inherited causes of infant mortality. SMA results from insufficient levels of the survival motor neuron (SMN) protein, and studies in animal models of the disease have shown that increasing SMN protein levels ameliorates the disease phenotype. Our group previously identified and optimized a new series of small molecules, with good potency and toxicity profiles and reasonable pharmacokinetics, that were able to increase SMN protein levels in SMA patient-derived cells. We show here that ML372, a representative of this series, almost doubles the half-life of residual SMN protein expressed from the SMN2 locus by blocking its ubiquitination and subsequent degradation by the proteasome. ML372 increased SMN protein levels in muscle, spinal cord, and brain tissue of SMA mice. Importantly, ML372 treatment improved the righting reflex and extended survival of a severe mouse model of SMA. These results demonstrate that slowing SMN degradation by selectively inhibiting its ubiquitination can improve the motor phenotype and lifespan of SMA model mice.


Subject(s)
Muscular Atrophy, Spinal/drug therapy , Survival of Motor Neuron 1 Protein/chemistry , Ubiquitination , Animals , Disease Models, Animal , HEK293 Cells , Humans , Male , Mice , Mice, Transgenic
12.
Assay Drug Dev Technol ; 12(6): 315-41, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25147906

ABSTRACT

Spinal muscular atrophy (SMA) is an autosomal recessive genetic disorder resulting in degeneration of α-motor neurons of the anterior horn and proximal muscle weakness. It is the leading cause of genetic mortality in children younger than 2 years. It affects ∼1 in 11,000 live births. In 95% of cases, SMA is caused by homozygous deletion of the SMN1 gene. In addition, all patients possess at least one copy of an almost identical gene called SMN2. A single point mutation in exon 7 of the SMN2 gene results in the production of low levels of full-length survival of motor neuron (SMN) protein at amounts insufficient to compensate for the loss of the SMN1 gene. Although no drug treatments are available for SMA, a number of drug discovery and development programs are ongoing, with several currently in clinical trials. This review describes the assays used to identify candidate drugs for SMA that modulate SMN2 gene expression by various means. Specifically, it discusses the use of high-throughput screening to identify candidate molecules from primary screens, as well as the technical aspects of a number of widely used secondary assays to assess SMN messenger ribonucleic acid (mRNA) and protein expression, localization, and function. Finally, it describes the process of iterative drug optimization utilized during preclinical SMA drug development to identify clinical candidates for testing in human clinical trials.


Subject(s)
Drug Discovery/methods , Genetic Therapy/methods , Muscular Atrophy, Spinal/genetics , Muscular Atrophy, Spinal/therapy , Animals , Drug Discovery/trends , Genetic Therapy/trends , High-Throughput Screening Assays/methods , High-Throughput Screening Assays/trends , Humans , Muscular Atrophy, Spinal/diagnosis , Survival of Motor Neuron 2 Protein/genetics
13.
PLoS One ; 8(12): e84506, 2013.
Article in English | MEDLINE | ID: mdl-24376816

ABSTRACT

Expression and function of the human papillomavirus (HPV) early protein 6 (E6) is necessary for viral replication and oncogenesis in cervical cancers. HPV E6 targets the tumor suppressor protein p53 for degradation. To achieve this, "high-risk" HPV E6 proteins bind to and modify the target specificity of the ubiquitin ligase E6AP (E6 associated protein). This E6-dependent loss of p53 enables the virus to bypass host cell defenses and facilitates virally induced activation of the cell cycle progression during viral replication. Disruption of the interaction between E6 and E6AP and stabilization of p53 should decrease viability and proliferation of HPV positive cells. A new in vitro high-throughput binding assay was developed to assay binding between HPV-16 E6 and E6AP and to identify compounds that inhibit this interaction. The compound luteolin emerged from the screen and a library of novel flavones based on its structure was synthesized and characterized using this in vitro binding assay. The compounds identified in this study disrupt the E6/E6AP interaction, increase the levels of p53 and p21(Cip1/Waf1), and decrease proliferation of HPV positive cell lines. The new class of flavonoid E6 inhibitors displays a high degree of specificity for HPV positive cells. Docking analyses suggest that these compounds bind in a hydrophobic pocket at the interface between E6 and E6AP and mimic the leucines in the conserved α-helical motif of E6AP. The activity and specificity of these compounds represent a promising new lead for development as an antiviral therapy in the treatment of HPV infection and cervical cancer.


Subject(s)
Flavonoids/chemistry , Human papillomavirus 16/drug effects , Models, Molecular , Oncogene Proteins, Viral/metabolism , Repressor Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Uterine Cervical Neoplasms/virology , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Drug Discovery , Female , Flavonoids/antagonists & inhibitors , Flavonoids/pharmacology , High-Throughput Screening Assays/methods , Humans , Luteolin/pharmacology , Molecular Structure , Oncogene Proteins, Viral/chemistry , Protein Binding/physiology , Repressor Proteins/chemistry , Tumor Suppressor Protein p53/metabolism , Uterine Cervical Neoplasms/drug therapy
14.
EMBO Mol Med ; 5(7): 1103-18, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23740718

ABSTRACT

Spinal muscular atrophy (SMA) is a neurodegenerative disease that causes progressive muscle weakness, which primarily targets proximal muscles. About 95% of SMA cases are caused by the loss of both copies of the SMN1 gene. SMN2 is a nearly identical copy of SMN1, which expresses much less functional SMN protein. SMN2 is unable to fully compensate for the loss of SMN1 in motor neurons but does provide an excellent target for therapeutic intervention. Increased expression of functional full-length SMN protein from the endogenous SMN2 gene should lessen disease severity. We have developed and implemented a new high-throughput screening assay to identify small molecules that increase the expression of full-length SMN from a SMN2 reporter gene. Here, we characterize two novel compounds that increased SMN protein levels in both reporter cells and SMA fibroblasts and show that one increases lifespan, motor function, and SMN protein levels in a severe mouse model of SMA.


Subject(s)
Drug Discovery , Muscular Atrophy, Spinal/drug therapy , Small Molecule Libraries/therapeutic use , Survival of Motor Neuron 2 Protein/genetics , Up-Regulation/drug effects , Animals , Cells, Cultured , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/pathology , High-Throughput Screening Assays , Humans , Mice , Muscular Atrophy, Spinal/genetics , Muscular Atrophy, Spinal/physiopathology , RNA, Messenger/genetics , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Survival of Motor Neuron 1 Protein/analysis , Survival of Motor Neuron 1 Protein/genetics , Survival of Motor Neuron 2 Protein/analysis
15.
Future Med Chem ; 4(13): 1733-50, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22924510

ABSTRACT

Spinal muscular atrophy (SMA) is an inherited neurodegenerative disease that results in progressive dysfunction of motor neurons of the anterior horn of the spinal cord. SMA is caused by the loss of full-length protein expression from the survival of motor neuron 1 (SMN1) gene. The disease has a unique genetic profile as it is autosomal recessive for the loss of SMN1, but a nearly identical homolog, SMN2, acts as a disease modifier whose expression is inversely correlated to clinical severity. Targeted therapeutic approaches primarily focus on increasing the levels of full-length SMN protein, through either gene replacement or regulation of SMN2 expression. There is currently no US FDA approved treatment for SMA. This is an exciting time as multiple efforts from academic and industrial laboratories are reaching the preclinical and clinical testing stages.


Subject(s)
Genetic Therapy/methods , Molecular Targeted Therapy/methods , Muscular Atrophy, Spinal/genetics , Muscular Atrophy, Spinal/therapy , SMN Complex Proteins/genetics , Animals , Gene Expression Regulation/drug effects , Humans , Motor Neurons/drug effects , Motor Neurons/metabolism , Motor Neurons/pathology , Muscular Atrophy, Spinal/pathology , Neuroprotective Agents/chemistry , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use
16.
J Biomol Screen ; 17(4): 481-95, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22233647

ABSTRACT

Spinal muscular atrophy (SMA) is a neurodegenerative disorder that is characterized by progressive loss of motor neuron function. It is caused by the homozygous loss of the SMN1 (survival of motor neuron 1) gene and a decrease in full-length SMN protein. SMN2 is a nearly identical homolog of SMN1 that, due to alternative splicing, expresses predominantly truncated SMN protein. SMN2 represents an enticing therapeutic target. Increasing expression of full-length SMN from the SMN2 gene might represent a treatment for SMA. We describe a newly designed cell-based reporter assay that faithfully and reproducibly measures full-length SMN expression from the SMN2 gene. This reporter can detect increases of SMN protein by an array of compounds previously shown to regulate SMN2 expression and by the overexpression of proteins that modulate SMN2 splicing. It also can be used to evaluate changes at both the transcriptional and splicing level. This assay can be a valuable tool for the identification of novel compounds that increase SMN2 protein levels and the optimization of compounds already known to modulate SMN2 expression. We present here preliminary data from a high-throughput screen using this assay to identify novel compounds that increase expression of SMN2.


