Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Oncotarget ; 8(6): 9243-9250, 2017 Feb 07.
Article in English | MEDLINE | ID: mdl-27999209

ABSTRACT

Oral cancers are easily accessible compared to many other cancers. Nevertheless, oral cancer is often diagnosed late, resulting in a poor prognosis. Most oral cancers are squamous cell carcinomas that predominantly develop from cell hyperplasias and dysplasias. DNA damage is induced in these tissues directly or indirectly in response to oncogene-induced deregulation of cellular proliferation. Consequently, a DNA Damage response (DDR) and a cell cycle checkpoint is activated. As dysplasia transitions to cancer, proteins involved in DNA damage and checkpoint signaling are mutated or silenced decreasing cell death while increasing genomic instability and allowing continued tumor progression. Hyperphosphorylation of Replication Protein A (RPA), including phosphorylation of Ser4 and Ser8 of RPA2, is a well-known indicator of DNA damage and checkpoint activation. In this study, we utilize S4S8-RPA phosphorylation as a marker for cancer development and progression in oral squamous cell carcinomas (OSCC). S4S8-RPA phosphorylation was observed to be low in normal cells, high in dysplasias, moderate in early grade tumors, and low in late stage tumors, essentially supporting the model of the DDR as an early barrier to tumorigenesis in certain types of cancers. In contrast, overall RPA expression was not correlative to DDR activation or tumor progression. Utilizing S4S8-RPA phosphorylation to indicate competent DDR activation in the future may have clinical significance in OSCC treatment decisions, by predicting the susceptibility of cancer cells to first-line platinum-based therapies for locally advanced, metastatic and recurrent OSCC.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma, Squamous Cell/metabolism , Head and Neck Neoplasms/metabolism , Mouth Neoplasms/metabolism , Replication Protein A/metabolism , Biomarkers, Tumor/genetics , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/therapy , Cell Line, Tumor , DNA Damage , DNA Repair , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/pathology , Head and Neck Neoplasms/therapy , Humans , Kaplan-Meier Estimate , Mouth Neoplasms/genetics , Mouth Neoplasms/pathology , Neoplasm Grading , Neoplasm Staging , Phosphorylation , Replication Protein A/genetics , Serine , Signal Transduction , Squamous Cell Carcinoma of Head and Neck , Time Factors
2.
J Antimicrob Chemother ; 71(12): 3432-3440, 2016 12.
Article in English | MEDLINE | ID: mdl-27609050

ABSTRACT

OBJECTIVES: The increasing threat of drug-resistant bacteria establishes a continuing need for the development of new strategies to fight infection. We examine the inhibition of the essential single-stranded DNA-binding proteins (SSBs) SSBA and SSBB as a potential antimicrobial therapy due to their importance in DNA replication, activating the SOS response and promoting competence-based mechanisms of resistance by incorporating new DNA. METHODS: Purified recombinant SSBs from Gram-positive (Staphylococcus aureus and Bacillus anthracis) and Gram-negative (Escherichia coli and Francisella tularensis) bacteria were assessed in a high-throughput screen for inhibition of duplex DNA unwinding by small molecule inhibitors. Secondary electrophoretic mobility shift assays further validated the top hits that were then tested for MICs using in vitro assays. RESULTS: We have identified compounds that show cross-reactivity in vitro, as well as inhibition of both F. tularensis and B. anthracis SSBA. Five compounds were moderately toxic to at least two of the four bacterial strains in vivo, including two compounds that were selectively non-toxic to human cells, 9-hydroxyphenylfluoron and purpurogallin. Three of the SSBA inhibitors also inhibited S. aureus SSBB in Gram-positive bacteria. CONCLUSIONS: Results from our study support the potential for SSB inhibitors as broad-spectrum antibacterial agents, with dual targeting capabilities against Gram-positive bacteria.


Subject(s)
Anti-Bacterial Agents/isolation & purification , Anti-Bacterial Agents/pharmacology , Bacteria/drug effects , Bacterial Proteins/antagonists & inhibitors , DNA-Binding Proteins/antagonists & inhibitors , Drug Evaluation, Preclinical/methods , High-Throughput Screening Assays , Microbial Sensitivity Tests
3.
Bioorg Med Chem ; 24(21): 5388-5392, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27642076

ABSTRACT

Ebola virus continues to be problematic as sporadic outbreaks in Africa continue to arise, and as terrorist organizations have considered the virus for bioterrorism use. Several proteins within the virus have been targeted for antiviral chemotherapy, including VP35, a dsRNA binding protein that promotes viral replication, protects dsRNA from degradation, and prevents detection of the viral genome by immune complexes. To augment the scope of our antiviral research, we have now employed molecular modeling techniques to enrich the population of compounds for further testing in vitro. In the initial docking of a static VP35 structure with an 80,000 compound library, 40 compounds were selected, of which four compounds inhibited VP35 with IC50 <200µM, with the best compounds having an IC50 of 20µM. By superimposing 26 VP35 structures, we determined four aspartic acid residues were highly flexible and the docking was repeated under flexible parameters. Of 14 compounds chosen for testing, five compounds inhibited VP35 with IC50 <200µM and one compound with an IC50 of 4µM. These studies demonstrate the value of docking in silico for enriching compounds for testing in vitro, and specifically using multiple structures as a guide for detecting flexibility and provide a foundation for further development of small molecule inhibitors directed towards VP35.


Subject(s)
Antiviral Agents/pharmacology , Computer Simulation , Nucleoproteins/antagonists & inhibitors , Viral Core Proteins/antagonists & inhibitors , Antiviral Agents/chemistry , Dose-Response Relationship, Drug , Molecular Docking Simulation , Molecular Structure , Nucleocapsid Proteins , Structure-Activity Relationship
4.
Cancer Res ; 74(18): 5165-72, 2014 Sep 15.
Article in English | MEDLINE | ID: mdl-25070753

ABSTRACT

The ATR/Chk1 pathway is a critical surveillance network that maintains genomic integrity during DNA replication by stabilizing the replication forks during normal replication to avoid replication stress. One of the many differences between normal cells and cancer cells is the amount of replication stress that occurs during replication. Cancer cells with activated oncogenes generate increased levels of replication stress. This creates an increased dependency on the ATR/Chk1 pathway in cancer cells and opens up an opportunity to preferentially kill cancer cells by inhibiting this pathway. In support of this idea, we have identified a small molecule termed HAMNO ((1Z)-1-[(2-hydroxyanilino)methylidene]naphthalen-2-one), a novel protein interaction inhibitor of replication protein A (RPA), a protein involved in the ATR/Chk1 pathway. HAMNO selectively binds the N-terminal domain of RPA70, effectively inhibiting critical RPA protein interactions that rely on this domain. HAMNO inhibits both ATR autophosphorylation and phosphorylation of RPA32 Ser33 by ATR. By itself, HAMNO treatment creates DNA replication stress in cancer cells that are already experiencing replication stress, but not in normal cells, and it acts synergistically with etoposide to kill cancer cells in vitro and slow tumor growth in vivo. Thus, HAMNO illustrates how RPA inhibitors represent candidate therapeutics for cancer treatment, providing disease selectivity in cancer cells by targeting their differential response to replication stress. Cancer Res; 74(18); 5165-72. ©2014 AACR.


Subject(s)
DNA Replication/drug effects , Naphthalenes/pharmacology , Replication Protein A/antagonists & inhibitors , Animals , Ataxia Telangiectasia Mutated Proteins/metabolism , Cell Growth Processes/drug effects , Checkpoint Kinase 1 , DNA Damage , Female , Humans , Mice , Mice, Nude , Models, Molecular , Phosphorylation , Protein Kinases/metabolism , Replication Protein A/genetics , Replication Protein A/metabolism , Signal Transduction/drug effects
5.
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
6.
Nucleic Acids Res ; 41(3): 2047-59, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23267009

ABSTRACT

Replication protein A (RPA), essential for DNA replication, repair and DNA damage signalling, possesses six ssDNA-binding domains (DBDs), including DBD-F on the N-terminus of the largest subunit, RPA70. This domain functions as a binding site for p53 and other DNA damage and repair proteins that contain amphipathic alpha helical domains. Here, we demonstrate direct binding of both ssDNA and the transactivation domain 2 of p53 (p53TAD2) to DBD-F, as well as DBD-F-directed dsDNA strand separation by RPA, all of which are inhibited by fumaropimaric acid (FPA). FPA binds directly to RPA, resulting in a conformational shift as determined through quenching of intrinsic tryptophan fluorescence in full length RPA. Structural analogues of FPA provide insight on chemical properties that are required for inhibition. Finally, we confirm the inability of RPA possessing R41E and R43E mutations to bind to p53, destabilize dsDNA and quench tryptophan fluorescence by FPA, suggesting that protein binding, DNA modulation and inhibitor binding all occur within the same site on DBD-F. The disruption of p53-RPA interactions by FPA may disturb the regulatory functions of p53 and RPA, thereby inhibiting cellular pathways that control the cell cycle and maintain the integrity of the human genome.


Subject(s)
Diterpenes/pharmacology , Replication Protein A/chemistry , Tumor Suppressor Protein p53/metabolism , Amino Acid Substitution , Binding Sites , Binding, Competitive , DNA, Single-Stranded/metabolism , Diterpenes/chemistry , Diterpenes/metabolism , Humans , Models, Molecular , Protein Structure, Tertiary , Replication Protein A/drug effects , Replication Protein A/genetics , Replication Protein A/metabolism , Tumor Suppressor Protein p53/chemistry
7.
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
8.
Bioorg Med Chem ; 19(8): 2589-95, 2011 Apr 15.
Article in English | MEDLINE | ID: mdl-21459001

ABSTRACT

The pharmacological suppression of the DNA damage response and DNA repair can increase the therapeutic indices of conventional chemotherapeutics. Replication Protein A (RPA), the major single-stranded DNA binding protein in eukaryotes, is required for DNA replication, DNA repair, DNA recombination, and DNA damage response signaling. Through the use of high-throughput screening of 1500 compounds, we have identified a small molecule inhibitor, 15-carboxy-13-isopropylatis-13-ene-17,18-dioic acid (NSC15520), that inhibited both the binding of Rad9-GST and p53-GST fusion proteins to the RPA N-terminal DNA binding domain (DBD), interactions that are essential for robust DNA damage signaling. NSC15520 competitively inhibited the binding of p53-GST peptide with an IC(50) of 10 µM. NSC15520 also inhibited helix destabilization of a duplex DNA (dsDNA) oligonucleotide, an activity dependent on the N-terminal domain of RPA70. NSC15520 did not inhibit RPA from binding single-stranded oligonucleotides, suggesting that the action of this inhibitor is specific for the N-terminal DBD of RPA, and does not bind to DBDs essential for single-strand DNA binding. Computer modeling implicates direct competition between NSC15520 and Rad9 for the same binding surface on RPA. Inhibitors of protein-protein interactions within the N-terminus of RPA are predicted to act synergistically with DNA damaging agents and inhibitors of DNA repair. Novel compounds such as NSC15520 have the potential to serve as chemosensitizing agents.


Subject(s)
Protein Interaction Domains and Motifs/drug effects , Replication Protein A/antagonists & inhibitors , Binding Sites , Computer Simulation , DNA Repair , DNA, Single-Stranded , Drug Evaluation, Preclinical/methods , High-Throughput Screening Assays , Humans , Oligonucleotides
9.
Head Neck ; 32(5): 636-45, 2010 May.
Article in English | MEDLINE | ID: mdl-19787780

ABSTRACT

BACKGROUND: Resistance to chemotherapy is a major limitation in the treatment of head and neck squamous cell carcinomas (HNSCCs), accounting for high mortality rates in patients. Here, we investigated the role of replication protein A (RPA) in cisplatin and etoposide resistance. METHODS: We used 6 parental HNSCC cell lines. We also generated 1 cisplatin-resistant progeny subline from a parental cisplatin-sensitive cell line, to examine cisplatin resistance and sensitivity with respect to RPA2 hyperphosphorylation and cell-cycle response. RESULTS: Cisplatin-resistant HNSCC cell levels of hyperphosphorylated RPA2 in response to cisplatin were 80% to 90% greater compared with cisplatin-sensitive cell lines. RPA2 hyperphosphorylation could be induced in the cisplatin-resistant HNSCC subline. The absence of RPA2 hyperphosphorylation correlated with a defect in cell-cycle progression and cell survival. CONCLUSION: Loss of RPA2 hyperphosphorylation occurs in HNSCC cells and may be a marker of cellular sensitivities to cisplatin and etoposide in HNSCC.


Subject(s)
Antineoplastic Agents/pharmacology , Cisplatin/pharmacology , Drug Resistance, Neoplasm , Etoposide/pharmacology , Replication Protein A/metabolism , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/pathology , Cell Cycle , Cell Line, Tumor , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/pathology , Humans , Phosphorylation
10.
Biochemistry ; 48(31): 7473-81, 2009 Aug 11.
Article in English | MEDLINE | ID: mdl-19586055

ABSTRACT

Replication protein A (RPA) is a heterotrimeric protein consisting of RPA1, RPA2, and RPA3 subunits that binds to single-stranded DNA (ssDNA) with high affinity. The response to replication stress requires the recruitment of RPA and the MRE11-RAD50-NBS1 (MRN) complex. RPA bound to ssDNA stabilizes stalled replication forks by recruiting checkpoint proteins involved in fork stabilization. MRN can bind DNA structures encountered at stalled or collapsed replication forks, such as ssDNA-double-stranded DNA (dsDNA) junctions or breaks, and promote the restart of DNA replication. Here, we demonstrate that RPA2 phosphorylation regulates the assembly of DNA damage-induced RPA and MRN foci. Using purified proteins, we observe a direct interaction between RPA with both NBS1 and MRE11. By utilizing RPA bound to ssDNA, we demonstrate that substituting RPA with phosphorylated RPA or a phosphomimetic weakens the interaction with the MRN complex. Also, the N-terminus of RPA1 is a critical component of the RPA-MRN protein-protein interaction. Deletion of the N-terminal oligonucleotide-oligosaccharide binding fold (OB-fold) of RPA1 abrogates interactions of RPA with MRN and individual proteins of the MRN complex. Further identification of residues critical for MRN binding in the N-terminus of RPA1 shows that substitution of Arg31 and Arg41 with alanines disrupts the RPA-MRN interaction and alters cell cycle progression in response to DNA damage. Thus, the N-terminus of RPA1 and phosphorylation of RPA2 regulate RPA-MRN interactions and are important in the response to DNA damage.


Subject(s)
DNA-Binding Proteins/metabolism , Peptide Fragments/metabolism , Protein Interaction Mapping , Protein Subunits/metabolism , Replication Protein A/metabolism , Amino Acid Sequence , Cell Cycle Proteins/metabolism , Cell Line, Tumor , DNA Damage , DNA-Binding Proteins/antagonists & inhibitors , HeLa Cells , Humans , MRE11 Homologue Protein , Molecular Sequence Data , Nuclear Proteins/metabolism , Phosphorylation , Replication Protein A/chemistry
11.
J Neuroimmune Pharmacol ; 3(2): 59-74, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18202920

ABSTRACT

Microglial neuroinflammatory responses affect the onset and progression of Parkinson's disease (PD). We posit that such neuroinflammatory responses are, in part, mediated by microglial interactions with nitrated and aggregated alpha-synuclein (alpha-syn) released from Lewy bodies as a consequence of dopaminergic neuronal degeneration. As disease progresses, secretions from alpha-syn-activated microglia can engage neighboring glial cells in a cycle of autocrine and paracrine amplification of neurotoxic immune products. Such pathogenic processes affect the balance between a microglial neurotrophic and neurotoxic signature. We now report that microglia secrete both neurotoxic and neuroprotective factors after exposure to nitrated alpha-syn (N-alpha-syn). Proteomic (surface enhanced laser desorption-time of flight, 1D sodium dodecyl sulfate electrophoresis, and liquid chromatography-tandem mass spectrometry) and limited metabolomic profiling demonstrated that N-alpha-syn-activated microglia secrete inflammatory, regulatory, redox-active, enzymatic, and cytoskeletal proteins. Increased extracellular glutamate and cysteine and diminished intracellular glutathione and secreted exosomal proteins were also demonstrated. Increased redox-active proteins suggest regulatory microglial responses to N-alpha-syn. These were linked to discontinuous cystatin expression, cathepsin activity, and nuclear factor-kappa B activation. Inhibition of cathepsin B attenuated, in part, N-alpha-syn microglial neurotoxicity. These data support multifaceted microglia functions in PD-associated neurodegeneration.


Subject(s)
Microglia/drug effects , Nerve Tissue Proteins/metabolism , Nitrates/pharmacology , alpha-Synuclein/pharmacology , Animals , Cathepsin B/antagonists & inhibitors , Cathepsin B/physiology , Cells, Cultured/drug effects , Cystatins/biosynthesis , Cystatins/genetics , Cysteine/metabolism , Cytoskeletal Proteins/biosynthesis , Cytoskeletal Proteins/genetics , Dopamine/physiology , Gene Expression Regulation/drug effects , Glutamic Acid/metabolism , Glutathione/analysis , Mice , Mice, Inbred C57BL , Microglia/metabolism , NF-kappa B/metabolism , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Nitrates/toxicity , Oxidation-Reduction , Oxidative Stress , Parkinson Disease/physiopathology , Protein Array Analysis , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Tandem Mass Spectrometry , alpha-Synuclein/toxicity
12.
J Neurochem ; 104(6): 1504-25, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18036154

ABSTRACT

Microglial neuroinflammatory processes play a primary role in dopaminergic neurodegeneration for Parkinson's disease (PD). This can occur, in part, by modulation of glial function following activation by soluble or insoluble modified alpha-synuclein (alpha-syn), a chief component of Lewy bodies that is released from affected dopaminergic neurons. alpha-Syn is nitrated during oxidative stress responses and in its aggregated form, induces inflammatory microglial functions. Elucidation of these microglial function changes in PD could lead to new insights into disease mechanisms. To this end, PD-associated inflammation was modeled by stimulation of microglia with aggregated and nitrated alpha-syn. These activated microglia were ameboid in morphology and elicited dopaminergic neurotoxicity. A profile of nitrated, aggregated alpha-syn-stimulated microglia was generated using combinations of genomic (microarrays) and proteomic (liquid chromatography-tandem mass spectrometry, differential gel electrophoresis, and protein array) assays. Genomic studies revealed a substantive role for nuclear factor-kappa B transcriptional activation. Qualitative changes in the microglial proteome showed robust increases in inflammatory, redox, enzyme, and cytoskeletal proteins supporting the genomic tests. Autopsy brain tissue acquired from substantia nigra and basal ganglia of PD patients demonstrated that parallel nuclear factor-kappa B-related inflammatory processes were, in part, active during human disease. Taken together, the transcriptome and proteome of nitrated alpha-syn activated microglia, shown herein, provide new potential insights into disease mechanisms.


Subject(s)
Microglia/metabolism , Oxidative Stress/physiology , Parkinson Disease/metabolism , Parkinson Disease/pathology , alpha-Synuclein/metabolism , Aged , Aged, 80 and over , Animals , Cell Nucleus/metabolism , Cells, Cultured , Female , Gene Expression/physiology , Humans , Male , Mice , Mice, Inbred C57BL , Microglia/immunology , Microglia/pathology , Middle Aged , NF-kappa B/genetics , NF-kappa B/metabolism , Neostriatum/metabolism , Neostriatum/pathology , Neuritis/immunology , Neuritis/metabolism , Neuritis/pathology , Nitrogen/metabolism , Parkinson Disease/immunology , Phenotype , Proteomics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Substantia Nigra/metabolism , Substantia Nigra/pathology , Transcription, Genetic/physiology , alpha-Synuclein/isolation & purification
13.
J Cell Sci ; 120(Pt 23): 4221-9, 2007 Dec 01.
Article in English | MEDLINE | ID: mdl-18003706

ABSTRACT

Post-translational phosphorylation of proteins provides a mechanism for cells to switch on or off many diverse processes, including responses to replication stress. Replication-stress-induced phosphorylation enables the rapid activation of numerous proteins involved in DNA replication, DNA repair and cell cycle checkpoints, including replication protein A (RPA). Here, we report that hydroxyurea (HU)-induced RPA phosphorylation requires both NBS1 (NBN) and NBS1 phosphorylation. Transfection of both phosphospecific and nonphosphospecific anti-NBS1 antibodies blocked hyperphosphorylation of RPA in HeLa cells. Nijmegen breakage syndrome (NBS) cells stably transfected with an empty vector or with S343A-NBS1 or S278A/S343A phospho-mutants were unable to hyperphosphorylate RPA in DNA-damage-associated foci following HU treatment. The stable transfection of fully functional NBS1 in NBS cells restored RPA hyperphosphorylation. Retention of ATR on chromatin in both NBS cells and in NBS cells expressing S278A/S343A NBS1 mutants decreased after DNA damage, suggesting that ATR is the kinase responsible for RPA phosphorylation. The importance of RPA hyperphosphorylation is demonstrated by the ability of cells expressing a phospho-mutant form of RPA32 (RPA2) to suppress and delay HU-induced apoptosis. Our findings suggest that RPA hyperphosphorylation requires NBS1 and is important for the cellular response to DNA damage.


Subject(s)
Cell Cycle Proteins/metabolism , DNA Replication , Nuclear Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Replication Protein A/metabolism , Annexin A5/metabolism , Ataxia Telangiectasia Mutated Proteins , Cell Line, Transformed , Cell Transformation, Viral , Fibroblasts/metabolism , Genetic Vectors , HeLa Cells , Humans , Phosphorylation , Retroviridae/genetics , Simian virus 40/physiology , Subcellular Fractions/metabolism , Transfection
14.
J Neurochem ; 102(3): 627-45, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17442053

ABSTRACT

Microglia, a primary immune effector cell of the central nervous system (CNS) affects homeostatic, neuroprotective, regenerative and degenerative outcomes in health and disease. Despite these broad neuroimmune activities linked to specific environmental cues, a precise cellular genetic profile for microglia in the context of disease and repair has not been elucidated. To this end we used nucleic acid microarrays, proteomics, immunochemical and histochemical tests to profile microglia in neuroprotective immune responses. Optic and sciatic nerve (ON and SN) fragments were used to stimulate microglia in order to reflect immune consequences of nervous system injury. Lipopolysaccharide and latex beads-induced microglial activation served as positive controls. Cytosolic and secreted proteins were profiled by surface enhanced laser desorption ionization-time of flight (SELDI-TOF) ProteinChip, 1D and 2D difference gel electrophoresis. Proteins were identified by peptide sequencing with tandem mass spectrometry, ELISA and western blot tests. Temporal expression of pro-inflammatory cytokines, antioxidants, neurotrophins, and lysosomal enzyme expression provided, for the first time, a unique profile of secreted microglia proteins with neuroregulatory functions. Most importantly, this molecular and biochemical signature supports a broad range of microglial functions for debris clearance and promotion of neural repair after injury.


Subject(s)
Brain Injury, Chronic/immunology , Encephalitis/immunology , Gene Expression Profiling , Gliosis/immunology , Microglia/immunology , Nerve Tissue Proteins/immunology , Animals , Animals, Newborn , Antioxidants/metabolism , Biomarkers/analysis , Biomarkers/metabolism , Brain Injury, Chronic/metabolism , Brain Injury, Chronic/physiopathology , Cells, Cultured , Cytokines/immunology , Cytokines/metabolism , Encephalitis/metabolism , Encephalitis/physiopathology , Gliosis/metabolism , Gliosis/physiopathology , Inflammation Mediators/pharmacology , Lysosomes/enzymology , Mice , Mice, Inbred C57BL , Microglia/metabolism , Nerve Growth Factors/immunology , Nerve Growth Factors/metabolism , Nerve Regeneration/immunology , Nerve Tissue Proteins/analysis , Nerve Tissue Proteins/metabolism , Phagocytosis/immunology , Proteomics
15.
Neurochem Res ; 29(6): 1161-8, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15176473

ABSTRACT

Microarray technology has become a common tool for developing expression profiles. Initially used in the analysis of cells lines and homogeneous tissues, this platform has been applied to more diverse tissues, such as the brain. Several neural disorders have already been profiled by microarrays using relatively large amounts of tissue. This data has unveiled many genes with differential expression between normal and diseased tissue that could potentially be used as gene markers for these afflictions. Because of the heterogeneity of the CNS, it is likely that small differences between gene expression in these studies would be enhanced by the sampling of a subset of cells based on these newly characterized gene markers. Subtraction of normal, unaffected cells from the sample may also result in a more accurate profile of a diseased cell. Expression profile studies from several neuropathological states are presented, with emphasis placed on those studies using small samples of cellular material and those using specialized methods of cell isolation and RNA amplification.


Subject(s)
Gene Expression Profiling/methods , Neurodegenerative Diseases/genetics , AIDS Dementia Complex/genetics , Brain/pathology , Brain/physiopathology , Humans , Parkinson Disease/genetics , Reproducibility of Results , Schizophrenia/genetics , Sensitivity and Specificity
16.
Neurobiol Aging ; 24(8): 1105-11, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14643382

ABSTRACT

Synaptogenesis and synaptic plasticity demonstrate the ability of neurons to mature and respond to various stimuli. Both of these events require protein synthesis, in particular, localized translation within dendrites. Dendrites localize specific mRNAs in proximity to dendritic spines and have the capacity to actively translate these mRNAs within various 'hotspots' in the dendritic space. Rates of dendritic translation are stimulated and inhibited by various agents, suggesting that several signaling pathways that modulate localized protein synthesis may exist within the dendrite. This review will cover several suggested pathways for regulation of dendritic translation and propose a correlation between deregulated dendritic translation and disease.


Subject(s)
Dendrites/metabolism , Nerve Tissue Proteins/biosynthesis , Neuronal Plasticity/physiology , Protein Biosynthesis/physiology , Animals , Humans , Nerve Tissue Proteins/genetics , RNA, Messenger/metabolism , Signal Transduction/physiology
17.
Gene ; 285(1-2): 149-56, 2002 Feb 20.
Article in English | MEDLINE | ID: mdl-12039041

ABSTRACT

ras GTPase activating protein (rasGAP) is highly conserved among mammalian species and is required for normal cardiovascular system development. Expression of this protein exhibits both quantitative and qualitative variability among tissues. Using a combination of DNA sequencing and database analyses, we have determined that the human rasGAP gene spans 122 kb and is composed of 25 exons; the size of each intron and the intron/exon junctions also have been elucidated. With one exception, all intron/exon boundaries conform to the GT/AG rule; the splice donor site of intron 3 is GC/AG. Results of RNA ligase mediated rapid amplification of cDNA ends followed by sequence determination indicate that the transcription start point (TSP) is approximately 588 bp upstream from the translational start site and is uninterrupted by introns; this extremely long 5' untranslated region is continuous with the first coding exon. Analysis of 1 kb of sequence upstream of the TSP did not identify any of the typical promoter elements (TATA or CAAT boxes). Sequential deletions of this 1 kb region followed by secreted alkaline phosphatase reporter gene analysis revealed that transcription is supported by this region of the rasGAP gene. Because the highest efficiency is demonstrated by a 213 bp sequence just upstream from the TSP (-786 to -584), this region is identified as containing the rasGAP minimal promoter. Sequence analysis of this 213 bp sequence shows few candidate sites for transcription factor binding. A 406 bp fragment surrounding the TSP exhibits characteristics of a CpG island (68% C+G; observed/expected ratio of CpG=0.95). RapidScan analysis revealed that high levels of rasGAP transcript are present in placenta and testis, but transcript is not detectable in kidney and intestinal tract. These data suggest that rasGAP transcription is regulated by an atypical mechanism capable of producing quantitative variability among tissue types.


Subject(s)
ras GTPase-Activating Proteins/genetics , Alkaline Phosphatase/genetics , Alkaline Phosphatase/metabolism , Base Sequence , CpG Islands/genetics , DNA/chemistry , DNA/genetics , Exons , Female , Gene Expression Regulation , Gene Expression Regulation, Developmental , Genes/genetics , HeLa Cells , Humans , Introns , Male , Molecular Sequence Data , Promoter Regions, Genetic/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Analysis, DNA , Transcription Initiation Site , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...