Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Front Genet ; 15: 1276365, 2024.
Article in English | MEDLINE | ID: mdl-38577247

ABSTRACT

Background: Maintenance of the genome is essential for cell survival, and impairment of the DNA damage response is associated with multiple pathologies including cancer and neurological abnormalities. DNA-PKcs is a DNA repair protein and a core component of the classical nonhomologous end-joining pathway, but it also has roles in modulating gene expression and thus, the overall cellular response to DNA damage. Methods: Using cells producing either wild-type (WT) or kinase-inactive (KR) DNA-PKcs, we assessed global alterations in gene expression in the absence or presence of DNA damage. We evaluated differential gene expression in untreated cells and observed differences in genes associated with cellular adhesion, cell cycle regulation, and inflammation-related pathways. Following exposure to etoposide, we compared how KR versus WT cells responded transcriptionally to DNA damage. Results: Downregulated genes were mostly involved in protein, sugar, and nucleic acid biosynthesis pathways in both genotypes, but enriched biological pathways were divergent, again with KR cells manifesting a more robust inflammatory response compared to WT cells. To determine what major transcriptional regulators are controlling the differences in gene expression noted, we used pathway analysis and found that many master regulators of histone modifications, proinflammatory pathways, cell cycle regulation, Wnt/ß-catenin signaling, and cellular development and differentiation were impacted by DNA-PKcs status. Finally, we have used qPCR to validate selected genes among the differentially regulated pathways to validate RNA sequence data. Conclusion: Overall, our results indicate that DNA-PKcs, in a kinase-dependent fashion, decreases proinflammatory signaling following genotoxic insult. As multiple DNA-PK kinase inhibitors are in clinical trials as cancer therapeutics utilized in combination with DNA damaging agents, understanding the transcriptional response when DNA-PKcs cannot phosphorylate downstream targets will inform the overall patient response to combined treatment.

2.
Pilot Feasibility Stud ; 10(1): 29, 2024 Feb 12.
Article in English | MEDLINE | ID: mdl-38347582

ABSTRACT

BACKGROUND: An advanced cancer diagnosis can be associated with a significant profile of distress. Psychedelic compounds have shown clinically significant effects in the treatment of psychological distress in patients with advanced-stage cancer. Given the challenges of delivering timely and effective intervention in the advanced cancer context, it is possible that an alternative, more pragmatic, approach lies in psychedelic 'microdosing'. Microdosing refers to repeated administration of psychedelics in sub-hallucinogenic doses. The purpose of this study is to evaluate the feasibility of conducting a full-scale randomised controlled trial comparing psychedelic microdose-assisted-meaning-centred psychotherapy (PA-MCP) to standard meaning-centred psychotherapy (MCP) in New Zealand indigenous (Maori) and non-indigenous people with advanced cancer and symptoms of anxiety and/or depression. Although MCP is a well-established psychotherapeutic treatment in advanced cancer populations, the potential efficacy and effectiveness of this therapy when delivered alongside a standardised microdose regimen of a psychedelic compound have not been investigated. METHODS: Participants with advanced-stage cancer and symptoms of anxiety and/or depression (N = 40; 20 Maori, 20 non-Maori) will be randomised under double-blind conditions to receive 7 sessions of MCP alongside 13 doses of either an LSD microdose (4-20 µg) (PA-MCP) or inactive placebo (placebo-MCP). The feasibility, acceptability, and safety of this intervention and physiological and psychological measures will be recorded at baseline, at each session of MCP, and at a 1-month and 6-month follow-up. DISCUSSION: Our findings will evaluate the feasibility, acceptability, and safety of a larger randomised controlled trial and provide an initial indication of the potential benefits of psychedelic microdosing for psychological distress in advanced-stage indigenous and non-indigenous cancer patients. TRIAL REGISTRATION: NZCTR, ACTRN12623000478617. Registered 11 May 2023.  https://www.anzctr.org.au/Trial/Registration/TrialReview.aspx?id=385810&isReview=true .

