Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Cancer Discov ; 2024 Mar 27.
Article in English | MEDLINE | ID: mdl-38533987

ABSTRACT

Cancer homeostasis depends on a balance between activated oncogenic pathways driving tumorigenesis and engagement of stress-response programs that counteract the inherent toxicity of such aberrant signaling. While inhibition of oncogenic signaling pathways has been explored extensively, there is increasing evidence that overactivation of the same pathways can also disrupt cancer homeostasis and cause lethality. We show here that inhibition of Protein Phosphatase 2A (PP2A) hyperactivates multiple oncogenic pathways and engages stress responses in colon cancer cells. Genetic and compound screens identify combined inhibition of PP2A and WEE1 as synergistic in multiple cancer models by collapsing DNA replication and triggering premature mitosis followed by cell death. This combination also suppressed the growth of patient-derived tumors in vivo. Remarkably, acquired resistance to this drug combination suppressed the ability of colon cancer cells to form tumors in vivo. Our data suggest that paradoxical activation of oncogenic signaling can result in tumor suppressive resistance.

2.
PLoS Genet ; 20(3): e1011187, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38457464

ABSTRACT

BACKGROUND: Recent developments in CRISPR/Cas9 genome-editing tools have facilitated the introduction of precise alleles, including genetic intervals spanning several kilobases, directly into the embryo. However, the introduction of donor templates, via homology directed repair, can be erroneous or incomplete and these techniques often produce mosaic founder animals. Thus, newly generated alleles must be verified at the sequence level across the targeted locus. Screening for the presence of the desired mutant allele using traditional sequencing methods can be challenging due to the size of the interval to be sequenced, together with the mosaic nature of founders. METHODOLOGY/PRINCIPAL FINDINGS: In order to help disentangle the genetic complexity of these animals, we tested the application of Oxford Nanopore Technologies long-read sequencing at the targeted locus and found that the achievable depth of sequencing is sufficient to offset the sequencing error rate associated with the technology used to validate targeted regions of interest. We have assembled an analysis workflow that facilitates interrogating the entire length of a targeted segment in a single read, to confirm that the intended mutant sequence is present in both heterozygous animals and mosaic founders. We used this workflow to compare the output of PCR-based and Cas9 capture-based targeted sequencing for validation of edited alleles. CONCLUSION: Targeted long-read sequencing supports in-depth characterisation of all experimental models that aim to produce knock-in or conditional alleles, including those that contain a mix of genome-edited alleles. PCR- or Cas9 capture-based modalities bring different advantages to the analysis.


Subject(s)
CRISPR-Cas Systems , Gene Editing , Animals , CRISPR-Cas Systems/genetics , Alleles , Gene Editing/methods , Recombinational DNA Repair , Polymerase Chain Reaction
3.
EMBO J ; 43(6): 1015-1042, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38360994

ABSTRACT

Targeting poly(ADP-ribose) glycohydrolase (PARG) is currently explored as a therapeutic approach to treat various cancer types, but we have a poor understanding of the specific genetic vulnerabilities that would make cancer cells susceptible to such a tailored therapy. Moreover, the identification of such vulnerabilities is of interest for targeting BRCA2;p53-deficient tumors that have acquired resistance to poly(ADP-ribose) polymerase inhibitors (PARPi) through loss of PARG expression. Here, by performing whole-genome CRISPR/Cas9 drop-out screens, we identify various genes involved in DNA repair to be essential for the survival of PARG;BRCA2;p53-deficient cells. In particular, our findings reveal EXO1 and FEN1 as major synthetic lethal interactors of PARG loss. We provide evidence for compromised replication fork progression, DNA single-strand break repair, and Okazaki fragment processing in PARG;BRCA2;p53-deficient cells, alterations that exacerbate the effects of EXO1/FEN1 inhibition and become lethal in this context. Since this sensitivity is dependent on BRCA2 defects, we propose to target EXO1/FEN1 in PARPi-resistant tumors that have lost PARG activity. Moreover, EXO1/FEN1 targeting may be a useful strategy for enhancing the effect of PARG inhibitors in homologous recombination-deficient tumors.


Subject(s)
Neoplasms , Tumor Suppressor Protein p53 , Humans , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , DNA Repair , DNA Damage , Neoplasms/drug therapy , Neoplasms/genetics , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Glycoside Hydrolases/genetics , Glycoside Hydrolases/metabolism , Flap Endonucleases/genetics , Flap Endonucleases/metabolism , Flap Endonucleases/therapeutic use , Exodeoxyribonucleases/genetics , DNA Repair Enzymes/genetics
4.
FEBS J ; 286(2): 322-341, 2019 01.
Article in English | MEDLINE | ID: mdl-29323786

ABSTRACT

Protein misfolding within the endoplasmic reticulum (ER stress) can be a cause or consequence of pulmonary disease. Mutation of proteins restricted to the alveolar type II pneumocyte can lead to inherited forms of pulmonary fibrosis, but even sporadic cases of pulmonary fibrosis appear to be strongly associated with activation of the unfolded protein response and/or the integrated stress response. Inhalation of smoke can impair protein folding and may be an important cause of pulmonary ER stress. Similarly, tissue hypoxia can lead to impaired protein homeostasis (proteostasis). But the mechanisms linking smoke and hypoxia to ER stress are only partially understood. In this review, we will examine the role of ER stress in the pathogenesis of lung disease by focusing on fibrosis, smoke, and hypoxia.


Subject(s)
Asphyxia/physiopathology , Endoplasmic Reticulum Stress , Endoplasmic Reticulum/pathology , Hypoxia/physiopathology , Pulmonary Fibrosis/physiopathology , Smoking/physiopathology , Unfolded Protein Response , Animals , Endoplasmic Reticulum/metabolism , Humans , Protein Folding
5.
BMC Biol ; 16(1): 34, 2018 04 03.
Article in English | MEDLINE | ID: mdl-29609607

ABSTRACT

BACKGROUND: Developmental pathways must be responsive to the environment. Phosphorylation of eIF2α enables a family of stress-sensing kinases to trigger the integrated stress response (ISR), which has pro-survival and developmental consequences. Bone morphogenetic proteins (BMPs) regulate multiple developmental processes in organisms from insects to mammals. RESULTS: Here we show in Drosophila that GCN2 antagonises BMP signalling through direct effects on translation and indirectly via the transcription factor crc (dATF4). Expression of a constitutively active GCN2 or loss of the eIF2α phosphatase dPPP1R15 impairs developmental BMP signalling in flies. In cells, inhibition of translation by GCN2 blocks downstream BMP signalling. Moreover, loss of d4E-BP, a target of crc, augments BMP signalling in vitro and rescues tissue development in vivo. CONCLUSION: These results identify a novel mechanism by which the ISR modulates BMP signalling during development.


Subject(s)
Drosophila Proteins/metabolism , Signal Transduction/physiology , Animals , Bone Morphogenetic Proteins/genetics , Bone Morphogenetic Proteins/metabolism , Drosophila , Drosophila Proteins/genetics , Eukaryotic Initiation Factor-2/genetics , Eukaryotic Initiation Factor-2/metabolism , Gene Expression Regulation/genetics , Gene Expression Regulation/physiology , Phosphorylation , Protein Kinases/genetics , Protein Kinases/metabolism , Signal Transduction/genetics
6.
FASEB J ; 30(12): 4083-4097, 2016 12.
Article in English | MEDLINE | ID: mdl-27601439

ABSTRACT

α1-Antitrypsin is a serine protease inhibitor produced in the liver that is responsible for the regulation of pulmonary inflammation. The commonest pathogenic gene mutation yields Z-α1-antitrypsin, which has a propensity to self-associate forming polymers that become trapped in inclusions of endoplasmic reticulum (ER). It is unclear whether these inclusions are connected to the main ER network in Z-α1-antitrypsin-expressing cells. Using live cell imaging, we found that despite inclusions containing an immobile matrix of polymeric α1-antitrypsin, small ER resident proteins can diffuse freely within them. Inclusions have many features to suggest they represent fragmented ER, and some are physically separated from the tubular ER network, yet we observed cargo to be transported between them in a cytosol-dependent fashion that is sensitive to N-ethylmaleimide and dependent on Sar1 and sec22B. We conclude that protein recycling occurs between ER inclusions despite their physical separation.-Dickens, J. A., Ordóñez, A., Chambers, J. E., Beckett, A. J., Patel, V., Malzer, E., Dominicus, C. S., Bradley, J., Peden, A. A., Prior, I. A., Lomas, D. A., Marciniak, S. J. The endoplasmic reticulum remains functionally connected by vesicular transport after its fragmentation in cells expressing Z-α1-antitrypsin.


Subject(s)
Biological Transport/physiology , Endoplasmic Reticulum/metabolism , Liver/metabolism , alpha 1-Antitrypsin/metabolism , Animals , Biological Transport/genetics , CHO Cells , Cells, Cultured , Cricetulus , Mutation/genetics , alpha 1-Antitrypsin/genetics
7.
Ann Am Thorac Soc ; 13 Suppl 4: S289-96, 2016 08.
Article in English | MEDLINE | ID: mdl-27564663

ABSTRACT

Alpha-1 antitrypsin deficiency is predominantly caused by point mutations that alter the protein's folding. These mutations fall into two broad categories: those that destabilize the protein dramatically and lead to its post-translational degradation and those that affect protein structure more subtly to promote protein polymerization within the endoplasmic reticulum (ER). This distinction is important because it determines the cell's response to each mutant. The severely misfolded mutants trigger an unfolded protein response (UPR) that promotes improved protein folding but can kill the cell in the chronic setting. In contrast, mutations that permit polymer formation fail to activate the UPR but instead promote a nuclear factor-κB-mediated ER overload response. The ability of polymers to increase a cell's sensitivity to ER stress likely explains apparent inconsistencies in the alpha-1 antitrypsin-signaling literature that have linked polymers with the UPR. In this review we discuss the use of mutant serpins to dissect each signaling pathway.


Subject(s)
Endoplasmic Reticulum Stress/genetics , alpha 1-Antitrypsin Deficiency/genetics , alpha 1-Antitrypsin/genetics , Endoplasmic Reticulum/metabolism , Humans , Mutation , NF-kappa B/metabolism , Signal Transduction/genetics , Unfolded Protein Response , alpha 1-Antitrypsin/metabolism , alpha 1-Antitrypsin Deficiency/metabolism
8.
Elife ; 42015 Mar 16.
Article in English | MEDLINE | ID: mdl-25774599

ABSTRACT

Four stress-sensing kinases phosphorylate the alpha subunit of eukaryotic translation initiation factor 2 (eIF2α) to activate the integrated stress response (ISR). In animals, the ISR is antagonised by selective eIF2α phosphatases comprising a catalytic protein phosphatase 1 (PP1) subunit in complex with a PPP1R15-type regulatory subunit. An unbiased search for additional conserved components of the PPP1R15-PP1 phosphatase identified monomeric G-actin. Like PP1, G-actin associated with the functional core of PPP1R15 family members and G-actin depletion, by the marine toxin jasplakinolide, destabilised the endogenous PPP1R15A-PP1 complex. The abundance of the ternary PPP1R15-PP1-G-actin complex was responsive to global changes in the polymeric status of actin, as was its eIF2α-directed phosphatase activity, while localised G-actin depletion at sites enriched for PPP1R15 enhanced eIF2α phosphorylation and the downstream ISR. G-actin's role as a stabilizer of the PPP1R15-containing holophosphatase provides a mechanism for integrating signals regulating actin dynamics with stresses that trigger the ISR.


Subject(s)
Actins/metabolism , Eukaryotic Initiation Factor-2/metabolism , Stress, Physiological , Amino Acid Sequence , Animals , Conserved Sequence , Depsipeptides/pharmacology , Drosophila melanogaster , HEK293 Cells , Humans , Mice , Molecular Sequence Data , Phosphorylation/drug effects , Protein Binding/drug effects , Protein Phosphatase 1/chemistry , Stress, Physiological/drug effects
9.
Hum Mol Genet ; 22(22): 4616-26, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-23814041

ABSTRACT

Mutants of neuroserpin are retained as polymers within the endoplasmic reticulum (ER) of neurones to cause the autosomal dominant dementia familial encephalopathy with neuroserpin inclusion bodies or FENIB. The cellular consequences are unusual in that the ordered polymers activate the ER overload response (EOR) in the absence of the canonical unfolded protein response. We use both cell lines and Drosophila models to show that the G392E mutant of neuroserpin that forms polymers is degraded by UBE2j1 E2 ligase and Hrd1 E3 ligase while truncated neuroserpin, a protein that lacks 132 amino acids, is degraded by UBE2g2 (E2) and gp78 (E3) ligases. The degradation of G392E neuroserpin results from SREBP-dependent activation of the cholesterol biosynthetic pathway in cells that express polymers of neuroserpin (G392E). Inhibition of HMGCoA reductase, the limiting enzyme of the cholesterol biosynthetic pathway, reduced the ubiquitination of G392E neuroserpin in our cell lines and increased the retention of neuroserpin polymers in both HeLa cells and primary neurones. Our data reveal a reciprocal relationship between cholesterol biosynthesis and the clearance of mutant neuroserpin. This represents the first description of a link between sterol metabolism and modulation of the proteotoxicity mediated by the EOR.


Subject(s)
Cholesterol/biosynthesis , Drosophila melanogaster/metabolism , Epilepsies, Myoclonic/metabolism , Heredodegenerative Disorders, Nervous System/metabolism , Neuropeptides/metabolism , Polymers/metabolism , Serpins/metabolism , Animals , Cells, Cultured , Disease Models, Animal , Drosophila melanogaster/genetics , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/metabolism , Epilepsies, Myoclonic/genetics , Epilepsies, Myoclonic/pathology , HeLa Cells , Heredodegenerative Disorders, Nervous System/genetics , Heredodegenerative Disorders, Nervous System/pathology , Humans , Mice , Mutant Proteins/metabolism , Neurons/metabolism , Neuropeptides/genetics , Protein Unfolding , Serpins/genetics , Signal Transduction , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitin-Protein Ligases/metabolism , Unfolded Protein Response , Neuroserpin
10.
J Cell Sci ; 126(Pt 6): 1406-15, 2013 Mar 15.
Article in English | MEDLINE | ID: mdl-23418347

ABSTRACT

Phosphorylation of eukaryotic translation initiation factor 2 alpha (eIF2α) by the kinase GCN2 attenuates protein synthesis during amino acid starvation in yeast, whereas in mammals a family of related eIF2α kinases regulate translation in response to a variety of stresses. Unlike single-celled eukaryotes, mammals also possess two specific eIF2α phosphatases, PPP1R15a and PPP1R15b, whose combined deletion leads to a poorly understood early embryonic lethality. We report the characterisation of the first non-mammalian eIF2α phosphatase and the use of Drosophila to dissect its role during development. The Drosophila protein demonstrates features of both mammalian proteins, including limited sequence homology and association with the endoplasmic reticulum. Of note, although this protein is not transcriptionally regulated, its expression is controlled by the presence of upstream open reading frames in its 5'UTR, enabling induction in response to eIF2α phosphorylation. Moreover, we show that its expression is necessary for embryonic and larval development and that this is to oppose the inhibitory effects of GCN2 on anabolic growth.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/growth & development , Eukaryotic Initiation Factor-2/metabolism , Protein Kinases/metabolism , Protein Phosphatase 1/metabolism , 5' Untranslated Regions/genetics , Amino Acid Sequence , Animals , Animals, Genetically Modified , COS Cells , Chlorocebus aethiops , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Embryo, Nonmammalian , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Stress , Eukaryotic Initiation Factor-2/genetics , HEK293 Cells , Humans , Molecular Sequence Data , Phosphorylation/genetics , Protein Kinases/genetics , Protein Phosphatase 1/genetics , RNA Processing, Post-Transcriptional/genetics , Sequence Homology, Amino Acid
11.
J Biol Chem ; 288(11): 7606-7617, 2013 Mar 15.
Article in English | MEDLINE | ID: mdl-23341460

ABSTRACT

Cell cycle checkpoints ensure that proliferation occurs only under permissive conditions, but their role in linking nutrient availability to cell division is incompletely understood. Protein folding within the endoplasmic reticulum (ER) is exquisitely sensitive to energy supply and amino acid sources because deficiencies impair luminal protein folding and consequently trigger ER stress signaling. Following ER stress, many cell types arrest within the G(1) phase, although recent studies have identified a novel ER stress G(2) checkpoint. Here, we report that ER stress affects cell cycle progression via two classes of signal: an early inhibition of protein synthesis leading to G(2) delay involving CHK1 and a later induction of G(1) arrest associated both with the induction of p53 target genes and loss of cyclin D(1). We show that substitution of p53/47 for p53 impairs the ER stress G(1) checkpoint, attenuates the recovery of protein translation, and impairs induction of NOXA, a mediator of cell death. We propose that cell cycle regulation in response to ER stress comprises redundant pathways invoked sequentially first to impair G(2) progression prior to ultimate G(1) arrest.


Subject(s)
Endoplasmic Reticulum/metabolism , Gene Expression Regulation , Genes, p53 , Tumor Suppressor Protein p53/genetics , Animals , Cell Cycle , Cell Line , Cell Line, Tumor , Cell Proliferation , Cell Separation , Drosophila melanogaster , Flow Cytometry , HEK293 Cells , HeLa Cells , Humans , Plasmids/metabolism , Protein Biosynthesis , Protein Phosphatase 1/metabolism , RNA Interference , Tumor Suppressor Protein p53/metabolism
12.
World J Diabetes ; 2(7): 114-8, 2011 Jul 15.
Article in English | MEDLINE | ID: mdl-21860696

ABSTRACT

Both environmental and genetic factors contribute to the development of diabetes mellitus and although monogenic disorders are rare, they offer unique insights into the fundamental biology underlying the disease. Mutations of the insulin gene or genes involved in the response to protein misfolding cause early onset diabetes. These have revealed an important role for endoplasmic reticulum stress in ß-cell survival. This form of cellular stress occurs when secretory proteins fail to fold efficiently. Of all the professional secretory cells we possess, ß-cells are the most sensitive to endoplasmic reticulum stress because of the large fluctuations in protein synthesis they face daily. Studies of endoplasmic reticulum stress signaling therefore offer the potential to identify new drug targets to treat diabetes.

13.
Methods Enzymol ; 499: 227-58, 2011.
Article in English | MEDLINE | ID: mdl-21683257

ABSTRACT

Transgenic Drosophila melanogaster have been used to model both the physiological and pathological behavior of serpins. The ability to generate flies expressing serpins and to rapidly assess associated phenotypes contributes to the power of this paradigm. While providing a whole-organism model of serpinopathies the powerful toolkit of genetic interventions allows precise molecular dissection of important biological pathways. In this chapter, we summarize the contribution that flies have made to the serpin field and then describe some of the experimental methods that are employed in these studies. In particular, we will describe the generation of transgenic flies, the assessment of phenotypes, and the principles of how to perform a genetic screen.


Subject(s)
Drosophila melanogaster/metabolism , Serpins/metabolism , Animals , Animals, Genetically Modified , Disease Models, Animal , Drosophila melanogaster/genetics , Humans , Protein Conformation , Serpins/genetics
14.
Diabetes Metab Res Rev ; 26(8): 611-21, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20922715

ABSTRACT

Endoplasmic reticulum (ER) stress is an integral part of life for all professional secretory cells, but it has been studied to greatest depth in the pancreatic ß-cell. This reflects both the crucial role played by ER stress in the pathogenesis of diabetes and also the exquisite vulnerability of these cells to ER dysfunction. The adaptive cellular response to ER stress, the unfolded protein response, comprises mechanisms to both regulate new protein translation and a transcriptional program to allow adaptation to the stress. The core of this response is a triad of stress-sensing proteins: protein kinase R-like endoplasmic reticulum kinase (PERK), inositol-requiring enzyme 1 (IRE1) and activating transcription factor 6. All three regulate portions of the transcriptional unfolded protein response, while PERK also attenuates protein synthesis during ER stress and IRE1 interacts directly with the c-Jun amino-terminal kinase stress kinase pathway. In this review we shall discuss these processes in detail, with emphasis given to their impact on diabetes and how recent findings indicate that ER stress may be responsible for the loss of ß-cell mass in the disease.


Subject(s)
Activating Transcription Factor 6/physiology , Diabetes Mellitus/physiopathology , Endoplasmic Reticulum/physiology , Endoribonucleases/physiology , Islets of Langerhans/physiology , Protein Serine-Threonine Kinases/physiology , Stress, Physiological/physiology , Animals , DNA-Binding Proteins/physiology , Diabetes Mellitus, Type 1/physiopathology , Epiphyses/abnormalities , Epiphyses/physiopathology , Glycoproteins/physiology , Humans , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/physiology , Mice , Osteochondrodysplasias/physiopathology , Oxidoreductases , Regulatory Factor X Transcription Factors , Transcription Factors/physiology , Unfolded Protein Response
15.
J Cell Sci ; 123(Pt 17): 2892-900, 2010 Sep 01.
Article in English | MEDLINE | ID: mdl-20682638

ABSTRACT

The integrated stress response (ISR) protects cells from numerous forms of stress and is involved in the growth of solid tumours; however, it is unclear how the ISR acts on cellular proliferation. We have developed a model of ISR signalling with which to study its effects on tissue growth. Overexpression of the ISR kinase PERK resulted in a striking atrophic eye phenotype in Drosophila melanogaster that could be rescued by co-expressing the eIF2alpha phosphatase GADD34. A genetic screen of 3000 transposon insertions identified grapes, the gene that encodes the Drosophila orthologue of checkpoint kinase 1 (CHK1). Knockdown of grapes by RNAi rescued eye development despite ongoing PERK activation. In mammalian cells, CHK1 was activated by agents that induce ER stress, which resulted in a G2 cell cycle delay. PERK was both necessary and sufficient for CHK1 activation. These findings indicate that non-genotoxic misfolded protein stress accesses DNA-damage-induced cell cycle checkpoints to couple the ISR to cell cycle arrest.


Subject(s)
Protein Kinases/physiology , Stress, Physiological/physiology , Animals , Cell Cycle/physiology , Cell Proliferation , Checkpoint Kinase 1 , DNA Damage , Drosophila melanogaster/enzymology , Drosophila melanogaster/genetics , Drosophila melanogaster/growth & development , Endoplasmic Reticulum/enzymology , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/physiology , Enzyme Activation , Eye/growth & development , Female , Gene Knockdown Techniques , Humans , Male , Phenotype , Protein Kinases/genetics , Protein Kinases/metabolism , Proteostasis Deficiencies/genetics , Proteostasis Deficiencies/metabolism , Stress, Physiological/genetics , cdc25 Phosphatases/metabolism , eIF-2 Kinase/biosynthesis , eIF-2 Kinase/genetics , eIF-2 Kinase/metabolism
16.
Neurobiol Aging ; 30(9): 1477-82, 2009 Sep.
Article in English | MEDLINE | ID: mdl-18162161

ABSTRACT

Mutations in the tau gene (MAPT) have been found in families with frontotemporal dementia with parkinsonism linked to chromosome 17. In addition, the MAPT H1-clade specific sub-haplotype, H1c, has been strongly associated with the tauopathies, progressive supranuclear palsy (PSP) and corticobasal degeneration (CBD) and, to a lesser extent, with Alzheimer's disease (AD). In Parkinson's disease (PD), there have been several reports of association with the MAPT H1-clade. Although weak to inconclusive, this association is supported by meta-analyses of the various studies. To further investigate this baffling role of MAPT in PD, six haplotype-tagging SNPs were genotyped in a large cohort of sporadic PD cases; 324 pathologically confirmed and 248 clinically diagnosed, and 660 controls. In the single-locus association analysis, the H1-clade was associated with an increased risk of PD (p=0.032). In the haplotype-analysis, the sole H2-derived haplotype was under-represented in all of the PD cases compared to controls (p=0.03). There was no significant difference in the distribution of any of the common haplotypes derived from the H1-clade background. Our study supports the hypothesis that genetic variability in the MAPT gene confers susceptibility to PD. However, the effect is not strong, and the H1c haplotype is not involved, suggesting a mechanism that is distinct to that involved in the associated tauopathies and may be explained by the H1/H2 inversion.


Subject(s)
Genetic Predisposition to Disease/genetics , Parkinson Disease/genetics , Polymorphism, Single Nucleotide/genetics , tau Proteins/genetics , Case-Control Studies , Cohort Studies , DNA Mutational Analysis , Gene Frequency/genetics , Genetic Markers/genetics , Genetic Testing , Genetic Variation/genetics , Genotype , Haplotypes/genetics , Humans , Nerve Degeneration/genetics
17.
Neurobiol Aging ; 30(3): 388-93, 2009 Mar.
Article in English | MEDLINE | ID: mdl-17723255

ABSTRACT

We identified a case of Alzheimer's disease with a deletion of the lysine residue at codon 280 (DeltaK280) in exon 10-encoded microtubule-binding repeat domain of the tau gene (MAPT). This mutation was originally identified in a sporadic case of frontotemporal dementia (FTD) with a family history of Parkinson's disease. In the original report, the authors were careful in their assessment of the pathogenicity and suggested one could not be sure whether the mutation was pathogenic or not. The mutation has always presented a conundrum because it is the only known mutation, of assumed pathogenicity, which increases the proportion of 3-repeat tau mRNA in in vitro assays. Here we present the clinical and pathological features of a new case with this mutation and discuss whether the mutation is indeed pathogenic.


Subject(s)
Alzheimer Disease/genetics , Alzheimer Disease/pathology , Gene Deletion , tau Proteins/genetics , Aged, 80 and over , Codon/genetics , Exons/genetics , Humans , Lysine/genetics , Male , Microtubule-Associated Proteins/genetics , Protein Structure, Tertiary/genetics , Repetitive Sequences, Amino Acid/genetics , tau Proteins/metabolism
18.
Neuro Oncol ; 10(6): 946-57, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18650488

ABSTRACT

Malignant peripheral nerve sheath tumors (MPNSTs) are sarcomas with poor prognosis and limited treatment options. Evidence for a role of epidermal growth factor receptor (EGFR) and receptor tyrosine kinase erbB2 in MPNSTs led us to systematically study these potential therapeutic targets in a larger tumor panel (n = 37). Multiplex ligation-dependent probe amplification and fluorescence in situ hybridization analysis revealed increased EGFR dosage in 28% of MPNSTs. ERBB2 and three tumor suppressor genes (PTEN [phosphatase and tensin homolog deleted on chromosome 10], CDKN2A [cyclin-dependent kinase inhibitor 2A], and TP53 [tumor protein p53]) were frequently lost or reduced. Reduction of CDKN2A was linked to appearance of metastasis. Comparison of corresponding neurofibromas and MPNSTs revealed an increase in genetic lesions in MPNSTs. No somatic mutations were found within tyrosine-kinase-encoding exons of EGFR and ERBB2. However, at the protein level, expression of EGFR and erbB2 was frequently detected in MPNSTs. EGFR expression was significantly associated with increased EGFR gene dosage. The EGFR ligands transforming growth factor alpha and EGF were more strongly expressed in MPNSTs than in neurofibromas. The effects of the drugs erlotinib and trastuzumab, which target EGFR and erbB2, were determined on MPNST cell lines. In contrast to trastuzumab, erlotinib mediated dose-dependent inhibition of cell proliferation. EGF-induced EGFR phosphorylation was attenuated by erlotinib. Summarized, our data indicate that EGFR and erbB2 are potential targets in treatment of MPNST patients.


Subject(s)
Antineoplastic Agents/administration & dosage , Drug Delivery Systems , ErbB Receptors/genetics , Nerve Sheath Neoplasms/genetics , Receptor, ErbB-2/genetics , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized , Blotting, Western , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Synergism , Epidermal Growth Factor/genetics , Epidermal Growth Factor/metabolism , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Erlotinib Hydrochloride , Gene Dosage , Genes, p16 , Genes, p53 , Humans , Immunohistochemistry , Nerve Sheath Neoplasms/metabolism , PTEN Phosphohydrolase/genetics , Polymorphism, Single-Stranded Conformational , Quinazolines/pharmacology , Receptor, ErbB-2/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transforming Growth Factor alpha/genetics , Transforming Growth Factor alpha/metabolism , Trastuzumab
19.
Neuro Oncol ; 9(3): 291-7, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17504929

ABSTRACT

A subset of glioblastomas (GBMs) carry gene amplifications on chromosomal segment 4q12. To characterize this amplicon in detail, we analyzed a set of 100 samples consisting of 65 GBMs, 10 WHO grade III astrocytomas, 12 oligodendrogliomas, and 13 glioma cell cultures. We applied multiplex ligation-dependent probe amplification to determine the gene dosage of PDGFRA, KIT, and KDR and the flanking genes USP46, RASL11B, LNX1, CHIC2, SEC3L1, and IGFBP7. The amplicon was highly variable in size and copy number and extended over a region of up to 5 Mb. Amplifications on 4q12 were observed in 15% of GBMs and 23% of GBM cell cultures but not in 22 other gliomas. We analyzed transcription and translation of some genes within this amplicon. Gene amplification generally correlated with high transcript levels but did not necessarily result in increased protein levels. However, we detected frequent expression of proteins encoded by PDGFRA, KIT, and KDR in GBMs and GBM cell cultures independent of the amplification status. Future treatment of GBM patients may include drugs targeting multiple kinases that are encoded by genes on chromosomal segment 4q12.


Subject(s)
Brain Neoplasms/genetics , Chromosomes, Human, Pair 4/genetics , Gene Amplification , Glioblastoma/genetics , Blotting, Western , Humans , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...