Subject(s)
Genes, Reporter , High-Throughput Screening Assays/methods , Small Molecule Libraries , Survival of Motor Neuron 2 Protein/genetics , Alternative Splicing , Cell Line , Drug Evaluation, Preclinical/methods , Gene Expression , Gene Order , Humans , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Reproducibility of Results , Survival of Motor Neuron 1 Protein/genetics , Survival of Motor Neuron 1 Protein/metabolism , Survival of Motor Neuron 2 Protein/metabolism , Transcription, Genetic
17.
J Med Chem ; 54(18): 6215-33, 2011 Sep 22.
Article in English | MEDLINE | ID: mdl-21819082

ABSTRACT

Spinal muscular atrophy (SMA) is an autosomal recessive disorder affecting the expression or function of survival motor neuron protein (SMN) due to the homozygous deletion or rare point mutations in the survival motor neuron gene 1 (SMN1). The human genome includes a second nearly identical gene called SMN2 that is retained in SMA. SMN2 transcripts undergo alternative splicing with reduced levels of SMN. Up-regulation of SMN2 expression, modification of its splicing, or inhibition of proteolysis of the truncated protein derived from SMN2 have been discussed as potential therapeutic strategies for SMA. In this manuscript, we detail the discovery of a series of arylpiperidines as novel modulators of SMN protein. Systematic hit-to-lead efforts significantly improved potency and efficacy of the series in the primary and orthogonal assays. Structure-property relationships including microsomal stability, cell permeability, and in vivo pharmacokinetics (PK) studies were also investigated. We anticipate that a lead candidate chosen from this series may serve as a useful probe for exploring the therapeutic benefits of SMN protein up-regulation in SMA animal models and a starting point for clinical development.


Subject(s)
Muscular Atrophy, Spinal/metabolism , Piperidines/chemical synthesis , Survival of Motor Neuron 1 Protein/metabolism , Survival of Motor Neuron 2 Protein/metabolism , Thiadiazoles/chemical synthesis , Thiazoles/chemical synthesis , Alternative Splicing , Caco-2 Cells , Cell Membrane Permeability , Drug Design , Exons , Fibroblasts/drug effects , Fibroblasts/metabolism , Genes, Reporter , HEK293 Cells , Humans , Luciferases, Firefly/biosynthesis , Luciferases, Firefly/genetics , Male , Microsomes, Liver/metabolism , Muscular Atrophy, Spinal/drug therapy , Piperidines/pharmacokinetics , Piperidines/pharmacology , Promoter Regions, Genetic , Structure-Activity Relationship , Survival of Motor Neuron 1 Protein/genetics , Survival of Motor Neuron 2 Protein/genetics , Thiadiazoles/pharmacokinetics , Thiadiazoles/pharmacology , Thiazoles/pharmacokinetics , Thiazoles/pharmacology , Transcription, Genetic
18.
J Mol Biol ; 386(4): 1123-37, 2009 Mar 06.
Article in English | MEDLINE | ID: mdl-19244625

ABSTRACT

E6 is an oncoprotein produced by human papillomavirus (HPV). The E6 protein from high-risk HPV type 16 contains two zinc-binding domains with two C-x-x-C motifs each. E6 exerts its transforming functions through formation of a complex with E6AP, which binds p53 and stimulates its degradation. There have been few biophysical and structural studies due to difficulty in preparation of soluble protein; here we describe the preparation of soluble E6 constructs including the two separated zinc-binding domains of E6. These proteins are used to examine the extent to which the two domains cooperate to mediate E6 function, how zinc influences the behavior of E6 protein, and which domains mediate aggregation. We demonstrate, using p53 degradation, E6AP binding, and hDlg (human homolog of the Drosophila discs large tumor suppressor protein) PDZ (postsynaptic density/disc large/zonula occludens) protein binding assays, that these soluble proteins are active, and, using NMR, circular dichroism, and fluorescence spectroscopies, we show that they are folded and stable. We show that the separated N-terminal and C-terminal domains interact, but nonproductively, for E6 function. The two domains bind zinc differently with higher affinity associated with the C-terminal domain. Analyses using surface plasmon resonance and circular dichroism and fluorescence spectroscopies show that aggregation is mediated more through the N-terminal domain than through the C-terminal domain. These studies allow a model in which the C-terminal zinc-binding domain of E6 recruits a target protein such as hDlg and the N-terminal domain is mostly responsible for recruiting a ubiquitin ligase to mediate target protein degradation.


Subject(s)
Oncogene Proteins, Viral/metabolism , Repressor Proteins/metabolism , Magnetic Resonance Spectroscopy , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Oncogene Proteins, Viral/chemistry , Oncogene Proteins, Viral/isolation & purification , Protein Binding/drug effects , Protein Denaturation/drug effects , Protein Stability/drug effects , Protein Structure, Quaternary , Protein Structure, Secondary , Protein Structure, Tertiary , Recombinant Fusion Proteins , Repressor Proteins/chemistry , Repressor Proteins/isolation & purification , Solubility/drug effects , Temperature , Urea/pharmacology , Zinc/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL