Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Hum Mutat ; 43(10): 1396-1407, 2022 10.
Article in English | MEDLINE | ID: mdl-35762214

ABSTRACT

Chordoma is a rare bone tumor with genetic risk factors largely unknown. We conducted a whole-exome sequencing (WES) analysis of germline DNA from 19 familial chordoma cases in five pedigrees and 137 sporadic chordoma patients and identified 17 rare germline variants in PALB2 and BRCA2, whose products play essential roles in homologous recombination (HR) and tumor suppression. One PALB2 variant showed disease cosegregation in a family with four affected people or obligate gene carrier. Chordoma cases had a significantly increased burden of rare variants in both genes when compared to population-based controls. Four of the six PALB2 variants identified from chordoma patients modestly affected HR function and three of the 11 BRCA2 variants caused loss of function in experimental assays. These results, together with previous reports of abnormal morphology and Brachyury expression of the notochord in Palb2 knockout mouse embryos and genomic signatures associated with HR defect and HR gene mutations in advanced chordomas, suggest that germline mutations in PALB2 and BRCA2 may increase chordoma susceptibility. Our data shed light on the etiology of chordoma and support the previous finding that PARP-1 inhibitors may be a potential therapy for some chordoma patients.


Subject(s)
BRCA2 Protein , Breast Neoplasms , Chordoma , Fanconi Anemia Complementation Group N Protein , Animals , BRCA2 Protein/genetics , Breast Neoplasms/genetics , Chordoma/genetics , Fanconi Anemia Complementation Group N Protein/genetics , Female , Genes, BRCA2 , Genetic Predisposition to Disease , Germ-Line Mutation , Humans , Mice
2.
Hum Mutat ; 42(2): 200-212, 2021 02.
Article in English | MEDLINE | ID: mdl-33314489

ABSTRACT

The discovery of high-risk breast cancer susceptibility genes, such as Breast cancer associated gene 1 (BRCA1) and Breast cancer associated gene 2 (BRCA2) has led to accurate identification of individuals for risk management and targeted therapy. The rapid decline in sequencing costs has tremendously increased the number of individuals who are undergoing genetic testing world-wide. However, given the significant differences in population-specific variants, interpreting the results of these tests can be challenging especially for novel genetic variants in understudied populations. Here we report the characterization of novel variants in the Malaysian and Singaporean population that consist of different ethnic groups (Malays, Chinese, Indian, and other indigenous groups). We have evaluated the functional significance of 14 BRCA2 variants of uncertain clinical significance by using multiple in silico prediction tools and examined their frequency in a cohort of 7840 breast cancer cases and 7928 healthy controls. In addition, we have used a mouse embryonic stem cell (mESC)-based functional assay to assess the impact of these variants on BRCA2 function. We found these variants to be functionally indistinguishable from wild-type BRCA2. These variants could fully rescue the lethality of Brca2-null mESCs and exhibited no sensitivity to six different DNA damaging agents including a poly ADP ribose polymerase inhibitor. Our findings strongly suggest that all 14 evaluated variants are functionally neutral. Our findings should be valuable in risk assessment of individuals carrying these variants.


Subject(s)
Breast Neoplasms , Animals , BRCA1 Protein/genetics , BRCA2 Protein/genetics , Breast Neoplasms/epidemiology , Cohort Studies , Female , Genes, BRCA2 , Genetic Predisposition to Disease , Genetic Testing , Humans , Malaysia , Mice
3.
Breast Cancer Res ; 22(1): 43, 2020 05 11.
Article in English | MEDLINE | ID: mdl-32393398

ABSTRACT

Next-generation sequencing of Sri Lankan families with inherited cancer syndromes resulted in the identification of five BRCA2 variants of unknown clinical significance. Interpreting such variants poses significant challenges for both clinicians and patients. Using a mouse embryonic stem cell-based functional assay, we found I785V, N830D, and K2077N to be functionally indistinguishable from wild-type BRCA2. Specific but mild sensitivity to olaparib and reduction in homologous recombination (HR) efficiency suggest partial loss of function of the A262T variant. This variant is located in the N-terminal DNA binding domain of BRCA2 that can facilitate HR by binding to dsDNA/ssDNA junctions. P3039P is clearly pathogenic because of premature protein truncation caused by exon 23 skipping. These findings highlight the value of mouse embryonic stem cell-based assays for determining the functional significance of variants of unknown clinical significance and provide valuable information regarding risk estimation and genetic counseling of families carrying these BRCA2 variants.


Subject(s)
BRCA2 Protein/genetics , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Mouse Embryonic Stem Cells/metabolism , Mutation , Neoplastic Syndromes, Hereditary/genetics , Neoplastic Syndromes, Hereditary/pathology , Animals , BRCA2 Protein/metabolism , Biological Assay/methods , Breast Neoplasms/epidemiology , Breast Neoplasms/metabolism , Cell Survival , Cohort Studies , Female , Homologous Recombination , Humans , Mice , Neoplastic Syndromes, Hereditary/epidemiology , Neoplastic Syndromes, Hereditary/metabolism , Sri Lanka/epidemiology , Xenograft Model Antitumor Assays
4.
J Hum Genet ; 65(9): 805-809, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32393813

ABSTRACT

A pathogenic mutation in BRCA2 significantly increases the risk of breast and ovarian cancers making it imperative to examine the functional consequences of variants of uncertain clinical significance. Variants that are predicted to result in a truncated protein are unambiguously classified as pathogenic. We have previously shown how a pathogenic splice site variant known to generate a premature termination codon (PTC) in exon 9 and a nonsense mutation at exon 7, can generate functional BRCA2 by skipping exons 4-7 and restoring the reading frame. Using a well-established mouse embryonic stem cell-based assay, we functionally characterize here one splice site mutation and 11 pathogenic BRCA2 variants that are either nonsense mutation or generate PTC in different exons ranging from exons 4 to 7. Our study shows that five variants can restore the open reading frame by exon skipping and generate a functional protein. This suggests further need to exercise prudence when classifying clearly pathogenic variants.


Subject(s)
BRCA2 Protein/genetics , Codon, Nonsense , Embryonic Stem Cells/metabolism , Ovarian Neoplasms/genetics , Alternative Splicing , Animals , BRCA2 Protein/metabolism , Cell Survival/genetics , Codon, Nonsense/genetics , Exons , Female , Gene Knockout Techniques , Mice , Mice, Knockout , Mutation , RNA Splice Sites
5.
Hum Mol Genet ; 21(18): 3993-4006, 2012 Sep 15.
Article in English | MEDLINE | ID: mdl-22678057

ABSTRACT

Single-nucleotide substitutions and small in-frame insertions or deletions identified in human breast cancer susceptibility genes BRCA1 and BRCA2 are frequently classified as variants of unknown clinical significance (VUS) due to the availability of very limited information about their functional consequences. Such variants can most reliably be classified as pathogenic or non-pathogenic based on the data of their co-segregation with breast cancer in affected families and/or their co-occurrence with a pathogenic mutation. Biological assays that examine the effect of variants on protein function can provide important information that can be used in conjunction with available familial data to determine the pathogenicity of VUS. In this report, we have used a previously described mouse embryonic stem (mES) cell-based functional assay to characterize eight BRCA2 VUS that affect highly conserved amino acid residues and map to the N-terminal PALB2-binding or the C-terminal DNA-binding domains. For several of these variants, very limited co-segregation information is available, making it difficult to determine their pathogenicity. Based on their ability to rescue the lethality of Brca2-deficient mES cells and their effect on sensitivity to DNA-damaging agents, homologous recombination and genomic integrity, we have classified these variants as pathogenic or non-pathogenic. In addition, we have used homology-based modeling as a predictive tool to assess the effect of some of these variants on the structural integrity of the C-terminal DNA-binding domain and also generated a knock-in mouse model to analyze the physiological significance of a residue reported to be essential for the interaction of BRCA2 with meiosis-specific recombinase, DMC1.


Subject(s)
BRCA2 Protein/genetics , Breast Neoplasms/genetics , Embryonic Stem Cells/metabolism , Mutation , Nuclear Proteins/metabolism , Tumor Suppressor Proteins/metabolism , Amino Acid Sequence , Animals , BRCA2 Protein/chemistry , Cell Cycle Proteins , Cell Survival , Cells, Cultured , Chromosome Mapping , Conserved Sequence , DNA Breaks, Double-Stranded , DNA Repair , DNA-Binding Proteins , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/physiology , Fanconi Anemia Complementation Group N Protein , Female , Genetic Association Studies , Humans , Likelihood Functions , Male , Mice , Mice, Transgenic , Mitomycin/pharmacology , Models, Molecular , Mutagens/pharmacology , Protein Binding , Protein Interaction Domains and Motifs/genetics , Protein Structure, Quaternary , Structural Homology, Protein
6.
Cancers (Basel) ; 16(13)2024 Jun 25.
Article in English | MEDLINE | ID: mdl-39001383

