Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Adv Sci (Weinh) ; 10(35): e2303535, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37904651

ABSTRACT

Mitochondrial dysfunction and abnormal energy metabolism are major features of cancer. However, the mechanisms underlying mitochondrial dysfunction during cancer progression are far from being clarified. Here, it is demonstrated that the expression level of succinyl-coenzyme A (CoA) synthetase GDP-forming subunit ß (SUCLG2) can affect the overall succinylation of lung adenocarcinoma (LUAD) cells. Succinylome analysis shows that the deletion of SUCLG2 can upregulate the succinylation level of mitochondrial proteins and inhibits the function of key metabolic enzymes by reducing either enzymatic activity or protein stability, thus dampening mitochondrial function in LUAD cells. Interestingly, SUCLG2 itself is also succinylated on Lys93, and this succinylation enhances its protein stability, leading to the upregulation of SUCLG2 and promoting the proliferation and tumorigenesis of LUAD cells. Sirtuin 5 (SIRT5) desuccinylates SUCLG2 on Lys93, followed by tripartite motif-containing protein 21 (TRIM21)-mediated ubiquitination through K63-linkage and degradation in the lysosome. The findings reveal a new role for SUCLG2 in mitochondrial dysfunction and clarify the mechanism of the succinylation-mediated protein homeostasis of SUCLG2 in LUAD, thus providing a theoretical basis for developing anti-cancer drugs targeting SUCLG2.


Subject(s)
Adenocarcinoma of Lung , Mitochondrial Diseases , Humans , Mitochondria/metabolism , Mitochondrial Diseases/metabolism , Adenocarcinoma of Lung/metabolism
2.
iScience ; 26(7): 107151, 2023 Jul 21.
Article in English | MEDLINE | ID: mdl-37416474

ABSTRACT

The dysregulation of glutamine metabolism provides survival advantages for tumors by supplementing tricarboxylic acid cycle. Glutamate dehydrogenase 1 (GLUD1) is one of the key enzymes in glutamine catabolism. Here, we found that enhanced protein stability was the key factor for the upregulation of GLUD1 in lung adenocarcinoma. We discovered that GLUD1 showed a high protein expression in lung adenocarcinoma cells or tissues. We elucidated that STIP1 homology and U-box-containing protein 1 (STUB1) was the key E3 ligase responsible for ubiquitin-mediated proteasomal degradation of GLUD1. We further showed that lysine 503 (K503) was the main ubiquitination site of GLUD1, inhibiting the ubiquitination at this site promoted the proliferation and tumor growth of lung adenocarcinoma cells. Taken together, this study clarifies the molecular mechanism of GLUD1 in maintaining protein homeostasis in lung adenocarcinoma, which provides a theoretical basis for the development of anti-cancer drugs targeting GLUD1.

4.
Cell Death Dis ; 13(12): 1075, 2022 12 27.
Article in English | MEDLINE | ID: mdl-36575176

ABSTRACT

Nutrient-limiting conditions are common during cancer development. The coordination of cellular glucose levels and cell survival is a fundamental question in cell biology and has not been completely understood. 4EBP1 is known as a translational repressor to regulate cell proliferation and survival by controlling translation initiation, however, whether 4EBP1 could participate in tumor survival by other mechanism except for translational repression function, especially under glucose starvation conditions remains unknown. Here, we found that protein levels of 4EBP1 was up-regulated in the central region of the tumor which always suffered nutrient deprivation compared with the peripheral region. We further discovered that 4EBP1 was dephosphorylated by PTPMT1 under glucose starvation conditions, which prevented 4EBP1 from being targeted for ubiquitin-mediated proteasomal degradation by HERC5. After that, 4EBP1 translocated to cytoplasm and interacted with STAT3 by competing with JAK and ERK, leading to the inactivation of STAT3 in the cytoplasm, resulting in apoptosis under glucose withdrawal conditions. Moreover, 4EBP1 knockdown increased the tumor volume and weight in xenograft models by inhibiting apoptosis in the central region of tumor. These findings highlight a novel mechanism for 4EBP1 as a new cellular glucose sensor in regulating cancer cell death under glucose deprivation conditions, which was different from its classical function as a translational repressor.


Subject(s)
Adaptor Proteins, Signal Transducing , Cell Cycle Proteins , Glucose , Lung Neoplasms , Humans , Cell Death , Cell Proliferation , Glucose/metabolism , Lung Neoplasms/genetics , PTEN Phosphohydrolase/metabolism , Signal Transduction , Animals , Adaptor Proteins, Signal Transducing/metabolism , Cell Cycle Proteins/metabolism
5.
Cell Death Dis ; 13(10): 870, 2022 10 14.
Article in English | MEDLINE | ID: mdl-36241648

ABSTRACT

Rho GTPases play an essential role in many cellular processes, including cell cycle progress, cell motility, invasion, migration, and transformation. Several studies indicated that the dysregulation of Rho GTPase signaling is closely related to tumorigenesis. Rho GEFs considered being positive regulators of Rho GTPase, promoting the dissociation of Rho protein from GDP and binding to GTP, thus activating the downstream signaling pathway. Herein, we demonstrated that ARHGEF3, a member of the Rho GEFs family, played an important role in non-small cell lung cancer (NSCLC). We found that ARHGEF3 was highly expressed in non-small cell lung cancer and facilitated cancer cell proliferation of NSCLC cells in vitro and in vivo. Further studies demonstrated that ARHGEF3 enhanced the protein homeostasis of ATP-citrate lyase (ACLY) by reducing its acetylation on Lys17 and Lys86, leading to the dissociation between ACLY and its E3 ligase-NEDD4. Interestingly, this function of ARHGEF3 on the protein homeostasis of ACLY was independent of its GEF activity. Taken together, our findings uncover a novel function of ARHGEF3, suggesting that ARHGEF3 is a promising therapeutic target in non-small cell lung cancer.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , ATP Citrate (pro-S)-Lyase/metabolism , Adenosine Triphosphate , Carcinoma, Non-Small-Cell Lung/genetics , Cell Proliferation , Guanosine Triphosphate , Humans , Lung Neoplasms/genetics , Rho Guanine Nucleotide Exchange Factors/genetics , Rho Guanine Nucleotide Exchange Factors/metabolism , Ubiquitin-Protein Ligases/metabolism , rho GTP-Binding Proteins/genetics , rho GTP-Binding Proteins/metabolism
6.
Cell Commun Signal ; 20(1): 133, 2022 08 30.
Article in English | MEDLINE | ID: mdl-36042481

ABSTRACT

BACKGROUND: Glycogen-Interacting Protein 1 (GNIP1), an E3 ligase, is a member of the tripartite motif (TRIM) family proteins. Current studies on GNIP1 mainly focus on glycogen metabolism. However, the function and molecular mechanisms of GNIP1 in regulating autophagy still remains unclear. This study aimed to investigate the regulatory mechanism of GNIP1 in regulating autophagy in non-small cell lung cancer (NSCLC). METHODS: Crystal violet staining assays were used to evaluate the ability of cell growth and proliferation. Transwell and scratch wound healing assays were used to evaluate the cell migration ability. The protein expressions were measured by western blot and immunohistochemistry. Co-immunoprecipitation assays determined the protein-protein interactions. The in vivo effect of GNIP1 on tumor growth was determined by xenograft assay. RESULTS: We found that GNIP1 was overexpressed in tumor tissues and the expression level of GNIP1 was related to the poor prognosis and the survival time of NSCLC patients. In non-small cell lung cancer (NSCLC), GNIP1 increased proliferation and migration of cancer cells by promoting autophagy. Mechanistic studies indicated that GNIP1, as a scaffold protein, recruited BECN1 and LC3B to promote the formation of autophagosomes. Besides, GNIP1 mediated the degradation of 14-3-3ζ, the negative regulator of VPS34 complex, thus promoting autophagy. Overexpressing GNIP1 promoted tumorigenesis and enhanced autophagy in xenograft models. CONCLUSION: GNIP1 promotes proliferation and migration of NSCLC cells through mediating autophagy, which provides theoretical basis for targeting GNIP1 as anti-cancer drugs. Video Abstract.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Tripartite Motif Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , 14-3-3 Proteins/metabolism , Autophagy , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Proliferation , Glycogen/metabolism , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology
7.
Int J Biol Sci ; 18(11): 4452-4465, 2022.
Article in English | MEDLINE | ID: mdl-35864951

ABSTRACT

Inhibiting cancer metabolism via glutaminase (GAC) is a promising strategy to disrupt tumor progression. However, mechanism regarding GAC acetylation remains mostly unknown. In this study, we demonstrate that lysine acetylation is a vital post-translational modification that inhibits GAC activity in non-small cell lung cancer (NSCLC). We identify that Lys311 is the key acetylation site on GAC, which is deacetylated by HDAC4, a class II deacetylase. Lys311 acetylation stimulates the interaction between GAC and TRIM21, an E3 ubiquitin ligase of the tripartite motif (TRIM) family, therefore promoting GAC K63-linked ubiquitination and inhibiting GAC activity. Furthermore, GACK311Q mutation in A549 cells decreases cell proliferation and alleviates tumor malignancy. Our findings reveal a novel mechanism of GAC regulation by acetylation and ubiquitination that participates in non-small cell lung cancer tumorigenesis.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Acetylation , Carcinogenesis/genetics , Carcinogenesis/metabolism , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Proliferation/genetics , Cell Transformation, Neoplastic , Glutaminase/genetics , Glutaminase/metabolism , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Humans , Lung Neoplasms/metabolism , Repressor Proteins/metabolism , Ubiquitination
8.
Autophagy ; 18(6): 1416-1432, 2022 06.
Article in English | MEDLINE | ID: mdl-34720024

ABSTRACT

Altered glutamine metabolism is an important aspect of cancer metabolic reprogramming. The GLS isoform GAC (glutaminase C), the rate-limiting enzyme in glutaminolysis, plays a vital role in cancer initiation and progression. Our previous studies demonstrated that phosphorylation of GAC was essential for its high enzymatic activity. However, the molecular mechanisms for GAC in maintaining its high enzymatic activity and protein stability still need to be further clarified. FAIM/FAIM1 (Fas apoptotic inhibitory molecule) is known as an important anti-apoptotic protein, but little is known about its function in tumorigenesis. Here, we found that knocking down FAIM induced macroautophagy/autophagy through suppressing the activation of the MTOR pathway in lung adenocarcinoma. Further studies demonstrated that FAIM could promote the tetramer formation of GAC through increasing PRKCE/PKCε-mediated phosphorylation. What's more, FAIM also stabilized GAC through sequestering GAC from degradation by protease ClpXP. These effects increased the production of α-ketoglutarate, leading to the activation of MTOR. Besides, FAIM also promoted the association of ULK1 and MTOR and this further suppressed autophagy induction. These findings discovered new functions of FAIM and elucidated an important molecular mechanism for GAC in maintaining its high enzymatic activity and protein stability.


Subject(s)
Adenocarcinoma of Lung , Apoptosis Regulatory Proteins , Glutamine , Lung Neoplasms , Adenocarcinoma of Lung/metabolism , Apoptosis Regulatory Proteins/metabolism , Autophagy , Glutaminase/metabolism , Glutamine/metabolism , Humans , Lung Neoplasms/metabolism , TOR Serine-Threonine Kinases
9.
Acta Biochim Biophys Sin (Shanghai) ; 54(11): 1610-1618, 2022 Nov 25.
Article in English | MEDLINE | ID: mdl-36604147

ABSTRACT

Abnormal proliferation and cell cycle perturbation are the main hallmarks of breast cancer. Cyclin-dependent kinase 1 (CDK1) is one of the key kinases for cell transition from the G2 phase to M phase during the cell cycle progression. However, little is known about the degradation mechanisms of CDK1. USP14 (ubiquitin-specific processing protease 14) is an important proteasome-associated deubiquitinase that is critical for proteome homeostasis and plays a crucial role in the initiation and development of cancer. In this study, we find that USP14 shows high expression in breast cancer cells and results in the abnormal proliferation of cancer cells. Furthermore, we examine cell cycle distribution by flow cytometry and find that inhibition of USP14 causes cell cycle arrest in G2/M phase. As CDK1 is the key kinase in G2/M phase, we detect the interaction between USP14 and CDK1 and the effect of USP14 on the deubiquitination of CDK1. The results reveal that USP14 interacts with CDK1 and stabilizes CDK1 by deubiquitinating K48-linked ubiquitination. In conclusion, our findings reveal an indispensable role of USP14 in regulating cell cycle progression by stabilizing CDK1 in breast cancer, suggesting that USP14 may be used as a potential therapeutic target in breast cancer therapy.


Subject(s)
Breast Neoplasms , Humans , Female , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , CDC2 Protein Kinase/metabolism , Ubiquitination , Cell Division , Cell Cycle , Cell Line, Tumor , Ubiquitin Thiolesterase/genetics
10.
Cell Death Dis ; 12(5): 430, 2021 04 30.
Article in English | MEDLINE | ID: mdl-33931584

ABSTRACT

Grainyhead-like 1 (GRHL1) is a transcription factor involved in embryonic development. However, little is known about the biological functions of GRHL1 in cancer. In this study, we found that GRHL1 was upregulated in non-small cell lung cancer (NSCLC) and correlated with poor survival of patients. GRHL1 overexpression promoted the proliferation of NSCLC cells and knocking down GRHL1 inhibited the proliferation. RNA sequencing showed that a series of cell cycle-related genes were altered when knocking down GRHL1. We further demonstrated that GRHL1 could regulate the expression of cell cycle-related genes by binding to the promoter regions and increasing the transcription of the target genes. Besides, we also found that EGF stimulation could activate GRHL1 and promoted its nuclear translocation. We identified the key phosphorylation site at Ser76 on GRHL1 that is regulated by the EGFR-ERK axis. Taken together, these findings elucidate a new function of GRHL1 on regulating the cell cycle progression and point out the potential role of GRHL1 as a drug target in NSCLC.


Subject(s)
Genes, cdc/genetics , Lung Neoplasms/genetics , Repressor Proteins/metabolism , Adult , Aged , Aged, 80 and over , Animals , Disease Progression , ErbB Receptors/metabolism , Humans , Male , Mice , Mice, Nude , Middle Aged , Up-Regulation , Young Adult
11.
Cell Signal ; 69: 109543, 2020 05.
Article in English | MEDLINE | ID: mdl-31958511

ABSTRACT

The gene trim7 encodes at least four isoforms Glycogenin-interacting protein 1 (GNIP1), GNIP2, GNIP3 and Tripartite motif containing 7 (TRIM7). GNIP1, the longest isoform, has been reported acting as an oncogene. However, it is very interesting that TRIM7, the shortest isoform, only 15 amino acids different from GNIP1 in C-terminal, acts in a completely different way from that of GNIP1 in our present study. TRIM7 expression was decreased in tumor compared with adjacent normal tissues, and the level of TRIM7 was negatively correlated with clinical stage of 94 patients with lung cancer. In vitro, TRIM7 dramatically inhibited the proliferation and migration of tumor cells, and promoted cell apoptosis. Further study showed that TRIM7 interacted with p65 via its C-terminal which is different from GNIP1. The interaction between TRIM7 and p65 promoted the ubiquitination of p65 and finally accelerated the degradation of p65 via 26S proteasome. In vivo, the tumor volume and weight were decreased by TRIM7 stable expression. Meanwhile, Ki67 was down-regulated, thyroid transcription factor 1 (TTF-1) and Caspase 3 were up-regulated in TRIM7 overexpression group in xenograft model. It is very impressive that TRIM7t (a truncated TRIM7 without C-terminal sequence that different with GNIP1) had little effect on the tumor growth in vivo. These findings highlight a curious mechanism for negative regulation of NF-kappa B signaling pathway by TRIM7 and demonstrate that TRIM7 would be a potential therapeutic target for lung cancer.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , Lung Neoplasms/metabolism , Signal Transduction , Transcription Factor RelA/metabolism , Tripartite Motif Proteins/physiology , Ubiquitin-Protein Ligases/physiology , Animals , Cell Line, Tumor , Cell Proliferation , Humans , Mice
12.
Pathol Res Pract ; 215(10): 152592, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31474315

ABSTRACT

Breast cancer is the most common malignant tumor among women in China, which seriously threatens women's physical and mental health. Tumorigenesis is closely related to the dysregulation of cell cycle. The cell cycle progression includes interphase and mitotic phase (M phase). Cyclin B1 is a key protein in regulating M phase, which is essential for the whole cell cycle progression. CyclinB1 can be degraded through ubiquitination mediated by the anaphase promoting complex/cyclosome (APC/C). However, the mechanism of how CyclinB1 is deubiquitinated in breast cancer still remains unclear. In this study, we discovered that CyclinB1 interacted with ubiquitin-specific peptidase 14 (USP14). Based on the deubiquitinating function of USP14, we detected the effect of USP14 on the ubiquitination of CyclinB1. Inhibiting the activity of USP14 or USP14 knockdown significantly increased the ubiquitination of CyclinB1. In accordance with this, knocking down USP14 arrested cell cycle at G2/M phase. Knocking down USP14 with siRNAs significantly inhibited the proliferation and migration of breast cancer cells. In conclusion, our study demonstrated that USP14 regulated the cell cycle of breast cancer cells by regulating the ubiquitination of CyclinB1, which will provide a solid theoretical basis for the development of anti-cancer drugs targeting USP14.


Subject(s)
Breast Neoplasms/metabolism , Cell Cycle/physiology , Cyclin B1/metabolism , Ubiquitin Thiolesterase/metabolism , Ubiquitination , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement/physiology , Cyclin B1/genetics , Female , Humans , Ubiquitin Thiolesterase/genetics
13.
Cell Cycle ; 18(9): 1019-1032, 2019 05.
Article in English | MEDLINE | ID: mdl-31023143

ABSTRACT

The anaphase promoting complex/cyclosome (APC/C), a cell cycle-regulated E3 ubiquitin ligase, is responsible for the transition from metaphase to anaphase and the exit from mitosis. The anaphase promoting complex subunit 10 (APC10), a subunit of the APC/C, executes a vital function in substrate recognition. However, no research has reported the connection between APC10 and cancer until now. In this study, we uncovered a novel, unprecedented role of APC10 in tumor progression, which is independent of APC/C. First, aberrant increase of APC10 expression was validated in non-small cell lung cancer (NSCLC) cells and tissues, and the absence of APC10 repressed cell proliferation and migration. Of great interest, we found that APC10 inhibition induced cell cycle arrest at the G0/G1 phase and reduced the expression of the APC/C substrate, Cyclin B1; this finding is different from the conventional concept of the accumulation of Cyclin B1 and cell cycle arrest in metaphase. Further, APC10 was found to interact with glutaminase C (GAC), and the inhibition of APC10 weakened glutamine metabolism and induced excessive autophagy. Taken together, these findings identify a novel function of APC10 in the regulation of NSCLC tumorigenesis and point to the possibility of APC10 as a new target for cancer therapy.


Subject(s)
Apc10 Subunit, Anaphase-Promoting Complex-Cyclosome/metabolism , Carcinogenesis/metabolism , Carcinoma, Non-Small-Cell Lung/metabolism , Disease Progression , Lung Neoplasms/metabolism , A549 Cells , Apc10 Subunit, Anaphase-Promoting Complex-Cyclosome/genetics , Autophagy/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Cell Movement/genetics , Cell Proliferation/genetics , Cytoplasm/metabolism , G1 Phase/genetics , Glutaminase/metabolism , Glutamine/metabolism , Humans , Lung Neoplasms/pathology , RNA, Small Interfering/genetics , Resting Phase, Cell Cycle/genetics , Signal Transduction/genetics , Transfection
14.
Autophagy ; 14(12): 2035-2048, 2018.
Article in English | MEDLINE | ID: mdl-30231667

ABSTRACT

Macroautophagy/autophagy is a multistep cellular process that sequesters cytoplasmic components for lysosomal degradation. BECN1/Beclin1 is a central protein that assembles cofactors for the formation of a BECN1-PIK3C3-PIK3R4 complex to trigger the autophagy protein cascade. Discovering the regulators of BECN1 is important for understanding the mechanism of autophagy induction. Here, we demonstrate that TRIM59, a tripartite motif protein, plays an important role in autophagy regulation in non-small cell lung cancer (NSCLC). On the one hand, TRIM59 regulates the transcription of BECN1 through negatively modulating the NFKB pathway. On the other hand, TRIM59 regulates TRAF6 induced K63-linked ubiquitination of BECN1, thus affecting the formation of the BECN1-PIK3C3 complex. We further demonstrate that TRIM59 can mediate K48-linked ubiquitination of TRAF6 and promote the proteasomal degradation of TRAF6. Taken together, our findings reveal novel dual roles for TRIM59 in autophagy regulation by affecting both the transcription and the ubiquitination of BECN1. Abbreviations: ACTB: actin beta; BECN1: beclin 1; CHX: cycloheximide; CQ: chloroquine; GFP: green fluorescent protein; HA: haemagglutinin tag; His: polyhistidine tag; LC3B: microtubule associated protein 1 light chain 3 beta; NFKB: nuclear factor kappa B; NFKBIA: NFKB inhibitor alpha; NSCLC: non-small cell lung cancer; PIK3C3: phosphatidylinositol 3-kinase catalytic subunit type 3; RELA: RELA proto-oncogene, NF-kB subunit; SQSTM1: sequestosome 1; tGFP: Turbo green fluorescent protein; TRAF6: TNF receptor associated factor 6; TRIM59: tripartite motif containing 59; B: ubiquitin.


Subject(s)
Autophagy/genetics , Beclin-1/genetics , Beclin-1/metabolism , Membrane Proteins/physiology , Metalloproteins/physiology , A549 Cells , Autophagy/physiology , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Gene Expression Regulation, Neoplastic/genetics , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Proto-Oncogene Mas , Signal Transduction/genetics , Transcription, Genetic/genetics , Tripartite Motif Proteins , Tumor Cells, Cultured , Ubiquitination/genetics
15.
Cell Res ; 28(6): 655-669, 2018 06.
Article in English | MEDLINE | ID: mdl-29515166

ABSTRACT

Glutamine metabolism plays an important role in cancer development and progression. Glutaminase C (GAC), the first enzyme in glutaminolysis, has emerged as an important target for cancer therapy and many studies have focused on the mechanism of enhanced GAC expression in cancer cells. However, little is known about the post-translational modification of GAC. Here, we report that phosphorylation is a crucial post-translational modification of GAC, which is responsible for the higher glutaminase activity in lung tumor tissues and cancer cells. We identify the key Ser314 phosphorylation site on GAC that is regulated by the NF-κB-PKCε axis. Blocking Ser314 phosphorylation by the S314A mutation in lung cancer cells inhibits the glutaminase activity, triggers genetic reprogramming, and alleviates tumor malignancy. Furthermore, we find that a high level of GAC phosphorylation correlates with poor survival rate of lung cancer patients. These findings highlight a previously unappreciated mechanism for activation of GAC by phosphorylation and demonstrate that targeting glutaminase activity can inhibit oncogenic transformation.


Subject(s)
Carcinogenesis/metabolism , Carcinoma, Non-Small-Cell Lung/metabolism , Glutaminase/metabolism , Lung Neoplasms/metabolism , Protein Kinase C-epsilon/metabolism , Animals , Carcinogenesis/pathology , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Humans , Lung Neoplasms/pathology , Mice, Inbred BALB C , Mice, Nude , Phosphorylation
16.
Am J Transl Res ; 9(9): 4217-4226, 2017.
Article in English | MEDLINE | ID: mdl-28979695

ABSTRACT

The transcription factor, Grainyhead-like 2 (GRHL2), is involved in wound healing, epidermal integrity, and epithelial-to-mesenchymal transition (EMT) in various biological processes; however, the biological function of GRHL2 in non-small cell lung cancer (NSCLC) is unknown. In the current study, we investigated the effect of GRHL2 on cell growth and migration in NSCLC cell lines and clinical tissues. Immunohistochemical analysis of clinical NSCLC specimens revealed that patients with high GRHL2 expression were associated with poor prognosis compared to patients with low GRHL2 expression. GRHL2 overexpression promoted cell growth and colony formation, and simultaneously suppressed cell migration in NSCLC cells. Furthermore, GRHL2 decreased the transcriptional activity of RhoG by directly binding to the RhoG promoter region. These findings confirm that GRHL2 plays an important role in regulating cell proliferation and migration in NSCLC.

17.
Oncotarget ; 8(17): 28063-28073, 2017 Apr 25.
Article in English | MEDLINE | ID: mdl-28039459

ABSTRACT

Metabolic reprogramming is critical for cancer cell proliferation. Glutaminolysis which provides cancer cells with bioenergetics and intermediates for macromolecular synthesis have been intensively studied in recent years. Glutaminase C (GAC) is the first and rate-limiting enzyme in glutaminolysis and plays important roles in cancer initiation and progression. We previously screened a small molecule named 968, a specific inhibitor of GAC, to block the proliferation of human breast cancer cells. In this study, we found that 968 effectively inhibited NSCLC cell proliferation and migration and arrested G0/G1 phase of cell cycle. Furthermore, we demonstrated that 968 inhibited the EGFR/ERK pathway via decreasing the expression of EGFR and phospho-ERK. Apart from this, we discovered that 968 treatment induced autophagy to protect cells against apoptosis and the combination of 968 with autophagy inhibitor Chloroquine (CQ) had synergistic effects on the growth of NSCLC cells. Thus, our study pointed out a new therapeutic strategy for NSCLC treatment by combination of 968 with CQ.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , Enzyme Inhibitors/pharmacology , ErbB Receptors/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Glutaminase/antagonists & inhibitors , Lung Neoplasms/metabolism , Signal Transduction/drug effects , Autophagy/drug effects , Beclin-1/metabolism , Cell Cycle/drug effects , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Humans
18.
Cell Signal ; 30: 59-66, 2017 01.
Article in English | MEDLINE | ID: mdl-27871936

ABSTRACT

COMMD protein family is an evolutionarily conserved gene family implicated in a number of critical processes including inflammation, copper homeostasis, sodium balance, endosomal sorting and cancer. In an effort to profile the expression pattern of COMMD family in several non-small cell lung cancer (NSCLC) cell lines, we found that compared with that in human bronchial epithelial (HBE) cells, the mRNA expression levels of five COMMD genes including COMMD3, COMMD4, COMMD5, COMMD6 and COMMD8 were significantly down-regulated, whereas COMMD9 was up-regulated in NSCLC cell lines. Here we reported that the expression of COMMD9 protein was significantly increased in various NSCLC cell lines and tissue samples. SiRNA-induced knocking down of COMMD9 inhibited proliferation and migration, arrested cell cycle at G1/S transition and induced autophagy in NSCLC cells. Mechanistically, COMMD9 interacted with the TFDP1 through COMM domain, and DNA-binding domain of TFDP1 was required for this interaction. Moreover, decreased expression COMMD9 attenuated TFDP1/E2F1 activation accompanied with enhanced p53 signaling pathway. Taken together, these findings demonstrate that COMMD9 participates in TFDP1/E2F1 activation and plays a critical role in non-small cell lung cancer.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Carcinoma, Non-Small-Cell Lung/genetics , E2F1 Transcription Factor/genetics , Lung Neoplasms/genetics , Transcription Factor DP1/genetics , Transcription, Genetic , Adaptor Proteins, Signal Transducing/genetics , Autophagy/genetics , Cell Adhesion/genetics , Cell Cycle Checkpoints/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , G1 Phase/genetics , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , HEK293 Cells , Humans , Protein Binding/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , S Phase/genetics , Signal Transduction/genetics , Tumor Stem Cell Assay , Tumor Suppressor Protein p53/metabolism
19.
Oncotarget ; 7(1): 610-21, 2016 Jan 05.
Article in English | MEDLINE | ID: mdl-26575584

ABSTRACT

The epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor (TKI) erlotinib has been approved based on the clinical benefit in non-small cell lung cancer (NSCLC) patients over the past decade. Unfortunately, cancer cells become resistant to this agent via various mechanisms, and this limits the improvement in patient outcomes. Thus, it is urgent to develop novel agents to overcome erlotinib resistance. Here, we propose a novel strategy to overcome acquired erlotinib resistance in NSCLC by inhibiting glutaminase activity. Compound 968, an inhibitor of the glutaminase C (GAC), when combined with erlotinib potently inhibited the cell proliferation of erlotinib-resistant NSCLC cells HCC827ER and NCI-H1975. The combination of compound 968 and erlotinib not only decreased GAC and EGFR protein expression but also inhibited GAC activity in HCC827ER cells. The growth of erlotinib-resistant cells was glutamine-dependent as proved by GAC gene knocked down and rescue experiment. More importantly, compound 968 combined with erlotinib down-regulated the glutamine and glycolysis metabolism in erlotinib-resistant cells. Taken together, our study provides a valuable approach to overcome acquired erlotinib resistance by blocking glutamine metabolism and suggests that combination of EGFR-TKI and GAC inhibitor maybe a potential treatment strategy for acquired erlotinib-resistant NSCLC.


Subject(s)
Benzophenanthridines/pharmacology , Drug Resistance, Neoplasm/drug effects , Erlotinib Hydrochloride/pharmacology , Glutaminase/antagonists & inhibitors , Apoptosis/drug effects , Apoptosis/genetics , Blotting, Western , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Dose-Response Relationship, Drug , Flow Cytometry , Glutaminase/genetics , Glutaminase/metabolism , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mitochondria/enzymology , Protein Kinase Inhibitors/pharmacology , RNA Interference , Time Factors
20.
PLoS One ; 10(11): e0142596, 2015.
Article in English | MEDLINE | ID: mdl-26599082

ABSTRACT

TRIM protein family is an evolutionarily conserved gene family implicated in a number of critical processes including inflammation, immunity, antiviral and cancer. In an effort to profile the expression patterns of TRIM superfamily in several non-small cell lung cancer (NSCLC) cell lines, we found that the expression of 10 TRIM genes including TRIM3, TRIM7, TRIM14, TRIM16, TRIM21, TRIM22, TRIM29, TRIM59, TRIM66 and TRIM70 was significantly upregulated in NSCLC cell lines compared with the normal human bronchial epithelial (HBE) cell line, whereas the expression of 7 other TRIM genes including TRIM4, TRIM9, TRIM36, TRIM46, TRIM54, TRIM67 and TRIM76 was significantly down-regulated in NSCLC cell lines compared with that in HBE cells. As TRIM59 has been reported to act as a proto-oncogene that affects both Ras and RB signal pathways in prostate cancer models, we here focused on the role of TRIM59 in the regulation of NSCLC cell proliferation and migration. We reported that TRIM59 protein was significantly increased in various NSCLC cell lines. SiRNA-induced knocking down of TRIM59 significantly inhibited the proliferation and migration of NSCLC cell lines by arresting cell cycle in G2 phase. Moreover, TRIM59 knocking down affected the expression of a number of cell cycle proteins including CDC25C and CDK1. Finally, we knocked down TRIM59 and found that p53 protein expression levels did not upregulate, so we proposed that TRIM59 may promote NSCLC cell growth through other pathways but not the p53 signaling pathway.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Cell Cycle Proteins/genetics , Membrane Proteins/genetics , Metalloproteins/genetics , Tumor Suppressor Protein p53/biosynthesis , Carcinoma, Non-Small-Cell Lung/pathology , Cell Cycle Proteins/biosynthesis , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Humans , Intracellular Signaling Peptides and Proteins , Membrane Proteins/biosynthesis , Metalloproteins/biosynthesis , Proto-Oncogene Mas , RNA, Small Interfering , Signal Transduction/genetics , Transcriptional Activation/genetics , Tripartite Motif Proteins , Tumor Suppressor Protein p53/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...