Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
ACS Chem Biol ; 19(4): 938-952, 2024 04 19.
Article in English | MEDLINE | ID: mdl-38565185

ABSTRACT

Phenotypic assays have become an established approach to drug discovery. Greater disease relevance is often achieved through cellular models with increased complexity and more detailed readouts, such as gene expression or advanced imaging. However, the intricate nature and cost of these assays impose limitations on their screening capacity, often restricting screens to well-characterized small compound sets such as chemogenomics libraries. Here, we outline a cheminformatics approach to identify a small set of compounds with likely novel mechanisms of action (MoAs), expanding the MoA search space for throughput limited phenotypic assays. Our approach is based on mining existing large-scale, phenotypic high-throughput screening (HTS) data. It enables the identification of chemotypes that exhibit selectivity across multiple cell-based assays, which are characterized by persistent and broad structure activity relationships (SAR). We validate the effectiveness of our approach in broad cellular profiling assays (Cell Painting, DRUG-seq, and Promotor Signature Profiling) and chemical proteomics experiments. These experiments revealed that the compounds behave similarly to known chemogenetic libraries, but with a notable bias toward novel protein targets. To foster collaboration and advance research in this area, we have curated a public set of such compounds based on the PubChem BioAssay dataset and made it available for use by the scientific community.


Subject(s)
Drug Discovery , High-Throughput Screening Assays , Small Molecule Libraries , Drug Discovery/methods , High-Throughput Screening Assays/methods , Cheminformatics/methods , Small Molecule Libraries/chemistry , Structure-Activity Relationship
2.
ACS Chem Biol ; 17(6): 1401-1414, 2022 06 17.
Article in English | MEDLINE | ID: mdl-35508359

ABSTRACT

Unbiased transcriptomic RNA-seq data has provided deep insights into biological processes. However, its impact in drug discovery has been narrow given high costs and low throughput. Proof-of-concept studies with Digital RNA with pertUrbation of Genes (DRUG)-seq demonstrated the potential to address this gap. We extended the DRUG-seq platform by subjecting it to rigorous testing and by adding an open-source analysis pipeline. The results demonstrate high reproducibility and ability to resolve the mechanism(s) of action for a diverse set of compounds. Furthermore, we demonstrate how this data can be incorporated into a drug discovery project aiming to develop therapeutics for schizophrenia using human stem cell-derived neurons. We identified both an on-target activation signature, induced by a set of chemically distinct positive allosteric modulators of the N-methyl-d-aspartate (NMDA) receptor, and independent off-target effects. Overall, the protocol and open-source analysis pipeline are a step toward industrializing RNA-seq for high-complexity transcriptomics studies performed at a saturating scale.


Subject(s)
Drug Discovery , Transcriptome , Drug Discovery/methods , Humans , RNA , Reproducibility of Results , Sequence Analysis, RNA/methods
3.
Cell Rep ; 27(2): 616-630.e6, 2019 04 09.
Article in English | MEDLINE | ID: mdl-30970262

ABSTRACT

Human pluripotent stem cells (hPSCs) generate a variety of disease-relevant cells that can be used to improve the translation of preclinical research. Despite the potential of hPSCs, their use for genetic screening has been limited by technical challenges. We developed a scalable and renewable Cas9 and sgRNA-hPSC library in which loss-of-function mutations can be induced at will. Our inducible mutant hPSC library can be used for multiple genome-wide CRISPR screens in a variety of hPSC-induced cell types. As proof of concept, we performed three screens for regulators of properties fundamental to hPSCs: their ability to self-renew and/or survive (fitness), their inability to survive as single-cell clones, and their capacity to differentiate. We identified the majority of known genes and pathways involved in these processes, as well as a plethora of genes with unidentified roles. This resource will increase the understanding of human development and genetics. This approach will be a powerful tool to identify disease-modifying genes and pathways.


Subject(s)
CRISPR-Cas Systems/genetics , Genetic Testing/methods , Genome/genetics , Pluripotent Stem Cells/metabolism , Humans
4.
Nat Commun ; 9(1): 4307, 2018 10 17.
Article in English | MEDLINE | ID: mdl-30333485

ABSTRACT

Here we report Digital RNA with pertUrbation of Genes (DRUG-seq), a high-throughput platform for drug discovery. Pharmaceutical discovery relies on high-throughput screening, yet current platforms have limited readouts. RNA-seq is a powerful tool to investigate drug effects using transcriptome changes as a proxy, yet standard library construction is costly. DRUG-seq captures transcriptional changes detected in standard RNA-seq at 1/100th the cost. In proof-of-concept experiments profiling 433 compounds across 8 doses, transcription profiles generated from DRUG-seq successfully grouped compounds into functional clusters by mechanism of actions (MoAs) based on their intended targets. Perturbation differences reflected in transcriptome changes were detected for compounds engaging the same target, demonstrating the value of using DRUG-seq for understanding on and off-target activities. We demonstrate DRUG-seq captures common mechanisms, as well as differences between compound treatment and CRISPR on the same target. DRUG-seq provides a powerful tool for comprehensive transcriptome readout in a high-throughput screening environment.


Subject(s)
Drug Discovery/methods , Gene Expression Profiling/methods , High-Throughput Screening Assays/methods , Sequence Analysis, RNA , Cell Line , Clustered Regularly Interspaced Short Palindromic Repeats , Humans
5.
Neurosci Lett ; 660: 109-114, 2017 Nov 01.
Article in English | MEDLINE | ID: mdl-28923481

ABSTRACT

Triggering receptor expressed in myeloid cells (TREM2) is a member of the immunoglobulin superfamily and is expressed in macrophages, dendritic cells, microglia, and osteoclasts. TREM2 plays a role in phagocytosis, regulates release of cytokine, contributes to microglia maintenance, and its ectodomain is shed from the cell surface. Here, the question was addressed at which position sheddases cleave TREM2 and what are the proteases involved in this process. Using both pharmacological and genetic approaches we report that the main protease contributing to the release of TREM2 ectodomain is ADAM17, (a disintegrin and metalloproteinase domain containing protein, also called TACE, TNFα converting enzyme) while ADAM10 plays a minor role. Complementary biochemical experiments reveal that cleavage occurs between histidine 157 and serine 158. Shedding is not altered for the R47H-mutated TREM2 protein that confers an increased risk for the development of Alzheimers disease. These findings reveal a link between shedding of TREM2 and its regulation during inflammatory conditions or chronic neurodegenerative disease like AD in which activity or expression of sheddases might be altered.


Subject(s)
ADAM17 Protein/metabolism , Histidine/metabolism , Membrane Glycoproteins/metabolism , Receptors, Immunologic/metabolism , ADAM10 Protein/metabolism , Amyloid Precursor Protein Secretases/metabolism , Animals , CHO Cells , Cell Line , Cricetulus , Humans , Membrane Proteins/metabolism
6.
Cell Stem Cell ; 19(6): 703-708, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27912091

ABSTRACT

Zika virus (ZIKV) can cross the placental barrier, resulting in infection of the fetal brain and neurological defects including microcephaly. The cellular tropism of ZIKV and the identity of attachment factors used by the virus to gain access to key cell types involved in pathogenesis are under intense investigation. Initial studies suggested that ZIKV preferentially targets neural progenitor cells (NPCs), providing an explanation for the developmental phenotypes observed in some pregnancies. The AXL protein has been nominated as a key attachment factor for ZIKV in several cell types including NPCs. However, here we show that genetic ablation of AXL has no effect on ZIKV entry or ZIKV-mediated cell death in human induced pluripotent stem cell (iPSC)-derived NPCs or cerebral organoids. These findings call into question the utility of AXL inhibitors for preventing birth defects after infection and suggest that further studies of viral attachment factors in NPCs are needed.


Subject(s)
Cerebrum/pathology , Gene Deletion , Neural Stem Cells/metabolism , Neural Stem Cells/virology , Neuroprotection , Organoids/virology , Proto-Oncogene Proteins/genetics , Receptor Protein-Tyrosine Kinases/genetics , Zika Virus Infection/prevention & control , Cell Death , Gene Knockout Techniques , Humans , Neural Stem Cells/pathology , Organoids/metabolism , Organoids/pathology , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Zika Virus Infection/pathology , Axl Receptor Tyrosine Kinase
7.
Genome Med ; 8(1): 5, 2016 Jan 19.
Article in English | MEDLINE | ID: mdl-26803900

ABSTRACT

BACKGROUND: Alzheimer's disease affects ~13% of people in the United States 65 years and older, making it the most common neurodegenerative disorder. Recent work has identified roles for environmental, genetic, and epigenetic factors in Alzheimer's disease risk. METHODS: We performed a genome-wide screen of DNA methylation using the Illumina Infinium HumanMethylation450 platform on bulk tissue samples from the superior temporal gyrus of patients with Alzheimer's disease and non-demented controls. We paired a sliding window approach with multivariate linear regression to characterize Alzheimer's disease-associated differentially methylated regions (DMRs). RESULTS: We identified 479 DMRs exhibiting a strong bias for hypermethylated changes, a subset of which were independently associated with aging. DMR intervals overlapped 475 RefSeq genes enriched for gene ontology categories with relevant roles in neuron function and development, as well as cellular metabolism, and included genes reported in Alzheimer's disease genome-wide and epigenome-wide association studies. DMRs were enriched for brain-specific histone signatures and for binding motifs of transcription factors with roles in the brain and Alzheimer's disease pathology. Notably, hypermethylated DMRs preferentially overlapped poised promoter regions, marked by H3K27me3 and H3K4me3, previously shown to co-localize with aging-associated hypermethylation. Finally, the integration of DMR-associated single nucleotide polymorphisms with Alzheimer's disease genome-wide association study risk loci and brain expression quantitative trait loci highlights multiple potential DMRs of interest for further functional analysis. CONCLUSION: We have characterized changes in DNA methylation in the superior temporal gyrus of patients with Alzheimer's disease, highlighting novel loci that facilitate better characterization of pathways and mechanisms underlying Alzheimer's disease pathogenesis, and improve our understanding of epigenetic signatures that may contribute to the development of disease.


Subject(s)
Alzheimer Disease/genetics , DNA Methylation , Gene Regulatory Networks , Genome-Wide Association Study/methods , Aged , Aged, 80 and over , Epigenesis, Genetic , Gene Ontology , Humans , Temporal Lobe/metabolism
8.
Antioxid Redox Signal ; 16(9): 855-68, 2012 May 01.
Article in English | MEDLINE | ID: mdl-21529244

ABSTRACT

AIMS: Mitochondrial damage due to Ca(2+) overload-induced opening of permeability transition pores (PTP) is believed to play a role in selective degeneration of nigrostriatal dopaminergic neurons in Parkinson's disease (PD). Genetic ablation of mitochondrial matrix protein cyclophilin D (CYPD) has been shown to increase Ca(2+) threshold of PTP in vitro and to prevent cell death in several in vivo disease models. We investigated the role of CYPD in a mouse model of MPTP (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine)-induced PD. RESULTS: We demonstrate that in vitro, brain mitochondria isolated from CYPD knockout mice were less sensitive to MPP+ (1-methyl-4-phenyl-pyridinium ion)-induced membrane depolarization, and free radical generation compared to wild-type mice. CYPD knockout mitochondria isolated from ventral midbrain of mice treated with MPTP in vivo exhibited less damage as judged from respiratory chain Complex I activity, State 3 respiration rate, and respiratory control index than wild-type mice, whereas assessment of apoptotic markers showed no differences between the two genotypes. However, CYPD knockout mice were significantly resistant only to an acute regimen of MPTP neurotoxicity in contrast to the subacute and chronic MPTP paradigms. INNOVATION: Inactivation of CYPD is beneficial in preserving mitochondrial functions only in an acute insult model of MPTP-induced dopaminergic neurotoxicity. CONCLUSION: Our results suggest that CYPD deficiency distinguishes the modes of dopaminergic neurodegeneration in various regimens of MPTP-neurotoxicity.


Subject(s)
Cyclophilins/genetics , Dopaminergic Neurons/metabolism , MPTP Poisoning/metabolism , Mitochondrial Membrane Transport Proteins/genetics , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/metabolism , 1-Methyl-4-phenylpyridinium/metabolism , Animals , Apoptosis/drug effects , Apoptosis/genetics , Astrocytes/drug effects , Basal Ganglia/metabolism , Calcium/metabolism , Cell Death/genetics , Peptidyl-Prolyl Isomerase F , Disease Models, Animal , Dopaminergic Neurons/drug effects , Humans , MPTP Poisoning/genetics , MPTP Poisoning/pathology , Mice , Mice, Knockout , Microglia/drug effects , Mitochondria/metabolism , Mitochondrial Permeability Transition Pore , Substantia Nigra/pathology , Tyrosine 3-Monooxygenase/metabolism , alpha-Synuclein/metabolism
9.
Exp Neurol ; 225(1): 74-84, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20561979

ABSTRACT

Sirtuins are NAD-dependent deacetylases that regulate important biologic processes including transcription, cell survival and metabolism. Activation of SIRT1, a mammalian sirtuin, extends longevity and increases neuronal survival. An important substrate of SIRT1 is peroxisome proliferator-activated receptor gamma co-activator-1alpha (PGC-1alpha), a principal regulator of energy metabolism, whose function is significantly impaired in Huntington's disease (HD). We studied the effects of a pharmacological preparation of the SIRT1 activator resveratrol (SRT501-M), in the N171-82Q transgenic mouse model of HD. We analyzed motor performance, survival, central and peripheral pathology and levels of PGC-1alpha expression. Administration of SRT501-M increased expression of PGC-1alpha, as well as its downstream targets, nuclear respiratory factor-1 (NRF-1) and uncoupling protein-1 (UCP-1) in brown adipose tissue (BAT), but there was no effect on PGC-1alpha, NRF-1 or the mitochondrial transcription factor (Tfam) in the striatum. SRT501-M administration also reduced BAT vacuolation and decreased elevated blood glucose levels. However, there was no significant improvement in weight loss, motor performance, survival and striatal atrophy. Activation of the PGC-1alpha signaling pathway via resveratrol-induced activation of SIRT1, therefore, is an effective therapy in BAT, but not in the central nervous system of HD transgenic mice.


Subject(s)
Disease Models, Animal , Huntington Disease/drug therapy , Huntington Disease/metabolism , Stilbenes/pharmacology , Stilbenes/therapeutic use , Adipose Tissue, Brown/drug effects , Adipose Tissue, Brown/metabolism , Animals , Antioxidants/pharmacology , Antioxidants/therapeutic use , Down-Regulation/drug effects , Down-Regulation/genetics , Huntington Disease/genetics , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Transgenic , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Resveratrol , Sirtuin 1/biosynthesis , Sirtuin 1/deficiency , Sirtuin 1/physiology , Trans-Activators/biosynthesis , Trans-Activators/deficiency , Trans-Activators/physiology , Transcription Factors , Treatment Outcome
10.
Neurobiol Dis ; 36(2): 320-30, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19660549

ABSTRACT

The activity of a key mitochondrial tricarboxylic acid cycle enzyme, alpha-ketoglutarate dehydrogenase complex (KGDHC), declines in many neurodegenerative diseases. KGDHC consists of three subunits. The dihydrolipoyl succinyl transferase (DLST) component is unique to KGDHC. DLST(+/-) mice showed reduced mRNA and protein levels and decreased brain mitochondrial KGDHC activity. Neurotoxic effects of mitochondrial toxins were exacerbated in DLST(+/-) mice. MPTP produced a significantly greater reduction of striatal dopamine and tyrosine hydroxylase-positive neurons in the substantia nigra pars compacta of DLST(+/-) mice. DLST deficiency enhanced the severity of lipid peroxidation in the substantia nigra after MPTP treatment. Striatal lesions induced by either malonate or 3-nitropropionic acid (3-NP) were significantly larger in DLST(+/-) mice than in wildtype controls. DLST deficiency enhanced the 3-NP inhibition of mitochondria enzymes, and 3-NP induced protein and DNA oxidations. These observations support the hypothesis that reductions in KGDHC may impair the adaptability of the brain and contribute to the pathogenesis of neurodegenerative diseases.


Subject(s)
Genetic Predisposition to Disease , Ketoglutarate Dehydrogenase Complex/deficiency , Ketoglutarate Dehydrogenase Complex/genetics , Mitochondria/enzymology , Mitochondria/genetics , Neurotoxins/toxicity , Animals , Brain/enzymology , Coenzyme A-Transferases/deficiency , Coenzyme A-Transferases/genetics , Coenzyme A-Transferases/metabolism , Energy Metabolism/genetics , Enzyme Activation/genetics , Female , Isoenzymes/deficiency , Isoenzymes/genetics , Isoenzymes/metabolism , Ketoglutarate Dehydrogenase Complex/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurodegenerative Diseases/enzymology , Neurodegenerative Diseases/genetics
11.
Free Radic Biol Med ; 47(7): 1019-27, 2009 Oct 01.
Article in English | MEDLINE | ID: mdl-19596066

ABSTRACT

Mitochondrial dysfunction and oxidative stress are involved in Alzheimer disease (AD) pathogenesis. In human AD brains, the activity of the alpha-ketoglutarate dehydrogenase enzyme complex (alpha-KGDHC) is reduced. KGDHC is mostly involved in NADH production. It can also participate in oxidative stress and reactive oxygen species (ROS) production. The mitochondrial dihydrolipoyl succinyltransferase enzyme (DLST) is a key subunit specific to the alpha-KGDHC. In cultured cells, reduction of DLST increased H(2)O(2)-induced ROS generation and cell death. Thus, we asked whether partial genetic deletion of DLST could accelerate the onset of AD pathogenesis, using a transgenic mouse model of amyloid deposition crossed with DLST(+/-) mice. Tg19959 mice, which carry the human amyloid precursor protein with two mutations, develop amyloid deposits and progressive behavioral abnormalities. We compared Tg19959 mice to Tg19959-DLST(+/-) littermates at 2-3 months of age and studied the effects of DLST deficiency on amyloid deposition, spatial learning and memory, and oxidative stress. We found that alpha-KGDHC activity was reduced in DLST(+/-) mice. We also found that DLST deficiency increased amyloid plaque burden, Abeta oligomers, and nitrotyrosine levels and accelerated the occurrence of spatial learning and memory deficits in female Tg19959 mice. Our data suggest that alpha-KGDHC may be involved in AD pathogenesis through increased mitochondrial oxidative stress.


Subject(s)
Acyltransferases/deficiency , Amyloid beta-Peptides/metabolism , Amyloidosis/pathology , Amyloidosis/physiopathology , Memory Disorders/metabolism , Mitochondria/enzymology , Peptide Fragments/metabolism , Acyltransferases/metabolism , Alzheimer Disease/metabolism , Amyloid beta-Peptides/genetics , Amyloidosis/metabolism , Animals , Disease Models, Animal , Female , Male , Memory Disorders/genetics , Memory Disorders/physiopathology , Mice , Mice, Transgenic , Mitochondria/metabolism , Oxidative Stress , Peptide Fragments/genetics
12.
Pancreas ; 36(2): 168-72, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18376308

ABSTRACT

OBJECTIVE: There is much accumulated evidence that EGFR, HER2, and their downstream signaling pathway members such as KRAS, BRAF, and PIK3CA are strongly implicated in cancer development and progression. Recently, mutations in the kinase domains of EGFR and HER2, associated with increased sensitivity to tyrosine kinase inhibitors, have been described. METHODS: To evaluate the mutational status of these genes in intraductal papillary mucinous neoplasm (IPMN)/intraductal papillary mucinous carcinoma (IPMC), EGFR and HER2 were analyzed in 36 IPMN/IPMC, and the results were correlated to the mutational status of the KRAS, BRAF, and PIK3CA genes in the samples. RESULTS: Together, we identified 1 silent mutation of HER2, 17 (43%) KRAS mutations, 1 (2.7%) BRAF mutation, and 4 (11%) mutations of PIK3CA in the IPMN/IPMC samples. CONCLUSIONS: The EGFR and ERBB2 (HER2) mutations are very infrequent in IPMN/IPMC, suggesting the limited possibility of targeting mutated ERBB2 and EGFR for therapy for these lesions. The KRAS, BRAF, and PIK3CA, however, could represent interesting targets for future therapies in these lesions.


Subject(s)
Adenocarcinoma, Mucinous/genetics , Carcinoma, Intraductal, Noninfiltrating/genetics , Gene Expression Regulation, Neoplastic , Mutation , Oncogenes , Pancreatic Neoplasms/genetics , Adenocarcinoma, Mucinous/enzymology , Adenocarcinoma, Mucinous/mortality , Adenocarcinoma, Mucinous/pathology , Aged , Aged, 80 and over , Carcinoma, Intraductal, Noninfiltrating/enzymology , Carcinoma, Intraductal, Noninfiltrating/mortality , Carcinoma, Intraductal, Noninfiltrating/pathology , Class I Phosphatidylinositol 3-Kinases , Cohort Studies , ErbB Receptors/genetics , Female , Gene Expression Regulation, Enzymologic , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Neoplasm Staging , Pancreatic Neoplasms/enzymology , Pancreatic Neoplasms/mortality , Pancreatic Neoplasms/pathology , Phosphatidylinositol 3-Kinases/genetics , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins p21(ras) , Receptor, ErbB-2/genetics , ras Proteins/genetics
13.
Clin Cancer Res ; 12(12): 3851-5, 2006 Jun 15.
Article in English | MEDLINE | ID: mdl-16778113

ABSTRACT

PURPOSE: Recent studies have reported high frequencies of somatic mutations in the phosphoinositide-3-kinase catalytic-alpha (PIK3CA) gene in various human solid tumors. More than 75% of those somatic mutations are clustered in the helical (exon 9) and kinase domains (exon 20). The three hot-spot mutations, E542K, E545K, and H1047R, have been proven to elevate the lipid kinase activity of PIK3CA and activate the Akt signaling pathway. The mutational status of PIK3CA in intraductal papillary mucinous neoplasm/carcinoma (IPMN/IPMC) has not been evaluated previously. EXPERIMENTAL DESIGN: To evaluate a possible role for PIK3CA in the tumorigenesis of IPMN and IPMC, exons 1, 4, 5, 6, 7, 9, 12, 18, and 20 were analyzed in 36 IPMN/IPMC and two mucinous cystadenoma specimens by direct genomic DNA sequencing. RESULTS: We identified four missense mutations in the nine screened exons of PIK3CA from 36 IPMN/IPMC specimens (11%). One of the four mutations, H1047R, has been previously reported as a hot-spot mutation. The remaining three mutations, T324I, W551G, and S1015F, were novel and somatic. CONCLUSION: This is the first report of PIK3CA mutation in pancreatic cancer. Our data provide evidence that the oncogenic properties of PIK3CA contribute to the tumorigenesis of IPMN/IPMC.


Subject(s)
Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Papillary/genetics , Mutation , Pancreatic Neoplasms/genetics , Phosphatidylinositol 3-Kinases/genetics , Adult , Aged , Aged, 80 and over , Class I Phosphatidylinositol 3-Kinases , Exons , Female , Humans , Male , Middle Aged
14.
Clin Cancer Res ; 12(5): 1441-6, 2006 Mar 01.
Article in English | MEDLINE | ID: mdl-16533766

ABSTRACT

PURPOSE: Recent studies have reported high frequencies of somatic mutations in the phosphoinositide-3-kinase catalytic alpha (PIK3CA) gene in several human solid tumors. Although gene amplifications of PIK3CA have been reported in head and neck squamous cell carcinoma (HNSCC), small mutation of the gene has not been evaluated in HNSCC previously. In this study, we examined the mutation frequency of PIK3CA in HNSCC. EXPERIMENTAL DESIGN: More than 75% of the somatic mutations of PIK3CA are clustered in the helical (exon 9) and kinase domains (exon 20). To investigate the possible role of PIK3CA in HNSCC tumorigenesis, exons 1, 4, 5, 6, 7, 9, and 20 of the gene were analyzed by direct genomic DNA sequencing in 38 HNSCC specimens. RESULTS: We identified four missense mutations in the seven exons of PIK3CA from 38 HNSCC specimens (11%). Three of the four mutations (i.e., H1047R, E542K, and E545K) have been previously reported as hotspot mutations. The remaining novel mutation, Y343C, is identified at exon 4 nucleotide 1028 A --> G. Three of the four mutations were shown to be somatic, whereas the fourth mutation (H1047R) was identified in a cell line. Interestingly, three of the four mutations identified were in pharyngeal cancer samples. CONCLUSIONS: These data provide evidence that oncogenic properties of PIK3CA contribute to the carcinogenesis of human head and neck cancers, especially in pharyngeal cancer. A specific kinase inhibitor to PIK3CA may potentially be an effective therapeutic reagent against HNSCC or pharyngeal cancer in particular.


Subject(s)
Carcinoma, Squamous Cell/genetics , Head and Neck Neoplasms/genetics , Mutation, Missense/genetics , Phosphatidylinositol 3-Kinases/genetics , Adult , Aged , Aged, 80 and over , Base Sequence , Class I Phosphatidylinositol 3-Kinases , Head and Neck Neoplasms/metabolism , Humans , Middle Aged , Molecular Sequence Data , Polymerase Chain Reaction , Sequence Homology, Nucleic Acid , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL