Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Sci Signal ; 17(844): eadn6052, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38980922

ABSTRACT

Inhibitors of the transforming growth factor-ß (TGF-ß) pathway are potentially promising antifibrotic therapies, but nonselective simultaneous inhibition of all three TGF-ß homologs has safety liabilities. TGF-ß1 is noncovalently bound to a latency-associated peptide that is, in turn, covalently bound to different presenting molecules within large latent complexes. The latent TGF-ß-binding proteins (LTBPs) present TGF-ß1 in the extracellular matrix, and TGF-ß1 is presented on immune cells by two transmembrane proteins, glycoprotein A repetitions predominant (GARP) and leucine-rich repeat protein 33 (LRRC33). Here, we describe LTBP-49247, an antibody that selectively bound to and inhibited the activation of TGF-ß1 presented by LTBPs but did not bind to TGF-ß1 presented by GARP or LRRC33. Structural studies demonstrated that LTBP-49247 recognized an epitope on LTBP-presented TGF-ß1 that is not accessible on GARP- or LRRC33-presented TGF-ß1, explaining the antibody's selectivity for LTBP-complexed TGF-ß1. In two rodent models of kidney fibrosis of different etiologies, LTBP-49247 attenuated fibrotic progression, indicating the central role of LTBP-presented TGF-ß1 in renal fibrosis. In mice, LTBP-49247 did not have the toxic effects associated with less selective TGF-ß inhibitors. These results establish the feasibility of selectively targeting LTBP-bound TGF-ß1 as an approach for treating fibrosis.


Subject(s)
Extracellular Matrix , Fibrosis , Latent TGF-beta Binding Proteins , Transforming Growth Factor beta1 , Transforming Growth Factor beta1/metabolism , Animals , Humans , Latent TGF-beta Binding Proteins/metabolism , Latent TGF-beta Binding Proteins/antagonists & inhibitors , Extracellular Matrix/metabolism , Mice , Male , Kidney Diseases/metabolism , Kidney Diseases/pathology , Kidney Diseases/drug therapy , Disease Progression , Kidney/pathology , Kidney/metabolism , Kidney/drug effects , Mice, Inbred C57BL
2.
Sci Transl Med ; 12(536)2020 03 25.
Article in English | MEDLINE | ID: mdl-32213632

ABSTRACT

Despite breakthroughs achieved with cancer checkpoint blockade therapy (CBT), many patients do not respond to anti-programmed cell death-1 (PD-1) due to primary or acquired resistance. Human tumor profiling and preclinical studies in tumor models have recently uncovered transforming growth factor-ß (TGFß) signaling activity as a potential point of intervention to overcome primary resistance to CBT. However, the development of therapies targeting TGFß signaling has been hindered by dose-limiting cardiotoxicities, possibly due to nonselective inhibition of multiple TGFß isoforms. Analysis of mRNA expression data from The Cancer Genome Atlas revealed that TGFΒ1 is the most prevalent TGFß isoform expressed in many types of human tumors, suggesting that TGFß1 may be a key contributor to primary CBT resistance. To test whether selective TGFß1 inhibition is sufficient to overcome CBT resistance, we generated a high-affinity, fully human antibody, SRK-181, that selectively binds to latent TGFß1 and inhibits its activation. Coadministration of SRK-181-mIgG1 and an anti-PD-1 antibody in mice harboring syngeneic tumors refractory to anti-PD-1 treatment induced profound antitumor responses and survival benefit. Specific targeting of TGFß1 was also effective in tumors expressing more than one TGFß isoform. Combined SRK-181-mIgG1 and anti-PD-1 treatment resulted in increased intratumoral CD8+ T cells and decreased immunosuppressive myeloid cells. No cardiac valvulopathy was observed in a 4-week rat toxicology study with SRK-181, suggesting that selectively blocking TGFß1 activation may avoid dose-limiting toxicities previously observed with pan-TGFß inhibitors. These results establish a rationale for exploring selective TGFß1 inhibition to overcome primary resistance to CBT.


Subject(s)
Neoplasms , Transforming Growth Factor beta/antagonists & inhibitors , Animals , CD8-Positive T-Lymphocytes , Cardiotoxicity , Cell Line, Tumor , Humans , Mice , Neoplasms/drug therapy , Rats , Signal Transduction
3.
Hum Mol Genet ; 28(7): 1076-1089, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30481286

ABSTRACT

Spinal muscular atrophy (SMA) is a neuromuscular disease characterized by loss of α-motor neurons, leading to profound skeletal muscle atrophy. Patients also suffer from decreased bone mineral density and increased fracture risk. The majority of treatments for SMA, approved or in clinic trials, focus on addressing the underlying cause of disease, insufficient production of full-length SMN protein. While restoration of SMN has resulted in improvements in functional measures, significant deficits remain in both mice and SMA patients following treatment. Motor function in SMA patients may be additionally improved by targeting skeletal muscle to reduce atrophy and improve muscle strength. Inhibition of myostatin, a negative regulator of muscle mass, offers a promising approach to increase muscle function in SMA patients. Here we demonstrate that muSRK-015P, a monoclonal antibody which specifically inhibits myostatin activation, effectively increases muscle mass and function in two variants of the pharmacological mouse model of SMA in which pharmacologic restoration of SMN has taken place either 1 or 24 days after birth to reflect early or later therapeutic intervention. Additionally, muSRK-015P treatment improves the cortical and trabecular bone phenotypes in these mice. These data indicate that preventing myostatin activation has therapeutic potential in addressing muscle and bone deficiencies in SMA patients. An optimized variant of SRK-015P, SRK-015, is currently in clinical development for treatment of SMA.


Subject(s)
Muscular Atrophy, Spinal/genetics , Myostatin/genetics , Myostatin/physiology , Animals , Antibodies, Monoclonal , Disease Models, Animal , Mice , Motor Neurons/metabolism , Muscle Strength/physiology , Muscle, Skeletal/metabolism , Muscular Atrophy, Spinal/physiopathology , Myostatin/antagonists & inhibitors , Survival of Motor Neuron 1 Protein/genetics , Survival of Motor Neuron 2 Protein/genetics
4.
Sci Signal ; 11(541)2018 07 31.
Article in English | MEDLINE | ID: mdl-30065029

ABSTRACT

Members of the family of nuclear factor κB (NF-κB) transcription factors are critical for multiple cellular processes, including regulating innate and adaptive immune responses, cell proliferation, and cell survival. Canonical NF-κB complexes are retained in the cytoplasm by the inhibitory protein IκBα, whereas noncanonical NF-κB complexes are retained by p100. Although activation of canonical NF-κB signaling through the IκBα kinase complex is well studied, few regulators of the NF-κB-inducing kinase (NIK)-dependent processing of noncanonical p100 to p52 and the subsequent nuclear translocation of p52 have been identified. We discovered a role for cyclin-dependent kinase 12 (CDK12) in transcriptionally regulating the noncanonical NF-κB pathway. High-content phenotypic screening identified the compound 919278 as a specific inhibitor of the lymphotoxin ß receptor (LTßR), and tumor necrosis factor (TNF) receptor superfamily member 12A (FN14)-dependent nuclear translocation of p52, but not of the TNF-α receptor-mediated nuclear translocation of p65. Chemoproteomics identified CDK12 as the target of 919278. CDK12 inhibition by 919278, the CDK inhibitor THZ1, or siRNA-mediated knockdown resulted in similar global transcriptional changes and prevented the LTßR- and FN14-dependent expression of MAP3K14 (which encodes NIK) as well as NIK accumulation by reducing phosphorylation of the carboxyl-terminal domain of RNA polymerase II. By coupling a phenotypic screen with chemoproteomics, we identified a pathway for the activation of the noncanonical NF-κB pathway that could serve as a therapeutic target in autoimmunity and cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Cyclin-Dependent Kinases/metabolism , Gene Expression Regulation, Neoplastic , NF-kappa B/metabolism , Osteosarcoma/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Bone Neoplasms/drug therapy , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclin-Dependent Kinases/genetics , Cyclins/genetics , Cyclins/metabolism , Gene Expression Profiling , High-Throughput Screening Assays , Humans , Indoles/pharmacology , Lymphotoxin beta Receptor/antagonists & inhibitors , Lymphotoxin beta Receptor/genetics , Lymphotoxin beta Receptor/metabolism , NF-kappa B/antagonists & inhibitors , NF-kappa B/genetics , NF-kappa B p52 Subunit/genetics , NF-kappa B p52 Subunit/metabolism , Osteosarcoma/drug therapy , Osteosarcoma/pathology , Propionates/pharmacology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Proteome , Signal Transduction , TWEAK Receptor/antagonists & inhibitors , TWEAK Receptor/genetics , TWEAK Receptor/metabolism , Tumor Cells, Cultured , NF-kappaB-Inducing Kinase
5.
Chem Biol ; 22(9): 1228-37, 2015 Sep 17.
Article in English | MEDLINE | ID: mdl-26364931

ABSTRACT

In an attempt to identify novel therapeutics and mechanisms to differentially kill tumor cells using phenotypic screening, we identified N-benzyl indole carbinols (N-BICs), synthetic analogs of the natural product indole-3-carbinol (I3C). To understand the mode of action for the molecules we employed Cancer Cell Line Encyclopedia viability profiling and correlative informatics analysis to identify and ultimately confirm the phase II metabolic enzyme sulfotransferase 1A1 (SULT1A1) as the essential factor for compound selectivity. Further studies demonstrate that SULT1A1 activates the N-BICs by rendering the compounds strong electrophiles which can alkylate cellular proteins and thereby induce cell death. This study demonstrates that the selectivity profile for N-BICs is through conversion by SULT1A1 from an inactive prodrug to an active species that induces cell death and tumor suppression.


Subject(s)
Arylsulfotransferase/metabolism , Benzyl Compounds/pharmacology , Indoles/pharmacology , Animals , Benzyl Compounds/pharmacokinetics , Cell Line, Tumor , Cell Survival/drug effects , Drug Screening Assays, Antitumor , Female , HCT116 Cells , Humans , Indoles/pharmacokinetics , Mice , Mice, Nude , Random Allocation , Xenograft Model Antitumor Assays
6.
J Biomol Screen ; 19(5): 791-802, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24518063

ABSTRACT

Gene-expression data are often used to infer pathways regulating transcriptional responses. For example, differentially expressed genes (DEGs) induced by compound treatment can help characterize hits from phenotypic screens, either by correlation with known drug signatures or by pathway enrichment. Pathway enrichment is, however, typically computed with DEGs rather than "upstream" nodes that are potentially causal of "downstream" changes. Here, we present graph-based models to predict causal targets from compound-microarray data. We test several approaches to traversing network topology, and show that a consensus minimum-rank score (SigNet) beat individual methods and could highly rank compound targets among all network nodes. In addition, larger, less canonical networks outperformed linear canonical interactions. Importantly, pathway enrichment using causal nodes rather than DEGs recovers relevant pathways more often. To further validate our approach, we used integrated data sets from the Cancer Genome Atlas to identify driving pathways in triple-negative breast cancer. Critical pathways were uncovered, including the epidermal growth factor receptor 2-phosphatidylinositide 3-kinase-AKT-MAPK growth pathway andATR-p53-BRCA DNA damage pathway, in addition to unexpected pathways, such as TGF-WNT cytoskeleton remodeling, IL12-induced interferon gamma production, and TNFR-IAP (inhibitor of apoptosis) apoptosis; the latter was validated by pooled small hairpin RNA profiling in cancer cells. Overall, our approach can bridge transcriptional profiles to compound targets and driving pathways in cancer.


Subject(s)
Drug Discovery , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Neoplasms/drug therapy , Triple Negative Breast Neoplasms/drug therapy , Female , Humans , Models, Theoretical , Neoplasms/metabolism , Neoplasms/pathology , Oligonucleotide Array Sequence Analysis , Phenotype , RNA, Messenger/metabolism , ROC Curve , Signal Transduction , Triple Negative Breast Neoplasms/metabolism
7.
J Biomol Screen ; 18(4): 407-19, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23150017

ABSTRACT

Translation initiation is a fine-tuned process that plays a critical role in tumorigenesis. The use of small molecules that modulate mRNA translation provides tool compounds to explore the mechanism of translational initiation and to further validate protein synthesis as a potential pharmaceutical target for cancer therapeutics. This report describes the development and use of a click beetle, dual luciferase cell-based assay multiplexed with a measure of compound toxicity using resazurin to evaluate the differential effect of natural products on cap-dependent or internal ribosome entry site (IRES)-mediated translation initiation and cell viability. This screen identified a series of cardiac glycosides as inhibitors of IRES-mediated translation using, in particular, the oncogene mRNA c-Myc IRES. Treatment of c-Myc-dependent cancer cells with these compounds showed a decrease in c-Myc protein associated with a significant modulation of cell viability. These findings suggest that inhibition of IRES-mediated translation initiation may be a strategy to inhibit c-Myc-driven tumorigenesis.


Subject(s)
Cardiac Glycosides/analysis , Cardiac Glycosides/pharmacology , Drug Evaluation, Preclinical , Protein Biosynthesis/drug effects , Protein Synthesis Inhibitors/pharmacology , Proto-Oncogene Proteins c-myc/metabolism , Ribosomes/metabolism , Apoptosis/drug effects , Base Sequence , Biological Assay , Cardiac Glycosides/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cymarine/chemistry , Cymarine/pharmacology , DNA Damage , Genes, Reporter , HEK293 Cells , Humans , Inhibitory Concentration 50 , Protein Synthesis Inhibitors/analysis , Protein Synthesis Inhibitors/chemistry , Proto-Oncogene Proteins c-myc/antagonists & inhibitors , Ribosomes/drug effects , Vascular Endothelial Growth Factor A/metabolism
8.
Cancer Discov ; 2(10): 948-59, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22874768

ABSTRACT

The overall power of kinase inhibitors is substantially overshadowed by the acquisition of drug resistance. To address this issue, we systematically assessed the potential of secreted proteins to induce resistance to kinase inhibitors. To this end, we developed a high-throughput platform for screening a cDNA library encoding 3,432 secreted proteins in cellular assays. Using cancer cells originally dependent on either MET, FGFR2, or FGFR3, we observed a bypass of dependence through ligand-mediated activation of alternative receptor tyrosine kinases (RTK). Our findings indicate a broad and versatile potential for RTKs from the HER and FGFR families as well as MET to compensate for loss of each other. We further provide evidence that combined inhibition of simultaneously active RTKs can lead to an added anticancer effect.


Subject(s)
Neoplasms/metabolism , Proto-Oncogene Proteins c-met/metabolism , Receptor, ErbB-2/metabolism , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Receptor, Fibroblast Growth Factor, Type 3/metabolism , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Resistance, Neoplasm/genetics , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , ErbB Receptors/metabolism , High-Throughput Screening Assays , Humans , Mice , Mutation , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/pathology , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Receptor, ErbB-2/genetics , Signal Transduction/drug effects , Transplantation, Heterologous
9.
Breast Cancer Res ; 10(2): R33, 2008.
Article in English | MEDLINE | ID: mdl-18430202

ABSTRACT

INTRODUCTION: Heat shock protein 90 (HSP90) is a key component of a multichaperone complex involved in the post-translational folding of a large number of client proteins, many of which play essential roles in tumorigenesis. HSP90 has emerged in recent years as a promising new target for anticancer therapies. METHODS: The concentrations of the HSP90 inhibitor NVP-AUY922 required to reduce cell numbers by 50% (GI50 values) were established in a panel of breast cancer cell lines and patient-derived human breast tumors. To investigate the properties of the compound in vivo, the pharmacokinetic profile, antitumor effect, and dose regimen were established in a BT-474 breast cancer xenograft model. The effect on HSP90-p23 complexes, client protein degradation, and heat shock response was investigated in cell culture and breast cancer xenografts by immunohistochemistry, Western blot analysis, and immunoprecipitation. RESULTS: We show that the novel small molecule HSP90 inhibitor NVP-AUY922 potently inhibits the proliferation of human breast cancer cell lines with GI50 values in the range of 3 to 126 nM. NVP-AUY922 induced proliferative inhibition concurrent with HSP70 upregulation and client protein depletion--hallmarks of HSP90 inhibition. Intravenous acute administration of NVP-AUY922 to athymic mice (30 mg/kg) bearing subcutaneous BT-474 breast tumors resulted in drug levels in excess of 1,000 times the cellular GI50 value for about 2 days. Significant growth inhibition and good tolerability were observed when the compound was administered once per week. Therapeutic effects were concordant with changes in pharmacodynamic markers, including HSP90-p23 dissociation, decreases in ERBB2 and P-AKT, and increased HSP70 protein levels. CONCLUSION: NVP-AUY922 is a potent small molecule HSP90 inhibitor showing significant activity against breast cancer cells in cellular and in vivo settings. On the basis of its mechanism of action, preclinical activity profile, tolerability, and pharmaceutical properties, the compound recently has entered clinical phase I breast cancer trials.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Isoxazoles/pharmacology , Resorcinols/pharmacology , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacokinetics , Blotting, Western , Breast Neoplasms/metabolism , Cell Line, Tumor , Disease Models, Animal , Female , Gene Expression Regulation, Neoplastic/drug effects , HSP70 Heat-Shock Proteins/metabolism , Humans , Immunohistochemistry , Immunoprecipitation , Isoxazoles/administration & dosage , Isoxazoles/pharmacokinetics , Mice , Mice, Nude , Molecular Chaperones , Proto-Oncogene Proteins c-akt/metabolism , Receptor, ErbB-2/metabolism , Receptors, Estrogen/metabolism , Resorcinols/administration & dosage , Resorcinols/pharmacokinetics , Transplantation, Heterologous , Up-Regulation/drug effects
10.
J Proteome Res ; 7(2): 640-7, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18186600

ABSTRACT

While numerous strategies exist for biomarker discovery, the bottleneck to product development and routine use at the clinic is in the verification phase of candidate biomarkers. The aim of this study was to establish a robust and high-throughput product ion monitoring (PIM) assay that is potentially capable of rapidly verifying candidates from discovery phase experiments. Using prostate-specific antigen (PSA), a model biomarker, and a routinely used mass spectrometer for discovery platforms, an ion trap (LTQ, Thermo), the utility of this instrument to perform PIM was explored. The proteotypic doubly charged intact peptide LSEPAELTDAVK ( m/ z 637) fragmenting to m/ z 943 (PAELTDAVK) was monitored. A limit of detection of 10 attomoles with a coefficient of variation (CV) of <20% was obtained for a purified recombinant PSA digest. Immunoextraction of endogenous PSA from serum using a monoclonal antibody on a 96-well microtiter plate, followed by PIM on the LTQ, enabled quantification of PSA down to less than 1 ng/mL with a limit of detection of 0.1 ng/mL and CVs < 20%. Mascot searching and ion ratio confirmation further supported the conclusion that the quantified moiety in serum was the PSA peptide. We conclude that this methodology could be adapted quickly and easily to other candidates, thus providing a much needed technology to bridge the gap between discovery and validation platforms.


Subject(s)
Ions , Prostate-Specific Antigen/analysis , Prostate-Specific Antigen/blood , Serum/chemistry , Amino Acid Sequence , Biomarkers , Humans , Male , Molecular Sequence Data , Peptides/analysis , Peptides/blood , Peptides/chemistry , Prostate-Specific Antigen/chemistry , Prostatic Neoplasms/blood , Tandem Mass Spectrometry
11.
Mutat Res ; 602(1-2): 43-53, 2006 Dec 01.
Article in English | MEDLINE | ID: mdl-17045619

ABSTRACT

The high mobility group A2 protein (HMGA2) has been implicated in the pathogenesis of mesenchymal tumors such as leiomyoma, lipoma and hamartoma. HMGA2 was pinpointed by mapping the breakpoints in the chromosomal translocations in 12q15, especially the t(12;14) that is commonly seen in uterine leiomyoma. It is generally assumed that altered expression of HMGA2 is an early event in the pathway to tumor formation. Here, we show evidence that three novel transcripts, A15, B6 and D12 are located within the HMGA2 gene itself and are transcribed from the opposite strand. These embedded transcripts are expressed at 6-20-fold higher levels in tumors compared to matched myometrium from the same patients. We estimate that the domain of increased expression extends 500kb on chromosome 12q15, and encompasses the majority of t(12;14) translocation breakpoints. However, a corresponding domain of consistently altered expression is not seen on chromosome 14 or outside of the chromosome 12 multiple aberration region. These data suggest that t(12;14) breakpoints contribute to the pathogenesis of uterine leiomyoma by interrupting a complex regulation of HMGA2 and other genes embedded within and around it. We also discovered a novel laminin receptor gene, transcribed from the opposite strand, within the promoter region of HMGA2. Although the roles for these embedded transcripts are still unknown, preliminary data suggest that they are members of the family of non-coding RNA and that they may play an important role in the pathology of uterine leiomyoma.


Subject(s)
Chromosomes, Human, Pair 12 , Chromosomes, Human, Pair 14 , HMGA2 Protein/genetics , Leiomyoma/genetics , Translocation, Genetic , Uterine Neoplasms/genetics , Alternative Splicing , Amino Acid Sequence , Base Sequence , Expressed Sequence Tags , Female , HMGA2 Protein/metabolism , Humans , Leiomyoma/metabolism , Leiomyoma/pathology , Models, Genetic , Molecular Sequence Data , Myometrium/metabolism , Promoter Regions, Genetic , Receptors, Laminin/genetics , Uterine Neoplasms/metabolism
12.
EMBO J ; 23(8): 1749-60, 2004 Apr 21.
Article in English | MEDLINE | ID: mdl-15057271

ABSTRACT

Receptor-mediated increases in the concentration of intracellular free calcium ([Ca2+]i) are responsible for controlling a plethora of physiological processes including gene expression, secretion, contraction, proliferation, neural signalling, and learning. Increases in [Ca2+]i often occur as repetitive Ca2+ spikes or oscillations. Induced by electrical or receptor stimuli, these repetitive Ca2+ spikes increase their frequency with the amplitude of the receptor stimuli, a phenomenon that appears critical for the induction of selective cellular functions. Here we report the characterisation of RASAL, a Ras GTPase-activating protein that senses the frequency of repetitive Ca2+ spikes by undergoing synchronous oscillatory associations with the plasma membrane. Importantly, we show that only during periods of plasma membrane association does RASAL inactivate Ras signalling. Thus, RASAL senses the frequency of complex Ca2+ signals, decoding them through a regulation of the activation state of Ras. Our data provide a hitherto unrecognised link between complex Ca2+ signals and the regulation of Ras.


Subject(s)
Calcium Signaling , Calcium/metabolism , ras GTPase-Activating Proteins/metabolism , Amino Acid Sequence , Animals , Cell Line , Cell Membrane/metabolism , Chlorocebus aethiops , Electrophysiology , Enzyme Activation , Humans , Liposomes/metabolism , Molecular Sequence Data , Phosphatidylcholines/metabolism , Phosphatidylserines/metabolism , Protein Binding , Protein Structure, Tertiary , Protein Transport , RNA, Small Interfering/genetics , Sequence Alignment , ras GTPase-Activating Proteins/chemistry , ras GTPase-Activating Proteins/genetics , ras Proteins/metabolism
13.
Somat Cell Mol Genet ; 20(1): 27-38, 1994 01.
Article in English | MEDLINE | ID: mdl-8197474

ABSTRACT

Huntington's disease, a neurodegenerative disorder characterized by loss of striatal neurons, is caused by an expanded, unstable trinucleotide repeat in a novel 4p16.3 gene. To lay the foundation for exploring the pathogenic mechanism in HD, we have determined the structure of the disease gene and examined its expression. The HD locus spans 180 kb and consists of 67 exons ranging in size from 48 bp to 341 bp with an average of 138 bp. Scanning of the HD transcript failed to reveal any additional sequence alterations characteristic of HD chromosomes. A codon loss polymorphism in linkage disequilibrium with the disorder revealed that both normal and HD alleles are represented in the mRNA population in HD heterozygotes, indicating that the defect does not eliminate transcription. The gene is ubiquitously expressed as two alternatively polyadenylated forms displaying different relative abundance in various fetal and adult tissues, suggesting the operation of interacting factors in determining specificity of cell loss. The HD gene was disrupted in a female carrying a balanced translocation with a breakpoint between exons 40 and 41. The absence of any abnormal phenotype in this individual argues against simple inactivation of the gene as the mechanism by which the expanded trinucleotide repeat causes HD. Taken together, these observations suggest that the dominant HD mutation either confers a new property on the mRNA or, more likely, alters an interaction at the protein level.


Subject(s)
Gene Expression , Huntington Disease/genetics , Repetitive Sequences, Nucleic Acid , Adult , Alleles , Base Sequence , Cell Line , Codon , DNA, Complementary , Exons , Female , Fetal Diseases/genetics , Humans , Huntington Disease/embryology , Introns , Molecular Sequence Data , Polymorphism, Genetic , RNA, Messenger/metabolism , Translocation, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...