Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Cytogenet Genome Res ; 161(8-9): 449-462, 2021.
Article in English | MEDLINE | ID: mdl-34657040

ABSTRACT

PIN2/TRF1-interacting telomerase inhibitor 1 (PinX1) can inhibit tumor growth by inhibiting telomerase activity. However, only few studies investigated the expression and function of PinX1 in nonalcoholic fatty liver disease (NAFLD). Thus, here we aimed to explore the roles of PinX1 in high-fat diet (HFD)-induced NAFLD in mice and in isolated hepatocytes. The mRNA expression of PinX1 and mTERT as well as telomere length were analyzed by RT-PCR. Pathological changes were detected by HE staining and oil red O staining. Triglyceride, cholesterol, alanine aminotransferase, aspartic aminotransferase, and telomerase activity were detected by ELISA. Hepatocyte apoptosis was determined by TUNEL and flow cytometry, and protein expression was analyzed by western blotting. We found that the expression of PinX1 was upregulated in the HFD group compared with the WT group. PinX1 knockout improved HFD-induced liver injury in mice and exhibited less lipid accumulation in hepatocytes. Moreover, telomere length, telomerase activity, and mTERT expression were significantly reduced in liver tissues of HFD-induced mice and palmitic acid-induced hepatocytes, while PinX1 knockout attenuated the effect. Furthermore, HFD-induced PinX1-/- mice exhibited less hepatocyte apoptosis than HFD-induced WT mice. Besides, PinX1 knockout inhibited the increase of cleaved caspase-3 and cleaved PARP expression in vivo and in vitro. Moreover, inhibition of mTERT reversed the effect of PinX1 knockout in hepatocytes. Taken together, our findings indicate that PinX1 promotes hepatocyte apoptosis and lipid accumulation by decreasing telomere length and telomerase activity in the development of NAFLD. PinX1 might be a target for the treatment of NAFLD.


Subject(s)
Apoptosis , Cell Cycle Proteins/deficiency , Liver , Non-alcoholic Fatty Liver Disease/enzymology , Non-alcoholic Fatty Liver Disease/pathology , Telomerase/metabolism , Tumor Suppressor Proteins/deficiency , Animals , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Disease Models, Animal , Hepatocytes/cytology , Hepatocytes/enzymology , Hepatocytes/pathology , Liver/cytology , Liver/enzymology , Liver/pathology , Male , Mice , Non-alcoholic Fatty Liver Disease/genetics , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
2.
Angiogenesis ; 22(4): 507-519, 2019 11.
Article in English | MEDLINE | ID: mdl-31254127

ABSTRACT

BACKGROUND: PIN2/TRF1-interacting telomerase inhibitor 1 (PinX1) is a tumor suppressor in various tumors. However, the molecular mechanism underlying PinX1's role in cancer development and progression remains unclear. In this study, we aimed to uncover the new molecular mechanism and role of PinX1 in renal cell carcinoma (RCC) progression. METHODS: We used miRNA microarray to detect the different expressed miRNAs upon PinX1 knockdown. Chromatin immunoprecipitation and Luciferase reporter assays were taken to identify the molecular mechanism of PinX1 in regulating mir-125-3p. In situ hybridization was performed to analyze the expression of mir-125a-3p in RCC using tissue microarray. The correlations between the mir-125a-3p expression level and clinicopathological features were evaluated using the χ2 test. The role and molecular mechanism of PinX1 in RCC angiogenesis were investigated through a series of in vitro and in vivo experiments. RESULTS: In this study, we discovered a new molecular mechanism of PinX1, in which PinX1 transcriptionally activated mir-125a-3p expression, thereby inhibiting the expression of vascular endothelial growth factor (VEGF), which is the target gene of mir-125a-3p. PinX1 also repressed tumor angiogenesis by increasing the mir-125a-3p expression in renal cancer. Moreover, the loss of mir-125a-3p expression was manifested in patients with RCC, and low miR-125a-3p levels correlated with poor survival of these patients. CONCLUSIONS: PinX1 represses renal cancer angiogenesis through mir-125a-3p/VEGF signal pathway. The miR-125a-3p may be a candidate clinical prognostic marker and a novel therapeutic target in RCC.


Subject(s)
Carcinoma, Renal Cell , Cell Cycle Proteins/metabolism , Kidney Neoplasms , MicroRNAs/metabolism , Neovascularization, Pathologic , RNA, Neoplasm/metabolism , Signal Transduction , Tumor Suppressor Proteins/metabolism , Vascular Endothelial Growth Factor A/metabolism , Carcinoma, Renal Cell/blood supply , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , Humans , Kidney Neoplasms/blood supply , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology
3.
Biochem Biophys Res Commun ; 514(1): 127-133, 2019 06 18.
Article in English | MEDLINE | ID: mdl-31027734

ABSTRACT

OBJECTIVE: Early malignant transformation of nasopharyngeal carcinoma(NPC) is associated with Epstein-Barr virus(EBV) infection and telomerase activation. The EBV latent membrane protein 1(LMP1) regulates expression of various genes by triggering NF-κB signaling pathway. PINX1 is a well-identified tumor suppressor gene by inhibiting telomerase activity and cancer cell growth. However, whether and how EBV inhibit PINX1 expression and activate telomerase in NPC is still incompletely elucidated. METHODS: Immunohistochemistry, real-time PCR and Western blotting were utilized to explore the expression of PINX1. Chromatin immunoprecipitation(ChIP) and Dual-luciferase reporter assay were used to elucidate the regulatory mechanism between NF-κB and PINX1. TRAP-SYBR Green assay and Southern blotting were utilized to detect telomerase activity and telomere length. CCK8 and EdU tests were conducted to measure proliferation ability. RESULTS: We demonstrated that PINX1 is down-regulated in NPC for the first time. Mechanistically, we found that LMP1 could inhibit the transcriptional activity of PINX1 by promoting the binding of p65 to three specific sites in PINX1 promoter, significantly, two(-1698/-1689, tgcaatttcc; -206/-197, cgggctttac) of which have not been reported. In addition, we also observed that LMP1 overexpression resulted in increased telomerase activity, prolonged telomere length and enhanced proliferation. CONCLUSION: We first discovered EBV led to reduced PINX1 expression through LMP1-NF-κB-PINX1 axis, which up-regulated telomerase activity in NPC. And hence, the tumor cells acquired the ability to proliferate more exuberantly. This signaling pathway illustrates the relationship between EBV latent infection and telomerase activation, and further provides new thinking for early diagnosis and treatment in NPC.


Subject(s)
Cell Cycle Proteins/genetics , Epstein-Barr Virus Infections/metabolism , NF-kappa B/metabolism , Nasopharyngeal Carcinoma/metabolism , Nasopharyngeal Neoplasms/metabolism , Telomerase/metabolism , Tumor Suppressor Proteins/genetics , Viral Matrix Proteins/metabolism , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Down-Regulation , Enzyme Activation , Epstein-Barr Virus Infections/complications , Epstein-Barr Virus Infections/genetics , Epstein-Barr Virus Infections/virology , Gene Expression Regulation, Neoplastic , Herpesvirus 4, Human/physiology , Humans , Nasopharyngeal Carcinoma/etiology , Nasopharyngeal Carcinoma/genetics , Nasopharyngeal Carcinoma/virology , Nasopharyngeal Neoplasms/etiology , Nasopharyngeal Neoplasms/genetics , Nasopharyngeal Neoplasms/virology , Signal Transduction , Tumor Suppressor Proteins/metabolism
4.
Cell Physiol Biochem ; 49(1): 282-294, 2018.
Article in English | MEDLINE | ID: mdl-30138944

ABSTRACT

BACKGROUND/AIMS: Cancer stem cells (CSCs) are important factors for the continuous growth, recurrence, and metastasis of malignant tumors. They are responsible for the ineffectiveness of traditional radiotherapy and chemotherapy toward malignant tumors. Currently, stem cells or side-population cells have been isolated from many cancer cell lines and malignant tumor tissues, including nasopharyngeal carcinoma. Exploring the biological characteristics of CSCs for CSC-targeted therapy has gained importance. CSCs possess higher telomerase activity; thus, the use of the gene encoding telomerase inhibitor PinX1 gene to target telomerase in CSCs and inhibit proliferation, invasion, and metastasis of CSCs has become an important means for the treatment of malignant tumors. PinX1 may regulate complex pathways, including TRF1, Mad1/c-Myc, and p53. METHODS: In this study, nasopharyngeal CD133+ CSCs were sorted using CD133 immunomagnetic beads by flow cytometry The successful isolation of CD133+ CSCs was confirmed by examining their surface markers, namely CD44, NaNOG, and SOX2 as well as their ability to undergo in vivo tumorigenesis and in vitro sphere formation, proliferation, migration, and invasion. In addition, CD133+ CSCs were transfected with the constructed PinX1 overexpression plasmid or siRNA and the resulting effects on their proliferation, migration, invasion, and apoptosis were detected using cell counting kit-8 (CCK-8), transwell assay, and scratch test, respectively. Furthermore, their effects on mRNA and protein levels of TRF1, TRF2, Mad1, c-Myc, and p53 were examined using quantitative real-time PCR and western blot, respectively. RESULTS: The overexpression of PinX1 in CD133+ CSCs significantly decreased hTERT (P < 0.001), inhibited proliferation, migration, and invasion, induced apoptosis, and significantly decreased c-Myc mRNA levels (P < 0.001), while it increased TRF1, Mad1, and p53 mRNA levels (all P < 0.001). On the other hand, PinX1 silencing in CD133+ CSCs significantly decreased TRF1, Mad1, and p53 mRNA levels (all P < 0.01), while it increased hTERT and c-Myc mRNA levels (all P < 0.05). CONCLUSION: These results indicate that PinX1 downregulates telomerase activity in CD133+ CSCs, inhibits its proliferation, migration, and invasion, and induces apoptosis possibly through TRF1, Mad1/c-Myc, and p53-mediated pathways.


Subject(s)
Cell Proliferation , Neoplastic Stem Cells/metabolism , Signal Transduction , Telomerase/metabolism , Tumor Suppressor Proteins/metabolism , AC133 Antigen/metabolism , Animals , Carcinoma/metabolism , Carcinoma/pathology , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Down-Regulation , Female , Humans , Mice , Mice, Nude , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms/metabolism , Nasopharyngeal Neoplasms/pathology , Neoplastic Stem Cells/cytology , Nuclear Proteins/metabolism , Proto-Oncogene Proteins c-myc/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Telomere-Binding Proteins/metabolism , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/antagonists & inhibitors , Tumor Suppressor Proteins/genetics
5.
J Virol ; 91(2)2017 Jan 15.
Article in English | MEDLINE | ID: mdl-27847364

ABSTRACT

Enterovirus 71 (EV71) is an emerging pathogen causing hand, foot, and mouth disease (HFMD) and fatal neurological diseases in infants and young children due to their underdeveloped immunocompetence. EV71 infection can induce cellular apoptosis through a variety of pathways, which promotes EV71 release. The viral protease 3C plays an important role in EV71-induced apoptosis. However, the molecular mechanism responsible for 3C-triggered apoptosis remains elusive. Here, we found that EV71 3C directly interacted with PinX1, a telomere binding protein. Furthermore, 3C cleaved PinX1 at the site of Q50-G51 pair through its protease activity. Overexpression of PinX1 reduced the level of EV71-induced apoptosis and EV71 release, whereas depletion of PinX1 by small interfering RNA promoted apoptosis induced by etoposide and increased EV71 release. Taken together, our study uncovered a mechanism that EV71 utilizes to promote host cell apoptosis through cleavage of cellular protein PinX1 by 3C. IMPORTANCE: EV71 3C plays an important role in processing viral proteins and interacting with host cells. In this study, we showed that 3C promoted apoptosis through cleaving PinX1, a telomere binding protein, and that this cleavage facilitated EV71 release. Our study demonstrated that PinX1 plays an important role in EV71 release and revealed a novel mechanism that EV71 utilizes to induce apoptosis. This finding is important in understanding EV71-host cell interactions and has potential impact on understanding other enterovirus-host cell interactions.


Subject(s)
Apoptosis , Coxsackievirus Infections/metabolism , Coxsackievirus Infections/virology , Cysteine Endopeptidases/metabolism , Enterovirus A, Human/physiology , Tumor Suppressor Proteins/metabolism , Viral Proteins/metabolism , 3C Viral Proteases , Apoptosis/drug effects , Cell Cycle Proteins , Cell Line , Coxsackievirus Infections/genetics , Etoposide/pharmacology , Humans , Protein Binding , Proteolysis , Telomere-Binding Proteins/genetics , Telomere-Binding Proteins/metabolism , Tumor Suppressor Proteins/genetics , Virus Release
6.
Mol Cancer ; 16(1): 74, 2017 04 04.
Article in English | MEDLINE | ID: mdl-28372542

ABSTRACT

BACKGROUND: The telomerase/telomere interacting protein PinX1 has been suggested as a tumor suppressor. However, the clinical and biological significance of PinX1 in human non-small cell lung cancer (NSCLC) is unclear. METHODS: PinX1 gene/expression pattern and its association with NSCLC patient survival were analyzed in cBioportal Web resource and two cohorts of NSCLC samples. A series of in vivo and in vitro assays were performed to elucidate the function of PinX1 on NSCLC cells proliferation and underlying mechanisms. RESULTS: More frequency of gene PinX1 homozygous deletion and heterozygote deficiency was first retrieved from cBioportal Web resource. Low expression of PinX1 correlated with smoking condition, histological type, T stage, N stage, M stage and TNM stage, and was an independent predictor for overall survival in a learning cohort (n = 93) and a validation cohort (n = 51) of NSCLC patients. Furthermore, knockdown of PinX1 dramatically accelerated NSCLC cell proliferation and G1/S transition, whereas ectopic overexpression of PinX1 substantially inhibited cell viability and cell cycle transition in vitro and in vivo. p15/cyclin D1 pathway and BMP5 might contribute to PinX1-associated cell proliferation and cell cycle transition. CONCLUSION: The cost-effective expression of PinX1 could constitute a novel molecular predictor/marker for NSCLC management.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cyclin D1/metabolism , Cyclin-Dependent Kinase Inhibitor p15/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Signal Transduction , Tumor Suppressor Proteins/genetics , Adult , Aged , Animals , Biomarkers, Tumor , Bone Morphogenetic Protein 5/genetics , Carcinoma, Non-Small-Cell Lung/mortality , Cell Cycle , Cell Cycle Proteins , Cell Line, Tumor , Cell Proliferation , Databases, Nucleic Acid , Disease Models, Animal , Female , Gene Deletion , Gene Silencing , Humans , Lung Neoplasms/mortality , Male , Mice , Middle Aged , Neoplasm Grading , Neoplasm Staging , Prognosis , Xenograft Model Antitumor Assays
7.
Biochim Biophys Acta ; 1839(2): 88-96, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24412852

ABSTRACT

The aberrant activation of telomerase is critical for the initiation and development of human cervical cancer, which is dependent on the activation of human telomerase reverse transcriptase (hTERT). Recently, Pin2/TRF1-interacting protein X1 (PinX1) has been identified as a suppressor of hTERT. It has been found that the telomerase is activated while the level of PinX1 is decreased in cervical cancer. However, the regulatory mechanism of PinX1 in cervical cancer cells remains unclear. In the present study, we demonstrated that the level of PinX1 is regulated by p53, and p53 functions as a transcriptional factor to directly activate the expression of PinX1 in cervical cancer cells. Moreover, we found that HPV16 E6 suppresses the expression of PinX1 via inhibiting p53 transcriptional activity, resulting in the enhancement of telomerase activity. This study not only for the first time shows that PinX1 is a novel target gene of p53 but also suggests that suppression of p53/PinX1 pathway may be a novel mechanism by which HPV16 E6 enhances the telomerase activity in cervical cancer cells.


Subject(s)
Oncogene Proteins, Viral/genetics , Repressor Proteins/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Proteins/genetics , Blotting, Western , Cell Cycle Proteins , Cell Line, Tumor , Cell Proliferation , Female , Gene Expression Regulation, Neoplastic , Humans , Mutation , Oncogene Proteins, Viral/metabolism , Promoter Regions, Genetic/genetics , Protein Binding , RNA Interference , Repressor Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Telomerase/metabolism , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/metabolism , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/metabolism , Uterine Cervical Neoplasms/pathology
8.
Neoplasma ; 62(6): 887-93, 2015.
Article in English | MEDLINE | ID: mdl-26458319

ABSTRACT

Telomeres have emerged as a promising and important factor modulating cellular and organism responses to ionizing radiation (IR). Pin2/TRF1 interacting protein X1 (PinX1) is an intrinsic telomerase inhibitor and a putative tumor suppressor gene in human cancers. The aim of this study is to investigate the role PinX1 in osteosarcoma (OS) radioresistance. A telomerase-positive OS cell line Saos-2 and a telomerase-negative OS cell line U2OS were used. PinX1 shRNA lentiviral vetors were constructed and transfected to cells. PinX1 expression was determined by real-time quantitative PCR (qPCR) and Western blotting. Relative telomere length (RTL) was detected by using qPCR. Flow cytometric analysis was used to detect cell cycle and apoptosis. Radiosensitivity was determined by colony formation assay. Data showed that, PinX1 knockdown resulted in telomere shortening, G1 phase arrest, increased apoptosis and enhanced IR sensitivity both in Saos-2 and U2OS cell lines, regardless of telomerase status. Our study concluded that PinX1 could serve as a novel predictor for radiotherapy response to OS patients, and the pathway of PinX1-mediated telomere stability might represent a new target to improve the radiotherapy effect of OS.

9.
Cancer Cell Int ; 14: 46, 2014.
Article in English | MEDLINE | ID: mdl-24936151

ABSTRACT

BACKGROUND: Pin2/TRF1 binding protein X1 (PinX1) has been identified as an endogenous telomerase inhibitor and a major haploinsufficient tumor suppressor gene. Increasing evidence suggests that reduced expression of PinX1 plays a key role in tumorigenesis. However, the PinX1 expression status and its correlation with the clinicopathological features in prostate cancer (PCa) have not been investigated. METHODS: PinX1 mRNA and protein expression in PCa and adjacent normal prostate tissues were evaluated by real-time quantitative RT-PCR (qRT-PCR) and western blotting. The clinicopathological significance of PinX1 was investigated by immunohistochemistry (IHC) analysis on a PCa tissue microarray (TMA). The cut-off score for positive expression of PinX1 was determined by the receiver operating characteristic (ROC) analysis. The correlation between PinX1 expression and clinicopathological features of PCa was analyzed by Chi-square test. RESULTS: Reduced expression of PinX1 mRNA and protein was observed in the majority of PCa, compared with their paired adjacent normal prostate tissues. When PinX1 positive expression percentage was determined to be above 60% (area under ROC curve = 0.833, P = 0.000), positive expression of PinX1 was observed in 100% (8/8) of normal prostate tissues and 32.5% (13/40) of PCa tissues by IHC. Reduced expression of PinX1 in patients was correlated with advanced clinical stage (χ(2) = 10.230, p = 0.017), high Gleason score (χ(2) = 4.019, p = 0.045), positive regional lymph node metastasis (χ(2) = 10.852, p = 0.004) and distant metastasis (χ(2) = 7.965, p = 0.005). CONCLUSIONS: Our findings suggest that reduced expression of PinX1 is correlates to progressive features in patients with PCa and may serve as a potential marker for diagnosis.

10.
Oncol Rep ; 49(2)2023 Feb.
Article in English | MEDLINE | ID: mdl-36633142

ABSTRACT

Following the publication of the above article, an interested reader drew to the authors' attention that Figs. 3C and E in the paper appeared to contain instances of duplicated data. The authors were able to consult their original data files, and realized that these figures had indeed been assembled incorrectly; subsequently, they requested that a corrigendum be published to take account of the errors that were made during the compilation of these figures. Having investigated this matter in the Editorial Office, however, additional panels of overlapping data were identified, comparing between Figs. 3 and 5; specifically, overlapping data panels were also identified in panels in Figs. 3C, E and F, and 5C and D. The Editor of Oncology Reports has considered the authors' request to publish a corrigendum, but has decided to decline this request on account of the large number of errors that have been identified; rather, the article is to be be retracted from the Journal on the basis of an overall lack of confidence in the presented data. The Editor apologizes to the readership of the Journal for any inconvenience caused. [Oncology Reports 40: 1533­1544, 2018; DOI: 10.3892/or.2018.6570].

11.
Front Pharmacol ; 13: 918665, 2022.
Article in English | MEDLINE | ID: mdl-35847001

ABSTRACT

New drugs or active leads with high efficiency and low toxicity are needed in the treatment of lung cancer. Natural products are an important source of anti-tumor drugs. At present, there are many molecular-targeted anti-tumor drugs derived from natural products or their derivatives for tumor treatment or in clinical trials. Hesperidin is a flavanone isolated from the Rutaceae plant lime Citrus aurantium L. or Citrus sinensis Osbeck. It has been considered to inhibit cancer cell viability in vitro. However, the effect of hesperidin on lung cancer and its underlying mechanism remain unclear. In this study, we found that the pinX1 expression level is closely related to overall survival and plays an important role in regulating lung cancer cell proliferation, migration, invasion, and senescence. More importantly, hesperidin significantly increased pinX1 protein expression, and knockdown pinX1 by its specific siRNA blocked the protective effects of hesperidin. Moreover, we also assessed that hesperidin at 100 mg/kg is safe in vivo. These findings showed that hesperidin is a potential therapeutic candidate for preventing the progression of lung cancer.

12.
J Steroid Biochem Mol Biol ; 210: 105858, 2021 06.
Article in English | MEDLINE | ID: mdl-33647521

ABSTRACT

Coregulators play an important role in prostate cancer (PCa), modulating androgen receptor (AR) action and representing a possible cause of androgen deprivation therapy failure. Pin2-interacting protein X1 (PinX1) is a nucleolar protein described as a steroid hormone receptor coregulator in breast cancer cell lines. In this work, we studied the effect of PinX1 on AR action in PCa. Our results demonstrate that PinX1 acts as an AR coactivator, increasing its transcriptional activity and target gene expression, as well as proliferation, migration and colony formation in PCa cell lines. These effects are observed in the presence and absence of AR agonist and antagonists, suggesting a possible androgen independent pathway for PinX1. We present the first oncogenic roles described for PinX1, acting as a coactivator of the AR.


Subject(s)
Cell Cycle Proteins/metabolism , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Receptors, Androgen/metabolism , Tumor Suppressor Proteins/metabolism , Binding Sites , Cell Cycle Proteins/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Nucleus/metabolism , Cell Proliferation/genetics , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic , Humans , Male , Promoter Regions, Genetic , Prostatic Neoplasms/genetics , Receptors, Androgen/genetics , Tumor Stem Cell Assay , Tumor Suppressor Proteins/genetics
13.
Aging (Albany NY) ; 13(7): 10175-10186, 2021 03 26.
Article in English | MEDLINE | ID: mdl-33819185

ABSTRACT

PIN2/TERF1-interacting telomerase inhibitor 1 (PINX1) is necessary for telomerase reverse transcriptase (TERT) elements to bind at telomeres and non-telomere sites. We aimed to investigate the role of PINX1 and TERT in lipopolysaccharide (LPS)-induced lung injury during acute stage and convalescent phase. Lung injury rat model was induced, and the expression of PINX1 and TERT in serum and lung tissues was examined using RT-qPCR on day 0 (D0), D3, and D14, respectively. The pathologic changes of lung tissues on D3 and D14 were detected using hematoxylin and eosin staining after TERT overexpression, PINX1 overexpression, or PINX1 silencing in lung injury rats. Results revealed that TERT was persistently reduced on D3 and D14, while PINX1 was decreased on D3 but increased on D14. TERT overexpression and PINX1 silencing led to the most serious lung damage, the highest levels of inflammatory factors and apoptosis on D3, while the best recovery was observed on D14. Simultaneously, PINX1 overexpression presented the opposite effects at acute stage and convalescent phase. Co-immunoprecipitation (co-IP) assay verified the connection between PINX1 and TERT. Taken together, these findings demonstrated that regulation of PINX1 expression ameliorates lung injury and alleviates cell senescence during the convalescent phase through affecting the telomerase activity.


Subject(s)
Cellular Senescence/physiology , Gene Expression Regulation , Lung Injury/metabolism , Lung/metabolism , Nuclear Proteins/metabolism , Telomerase/metabolism , Animals , Apoptosis/physiology , Cell Proliferation/physiology , Lipopolysaccharides , Lung Injury/genetics , Nuclear Proteins/genetics , Rats , Rats, Sprague-Dawley
14.
Am J Cancer Res ; 11(11): 5485-5495, 2021.
Article in English | MEDLINE | ID: mdl-34873474

ABSTRACT

Although thyroid cancer is the most prevalent endocrine malignancy, overall patients with thyroid cancer have a good long-term survival. However, a small percentage of patients with progressive thyroid cancer have poor outcomes, and the genetic drivers playing a key role thyroid cancer progression are mostly unknown. Here, we investigated the role of the PINX1 in thyroid cancer progression. Interestingly, PINX1 expression was significantly higher in ATC than in PTC in both patients and cell lines. When PINX1 was knockdown in ATC cells, cell proliferation rates, colony formation capacity, and cell cycle progression were significantly reduced. Furthermore, cell motility and the expression of EMT drivers were reduced by PINX1 downregulation. In contrast, the overexpression of PINX1 in PTC cells significantly increased those phenotypes of tumor progression, which demonstrates that PINX1 could promote tumor proliferation and malignant transformation in both PTC and ATC cells. To further understand whether PINX1 is also involved in the progression of PTC to ATC, we examined PI3K/AKT, MAPK, and ß-catenin signaling activation after PINX1 modulation. Decreased PINX1 expression reduced the levels of p-AKT, p-ERK, p-p38, and ß-catenin in ATC cells, but the increase of PINX1 expression upregulated the phosphorylation of AKT, ERK, and p38 and the levels of ß-catenin in PTC cells. These results were all confirmed in xenograft mouse tumors. Our findings suggest that PINX1 regulates thyroid cancer progression by promoting cell proliferation, EMT, and signaling activation, and support the hypothesis that PINX1 could be a prognostic marker and a therapeutic target of thyroid cancer.

15.
J Am Heart Assoc ; 9(6): e010240, 2020 03 17.
Article in English | MEDLINE | ID: mdl-32157956

ABSTRACT

Background Pin2/TRF1-interacting protein, PinX1, was previously identified as a tumor suppressor. Here, we discovered a novel transcript variant of mPinX1 (mouse PinX1), mPinX1t (mouse PinX1t), in embryonic stem cells (ESCs). The aims of this investigation were (1) to detect the presence of mPinX1 and mPinX1t in ESCs and their differentiation derivatives; (2) to investigate the role of mPinX1 and mPinX1t on regulating the characteristics of undifferentiated ESCs and the cardiac differentiation of ESCs; (3) to elucidate the molecular mechanisms of how mPinX1 and mPinX1t regulate the cardiac differentiation of ESCs. Methods and Results By 5' rapid amplification of cDNA ends, 3' rapid amplification of cDNA ends, and polysome fractionation followed by reverse transcription-polymerase chain reaction, mPinX1t transcript was confirmed to be an intact mRNA that is actively translated. Western blot confirmed the existence of mPinX1t protein. Overexpression or knockdown of mPinX1 (both decreased mPinX1t expression) both decreased while overexpression of mPinX1t increased the cardiac differentiation of ESCs. Although both mPinX1 and mPinX1t proteins were found to bind to cardiac transcription factor mRNAs, only mPinX1t protein but not mPinX1 protein was found to bind to nucleoporin 133 protein, a nuclear pore complex component. In addition, mPinX1t-containing cells were found to have a higher cytosol-to-nucleus ratio of cardiac transcription factor mRNAs when compared with that in the control cells. Our data suggested that mPinX1t may positively regulate cardiac differentiation by enhancing export of cardiac transcription factor mRNAs through interacting with nucleoporin 133. Conclusions We discovered a novel transcript variant of mPinX1, the mPinX1t, which positively regulates the cardiac differentiation of ESCs.


Subject(s)
Cell Cycle Proteins/metabolism , Cell Differentiation , Mouse Embryonic Stem Cells/metabolism , Myocytes, Cardiac/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Cell Cycle Proteins/genetics , Cell Line , Cell Lineage , Gene Expression Regulation, Developmental , Mice , Minor Histocompatibility Antigens/genetics , Minor Histocompatibility Antigens/metabolism , Morphogenesis , Nuclear Pore Complex Proteins/genetics , Nuclear Pore Complex Proteins/metabolism , Protein Isoforms , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription, Genetic , Tumor Suppressor Proteins/genetics
16.
J Exp Clin Cancer Res ; 39(1): 31, 2020 Feb 07.
Article in English | MEDLINE | ID: mdl-32028978

ABSTRACT

BACKGROUND: Recurrence and distant metastasis are still the main factors leading to treatment failure for malignant tumors including nasopharyngeal carcinoma (NPC). Therefore, elucidating the molecular mechanisms underlying nasopharyngeal carcinoma metastasis is of great clinical significance for targeted gene therapy and prognostic evaluation. PinX1, a tumor suppressor gene, was previously demonstrated to be a powerful tool for targeting telomerase in order to resist malignant tumor proliferation and migration. The aim of this study was to explore the mechanism through which PinX1 regulates epithelial-mesenchymal transition (EMT) and tumor metastasis in NPC and investigate its clinical significance and biological role with respect to disease progression. METHODS: Cell Counting Kit-8 (CCK8), Transwell assays, Colony formation analysis and Xenograft tumorigenicity assay were used to measure the nasopharyngeal CD133+ cancer stem cell proliferation, migration, and invasion abilities. Reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and western blot assays were conducted to investigate the underlying mechanism that PinX1 inhibits cell proliferation, migration, and invasion via regulating EMT in nasopharyngeal CD133+ CSCs. RESULTS: We found that the overexpression of PinX1 and P53 inhibited cell proliferation, migration, and invasion, but that the inhibition of miR-200b blocked these effects, in nasopharyngeal CD133+ cancer stem cells (CSCs). Mechanistic investigations elucidated that PinX1 inhibits cell proliferation, migration, and invasion by regulating the P53/miR-200b-mediated transcriptional suppression of Snail1, Twist1, and Zeb1, consequently inhibiting EMT in nasopharyngeal CD133+ CSCs. CONCLUSIONS: Our findings indicate that PinX1 inhibits cell proliferation, migration, and invasion via P53/miR-200b-regulated EMT in the malignant progression of human NPC, which might suggest novel clinical implications for disease treatment.


Subject(s)
Biomarkers, Tumor , Cell Cycle Proteins/genetics , Nasopharyngeal Neoplasms/genetics , Neoplastic Stem Cells/metabolism , Tumor Suppressor Proteins/genetics , Animals , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Disease Models, Animal , Disease Progression , Epithelial-Mesenchymal Transition/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Mice , MicroRNAs/genetics , Nasopharyngeal Neoplasms/drug therapy , Nasopharyngeal Neoplasms/pathology , Neoplastic Stem Cells/pathology , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Proteins/metabolism
17.
Cell Biosci ; 9: 47, 2019.
Article in English | MEDLINE | ID: mdl-31210926

ABSTRACT

BACKGROUND: Telomere maintenance is an important factor in tumorigenesis. PinX1 is a potent telomerase regulator which also involves in telomerase loading to telomeres. Nucleophosmin (NPM) can partially attenuate PinX1 inhibition of telomerase activity and NPM loading to hTERT requires PinX1. However, the role of the PinX1/NPM interaction in telomerase activity is not fully understood. METHOD: The long-term effects of PinX1 and NPM down-regulation on telomere length were investigated by TRF assay. The localization of the PinX1/NPM association and the NPM/PinX1/hTERT complex formation were examined by immunofluorescence studies. RESULTS: Concurrent long-term down-regulation of PinX1 and NPM led to a substantial decrease in telomere length. The interaction with PinX1 was crucial in NPM localization in the nucleolus during the S phase. PinX1 and NPM associated throughout S phase and the NPM/PinX1/hTERT complex formation peaked during the early-S phase. The PinX1/NPM interaction was shown to localize away from Cajal Bodies at the start of S phase. CONCLUSION: PinX1/NPM interaction is important in telomerase regulation during catalysis. NPM is recruited to hTERT by PinX1 and is required in the proposed telomerase modulating unit to activate telomerase when telomere extension occurs during S phase.

18.
Hum Pathol ; 81: 176-183, 2018 11.
Article in English | MEDLINE | ID: mdl-30026037

ABSTRACT

PIN2/TERF1 interacting telomerase inhibitor 1 (PINX1) is a telomerase inhibitor located on human chromosome 8p23 and also acts as a tumor suppressor in several types of cancers, including breast, gastric, ovarian, and bladder cancer. However, the role of PINX1 expression in papillary thyroid carcinoma (PTC) has not been defined. Therefore, we investigated the role of PINX1 expression in PTC by analyzing the correlation between PINX1 expression and various clinicopathological factors. Immunohistochemistry for PINX1 was performed using a tissue microarray of samples taken from the 160 patients with PTC. We also assessed mRNA and protein expression for PINX1 via quantitative real-time polymerase chain reaction and immunohistochemical analysis. Positive staining for PINX1 was found in 16.3% of PTC cases. PINX1 expression was significantly associated with tumor size, lymph node metastasis, telomerase reverse transcriptase, promoter mutation and recurrence. PINX1 mRNA expression was more pronounced in the recurrent group than in the nonrecurrent group. In addition, results of the binary logistic regression model showed that PINX1 protein expression had a significant influence on recurrence. We concluded that PINX1 expression was associated with several clinicopathological factors and had a significant influence on recurrence in patients with PTC. Therefore, PINX1 expression could be a useful prognostic marker in PTC patients.


Subject(s)
Biomarkers, Tumor/analysis , Thyroid Cancer, Papillary/chemistry , Thyroid Neoplasms/chemistry , Tumor Suppressor Proteins/analysis , Adult , Aged , Biomarkers, Tumor/genetics , Cell Cycle Proteins , Female , Humans , Immunohistochemistry , Lymph Node Excision , Lymphatic Metastasis , Male , Middle Aged , Neoplasm Recurrence, Local , Real-Time Polymerase Chain Reaction , Retrospective Studies , Reverse Transcriptase Polymerase Chain Reaction , Risk Factors , Thyroid Cancer, Papillary/genetics , Thyroid Cancer, Papillary/secondary , Thyroid Cancer, Papillary/therapy , Thyroid Neoplasms/genetics , Thyroid Neoplasms/pathology , Thyroid Neoplasms/therapy , Thyroidectomy , Tissue Array Analysis , Treatment Outcome , Tumor Burden , Tumor Suppressor Proteins/genetics , Young Adult
19.
Oncotarget ; 8(40): 67241-67253, 2017 Sep 15.
Article in English | MEDLINE | ID: mdl-28978030

ABSTRACT

Pin2/TRF1-interacting telomere inhibitor 1 (PinX1) was originally identified as a telomerase inhibitor, involved in maintaining telomerase activity, telomere length, and chromosomal stability. However, research has shown that PinX1 can have opposing molecular status in its expression patterns in several other tumor types. We thus investigated the genetic profile and biological implication of PinX1 in several human cancers using the cBioportal database. Our results showed that PinX1 deletion accounted for the most alterations, with the frequency of its deletion regularly occurring in pathological types of carcinosarcoma and adenocarcinoma. We found few instances of PinX1 gene mutations and 3D structural analysis demonstrated that these mutation sites were always located within telomerase inhibitor domains. Furthermore, our analysis of several human cancers from the cBioportal database revealed more frequent PinX1 homozygous depletion and PinX1 heterozygous deficiency, but both more infrequent PinX1 gain and rare instances of PinX1 amplification. The status of PinX1 genetic alterations was correlated with prognosis and may be tumor-type specific. As such, its biological function in tumorigenesis and later prognosis is complicated and may involve co-worked with NEIL2, R3HCC1, POLR3D, GTF2E2, and INTS10. In addition, we observed that PinX1 interacts with TERT, DKC1, PTGES3, and HSP90AA1. PinX1 mRNA expression was decreased in most selected cancer tissues, which could promote tumor growth and enhance tumorigenicity. Collectively, our data reveal PinX1 expression patterns and potential mechanisms in various human cancers. Further work will be needed to comprehensively examine its role in tumor genesis and progression.

20.
Oncotarget ; 7(40): 66267-66275, 2016 Oct 04.
Article in English | MEDLINE | ID: mdl-27556185

ABSTRACT

PIN2/TRF1-interacting telomerase inhibitor 1 (PinX1) is a novel cloned gene located at human chromosome 8p23, playing a vital role in maintaining telomeres length and chromosome stability. It has been demonstrated to be involved in tumor genesis and progression in most malignancies. However, some researches showed opposing molecular status of PinX1 gene and its expression patterns in several other types of tumors. The pathogenic mechanism of PinX1 expression in human malignancy is not yet clear. Moreover, emerging evidence suggest that PinX1 (especially its TID domain) might be a potential new target cancer treatment. Therefore, PinX1 may be a new potential diagnostic biomarker and therapeutic target for human cancers, and may play different roles in different human cancers. The functions and the mechanisms of PinX1 in various human cancers remain unclear, suggesting the necessity of further extensive works of its role in tumor genesis and progression.


Subject(s)
Gene Expression Regulation, Neoplastic , Neoplasms/metabolism , Telomerase/antagonists & inhibitors , Tumor Suppressor Proteins/chemistry , Tumor Suppressor Proteins/metabolism , Cell Cycle Proteins , Humans , Neoplasms/diagnosis , Neoplasms/genetics , Neoplasms/therapy , Tumor Suppressor Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL