Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 176
Filter
1.
Nucleic Acids Res ; 49(18): 10507-10523, 2021 10 11.
Article in English | MEDLINE | ID: mdl-34534348

ABSTRACT

A DNA replication program, which ensures that the genome is accurately and wholly replicated, is established during G1, before the onset of S phase. In G1, replication origins are licensed, and upon S phase entry, a subset of these will form active replisomes. Tight regulation of the number of active replisomes is crucial to prevent replication stress-induced DNA damage. TICRR/TRESLIN is essential for DNA replication initiation, and the level of TICRR and its phosphorylation determine the number of origins that initiate during S phase. However, the mechanisms regulating TICRR protein levels are unknown. Therefore, we set out to define the TICRR/TRESLIN protein dynamics throughout the cell cycle. Here, we show that TICRR levels are high during G1 and dramatically decrease as cells enter S phase and begin DNA replication. We show that degradation of TICRR occurs specifically during S phase and depends on ubiquitin ligases and proteasomal degradation. Using two targeted siRNA screens, we identify CRL4DTL as a cullin complex necessary for TICRR degradation. We propose that this mechanism moderates the level of TICRR protein available for replication initiation, ensuring the proper number of active origins as cells progress through S phase.


Subject(s)
Cell Cycle Proteins/metabolism , S Phase , Ubiquitin-Protein Ligases/metabolism , CDC2 Protein Kinase/antagonists & inhibitors , Carrier Proteins/physiology , Cell Cycle , Cell Cycle Proteins/physiology , Cell Line, Tumor , Cullin Proteins/metabolism , Cullin Proteins/physiology , Cyclin-Dependent Kinase 2/antagonists & inhibitors , DNA-Binding Proteins/physiology , Humans , Proliferating Cell Nuclear Antigen/physiology , Ubiquitin-Protein Ligases/physiology
2.
Mol Biol Cell ; 32(20): br6, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34379448

ABSTRACT

On induction of DNA damage with 405-nm laser light, proteins involved in base excision repair (BER) are recruited to DNA lesions. We find that the dynamics of factors typical of either short-patch (XRCC1) or long-patch (PCNA) BER are altered by chemicals that perturb actin or tubulin polymerization in human cells. Whereas the destabilization of actin filaments by latrunculin B, cytochalasin B, or Jasplakinolide decreases BER factor accumulation at laser-induced damage, inhibition of tubulin polymerization by nocodazole increases it. We detect no recruitment of actin to sites of laser-induced DNA damage, yet the depolymerization of cytoplasmic actin filaments elevates both actin and tubulin signals in the nucleus. While published evidence suggested a positive role for F-actin in double-strand break repair in mammals, the enrichment of actin in budding yeast nuclei interferes with BER, augmenting sensitivity to Zeocin. Our quantitative imaging results suggest that the depolymerization of cytoplasmic actin may compromise BER efficiency in mammals not only due to elevated levels of nuclear actin but also of tubulin, linking cytoskeletal integrity to BER.


Subject(s)
DNA Repair/physiology , Proliferating Cell Nuclear Antigen/metabolism , X-ray Repair Cross Complementing Protein 1/metabolism , Actins/metabolism , Cell Line, Tumor , Cell Nucleus/metabolism , Cytoskeleton/physiology , DNA/metabolism , DNA Damage/genetics , DNA Damage/physiology , DNA Repair/genetics , DNA-Binding Proteins/metabolism , HeLa Cells , Humans , Proliferating Cell Nuclear Antigen/physiology , Tubulin/metabolism , X-ray Repair Cross Complementing Protein 1/physiology
3.
Nucleic Acids Res ; 47(13): 6826-6841, 2019 07 26.
Article in English | MEDLINE | ID: mdl-31114918

ABSTRACT

Proliferating cell nuclear antigen (PCNA) is a sliding clamp that acts as a central co-ordinator for mismatch repair (MMR) as well as DNA replication. Loss of Elg1, the major subunit of the PCNA unloader complex, causes over-accumulation of PCNA on DNA and also increases mutation rate, but it has been unclear if the two effects are linked. Here we show that timely removal of PCNA from DNA by the Elg1 complex is important to prevent mutations. Although premature unloading of PCNA generally increases mutation rate, the mutator phenotype of elg1Δ is attenuated by PCNA mutants PCNA-R14E and PCNA-D150E that spontaneously fall off DNA. In contrast, the elg1Δ mutator phenotype is exacerbated by PCNA mutants that accumulate on DNA due to enhanced electrostatic PCNA-DNA interactions. Epistasis analysis suggests that PCNA over-accumulation on DNA interferes with both MMR and MMR-independent process(es). In elg1Δ, over-retained PCNA hyper-recruits the Msh2-Msh6 mismatch recognition complex through its PCNA-interacting peptide motif, causing accumulation of MMR intermediates. Our results suggest that PCNA retention controlled by the Elg1 complex is critical for efficient MMR: PCNA needs to be on DNA long enough to enable MMR, but if it is retained too long it interferes with downstream repair steps.


Subject(s)
Carrier Proteins/physiology , DNA Mismatch Repair , DNA, Fungal/metabolism , Mutation , Proliferating Cell Nuclear Antigen/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/physiology , Saccharomyces cerevisiae/genetics , Carrier Proteins/genetics , Crystallography, X-Ray , DNA Replication , DNA, Fungal/genetics , DNA-Binding Proteins/metabolism , Gene Editing , Genes, Fungal , MutS Homolog 2 Protein/metabolism , MutS Homolog 3 Protein/metabolism , Nucleic Acid Conformation , Point Mutation , Proliferating Cell Nuclear Antigen/physiology , Protein Binding , Protein Conformation , Recombinant Proteins/metabolism , S Phase , Saccharomyces cerevisiae Proteins/metabolism , Sumoylation
4.
FEBS J ; 286(14): 2711-2725, 2019 07.
Article in English | MEDLINE | ID: mdl-30963698

ABSTRACT

Rev1, a Y-family DNA polymerase, is involved in the tolerance of DNA damage by translesion DNA synthesis (TLS). Previous studies have shown that the C-terminal domain (CTD) and ubiquitin (Ub)-binding (UBM) domains of Rev1 play important roles in UV-damage tolerance, but how these domains contribute to the process remains unclear. In this study, we created Ub mutations in a proliferating cell nuclear antigen (PCNA)-Ub fusion that differentially affect its interaction with Rev1 and Polη and found that UV-damage tolerance depends on its interaction with Rev1 but not Polη. We also created Rev1-UBM mutations altering its interaction with a PCNA-Ub fusion and Rev1-CTD mutations affecting its interaction with Polη and the Rev7 subunit of Polζ. We thus demonstrated that elevated expression of Rev1 alone is sufficient to confer enhanced UV-damage tolerance and that this tolerance depends on its physical interaction with monoubiquitinated PCNA and Polζ but is independent of Polη. Collectively, these studies reveal central roles played by Rev1 in coordinating UV-damage response pathway choice in mammalian cells.


Subject(s)
DNA Damage , Nucleotidyltransferases/physiology , Ultraviolet Rays , DNA-Directed DNA Polymerase/physiology , HCT116 Cells , Humans , Mutation , Nucleotidyltransferases/chemistry , Nucleotidyltransferases/genetics , Proliferating Cell Nuclear Antigen/physiology , Ubiquitin/physiology
5.
Cell Rep ; 27(1): 48-58.e7, 2019 04 02.
Article in English | MEDLINE | ID: mdl-30943414

ABSTRACT

To enable reliable cell fate decisions, mammalian cells need to adjust their responses to dynamically changing internal states by rewiring the corresponding signaling networks. Here, we combine time-lapse microscopy of endogenous fluorescent reporters with computational analysis to understand at the single-cell level how the p53-mediated DNA damage response is adjusted during cell cycle progression. Shape-based clustering revealed that the dynamics of the CDK inhibitor p21 diverges from the dynamics of its transcription factor p53 during S phase. Using mathematical modeling, we predict and experimentally validate that S phase-specific degradation of p21 by PCNA-CRL4cdt2 is sufficient to explain these heterogeneous responses. This highlights how signaling pathways and cell regulatory networks intertwine to adjust the cellular response to the individual needs of a given cell.


Subject(s)
Cell Cycle/physiology , Cyclin-Dependent Kinase Inhibitor p21/metabolism , DNA Damage/physiology , Proliferating Cell Nuclear Antigen/physiology , Proteolysis , A549 Cells , Cell Cycle Checkpoints/physiology , Cells, Cultured , DNA Repair/physiology , Female , HEK293 Cells , Humans , MCF-7 Cells , Proliferating Cell Nuclear Antigen/genetics , Signal Transduction/genetics , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Protein p53/physiology
6.
Appl Physiol Nutr Metab ; 44(5): 507-511, 2019 May.
Article in English | MEDLINE | ID: mdl-30286297

ABSTRACT

Flaxseed is useful as a functional food and alternative medicine owing to its beneficial health effects. Its action on ovarian cell functions and interrelationships with the upstream hormonal regulators remain unknown. Our aim was to examine the direct influence of flaxseed extract on basal porcine ovarian functions (proliferation, apoptosis), leptin release, and response to insulin-like growth factor I (IGF-I). First, we examined the effect of flaxseed extract on the accumulation of proliferation (PCNA) and apoptosis (Bax) markers and on leptin release in cultured porcine ovarian granulosa cells. Next, granulosa cells were cultured with IGF-I with and without flaxseed extract and analyzed for PCNA and Bax accumulation by quantitative immunocytochemistry and for leptin release by radioimmunoassay. Flaxseed decreased the accumulation of PCNA and increased that of Bax at all doses and reduced leptin output at 100 µg/mL. In contrast, IGF-I promoted PCNA accumulation and suppressed Bax. Flaxseed did not modify IGF-I action on these parameters. Thus, we showed that flaxseed influences porcine reproductive processes, having a direct effect on the ovary and the ability to affect ovarian cell proliferation, apoptosis, and leptin release. Furthermore, we confirmed the pro-proliferative and antiapoptotic actions of IGF-I but showed that flaxseed action on ovarian cell proliferation and apoptosis is not due to changes in the cell response to IGF-I. The potential direct anti-reproductive action of flaxseed needs to be considered during its application in nutrition, medicine, and animal production.


Subject(s)
Flax , Granulosa Cells/physiology , Ovary/cytology , Plant Extracts/pharmacology , Animals , Apoptosis , Cell Proliferation , Cells, Cultured , Female , Insulin-Like Growth Factor I/physiology , Leptin/physiology , Proliferating Cell Nuclear Antigen/physiology , Swine , bcl-2-Associated X Protein/physiology
7.
Anim Sci J ; 88(12): 1943-1954, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28782242

ABSTRACT

Thyroid hormones are important in the development and regulation of testes. This study was conducted to determine the effects of hyper- and hypothyroidism on testicular development in prepubertal rats aged 20-70 days. Weaning male rats (20 days old) until day 70 age were randomly divided into four groups: control, hyperthyroid (hyper-T), hypothyroid (hypo-T) and hypothyroid treated with thyroxine (T4) (hypo-T+T4). The results indicated that thyroid hormones caused a significant effect in body and testis weights, and food and water consumption. In addition there were changes in serum concentrations of tri-iodothyronine, T4, thyroid stimulating hormone (TSH) and testosterone. Histomorphology showed a significant decrease in seminiferous tubule diameter in hyper-T compared to the other groups. Leydig cell numbers showed a significant elevation in hyper-T but not in hypo-T groups. Immunostaining indicated that TSH receptor (TSHR), thyroid hormone receptors α/ß (TRαß) and proliferating cell nuclear antigen (PCNA) have the roles in testicular development. Our findings suggest that hyper- and hypo-thyroidism regulate testicular cell proliferation and spermatogenesis in prepubertal rats, indicating that expression of TSHR, TRαß and PCNA may be regulated by thyroid hormones that are involved in testicular development; and that the administration of T4 to the hypo-T+T4 group leads to an improvement in the testicular condition.


Subject(s)
Cell Proliferation , Hyperthyroidism/physiopathology , Hypothyroidism/physiopathology , Puberty/physiology , Testis/cytology , Testis/growth & development , Thyroid Hormones/physiology , Animals , Cell Proliferation/drug effects , Cell Proliferation/physiology , Hyperthyroidism/pathology , Hypothyroidism/drug therapy , Hypothyroidism/pathology , Leydig Cells/cytology , Male , Proliferating Cell Nuclear Antigen/metabolism , Proliferating Cell Nuclear Antigen/physiology , Rats , Rats, Sprague-Dawley , Receptors, Thyrotropin/metabolism , Spermatogenesis , Testis/pathology , Testosterone/blood , Thyroid Hormone Receptors alpha/metabolism , Thyroid Hormone Receptors alpha/physiology , Thyroid Hormone Receptors beta/metabolism , Thyroid Hormone Receptors beta/physiology , Thyroid Hormones/blood , Thyroxine/administration & dosage , Thyroxine/pharmacology
8.
Chromosome Res ; 25(2): 89-100, 2017 06.
Article in English | MEDLINE | ID: mdl-27987109

ABSTRACT

Heterochromatin plays important roles in the structure, maintenance, and function of the eukaryotic genome. It is associated with special histone modifications and specialized non-histone proteins and assumes a more compact structure than euchromatin. Genes embedded in heterochromatin are generally transcriptionally silent. It was found previously that several mutations of proliferating cell nuclear antigen (PCNA), a DNA replication processivity factor, reduce transcriptional silencing at heterochromatin loci in Saccharomyces cerevisiae. However, the notion that PCNA plays a role in transcriptional silencing was recently questioned because of a potential problem concerning the silencing assays used in prior studies. To determine if PCNA is a bona fide contributor to heterochromatin-mediated transcriptional silencing, we examined the effects of PCNA mutations on heterochromatin structure. We found evidence implicating PCNA in the maintenance of the high-order structure and stability of heterochromatin, which indicates a role of DNA replication in heterochromatin maintenance.


Subject(s)
Heterochromatin/chemistry , Proliferating Cell Nuclear Antigen/physiology , Saccharomyces cerevisiae/genetics , DNA Repair , DNA Replication , Gene Silencing , Heterochromatin/ultrastructure
9.
J Neurochem ; 140(4): 576-588, 2017 02.
Article in English | MEDLINE | ID: mdl-27926980

ABSTRACT

It is necessary for the cell-cycle machinery of neurons to be suppressed to promote differentiation and maintenance of their terminally differentiated state. Reactivation of the cell cycle in response to neurotoxic insults leads to neuronal cell death and some cell-cycle-related proteins contribute to the process. p27 kip1 (p27), an inhibitor of cyclin-dependent kinases, prevents unwarranted cyclin-dependent kinase activation. In this study, we have elucidated a novel mechanism via which p27 promotes apoptosis of neurons stimulated by neurotoxic amyloid peptide Aß42 (Amyloid ß1-42 peptide). Co-immunoprecipitation analysis revealed that p27 promotes interaction between Cyclin-dependent kinase 5 (Cdk5) and cyclin D1, which is induced by Aß42 in cortical neurons. As a result, Cdk5 is sequestered from its neuronal activator p35 resulting in kinase deactivation. The depletion of p27, which was achieved by specific siRNA, restored Cdk5/p35 interaction by preventing association between Cdk5 and cyclin D1 and also abrogated Aß42 induced apoptosis of cortical neurons. Furthermore, analysis of cell cycle markers suggested that p27 may play a role in Aß42 induced aberrant cell cycle progression of neurons, which may result in apoptosis. These findings provide novel insights into how p27, which otherwise performs important neuronal functions, may become deleterious to neurons under neurotoxic conditions.


Subject(s)
Amyloid beta-Peptides/toxicity , Apoptosis/physiology , Neurons/physiology , Peptide Fragments/toxicity , Proliferating Cell Nuclear Antigen/physiology , Animals , Apoptosis/drug effects , Cell Movement/drug effects , Cell Movement/physiology , Female , HEK293 Cells , Humans , Male , Mice , Mice, Transgenic , NIH 3T3 Cells , Neurons/drug effects , PC12 Cells , Rats , Rats, Sprague-Dawley
10.
PLoS One ; 11(8): e0161307, 2016.
Article in English | MEDLINE | ID: mdl-27537501

ABSTRACT

Proliferating cell nuclear antigen (PCNA) plays a key role in many cellular processes and due to that it interacts with a plethora of proteins. The main interacting surfaces of Saccharomyces cerevisiae PCNA have been mapped to the interdomain connecting loop and to the carboxy-terminal domain. Here we report that the subunit interface of yeast PCNA also has regulatory roles in the function of several DNA damage response pathways. Using site-directed mutagenesis we engineered mutations at both sides of the interface and investigated the effect of these alleles on DNA damage response. Genetic experiments with strains bearing the mutant alleles revealed that mutagenic translesion synthesis, nucleotide excision repair, and homologous recombination are all regulated through residues at the subunit interface. Moreover, genetic characterization of one of our mutants identifies a new sub-branch of nucleotide excision repair. Based on these results we conclude that residues at the subunit boundary of PCNA are not only important for the formation of the trimer structure of PCNA, but they constitute a regulatory protein domain that mediates different DNA damage response pathways, as well.


Subject(s)
Proliferating Cell Nuclear Antigen/genetics , Saccharomyces cerevisiae Proteins/genetics , DNA Damage/genetics , DNA Damage/physiology , DNA Repair/genetics , DNA Repair/physiology , Mutagenesis, Site-Directed , Mutation/genetics , Mutation/physiology , Proliferating Cell Nuclear Antigen/physiology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae/physiology , Saccharomyces cerevisiae Proteins/physiology , Transcription, Genetic/genetics , Transcription, Genetic/physiology
11.
J Microbiol Biotechnol ; 26(4): 637-47, 2016 Apr 28.
Article in English | MEDLINE | ID: mdl-26699741

ABSTRACT

Proliferating cell nuclear antigen (PCNA) is a critical eukaryotic replication accessory factor that supports DNA binding in DNA processing, such as DNA replication, repair, and recombination. PCNA consists of three toroidal-shaped monomers that encircle doublestranded DNA. The diverse functions of PCNA may be regulated by its interactions with partner proteins. Many of the PCNA partner proteins generally have a conserved PCNAinteracting peptide (PIP) motif, located at the N- or C- terminal region. The PIP motif forms a 310 helix that enters into the hydrophobic groove produced by an interdomain-connecting loop, a central loop, and a C-terminal tail in the PCNA. Post-translational modification of PCNA also plays a critical role in regulation of its function and binding partner proteins. Structural and biochemical studies of PCNA-protein will be useful in designing therapeutic agents, as well as estimating the outcome of anticancer drug development. This review summarizes the characterization of eukaryotic PCNA in relation to the protein structures, functions, and modifications, and interaction with proteins.


Subject(s)
Proliferating Cell Nuclear Antigen/chemistry , Proliferating Cell Nuclear Antigen/physiology , Protein Processing, Post-Translational/genetics , Amino Acid Sequence , Animals , DNA Replication , DNA-Binding Proteins/chemistry , Humans , Proliferating Cell Nuclear Antigen/genetics , Protein Binding , Saccharomyces cerevisiae Proteins/genetics
12.
Biochem J ; 470(1): 115-29, 2015 Aug 15.
Article in English | MEDLINE | ID: mdl-26251451

ABSTRACT

Eukaryotic PCNAs (proliferating-cell nuclear antigens) play diverse roles in nucleic acid metabolism in addition to DNA replication. Plasmodium falciparum, which causes human malaria, harbours two PCNA homologues: PfPCNA1 and PfPCNA2. The functional role of two distinct PCNAs in the parasite still eludes us. In the present study, we show that, whereas both PfPCNAs share structural and biochemical properties, only PfPCNA1 functionally complements the ScPCNA mutant and forms distinct replication foci in the parasite, which PfPCNA2 fails to do. Although PfPCNA1 appears to be the primary replicative PCNA, both PfPCNA1 and PfPCNA2 participate in an active DDR (DNA-damage-response) pathway with significant accumulation in the parasite upon DNA damage induction. Interestingly, PfPCNA genes were found to be regulated not at the transcription level, but presumably at the protein stability level upon DNA damage. Such regulation of PCNA has not been shown in eukaryotes before. Moreover, overexpression of PfPCNA1 and PfPCNA2 in the parasite confers a survival edge on the parasite in a genotoxic environment. This is the first evidence of a PfPCNA-mediated DDR in the parasite and gives new insights and rationale for the presence of two PCNAs as a parasite survival strategy and its probable success.


Subject(s)
DNA Damage/physiology , DNA Replication/physiology , Plasmodium falciparum/physiology , Proliferating Cell Nuclear Antigen/physiology , Cells, Cultured , Humans , Proliferating Cell Nuclear Antigen/chemistry , Protein Structure, Secondary
13.
J Anim Sci ; 93(4): 1679-88, 2015 Apr.
Article in English | MEDLINE | ID: mdl-26020189

ABSTRACT

Polyamines are necessary for normal integrity and the restitution after injury of the gastrointestinal epithelium. The objective of this study was to investigate the effects of oral administration of putrescine and proline during the suckling period on epithelial restitution after early weaning in piglets. Eighteen neonatal piglets (Duroc × Landrace × Large Yorkshire) from 3 litters (6 piglets per litter) were assigned to 3 groups, representing oral administration with an equal volume of saline (control), putrescine (5 mg/kg BW), and proline (25 mg/kg BW) twice daily from d 1 to weaning at 14 d of age. Plasma and intestinal samples were obtained 3 d after weaning. The results showed that oral administration of putrescine or proline increased the final BW and ADG of piglets compared with the control (P < 0.05). Proline treatment decreased plasma D-lactate concentration but increased the villus height in the jejunum and ileum, as well as the percentage of proliferating cell nuclear antigen (PCNA) positive cells and alkaline phosphatase (AKP) activity in the jejunal mucosa (P < 0.05). The protein expressions for zonula occludens (ZO-1), occludin, and claudin-3 (P < 0.05) but not mRNA were increased in the jejunum of putrescine- and proline-treated piglets compared with those of control piglets. The voltage-gated K+ channel (Kv) 1.1 protein expression in the jejunum of piglets administrated with putrescine and the Kv1.5 mRNA and Kv1.1 protein levels in the ileum of piglets administrated with proline were greater than those in control piglets (P < 0.05). These findings indicate that polyamine or its precursor could improve mucosal proliferation, intestinal morphology, as well as tight junction and potassium channel protein expressions in early-weaned piglets, with implications for epithelial restitution and barrier function after stress injury.


Subject(s)
Animals, Newborn/physiology , Animals, Suckling/physiology , Cell Proliferation/drug effects , Intestinal Mucosa/drug effects , Proline/pharmacology , Putrescine/pharmacology , Swine/physiology , Weaning , Administration, Oral , Alkaline Phosphatase/drug effects , Alkaline Phosphatase/physiology , Animals , Cell Proliferation/physiology , Endothelium/cytology , Endothelium/drug effects , Ileum/cytology , Ileum/drug effects , Ileum/physiology , Intestinal Mucosa/cytology , Intestinal Mucosa/physiology , Jejunum/cytology , Jejunum/drug effects , Jejunum/physiology , Potassium Channels/drug effects , Potassium Channels/physiology , Proliferating Cell Nuclear Antigen/drug effects , Proliferating Cell Nuclear Antigen/physiology , Proline/administration & dosage , Putrescine/administration & dosage , Tight Junction Proteins/drug effects , Tight Junction Proteins/physiology
14.
Braz. j. vet. res. anim. sci ; 52(4): 356-362, 2015.
Article in English | LILACS | ID: lil-780262

ABSTRACT

Immunohistochemical expression of proliferating cell nuclear antigen (PCNA) was evaluated in the endometrium of mares during estrus and at early diestrus. Three samples were collected by endometrial biopsy from 10 mares, on estrus/ second day, in the ovulation day and seven days after the ovulation day. PCNA expression was high in luminal epithelium and low in endometrial glands on samples taken on estrus/second day and on the ovulation day (p 0.05). For samples collected on the seventh day following ovulation, the averaged PCNA immunostaining was higher in glandular epithelium (p 0.05). The study revealed that luminal epithelial cells exhibit higher proliferation during estrus and glandular epithelial cells exhibited higher proliferation during diestrus...


A expressão do antígeno núcleo celular proliferante (ANCP) foi avaliada no endométrio de éguas durante o estro e início do diestro. Em cada uma de dez éguas foram efetuadas biópsias do endométrio em três momentos dos respectivos ciclos reprodutivos: segundo dia do estro, dia da ovulação e sete dias após a ovulação. Nas amostras colhidas no segundo dia do estro e no dia da ovulação, a expressão do ANCP foi elevada no epitélio luminal e baixa nas glândulas endometriais (p 0,05). Nas amostras colhidas no sétimo dia após a ovulação, a média de ANCP imunologicamente corado foi maior no epitélio glandular (p 0,05). O estudo revelou que as células do epitélio luminal apresentaram a maior proliferação durante o estro e que as células epiteliais glandulares apresentaram a maior proliferação durante o diestro...


Subject(s)
Animals , Female , Proliferating Cell Nuclear Antigen/physiology , Horses/physiology , Endometrium/physiology , Biopsy/veterinary , Epithelial Cells/physiology , Diestrus/physiology , Estrus/physiology , Fertility/physiology , Cell Proliferation
15.
Proc Natl Acad Sci U S A ; 111(32): 11816-21, 2014 Aug 12.
Article in English | MEDLINE | ID: mdl-25071216

ABSTRACT

Kaposi's sarcoma-associated herpesvirus (KSHV) latently infects tumor cells and persists as a multiple-copy, extrachromosomal, circular episome. To persist, the viral genome must replicate with each cell cycle. The KSHV latency-associated nuclear antigen (LANA) mediates viral DNA replication and persistence, but little is known regarding the underlying mechanisms. We find that LANA recruits replication factor C (RFC), the DNA polymerase clamp [proliferating cell nuclear antigen (PCNA)] loader, to drive DNA replication efficiently. Mutated LANA lacking RFC interaction was deficient for LANA-mediated DNA replication and episome persistence. RFC depletion had a negative impact on LANA's ability to replicate and maintain viral DNA in cells containing artificial KSHV episomes or in infected cells, leading to loss of virus. LANA substantially increased PCNA loading onto DNA in vitro and recruited RFC and PCNA to KSHV DNA in cells. These findings suggest that PCNA loading is a rate-limiting step in DNA replication that is incompatible with viral survival. LANA enhancement of PCNA loading permits efficient virus replication and persistence, revealing a previously unidentified mechanism for KSHV latency.


Subject(s)
Antigens, Viral/physiology , Herpesvirus 8, Human/physiology , Herpesvirus 8, Human/pathogenicity , Nuclear Proteins/physiology , Replication Protein C/physiology , Virus Replication/physiology , Cell Line, Tumor , DNA Replication/physiology , DNA, Viral/biosynthesis , DNA, Viral/genetics , DNA-Directed DNA Polymerase/physiology , Gene Knockdown Techniques , Herpesvirus 8, Human/genetics , Host-Pathogen Interactions , Humans , Proliferating Cell Nuclear Antigen/physiology , Replication Protein C/antagonists & inhibitors , Replication Protein C/genetics , Sarcoma, Kaposi/physiopathology , Sarcoma, Kaposi/virology , Virus Latency/physiology
16.
Int J Clin Exp Pathol ; 7(4): 1534-43, 2014.
Article in English | MEDLINE | ID: mdl-24817949

ABSTRACT

FOXL2 is a transcription factor that is essential for ovarian function and maintenance, the germline mutations of which give rise to the blepharophimosis ptosis epicanthus inversus syndrome (BPES), often associated with premature ovarian failure. Recently, its mutations have been found in ovarian granulosa cell tumors (OGCTs). In this study, we measured the expression of FOXL2 in cervical cancer by immunohistochemistry and its mRNA level in cervical cancer cell lines Hela and Siha by RT-PCR. Then we overexpressed FOXL2 in Hela cells and silenced it in Siha cells by plasmid transfection and verified using western blotting. When FOXL2 was overexpressed or silenced, cells proliferation and apoptosis were determined by Brdu assay and Annexin V/PI detection kit, respectively. In addition, we investigated the effects of FOXL2 on the adhesion and invasion of Hela and Siha cells. Finally, we analyzed the influences of FOXL2 on Ki67, PCNA and FasL by flow cytometry. The results showed that FOXL2 was highly expressed in cervical squamous cancer. Overexpressing FOXL2 suppressed Hela proliferation and facilitated its apoptosis. Silencing FOXL2 enhanced Siha proliferation and inhibited its apoptosis. Meanwhile, silencing FOXL2 promoted Siha invasion, but it had no effect on cells adhesion. In addition, overexpressing FOXL2 decreased the expression of Ki67 in Hela and Siha cells. Therefore, our results suggested that FOXL2 restrained cells proliferation and enhanced cells apoptosis mainly through decreasing Ki67 expression.


Subject(s)
Adenocarcinoma/physiopathology , Apoptosis/physiology , Carcinoma, Squamous Cell/physiopathology , Cell Movement/physiology , Cell Proliferation/physiology , Forkhead Transcription Factors/physiology , Uterine Cervical Neoplasms/physiopathology , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Adult , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Cells, Cultured , Fas Ligand Protein/genetics , Fas Ligand Protein/physiology , Female , Forkhead Box Protein L2 , Forkhead Transcription Factors/genetics , Gene Expression Regulation, Neoplastic/physiology , Gene Silencing , HeLa Cells , Humans , Ki-67 Antigen/genetics , Ki-67 Antigen/physiology , Middle Aged , Proliferating Cell Nuclear Antigen/genetics , Proliferating Cell Nuclear Antigen/physiology , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/pathology
17.
Radiother Oncol ; 108(3): 370-7, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23830194

ABSTRACT

The epidermal growth factor receptor (EGFR) has been one of the most targeted receptors in the field of oncology. While anti-EGFR inhibitors have demonstrated clinical success in specific cancers, most patients demonstrate either intrinsic or acquired resistance within one year of treatment. Many mechanisms of resistance to EGFR inhibitors have been identified, one of these being attributed to alternatively localized EGFR from the cell membrane into the cell's nucleus. Inside the nucleus, EGFR functions as a co-transcription factor for several genes involved in cell proliferation and angiogenesis, and as a tyrosine kinase to activate and stabilize proliferating cell nuclear antigen and DNA dependent protein kinase. Nuclear localized EGFR is highly associated with disease progression, worse overall survival in numerous cancers, and enhanced resistance to radiation, chemotherapy, and the anti-EGFR therapies gefitinib and cetuximab. In this review the current knowledge of how nuclear EGFR enhances resistance to cancer therapeutics is discussed, in addition to highlighting ways to target nuclear EGFR as an anti-cancer strategy in the future.


Subject(s)
ErbB Receptors/antagonists & inhibitors , Neoplasms/drug therapy , Animals , Celecoxib , Cell Line, Tumor , Drug Resistance, Neoplasm , ErbB Receptors/physiology , Humans , Proliferating Cell Nuclear Antigen/physiology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Pyrazoles/pharmacology , Radiation Tolerance , Sulfonamides/pharmacology
18.
DNA Repair (Amst) ; 12(9): 691-8, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23731732

ABSTRACT

Homologous recombination (HR) is essential for maintaining genomic integrity, which is challenged by a wide variety of potentially lethal DNA lesions. Regardless of the damage type, recombination is known to proceed by RAD51-mediated D-loop formation, followed by DNA repair synthesis. Nevertheless, the participating polymerases and extension mechanism are not well characterized. Here, we present a reconstitution of this step using purified human proteins. In addition to Pol δ, TLS polymerases, including Pol η and Pol κ, also can extend D-loops. In vivo characterization reveals that Pol η and Pol κ are involved in redundant pathways for HR. In addition, the presence of PCNA on the D-loop regulates the length of the extension tracks by recruiting various polymerases and might present a regulatory point for the various recombination outcomes.


Subject(s)
DNA-Directed DNA Polymerase/chemistry , Homologous Recombination , Proliferating Cell Nuclear Antigen/chemistry , DNA Damage , DNA Polymerase III/chemistry , DNA Polymerase III/physiology , DNA Replication , DNA, Single-Stranded/biosynthesis , DNA-Directed DNA Polymerase/physiology , HeLa Cells , Humans , Osmolar Concentration , Proliferating Cell Nuclear Antigen/physiology , RNA-Binding Protein FUS/chemistry , RNA-Binding Protein FUS/physiology , Rad51 Recombinase/chemistry , DNA Polymerase iota
19.
Bioessays ; 35(7): 586-96, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23592240

ABSTRACT

The KCTD family includes tetramerization (T1) domain containing proteins with diverse biological effects. We identified a novel member of the KCTD family, BTBD10. A comprehensive analysis of protein-protein interactions (PPIs) allowed us to put forth a number of testable hypotheses concerning the biological functions for individual KCTD proteins. In particular, we predict that KCTD20 participates in the AKT-mTOR-p70 S6k signaling cascade, KCTD5 plays a role in cytokinesis in a NEK6 and ch-TOG-dependent manner, KCTD10 regulates the RhoA/RhoB pathway. Developmental regulator KCTD15 represses AP-2α and contributes to energy homeostasis by suppressing early adipogenesis. TNFAIP1-like KCTD proteins may participate in post-replication DNA repair through PCNA ubiquitination. KCTD12 may suppress the proliferation of gastrointestinal cells through interference with GABAb signaling. KCTD9 deserves experimental attention as the only eukaryotic protein with a DNA-like pentapeptide repeat domain. The value of manual curation of PPIs and analysis of existing high-throughput data should not be underestimated.


Subject(s)
Cell Differentiation/genetics , Nuclear Proteins/physiology , Repressor Proteins/physiology , Vertebrates/genetics , Animals , DNA Replication , Gene Expression Regulation, Developmental , Humans , Intracellular Signaling Peptides and Proteins , NIMA-Related Kinases , Nuclear Proteins/genetics , Potassium Channels/genetics , Potassium Channels/physiology , Potassium Channels, Voltage-Gated/genetics , Potassium Channels, Voltage-Gated/physiology , Proliferating Cell Nuclear Antigen/genetics , Proliferating Cell Nuclear Antigen/physiology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/physiology , Repressor Proteins/genetics , Ribosomal Protein S6 Kinases, 70-kDa/genetics , Ribosomal Protein S6 Kinases, 70-kDa/physiology , Signal Transduction , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/physiology , Ubiquitination
20.
Am J Physiol Endocrinol Metab ; 304(10): E1064-76, 2013 May 15.
Article in English | MEDLINE | ID: mdl-23531612

ABSTRACT

Adult mice lacking functional GABAB receptors (GABAB1KO) have glucose metabolism alterations. Since GABAB receptors (GABABRs) are expressed in progenitor cells, we evaluated islet development in GABAB1KO mice. Postnatal day 4 (PND4) and adult, male and female, GABAB1KO, and wild-type littermates (WT) were weighed and euthanized, and serum insulin and glucagon was measured. Pancreatic glucagon and insulin content were assessed, and pancreas insulin, glucagon, PCNA, and GAD65/67 were determined by immunohistochemistry. RNA from PND4 pancreata and adult isolated islets was obtained, and Ins1, Ins2, Gcg, Sst, Ppy, Nes, Pdx1, and Gad1 transcription levels were determined by quantitative PCR. The main results were as follows: 1) insulin content was increased in PND4 GABAB1KO females and in both sexes in adult GABAB1KOs; 2) GABAB1KO females had more clusters (<500 µm(2)) and less islets than WT females; 3) cluster proliferation was decreased at PND4 and increased in adult GABAB1KO mice; 4) increased ß-area at the expense of the α-cell area was present in GABAB1KO islets; 5) Ins2, Sst, and Ppy transcription were decreased in PND4 GABAB1KO pancreata, adult GABAB1KO female islets showed increased Ins1, Ins2, and Sst expression, Pdx1 was increased in male and female GABAB1KO islets; and 6) GAD65/67 was increased in adult GABAB1KO pancreata. We demonstrate that several islet parameters are altered in GABAB1KO mice, further pinpointing the importance of GABABRs in islet physiology. Some changes persist from neonatal ages to adulthood (e.g., insulin content in GABAB1KO females), whereas other features are differentially regulated according to age (e.g., Ins2 was reduced in PND4, whereas it was upregulated in adult GABAB1KO females).


Subject(s)
Insulin Resistance/physiology , Islets of Langerhans/physiology , Receptors, GABA-B/deficiency , Animals , Animals, Newborn , Body Weight/physiology , Female , Gene Expression Regulation , Glucagon/blood , Glucagon/genetics , Glucagon/physiology , Glutamate Decarboxylase/physiology , Insulin/blood , Insulin/genetics , Insulin/physiology , Islets of Langerhans/growth & development , Islets of Langerhans/metabolism , Islets of Langerhans/ultrastructure , Male , Mice , Mice, Inbred BALB C , Mice, Knockout , Organ Size/physiology , Proliferating Cell Nuclear Antigen/analysis , Proliferating Cell Nuclear Antigen/physiology , RNA/chemistry , RNA/genetics , Real-Time Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...