1
|
Verma A, Sumi S, Seervi M. Heat shock proteins-driven stress granule dynamics: yet another avenue for cell survival. Apoptosis 2021; 26:371-384. [PMID: 33978921 DOI: 10.1007/s10495-021-01678-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/04/2021] [Indexed: 12/24/2022]
Abstract
Heat shock proteins (HSPs) are evolutionary conserved 'stress-response' proteins that facilitate cell survival against various adverse conditions. HSP-mediated cytoprotection was hitherto reported to occur principally in two ways. Firstly, HSPs interact directly or indirectly with apoptosis signaling components and suppress apoptosis. Secondly, through chaperon activity, HSPs suppress proteotoxicity and maintain protein-homeostasis. Recent studies highlight the interaction of HSPs with cytoplasmic stress granules (SGs). SGs are conserved cytoplasmic mRNPs granules that aid in cell survival under stressful conditions. We primarily aim to describe the distinct cell survival strategy mediated by HSPs as the crucial regulators of SGs assembly and disassembly. Based on the growing evidence, HSPs and associated co-chaperones act as important determinants of SG assembly, composition and dissolution. Under cellular stress, as a 'stress-coping mechanism', the formation of SGs reprograms protein translation machinery and modulates signaling pathways indispensable for cell survival. Besides their role in suppressing apoptosis, HSPs also regulate protein-homeostasis by their chaperone activity as well as by their tight regulation of SG dynamics. The intricate molecular signaling in and around the nexus of HSPs-SGs and its importance in diseases has to be unearthed. These studies have significant implications in the management of chronic diseases such as cancer and neurodegenerative diseases where SGs possess pathological functions.
Collapse
Affiliation(s)
- Akanksha Verma
- Department of Biotechnology, All India Institute of Medical Sciences (AIIMS), New Delhi, 110029, India
| | - S Sumi
- Cardiovascular Diseases and Diabetes Biology, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram, Kerala, India
| | - Mahendra Seervi
- Department of Biotechnology, All India Institute of Medical Sciences (AIIMS), New Delhi, 110029, India.
| |
Collapse
|
2
|
Forouzanfar F, Barreto G, Majeed M, Sahebkar A. Modulatory effects of curcumin on heat shock proteins in cancer: A promising therapeutic approach. Biofactors 2019; 45:631-640. [PMID: 31136038 DOI: 10.1002/biof.1522] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 05/12/2019] [Indexed: 12/23/2022]
Abstract
Cancer metastasis represents a multistep process, including alteration of cell adhesion/motility in the microenvironment and sustained angiogenesis, which is essential for supporting cancer growth in tissues that are distant from the primary tumor. There is growing evidence suggesting that heat shock proteins (HSPs) (also known as heat stress proteins), which constitute a family of stress-inducible proteins, may be involved in the pathogenesis of cancer. Curcumin (diferuloylmethane) is a potent anti-inflammatory, antioxidant, antimicrobial, and antitumor agent. Curcumin has been shown to regulate different members of HSPs including HSP27, HSP40, HSP60, HSP70, and HSP90 in cancer. Here, we present extent findings suggesting that curcumin may act as a potential therapeutic agent for the treatment of cancer through its regulation of HSPs.
Collapse
Affiliation(s)
- Fatemeh Forouzanfar
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Neuroscience, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - George Barreto
- Departamento de Nutrición yBioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, D.C., Colombia
- Instituto de Ciencias Biomédicas, Universidad Autónoma de Chile, Santiago, Chile
| | | | - Amirhossein Sahebkar
- Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Medical Biotechnology,School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
3
|
Glucocorticoid modulatory element-binding protein 1 (GMEB1) interacts with the de-ubiquitinase USP40 to stabilize CFLAR L and inhibit apoptosis in human non-small cell lung cancer cells. J Exp Clin Cancer Res 2019; 38:181. [PMID: 31046799 PMCID: PMC6498657 DOI: 10.1186/s13046-019-1182-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 04/17/2019] [Indexed: 02/05/2023] Open
Abstract
Background GMEB1 was originally identified via its interaction with GMEB2, which binds to the promoter region of the tyrosine aminotransferase (TAT) gene and modulates transactivation of the glucocorticoid receptor gene. In the cytosol, GMEB1 interacts with and inhibits CASP8, but the molecular mechanism is currently unknown. Methods Human non-small cell lung cancer cells and 293FT cells were used to investigate the function of GMEB1/USP40/CFLARL complex by WB, GST Pull-Down Assay, Immunoprecipitation, Immunofluorescence and Flow cytometry analysis. A549 cells overexpressing green fluorescent protein and GMEB1 shRNA were used for tumor xenograft using female athymic nu/nu 4-week-old mice. Results We found GMEB1 interacted with CFLARL (also known as c-FLIPL) in the cytosol and promoted its stability. USP40 targeted CFLARL for K48-linked de-ubiquitination. GMEB1 promoted the binding of USP40 to CFLARL. USP40 knockdown did not increase CFLARL protein level despite GMEB1 overexpression, suggesting GMEB1 promotes CFLARL stability via USP40. Additionally, GMEB1 inhibited the activation of pro-caspase 8 and apoptosis in non-small cell lung cancer (NSCLC) cell via CFLARL stabilization. Also, GMEB1 inhibited the formation of DISC upon TRAIL activation. CFLARL enhanced the binding of GMEB1 and CASP8. Downregulation of GMEB1 inhibited A549 xenograft tumor growth in vivo. Conclusions Our findings show the de-ubiquitinase USP40 regulates the ubiquitination and degradation of CFLARL; and GMEB1 acts as a bridge protein for USP40 and CFLARL. Mechanistically, we found GMEB1 inhibits the activation of CASP8 by modulating ubiquitination and degradation of CFLARL. These findings suggest a novel strategy to induce apoptosis through CFLARL targeting in human NSCLC cells. Electronic supplementary material The online version of this article (10.1186/s13046-019-1182-3) contains supplementary material, which is available to authorized users.
Collapse
|
4
|
Li H, Liu Y, Wen M, Zhao F, Zhao Z, Liu Y, Lin X, Wang L. Hydroxysafflor yellow A (HSYA) alleviates apoptosis and autophagy of neural stem cells induced by heat stress via p38 MAPK/MK2/Hsp27-78 signaling pathway. Biomed Pharmacother 2019; 114:108815. [PMID: 30954890 DOI: 10.1016/j.biopha.2019.108815] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Revised: 03/22/2019] [Accepted: 03/26/2019] [Indexed: 01/01/2023] Open
Abstract
This study aimed to explore mechanisms of the effects of hydroxysafflor yellow A (HSYA) on neural stem cells (NSCs) after heat stress (HS). Rat NSCs cells were cultured at 42 °C to impose heat stress. Cell counting kit-8 and Edu assay were used to analyze NSC proliferation. Annexin V/PI apoptosis kit was used to detect NSC apoptosis. Expression and phosphorylation of autophagy and apoptosis-associated proteins were determined by western blotting. We showed that HSYA significantly promoted proliferation and attenuated apoptosis of NSCs after heat stress. HSYA also increased Bcl-2 expression but decreased the expression of Bax and cleaved caspase-3 in NSCs induced by heat stress. In addition, HSYA decreased p38 and Hsp27-78 phosphorylation and MK-2 expression after heat stress, which was consistent with NSCs treated with SB203850 treatment or p38 knockdown. Furthermore, we demonstrated that heat stress increased LC3-II expression and mTOR phosphorylation, and decreased the expression of p62 in NSCs, while HSYA, SB203850 treatment or p38 knockdown reversed these alterations. In conclusion, HSYA significantly reversed the apoptosis and autophagy of NSCs induced by heat stress (P < 0.05), via downregulating MK2 expression and p38 and Hsp27-78 phosphorylation.
Collapse
Affiliation(s)
- Hongbo Li
- Department of Intensive Care Unit, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Yanan Liu
- Department of Intensive Care Unit, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Minyong Wen
- Department of Intensive Care Unit, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Fu Zhao
- Department of Intensive Care Unit, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Zhihui Zhao
- Department of Traditional Chinese Medicine Surgery, Jilin People's Hospital, Jilin, 132000, China
| | - Yunsong Liu
- Intensive Care Unit, Clifford Hospital, Guangzhou University of Chinese Medicine, No.3 Hongfu Road, Panyu District, Guangzhou 511495, PR China.
| | - Xinfeng Lin
- Department of Intensive Care Unit, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
| | - Lin Wang
- Department of Emergency, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, No. 16 Jichang Road, Baiyun District, Guangzhou, 510405, China.
| |
Collapse
|
5
|
TGF-β downregulation-induced cancer cell death is finely regulated by the SAPK signaling cascade. Exp Mol Med 2018; 50:1-19. [PMID: 30523245 PMCID: PMC6283885 DOI: 10.1038/s12276-018-0189-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2018] [Revised: 09/06/2018] [Accepted: 09/11/2018] [Indexed: 02/08/2023] Open
Abstract
Transforming growth factor (TGF)-β signaling is increasingly recognized as a key driver in cancer. In progressive cancer tissues, TGF-β promotes tumor formation, and its increased expression often correlates with cancer malignancy. In this study, we utilized adenoviruses expressing short hairpin RNAs against TGF-β1 and TGF-β2 to investigate the role of TGF-β downregulation in cancer cell death. We found that the downregulation of TGF-β increased the phosphorylation of several SAPKs, such as p38 and JNK. Moreover, reactive oxygen species (ROS) production was also increased by TGF-β downregulation, which triggered Akt inactivation and NOX4 increase-derived ROS in a cancer cell-type-specific manner. We also revealed the possibility of substantial gene fluctuation in response to TGF-β downregulation related to SAPKs. The expression levels of Trx and GSTM1, which encode inhibitory proteins that bind to ASK1, were reduced, likely a result of the altered translocation of Smad complex proteins rather than from ROS production. Instead, both ROS and ROS-mediated ER stress were responsible for the decrease in interactions between ASK1 and Trx or GSTM1. Through these pathways, ASK1 was activated and induced cytotoxic tumor cell death via p38/JNK activation and (or) induction of ER stress. Reducing the levels of the multifunctional protein transforming growth factor (TGF)-β in cancer cells prevents tumor growth in mice. Previous studies have shown that high levels of TGF-β in cancerous tissue are associated with accelerated disease progression. Hye Jin Choi and Jae J Song at Yonsei University in Seoul, South Korea, and colleagues infected cancer cells with genetically modified viruses that reduced the expression of the gene encoding TGF-β. The resulting decrease in TGF-β protein led to cell death by stimulating the production of reactive oxygen species and signaling through the apoptosis signal-regulating kinase 1 (ASK1) pathway. When tumor-bearing mice were infected with these modified viruses, their overall survival was improved. Further understanding the mechanisms through which TGF-β regulates cancer cell survival will contribute to the development of new approaches in cancer treatment.
Collapse
|
6
|
Park SH, Lee DH, Kim JL, Kim BR, Na YJ, Jo MJ, Jeong YA, Lee SY, Lee SI, Lee YY, Oh SC. Metformin enhances TRAIL-induced apoptosis by Mcl-1 degradation via Mule in colorectal cancer cells. Oncotarget 2018; 7:59503-59518. [PMID: 27517746 PMCID: PMC5312327 DOI: 10.18632/oncotarget.11147] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2015] [Accepted: 07/06/2016] [Indexed: 01/09/2023] Open
Abstract
Metformin is an anti-diabetic drug with a promising anti-cancer potential. In this study, we show that subtoxic doses of metformin effectively sensitize human colorectal cancer (CRC) cells to tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL), which induces apoptosis. Metformin alone did not induce apoptosis, but significantly potentiated TRAIL-induced apoptosis in CRC cells. CRC cells treated with metformin and TRAIL showed activation of the intrinsic and extrinsic pathways of caspase activation. We attempted to elucidate the underlying mechanism, and found that metformin significantly reduced the protein levels of myeloid cell leukemia 1 (Mcl-1) in CRC cells and, the overexpression of Mcl-1 inhibited cell death induced by metformin and/or TRAIL. Further experiments revealed that metformin did not affect mRNA levels, but increased proteasomal degradation and protein stability of Mcl-1. Knockdown of Mule triggered a significant decrease of Mcl-1 polyubiquitination. Metformin caused the dissociation of Noxa from Mcl-1, which allowed the binding of the BH3-containing ubiquitin ligase Mule followed by Mcl-1ubiquitination and degradation. The metformin-induced degradation of Mcl-1 required E3 ligase Mule, which is responsible for the polyubiquitination of Mcl-1. Our study is the first report indicating that metformin enhances TRAIL-induced apoptosis through Noxa and favors the interaction between Mcl-1 and Mule, which consequently affects Mcl-1 ubiquitination.
Collapse
Affiliation(s)
- Seong Hye Park
- Brain Korea 21 Program for Biomedicine Science, Korea University College of Medicine, Korea University, Seoul, Republic of Korea
| | - Dae-Hee Lee
- Brain Korea 21 Program for Biomedicine Science, Korea University College of Medicine, Korea University, Seoul, Republic of Korea.,Division of Oncology/Hematology, Department of Internal Medicine, Korea University College of Medicine, Seoul, Republic of Korea
| | - Jung Lim Kim
- Division of Oncology/Hematology, Department of Internal Medicine, Korea University College of Medicine, Seoul, Republic of Korea
| | - Bo Ram Kim
- Brain Korea 21 Program for Biomedicine Science, Korea University College of Medicine, Korea University, Seoul, Republic of Korea
| | - Yoo Jin Na
- Brain Korea 21 Program for Biomedicine Science, Korea University College of Medicine, Korea University, Seoul, Republic of Korea
| | - Min Jee Jo
- Brain Korea 21 Program for Biomedicine Science, Korea University College of Medicine, Korea University, Seoul, Republic of Korea
| | - Yoon A Jeong
- Division of Oncology/Hematology, Department of Internal Medicine, Korea University College of Medicine, Seoul, Republic of Korea
| | - Suk-Young Lee
- Division of Oncology/Hematology, Department of Internal Medicine, Korea University College of Medicine, Seoul, Republic of Korea
| | - Sun Il Lee
- Department of Surgery, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Republic of Korea
| | - Yong Yook Lee
- The Korean Ginseng Research Institute, Daejeon, Republic of Korea
| | - Sang Cheul Oh
- Brain Korea 21 Program for Biomedicine Science, Korea University College of Medicine, Korea University, Seoul, Republic of Korea.,Division of Oncology/Hematology, Department of Internal Medicine, Korea University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
7
|
Zhang L, Shan Y, Li C, Sun Y, Su P, Wang J, Li L, Pan X, Zhang J. Discovery of novel anti-angiogenesis agents. Part 6: Multi-targeted RTK inhibitors. Eur J Med Chem 2017; 127:275-285. [DOI: 10.1016/j.ejmech.2016.12.059] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Revised: 12/29/2016] [Accepted: 12/30/2016] [Indexed: 01/20/2023]
|
8
|
Kang D, Choi HJ, Kang S, Kim SY, Hwang YS, Je S, Han Z, Kim JH, Song JJ. Ratio of phosphorylated HSP27 to nonphosphorylated HSP27 biphasically acts as a determinant of cellular fate in gemcitabine-resistant pancreatic cancer cells. Cell Signal 2015; 27:807-17. [PMID: 25615626 DOI: 10.1016/j.cellsig.2015.01.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Revised: 12/15/2014] [Accepted: 01/13/2015] [Indexed: 01/24/2023]
Abstract
Gemcitabine has been used most commonly as an anticancer drug to treat advanced pancreatic cancer patients. However, intrinsic or acquired resistance of pancreatic cancer to gemcitabine was also developed, which leads to very low five-year survival rates. Here, we investigated whether cellular levels of HSP27 phosphorylation act as a determinant of cellular fate with gemcitabine. In addition we have demonstrated whether HSP27 downregulation effectively could overcome the acquisition of gemcitabine resistance by using transcriptomic analysis. We observed that gemcitabine induced p38/HSP27 phosphorylation and caused acquired resistance. After acquisition of gemcitabine resistance, cancer cells showed higher activity of NF-κB. NF-κB activity, as well as colony formation in gemcitabine-resistant pancreatic cancer cells, was significantly decreased by HSP27 downregulation and subsequent TRAIL treatment, showing that HSP27 was a common network mediator of gemcitabine/TRAIL-induced cell death. After transcriptomic analysis, gene fluctuation after HSP27 downregulation was very similar to that of pancreatic cancer cells susceptible to gemcitabine, and then in opposite position to that of acquired gemcitabine resistance, which makes it possible to downregulate HSP27 to overcome the acquired gemcitabine resistance to function as an overall survival network inhibitor. Most importantly, we demonstrated that the ratio of phosphorylated HSP27 to nonphosphorylated HSP27 rather than the cellular level of HSP27 itself acts biphasically as a determinant of cellular fate in gemcitabine-resistant pancreatic cancer cells.
Collapse
Affiliation(s)
- Dongxu Kang
- Institute for Cancer Research, Yonsei University College of Medicine, Seoul, Republic of Korea; Department of Oncology, Affiliated Hospital of Yanbian University, Yanji, Jilin Province, PR China
| | - Hye Jin Choi
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Sujin Kang
- Institute for Cancer Research, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - So Young Kim
- Institute for Cancer Research, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Yong-Sic Hwang
- Department of Bioscience and Biotechnology, Konkuk University, Seoul, Republic of Korea
| | - Suyeon Je
- Institute for Cancer Research, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Zhezhu Han
- Institute for Cancer Research, Yonsei University College of Medicine, Seoul, Republic of Korea; Department of Oncology, Affiliated Hospital of Yanbian University, Yanji, Jilin Province, PR China
| | - Joo-Hang Kim
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea.
| | - Jae J Song
- Institute for Cancer Research, Yonsei University College of Medicine, Seoul, Republic of Korea; Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
9
|
Chen HF, Liu SJ, Chen G. Heat shock protein 27 phosphorylation in the proliferation and apoptosis of human umbilical vein endothelial cells induced by high glucose through the phosphoinositide 3‑kinase/Akt and extracellular signal‑regulated kinase 1/2 pathways. Mol Med Rep 2014; 11:1504-8. [PMID: 25373458 DOI: 10.3892/mmr.2014.2884] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Accepted: 09/18/2014] [Indexed: 11/06/2022] Open
Abstract
In the present study, the effect of the heat shock protein 27 (HSP27) signaling pathway on the proliferation and apoptosis of human umbilical vein endothelial cells (HUVECs) induced by high glucose (HG) was investigated. HUVEC proliferation in the indicated conditions was measured by the alamarBlue® assay. Apoptosis in HUVECs cultured with HG was analyzed by an Annexin V‑fluorescein isothiocyanate/propidium iodide apoptosis detection kit. HSP27 activity was evaluated by western blotting with specific phospho‑HSP27 antibody. HUVEC proliferation induced by HG was observed to be reduced by the HSP27 inhibitor quercetin in a concentration‑dependent manner, with a concomitant increase in apoptosis. The phosphorylation of HSP27 induced by HG was blocked by the specific phosphoinositide 3‑kinase (PI3K) inhibitor LY294002 and the specific extracellular signal‑regulated kinase (ERK) 1/2 inhibitor U0126 in a concentration‑dependent manner, with peak inhibition rates of 62.6 and 56.1%, respectively. LY294002 and U0126 also reduced HUVEC proliferation with a concomitant increase in apoptotic rate. In conclusion, HSP27 phosphorylation is important in mediating the proliferation and apoptosis of HUVECs induced by high glucose, and PI3K/Akt and ERK1/2 are important signaling pathways that contribute to HSP27 phosphorylation.
Collapse
Affiliation(s)
- Hai-Feng Chen
- Department of Cardiology, Fujian Provincial Clinical College, Fujian Medical University, Fuzhou, Fujian 350001, P.R. China
| | - Shu-Juan Liu
- Department of Endocrinology, Fujian Provincial Clinical College, Fujian Medical University, Fuzhou, Fujian 350001, P.R. China
| | - Gang Chen
- Department of Endocrinology, Fujian Provincial Clinical College, Fujian Medical University, Fuzhou, Fujian 350001, P.R. China
| |
Collapse
|
10
|
Qi Z, Shen L, Zhou H, Jiang Y, Lan L, Luo L, Yin Z. Phosphorylation of heat shock protein 27 antagonizes TNF-α induced HeLa cell apoptosis via regulating TAK1 ubiquitination and activation of p38 and ERK signaling. Cell Signal 2014; 26:1616-25. [PMID: 24686082 DOI: 10.1016/j.cellsig.2014.03.015] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2014] [Revised: 03/07/2014] [Accepted: 03/16/2014] [Indexed: 11/28/2022]
Abstract
Tumor necrosis factor (TNF)-α is a potent cytokine that regulates critical cellular processes including apoptosis. TNF-α usually triggers both survival and apoptotic signals in various cell types. Heat shock protein 27 (HSP27), an important cellular chaperone, is believed to protect cells from apoptosis. HSP27 can be phosphorylated and changed its cellular function according to different stimuli. However, available reports on the role of HSP27 phosphorylation in apoptosis remain elusive. In this study, we investigated the role of HSP27 phosphorylation in TNF-α induced apoptosis in human cervical carcinoma (HeLa) cells. We found that TNF-α induced apoptosis was enhanced if we suppressed the TNF-α induced HSP27 phosphorylation by specific inhibitor CMPD1 or MAPKAPK2 (MK2) knockdown or by overexpression of non-phosphorylatable mutant HSP27-3A. Through co-immunoprecipitation and confocal microscopy, we observed that HSP27 associated with transforming growth factor-β (TGF-β)-activated kinase 1 (TAK1) in response to TNF-α stimulation. By blocking MK2 activity or overexpressing phospho-mimetic mutant Hsp27-3D, we further showed that HSP27 phosphorylation facilitated the TNF-α induced ubiquitination and phosphorylation of TAK1 and the activations of p38 MAPK and ERK, the TAK1 downstream pro-survival signaling. In addition, we also found that increased HSP27 phosphorylation inhibited TRADD ubiquitination but did not influence the binding between TRADD and FADD in a pro-apoptotic complex. Taken together, our data indicated that HSP27 phosphorylation was involved in modulating the TNF-α induced apoptosis via interacting with TAK1 and regulating TAK1 post-translational modifications in HeLa cells. This study demonstrates that HSP27 phosphorylation serves as a novel regulator in TNF-α-induced apoptosis, and provides a new insight into the cytoprotective role of HSP27 phosphorylation.
Collapse
Affiliation(s)
- Zhilin Qi
- Jiangsu Province Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, Jiangsu, PR China; Department of Biochemistry, Wannan Medical College, Wuhu, Anhui, PR China
| | - Lei Shen
- Jiangsu Province Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, Jiangsu, PR China
| | - Huiting Zhou
- Jiangsu Province Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, Jiangsu, PR China
| | - Yi Jiang
- Jiangsu Province Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, Jiangsu, PR China
| | - Lei Lan
- Jiangsu Province Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, Jiangsu, PR China.
| | - Lan Luo
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, PR China.
| | - Zhimin Yin
- Jiangsu Province Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, Jiangsu, PR China.
| |
Collapse
|
11
|
Qi S, Xin Y, Qi Z, Xu Y, Diao Y, Lan L, Luo L, Yin Z. HSP27 phosphorylation modulates TRAIL-induced activation of Src-Akt/ERK signaling through interaction with β-arrestin2. Cell Signal 2014; 26:594-602. [DOI: 10.1016/j.cellsig.2013.11.033] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2013] [Revised: 11/25/2013] [Accepted: 11/25/2013] [Indexed: 11/29/2022]
|
12
|
Mellier G, Liu D, Bellot G, Holme AL, Pervaiz S. Small molecule sensitization to TRAIL is mediated via nuclear localization, phosphorylation and inhibition of chaperone activity of Hsp27. Cell Death Dis 2013; 4:e890. [PMID: 24176848 PMCID: PMC3920951 DOI: 10.1038/cddis.2013.413] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2013] [Revised: 09/19/2013] [Accepted: 09/20/2013] [Indexed: 02/05/2023]
Abstract
The small chaperone protein Hsp27 confers resistance to apoptosis, and therefore is an attractive anticancer drug target. We report here a novel mechanism underlying the tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) sensitizing activity of the small molecule LY303511, an inactive analog of the phosphoinositide 3-kinase inhibitor inhibitor LY294002, in HeLa cells that are refractory to TRAIL-induced apoptosis. On the basis of the fact that LY303511 is derived from LY294002, itself derived from quercetin, and earlier findings indicating that quercetin and LY294002 affected Hsp27 expression, we investigated whether LY303511 sensitized cancer cells to TRAIL via a conserved inhibitory effect on Hsp27. We provide evidence that upon treatment with LY303511, Hsp27 is progressively sequestered in the nucleus, thus reducing its protective effect in the cytosol during the apoptotic process. LY303511-induced nuclear translocation of Hsp27 is linked to its sustained phosphorylation via activation of p38 kinase and MAPKAP kinase 2 and the inhibition of PP2A. Furthermore, Hsp27 phosphorylation leads to the subsequent dissociation of its large oligomers and a decrease in its chaperone activity, thereby further compromising the death inhibitory activity of Hsp27. Furthermore, genetic manipulation of Hsp27 expression significantly affected the TRAIL sensitizing activity of LY303511, which corroborated the Hsp27 targeting activity of LY303511. Taken together, these data indicate a novel mechanism of small molecule sensitization to TRAIL through targeting of Hsp27 functions, rather than its overall expression, leading to decreased cellular protection, which could have therapeutic implications for overcoming chemotherapy resistance in tumor cells.
Collapse
Affiliation(s)
- G Mellier
- 1] ROS, Apoptosis and Cancer Biology Laboratory, Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore [2] Cancer and Stem Cell Biology Program, Duke-NUS Graduate Medical School, Singapore, Singapore
| | | | | | | | | |
Collapse
|
13
|
Kim J, Kang D, Sun BK, Kim JH, Song JJ. TRAIL/MEKK4/p38/HSP27/Akt survival network is biphasically modulated by the Src/CIN85/c-Cbl complex. Cell Signal 2012; 25:372-9. [PMID: 23085457 DOI: 10.1016/j.cellsig.2012.10.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2012] [Revised: 10/01/2012] [Accepted: 10/16/2012] [Indexed: 12/30/2022]
Abstract
Previously, we showed that mitogen-activated protein kinase/extracellular signal-related kinase 4 (MEKK4) is responsible for p38 activation and that its activation during tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) treatment also increases the catalytic activity of Akt. Here, we further investigated how the TRAIL-induced MEKK4/p38/heat shock protein (HSP27)/Akt survival network is modulated by the Src/c-Cbl interacting protein of 85kDa (CIN85)/c-Cbl complex. TRAIL-induced activation of Akt catalytic activity and phosphorylation were highly correlated with p38/HSP27 phosphorylation, whereas the phosphorylation of p38/HSP27 increased further during incubation with curcumin and TRAIL, which caused significant apoptotic cell death. CIN85, a c-Cbl-binding protein, plays an essential role in connecting cell survival to cell death. The interaction of CIN85 with MEKK4 was increased during the late phase of TRAIL incubation, suggesting that sustained p38 and HSP27 phosphorylation protects cells by preventing further cell death. However, further increases in p38/HSP27 phosphorylation induced by cotreatment with curcumin and TRAIL converted cell fate to death. Taken together, these data demonstrate that phosphorylated p38/HSP27 as biphasic modulators act in conjunction with CIN85 to determine whether cells survive or die in response to apoptotic stress.
Collapse
Affiliation(s)
- Jina Kim
- Institute for Cancer Research, College of Medicine, Yonsei University, Seoul, Republic of Korea
| | | | | | | | | |
Collapse
|