ABSTRACT

Activating mutations in the RAS/MAPK pathway are observed in relapsed neuroblastoma. Preclinical studies indicate that these tumors have an increased sensitivity to inhibitors of the RAS/MAPK pathway, such as MEK inhibitors. MEK inhibitors do not induce durable responses as single agents, indicating a need to identify synergistic combinations of targeted agents to provide therapeutic benefit. We previously showed preclinical therapeutic synergy between a MEK inhibitor, trametinib, and a monoclonal antibody specific for IGF1R, ganitumab in RAS-mutated rhabdomyosarcoma. Neuroblastoma cells, like rhabdomyosarcoma cells, are sensitive to the inhibition of the RAS/MAPK and IGF1R/AKT/mTOR pathways. We hypothesized that the combination of trametinib and ganitumab would be effective in RAS-mutated neuroblastoma. In this study, trametinib and ganitumab synergistically suppressed neuroblastoma cell proliferation and induced apoptosis in cell culture. We also observed a delay in tumor initiation and prolongation of survival in heterotopic and orthotopic xenograft models treated with trametinib and ganitumab. However, the growth of both primary and metastatic tumors was observed in animals receiving the combination of trametinib and ganitumab. Therefore, more preclinical work is necessary before testing this combination in patients with relapsed or refractory RAS-mutated neuroblastoma.

7.
Blood ; 118(9): 2430-42, 2011 Sep 01.
Article in English | MEDLINE | ID: mdl-21719596

ABSTRACT

Biallelic mutations in the human breast cancer susceptibility gene, BRCA2, are associated with Fanconi anemia, implying that some persons who inherit 2 deleterious variants of BRCA2 are able to survive even though it is well established that BRCA2 is indispensable for viability in mice. One such variant, IVS7 + 2T > G, results in premature protein truncation because of skipping of exon 7. Surprisingly, the persons who are either IVS7 + 2T > G homozygous or compound heterozygous are born alive but die of malignancy associated with Fanconi anemia. Using a mouse embryonic stem cell-based functional assay, we found that the IVS7 + 2T > G allele produces an alternatively spliced transcript lacking exons 4-7, encoding an in-frame BRCA2 protein with an internal deletion of 105 amino acids (BRCA2(Δ105)). We demonstrate that BRCA2(Δ105) is proficient in homologous recombination-mediated DNA repair as measured by different functional assays. Evaluation of this transcript in normal and leukemia cells suggests that BRCA2(Δ105) may contribute to the viability of persons inheriting this mutation. In this study, we have also characterized 5 other BRCA2 variants and found 3 of these (p.L2510P, p.R2336H, and p.W2626C) to be deleterious and 2 (p.I2490T and p.K2729N) probably neutral. Such studies are important to understand the functional significance of unclassified BRCA2 variants.


Subject(s)
Fanconi Anemia/genetics , Genes, BRCA2 , Genetic Complementation Test , Alleles , Alternative Splicing , Amino Acid Substitution , Animals , Cell Line , Cell Line, Tumor , Chromosomes, Artificial, Bacterial/genetics , Embryonic Stem Cells , Exons/genetics , Genotype , Humans , Leukemia, Myeloid, Acute/pathology , Mice , Mutation , RNA Splice Sites/genetics , Sequence Deletion , Sequence Homology, Nucleic Acid
8.
Cell Rep Methods ; 3(11): 100628, 2023 Nov 20.
Article in English | MEDLINE | ID: mdl-37922907

ABSTRACT

Sequencing of genes, such as BRCA1 and BRCA2, is recommended for individuals with a personal or family history of early onset and/or bilateral breast and/or ovarian cancer or a history of male breast cancer. Such sequencing efforts have resulted in the identification of more than 17,000 BRCA2 variants. The functional significance of most variants remains unknown; consequently, they are called variants of uncertain clinical significance (VUSs). We have previously developed mouse embryonic stem cell (mESC)-based assays for functional classification of BRCA2 variants. We now developed a next-generation sequencing (NGS)-based approach for functional evaluation of BRCA2 variants using pools of mESCs expressing 10-25 BRCA2 variants from a given exon. We use this approach for functional evaluation of 223 variants listed in ClinVar. Our functional classification of BRCA2 variants is concordant with the classification reported in ClinVar or those reported by other orthogonal assays.


Subject(s)
Genes, BRCA2 , Ovarian Neoplasms , Humans , Female , Male , Animals , Mice , Mouse Embryonic Stem Cells , Ovarian Neoplasms/genetics , BRCA2 Protein/genetics
9.
Clin Cancer Res ; 29(2): 472-487, 2023 01 17.
Article in English | MEDLINE | ID: mdl-36322002

ABSTRACT

PURPOSE: PAX-fusion negative rhabdomyosarcoma (FN RMS) is driven by alterations in the RAS/MAP kinase pathway and is partially responsive to MEK inhibition. Overexpression of IGF1R and its ligands is also observed in FN RMS. Preclinical and clinical studies have suggested that IGF1R is itself an important target in FN RMS. Our previous studies revealed preclinical efficacy of the MEK1/2 inhibitor, trametinib, and an IGF1R inhibitor, BMS-754807, but this combination was not pursued clinically due to intolerability in preclinical murine models. Here, we sought to identify a combination of an MEK1/2 inhibitor and IGF1R inhibitor, which would be tolerated in murine models and effective in both cell line and patient-derived xenograft models of RAS-mutant FN RMS. EXPERIMENTAL DESIGN: Using proliferation and apoptosis assays, we studied the factorial effects of trametinib and ganitumab (AMG 479), a mAb with specificity for human and murine IGF1R, in a panel of RAS-mutant FN RMS cell lines. The molecular mechanism of the observed synergy was determined using conventional and capillary immunoassays. The efficacy and tolerability of trametinib/ganitumab was assessed using a panel of RAS-mutated cell-line and patient-derived RMS xenograft models. RESULTS: Treatment with trametinib and ganitumab resulted in synergistic cellular growth inhibition in all cell lines tested and inhibition of tumor growth in four of six models of RAS-mutant RMS. The combination had little effect on body weight and did not produce thrombocytopenia, neutropenia, or hyperinsulinemia in tumor-bearing SCID beige mice. Mechanistically, ganitumab treatment prevented the phosphorylation of AKT induced by MEK inhibition alone. Therapeutic response to the combination was observed in models without a mutation in the PI3K/PTEN axis. CONCLUSIONS: We demonstrate that combined trametinib and ganitumab is effective in a genomically diverse panel of RAS-mutated FN RMS preclinical models. Our data also show that the trametinib/ganitumab combination likely has a favorable tolerability profile. These data support testing this combination in a phase I/II clinical trial for pediatric patients with relapsed or refractory RAS-mutated FN RMS.


Subject(s)
Rhabdomyosarcoma , Humans , Animals , Mice , Child , Cell Line, Tumor , Mice, SCID , Rhabdomyosarcoma/drug therapy , Rhabdomyosarcoma/genetics , Rhabdomyosarcoma/pathology , Protein Kinase Inhibitors/pharmacology , Mitogen-Activated Protein Kinase Kinases
10.
Commun Biol ; 4(1): 355, 2021 03 19.
Article in English | MEDLINE | ID: mdl-33742095

ABSTRACT

Mechanical stress induced by contractions constantly threatens the integrity of muscle Z-disc, a crucial force-bearing structure in striated muscle. The PDZ-LIM proteins have been proposed to function as adaptors in transducing mechanical signals to preserve the Z-disc structure, however the underlying mechanisms remain poorly understood. Here, we show that LDB3, a well-characterized striated muscle PDZ-LIM protein, modulates mechanical stress signaling through interactions with the mechanosensing domain in filamin C, its chaperone HSPA8, and PKCα in the Z-disc of skeletal muscle. Studies of Ldb3Ala165Val/+ mice indicate that the myopathy-associated LDB3 p.Ala165Val mutation triggers early aggregation of filamin C and its chaperones at muscle Z-disc before aggregation of the mutant protein. The mutation causes protein aggregation and eventually Z-disc myofibrillar disruption by impairing PKCα and TSC2-mTOR, two important signaling pathways regulating protein stability and disposal of damaged cytoskeletal components at a major mechanosensor hub in the Z-disc of skeletal muscle.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , LIM Domain Proteins/genetics , Mechanotransduction, Cellular , Muscle, Skeletal/enzymology , Myopathies, Structural, Congenital/enzymology , Point Mutation , Protein Kinase C-alpha/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Autophagy , Disease Models, Animal , Down-Regulation , Filamins/metabolism , HSC70 Heat-Shock Proteins/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Muscle Contraction , Muscle Strength , Muscle, Skeletal/pathology , Muscle, Skeletal/physiopathology , Myopathies, Structural, Congenital/genetics , Myopathies, Structural, Congenital/pathology , Myopathies, Structural, Congenital/physiopathology , Protein Aggregates , Protein Aggregation, Pathological , Protein Kinase C-alpha/genetics , TOR Serine-Threonine Kinases/genetics , Tuberous Sclerosis Complex 2 Protein/genetics , Tuberous Sclerosis Complex 2 Protein/metabolism
11.
Cell Death Dis ; 12(9): 838, 2021 09 06.
Article in English | MEDLINE | ID: mdl-34489406

ABSTRACT

Hereditary non-polyposis colorectal cancer, now known as Lynch syndrome (LS) is one of the most common cancer predisposition syndromes and is caused by germline pathogenic variants (GPVs) in DNA mismatch repair (MMR) genes. A common founder GPV in PMS2 in the Canadian Inuit population, NM_000535.5: c.2002A>G, leads to a benign missense (p.I668V) but also acts as a de novo splice site that creates a 5 bp deletion resulting in a truncated protein (p.I668*). Individuals homozygous for this GPV are predisposed to atypical constitutional MMR deficiency with a delayed onset of first primary malignancy. We have generated mice with an equivalent germline mutation (Pms2c.1993A>G) and demonstrate that it results in a splicing defect similar to those observed in humans. Homozygous mutant mice are viable like the Pms2 null mice. However, unlike the Pms2 null mice, these mutant mice are fertile, like humans homozygous for this variant. Furthermore, these mice exhibit a significant increase in microsatellite instability and intestinal adenomas on an Apc mutant background. Rectification of the splicing defect in human and murine fibroblasts using antisense morpholinos suggests that this novel mouse model can be valuable in evaluating the efficacy aimed at targeting the splicing defect in PMS2 that is highly prevalent among the Canadian Inuits.


Subject(s)
DNA Mismatch Repair/genetics , Founder Effect , Mismatch Repair Endonuclease PMS2/genetics , Mutation/genetics , RNA Splicing/genetics , Adenomatous Polyposis Coli Protein/genetics , Animals , Base Sequence , Disease Models, Animal , Exons/genetics , Fertility/genetics , Fibroblasts/metabolism , Male , Meiosis , Mice, Inbred C57BL , Microsatellite Instability , Mismatch Repair Endonuclease PMS2/metabolism , Morpholinos/pharmacology , Polyps/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Spermatozoa/pathology , Testis/pathology
12.
NPJ Genom Med ; 5(1): 52, 2020 Dec 08.
Article in English | MEDLINE | ID: mdl-33293522

ABSTRACT

Sequencing-based genetic tests to identify individuals at increased risk of hereditary breast and ovarian cancers have resulted in the identification of more than 40,000 sequence variants of BRCA1 and BRCA2. A majority of these variants are considered to be variants of uncertain significance (VUS) because their impact on disease risk remains unknown, largely due to lack of sufficient familial linkage and epidemiological data. Several assays have been developed to examine the effect of VUS on protein function, which can be used to assess their impact on cancer susceptibility. In this study, we report the functional characterization of 88 BRCA2 variants, including several previously uncharacterized variants, using a well-established mouse embryonic stem cell (mESC)-based assay. We have examined their ability to rescue the lethality of Brca2 null mESC as well as sensitivity to six DNA damaging agents including ionizing radiation and a PARP inhibitor. We have also examined the impact of BRCA2 variants on splicing. In addition, we have developed a computational model to determine the probability of impact on function of the variants that can be used for risk assessment. In contrast to the previous VarCall models that are based on a single functional assay, we have developed a new platform to analyze the data from multiple functional assays separately and in combination. We have validated our VarCall models using 12 known pathogenic and 10 neutral variants and demonstrated their usefulness in determining the pathogenicity of BRCA2 variants that are listed as VUS or as variants with conflicting functional interpretation.

13.
Hum Mutat ; 30(11): 1543-50, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19795481

ABSTRACT

Variants of unknown significance (VUS) in BRCA1 and BRCA2 are common, and present significant challenges for genetic counseling. We observed that BRCA2: c.6853A>G (p.I2285V) (Breast Cancer Information Core [BIC] name: 7081A>G; http://research.nhgri.nih.gov/bic/) co-occurs in trans with the founder mutation c.5946delT (p.S1982RfsX22) (BIC name: 6174delT), supporting the published classification of p.I2285V as a neutral variant. However, we also noted that when compared with wild-type BRCA2, p.I2285V resulted in increased exclusion of exon 12. Functional assay using allelic complementation in Brca2-null mouse embryonic stem cells revealed that p.I2285V, an allele with exon 12 deleted and wild-type BRCA2 were all phenotypically indistinguishable, as measured by sensitivity to DNA-damaging agents, effect on irradiation-induced Rad51 foci formation, homologous recombination, and overall genomic integrity. An allele frequency study showed the p.I2285V variant was identified in 15 out of 722 (2.1%) Ashkenazi Jewish cases and 10 out of 475 (2.1%) ethnically-matched controls (odds ratio, 0.99; 95% confidence interval: 0.44-2.21; P=0.97). Thus the p.I2285V variant is not associated with an increased risk for breast cancer. Taken together, our clinical and functional studies strongly suggest that exon 12 is functionally redundant and therefore missense variants in this exon are likely to be neutral. Such comprehensive functional studies will be important adjuncts to genetic studies of variants.


Subject(s)
Exons , Genes, BRCA2 , Animals , BRCA2 Protein/chemistry , BRCA2 Protein/genetics , BRCA2 Protein/physiology , DNA Mutational Analysis , Gene Frequency , Genetic Variation , Humans , Mice , RNA Splicing , RNA, Messenger/chemistry
14.
Curr Biol ; 16(2): 130-9, 2006 Jan 24.
Article in English | MEDLINE | ID: mdl-16431365

ABSTRACT

BACKGROUND: Arf GAPs are multidomain proteins that function in membrane traffic by inactivating the GTP binding protein Arf1. Numerous Arf GAPs contain a BAR domain, a protein structural element that contributes to membrane traffic by either inducing or sensing membrane curvature. We have examined the role of a putative BAR domain in the function of the Arf GAP ASAP1. RESULTS: ASAP1's N terminus, containing the putative BAR domain together with a PH domain, dimerized to form an extended structure that bound to large unilamellar vesicles containing acidic phospholipids, properties that define a BAR domain. A recombinant protein containing the BAR domain of ASAP1, together with the PH and Arf GAP domains, efficiently bent the surface of large unilamellar vesicles, resulting in the formation of tubular structures. This activity was regulated by Arf1*GTP binding to the Arf GAP domain. In vivo, the tubular structures induced by ASAP1 mutants contained epidermal growth factor receptor (EGFR) and Rab11, and ASAP1 colocalized in tubular structures with EGFR during recycling of receptor. Expression of ASAP1 accelerated EGFR trafficking and slowed cell spreading. An ASAP1 mutant lacking the BAR domain had no effect. CONCLUSIONS: The N-terminal BAR domain of ASAP1 mediates membrane bending and is necessary for ASAP1 function. The Arf dependence of the bending activity is consistent with ASAP1 functioning as an Arf effector.


Subject(s)
Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/physiology , Cell Membrane/ultrastructure , ErbB Receptors/metabolism , GTPase-Activating Proteins/chemistry , GTPase-Activating Proteins/physiology , ADP-Ribosylation Factors/physiology , Adaptor Proteins, Signal Transducing/analysis , Amino Acid Sequence , Animals , GTPase-Activating Proteins/analysis , Mice , Models, Biological , Models, Molecular , Molecular Sequence Data , Protein Structure, Tertiary , Protein Transport , Sequence Alignment
15.
Cancer Genomics Proteomics ; 6(1): 41-9, 2009.
Article in English | MEDLINE | ID: mdl-19451089

ABSTRACT

Celecoxib, a selective inhibitor of cyclooxygenase-2 (Cox-2), was efficacious in clinical prevention trials of patients with familial adenomatous polyposis (FAP) and sporadic colorectal cancer. To identify as yet poorly defined molecular determinants of celecoxib efficacy, a multidimensional serum fractionation approach was used coupled with nanospray tandem mass spectrometry to perform label-free global proteomic profiling of serum samples from the FAP/celecoxib prevention trial. Subsequently, the application of an algorithm for large-scale biomarker discovery on comparative serum proteomic profiles of pre- and post-celecoxib treatment samples identified 83 potentially celecoxib-responsive proteins from various cellular compartments, biological processes and molecular functions. Celecoxib modulation of some of these proteins was confirmed in serum samples of FAP patients and colorectal cancer cell lines by Western blotting. Thus, using a shotgun procedure to rapidly identify important celecoxib-modulated proteins, this pilot study has uncovered novel systemic changes some of which are highly relevant for carcinogenesis and vascular biology. Validation of selected markers, especially those involved in key signaling networks and those considered molecular indicators of cardiovascular pathology, in larger celecoxib clinical trials is expected to provide insights into the molecular mechanisms of celecoxib and the efficacy/toxicity issues related to its use as a chemopreventive agent.


Subject(s)
Adenomatous Polyposis Coli/drug therapy , Adenomatous Polyposis Coli/metabolism , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Blood Proteins/metabolism , Proteomics , Pyrazoles/pharmacology , Sulfonamides/pharmacology , Adenomatous Polyposis Coli/pathology , Blotting, Western , Celecoxib , Chromatography, Liquid , Computational Biology , Humans , Peptide Fragments , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
16.
Curr Biol ; 15(23): 2164-9, 2005 Dec 06.
Article in English | MEDLINE | ID: mdl-16332543

ABSTRACT

Arf1 is a GTP binding protein that functions at a number of cellular sites to control membrane traffic and actin remodeling. Arf1 is regulated by site-specific GTPase-activating proteins (GAPs). The combined results of crystallographic and biochemical studies have led to the proposal that Arf1 GAPs differ in the specific interface formed with Arf1. To test this hypothesis, we have used mutagenesis to examine the interaction of three Arf GAPs (ASAP1, AGAP1, and ArfGAP1) with switch 1, switch 2, and alpha helix3 of Arf1. The GAPs were similar in being affected by mutations in switch 1 and 2. However, effects of a mutation within alpha helix3 and specific mutations within switch 1 and 2 differed among the GAPs. The largest differences were observed with a change of isoleucine 46 to aspartate ([I46D]Arf1), which reduced ASAP1-induced catalysis by approximately 10,000-fold but had a 3-fold effect on AGAP1. The reduction was due to an isolated effect on the catalytic rate, k(cat). In vivo [I46D]Arf1 had no detectable effect on the Golgi apparatus but, instead, functioned as a constitutively active mutant in the cell periphery, affecting the localization of ASAP1 and paxillin. Based on our results, we conclude that the contribution of specific residues within switch 1 of Arf to binding and achieving a transition state toward GTP hydrolysis differs among Arf GAPs.


Subject(s)
ADP-Ribosylation Factor 1/genetics , ADP-Ribosylation Factor 1/metabolism , ADP-Ribosylation Factors/metabolism , Adaptor Proteins, Signal Transducing/metabolism , GTPase-Activating Proteins/metabolism , Models, Molecular , Animals , Catalysis , DNA Mutational Analysis , Fluorescent Antibody Technique , Golgi Apparatus/metabolism , Guanosine Triphosphate/metabolism , Humans , Mice , Mutation/genetics , NIH 3T3 Cells
17.
Cancer Epidemiol Biomarkers Prev ; 17(11): 3051-61, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18957522

ABSTRACT

It is well established that celecoxib, a selective inhibitor of cyclooxygenase-2 (COX-2) and a tested chemopreventive agent, has several COX-2-independent activities. In an attempt to better understand COX-2-independent molecular mechanisms underlying the chemopreventive activity of celecoxib, we did global transcription profiling of celecoxib-treated COX-2-positive and COX-2-deficient colorectal cancer cell lines. Celecoxib treatment resulted in significantly altered expression levels of over 1,000 to 3,000 transcripts in these cell lines, respectively. A pathway/functional analysis of celecoxib-affected transcripts, using Gene Ontology and Biocarta Pathways and exploring biological association networks, revealed that celecoxib modulates expression of numerous genes involved in a variety of cellular processes, including metabolism, cell proliferation, apoptotic signaling, cell cycle check points, lymphocyte activation, and signaling pathways. Among these processes, cell proliferation and apoptotic signaling consistently ranked as the highest-scoring Gene Ontology terms and Biocarta Pathways in both COX-2 expresser and nonexpresser cell lines. Altered expression of many of the genes by celecoxib was confirmed by quantitative PCR and at the protein level by Western blotting. Many novel genes emerged from our analysis of global transcription patterns that were not previously reported to be affected by celecoxib. In the future, in-depth work on selected genes will determine if these genes may serve as potential molecular targets for more effective chemopreventive strategies.


Subject(s)
Colonic Neoplasms/genetics , Cyclooxygenase 2 Inhibitors/pharmacology , Gene Expression Profiling , Pyrazoles/pharmacology , Sulfonamides/pharmacology , Blotting, Western , Celecoxib , Cell Line, Tumor/metabolism , Colonic Neoplasms/chemistry , Colonic Neoplasms/prevention & control , Humans , Microarray Analysis , Polymerase Chain Reaction , Tumor Cells, Cultured
18.
Cell Death Dis ; 8(6): e2875, 2017 06 15.
Article in English | MEDLINE | ID: mdl-28617445

ABSTRACT

BRCA2 encodes a protein with a fundamental role in homologous recombination that is essential for normal development. Carrier status of mutations in BRCA2 is associated with familial breast and ovarian cancer, while bi-allelic BRCA2 mutations can cause Fanconi anemia (FA), a cancer predisposition syndrome with cellular cross-linker hypersensitivity. Cancers associated with BRCA2 mutations can acquire chemo-resistance on relapse. We modeled acquired cross-linker resistance with an FA-derived BRCA2-mutated acute myeloid leukemia (AML) platform. Associated with acquired cross-linker resistance was the expression of a functional BRCA2 protein variant lacking exon 5 and exon 7 (BRCA2ΔE5+7), implying a role for BRCA2 splicing for acquired chemo-resistance. Integrated network analysis of transcriptomic and proteomic differences for phenotyping of BRCA2 disruption infers impact on transcription and chromatin remodeling in addition to the DNA damage response. The striking overlap with transcriptional profiles of FA patient hematopoiesis and BRCA mutation associated ovarian cancer helps define and explicate the 'BRCAness' profile.


