Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 82
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Mol Cell Proteomics ; 20: 100115, 2021.
Article in English | MEDLINE | ID: mdl-34129943

ABSTRACT

The occurrence and prevalence of colorectal cancer (CRC) is closely associated with age. More than 90% of patients with CRC are diagnosed after 50 years of age. However, CRC incidence of young individuals has been increasing since 1990s, whereas the overall CRC frequency is declining. Distinct overall survival rates between young and aged patients with CRC have been established. Tremendous efforts have been made to clarify the underlying mechanisms of age-dependent clinical differences, but it still remains elusive. Here, we performed proteomic profiling of 50 patients with CRC and revealed proteomic signatures of CRC across age groups. Gene set enrichment analysis showed that distinct age-dependent clinical outcomes might mainly attribute to varied MYC targets V1/V2, E2F targets and G2M checkpoint gene sets, which were associated with cancer cell proliferation, cell apoptosis, tumor growth, and tumor metastasis. Multiple linear regression analysis revealed a large number of functional proteins, such as NOP2, CSE1L, NHP2, NOC2L and CDK1, with adjusted expression significantly correlated with age (p < 0.05). Among them, NHP2 is a core component of the telomerase complex associated with age. High NHP2 expression predicted poor overall survival, with a more significant correlation in aged patients with CRC. Knockdown of NHP2 significantly suppressed cancer cell proliferation. In addition, we revealed some age-related potential clinically actionable targets, such as PSEN1, TSPO, and CDK1, which might be more suitable for patients with late-onset CRC. Collectively, this study identifies age-associated proteomic signatures and potential therapeutic targets of CRC and may help make a precise decision on CRC treatment.


Subject(s)
Aging/metabolism , Colorectal Neoplasms/metabolism , Adult , Aging/genetics , CDC2 Protein Kinase/metabolism , Cell Line, Tumor , Cellular Apoptosis Susceptibility Protein/metabolism , Colorectal Neoplasms/genetics , Female , Humans , Male , Middle Aged , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Presenilin-1/metabolism , Proteomics , Receptors, GABA/metabolism , Repressor Proteins/metabolism , Ribonucleoproteins, Small Nuclear/genetics , Ribonucleoproteins, Small Nuclear/metabolism , tRNA Methyltransferases/genetics , tRNA Methyltransferases/metabolism
2.
J Biol Chem ; 297(1): 100803, 2021 07.
Article in English | MEDLINE | ID: mdl-34022224

ABSTRACT

The transcriptional coactivator with PDZ-binding motif (TAZ) (WWTR1) induces epithelial-mesenchymal transition and enhances drug resistance in multiple cancers. TAZ has been shown to interact with transcription factors in the nucleus, but when phosphorylated, translocates to the cytoplasm and is degraded through proteasomes. Here, we identified a compound TAZ inhibitor 4 (TI-4) that shifted TAZ localization to the cytoplasm independently of its phosphorylation. We used affinity beads to ascertain a putative target of TI-4, chromosomal segregation 1 like (CSE1L), which is known to be involved in the recycling of importin α and as a biomarker of cancer malignancy. We found that TI-4 suppressed TAZ-mediated transcription in a CSE1L-dependent manner. CSE1L overexpression increased nuclear levels of TAZ, whereas CSE1L silencing delayed its nuclear import. We also found via the in vitro coimmunoprecipitation experiments that TI-4 strengthened the interaction between CSE1L and importin α5 and blocked the binding of importin α5 to TAZ. WWTR1 silencing attenuated CSE1L-promoted colony formation, motility, and invasiveness of human lung cancer and glioblastoma cells. Conversely, CSE1L silencing blocked TAZ-promoted colony formation, motility, and invasiveness in human lung cancer and glioblastoma cells. In human cancer tissues, the expression level of CSE1L was found to correlate with nuclear levels of TAZ. These findings support that CSE1L promotes the nuclear accumulation of TAZ and enhances malignancy in cancer cells.


Subject(s)
Cell Nucleus/metabolism , Cellular Apoptosis Susceptibility Protein/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Trans-Activators/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation , Gene Expression Regulation, Neoplastic , Gene Silencing , Green Fluorescent Proteins/metabolism , Humans , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Models, Biological , Neoplasm Invasiveness , Neoplasms/genetics , Phosphorylation , Photobleaching , Protein Binding , Protein Transport , Subcellular Fractions/metabolism , Transcriptional Coactivator with PDZ-Binding Motif Proteins , Tumor Stem Cell Assay , alpha Karyopherins/metabolism
3.
BMC Cancer ; 21(1): 175, 2021 Feb 18.
Article in English | MEDLINE | ID: mdl-33602168

