Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
Int J Biol Macromol ; 127: 306-310, 2019 Apr 15.
Article in English | MEDLINE | ID: mdl-30611803

ABSTRACT

Telomeres are the tandem repeats (TTAGGG) present at the ends of the chromosomes that ensure chromosome stability and protect chromosomes from degradation. Telomeres in somatic human cells shorten after every cellular division and are linked to the cellular senescence. In this study we have investigated telomere length and expression of shelterin genes in aborted fetus material from idiopathic recurrent pregnancy losses. Telomere length was measured using Telomere Restriction Fragment Length (TRF) analysis. The gene expression levels for important shelterin complex proteins (TRF1, TRF2, POT1, and TPP1) were determined by Real-time Quantitative Reverse Transcriptase PCR (qRT-PCR). Our results demonstrated down regulation of TRF2 and TPP1 and a strong decline in average telomere length in abort material from women suffering from idiopathic recurrent pregnancy loss. We suggest that shorter telomere length and downregulation of the major shelterin components TRF2 and TPP1 leading to "telomere uncapping", might play a critical role in recurrent pregnancy loss.


Subject(s)
Abortion, Habitual/metabolism , Aminopeptidases/biosynthesis , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/biosynthesis , Down-Regulation , Fetus/metabolism , Gene Expression Regulation, Developmental , Telomere Homeostasis , Telomere-Binding Proteins/biosynthesis , Telomeric Repeat Binding Protein 2/biosynthesis , Abortion, Habitual/pathology , Adult , Female , Fetus/pathology , Humans , Pregnancy , Shelterin Complex
2.
J Exp Clin Cancer Res ; 37(1): 182, 2018 Aug 03.
Article in English | MEDLINE | ID: mdl-30075819

ABSTRACT

BACKGROUND: Human telomerase reverse transcriptase (hTERT) is highly expressed in over 80% of tumors, including human epithelial ovarian cancer (EOC). However, the mechanisms through which hTERT is up-regulated in EOC and promotes tumor progression remain unclear. The aim of this study is to identify RIF1 as a novel molecular target that modulate hTERT signaling and EOC growth. METHODS: RIF1 expression in ovarian cancer, benign and normal ovarian tissues was examined by immunohistochemistry. The biological role of RIF1 was revealed by MTS, colony formation and sphere formation assays. Luciferase reporter assay and chromatin immunoprecipitation (CHIP) assay were used to verify RIF1 as a novel hTERT promoter-binding protein in EOC cells. The role of RIF1 on tumorigenesis in vivo was detected by the xenograft model. RESULTS: RIF1 expression is upregulated in EOC tissues and is closely correlated with FIGO stage and prognosis of EOC patients. Functionally, RIF1 knockdown suppressed the expression and promoter activity of hTERT and consequently inhibited the growth and CSC-like traits of EOC cells. RIF1 knockdown also inhibited tumorigenesis in xenograft model. RIF1 overexpression had the opposite effect. Luciferase reporter assay and ChIP assay verified RIF1 directly bound to hTERT promoter to upregulate its expression. The rescue experiments suggested hTERT overexpression rescued the inhibition of EOC cell growth and CSC-like traits mediated by RIF1 knockdown. Consistently, hTERT knockdown abrogated the RIF1-induced promotion of EOC cell growth and CSC-like traits. CONCLUSIONS: RIF1 promotes EOC progression by activating hTERT and the RIF1/hTERT pathway may be a potential therapeutic target for EOC patients.


Subject(s)
Carcinoma, Ovarian Epithelial/metabolism , Ovarian Neoplasms/metabolism , Telomerase/biosynthesis , Telomere-Binding Proteins/metabolism , Animals , Carcinoma, Ovarian Epithelial/enzymology , Carcinoma, Ovarian Epithelial/genetics , Carcinoma, Ovarian Epithelial/pathology , Cell Line, Tumor , Disease Progression , Female , Gene Knockdown Techniques , Humans , Immunohistochemistry , Mice , Mice, Inbred BALB C , Mice, Nude , Ovarian Neoplasms/enzymology , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Telomerase/genetics , Telomerase/metabolism , Telomere-Binding Proteins/biosynthesis , Telomere-Binding Proteins/genetics , Transfection
3.
PLoS One ; 13(6): e0197154, 2018.
Article in English | MEDLINE | ID: mdl-29870526

ABSTRACT

BACKGROUND: Colorectal cancer is the leading cause of cancer-related deaths in Saudi Arabia. Cancer has a multifactorial nature and can be described as a disease of altered gene expression. The profiling of gene expression has been used to identify cancer subtypes and to predict patients' responsiveness. Telomere-associated proteins that regulate telomere biology are essential molecules in cancer development. Thus, the present study examined their contributions to colorectal cancer progression in Saudi patients. METHODS: The expression of hTERT, TRF1, TRF2, POT1, ATR, ATM, Chk1 and Chk2 were measured via real-time PCR in matched cancerous and adjacent tissues of CRC patients. The protein level of hTERT, TRF1, TRF2, ATR, ATM, Chk1 and Chk2 were measured using immunohistochemistry. A region of hTERT core promoter was sequenced via Sanger sequencing. Methylation of CTCF binding site was examined via methylation-specific PCR. Finally, the length of telomere was estimated using q-PCR. RESULTS: Our results showed that POT1, ATR, Chk1 and Chk2 show increased expression in CRC relative to the adjacent mucosa. The expression levels of each gene were associated with clinicopathological characteristics of patients with CRC. There was a positive correlation between the age of the patients and hTERT expression. Regarding tumor site, telomere length, ATR, ATM and Chk1 were shown to be altered. No somatic mutation was detected in hTERT core promoter, and no differences in methylation patterns at CTCF binding site in the promoter between normal and cancer tissues. CONCLUSION: Analysis of targeted genes expression in colorectal cancer based on the clinical variables revealed that tumor location and age could have a role in gene expression and telomere length variations and this could be taken under consideration during CRC diagnosis and therapy. Other epigenetic mechanisms could influence hTERT expression in cancers. Our findings warrant further validation through experiments involving a larger number of patients.


Subject(s)
Colorectal Neoplasms/metabolism , DNA Damage , Gene Expression Regulation, Neoplastic , Neoplasm Proteins/biosynthesis , Telomere Homeostasis , Telomere-Binding Proteins/biosynthesis , Adult , Aged , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Female , Humans , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Male , Middle Aged , Neoplasm Proteins/genetics , Telomere-Binding Proteins/genetics
4.
Turk J Haematol ; 34(3): 200-206, 2017 Aug 02.
Article in English | MEDLINE | ID: mdl-28404540

ABSTRACT

OBJECTIVE: As evidence was shown that abnormal shortening of telomeres begins to accumulate in myelodysplastic syndrome (MDS) patients, this study was conducted to determine the relationship between the mRNA expression levels of telomere-binding proteins (TRF1/TRF2/TIN2/TPP1/POT1/RAP1) and the risk level in MDS. MATERIALS AND METHODS: There were 40 patients with MDS and 40 normal controls in this study. Methods including telomere content assays and quantitative reverse transcription-polymerase chain reaction were used to examine the mRNA levels of TRF1/TRF2/TIN2/TPP1/POT1/RAP1 in patients with MDS. RESULTS: Compared to the normal group used as a control, the mRNA expression levels of RAP1/POT1/TPP1 of the patients with MDS were decreased, whereas their levels of TRF1/TRF2 and TIN2 were increased. A positive correlation was found between the TRF1, TRF2, and TIN2 mRNA expression levels and the risk level of the International Prognostic Scoring System (IPSS) and the World Health Organization Prognostic Scoring System (WPSS) criteria; however, a negative correlation was found between RAP1/POT1/TPP1 mRNA expression levels and the risk levels of IPSS and WPSS criteria. CONCLUSION: Because the reduction of TRF1/TRF2/TIN2 mRNA expression and the increase of RAP1/POT1/TPP1 mRNA expression are closely related to the risk levels of the IPSS and WPSS criteria in MDS, it is thought that these telomere-binding proteins could lead to abnormal telomere length and function, which cause chromosomal abnormalities in MDS. With this evidence, we suggest that those proteins' mRNA expressions could be used as biomarkers for the assessment of the risk degree of MDS patients.


Subject(s)
Gene Expression Regulation , Myelodysplastic Syndromes/metabolism , RNA, Messenger/biosynthesis , Shelterin Complex/genetics , Telomere Homeostasis , Telomere-Binding Proteins/biosynthesis , Telomere/metabolism , Adult , Aged , Biomarkers/metabolism , Case-Control Studies , Female , Humans , Male , Middle Aged , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/pathology , RNA, Messenger/genetics , Risk Factors , Telomere/genetics , Telomere-Binding Proteins/genetics
5.
PLoS Genet ; 12(10): e1006373, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27741243

ABSTRACT

Filamentous growth is a hallmark of C. albicans pathogenicity compared to less-virulent ascomycetes. A multitude of transcription factors regulate filamentous growth in response to specific environmental cues. Our work, however, suggests the evolutionary history of C. albicans that resulted in its filamentous growth plasticity may be tied to a change in the general transcription machinery rather than transcription factors and their specific targets. A key genomic difference between C. albicans and its less-virulent relatives, including its closest relative C. dubliniensis, is the unique expansion of the TLO (TeLOmere-associated) gene family in C. albicans. Individual Tlo proteins are fungal-specific subunits of Mediator, a large multi-subunit eukaryotic transcriptional co-activator complex. This amplification results in a large pool of 'free,' non-Mediator associated, Tlo protein present in C. albicans, but not in C. dubliniensis or other ascomycetes with attenuated virulence. We show that engineering a large 'free' pool of the C. dubliniensis Tlo2 (CdTlo2) protein in C. dubliniensis, through overexpression, results in a number of filamentation phenotypes typically associated only with C. albicans. The amplitude of these phenotypes is proportional to the amount of overexpressed CdTlo2 protein. Overexpression of other C. dubliniensis and C. albicans Tlo proteins do result in these phenotypes. Tlo proteins and their orthologs contain a Mediator interaction domain, and a potent transcriptional activation domain. Nuclear localization of the CdTlo2 activation domain, facilitated naturally by the Tlo Mediator binding domain or artificially through an appended nuclear localization signal, is sufficient for the CdTlo2 overexpression phenotypes. A C. albicans med3 null mutant causes multiple defects including the inability to localize Tlo proteins to the nucleus and reduced virulence in a murine systemic infection model. Our data supports a model in which the activation domain of 'free' Tlo protein competes with DNA bound transcription factors for targets that regulate key aspects of C. albicans cell physiology.


Subject(s)
Candida albicans/genetics , Candidiasis/genetics , Telomere-Binding Proteins/genetics , Telomere/genetics , Animals , Candida albicans/growth & development , Candida albicans/pathogenicity , Candidiasis/microbiology , Candidiasis/pathology , Fungi/genetics , Fungi/growth & development , Fungi/pathogenicity , Gene Expression Regulation, Fungal , Genomics , Humans , Hyphae/genetics , Hyphae/growth & development , Hyphae/pathogenicity , Mice , Phenotype , Telomere-Binding Proteins/biosynthesis
6.
J Exp Med ; 213(8): 1429-40, 2016 07 25.
Article in English | MEDLINE | ID: mdl-27432940

ABSTRACT

The analysis of individuals with telomere defects may shed light on the delicate interplay of factors controlling genome stability, premature aging, and cancer. We herein describe two Coats plus patients with telomere and genomic defects; both harbor distinct, novel mutations in STN1, a member of the human CTC1-STN1-TEN1 (CST) complex, thus linking this gene for the first time to a human telomeropathy. We characterized the patients' phenotype, recapitulated it in a zebrafish model and rescued cellular and clinical aspects by the ectopic expression of wild-type STN1 or by thalidomide treatment. Interestingly, a significant lengthy control of the gastrointestinal bleeding in one of our patients was achieved by thalidomide treatment, exemplifying a successful bed-to-bench-and-back approach.


Subject(s)
Ataxia , Brain Neoplasms , Calcinosis , Central Nervous System Cysts , Gene Expression Regulation/drug effects , Leukoencephalopathies , Muscle Spasticity , Mutation , Retinal Diseases , Seizures , Telomere-Binding Proteins , Telomere , Thalidomide/administration & dosage , Animals , Ataxia/drug therapy , Ataxia/genetics , Ataxia/metabolism , Ataxia/pathology , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Calcinosis/drug therapy , Calcinosis/genetics , Calcinosis/metabolism , Calcinosis/pathology , Central Nervous System Cysts/drug therapy , Central Nervous System Cysts/genetics , Central Nervous System Cysts/metabolism , Central Nervous System Cysts/pathology , Disease Models, Animal , Female , Humans , Leukoencephalopathies/drug therapy , Leukoencephalopathies/genetics , Leukoencephalopathies/metabolism , Leukoencephalopathies/pathology , Male , Muscle Spasticity/drug therapy , Muscle Spasticity/genetics , Muscle Spasticity/metabolism , Muscle Spasticity/pathology , Retinal Diseases/drug therapy , Retinal Diseases/genetics , Retinal Diseases/metabolism , Retinal Diseases/pathology , Seizures/drug therapy , Seizures/genetics , Seizures/metabolism , Seizures/pathology , Telomere/genetics , Telomere/metabolism , Telomere/pathology , Telomere-Binding Proteins/biosynthesis , Telomere-Binding Proteins/genetics , Thalidomide/adverse effects , Zebrafish
7.
In Vitro Cell Dev Biol Anim ; 52(7): 772-81, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27251157

ABSTRACT

The proliferation and differentiation potential of aged bone marrow stromal cells (BMSCs) are significantly reduced. In order to improve the performance of the aged BMSCs, these cells were treated with 2 mM glutathione monoethyl ester (GSH-MEE) for 24 h. Proliferation rate, telomerase activity, telomere length, and differentiation to cholinergic neuron-like cells (CNLCs) were observed to increase. Though, the expression level of telomerase reverse transcriptase gene increased, but CTC1 and TEN1 genes from Ctc1-Stn1-Ten1 complex encoding proteins with regulatory function significantly decreased. Trypan blue exclusion assay was used to analyze the proliferation and, while telomere length, its several related gene expressions, and telomerase activity were measured using the real time reverse transcription-polymerase chain reaction and polymerase chain reaction enzyme-linked immunosorbent assay techniques, respectively. CNLCs differentiation potential was evaluated by estimating the percentage of choline acetyltransferase immunereactive cells.The results suggested that GSH-MEE could improve aged rat BMSC properties and would be of potential benefit for enhancing the performance of aged people's BMSCs.


Subject(s)
Glutathione/analogs & derivatives , Telomerase/biosynthesis , Telomere-Binding Proteins/biosynthesis , Telomere/genetics , Animals , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Choline O-Acetyltransferase/biosynthesis , Gene Expression Regulation, Developmental/drug effects , Glutathione/administration & dosage , Humans , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Neurons/cytology , Neurons/metabolism , Rats , Telomerase/genetics , Telomere/metabolism , Telomere-Binding Proteins/genetics
9.
Sci Rep ; 5: 10679, 2015 May 29.
Article in English | MEDLINE | ID: mdl-26022086

ABSTRACT

Rif1, identified as a regulator of telomerase in yeast, is an evolutionarily conserved protein and functions in diverse processes including telomere length regulation, epigenetic gene regulation, anti-checkpoint activity, DNA repair and establishing timing of firing at replication origins. Previously we had identified that all Rif1 homologues have PP1 interacting SILK-RVxF motif. In the present study, we show that Drosophila Rif1 is essential for normal fly development and loss of dRif1 impairs temporal regulation of initiation of DNA replication. In multiple tissues dRif1 colocalizes with HP1, a protein known to orchestrate timing of replication in fly. dRif1 associates with chromosomes in a mitotic stage-dependent manner coinciding with dephosphorylation of histones. Ectopic expression of dRif1 causes enlarged larval imaginal discs and early pupal lethality which is completely reversed by co-expression of PP1 87B, the major protein phosphatase in Drosophila, indicating genetic and functional interaction. These findings suggest that dRif1 is an adaptor for PP1 and functions by recruiting PP1 to multiple sites on the chromosome.


Subject(s)
Carrier Proteins/genetics , DNA Replication/genetics , Drosophila Proteins/genetics , Drosophila/growth & development , Protein Phosphatase 1/genetics , Telomere-Binding Proteins/biosynthesis , Animals , Chromosomal Proteins, Non-Histone/biosynthesis , Chromosomal Proteins, Non-Histone/genetics , Drosophila/genetics , Drosophila Proteins/biosynthesis , Gene Expression Regulation, Developmental , Phosphorylation , Protein Phosphatase 1/biosynthesis , Telomere/genetics , Telomere-Binding Proteins/genetics
10.
PLoS One ; 10(4): e0123531, 2015.
Article in English | MEDLINE | ID: mdl-25856570

ABSTRACT

RAP1 (RAS proximate 1), a small GTP-binding protein of the RAS superfamily, is a putative oncogene that is highly expressed in several malignant cell lines and types of cancers, including some types of squamous cell carcinoma. However, the participation of RAP1 in cervical carcinogenesis is unknown. We conducted a cross-sectional study of paraffin-embedded cervical biopsies to determine the association of RAP1 with cervical intraepithelial neoplasia (CIN). Standard and quantitative immunohistochemistry assessment of RAP1 expression in fixed tissue was performed on 183 paraffin-embedded cervical biopsies that were classified as normal or non-dysplastic mucosa (NDM) (n = 33); CIN grade 1 (n = 84) and CIN grade 2/3 (n = 66). A gradual increase in RAP1 expression in NDM < CIN 1 < CIN 2/3 (p<0.001) specimens was observed and was in agreement with the histopathologic diagnosis. A progressive increase in the RAP1 expression levels increased the risk of CIN 1 [odds ratio (OR) = 3.50; 95% confidence interval (CI) 1.30-10.64] 3.5 fold and the risk of CIN 2/3 (OR = 19.86, 95% CI 6.40-70.79) nearly 20 fold when compared to NDM. In addition, stereotype ordinal regression analysis showed that this progressive increase in RAP1 expression more strongly impacted CIN 2/3 than CIN 1. Our findings suggest that RAP1 may be a useful biomarker for the diagnosis of CIN.


Subject(s)
Biomarkers, Tumor/biosynthesis , Carcinoma, Squamous Cell/genetics , Telomere-Binding Proteins/biosynthesis , Uterine Cervical Dysplasia/genetics , Adolescent , Adult , Aged , Biomarkers, Tumor/genetics , Carcinogenesis/genetics , Carcinoma, Squamous Cell/diagnosis , Carcinoma, Squamous Cell/pathology , Female , Gene Expression Regulation, Neoplastic , Humans , Middle Aged , Neoplasm Staging , Risk Factors , Shelterin Complex , Telomere-Binding Proteins/genetics , Uterine Cervical Dysplasia/diagnosis , Uterine Cervical Dysplasia/pathology
11.
Nat Cell Biol ; 17(4): 458-69, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25774833

ABSTRACT

The role of the conserved meiotic telomere bouquet has been enigmatic for over a century. We showed previously that disruption of the fission yeast bouquet impairs spindle formation in approximately half of meiotic cells. Surprisingly, bouquet-deficient meiocytes with functional spindles harbour chromosomes that fail to achieve spindle attachment. Kinetochore proteins and the centromeric histone H3 variant Cnp1 fail to localize to those centromeres that exhibit spindle attachment defects in the bouquet's absence. The HP1 orthologue Swi6 also fails to bind these centromeres, suggesting that compromised pericentromeric heterochromatin underlies the kinetochore defects. We find that centromeres are prone to disassembly during meiosis, but this is reversed by localization of centromeres to the telomere-proximal microenvironment, which is conducive to heterochromatin formation and centromere reassembly. Accordingly, artificially tethering a centromere to a telomere rescues the tethered centromere but not other centromeres. These results reveal an unanticipated level of control of centromeres by telomeres.


Subject(s)
Centromere/metabolism , Meiosis/genetics , Schizosaccharomyces/genetics , Spindle Apparatus/genetics , Telomere/metabolism , Chromosomal Proteins, Non-Histone/biosynthesis , Chromosomal Proteins, Non-Histone/genetics , Heterochromatin/metabolism , Histones/metabolism , Kinetochores/metabolism , Schizosaccharomyces pombe Proteins/biosynthesis , Schizosaccharomyces pombe Proteins/genetics , Telomere-Binding Proteins/biosynthesis , Telomere-Binding Proteins/genetics
12.
J Cell Biol ; 208(4): 415-28, 2015 Feb 16.
Article in English | MEDLINE | ID: mdl-25688135

ABSTRACT

Telomeres and centromeres have traditionally been considered to perform distinct roles. During meiotic prophase, in a conserved chromosomal configuration called the bouquet, telomeres gather to the nuclear membrane (NM), often near centrosomes. We found previously that upon disruption of the fission yeast bouquet, centrosomes failed to insert into the NM at meiosis I and nucleate bipolar spindles. Hence, the trans-NM association of telomeres with centrosomes during prophase is crucial for efficient spindle formation. Nonetheless, in approximately half of bouquet-deficient meiocytes, spindles form properly. Here, we show that bouquet-deficient cells can successfully undergo meiosis using centromere-centrosome contact instead of telomere-centrosome contact to generate spindle formation. Accordingly, forced association between centromeres and centrosomes fully rescued the spindle defects incurred by bouquet disruption. Telomeres and centromeres both stimulate focal accumulation of the SUN domain protein Sad1 beneath the centrosome, suggesting a molecular underpinning for their shared spindle-generating ability. Our observations demonstrate an unanticipated level of interchangeability between the two most prominent chromosomal landmarks.


Subject(s)
Centromere/genetics , Centrosome/metabolism , Meiosis , Schizosaccharomyces/genetics , Spindle Apparatus/genetics , Telomere/genetics , Cell Cycle Proteins/biosynthesis , Cell Cycle Proteins/genetics , Chromatin/genetics , Chromosomes, Fungal/genetics , Dyneins/genetics , Endoribonucleases/genetics , Green Fluorescent Proteins/biosynthesis , Green Fluorescent Proteins/genetics , Nuclear Envelope/metabolism , Nuclear Proteins/genetics , Schizosaccharomyces/cytology , Schizosaccharomyces pombe Proteins/biosynthesis , Schizosaccharomyces pombe Proteins/genetics , Schizosaccharomyces pombe Proteins/metabolism , Telomere-Binding Proteins/biosynthesis , Telomere-Binding Proteins/genetics
13.
Invest Ophthalmol Vis Sci ; 56(2): 711-21, 2015 Jan 08.
Article in English | MEDLINE | ID: mdl-25574050

ABSTRACT

PURPOSE: Recently, RAP1 (Telomeric Repeat Binding Factor 2, Interacting Protein [TERF2IP]) was discovered as a modulator that selectively regulates nuclear factor light chain kappa enhancer of activated B cells (NFκB) signaling. The roles of RAP1 in regulation of inflammation and angiogenesis for corneal recovery following corneal injury remain poorly understood. The effects of RAP1 deletion on corneal resurfacing and neovascularization in a corneal alkali burn mouse model were examined. METHODS: Corneal defects and neovascularization were induced in vivo by infliction of an alkali burn to the cornea with 1 N sodium hydroxide solution in RAP1 knock-out (RKO) and wild-type (RWT) mice. Corneal resurfacing was evaluated using slit-lamp biomicroscopy. Neovascularization following injury was evaluated by bright view biomicroscopy and immunofluorescence staining with the endothelial marker platelet endothelial cell adhesion molecule (PECAM). The cytokine profiles of corneal tissue involved in inflammation and neovascularization following injury was compared between RKO and RWT mice. Corneal epithelial cells were isolated for classic scratch wound healing assay and further testing with lipopolysaccharide stimulation. RESULTS: Resurfacing of the burned cornea was accelerated and angiogenesis was suppressed, faster recovery of corneal epithelial cells from classic scratch wound healing and superior tolerance of lipopolysaccharides challenge was observed in the RKO compared to RWT. Molecular investigation revealed that deletion of RAP1 reduced upregulation of inflammatory cytokine (IL1A), finely regulated the expression of angiogenic factor (VEGF), and antiangiogenic factor (PEDF), following injury for better corneal recovery. CONCLUSIONS: Deficiency of RAP1 facilitates corneal recovery after injury. Specificity of RAP1 inhibition may lead to design of specific inhibitors of NFκB in the treatment of ocular injuries.


Subject(s)
Burns, Chemical/metabolism , Corneal Injuries/metabolism , Corneal Neovascularization/genetics , Eye Burns/metabolism , Telomere-Binding Proteins/genetics , Up-Regulation , Wound Healing , Alkalies/toxicity , Animals , Burns, Chemical/complications , Burns, Chemical/pathology , Corneal Injuries/complications , Corneal Injuries/pathology , Corneal Neovascularization/metabolism , Corneal Neovascularization/pathology , Disease Models, Animal , Epithelium, Corneal/drug effects , Epithelium, Corneal/metabolism , Epithelium, Corneal/pathology , Eye Burns/complications , Eye Burns/pathology , Mice , Mice, Knockout , RNA, Messenger/genetics , Shelterin Complex , Signal Transduction , Telomere-Binding Proteins/biosynthesis
14.
Breast Cancer Res Treat ; 145(3): 581-91, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24807106

ABSTRACT

Mammalian telomeric DNA consists of tandem repeats of the sequence TTAGGG associated with a specialized set of proteins, known collectively as Shelterin. These telosomal proteins protect the ends of chromosomes against end-to-end fusion and degradation. Short telomeres in breast cancer cells confer telomere dysfunction and this can be related to Shelterin proteins and their level of expression in breast cancer cell lines. This study investigates whether expression of Shelterin and Shelterin-associated proteins are altered, and influence the protection and maintenance of telomeres, in breast cancer cells. 5-aza-2'-deoxycytidine (5-aza-CdR) and trichostatin A (TSA) were used in an attempt to reactivate the expression of silenced genes. Our studies have shown that Shelterin and Shelterin-associated genes were down-regulated in breast cancer cell lines; this may be due to epigenetic modification of DNA as the promoter region of POT1 was found to be partially methylated. Shelterin genes expression was up-regulated upon treatment of 21NT breast cancer cells with 5-aza-CdR and TSA. The telomere length of treated 21NT cells was measured by q-PCR showed an increase in telomere length at different time points. Our studies have shown that down-regulation of Shelterin genes is partially due to methylation in some epithelial breast cancer cell lines. Removal of epigenetic silencing results in up-regulation of Shelterin and Shelterin-associated genes which can then lead to telomere length elongation and stability.


Subject(s)
Azacitidine/analogs & derivatives , Breast Neoplasms/drug therapy , Hydroxamic Acids/pharmacology , Telomere Homeostasis/drug effects , Telomere-Binding Proteins/biosynthesis , Antineoplastic Agents/pharmacology , Azacitidine/pharmacology , Cell Line, Tumor , DNA/genetics , DNA Methylation/genetics , Decitabine , Epigenesis, Genetic , Female , Histones/metabolism , Humans , MCF-7 Cells , Methyltransferases/antagonists & inhibitors , Promoter Regions, Genetic/genetics , Shelterin Complex , Telomere/drug effects , Telomere/physiology , Telomere-Binding Proteins/genetics
15.
Biochim Biophys Acta ; 1840(7): 2222-33, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24631651

ABSTRACT

BACKGROUND: Telomere is protected by its G-quadruplex, T-loop structure, telomerase, and binding protein complex. Protein POT1 (protection of telomeres 1) is one subunit of telomere binding protein complex Shelterin. POT1 acts as a regulator of telomerase-dependent telomere length, and it can help telomere to form D-loop structure to stabilize telomere. POT1 protects telomere ends from ATR-dependent DNA damage response as well. METHODS: Extensive methods were used, including CD, EMSA, ITC, PCR stop assay, luciferase reporter assay, quantitative real-time PCR, Western blot, chromatin immunoprecipitation (Ch-IP), cloning, expression and purification of proteins. RESULTS: We found a new G-rich 30-base-pair long sequence (P-pot1 G18) located from -165 to -136 base pairs upstream of the translation starting site of protein POT1. This sequence in the promoter region of pot1 gene formed G-quadruplex resulting in down-regulation of pot1 gene transcription. This G-rich sequence is close to a binding site "TCCC" for transcription factor hnRNP K (heterogeneous nuclear ribonucleoprotein K), and its conversion to G-quadruplex prevented the access of hnRNP K to this binding site. The binding of hnRNP K could up-regulate pot1 gene transcription. TMPyP4 (meso-tetra(N-methyl-4-pyridyl)porphine) has been widely used as G-quadruplex binding ligand, which stabilized the G-quadruplex in vitro and in cellulo, resulting in down-regulation of pot1 gene transcription. CONCLUSIONS: This G-quadruplex might become a potentially new drug target for antitumor agents. GENERAL SIGNIFICANCE: Our results first demonstrated that G-quadruplex formation can affect the binding of transcription factor to its nearby binding site, and thus making additional influence to gene transcription.


Subject(s)
G-Quadruplexes/drug effects , Telomere-Binding Proteins/biosynthesis , Telomere-Binding Proteins/genetics , Telomere/genetics , Binding Sites , DNA, Single-Stranded/genetics , Gene Expression Regulation , Humans , Porphyrins/pharmacology , Protein Binding , Shelterin Complex , Telomerase/genetics , Telomerase/metabolism , Telomere-Binding Proteins/chemistry
16.
J Immunol ; 191(3): 1496-504, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23804711

ABSTRACT

Trafficking of malignant lymphocytes is fundamental to the biology of chronic lymphocytic leukemia (CLL). Transendothelial migration (TEM) of normal lymphocytes into lymph nodes requires the chemokine-induced activation of Rap1 and αLß2 integrin. However, in most cases of CLL, Rap1 is refractory to chemokine stimulation, resulting in failed αLß2 activation and TEM unless α4ß1 is coexpressed. In this study, we show that the inability of CXCL12 to induce Rap1 GTP loading in CLL cells results from failure of Rap1-containing endosomes to translocate to the plasma membrane. Furthermore, failure of chemokine-induced Rap1 translocation/GTP loading was associated with a specific pattern of cellular IgD distribution resembling that observed in normal B cells anergized by DNA-based Ags. Anergic features and chemokine unresponsiveness could be simultaneously reversed by culturing CLL cells ex vivo, suggesting that these two features are coupled and driven by stimuli present in the in vivo microenvironment. Finally, we show that failure of Rap1 translocation/GTP loading is linked to defective activation of phospholipase D1 and its upstream activator Arf1. Taken together, our findings indicate that chemokine unresponsiveness in CLL lymphocytes results from failure of Arf1/phospholipase D1-mediated translocation of Rap1 to the plasma membrane for GTP loading and may be a specific feature of anergy induced by DNA Ags.


Subject(s)
ADP-Ribosylation Factor 1/metabolism , Clonal Anergy/immunology , Leukemia, Lymphocytic, Chronic, B-Cell/immunology , Phospholipase D/metabolism , Telomere-Binding Proteins/metabolism , B-Lymphocytes/immunology , Cell Membrane/metabolism , Chemokine CXCL12/metabolism , Endosomes/metabolism , Enzyme Activation , Humans , Immunoglobulin D/immunology , Immunoglobulin D/metabolism , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Lymph Nodes/cytology , Lymph Nodes/immunology , Lymphocyte Function-Associated Antigen-1/metabolism , Lymphocytes/cytology , Lymphocytes/immunology , Lymphocytes/metabolism , Shelterin Complex , Telomere-Binding Proteins/biosynthesis , Transendothelial and Transepithelial Migration/immunology , Tumor Cells, Cultured , rap GTP-Binding Proteins/biosynthesis
17.
Oncol Rep ; 28(2): 622-8, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22614916

ABSTRACT

Rap1GAP which regulates the GTP-GDP form switch of Rap1 is a member of the GTPase-activating protein (GAP) family and has recently received substantial attention. Rap1GAP is thought of as a putative tumor suppressor gene and plays an important role in human tumor progression including pancreatic cancer, thyroid cancer and melanoma. In the current study, we found that the expression of Rap1GAP was lower in acute myeloid leukemia (AML) patients compared to non-malignant blood disease patients. The expression of Rap1GAP was also low in HL-60, NB4, U937 and SHI-1 myeloid leukemia cell lines. Upregulated Rap1GAP in NB4 and HL-60 cells promoted cell differentiation induced by ATRA or TPA compared to the empty vector control cells. Furthermore, Rap1GAP-transfected cells also showed a higher rate of apoptosis in response to arsenic trioxide compared to the control counterpart cells. In addition, we found that increased expression of Rap1GAP promoted leukemia cell invasion may be due to matrix metalloproteinase 9 (MMP9). In conclusion, these results demonstrated that Rap1GAP promoted leukemia cell differentiation and apoptosis, but increased leukemia cell invasion in vitro.


Subject(s)
GTPase-Activating Proteins/biosynthesis , Leukemia, Megakaryoblastic, Acute/metabolism , Leukemia, Myeloid, Acute/metabolism , Leukemia, Promyelocytic, Acute/metabolism , Apoptosis/physiology , Cell Differentiation/physiology , Down-Regulation , GTPase-Activating Proteins/genetics , HeLa Cells , Humans , Leukemia, Megakaryoblastic, Acute/genetics , Leukemia, Megakaryoblastic, Acute/pathology , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Leukemia, Promyelocytic, Acute/genetics , Leukemia, Promyelocytic, Acute/pathology , Matrix Metalloproteinase 9/metabolism , Neoplasm Invasiveness , Shelterin Complex , Telomere-Binding Proteins/antagonists & inhibitors , Telomere-Binding Proteins/biosynthesis , Telomere-Binding Proteins/genetics , Transduction, Genetic , U937 Cells , Up-Regulation
18.
Cancer Med ; 1(2): 165-75, 2012 Oct.
Article in English | MEDLINE | ID: mdl-23342266

ABSTRACT

Telomere dysregulation occurs in both the in situ and invasive stages of many carcinomas, including breast. Knockout experiments have identified several telomere-associated proteins required for proper telomere function and maintenance, including telomere repeat-binding factor 1 and 2 (TRF1 and TRF2), protection of telomeres (POT1), and TRF1-interacting nuclear factor 2 (TIN2). Using telomere content assays and quantitative reverse transcription-polymerase chain reaction (RT-PCR), we examined the relationship between telomere length and the mRNA levels of telomere-associated proteins in breast tumors. The levels of TRF2, TRF1, TIN2, and POT1 mRNA, but not telomerase reverse transcriptase (TERT) RNA, are inversely correlated with telomere content in breast tumors. Significant associations were identified between the mRNA levels of TRF1, TIN2, and POT1; however, there were no significant associations with the mRNA levels of TRF2 or TERT. These associations suggest that a complex transcriptional program coordinately regulates the expression of these mRNAs. We examined the promoter regions of the telomere-associated proteins to identify transcription factors consistent with the observed patterns of presumed coordinate expression. We demonstrated in human breast cancer cell lines that expressions of TRF1, TIN2, and POT1 are upregulated by dexamethasone, suggesting activation of the glucocorticoid receptor, whereas TERT, TRF2, TRF1, TIN2, and POT1 are upregulated by tumor necrosis factor-α (TNF-α), suggesting activation of the nuclear factor kappa B transcription factor. These findings link telomere content in breast tumors to the coordinate expression of several telomere-associated proteins previously shown to be negative regulators of telomere length in cell lines. The results further suggest a possible link between the expressions of the telomere-associated proteins and mediators of stress and inflammation.Telomere content assays and quantitative RT-PCR demonstrate that the levels of TRF2, TRF1, TIN2, and POT1 mRNA, but not telomerase reverse transcriptase (TERT) RNA, are inversely correlated with telomere content in breast tumors. Within human breast cancer cell lines, expressions of TRF1, TIN2, and POT1 are upregulated by dexamethasone, suggesting activation of the glucocorticoid receptor, whereas TERT, TRF2, TRF1, TIN2, and POT1 are upregulated by TNF-α, suggesting activation of the NFκB transcription factor. These findings link telomere content in breast tumors to the expression of several telomere-associated proteins previously shown to be negative regulators of telomere length in cell lines and suggest a link between the expressions of the telomere-associated proteins and mediators of stress and inflammation.


Subject(s)
Breast Neoplasms/metabolism , Carcinoma/metabolism , Telomere Homeostasis , Telomere-Binding Proteins/biosynthesis , Telomere/genetics , Adult , Aged , Aged, 80 and over , Breast Neoplasms/genetics , Carcinoma/genetics , Cell Line, Tumor , Dexamethasone/metabolism , Female , Humans , MCF-7 Cells , Middle Aged , NF-kappa B/metabolism , Promoter Regions, Genetic , RNA, Messenger/biosynthesis , Shelterin Complex , Telomerase/genetics , Telomerase/metabolism , Telomere-Binding Proteins/genetics , Telomere-Binding Proteins/metabolism , Telomeric Repeat Binding Protein 1/genetics , Telomeric Repeat Binding Protein 1/metabolism , Telomeric Repeat Binding Protein 2/genetics , Telomeric Repeat Binding Protein 2/metabolism , Tumor Necrosis Factor-alpha/metabolism
19.
J Appl Physiol (1985) ; 112(5): 773-81, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22162529

ABSTRACT

Located at the end of chromosomes, telomeres are progressively shortened with each replication of DNA during aging. Integral to the regulation of telomere length is a group of proteins making up the shelterin complex, whose tissue-specific function during physiological stress is not well understood. In this study, we examine the mRNA and protein levels of proteins within and associated with the shelterin complex in subjects (n = 8, mean age = 44 yr) who completed a physiological stress of seven marathons in 7 days. Twenty-two to 24 h after the last marathon, subjects had increased mRNA levels of DNA repair enzymes Ku70 and Ku80 (P < 0.05) in both skeletal muscle and peripheral blood mononuclear cells (PBMCs). Additionally, the PBMCs displayed an increment in three shelterin protein mRNA levels (TRF1, TRF2, and Pot-1, P < 0.05) following the event. Seven days of ultrarunning did not result in changes in mean telomere length, telomerase activity, hTert mRNA, or hterc mRNAs found in PBMCs. Higher protein concentrations of TRF2 were found in skeletal muscle vs. PBMCs at rest. Mean telomere length in skeletal muscle did not change and did not contain detectable levels of htert mRNA or telomerase activity. Furthermore, changes in the PBMCs could not be attributed to changes in the proportion of subtypes of CD4(+) or CD8(+) cells. We have provided the first evidence that, in humans, proteins within and associated with the shelterin complex increase at the mRNA level in response to a physiological stress differentially in PBMCs and skeletal muscle.


Subject(s)
Leukocytes, Mononuclear/metabolism , Muscle, Skeletal/metabolism , RNA, Messenger/blood , Running/physiology , Telomere-Binding Proteins/genetics , Adult , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/physiology , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/physiology , DNA Repair Enzymes/genetics , DNA Repair Enzymes/metabolism , Female , Humans , Leukocytes, Mononuclear/physiology , Male , Muscle, Skeletal/physiology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Shelterin Complex , Stress, Physiological/physiology , Telomerase/genetics , Telomerase/metabolism , Telomere/metabolism , Telomere/physiology , Telomere-Binding Proteins/biosynthesis
20.
Asia Pac J Clin Oncol ; 7(4): 339-45, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22151982

ABSTRACT

AIM: The aim of this study was to investigate the expression of telomere repeat binding factor 1 (TRF1), TRF2 and protection of telomeres 1 (POT1) in gastric cancer and their relationships with clinicopathological features and telomerase activity. METHODS: In total 36 gastric cancer tissue and paired adjacent normal tissue were analyzed. The mRNA expression of telomere binding proteins TRF1, TRF2 and POT1 were measured using quantitative reverse transcription polymerase chain reaction, and telomerase activity was assessed by the telomeric repeat amplification protocol/enzyme linked immunosorbent assay method. RESULTS: The expression of POT1 was significantly higher in tumor tissue than in adjacent normal tissue (P < 0.001). Levels of TRF2 mRNA were significantly higher in bigger tumors (diameter ≥ 5 cm) than in small tumors (diameter < 5 cm) (P = 0.043). POT1 mRNA transcription levels were higher in tumors with lymph nodes metastases than in those without lymph nodes metastases (P = 0.048). POT1 expression was significantly correlated with tumor stage (P = 0.008). A higher level of expression of POT1 was observed in late-stage tumors (III, IV) than in early stage tumors (I, II). Telomerase activity was significantly higher in gastric cancers than in corresponding normal tissue (P < 0.001). Moreover, POT1 expression was significantly positive correlated with telomerase activity (r = 0.572, P < 0.01). CONCLUSION: POT1 was overexpressed in gastric cancer and may be associated with stomach carcinogenesis and gastric cancer progression.


Subject(s)
Stomach Neoplasms/metabolism , Telomere-Binding Proteins/biosynthesis , Adult , Aged , Female , Humans , Male , Middle Aged , Reverse Transcriptase Polymerase Chain Reaction , Shelterin Complex , Stomach Neoplasms/enzymology , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Telomerase/metabolism , Telomere-Binding Proteins/genetics , Telomere-Binding Proteins/metabolism , Telomeric Repeat Binding Protein 1/biosynthesis , Telomeric Repeat Binding Protein 1/genetics , Telomeric Repeat Binding Protein 1/metabolism , Telomeric Repeat Binding Protein 2/biosynthesis , Telomeric Repeat Binding Protein 2/genetics , Telomeric Repeat Binding Protein 2/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...