3.
Oncogene ; 41(24): 3355-3369, 2022 06.
Article in English | MEDLINE | ID: mdl-35538224

ABSTRACT

The oncogene Ras and the tumor suppressor gene p53 are frequently co-mutated in human cancer and mutations in Ras and p53 can cooperate to generate a more malignant cell state. To discover novel druggable targets for cancers carrying co-mutations in Ras and p53, we performed arrayed, kinome focused siRNA and oncology drug phenotypic screening utilizing a set of syngeneic Ras mutant squamous cell carcinoma (SCC) cell lines that also carried co-mutations in selected p53 pathway genes. These cell lines were derived from SCCs from carcinogen-treated inbred mice which harbored germline deletions or mutations in Trp53, p19Arf, Atm, or Prkdc. Both siRNA and drug phenotypic screening converge to implicate the phosphoinositol kinases, receptor tyrosine kinases, MAP kinases, as well as cell cycle and DNA damage response genes as targetable dependencies in SCC. Differences in functional kinome profiles between Ras mutant cell lines reflect incomplete penetrance of Ras synthetic lethal kinases and indicate that co-mutations cause a rewiring of survival pathways in Ras mutant tumors. This study describes the functional kinomic landscape of Ras/p53 mutant chemically-induced squamous cell carcinoma in both the baseline unperturbed state and following DNA damage and nominates candidate therapeutic targets, including the Nek4 kinase, for further development.


Subject(s)
Carcinoma, Squamous Cell , Tumor Suppressor Protein p53 , ras Proteins , Animals , Carcinoma, Squamous Cell/enzymology , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p16/genetics , Humans , Mice , Mutation , RNA, Small Interfering , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , ras Proteins/genetics
4.
J Appl Genet ; 63(1): 141-144, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34817771

ABSTRACT

The nonhomologous end-joining pathway is a primary DNA double-strand break repair pathway in eukaryotes. DNA ligase IV (Lig4) catalyzes the final step of DNA end ligation in this pathway. Partial loss of Lig4 in mammals causes Lig4 syndrome, while complete loss is embryonically lethal. DNA ligase 4 (DNAlig4) null Drosophila melanogaster is viable, but sensitive to ionizing radiation during early development. We proposed to explore if DNAlig4 loss induced other long-term sensitivities and defects in D. melanogaster. We demonstrated that DNAlig4 mutant strains had decreased lifespan and lower resistance to nutrient deprivation, indicating Lig4 is required for maintaining health and longevity in D. melanogaster.


Subject(s)
Drosophila melanogaster , Longevity , Animals , DNA End-Joining Repair , DNA Ligase ATP/genetics , DNA Ligase ATP/metabolism , DNA Ligases/genetics , DNA Ligases/metabolism , DNA Repair/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Longevity/genetics , Mutation , Nutrients
5.
J Nucleic Acids ; 2019: 9170341, 2019.
Article in English | MEDLINE | ID: mdl-30838131

ABSTRACT

DNA damage is a hallmark of cancer, and mutation and misregulation of proteins that maintain genomic fidelity are associated with the development of multiple cancers. DNA double strand breaks are arguably considered the most deleterious type of DNA damage. The nonhomologous end-joining (NHEJ) pathway is one mechanism to repair DNA double strand breaks, and proteins involved in NHEJ may also regulate DNA replication. We previously established that DNA-PKcs, a NHEJ protein, promotes genomic stability and cell viability following cellular exposure to replication stress; we wanted to discern whether another NHEJ protein, DNA ligase IV (Lig4), shares this phenotype. Our investigations focused on triple negative breast cancer cells, as, compared to nonbasal breast cancer, LIG4 is frequently amplified, and an increased gene dose is associated with higher Lig4 expression. We depleted Lig4 using siRNA and confirmed our knockdown by qPCR and western blotting. Cell survival diminished with Lig4 depletion alone, and this was associated with increased replication fork stalling. Checkpoint protein Chk1 activation and dephosphorylation were unchanged in Lig4-depleted cells. Lig4 depletion resulted in sustained DNA-PKcs phosphorylation following hydroxyurea exposure. Understanding the effect of Lig4 on genomic replication and the replication stress response will clarify the biological ramifications of inhibiting Lig4 activity. In addition, Lig4 is an attractive clinical target for directing CRISPR/Cas9-mediated repair towards homology-directed repair and away from NHEJ, thus understanding of how diminishing Lig4 impacts cell biology is critical.

6.
Cell Cycle ; 17(3): 275-276, 2018.
Article in English | MEDLINE | ID: mdl-29265907
7.
Cell Death Differ ; 24(11): 1853-1860, 2017 11.
Article in English | MEDLINE | ID: mdl-28686579

ABSTRACT

Ionizing radiation (IR) is one of the most widely used treatments for cancer. However, acute damage to the gastrointestinal tract or gastrointestinal acute radiation syndrome (GI-ARS) is a major dose-limiting side effect, and the mechanisms that underlie this remain unclear. Here we use mouse models to explore the relative roles of DNA repair, apoptosis, and cell cycle arrest in radiation response. IR induces DNA double strand breaks and DNA-PK mutant Prkdcscid/scid mice are sensitive to GI-ARS due to an inability to repair these breaks. IR also activates the tumor suppressor p53 to trigger apoptotic cell death within intestinal crypt cells and p53 deficient mice are resistant to apoptosis. To determine if DNA-PK and p53 interact to govern radiosensitivity, we compared the response of single and compound mutant mice to 8 Gy IR. Compound mutant Prkdcscid/scid/Trp53-/-mice died earliest due to severe GI-ARS. While both Prkdcscid/scid and Prkdcscid/scid/Trp53-/-mutant mice had higher levels of IR-induced DNA damage, particularly within the stem cell compartment of the intestinal crypt, in Prkdcscid/scid/Trp53-/-mice these damaged cells abnormally progressed through the cell cycle resulting in mitotic cell death. This led to a loss of Paneth cells and a failure to regenerate the differentiated epithelial cells required for intestinal function. IR-induced apoptosis did not correlate with radiosensitivity. Overall, these data reveal that DNA repair, mediated by DNA-PK, and cell cycle arrest, mediated by p53, cooperate to protect the stem cell niche after DNA damage, suggesting combination approaches to modulate both pathways may be beneficial to reduce GI-ARS. As many cancers harbor p53 mutations, this also suggests targeting DNA-PK may be effective to enhance sensitivity of p53 mutant tumors to radiation.


Subject(s)
Acute Radiation Syndrome/metabolism , Acute Radiation Syndrome/pathology , DNA-Activated Protein Kinase/metabolism , DNA-Binding Proteins/metabolism , Gastrointestinal Tract/pathology , Nuclear Proteins/metabolism , Radiation, Ionizing , Stem Cells/metabolism , Stem Cells/pathology , Tumor Suppressor Protein p53/metabolism , Animals , Apoptosis/radiation effects , Cell Proliferation/radiation effects , DNA Damage , Gastrointestinal Tract/radiation effects , Histones/metabolism , Inhibitor of Apoptosis Proteins/metabolism , Mice, Inbred C57BL , Mitosis , Mitotic Index , Paneth Cells/metabolism , Phenotype , Phosphorylation/radiation effects , Radiation Tolerance/radiation effects , Receptors, G-Protein-Coupled/metabolism , Repressor Proteins/metabolism , Staining and Labeling , Stem Cell Niche/radiation effects , Stem Cells/radiation effects , Survivin
8.
Gene ; 591(1): 6-13, 2016 Oct 10.
Article in English | MEDLINE | ID: mdl-27349565

ABSTRACT

Progesterone (P4), a steroid produced during estrous cycles and gestation for maintenance of pregnancy, also plays key roles in breast development to allow lactation post-parturition. Progestins (P4 and related steroids) are also implicated in breast cancer etiology. Hormone replacement therapy containing both estrogen and progestins increases breast cancer incidence while estrogen hormone therapy lowers breast cancer risk. P4 signaling via nuclear P4 receptors (PRs) has been extensively studied in breast cancer, however, progestin signaling via non-classical membrane bound progestin receptors (MPRs and PGRMC1) remains unclear. Moreover, P4 metabolites and synthetic progestins may bind membrane progestin receptors. We hypothesized that PR-negative breast epithelial cells express non-classical progestin receptors, which activate intracellular signaling pathways differently depending on nature of progestin. Therefore, our objectives for the current study were to determine expression of MPRs and PGRMC1 in two PR-negative non-tumorigenic breast epithelial cell lines, assess progestin-mediated signaling and biological functions. We determined five MPR isoforms and PGRMC1 were present in MCF10A cells and all progestin receptors but MPRß in MCF12A cells. MCF10A and MCF12A cells were treated with P4, select P4 metabolites (5αP and 3αHP), medroxyprogesterone acetate (MPA), or a specific MPR-Agonist (MPR-Ag) and phosphorylation of ERK, p38, JNK, and AKT was characterized following treatment. To our knowledge this is the first report of ERK and JNK activation in MCF10A and MCF12A cells with P4, P4 metabolites, MPA, and MPR-Ag. Activation of ERK and JNK in cells treated with MPR-Ag implicates MPRs may serve as the receptors responsible for their activation. In contrast, p38 activation varied with cell type and with progestin treatment. P4 and MPA promoted AKT phosphorylation in the MCF12A cell line only whereas no activation was observed in MCF10A cells. Interestingly, cellular proliferation increased in MCF10A cells treated with MPA or 5αP, while MPR-Ag tended to slightly decrease proliferation. Collectively, our data highlights the importance of investigating the effects of synthetic progestins in breast cancer biology. Our results add to the understanding that various progestins have on breast epithelial cells and underscores the importance of considering both membrane bound receptors and progestin type in breast cancer development.


Subject(s)
Breast/cytology , Cell Membrane/metabolism , Epithelial Cells/metabolism , Mitogen-Activated Protein Kinases/metabolism , Progestins/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Progesterone/metabolism , 5-alpha-Dihydroprogesterone , Cell Line , Cell Membrane/drug effects , Cell Survival/drug effects , Enzyme Activation/drug effects , Epithelial Cells/drug effects , Female , Humans , Medroxyprogesterone Acetate , Phosphorylation/drug effects
9.
Reproduction ; 150(1): 43-52, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25861798

ABSTRACT

Early gestation is a critical period when implantation and placental vascularization are established, processes influenced by progesterone (P4). Although human chorionic gonadotropin (hCG) is not endogenously synthesized by livestock, it binds the LH receptor, stimulating P4 synthesis. We hypothesized treating pregnant ewes with hCG would increase serum P4, number of corpora lutea (CLs) and concepti, augment steroidogenic enzymes, and increase membrane P4 receptors (PAQRs) and angiogenic factors in reproductive tissues. The objective was to determine molecular alterations induced by hCG in pregnant sheep that may promote pregnancy. Ewes received either 600 IU of hCG or saline i.m. on day 4 post mating. Blood samples were collected daily from day 0 until tissue collection for serum P4 analysis. Reproductive tissues were collected on either day 13 or 25 of gestation and analyzed for PAQRs, CXCR4, proangiogenic factors and steroidogenic enzymes. Ewes receiving hCG had more CL and greater serum P4, which remained elevated. On day 25, StAR protein production decreased in CL from hCG-treated ewes while HSD3B1 was unchanged; further, expression of CXCR4 significantly increased and KDR tended to increase. PAQR7 and CXCR4 protein was increased in caruncle tissue from hCG-treated ewes. Maternal hCG exposure influenced fetal extraembryonic tissues, as VEGFA, VEGFB, FLT1, and ANGPT1 expression increased. Our results indicate hCG increases serum P4 due to augmented CL number per ewe. hCG treatment resulted in greater PAQR7 and CXCR4 in maternal endometrium and promoted expression of proangiogenic factors in fetal extraembryonic membranes. Supplementing livestock with hCG may boost P4 levels and improve reproductive efficiency.


Subject(s)
Chorionic Gonadotropin/pharmacology , Corpus Luteum/drug effects , Progesterone/blood , Receptors, Vascular Endothelial Growth Factor/metabolism , Vascular Endothelial Growth Factor A/metabolism , Animals , Corpus Luteum/metabolism , Female , Multienzyme Complexes/metabolism , Phosphoproteins/metabolism , Placenta/metabolism , Pregnancy , Progesterone Reductase/metabolism , Receptors, LH/metabolism , Receptors, Progesterone/metabolism , Sheep , Steroid Isomerases/metabolism
10.
DNA Repair (Amst) ; 21: 131-9, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24819595

ABSTRACT

Genotoxins and other factors cause replication stress that activate the DNA damage response (DDR), comprising checkpoint and repair systems. The DDR suppresses cancer by promoting genome stability, and it regulates tumor resistance to chemo- and radiotherapy. Three members of the phosphatidylinositol 3-kinase-related kinase (PIKK) family, ATM, ATR, and DNA-PK, are important DDR proteins. A key PIKK target is replication protein A (RPA), which binds single-stranded DNA and functions in DNA replication, DNA repair, and checkpoint signaling. An early response to replication stress is ATR activation, which occurs when RPA accumulates on ssDNA. Activated ATR phosphorylates many targets, including the RPA32 subunit of RPA, leading to Chk1 activation and replication arrest. DNA-PK also phosphorylates RPA32 in response to replication stress, and we demonstrate that cells with DNA-PK defects, or lacking RPA32 Ser4/Ser8 targeted by DNA-PK, confer similar phenotypes, including defective replication checkpoint arrest, hyper-recombination, premature replication fork restart, failure to block late origin firing, and increased mitotic catastrophe. We present evidence that hyper-recombination in these mutants is ATM-dependent, but the other defects are ATM-independent. These results indicate that DNA-PK and ATR signaling through RPA32 plays a critical role in promoting genome stability and cell survival in response to replication stress.


Subject(s)
DNA Replication , DNA-Activated Protein Kinase/metabolism , G2 Phase Cell Cycle Checkpoints , Homologous Recombination , Nuclear Proteins/metabolism , Replication Protein A/metabolism , Animals , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , CHO Cells , Cell Line, Tumor , Checkpoint Kinase 1 , Cricetinae , Cricetulus , DNA-Activated Protein Kinase/genetics , Humans , Mutation , Nuclear Proteins/genetics , Phosphorylation , Protein Kinases/genetics , Protein Kinases/metabolism , Replication Protein A/genetics , Serine/genetics , Serine/metabolism
11.
Nucleic Acids Res ; 40(21): 10780-94, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22977173

ABSTRACT

DNA damage encountered by DNA replication forks poses risks of genome destabilization, a precursor to carcinogenesis. Damage checkpoint systems cause cell cycle arrest, promote repair and induce programed cell death when damage is severe. Checkpoints are critical parts of the DNA damage response network that act to suppress cancer. DNA damage and perturbation of replication machinery causes replication stress, characterized by accumulation of single-stranded DNA bound by replication protein A (RPA), which triggers activation of ataxia telangiectasia and Rad3 related (ATR) and phosphorylation of the RPA32, subunit of RPA, leading to Chk1 activation and arrest. DNA-dependent protein kinase catalytic subunit (DNA-PKcs) [a kinase related to ataxia telangiectasia mutated (ATM) and ATR] has well characterized roles in DNA double-strand break repair, but poorly understood roles in replication stress-induced RPA phosphorylation. We show that DNA-PKcs mutant cells fail to arrest replication following stress, and mutations in RPA32 phosphorylation sites targeted by DNA-PKcs increase the proportion of cells in mitosis, impair ATR signaling to Chk1 and confer a G2/M arrest defect. Inhibition of ATR and DNA-PK (but not ATM), mimic the defects observed in cells expressing mutant RPA32. Cells expressing mutant RPA32 or DNA-PKcs show sustained H2AX phosphorylation in response to replication stress that persists in cells entering mitosis, indicating inappropriate mitotic entry with unrepaired damage.


Subject(s)
Cell Cycle Proteins/metabolism , DNA Replication , DNA-Activated Protein Kinase/metabolism , DNA-Binding Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Replication Protein A/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Ataxia Telangiectasia Mutated Proteins , CHO Cells , Cell Cycle Checkpoints , Checkpoint Kinase 1 , Cricetinae , Cricetulus , DNA Breaks, Double-Stranded , DNA Repair , Humans , Mitosis , Mutation , Phosphorylation , Protein Kinases/metabolism , Replication Protein A/chemistry , Replication Protein A/genetics , Serine/metabolism , Signal Transduction , Stress, Physiological
12.
Mar Drugs ; 9(11): 2397-2408, 2011.
Article in English | MEDLINE | ID: mdl-22163192

ABSTRACT

Type IIα DNA topoisomerase (TopoIIα) is among the most important clinical drug targets for the treatment of cancer. Recently, the DNA repair protein Metnase was shown to enhance TopoIIα activity and increase resistance to TopoIIα poisons. Using in vitro DNA decatenation assays we show that neoamphimedine potently inhibits TopoIIα-dependent DNA decatenation in the presence of Metnase. Cell proliferation assays demonstrate that neoamphimedine can inhibit Metnase-enhanced cell growth with an IC(50) of 0.5 µM. Additionally, we find that the apparent K(m) of TopoIIα for ATP increases linearly with higher concentrations of neoamphimedine, indicating ATP-competitive inhibition, which is substantiated by molecular modeling. These findings support the continued development of neoamphimedine as an anticancer agent, particularly in solid tumors that over-express Metnase.


Subject(s)
Acridines/pharmacology , Adenosine Triphosphate/metabolism , Antigens, Neoplasm/drug effects , DNA Topoisomerases, Type II/drug effects , DNA-Binding Proteins/drug effects , Histone-Lysine N-Methyltransferase/metabolism , Acridines/administration & dosage , Antigens, Neoplasm/metabolism , Cell Proliferation/drug effects , DNA Topoisomerases, Type II/metabolism , DNA-Binding Proteins/metabolism , Dose-Response Relationship, Drug , HEK293 Cells , Humans , In Vitro Techniques , Inhibitory Concentration 50 , Models, Molecular
13.
J Cancer ; 2: 262-70, 2011 May 04.
Article in English | MEDLINE | ID: mdl-21552385

ABSTRACT

Canine and human osteosarcoma (OSA) have many similarities, with the majority of reported cases occurring in the appendicular skeleton, gender predominance noted, high rate of metastasis at the time of presentation, and a lack of known etiology for this devastating disease. Due to poor understanding of the molecular mechanisms underlying OSA, we have characterized seven different OSA canine cell lines: Abrams, D17, Grey, Hughes, Ingles, Jarques, and Marisco and compared them to U2, a human OSA cell line, for the following parameters: morphology, growth, contact inhibition, migrational tendencies, alkaline phosphatase staining, heterologous tumor growth, double-strand DNA breaks, and oxidative damage. All results demonstrated the positive characteristics of the Abrams cell line for use in future studies of OSA. Of particular interest, the robust growth of a subcutaneous tumor and rapid pulmonary metastasis of the Abrams cell line in an immunocompromised mouse shows incredible potential for the future use of Abrams as a canine OSA model. Further investigations utilizing a canine cell model of OSA, such as Abrams, will be invaluable to understanding the molecular events underlying OSA, pharmaceutical inhibition of metastasis, and eventual prevention of this devastating disease.

14.
J Mol Cell Biol ; 3(1): 4-12, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21278446

ABSTRACT

High-fidelity replication of DNA, and its accurate segregation to daughter cells, is critical for maintaining genome stability and suppressing cancer. DNA replication forks are stalled by many DNA lesions, activating checkpoint proteins that stabilize stalled forks. Stalled forks may eventually collapse, producing a broken DNA end. Fork restart is typically mediated by proteins initially identified by their roles in homologous recombination repair of DNA double-strand breaks (DSBs). In recent years, several proteins involved in DSB repair by non-homologous end joining (NHEJ) have been implicated in the replication stress response, including DNA-PKcs, Ku, DNA Ligase IV-XRCC4, Artemis, XLF and Metnase. It is currently unclear whether NHEJ proteins are involved in the replication stress response through indirect (signaling) roles, and/or direct roles involving DNA end joining. Additional complexity in the replication stress response centers around RPA, which undergoes significant post-translational modification after stress, and RAD52, a conserved HR protein whose role in DSB repair may have shifted to another protein in higher eukaryotes, such as BRCA2, but retained its role in fork restart. Most cancer therapeutic strategies create DNA replication stress. Thus, it is imperative to gain a better understanding of replication stress response proteins and pathways to improve cancer therapy.


Subject(s)
DNA Replication , Animals , DNA Breaks, Double-Stranded , DNA Damage , DNA Ligases/metabolism , DNA Repair , DNA-Activated Protein Kinase/genetics , DNA-Activated Protein Kinase/metabolism , Humans , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Rad52 DNA Repair and Recombination Protein/genetics , Rad52 DNA Repair and Recombination Protein/metabolism , Signal Transduction
15.
Toxicol Lett ; 199(1): 17-21, 2010 Nov 10.
Article in English | MEDLINE | ID: mdl-20688138

ABSTRACT

Atrazine (ATRA) is the most commonly applied herbicide in the United States and is detected frequently in drinking water at significant levels. Following oral exposure, metabolism of ATRA generates diaminochlorotriazine (DACT), an electrophilic molecule capable of forming covalent protein adducts. At high doses, both ATRA and DACT can disrupt the preovulatory luteinizing hormone (LH) surge in rats, thereby altering normal reproductive function. This research was designed to identify DACT protein adducts formed in three distinct brain regions of ATRA-exposed rats, including the preoptic area (POA), medial basal hypothalamus (MBH), and cortex (CTX). Proteins with DACT adducts were identified following 2-dimensional electrophoresis (2-DE), immunodetection, and MALDI-TOF mass spectrometry analysis. Western blots from exposed animals revealed over 30 DACT-modified spots that were absent in controls. Protein spots were matched to concurrently run 2-DE gels stained with Sypro Ruby, excised, and in-gel digested with trypsin.


Subject(s)
Atrazine/analogs & derivatives , Atrazine/toxicity , Cerebral Cortex/drug effects , Hypothalamus, Middle/drug effects , Preoptic Area/drug effects , Proteins/metabolism , Proteomics/methods , Administration, Oral , Animals , Atrazine/administration & dosage , Atrazine/chemistry , Atrazine/metabolism , Blotting, Western , Cerebral Cortex/chemistry , Cerebral Cortex/metabolism , Electrophoresis, Gel, Two-Dimensional , Female , Herbicides/administration & dosage , Herbicides/toxicity , Hypothalamus, Middle/chemistry , Hypothalamus, Middle/metabolism , Immunoenzyme Techniques , Peptide Mapping , Preoptic Area/chemistry , Preoptic Area/metabolism , Proteins/chemistry , Rats , Rats, Wistar , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
16.
Toxicol Lett ; 184(3): 186-91, 2009 Feb 10.
Article in English | MEDLINE | ID: mdl-19063952

ABSTRACT

DJ-1 mutation induces early-onset Parkinson's disease, and conversely over-expression of DJ-1 is associated with cancer in numerous tissues. A gene-trap screening library conducted in embryonic stem cells was utilized for generation of a DJ-1 mutant mouse. Real-time PCR and immunoblotting were utilized to confirm functional mutation of the DJ-1 gene. Normal DJ-1 protein expression in adult mouse tissue was characterized and demonstrates high expression in brain tissue with wide systemic distribution. Primary astrocytes isolated from DJ-1(-/-) mice reveal a decreased nuclear localization of DJ-1 protein in response to rotenone or LPS, with a concomitant increase in mitochondrial localization of DJ-1 found only in the rotenone exposure. Resting mitochondrial membrane potential was significantly lower in DJ-1(-/-) astrocytes, as compared to controls. Our DJ-1 knockout mouse provides an exciting tool for exploring the molecular and physiological roles of DJ-1 to further explicate its functions in neurodegeneration.


Subject(s)
Astrocytes/metabolism , Cerebral Cortex/metabolism , Mutation , Oncogene Proteins/genetics , Animals , Astrocytes/drug effects , Cell Nucleus/metabolism , Cells, Cultured , Cerebral Cortex/drug effects , Dose-Response Relationship, Drug , Genotype , Lipopolysaccharides/pharmacology , Membrane Potential, Mitochondrial , Mice , Mice, Knockout , Mitochondria/metabolism , Oncogene Proteins/metabolism , Peroxiredoxins , Phenotype , Protein Deglycase DJ-1 , Protein Transport , Rotenone/pharmacology
17.
Arch Toxicol ; 82(2): 103-16, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17874069

ABSTRACT

The objectives of this study were twofold: (1) evaluating the carcinogenic potential of the mixture of two persistent environmental pollutants, hexachlorobenzene (HCB) and 3,3',4,4',5-pentachlorobiphenyl (PCB 126), in an initiation-promotion bioassay involving the development of pi glutathione S-transferase (GST-P) liver foci, and (2) analyzing the GST-P foci data using a biologically-based computer model (i.e., clonal growth model) with an emphasis on the effect of focal size on the growth kinetics of initiated cells. The 8-week bioassay involved a series of treatments of initiator, two-thirds partial hepatectomy, and daily oral gavage of the mixture of two doses in male F344 rats. The mixture treatment significantly increased liver GST-P foci development, indicating carcinogenic potential of this mixture. Our clonal growth model was developed to simulate the appearance and development of initiated GST-P cells in the liver over time. In the model, the initiated cells were partitioned into two subpopulations with the same division rate but different death rates. Each subpopulation was further categorized into single cells, mini- (2-11 cells), medium- (12-399 cells), and large-foci (>399 cells) with different growth kinetics. Our modeling suggested that the growth of GST-P foci is size-dependent; in general, the larger the foci, the higher the rate constants of division and death. In addition, the modeling implied that the two doses promoted foci development in different manners even though the experimental foci data appeared to be similar between the two doses. This study further illustrated how clonal growth modeling may facilitate our understanding in chemical carcinogenic process.


Subject(s)
Carcinogens/toxicity , Glutathione S-Transferase pi/drug effects , Hexachlorobenzene/toxicity , Liver/drug effects , Polychlorinated Biphenyls/toxicity , Administration, Oral , Animals , Biological Assay , Carcinogenicity Tests , Carcinogens/administration & dosage , Computer Simulation , Dose-Response Relationship, Drug , Environmental Pollutants/administration & dosage , Environmental Pollutants/toxicity , Glutathione S-Transferase pi/metabolism , Hexachlorobenzene/administration & dosage , Liver/pathology , Male , Models, Biological , Polychlorinated Biphenyls/administration & dosage , Rats , Rats, Inbred F344
18.
Fam Cancer ; 6(4): 473-82, 2007.
Article in English | MEDLINE | ID: mdl-17636425

ABSTRACT

CONTEXT: Models have been developed to predict the probability that a person carries a detectable germline mutation in the BRCA1 or BRCA2 genes. Their relative performance in a clinical setting is unclear. OBJECTIVE: To compare the performance characteristics of four BRCA1/BRCA2 gene mutation prediction models: LAMBDA, based on a checklist and scores developed from data on Ashkenazi Jewish (AJ) women; BRCAPRO, a Bayesian computer program; modified Couch tables based on regression analyses; and Myriad II tables collated by Myriad Genetics Laboratories. DESIGN AND SETTING: Family cancer history data were analyzed from 200 probands from the Mayo Clinic Familial Cancer Program, in a multispecialty tertiary care group practice. All probands had clinical testing for BRCA1 and BRCA2 mutations conducted in a single laboratory. MAIN OUTCOMES MEASURES: For each model, performance was assessed by the area under the receiver operator characteristic curve (ROC) and by tests of accuracy and dispersion. Cases "missed" by one or more models (model predicted less than 10% probability of mutation when a mutation was actually found) were compared across models. RESULTS: All models gave similar areas under the ROC curve of 0.71 to 0.76. All models except LAMBDA substantially under-predicted the numbers of carriers. All models were too dispersed. CONCLUSIONS: In terms of ranking, all prediction models performed reasonably well with similar performance characteristics. Model predictions were widely discrepant for some families. Review of cancer family histories by an experienced clinician continues to be vital to ensure that critical elements are not missed and that the most appropriate risk prediction figures are provided.


Subject(s)
BRCA1 Protein/genetics , BRCA2 Protein/genetics , Heterozygote , Models, Genetic , Mutation/genetics , Software , Adult , Computer Simulation , Female , Humans , Jews/genetics , Middle Aged
19.
Cancer ; 110(3): 670-7, 2007 Aug 01.
Article in English | MEDLINE | ID: mdl-17559146

ABSTRACT

BACKGROUND: Efficacy and toxicity of oxaliplatin (Eloxatin; Sanofi-Aventis, Paris, France) combined with irinotecan (IROX) were examined in 383 patients enrolled on the IROX arm of Intergroup Study N9741. METHODS: This IROX regimen was oxaliplatin 85 mg/m(2) and irinotecan 200 mg/m(2) administered every 3 weeks. The relation between adverse events on IROX to selected characteristics was analyzed. Time to progression (TTP), response rate, and overall survival for patients treated with IROX compared with patients treated with oxaliplatin with 5- fluorouracil (FOLFOX) were updated in this article. RESULTS: Grade >or=3 gastrointestinal and hematologic toxicities were common with 39% patients experiencing neutropenia, 28% diarrhea, and 21% vomiting. Patients ages >70 years experienced higher rates of grade >or=3 toxicity, with significantly higher rates of grade >or=3 hematologic toxicities (P = .02). Long-term toxicity was uncommon, and nearly all cases of grade >or=3 neurotoxicity resolved within 10 months. Fifty-two percent of patients required dose reductions for adverse events, and 26% experienced 119 hospitalizations related to complications of treatment or their disease, with 5 treatment-related deaths. This analysis confirmed prior findings that FOLFOX is superior to IROX in terms of response rate (43% vs 36%, p = 0.002), TTP (9.2 months vs 6.7 months, P < .0001), and overall survival (19.5 months vs 17.3 months, P = .0001). CONCLUSIONS: IROX was found to be less active than FOLFOX but with a similar toxicity profile except in patients ages >70 years. Although IROX may be considered in patients intolerant of 5-FU or in patients known to have a dihydropyrimidine dehydrogenase (DPD) deficiency, it should be used with caution in older patients.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Colorectal Neoplasms/drug therapy , Adult , Aged , Aged, 80 and over , Camptothecin/analogs & derivatives , Camptothecin/therapeutic use , Colorectal Neoplasms/secondary , Disease Progression , Female , Fluorouracil/therapeutic use , Humans , Leucovorin/therapeutic use , Male , Middle Aged , Organoplatinum Compounds/therapeutic use , Survival Rate , Treatment Outcome
20.
Clin Colorectal Cancer ; 6(1): 32-7, 2006 May.
Article in English | MEDLINE | ID: mdl-16796789

ABSTRACT

Colorectal cancer is the second leading cause of cancer-related deaths in Western countries. Approximately 35% of patients will have metastatic disease at diagnosis, and an additional 25% of patients with resected stage II/III disease will develop recurrence. In approximately 30% of patients, metastatic disease will be restricted to a single organ, with the liver and lungs accounting for the majority of single organ-site metastases. In recent years, aggressive surgical resection of pulmonary metastases has become increasingly common with the recognition that this offers the best chance of long-term cure despite recent chemotherapeutic advances. Unfortunately, relapse after pulmonary resection remains approximately 70% despite advances in imaging and surgical technique. This review examines prognostic factors that influence survival after resection and repeat resection of pulmonary colorectal metastases and examines the impact of lymph node metastases, chemotherapy, and hepatic metastases on outcome. Pathologic markers that might determine outcome and current literature, which consists mainly of retrospective institutional reports, is reviewed.


Subject(s)
Colorectal Neoplasms/pathology , Lung Neoplasms/secondary , Lung Neoplasms/surgery , Colorectal Neoplasms/microbiology , Humans , Lung Neoplasms/mortality , Lymphatic Metastasis , Prognosis , Survival Rate
SELECTION OF CITATIONS
SEARCH DETAIL
...