Subject(s)
Alternative Splicing , BRCA2 Protein/genetics , BRCA2 Protein/metabolism , Drug Resistance, Neoplasm , Genes, BRCA2 , Mutation , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line, Tumor , DNA Damage , Exons , Fanconi Anemia/drug therapy , Fanconi Anemia/genetics , Fanconi Anemia/metabolism , Female , Genetic Predisposition to Disease , Humans , Introns , K562 Cells , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Mice , Neoplasm Recurrence, Local , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Phenotype , RNA Splicing , Transcription, Genetic
19.
Cell Signal ; 17(10): 1276-88, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16038802

ABSTRACT

ASAP1 is an Arf GAP with a PH domain immediately N-terminal to the catalytic Arf GAP domain. PH domains are thought to regulate enzymes by binding to specific phosphoinositide lipids in membranes, thereby recruiting the enzyme to a site of action. Here, we have examined the functional relationship between the PH and Arf GAP domains. We found that GAP activity requires the cognate PH domain of ASAP1, leading us to hypothesize that the Arf GAP and PH domains directly interact to form the substrate binding site. This hypothesis was supported by the combined results of protection and hydrodynamic studies. We then examined the role of the PH domain in the regulation of Arf GAP activity. The results of saturation kinetics, limited proteolysis, FRET and fluorescence spectrometry support a model in which regulation of the GAP activity of ASAP1 involves a conformational change coincident with recruitment to a membrane surface, and a second conformational change following the specific binding of phosphatidylinositol 4,5-bisphosphate.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Phospholipids/metabolism , ADP-Ribosylation Factor 1/metabolism , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/genetics , Amino Acid Sequence , Binding Sites/genetics , Binding, Competitive/drug effects , Biotin/analogs & derivatives , Biotin/chemistry , Blood Proteins/metabolism , Catalytic Domain , Fluorescence Resonance Energy Transfer , GTPase-Activating Proteins/metabolism , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Guanosine 5'-O-(3-Thiotriphosphate)/pharmacology , Guanosine Triphosphate/metabolism , Guanosine Triphosphate/pharmacology , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , Kinetics , Liposomes/metabolism , Liposomes/pharmacology , Lysine/chemistry , Models, Molecular , Molecular Sequence Data , Mutation/genetics , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Phosphatidic Acids/metabolism , Phosphatidylinositol 4,5-Diphosphate/analogs & derivatives , Phosphatidylinositol 4,5-Diphosphate/metabolism , Phospholipase C delta , Phospholipids/pharmacology , Phosphoproteins/metabolism , Plasmids/genetics , Protein Binding/drug effects , Protein Conformation , Protein Structure, Tertiary , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Homology, Amino Acid , Succinimides/chemistry , Type C Phospholipases/genetics , Type C Phospholipases/metabolism
20.
Nat Commun ; 7: 12425, 2016 08 08.
Article in English | MEDLINE | ID: mdl-27498558

ABSTRACT

Poly (ADP-ribose) polymerase (PARP) inhibitor (PARPi) olaparib has been approved for treatment of advanced ovarian cancer associated with BRCA1 and BRCA2 mutations. BRCA1- and BRCA2-mutated cells, which are homologous recombination (HR) deficient, are hypersensitive to PARPi through the mechanism of synthetic lethality. Here we examine the effect of PARPi on HR-proficient cells. Olaparib pretreatment, PARP1 knockdown or Parp1 heterozygosity of Brca2(cko/ko) mouse embryonic stem cells (mESCs), carrying a null (ko) and a conditional (cko) allele of Brca2, results in viable Brca2(ko/ko) cells. PARP1 deficiency does not restore HR in Brca2(ko/ko) cells, but protects stalled replication forks from MRE11-mediated degradation through its impaired recruitment. The functional consequence of Parp1 heterozygosity on BRCA2 loss is demonstrated by a significant increase in tumorigenesis in Brca2(cko/cko) mice. Thus, while olaparib efficiently kills BRCA2-deficient cells, we demonstrate that it can also contribute to the synthetic viability if PARP is inhibited before BRCA2 loss.


Subject(s)
BRCA2 Protein/deficiency , Poly (ADP-Ribose) Polymerase-1/deficiency , Animals , BRCA2 Protein/metabolism , Cell Survival/drug effects , DNA Replication/drug effects , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Homologous Recombination/drug effects , Humans , Integrases/metabolism , MRE11 Homologue Protein/metabolism , Mice , Models, Biological , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/drug effects , Mouse Embryonic Stem Cells/metabolism , Poly (ADP-Ribose) Polymerase-1/antagonists & inhibitors , Poly (ADP-Ribose) Polymerase-1/metabolism , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL