17 results on '"Jia L"'
Search Results
2. PTEN activation through K163 acetylation by inhibiting HDAC6 contributes to tumour inhibition
- Author
-
Meng, Z, Jia, L-F, and Gan, Y-H
- Published
- 2016
- Full Text
- View/download PDF
3. KLF5 promotes breast cancer proliferation, migration and invasion in part by upregulating the transcription of TNFAIP2
- Author
-
Jia, L, Zhou, Z, Liang, H, Wu, J, Shi, P, Li, F, Wang, Z, Wang, C, Chen, W, Zhang, H, Wang, Y, Liu, R, Feng, J, and Chen, C
- Published
- 2016
- Full Text
- View/download PDF
4. Modification of sialylation is associated with multidrug resistance in human acute myeloid leukemia
- Author
-
Ma, H, Zhou, H, Song, X, Shi, S, Zhang, J, and Jia, L
- Published
- 2015
- Full Text
- View/download PDF
5. PTEN activation through K163 acetylation by inhibiting HDAC6 contributes to tumour inhibition
- Author
-
Meng, Z, primary, Jia, L-F, additional, and Gan, Y-H, additional
- Published
- 2015
- Full Text
- View/download PDF
6. KLF5 promotes breast cancer proliferation, migration and invasion in part by upregulating the transcription of TNFAIP2
- Author
-
Jia, L, primary, Zhou, Z, additional, Liang, H, additional, Wu, J, additional, Shi, P, additional, Li, F, additional, Wang, Z, additional, Wang, C, additional, Chen, W, additional, Zhang, H, additional, Wang, Y, additional, Liu, R, additional, Feng, J, additional, and Chen, C, additional
- Published
- 2015
- Full Text
- View/download PDF
7. Modification of sialylation is associated with multidrug resistance in human acute myeloid leukemia
- Author
-
Ma, H, primary, Zhou, H, additional, Song, X, additional, Shi, S, additional, Zhang, J, additional, and Jia, L, additional
- Published
- 2014
- Full Text
- View/download PDF
8. Bone marrow stromal cells dictate lanosterol biosynthesis and ferroptosis of multiple myeloma.
- Author
-
Jiang H, Wang L, Zhang Q, Wang S, Jia L, Cheng H, Wang J, Li X, Xie Y, Wang Y, Hu M, Guo J, Li Q, Peng Z, Wang M, Xie Y, Li T, Wang Y, Geng BD, Swaminathan S, Bergsagel PL, and Liu Z
- Subjects
- Animals, Humans, Mice, Reactive Oxygen Species metabolism, Cell Line, Tumor, Iron metabolism, Signal Transduction, Multiple Myeloma pathology, Multiple Myeloma metabolism, Ferroptosis, Lanosterol pharmacology, Mesenchymal Stem Cells metabolism
- Abstract
Ferroptosis has been demonstrated a promising way to counteract chemoresistance of multiple myeloma (MM), however, roles and mechanism of bone marrow stromal cells (BMSCs) in regulating ferroptosis of MM cells remain elusive. Here, we uncovered that MM cells were more susceptible to ferroptotic induction under the interaction of BMSCs using in vitro and in vivo models. Mechanistically, BMSCs elevated the iron level in MM cells, thereby activating the steroid biosynthesis pathway, especially the production of lanosterol, a major source of reactive oxygen species (ROS) in MM cells. We discovered that direct coupling of CD40 ligand and CD40 receptor constituted the key signaling pathway governing lanosterol biosynthesis, and disruption of CD40/CD40L interaction using an anti-CD40 neutralizing antibody or conditional depletion of Cd40l in BMSCs successfully eliminated the iron level and lanosterol production of MM cells localized in the Vk*MYC Vk12653 or NSG mouse models. Our study deciphers the mechanism of BMSCs dictating ferroptosis of MM cells and highlights the therapeutic potential of non-apoptosis strategies for managing refractory or relapsed MM patients., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
9. WDR74 modulates melanoma tumorigenesis and metastasis through the RPL5-MDM2-p53 pathway.
- Author
-
Li Y, Zhou Y, Li B, Chen F, Shen W, Lu Y, Zhong C, Zhang C, Xie H, Katanaev VL, and Jia L
- Subjects
- Animals, Apoptosis genetics, Carcinogenesis genetics, Cell Line, Tumor, Cell Proliferation genetics, Female, Gene Knockout Techniques, Humans, Melanoma genetics, Mice, Phosphorylation, Proteolysis, Proto-Oncogene Proteins genetics, Proto-Oncogene Proteins c-mdm2 metabolism, RNA, Small Interfering metabolism, RNA-Binding Proteins genetics, Ribosomal Proteins metabolism, Signal Transduction genetics, Skin pathology, Skin Neoplasms genetics, Tumor Suppressor Protein p53 metabolism, Ubiquitination genetics, Xenograft Model Antitumor Assays, Carcinogenesis pathology, Melanoma pathology, Proto-Oncogene Proteins metabolism, RNA-Binding Proteins metabolism, Skin Neoplasms pathology
- Abstract
The key molecules and underlying mechanisms of melanoma metastasis remain poorly understood. Using isobaric tag for relative and absolute quantitation (iTRAQ) proteomic screening, probing of patients' samples, functional verification, and mechanistic validation, we identified the important role of the WD repeat-containing protein 74 (WDR74) in melanoma progression and metastasis. Through gain- and loss-of-function approaches, WDR74 was found to promote cell proliferation, apoptosis resistance, and aggressive behavior in vitro. Moreover, WDR74 contributed to melanoma growth and metastasis in vivo. Mechanistically, WDR74 modulates RPL5 protein levels and consequently regulates MDM2 and insulates the ubiquitination degradation of p53 by MDM2. Our study is the first to reveal the oncogenic role of WDR74 in melanoma progression and the regulatory effect of WDR74 on the RPL5-MDM2-p53 pathway. Collectively, WDR74 can serve as a candidate target for the prevention and treatment of melanoma in the clinic.
- Published
- 2020
- Full Text
- View/download PDF
10. Promotion of tumor-associated macrophages infiltration by elevated neddylation pathway via NF-κB-CCL2 signaling in lung cancer.
- Author
-
Zhou L, Jiang Y, Liu X, Li L, Yang X, Dong C, Liu X, Lin Y, Li Y, Yu J, He R, Huang S, Liu G, Zhang Y, Jeong LS, Hoffman RM, and Jia L
- Subjects
- Adenocarcinoma metabolism, Animals, Cell Line, Tumor, Chemokine CCL2 genetics, Humans, Lung Neoplasms metabolism, Mice, NEDD8 Protein genetics, Tumor Microenvironment, Adenocarcinoma pathology, Chemokine CCL2 metabolism, Lung Neoplasms pathology, Macrophages pathology, NEDD8 Protein metabolism, NF-kappa B metabolism, Signal Transduction
- Abstract
Tumor-associated macrophages (TAMs) are the most abundant cancer stromal cells and play an essential role in tumor immunosuppression, providing a suitable microenvironment for cancer development and progression. However, mechanisms of regulating TAMs infiltration in tumor sites are not fully understood. Here, we show that inactivation of neddylation pathway significantly inhibits infiltration of TAMs, leading to the suppression of lung cancer metastasis. RNA-sequencing analysis revealed that neddylation inactivation suppresses the transactivation of chemotactic cytokine ligand 2 (CCL2). Mechanistically, neddylation inactivation inhibits the activity of Cullin-RING ligases (CRLs) and induces the accumulation of its substrate IκBα to block NF-κB transcriptional activity and CCL2 transactivation. As a result, neddylation inactivation exhibits lower chemotaxis of monocytes, thereby decreasing TAMs infiltration, which can be alleviated by CCL2 addition. Moreover, the expression level of NEDD8 is positively correlated with high CCL2 expression in lung adenocarcinoma, conferring a worse overall patient survival. Together, neddylation pathway promotes CCL2 transactivation and TAMs infiltration in lung cancer to provide a tumor-promoting microenvironment, which validates neddylation pathway as a promising target for anti-TAMs therapeutic strategies.
- Published
- 2019
- Full Text
- View/download PDF
11. mTORC2-mediated PDHE1α nuclear translocation links EBV-LMP1 reprogrammed glucose metabolism to cancer metastasis in nasopharyngeal carcinoma.
- Author
-
Zhang J, Jia L, Liu T, Yip YL, Tang WC, Lin W, Deng W, Lo KW, You C, Lung ML, Lung HL, Cheung AL, Tsao SW, and Tsang CM
- Subjects
- Active Transport, Cell Nucleus genetics, Animals, Cell Proliferation genetics, Cell Transformation, Viral physiology, Cells, Cultured, Epstein-Barr Virus Infections complications, Epstein-Barr Virus Infections genetics, Epstein-Barr Virus Infections metabolism, Glycolysis genetics, Herpesvirus 4, Human physiology, Humans, Male, Mice, Mice, Inbred NOD, Mice, Nude, Mice, SCID, Nasopharyngeal Carcinoma genetics, Nasopharyngeal Carcinoma metabolism, Nasopharyngeal Carcinoma virology, Nasopharyngeal Neoplasms genetics, Nasopharyngeal Neoplasms metabolism, Nasopharyngeal Neoplasms virology, Neoplasm Metastasis, Protein Transport, Pyruvate Dehydrogenase (Lipoamide) genetics, Signal Transduction genetics, Cell Nucleus metabolism, Glucose metabolism, Mechanistic Target of Rapamycin Complex 2 physiology, Nasopharyngeal Carcinoma pathology, Nasopharyngeal Neoplasms pathology, Pyruvate Dehydrogenase (Lipoamide) metabolism, Viral Matrix Proteins physiology
- Abstract
EBV infection of preinvasive nasopharyngeal epithelium is believed to be an initiation step during pathogenesis of nasopharyngeal carcinoma (NPC), but the mechanisms remain poorly understood. Here we report a novel mechanism driving NPC metastasis through the EBV-encoded LMP1-mediated metabolic reprogramming, via activation of IGF1-mTORC2 signaling and nuclear acetylation of the Snail promoter by the PDHE1α, an enzyme involved in glucose metabolism. Mechanistically, EBV-LMP1 increases the cellular secretion of IGF1 which promotes phosphorylation of IGF1R to activate mTORC2/AKT signaling linking glucose metabolism to cell motility. LMP1 expression facilitates translocation of mitochondrial PDHE1α into the nucleus in a phosphorylation-dependent manner at Ser
293 residue. Functionally, nuclear PDHE1α promotes H3K9 acetylation on the Snail promoter to enhance cell motility, thereby driving cancer metastasis. Importantly, the IGF1/mTORC2/PDHE1α/Snail axis correlates significantly with disease progression and poor prognosis in NPC patients. This study highlights the functional importance of IGF1-mTORC2-PDHE1α signaling mediated by EBV-LMP1 in NPC pathogenesis.- Published
- 2019
- Full Text
- View/download PDF
12. Long noncoding RNA CAR10 promotes lung adenocarcinoma metastasis via miR-203/30/SNAI axis.
- Author
-
Ge X, Li GY, Jiang L, Jia L, Zhang Z, Li X, Wang R, Zhou M, Zhou Y, Zeng Z, Xiang J, and Li Z
- Subjects
- A549 Cells, Animals, Cell Line, Cell Line, Tumor, Chromatin genetics, Epithelial-Mesenchymal Transition genetics, Female, Gene Expression Regulation, Neoplastic genetics, HEK293 Cells, HeLa Cells, Hep G2 Cells, Humans, Lung Neoplasms genetics, MCF-7 Cells, Melanoma, Experimental genetics, Mice, Inbred BALB C, Mice, Nude, Prognosis, Up-Regulation genetics, Adenocarcinoma of Lung genetics, MicroRNAs genetics, RNA, Long Noncoding genetics, Snail Family Transcription Factors genetics
- Abstract
Long noncoding RNAs (lncRNAs) play an important role in lung adenocarcinoma (LUAD) metastasis. Here, we found that lncRNA chromatin-associated RNA 10 (CAR10) was upregulated in the tumor tissue of patients with LUAD and enhanced tumor metastasis in vitro and in vivo. Mechanistically, CAR10 induced epithelial-to-mesenchymal transition (EMT) by directly binding with miR-30 and miR-203 and then regulating the expression of SNAI1 and SNAI2. CAR10 overexpression was positively correlated with a poor prognosis in LUAD patients, whereas overexpression of both CAR10 and SNAI was correlated with even worse clinical outcomes. In conclusion, the CAR10/miR-30/203/SNAI axis is a novel and potential therapeutic target for LUAD.
- Published
- 2019
- Full Text
- View/download PDF
13. Orphan nuclear receptor TLX contributes to androgen insensitivity in castration-resistant prostate cancer via its repression of androgen receptor transcription.
- Author
-
Jia L, Wu D, Wang Y, You W, Wang Z, Xiao L, Cai G, Xu Z, Zou C, Wang F, Teoh JY, Ng CF, Yu S, and Chan FL
- Subjects
- Androgen Antagonists pharmacology, Animals, Apoptosis, Biomarkers, Tumor genetics, Cell Proliferation, Humans, Male, Mice, Mice, SCID, Orphan Nuclear Receptors, Prognosis, Prostatic Neoplasms, Castration-Resistant drug therapy, Prostatic Neoplasms, Castration-Resistant genetics, Receptors, Androgen chemistry, Receptors, Androgen metabolism, Receptors, Cytoplasmic and Nuclear genetics, Transcriptional Activation, Tumor Cells, Cultured, Xenograft Model Antitumor Assays, Biomarkers, Tumor metabolism, Gene Expression Regulation, Neoplastic, Prostatic Neoplasms, Castration-Resistant pathology, Receptors, Androgen genetics, Receptors, Cytoplasmic and Nuclear metabolism, Transcription, Genetic
- Abstract
The metastatic castration-resistant prostate cancer (CRPC) is a lethal form of prostate cancer, in which the expression of androgen receptor (AR) is highly heterogeneous. Indeed, lower AR expression and attenuated AR signature activity is shown in CRPC tissues, especially in the subset of neuroendocrine prostate cancer (NEPC) and prostate cancer stem-like cells (PCSCs). However, the significance of AR downregulation in androgen insensitivity and de-differentiation of tumor cells in CRPC is poorly understood and much neglected. Our previous study shows that the orphan nuclear receptor TLX (NR2E1), which is upregulated in prostate cancer, plays an oncogenic role in prostate carcinogenesis by suppressing oncogene-induced senescence. In the present study, we further established that TLX exhibited an increased expression in metastatic CRPC. Further analyses showed that overexpression of TLX could confer resistance to androgen deprivation and anti-androgen in androgen-dependent prostate cancer cells in vitro and in vivo, whereas knockdown of endogenous TLX could potentiate the sensitivity to androgen deprivation and anti-androgen in prostate cancer cells. Our study revealed that the TLX-induced resistance to androgen deprivation and anti-androgen was mediated through its direct suppression of AR gene transcription and signaling in both androgen-stimulated and -unstimulated prostate cancer cells. We also characterized that TLX could bind directly to AR promoter and repress AR transcription by recruitment of histone modifiers, including HDAC1, HDAC3, and LSD1. Together, our present study shows, for the first time, that TLX can contribute to androgen insensitivity in CRPC via repression of AR gene transcription and signaling, and also implicates that targeting the druggable TLX may have a potential therapeutic significance in CRPC management, particularly in NEPC and PCSCs.
- Published
- 2018
- Full Text
- View/download PDF
14. Lung fibroblasts promote metastatic colonization through upregulation of stearoyl-CoA desaturase 1 in tumor cells.
- Author
-
Liu G, Feng S, Jia L, Wang C, Fu Y, and Luo Y
- Subjects
- Animals, Cells, Cultured, Fibroblasts metabolism, Fibroblasts pathology, Gene Expression Regulation, Neoplastic, HEK293 Cells, Humans, Lung Neoplasms genetics, Lung Neoplasms metabolism, Mice, Mice, Inbred BALB C, Mice, Inbred C57BL, Mice, Nude, Neoplasms genetics, Neoplasms metabolism, Stearoyl-CoA Desaturase metabolism, Tumor Microenvironment genetics, Up-Regulation genetics, Cell Proliferation genetics, Fibroblasts physiology, Lung pathology, Lung Neoplasms secondary, Neoplasms pathology, Stearoyl-CoA Desaturase genetics
- Abstract
As a rate-limiting step in metastasis, metastatic colonization requires survival signals from supportive stroma. However, the mechanisms driving this process are incompletely understood. Here, we showed that the proliferation of B16F10 cells was promoted when cocultured with lung fibroblasts. Meanwhile, co-injection of B16F10 tumor cells with mouse lung fibroblasts significantly increased lung metastasis. Based on GEO database, we identified stearoyl-CoA desaturase 1 (SCD1) as a novel factor promoting metastatic colonization. Importantly, we found that fibroblast-secreted cathepsin B (CTSB) induced the upregulation of SCD1 in B16F10 through Annexin A2 (ANXA2) and PI3K/Akt/mTOR pathway. The elevated SCD1 induced a higher ratio of monounsaturated fatty acids to saturated fatty acids in B16F10 cells. The changes in fatty acid composition contributed to tumor cell proliferation and metastatic colonization. Furthermore, targeting SCD1 effectively inhibited lung metastasis and prolonged the overall survival of mice. Meanwhile, the expression of SCD1 was negatively correlated with disease-free survival in five types of cancer patients. Collectively, our study identifies SCD1 as a critical modulator of tumor cell proliferation that is activated by cathepsin B, secreted by lung fibroblasts at the metastatic niche. Our novel findings provide potential therapeutic targets to prevent tumor metastasis.
- Published
- 2018
- Full Text
- View/download PDF
15. Role of Smac in human leukaemic cell apoptosis and proliferation.
- Author
-
Jia L, Patwari Y, Kelsey SM, Srinivasula SM, Agrawal SG, Alnemri ES, and Newland AC
- Subjects
- Apoptosis radiation effects, Apoptosis Regulatory Proteins, Apoptotic Protease-Activating Factor 1, Carrier Proteins genetics, Caspases metabolism, Cytochrome c Group metabolism, Enzyme Activation, Humans, Intracellular Signaling Peptides and Proteins, Leukemia enzymology, Mitochondrial Proteins genetics, Proteins metabolism, Recombinant Proteins metabolism, Transfection, Tumor Cells, Cultured, X-Linked Inhibitor of Apoptosis Protein, Apoptosis physiology, Carrier Proteins physiology, Cell Division physiology, Leukemia pathology, Mitochondrial Proteins physiology
- Abstract
Smac (or DIABLO) is a recently identified, novel proapoptotic molecule, which is released from mitochondria into the cytosol during apoptosis. Smac functions by eliminating the caspase-inhibitory properties of the inhibitors of apoptosis proteins (IAP), particularly XIAP. In this study, we stably transfected both full-length (FL) and mature (MT) Smac genes into the K562 and CEM leukaemic cell lines. Both FL and MT Smac transfectants increased the sensitivity of leukaemic cells to UV light-induced apoptosis and the activation of caspase-9 and caspase-3. Purified cytosol from the mature Smac transfectants, or the addition of human recombinant Smac protein or N-7 peptide into nontransfected cytosol, showed an increased sensitivity to cytochrome c-induced activation of caspase-3. The mature Smac enhanced the susceptibility of both K562 and CEM cells to TRAIL-induced apoptosis. Overexpression of the mature Smac protein also inhibited proliferation, as detected by reduced colony formation and Ki-67 expression in leukaemic cells. Cell cycle analysis revealed that Smac transfectants displayed significant G0/G1 arrest and reduction in 5-bromo-2'-deoxyuridine (BrdU) incorporation. Smac sensitized human acute myeloid leukaemia blasts to cytochrome c-induced activation of caspase-3. However, Smac failed to overcome Apaf-1-deficiency-mediated resistance to cytochrome c in primary leukaemic blasts. In summary, this study reveals that Smac/DIABLO exhibits a potential role in increasing apoptosis and suppressing proliferation in human leukaemic cells. Importantly, it also indicates that it is crucial to evaluate the levels of Apaf-1 and XIAP proteins in patient samples before using Smac peptide therapy in the treatment of human leukaemia.
- Published
- 2003
- Full Text
- View/download PDF
16. Role of DNA methylation in the suppression of Apaf-1 protein in human leukaemia.
- Author
-
Fu WN, Bertoni F, Kelsey SM, McElwaine SM, Cotter FE, Newland AC, and Jia L
- Subjects
- Apoptosis drug effects, Apoptosis genetics, Apoptosis radiation effects, Apoptotic Protease-Activating Factor 1, Azacitidine pharmacology, Cytosine metabolism, DNA Methylation, Decitabine, Humans, Leukemia pathology, Leukemia therapy, Polymerase Chain Reaction methods, Promoter Regions, Genetic, Proteins genetics, Reference Values, Sequence Analysis, DNA methods, Tumor Cells, Cultured, Ultraviolet Rays, Azacitidine analogs & derivatives, Leukemia genetics, Proteins metabolism
- Abstract
Apaf-1 protein deficiency occurs in human leukaemic blasts and confers resistance to cytochrome-c-dependent apoptosis. Demethylation treatment with 5-aza-2'-deoxycytidine (5aza2dc) increased the sensitivity of the K562 leukaemic cell line to UV light-induced apoptosis in association with increased Apaf-1 protein levels. There was no correlation between Apaf-1 protein expression and Apaf-1 mRNA levels after the demethylation treatment. Methylation-specific polymerase chain reaction was used to show that the methylation can occur within the Apaf-1 promoter region in leukaemic blasts. Apaf-1 DNA methylation was demonstrated in acute myeloid leukaemia, chronic myeloid leukaemia and acute lymphoid leukaemia, suggesting that it is not specific to a particular leukaemia subtype. Apaf-1 protein expression did not correlate with Apaf-1 mRNA levels in human leukaemic blasts. Some leukaemic cells expressed high levels of Apaf-1 mRNA but low levels of Apaf-1 protein. This study suggests that Apaf-1 DNA promoter methylation might contribute to the inactivation of Apaf-1 expression. However, Apaf-1 protein levels might also be controlled at post-transcription level.
- Published
- 2003
- Full Text
- View/download PDF
17. Bax translocation is crucial for the sensitivity of leukaemic cells to etoposide-induced apoptosis.
- Author
-
Jia L, Patwari Y, Srinivasula SM, Newland AC, Fernandes-Alnemri T, Alnemri ES, and Kelsey SM
- Subjects
- Apoptotic Protease-Activating Factor 1, Biological Transport, Caspases physiology, DNA Damage, Humans, K562 Cells, Leukemia pathology, Proteins physiology, bcl-2-Associated X Protein, Antineoplastic Agents, Phytogenic pharmacology, Apoptosis drug effects, Etoposide pharmacology, Leukemia drug therapy, Proto-Oncogene Proteins metabolism, Proto-Oncogene Proteins c-bcl-2
- Abstract
Bax translocation from cytosol to mitochondria is believed to be a crucial step for triggering cytochrome c release from mitochondria. However, it is unclear whether Bax translocation is associated with Bax induction by DNA damaging agents. The induction of Bax in response to DNA damaging agents has been considered to be linked with p53. In this study, we used the p53 negative human chronic myeloid leukaemia K562 cell line. Bax up-regulation occurred at the whole cell level after DNA damage induced by etoposide. However, after incubation with etoposide, Bax failed to translocate to mitochondria and as a result, the apoptotic process was blocked. A Bax stable transfectant, the K/Bax cell line, expressed more Bax protein in the cytosol, mitochondria and nuclei. This Bax overexpression induced cytochrome c release, a reduction of cytochrome c oxidase activity and mitochondrial membrane potential (Delta(Psi)m). However, Bax-induced apoptosis was blocked downstream of mitochondria in K562 cells. The increased levels of mitochondrial Bax sensitized cells to etoposide-induced activation of caspases-2, -3 and -9 and apoptosis. However, after transient transfection with the Apaf-1 gene, K/Bax cells were sensitized to etoposide-induced caspase activation and apoptosis to a larger extent compared with Bax or Apaf-1 transfection alone. We therefore conclude that two mechanisms contribute to the resistance of K562 cells to etoposide-induced apoptosis; firstly failure of Bax targeting to mitochondria and, secondly, deficiency of Apaf-1. Uncoupling of Bax translocation from Bax induction can occur in response to etoposide-induced DNA damage.
- Published
- 2001
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.