ABSTRACT

BACKGROUND: Apoptosis-related genes(Args)play an essential role in the occurrence and progression of hepatocellular carcinoma(HCC). However, few studies have focused on the prognostic significance of Args in HCC. In the study, we aim to explore an efficient prognostic model of Asian HCC patients based on the Args. METHODS: We downloaded mRNA expression profiles and corresponding clinical data of Asian HCC patients from The Cancer Genome Atlas (TCGA) and International Cancer Genome Consortium (ICGC) databases. The Args were collected from Deathbase, a database related to cell death, combined with the research results of GeneCards、National Center for Biotechnology Information (NCBI) databases and a lot of literature. We used Wilcoxon-test and univariate Cox analysis to screen the differential expressed genes (DEGs) and the prognostic related genes (PRGs) of HCC. The intersection genes of DEGs and PGGs were seen as crucial Args of HCC. The prognostic model of Asian HCC patients was constructed by least absolute shrinkage and selection operator (lasso)- proportional hazards model (Cox) regression analysis. Kaplan-Meier curve, Principal Component Analysis (PCA) analysis, t-distributed Stochastic Neighbor Embedding (t-SNE) analysis, risk score curve, receiver operating characteristic (ROC) curve, and the HCC data of ICGC database and the data of Asian HCC patients of Kaplan-Meier plotter database were used to verify the model. RESULTS: A total of 20 of 56 Args were differentially expressed between HCC and adjacent normal tissues (p < 0.05). Univariate Cox regression analysis showed that 10 of 56 Args were associated with survival time and survival status of HCC patients (p < 0.05). There are seven overlapping genes of these 20 and 10 genes, including BAK1, BAX, BNIP3, CRADD, CSE1L, FAS, and SH3GLB1. Through Lasso-Cox analysis, an HCC prognostic model composed of BAK1, BNIP3, CSE1L, and FAS was constructed. Kaplan-Meier curve, PCA, t-SNE analysis, risk score curve, ROC curve, and secondary verification of ICGC database and Kaplan-Meier plotter database all support the reliability of the model. CONCLUSIONS: Lasso-Cox regression analysis identified a 4-gene prognostic model, which integrates clinical and gene expression and has a good effect. The expression of Args is related to the prognosis of HCC patients, but the specific mechanism remains to be further verified.


Subject(s)
Apoptosis Regulatory Proteins/genetics , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/genetics , Apoptosis/genetics , Apoptosis Regulatory Proteins/metabolism , Asian People/genetics , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Carcinoma, Hepatocellular/metabolism , Cellular Apoptosis Susceptibility Protein/genetics , Cellular Apoptosis Susceptibility Protein/metabolism , Computational Biology/methods , Female , Gene Expression Profiling/methods , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Middle Aged , Prognosis , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , bcl-2 Homologous Antagonist-Killer Protein/genetics , bcl-2 Homologous Antagonist-Killer Protein/metabolism , fas Receptor/genetics , fas Receptor/metabolism
4.
Proc Natl Acad Sci U S A ; 115(17): E4013-E4022, 2018 04 24.
Article in English | MEDLINE | ID: mdl-29636421

ABSTRACT

Epigenetic silencing can be mediated by various mechanisms, and many regulators remain to be identified. Here, we report a genome-wide siRNA screening to identify regulators essential for maintaining gene repression of a CMV promoter silenced by DNA methylation. We identified CSE1L (chromosome segregation 1 like) as an essential factor for the silencing of the reporter gene and many endogenous methylated genes. CSE1L depletion did not cause DNA demethylation. On the other hand, the methylated genes derepressed by CSE1L depletion largely overlapped with methylated genes that were also reactivated by treatment with histone deacetylase inhibitors (HDACi). Gene silencing defects observed upon CSE1L depletion were linked to its nuclear import function for certain protein cargos because depletion of other factors involved in the same nuclear import pathway, including KPNAs and KPNB1 proteins, displayed similar derepression profiles at the genome-wide level. Therefore, CSE1L appears to be critical for the nuclear import of certain key repressive proteins. Indeed, NOVA1, HDAC1, HDAC2, and HDAC8, genes known as silencing factors, became delocalized into cytosol upon CSE1L depletion. This study suggests that the cargo specificity of the protein nuclear import system may impact the selectivity of gene silencing.


Subject(s)
Cell Nucleus/metabolism , Cellular Apoptosis Susceptibility Protein/metabolism , Gene Silencing/physiology , Active Transport, Cell Nucleus/drug effects , Active Transport, Cell Nucleus/genetics , Cell Line , Cell Nucleus/genetics , Cellular Apoptosis Susceptibility Protein/genetics , DNA Methylation/drug effects , Gene Silencing/drug effects , Genome-Wide Association Study , Histone Deacetylase 1/genetics , Histone Deacetylase 1/metabolism , Histone Deacetylase 2/genetics , Histone Deacetylase 2/metabolism , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Humans , Neuro-Oncological Ventral Antigen , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , beta Karyopherins/genetics , beta Karyopherins/metabolism
5.
J Cell Physiol ; 235(3): 2071-2079, 2020 03.
Article in English | MEDLINE | ID: mdl-31347172

ABSTRACT

Human chromosomal segregation 1-like (CSE1L) gene functions as a key molecular mediator in cellular proliferation, invasion, and apoptosis. The association of CSE1L with tumor progression has been reported in diverse human cancers. A greater understanding of CSE1L molecular mechanism is beneficial for cancer treatment. In the current study, we show that CSE1L was highly expressed in gastric cancer (GC) cell lines. CSE1L silence promoted apoptosis and inhibited cell proliferation and invasion. Overexpression of glycoprotein nonmetastatic melanoma protein B (GPNMB) reversed the anticancer effect of CSE1L inhibition. CSE1L inhibition decreased GPNMB by microphthalmia-associated transcription factor (MITF). Moreover, GPNMB regulates the phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt)/mammalian target of rapamycin (mTOR) and mitogen-activated protein kinase kinase (MEK)/extracellular signal-regulated kinase (ERK) signaling pathway. Taken together, our study revealed that CSE1L inhibition decreased MITF and suppressed GPNMB expression, thereby activating the PI3K/Akt/mTOR and MEK/ERK signaling pathway, ultimately inhibiting the tumor growth and metastasis in GC.


Subject(s)
Cell Proliferation/physiology , Cellular Apoptosis Susceptibility Protein/metabolism , Membrane Glycoproteins/metabolism , Microphthalmia-Associated Transcription Factor/metabolism , Neoplasm Metastasis/pathology , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Apoptosis/physiology , Cell Line , Cell Line, Tumor , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , MAP Kinase Signaling System/physiology , Mitogen-Activated Protein Kinase Kinases/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/metabolism
6.
Am J Pathol ; 188(7): 1597-1607, 2018 07.
Article in English | MEDLINE | ID: mdl-29630856

ABSTRACT

Epithelial-to-mesenchymal transition (EMT) is postulated to be a prerequisite for the establishment of endometriosis (EMS), a common reproductive disorder in women. Our previous studies have demonstrated the elevated expression of transmembrane glycoprotein CD147 and its prosurvival effect on abnormal cells in endometriosis. Intriguingly, CD147 is known to promote EMT in cancers. However, the involvement of CD147 in EMT during the establishment of endometriosis remains incompletely understood. We found that CD147 promotes EMT in human endometrial adenocarcinoma cell line Ishikawa. We identified a novel CD147-interacting partner, cellular apoptosis susceptibility protein (CAS), which stabilized the interaction between E-cadherin (E-cad) and ß-catenin (ß-cat) by forming the CAS/E-cad/ß-cat complex. Down-regulation of CAS led to the release and nuclear translocation of ß-cat from E-cad, resulting in the overexpression of the EMT-promoting gene SNAIL. Interestingly, overexpression of CD147 impaired the interaction between CAS and E-cad and triggered the release of ß-cat from the CAS/E-cad/ß-cat complex, which in turn led to EMT. Furthermore, CAS was down-regulated in EMS, with elevated levels of CD147 and nuclear ß-cat. These findings suggest a previously undefined role of CAS in regulating EMT and reveal the involvement of a CD147-induced EMT signaling pathway in pathogenic progression of EMS.


Subject(s)
Antigens, CD/metabolism , Basigin/metabolism , Cadherins/metabolism , Cellular Apoptosis Susceptibility Protein/metabolism , Endometrial Neoplasms/pathology , Endometriosis/pathology , Epithelial-Mesenchymal Transition , beta Catenin/metabolism , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Adult , Antigens, CD/genetics , Basigin/genetics , Cadherins/genetics , Case-Control Studies , Cell Membrane , Cellular Apoptosis Susceptibility Protein/genetics , Endometrial Neoplasms/genetics , Endometrial Neoplasms/metabolism , Endometriosis/genetics , Endometriosis/metabolism , Female , Gene Expression Regulation, Neoplastic , Humans , Middle Aged , Protein Interaction Domains and Motifs , Signal Transduction , Tumor Cells, Cultured , beta Catenin/genetics
7.
J Biol Chem ; 291(5): 2379-88, 2016 Jan 29.
Article in English | MEDLINE | ID: mdl-26668314

ABSTRACT

A signature event during the cell intrinsic apoptotic pathway is mitochondrial outer membrane permeabilization, leading to formation of the apoptosome, a caspase activation complex. The cellular apoptosis susceptibility protein (CAS) can facilitate apoptosome assembly by stimulating nucleotide exchange on Apaf-1 following binding of cytochrome c. We report here that CAS expression itself is up-regulated during tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-induced apoptosis, and knockdown of CAS renders cells resistant to TRAIL. We find that TRAIL induces up-regulation of CAS in a posttranscriptional, caspase-8-dependent manner through degradation of cIAP1, an E3 ligase that targets CAS for ubiquitin-dependent proteasomal degradation. We identified a novel signaling pathway whereby caspase-8 engages a feedforward cascade that leads to CAS up-regulation and amplifies the apoptotic signal. Furthermore, in silico analysis revealed that expression of CAS is up-regulated at both the mRNA and DNA levels in human breast tumors, consistent with its role in promoting cell proliferation. Overexpression of various oncogenes led to CAS up-regulation in non-transformed cells. Intriguingly, oncogene-induced CAS up-regulation also resulted in greater susceptibility to TRAIL-induced cell death, consistent with its proapoptotic function. These findings suggest that CAS plays contrasting roles in proliferation and apoptosis and that overexpression of CAS in tumors could serve as a potential biomarker to guide therapeutic choices.


Subject(s)
Apoptosis , Cell Proliferation , Cellular Apoptosis Susceptibility Protein/metabolism , Gene Expression Regulation, Neoplastic , TNF-Related Apoptosis-Inducing Ligand/metabolism , Apoptosomes , Apoptotic Protease-Activating Factor 1/metabolism , Caspase 8/metabolism , Cell Line, Tumor , Cell Survival , HEK293 Cells , HT29 Cells , Humans , Inhibitor of Apoptosis Proteins/metabolism , Mitochondrial Membranes/metabolism , Permeability , RNA, Small Interfering/metabolism , Signal Transduction , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/metabolism , Up-Regulation
8.
Am J Pathol ; 186(10): 2761-8, 2016 10.
Article in English | MEDLINE | ID: mdl-27521996

ABSTRACT

Human cellular apoptosis susceptibility (chromosomal segregation 1-like, CSE1L) gene plays a role in nuclear-to-cytoplasm transport and chromosome segregation during mitosis, cellular proliferation, and apoptosis. CSE1L is involved in colon carcinogenesis. CSE1L gene expression was assessed with three data sets using Affymetrix U133 + gene chips on normal human colonic mucosa (NR), adenomas (ADs), and colorectal carcinoma (CRC). CSE1L protein expression in CRC, AD, and NR from the same patients was measured by immunohistochemistry using a tissue microarray. We evaluated CSE1L expression in CRC cells (HCT116, SW480, and HT29) and its biological functions. CSE1L mRNA was significantly increased in all AD and CRC compared with NR (P < 0.001 and P = 0.02, respectivly). We observed a change in CSE1L staining intensity and cellular localization by immunohistochemistry. CSE1L was significantly increased during the transition from AD to CRC when compared with NR in a CRC tissue microarray (P = 0.01 and P < 0.001). HCT116, SW480, and HT29 cells also expressed CSE1L protein. CSE1L knockdown by shRNA inhibited protein, resulting in decreased cell proliferation, reduced colony formation in soft agar, and induction of apoptosis. CSE1L protein is expressed early and across all stages of CRC development. shRNA knockdown of CSE1L was associated with inhibition of tumorigenesis in CRC cells. CSE1L may represent a potential target for treatment of CRC.


Subject(s)
Adenoma/pathology , Carcinogenesis/genetics , Cellular Apoptosis Susceptibility Protein/genetics , Colorectal Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Adenoma/genetics , Adenoma/metabolism , Adult , Aged , Aged, 80 and over , Apoptosis/genetics , Cell Line, Tumor , Cell Nucleus/metabolism , Cell Proliferation , Cellular Apoptosis Susceptibility Protein/metabolism , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Cytoplasm/metabolism , Epithelial Cells/metabolism , Epithelial Cells/pathology , Female , Humans , Male , Middle Aged , Protein Transport , Tissue Array Analysis , Young Adult
9.
Genes Cells ; 20(9): 681-94, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26123175

ABSTRACT

Homologous recombinational repair (HR) is one of the major repair systems for DNA double-strand breaks. RAD51 is a key molecule in HR, and the RAD51 concentration in the cell nucleus increases after DNA damage induction. However, the mechanism that regulates the intracellular distribution of RAD51 is still unclear. Here, we show that hCAS/CSE1L associates with RAD51 in human cells. We found that hCAS/CSE1L negatively regulates the nuclear protein level of RAD51 under normal conditions. hCAS/CSE1L is also required to repress the DNA damage-induced focus formation of RAD51. Moreover, we show that hCAS/CSE1L plays roles in the regulation of the HR activity and in chromosome stability. These findings suggest that hCAS/CSE1L is responsible for controlling the HR activity by directly interacting with RAD51.


Subject(s)
Cellular Apoptosis Susceptibility Protein/metabolism , Homologous Recombination , Rad51 Recombinase/metabolism , Recombinational DNA Repair , Cell Line, Tumor , Cell Nucleus/metabolism , Chromosome Aberrations , DNA Breaks, Double-Stranded , Humans
10.
Mol Carcinog ; 55(11): 1542-1552, 2016 Nov.
Article in English | MEDLINE | ID: mdl-26331446

ABSTRACT

The Ras/ERK (extracellular signal-regulated protein kinase) and cAMP/PKA (protein kinase A) pathways are essential for the transcriptional activities of CREB (cAMP response element binding protein) and MITF (microphthalmia-associated transcription factor) in melanogenesis and the progression of melanoma. However, the interaction between Ras/ERK and cAMP/PKA pathways in the melanogenesis and progression of melanoma is not fully known. Here, we report that CSE1L (chromosome segregation 1-like protein) regulates cAMP/PKA-induced CREB and MITF expressions as well as Ras-induced ERK1/2 phosphorylation. IBMX, a cAMP/PKA activator, treatment induced CSE1L phosphorylation and augmented Ras-induced ERK1/2 phosphorylation. CSE1L knockdown by CSE1L shRNA expression vectors inhibited Ras-induced ERK1/2 phosphorylation and melanogenesis in melanoma cells. CSE1L overexpression increased phospho-CREB expression; CSE1L knockdown also inhibited Ras-induced phospho-CREB, MITF, and tyrosinase expressions, regardless of the presence of IBMX. This study identifies CSE1L links and controls the Ras/ERK and cAMP/PKA pathways in the melanogenesis of melanoma cells. Melanomas frequently develop drug resistance via paradoxical activation of Ras/Raf/MEK/ERK or alternatively activated Ras/ERK and cAMP/PKA pathways. Thus CSE1L may be a potential target for treating melanomas that harbor Ras mutations or are resistant to drugs targeting Raf/MEK/ERK. © 2015 Wiley Periodicals, Inc.


Subject(s)
Cellular Apoptosis Susceptibility Protein/metabolism , MAP Kinase Signaling System , Melanoma/metabolism , Skin Neoplasms/metabolism , 1-Methyl-3-isobutylxanthine/pharmacology , Aged , Aged, 80 and over , Animals , Cell Line, Tumor , Female , Humans , Male , Melanoma/pathology , Mice , Middle Aged , Mutation , Neoplasm Transplantation , Phosphorylation , Skin Neoplasms/pathology , ras Proteins/metabolism
11.
Tumour Biol ; 37(10): 13077-13090, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27596143

ABSTRACT

CSE1L (chromosome segregation 1-like protein), also named as CAS (cellular apoptosis susceptibility protein), is highly expressed in most cancer types. CSE1L/CAS is a multiple functional protein that plays roles in apoptosis, cell survival, chromosome assembly, nucleocytoplasmic transport, microvesicle formation, and cancer metastasis; some of the functions are explicitly correlated. CSE1L is also a cancer serum biomarker. The phosphorylation of CAS is regulated by the extracellular signal-regulated kinase (ERK). The RAS/RAF/MAPK/ERK signaling pathways are the essential targets of most targeted cancer drugs, thus serum phosphorylated CSE1L may be a potential biomarker for monitoring drug resistance in targeted therapy. CSE1L can regulate Ras-induced ERK phosphorylation. CSE1L also regulates the expression and phosphorylation of CREB (cAMP response element binding protein) and MITF (microphthalmia-associated transcription factor) and is thus involved in the melanogenesis and progression of melanoma. CAS is an exosome/microvesicle membrane protein. Tumor cells consistently secrete microvesicles and tumor-derived microvesicles may be accumulated around tumors. Therefore, microvesicle membrane CSE1L may be a potential target for the development of high-efficacy antibody-drug conjugates (ADCs) for cancer therapy. This review will focus on CSE1L expression in cancers, its relationship to Ras/ERK and cAMP/PKA signaling pathways in melanoma development, its potential for the development of ADCs and tumor imaging reagents, and secretory phosphorylated CSE1L for monitoring the emergence of drug resistance in targeted cancer therapy.


Subject(s)
Antineoplastic Agents/therapeutic use , Biomarkers/metabolism , Cellular Apoptosis Susceptibility Protein/metabolism , Molecular Targeted Therapy , Neoplasms/diagnosis , Neoplasms/drug therapy , Humans , Neoplasms/metabolism , Neoplasms/pathology , Signal Transduction/drug effects
12.
Mol Cell ; 30(2): 239-47, 2008 Apr 25.
Article in English | MEDLINE | ID: mdl-18439902

ABSTRACT

During apoptosis, cytochrome c is released from mitochondria to the cytosol, where it binds Apaf-1. The Apaf-1/cytochrome c complex then oligomerizes either into heptameric caspase-9-activating apoptosome, which subsequently activates caspase-3 and caspase-7, or bigger inactive aggregates, depending on the availability of nucleotide dATP/ATP. A tumor suppressor protein, PHAPI, enhances caspase-9 activation by promoting apoptosome formation through an unknown mechanism. We report here the identification of cellular apoptosis susceptibility protein (CAS) and heat shock protein 70 (Hsp70) as mediators of PHAPI activity. PHAPI, CAS, and Hsp70 function together to accelerate nucleotide exchange on Apaf-1 and prevent inactive Apaf-1/cytochrome c aggregation. CAS expression is induced by multiple apoptotic stimuli including UV irradiation. Knockdown of CAS by RNA interference (RNAi) in cells attenuates apoptosis induced by UV light and causes endogenous Apaf-1 to form aggregates. These studies indicated that PHAPI, CAS, and Hsp70 play an important regulatory role during apoptosis.


Subject(s)
Apoptosis , Apoptosomes/metabolism , Apoptotic Protease-Activating Factor 1/metabolism , Cellular Apoptosis Susceptibility Protein/metabolism , HSP70 Heat-Shock Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Apoptotic Protease-Activating Factor 1/antagonists & inhibitors , Caspase 9/metabolism , Caspase Inhibitors , Cellular Apoptosis Susceptibility Protein/antagonists & inhibitors , Cellular Apoptosis Susceptibility Protein/genetics , Enzyme Activation , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins/genetics , Nuclear Proteins , Nucleotides/metabolism , RNA Interference , RNA-Binding Proteins
13.
Proc Natl Acad Sci U S A ; 110(17): E1584-93, 2013 Apr 23.
Article in English | MEDLINE | ID: mdl-23569239

ABSTRACT

Nuclear pore complexes (NPCs) mediate the exchange of macromolecules between the cytoplasm and the nucleoplasm. Soluble nuclear transport receptors bind signal-dependent cargos to form transport complexes that diffuse through the NPC and are then disassembled. Although transport receptors enable the NPC's permeability barrier to be overcome, directionality is established by complex assembly and disassembly. Here, we delineate the choreography of importin-α/CAS complex assembly and disassembly in permeabilized cells, using single-molecule fluorescence resonance energy transfer and particle tracking. Monitoring interaction sequences in intact NPCs ensures spatiotemporal preservation of structures and interactions critical for activity in vivo. We show that key interactions between components are reversible, multiple outcomes are often possible, and the assembly and disassembly of complexes are precisely controlled to occur at the appropriate place and time. Importin-α mutants that impair interactions during nuclear import were used together with cytoplasmic Ran GTPase-activating factors to demonstrate that importin-α/CAS complexes form in the nuclear basket region, at the termination of protein import, and disassembly of importin-α/CAS complexes after export occurs in the cytoplasmic filament region of the NPC. Mathematical models derived from our data emphasize the intimate connection between transport and the coordinated assembly and disassembly of importin-α/CAS complexes for generating productive transport cycles.


Subject(s)
Active Transport, Cell Nucleus/physiology , Cellular Apoptosis Susceptibility Protein/metabolism , Models, Biological , Multiprotein Complexes/metabolism , Nuclear Pore/metabolism , alpha Karyopherins/metabolism , Electrophoresis, Polyacrylamide Gel , Fluorescence Resonance Energy Transfer , Green Fluorescent Proteins , Protein Binding
14.
Exp Cell Res ; 319(17): 2627-36, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-23948303

ABSTRACT

The human homolog of the yeast cse1 gene (CSE1L) is over-expressed in ovarian cancer. CSE1L forms complex with Ran and importin-α and has roles in nucleocytoplasmic traffic and gene expression. CSE1L accumulated in the nucleus of ovarian cancer cell lines, while it was localized also in the cytoplasm of other cancer cell lines. Nuclear localization depended on AKT, which was constitutively active in ovarian cancer cells, as the CSE1L protein translocated to the cytoplasm when AKT was inactivated. Moreover, the expression of a constitutively active AKT forced the translocation of CSE1L from the cytoplasm to the nucleus in other cancer cells. Nuclear accrual of CSE1L was associated to the nuclear accumulation of the phosphorylated Ran Binding protein 3 (RanBP3), which depended on AKT as well. Also in samples of human ovarian cancer, AKT activation was associated to nuclear accumulation of CSE1L and phosphorylation of RanBP3. Expression profiling of ovarian cancer cells after CSE1L silencing showed that CSE1L was required for the expression of genes promoting invasion and metastasis. In agreement, CSE1L silencing impaired motility and invasiveness of ovarian cancer cells. Altogether these data show that in ovarian cancer cells activated AKT by affecting RanBP3 phosphorylation determines the nuclear accumulation of CSE1L and likely the nuclear concentration of transcription factors conveying pro-oncogenic signals.


Subject(s)
Cell Nucleus/metabolism , Cellular Apoptosis Susceptibility Protein/metabolism , Ovarian Neoplasms/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Transcriptional Activation , Active Transport, Cell Nucleus , Cell Line, Tumor , Cell Movement , Cellular Apoptosis Susceptibility Protein/genetics , Cytoplasm/metabolism , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Nuclear Proteins/metabolism , Nucleocytoplasmic Transport Proteins/metabolism , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Phosphorylation , Transcription, Genetic
15.
J Cell Biol ; 223(2)2024 02 05.
Article in English | MEDLINE | ID: mdl-38241019

ABSTRACT

Exportin receptors are concentrated in the nucleus to transport essential cargoes out of it. A mislocalization of exportins to the cytoplasm is linked to disease. Hence, it is important to understand how their containment within the nucleus is regulated. Here, we have studied the nuclear efflux of exportin2 (cellular apoptosis susceptibility protein or CAS) that delivers karyopherinα (Kapα or importinα), the cargo adaptor for karyopherinß1 (Kapß1 or importinß1), to the cytoplasm in a Ran guanosine triphosphate (RanGTP)-mediated manner. We show that the N-terminus of CAS attenuates the interaction of RanGTPase activating protein 1 (RanGAP1) with RanGTP to slow GTP hydrolysis, which suppresses CAS nuclear exit at nuclear pore complexes (NPCs). Strikingly, a single phosphomimetic mutation (T18D) at the CAS N-terminus is sufficient to abolish its nuclear retention and coincides with metastatic cellular behavior. Furthermore, downregulating Kapß1 disrupts CAS nuclear retention, which highlights the balance between their respective functions that is essential for maintaining the Kapα transport cycle. Therefore, NPCs play a functional role in selectively partitioning exportins in the cell nucleus.


Subject(s)
Cell Nucleus , Cellular Apoptosis Susceptibility Protein , Karyopherins , ran GTP-Binding Protein , Active Transport, Cell Nucleus/physiology , Biological Transport , Cell Nucleus/metabolism , Cytoplasm/metabolism , Karyopherins/metabolism , Nuclear Pore/metabolism , ran GTP-Binding Protein/metabolism , Humans , Cellular Apoptosis Susceptibility Protein/genetics , Cellular Apoptosis Susceptibility Protein/metabolism
16.
Expert Rev Clin Immunol ; 20(9): 1113-1125, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38752776

ABSTRACT

BACKGROUND: Rising cancer-related mortality underscores the importance of biomarkers for treatment and prognosis, with Chromosome Segregation 1 Like (CSE1L) linked to various cancers yet its roles remain partially understood. This study investigates CSE1L's expression and oncogenic mechanisms in solid tumors. RESEARCH DESIGN AND METHODS: We analyzed multi-omics data from 31 solid tumors, measured CSE1L in 41 head and neck carcinoma patients post-chemotherapy via qRT-PCR, and evaluated the impact of CSE1L knockdown on cell proliferation in A549 and HepG2 cells. RESULTS: In this study, we observed significantly elevated levels of CSE1L RNA in 13 tumor tissues and protein levels in 8 tumor tissues compared to their corresponding adjacent normal tissues. Additionally, our investigation unveiled a correlation between heightened CSE1L expression in tumor tissues and worsened patient prognosis, poor response to immunotherapy, and diminished effectiveness of neoadjuvant chemotherapy. Through an analysis of CSE1L mechanisms, we discovered its potential involvement in promoting tumor cell proliferation, enhancing drug resistance, and influencing immune infiltration, thereby impacting patient prognosis and treatment outcomes. Finally, we delved into the potential mechanisms underlying upregulation of CSE1L in tumor tissues. CONCLUSION: Our findings demonstrate that CSE1L promotes tumor development in various malignancies, highlighting its potential as both a therapeutic target and prognostic indicator.


Subject(s)
Biomarkers, Tumor , Cellular Apoptosis Susceptibility Protein , Drug Resistance, Neoplasm , Neoplasms , Humans , Biomarkers, Tumor/metabolism , Biomarkers, Tumor/genetics , Prognosis , Cellular Apoptosis Susceptibility Protein/metabolism , Cellular Apoptosis Susceptibility Protein/genetics , Drug Resistance, Neoplasm/genetics , Neoplasms/immunology , Neoplasms/drug therapy , Cell Proliferation , Hep G2 Cells , Gene Expression Regulation, Neoplastic , A549 Cells
17.
Life Sci ; 340: 122479, 2024 Mar 01.
Article in English | MEDLINE | ID: mdl-38301874

ABSTRACT

THE HEADINGS AIMS: DEAD-box helicase 27 (DDX27), a member of the DEAD-Box nucleic acid helicase family, holds an elusive role in oral squamous cell carcinoma (OSCC). This study aims to unravel the regulatory functions of DDX27 in OSCC and explore its downstream targets. MATERIALS AND METHODS: A commercial oral squamous cell carcinoma (OSCC) tissue microarray (TMA) was utilized. We analyzed differentially expressed genes in OSCC through the GEO database. Target gene silencing was achieved using the shRNA-mediated lentivirus method. Coexpedia analysis identified co-expressed genes associated with DDX27. Additionally, a Co-Immunoprecipitation (Co-IP) experiment confirmed the protein interaction between DDX27 and CSE1L. Xenograft tumor models were employed to evaluate DDX27's role in OSCC tumor formation. KEY FINDINGS: Elevated DDX27 expression in OSCC correlated with a higher pathological grade. DDX27 knockdown resulted in decreased cell proliferation, increased apoptosis, inhibited cell migration, and induced G2/M phase cell cycle arrest, as well as impaired tumor outgrowth. Coexpedia analysis identified STAU1, NELFCD, and CSE1L as top co-expressed genes. Lentiviral vectors targeting STAU1, NELFCD, and CSE1L revealed that silencing CSE1L significantly impaired cell growth, indicating it as a downstream target of DDX27. Cell rescue experiments demonstrated that increased DDX27 levels ameliorated cell proliferation, attenuated apoptosis, and CSE1L depletion blocked cell development induced by DDX27 overexpression. SIGNIFICANCES: This study highlighted DDX27 as a potential therapeutic target for OSCC treatment, shedding light on its crucial role in OSCC development. Targeting DDX27 or its downstream effector, CSE1L, holds promise for innovative OSCC therapies.


Subject(s)
Carcinoma, Squamous Cell , Cellular Apoptosis Susceptibility Protein , DEAD-box RNA Helicases , Head and Neck Neoplasms , Mouth Neoplasms , Humans , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Cytoskeletal Proteins/genetics , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , Gene Expression Regulation, Neoplastic , Head and Neck Neoplasms/genetics , Mouth Neoplasms/pathology , RNA-Binding Proteins/metabolism , Squamous Cell Carcinoma of Head and Neck/genetics , Transcription Factors/metabolism , Cellular Apoptosis Susceptibility Protein/metabolism
18.
J Transl Med ; 11: 29, 2013 Jan 31.
Article in English | MEDLINE | ID: mdl-23369209

ABSTRACT

BACKGROUND: Colorectal carcinomas spread easily to nearby tissues around the colon or rectum, and display strong potential for invasion and metastasis. CSE1L, the chromosome segregation 1-like protein, is implicated in cancer progression and is located in both the cytoplasm and nuclei of tumor cells. We investigated the prognostic significance of cytoplasmic vs. nuclear CSE1L expression in colorectal cancer. METHODS: The invasion- and metastasis-stimulating activities of CSE1L were studied by in vitro invasion and animal experiments. CSE1L expression in colorectal cancer was assayed by immunohistochemistry, with tissue microarray consisting of 128 surgically resected specimens; and scored using a semiquantitative method. The correlations between CSE1L expression and clinicopathological parameters were analyzed. RESULTS: CSE1L overexpression was associated with increased invasiveness and metastasis of cancer cells. Non-neoplastic colorectal glands showed minimal CSE1L staining, whereas most colorectal carcinomas (99.2%, 127/128) were significantly positive for CSE1L staining. Cytoplasmic CSE1L was associated with cancer stage (P=0.003) and depth of tumor penetration (P=0.007). Cytoplasmic CSE1L expression also correlated with lymph node metastasis of the disease in Cox regression analysis CONCLUSIONS: CSE1L regulates the invasiveness and metastasis of cancer cells, and immunohistochemical analysis of cytoplasmic CSE1L in colorectal tumors may provide a useful aid to prognosis.


Subject(s)
Cellular Apoptosis Susceptibility Protein/metabolism , Colorectal Neoplasms/metabolism , Cytoplasm/metabolism , Neoplasm Invasiveness , Adult , Aged , Aged, 80 and over , Animals , Cell Line, Tumor , Colorectal Neoplasms/pathology , Female , Humans , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Middle Aged , Tissue Array Analysis
19.
Angew Chem Int Ed Engl ; 52(1): 410-4, 2013 Jan 02.
Article in English | MEDLINE | ID: mdl-23080551

ABSTRACT

A Prins cyclization between a polymer-bound aldehyde and a homoallylic alcohol served as the key step in the synthesis of tetrahydropyran derivatives. A phenotypic screen led to the identification of compounds that inhibit mitosis (as seen by the accumulation of round cells with condensed DNA and membrane blebs). These compounds were termed tubulexins as they target the CSE1L protein and the vinca alkaloid binding site of tubulin.


Subject(s)
Cellular Apoptosis Susceptibility Protein/metabolism , Pyrans/chemical synthesis , Pyrans/pharmacology , Tubulin/metabolism , Animals , Binding Sites , Cellular Apoptosis Susceptibility Protein/chemistry , HeLa Cells , Humans , MCF-7 Cells , Mitosis/drug effects , Mitosis Modulators , Pyrans/chemistry , Tubulin/chemistry , Tubulin Modulators/chemical synthesis , Tubulin Modulators/chemistry , Tubulin Modulators/pharmacology
20.
Mol Med ; 18: 1269-80, 2012 Dec 06.
Article in English | MEDLINE | ID: mdl-22952058

ABSTRACT

Tumor-derived microvesicles are rich in metastasis-related proteases and play a role in the interactions between tumor cells and tumor microenvironment in tumor metastasis. Because shed microvesicles may remain in the extracellular environment around tumor cells, the microvesicle membrane protein may be the potential target for cancer therapy. Here we report that chromosome segregation 1-like (CSE1L) protein is a microvesicle membrane protein and is a potential target for cancer therapy. v-H-Ras expression induced extracellular signal-regulated kinase (ERK)-dependent CSE1L phosphorylation and microvesicle biogenesis in various cancer cells. CSE1L overexpression also triggered microvesicle generation, and CSE1L knockdown diminished v-H-Ras-induced microvesicle generation, matrix metalloproteinase (MMP)-2 and MMP-9 secretion and metastasis of B16F10 melanoma cells. CSE1L was preferentially accumulated in microvesicles and was located in the microvesicle membrane. Furthermore, anti-CSE1L antibody-conjugated quantum dots could target tumors in animal models. Our findings highlight a novel role of Ras-ERK signaling in tumor progression and suggest that CSE1L may be involved in the "early" and "late" metastasis of tumor cells in tumorigenesis. Furthermore, the novel microvesicle membrane protein, CSE1L, may have clinical utility in cancer diagnosis and targeted cancer therapy.


Subject(s)
Cell-Derived Microparticles/metabolism , Cellular Apoptosis Susceptibility Protein/metabolism , Neoplasms/pathology , ras Proteins/metabolism , Animals , Antibodies/pharmacology , Cell Line, Tumor , Cellular Apoptosis Susceptibility Protein/blood , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Neoplasm Metastasis , Neoplasms/blood , Phosphorylation/drug effects , Phosphothreonine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL