1
|
Clathrin- and dynamin-dependent endocytosis limits canonical NF-κB signaling triggered by lymphotoxin β receptor. Cell Commun Signal 2020; 18:176. [PMID: 33148272 PMCID: PMC7640449 DOI: 10.1186/s12964-020-00664-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 09/18/2020] [Indexed: 02/08/2023] Open
Abstract
Background Lymphotoxin β receptor (LTβR) is a member of tumor necrosis factor receptor (TNFR) superfamily which regulates the immune response. At the cellular level, upon ligand binding, the receptor activates the pro-inflammatory NF-κB and AP-1 pathways. Yet, the intracellular distribution of LTβR, the routes of its endocytosis and their connection to the signaling activation are not characterized. Here, we investigated the contribution of LTβR internalization to its signaling potential. Methods Intracellular localization of LTβR in unstimulated and stimulated cells was analyzed by confocal microscopy. Endocytosis impairment was achieved through siRNA- or CRISPR/Cas9-mediated depletion, or chemical inhibition of proteins regulating endocytic routes. The activation of LTβR-induced signaling was examined. The levels of effector proteins of the canonical and non-canonical branches of the NF-κB pathway, and the phosphorylation of JNK, Akt, ERK1/2, STAT1 and STAT3 involved in diverse signaling cascades, were measured by Western blotting. A transcriptional response to LTβR stimulation was assessed by qRT-PCR analysis. Results We demonstrated that LTβR was predominantly present on endocytic vesicles and the Golgi apparatus. The ligand-bound pool of the receptor localized to endosomes and was trafficked towards lysosomes for degradation. Depletion of regulators of different endocytic routes (clathrin-mediated, dynamin-dependent or clathrin-independent) resulted in the impairment of LTβR internalization, indicating that this receptor uses multiple entry pathways. Cells deprived of clathrin and dynamins exhibited enhanced activation of canonical NF-κB signaling represented by increased degradation of IκBα inhibitor and elevated expression of LTβR target genes. We also demonstrated that clathrin and dynamin deficiency reduced to some extent LTβR-triggered activation of the non-canonical branch of the NF-κB pathway. Conclusions Our work shows that the impairment of clathrin- and dynamin-dependent internalization amplifies a cellular response to LTβR stimulation. We postulate that receptor internalization restricts responsiveness of the cell to subthreshold stimuli. Video Abstract
Graphical abstract ![]()
Supplementary information Supplementary information accompanies this paper at 10.1186/s12964-020-00664-0.
Collapse
|
2
|
Zheng P, Chen X, Xie J, Chen X, Lin S, Ye L, Chen L, Lin J, Yu X, Zheng M. Capn4 is induced by and required for Epstein-Barr virus latent membrane protein 1 promotion of nasopharyngeal carcinoma metastasis through ERK/AP-1 signaling. Cancer Sci 2019; 111:72-83. [PMID: 31691433 PMCID: PMC6942433 DOI: 10.1111/cas.14227] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 10/22/2019] [Accepted: 10/31/2019] [Indexed: 12/11/2022] Open
Abstract
Capn4, also known as CapnS1, is a member of the calpain family, which plays a crucial role in maintaining the activity and function of calpain. We previously reported that Capn4 also plays an essential role in the migration of nasopharyngeal carcinoma (NPC) cells through regulation of (MMP‐2) by nuclear factor‐kappa B activation. Epstein‐Barr virus latent membrane protein 1 (LMP1) is closely related to the malignant functions of NPC; however, the relationship between LMP1 and Capn4 in NPC remain unclear. Immunohistochemical studies showed that the level of LMP1 and Capn4 expression was high in both primary and metastatic NPC tissues, with a significantly positive correlation. We further found that LMP1 was able to upregulate the Capn4 promoter in a dose‐dependent way through the C‐terminal activation region (CTAR)1 and CTAR2 domains to activate AP‐1. Moreover, we also found that LMP1 activated AP‐1 through ERK/JNK phosphorylation. These findings indicate that Capn4 coordination with LMP1 promotes actin rearrangement and, ultimately, cellular migration. These results show that Capn4 coordination with LMP1 enhances NPC migration by increasing actin rearrangement involving ERK/JNK/AP‐1 signaling. Therapeutically, additional and more specific LMP1 and Capn4 targeted inhibitors could be exploited to treat NPC.
Collapse
Affiliation(s)
- Peichan Zheng
- Fujian Center for Safety Evaluation of New Drug, Fujian Medical University, Fuzhou, China
| | - Xiong Chen
- Department of Medical Oncology, The 900th Hospital of the People's Liberation Army Joint Service Support Force, Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Jianqin Xie
- Fujian Center for Safety Evaluation of New Drug, Fujian Medical University, Fuzhou, China
| | - Xi Chen
- Department of Medical Oncology, The 900th Hospital of the People's Liberation Army Joint Service Support Force, Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Shanshan Lin
- Fujian Center for Safety Evaluation of New Drug, Fujian Medical University, Fuzhou, China
| | - Lixiang Ye
- Fujian Center for Safety Evaluation of New Drug, Fujian Medical University, Fuzhou, China
| | - Lingfan Chen
- Fujian Center for Safety Evaluation of New Drug, Fujian Medical University, Fuzhou, China
| | - Jing Lin
- Fujian Center for Safety Evaluation of New Drug, Fujian Medical University, Fuzhou, China
| | - Xiangbin Yu
- Fujian Center for Safety Evaluation of New Drug, Fujian Medical University, Fuzhou, China
| | - Ming Zheng
- College of Pharmacy, Fujian Medical University, Fuzhou, China
| |
Collapse
|
3
|
Prostate tumor overexpressed-1, in conjunction with human papillomavirus status, predicts outcome in early-stage human laryngeal squamous cell carcinoma. Oncotarget 2017; 7:31878-91. [PMID: 26992242 PMCID: PMC5077983 DOI: 10.18632/oncotarget.8103] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Accepted: 03/01/2016] [Indexed: 12/14/2022] Open
Abstract
In human cancer, molecular markers combined with clinical characteristics are used increasingly to predict prognosis. Prostate tumor overexpressed-1 (PTOV1), first identified in prostate cancer, is a key factor in tumor progression and correlates with unfavorable clinical outcomes. HPV infection status was tested by HPV E6-targeted multiplex real-time PCR and p16 immunohistochemistry (IHC). Real-time PCR and western blotting analyses were used to examine the mRNA and protein expression levels of PTOV1 in eight paired LSCC samples. IHC was performed to assess PTOV1 protein expression in 196 paraffin-embedded, archived LSCC samples. PTOV1 protein and mRNA expression was increased in LSCC tissues compared with adjacent noncancerous tissue samples. High expression of PTOV1was significantly associated with advanced TNM stage by the X2 test. Multivariate analysis revealed that PTOV1 and HPV status were independent prognostic indicators of overall survival (OS) and progression-free survival (PFS) (P = 0.001, P = 0.009 for OS, P = 0.005, P = 0.012 for PFS, respectively). Our study provides the first evidence that the combination of PTOV1 expression level and HPV status provides more prognostic information compared with HPV status alone with the significance still exists in the HPV negative subgroup.
Collapse
|
4
|
Li R, Leng AM, Liu XM, Hu TZ, Zhang LF, Li M, Jiang XX, Zhou YW, Xu CX. Overexpressed PTOV1 associates with tumorigenesis and progression of esophageal squamous cell carcinoma. Tumour Biol 2017. [PMID: 28651486 DOI: 10.1177/1010428317705013] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
PTOV1 has been demonstrated to play an extensive role in many types of cancers. This study takes the first step to clarify the potential relationship between esophageal squamous cell carcinoma and PTOV1 expression and highlight the link between PTOV1 and the tumorigenesis, progression, and prognosis of esophageal squamous cell carcinoma. PTOV1 expression was detected by quantitative reverse transcription polymerase chain reaction and western blotting or immunohistochemical staining in esophageal squamous cell carcinoma cell lines, esophageal squamous cell carcinoma tissues, and its paired adjacent non-cancerous tissues. Moreover, we have analyzed the relationship between PTOV1 expression and clinicopathological features of esophageal squamous cell carcinoma. Survival analysis and Cox regression analysis were used to assess its prognostic significance. We found that PTOV1 expression was significantly higher in the esophageal squamous cell carcinoma cell lines and tissues at messenger RNA level (p < 0.001) and protein level (p < 0.001). Gender, tumor size, or differentiation was tightly associated with the PTOV1 expression. Lymph node involvement (p < 0.001) and TNM stage (p < 0.001) promoted a high PTOV1 expression. A prognostic significance of PTOV1 was also found by Log-rank method, and the overexpression of PTOV1 was related to a shorter OS and DFS. Multiple Cox regression analysis indicated overexpressed PTOV1 as an independent indicator for adverse prognosis. In conclusion, this study takes the lead to demonstrate that the overexpressed PTOV1 plays a vital role in the tumorigenesis and progression of esophageal squamous cell carcinoma, and it is potentially a valuable prognostic predicator and new chemotherapeutic target for esophageal squamous cell carcinoma.
Collapse
Affiliation(s)
- Rong Li
- 1 Department of Gastroenterology, Third Xiangya Hospital of Central South University, Changsha, People's Republic of China.,2 Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Changsha, People's Republic of China
| | - Ai-Min Leng
- 3 Department of Gastroenterology, Xiangya Hospital of Central South University, Changsha, People's Republic of China
| | - Xiao-Ming Liu
- 1 Department of Gastroenterology, Third Xiangya Hospital of Central South University, Changsha, People's Republic of China.,2 Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Changsha, People's Republic of China
| | - Ting-Zi Hu
- 1 Department of Gastroenterology, Third Xiangya Hospital of Central South University, Changsha, People's Republic of China.,2 Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Changsha, People's Republic of China
| | - Lin-Fang Zhang
- 1 Department of Gastroenterology, Third Xiangya Hospital of Central South University, Changsha, People's Republic of China.,2 Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Changsha, People's Republic of China
| | - Ming Li
- 1 Department of Gastroenterology, Third Xiangya Hospital of Central South University, Changsha, People's Republic of China.,2 Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Changsha, People's Republic of China
| | - Xiao-Xia Jiang
- 1 Department of Gastroenterology, Third Xiangya Hospital of Central South University, Changsha, People's Republic of China.,2 Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Changsha, People's Republic of China
| | - Yan-Wu Zhou
- 4 Department of Thoracic Surgery, Xiangya Hospital of Central South University, Changsha, People's Republic of China
| | - Can-Xia Xu
- 1 Department of Gastroenterology, Third Xiangya Hospital of Central South University, Changsha, People's Republic of China.,2 Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Changsha, People's Republic of China
| |
Collapse
|
5
|
Wang R, Ding Q, De Assuncao TM, Mounajjed T, Maiers JL, Dou C, Cao S, Yaqoob U, Huebert RC, Shah VH. Hepatic Stellate Cell Selective Disruption of Dynamin-2 GTPase Increases Murine Fibrogenesis through Up-Regulation of Sphingosine-1 Phosphate-Induced Cell Migration. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 187:134-145. [PMID: 27840081 PMCID: PMC5225297 DOI: 10.1016/j.ajpath.2016.09.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Revised: 08/22/2016] [Accepted: 09/01/2016] [Indexed: 01/04/2023]
Abstract
Dynamin-2 (Dyn2) is implicated in endocytosis of receptor tyrosine kinases, which contribute to hepatic stellate cell (HSC) activation and liver fibrosis. A point mutation converting lysine 44 of Dyn2 to alanine (Dyn2K44A) disrupts its GTPase activity. We hypothesized that Dyn2K44A expression in HSCs would decrease HSC activation and fibrogenesis in vivo by disrupting receptor tyrosine kinase endocytosis and signaling. Dyn2K44Afl/fl mice were crossed with Collagen1-Cre (Col1Cre) mice to generate offspring with HSC selective expression of Dyn2K44A (Col1Cre/Dyn2K44Afl/fl). Contrary to our hypothesis, Col1Cre/Dyn2K44Afl/fl mice showed increased hepatic fibrosis in response to liver injury. To elucidate mechanisms, we conducted in vitro experiments with HSCs infected with adenoviral vectors encoding LacZ, Dyn2K44A, or Dyn2WT. HSC-expressing Dyn2K44A displayed increased mRNA and protein levels of sphingosine kinase-1 (SK1), an enzyme previously implicated in the pathogenesis of fibrosis. To study the functional effects of Dyn2K44A regulation of SK1, we examined effects of AKT signaling and migration in HSCs. Dyn2K44A promoted both AKT phosphorylation and HSC migration in an SK1-dependent manner. Genetic disruption of Dyn2 GTPase activity selectively in HSC enhances fibrogenesis, driven at least in part through up-regulation of the SK1 pathway and cell migration in HSCs.
Collapse
Affiliation(s)
- Ruisi Wang
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota
| | - Qian Ding
- Department of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Thiago M De Assuncao
- Department of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Taofic Mounajjed
- Laboratory of Anatomic Pathology, Mayo Clinic, Rochester, Minnesota
| | - Jessica L Maiers
- Department of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Changwei Dou
- Department of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Sheng Cao
- Department of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Usman Yaqoob
- Department of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Robert C Huebert
- Department of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Vijay H Shah
- Department of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota.
| |
Collapse
|
6
|
Chen X, Li J, Cheng Z, Xu Y, Wang X, Li X, Xu D, Kapron CM, Liu J. Low Dose Cadmium Inhibits Proliferation of Human Renal Mesangial Cells via Activation of the JNK Pathway. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2016; 13:ijerph13100990. [PMID: 27739415 PMCID: PMC5086729 DOI: 10.3390/ijerph13100990] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/23/2016] [Accepted: 09/29/2016] [Indexed: 12/15/2022]
Abstract
Cadmium (Cd) is a heavy metal and environmental pollutant. The kidney is the principal target organ of Cd exposure. Previously, we found that low concentration of Cd damages the integrity of the glomerular filtration barrier. However, little is known about the effects of Cd on renal mesangial cells, which provide structural support for the glomerular capillary loops and regulate intraglomerular blood flow. In this study, human renal mesangial cells (HRMCs) were cultured in the presence of serum and treated with 4 μM Cd. We found that Cd activates the c-Jun N-terminal kinase (JNK) pathway, and increases the protein levels of c-Jun and c-Fos. Cd treatment also induces a decrease in proliferation and an increase in apoptosis of HRMCs, but only the decrease in HRMC proliferation was reversed by pretreatment with SP600125, an inhibitor of the JNK pathway. In addition, Cd does not change the expression of α-smooth muscle actin and platelet-derived growth factor receptor-β, the markers of mesangial cells, or the alignment of the filamentous actin (F-actin) cytoskeleton of HRMCs. Our data indicate that the JNK pathway mediates the inhibitory effects of Cd on HRMC proliferation.
Collapse
Affiliation(s)
- Xiaocui Chen
- Medical Research Center, Shandong Provincial Qianfoshan Hospital, Shandong University, 16766 Jingshi Road, Jinan 250014, China.
| | - Jing Li
- Key Laboratory of Molecular and Nano Probes, Ministry of Education, College of Chemistry, Chemical Engineering and Materials Science, Shandong Normal University, Jinan 250014, China.
| | | | - Yinghua Xu
- Taishan Medical College, Taian 271000, China.
| | - Xia Wang
- Medical Research Center, Shandong Provincial Qianfoshan Hospital, Shandong University, 16766 Jingshi Road, Jinan 250014, China.
| | - Xiaorui Li
- Taishan Medical College, Taian 271000, China.
| | - Dongmei Xu
- Department of Nephrology, Shandong Provincial Qianfoshan Hospital, Shandong University, 16766 Jingshi Road, Jinan 250014, China.
| | - Carolyn M Kapron
- Department of Biology, Trent University, Peterborough, ON K9L0G2, Canada.
| | - Ju Liu
- Medical Research Center, Shandong Provincial Qianfoshan Hospital, Shandong University, 16766 Jingshi Road, Jinan 250014, China.
| |
Collapse
|
7
|
Mo EP, Zhang RR, Xu J, Zhang H, Wang XX, Tan QT, Liu FL, Jiang RW, Cai SH. Calotropin from Asclepias curasavica induces cell cycle arrest and apoptosis in cisplatin-resistant lung cancer cells. Biochem Biophys Res Commun 2016; 478:710-5. [PMID: 27498029 DOI: 10.1016/j.bbrc.2016.08.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Accepted: 08/03/2016] [Indexed: 12/14/2022]
Abstract
Calotropin (M11), an active compound isolated from Asclepias curasavica L., was found to exert strong inhibitory and pro-apoptotic activity specifically against cisplatin-induced resistant non-small cell lung cancer (NSCLC) cells (A549/CDDP). Molecular mechanism study revealed that M11 induced cell cycle arrest at the G2/M phase through down-regulating cyclins, CDK1, CDK2 and up-regulating p53 and p21. Furthermore, M11 accelerated apoptosis through the mitochondrial apoptotic pathway which was accompanied by increase Bax/Bcl-2 ratio, decrease in mitochondrial membrane potential, increase in reactive oxygen species production, activations of caspases 3 and 9 as well as cleavage of poly ADP-ribose polymerase (PARP). The activation and phosphorylation of JNK was also found to be involved in M11-induced apoptosis, and SP610025 (specific JNK inhibitor) partially prevented apoptosis induced by M11. In contrast, all of the effects that M11 induce cell cycle arrest and apoptosis in A549/CDDP cells were not significant in A549 cells. Drugs with higher sensitivity against resistant tumor cells than the parent cells are rather rare. Results of this study supported the potential application of M11 on the non-small lung cancer (NSCLC) with cisplatin resistance.
Collapse
Affiliation(s)
- En-Pan Mo
- College of Pharmacy, Jinan University, Guangzhou, 510632, PR China
| | - Rong-Rong Zhang
- College of Pharmacy, Jinan University, Guangzhou, 510632, PR China
| | - Jun Xu
- College of Pharmacy, Jinan University, Guangzhou, 510632, PR China
| | - Huan Zhang
- College of Pharmacy, Jinan University, Guangzhou, 510632, PR China
| | - Xiao-Xiong Wang
- College of Pharmacy, Jinan University, Guangzhou, 510632, PR China
| | - Qiu-Tong Tan
- College of Pharmacy, Jinan University, Guangzhou, 510632, PR China
| | - Fang-Lan Liu
- College of Pharmacy, Jinan University, Guangzhou, 510632, PR China
| | - Ren-Wang Jiang
- College of Pharmacy, Jinan University, Guangzhou, 510632, PR China.
| | - Shao-Hui Cai
- College of Pharmacy, Jinan University, Guangzhou, 510632, PR China.
| |
Collapse
|