1
|
Huo Q, Wang J, Xie N. High HSPB1 expression predicts poor clinical outcomes and correlates with breast cancer metastasis. BMC Cancer 2023; 23:501. [PMID: 37268925 DOI: 10.1186/s12885-023-10983-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 05/19/2023] [Indexed: 06/04/2023] Open
Abstract
BACKGROUND Heat shock protein beta-1 (HSPB1) is a crucial biomarker for pathological processes in various cancers. However, the clinical value and function of HSPB1 in breast cancer has not been extensively explored. Therefore, we adopted a systematic and comprehensive approach to investigate the correlation between HSPB1 expression and clinicopathological features of breast cancer, as well as determine its prognostic value. We also examined the effects of HSPB1 on cell proliferation, invasion, apoptosis, and metastasis. METHODS We investigated the expression of HSPB1 in patients with breast cancer using The Cancer Genome Atlas and immunohistochemistry. Chi-squared test and Wilcoxon signed-rank test were used to examine the relationship between HSPB1 expression and clinicopathological characteristics. RESULTS We observed that HSPB1 expression was significantly correlated with the stage N, pathologic stages, as well as estrogen and progesterone receptors. Furthermore, high HSPB1 expression resulted in a poor prognosis for overall survival, relapse-free survival, and distant metastasis-free survival. Multivariable analysis showed that patients with poor survival outcomes had higher tumor, node, metastasis, and pathologic stages. Pathway analysis of HSPB1 and the altered neighboring genes suggested that HSPB1 is involved in the epithelial-to-mesenchymal transition. Functional analysis revealed showed that transient knockdown of HSPB1 inhibited the cell migration/invasion ability and promoted apoptosis. CONCLUSIONS HSPB1 may be involved in breast cancer metastasis. Collectively, our study demonstrated that HSPB1 has prognostic value for clinical outcomes and may serve as a therapeutic biomarker for breast cancer.
Collapse
Affiliation(s)
- Qin Huo
- Biobank, Shenzhen Institute of Translational Medicine, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Health Science Center, Shenzhen University, 518035, Shenzhen, China
| | - Juan Wang
- Department of General Practice, Army Medical Center of PLA, Chongqing, 400042, China
| | - Ni Xie
- Biobank, Shenzhen Institute of Translational Medicine, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Health Science Center, Shenzhen University, 518035, Shenzhen, China.
| |
Collapse
|
2
|
Rizvi SF, Hasan A, Parveen S, Mir SS. Untangling the complexity of heat shock protein 27 in cancer and metastasis. Arch Biochem Biophys 2023; 736:109537. [PMID: 36738981 DOI: 10.1016/j.abb.2023.109537] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 12/27/2022] [Accepted: 02/01/2023] [Indexed: 02/05/2023]
Abstract
Heat shock protein 27 is a type of molecular chaperone whose expression gets up-regulated due to reaction towards different stressful triggers including anticancer treatments. It is known to be a major player of resistance development in cancer cells, whereby cells are sheltered against the therapeutics that normally activate apoptosis. Heat shock protein 27 (HSP27) is one of the highly expressed proteins during various cellular insults and is a strong tumor survival factor. HSP27 influences various cellular pathways associated with cancer cell survival and growth such as apoptosis, autophagy, metastasis, angiogenesis, epithelial to mesenchymal transition, etc. HSP27 is molecular machinery which prevents the clumping of numerous substrates or client proteins which get mutated in cancer. It has been reported in several studies that targeting HSP27 is difficult because of its dynamic structure and absence of an ATP-binding site. Here, in this review, we have summarized different modulators of HSP27 and their mechanism of action as well. Effect of deregulated HSP27 in various cancer models, limitations of targeting HSP27, resistance against the conventional drugs generated due to the overexpression of HSP27, and measures to counteract this effect have also been discussed here in detail.
Collapse
Affiliation(s)
- Suroor Fatima Rizvi
- Molecular Cell Biology Laboratory, Integral Information and Research Centre-4 (IIRC-4), Integral University, Kursi Road, Lucknow, 226026, India; Department of Bioengineering, Faculty of Engineering, Integral University, Kursi Road, Lucknow, 226026, India.
| | - Adria Hasan
- Molecular Cell Biology Laboratory, Integral Information and Research Centre-4 (IIRC-4), Integral University, Kursi Road, Lucknow, 226026, India; Department of Bioengineering, Faculty of Engineering, Integral University, Kursi Road, Lucknow, 226026, India.
| | - Sana Parveen
- Molecular Cell Biology Laboratory, Integral Information and Research Centre-4 (IIRC-4), Integral University, Kursi Road, Lucknow, 226026, India; Department of Biosciences, Faculty of Science, Integral University, Kursi Road, Lucknow, 226026, India.
| | - Snober S Mir
- Molecular Cell Biology Laboratory, Integral Information and Research Centre-4 (IIRC-4), Integral University, Kursi Road, Lucknow, 226026, India; Department of Biosciences, Faculty of Science, Integral University, Kursi Road, Lucknow, 226026, India.
| |
Collapse
|
3
|
Alvarez FA, Kaddour H, Lyu Y, Preece C, Cohen J, Baer L, Stopeck AT, Thompson P, Okeoma CM. Blood plasma derived extracellular vesicles (BEVs): particle purification liquid chromatography (PPLC) and proteomic analysis reveals BEVs as a potential minimally invasive tool for predicting response to breast cancer treatment. Breast Cancer Res Treat 2022; 196:423-437. [PMID: 36114323 PMCID: PMC10560447 DOI: 10.1007/s10549-022-06733-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 08/28/2022] [Indexed: 11/02/2022]
Abstract
PURPOSE Circulating blood plasma derived extracellular vesicles (BEVs) containing proteins hold promise for their use as minimally invasive biomarkers for predicting response to cancer therapy. The main goal of this study was to establish the efficiency and utility of the particle purification liquid chromatography (PPLC) BEV isolation method and evaluate the role of BEVs in predicting breast cancer (BC) patient response to neoadjuvant chemotherapy (NAC). METHODS PPLC isolation was used to separate BEVs from non-EV contaminants and characterize BEVs from 17 BC patients scheduled to receive NAC. Using LC-MS/MS, we compared the proteome of PPLC-isolated BEVs from patients (n = 7) that achieved a pathological complete response (pCR) after NAC (responders [R]) to patients (n = 10) who did not achieve pCR (non-responders [NR]). Luminal MCF7 and basaloid MDA-MB-231 BC cells were treated with isolated BEVs and evaluated for metabolic activity by MTT assay. RESULTS NR had elevated BEV concentrations and negative zeta potential (ζ-potential) prior to receipt of NAC. Eight proteins were enriched in BEVs of NR. GP1BA (CD42b), PECAM-1 (CD31), CAPN1, HSPB1 (HSP27), and ANXA5 were validated using western blot. MTT assay revealed BEVs from R and NR patients increased metabolic activity of MCF7 and MDA-MB-231 BC cells and the magnitude was highest in MCF7s treated with NR BEVs. CONCLUSION PPLC-based EV isolation provides a preanalytical separation process for BEVs devoid of most contaminants. Our findings suggest that PPLC-isolated BEVs and the five associated proteins may be established as predictors of chemoresistance, and thus serve to identify NR to spare them the toxic effects of NAC.
Collapse
Affiliation(s)
- Folnetti A Alvarez
- Department of Pharmacology, Stony Brook University Renaissance School of Medicine, Stony Brook, NY, 11794-8651, USA
| | - Hussein Kaddour
- Department of Pharmacology, Stony Brook University Renaissance School of Medicine, Stony Brook, NY, 11794-8651, USA
- Regeneron Pharmaceuticals, Inc, Tarrytown, NY, 10591, USA
| | - Yuan Lyu
- Department of Pharmacology, Stony Brook University Renaissance School of Medicine, Stony Brook, NY, 11794-8651, USA
- Medical Research Center, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Christina Preece
- Department of Pathology, Stony Brook University Renaissance School of Medicine, Stony Brook, NY, 11794-8651, USA
- Department of Medicine, Cedars Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Jules Cohen
- Department of Medicine, Division of Hematology and Medical Oncology, Stony Brook University, Stony Brook, NY, 11794-8651, USA
- Stony Brook University Cancer Center, Stony Brook, NY, 11794-8651, USA
| | - Lea Baer
- Department of Medicine, Division of Hematology and Medical Oncology, Stony Brook University, Stony Brook, NY, 11794-8651, USA
- Stony Brook University Cancer Center, Stony Brook, NY, 11794-8651, USA
| | - Alison T Stopeck
- Department of Medicine, Division of Hematology and Medical Oncology, Stony Brook University, Stony Brook, NY, 11794-8651, USA
- Stony Brook University Cancer Center, Stony Brook, NY, 11794-8651, USA
| | - Patricia Thompson
- Department of Pathology, Stony Brook University Renaissance School of Medicine, Stony Brook, NY, 11794-8651, USA
- Stony Brook University Cancer Center, Stony Brook, NY, 11794-8651, USA
| | - Chioma M Okeoma
- Department of Pharmacology, Stony Brook University Renaissance School of Medicine, Stony Brook, NY, 11794-8651, USA.
- Department of Pathology, Microbiology, and Immunology, New York Medical College, Valhalla, NY, 10595-1524, USA.
| |
Collapse
|
4
|
Alternative ANKHD1 transcript promotes proliferation and inhibits migration in uterine corpus endometrial carcinoma. NPJ Genom Med 2022; 7:56. [PMID: 36171217 PMCID: PMC9519915 DOI: 10.1038/s41525-022-00321-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Accepted: 08/08/2022] [Indexed: 11/22/2022] Open
Abstract
Alternative splicing (AS) is common in gene expression, and abnormal splicing often results in several cancers. Overall survival-associated splicing events (OS-SEs) have been used to predict prognosis in cancer. The aim of this study was to investigate the presence and function of OS-SEs in uterine corpus endometrial carcinoma (UCEC). Based on TCGA and TCGASpliceSeq databases, gene expression and the AS data of UCEC samples were retrieved. An alternate terminator of ANKHD1 transcripts named ANKHD1-BP3 was found to be significantly related to metastasis and OS in UCEC and significantly associated with HSPB1. The upregulated expression of HSPB1 induced downregulation of ANKHD1-BP3 and promoted tumor metastasis. These findings indicate that HSPB1, a splicing factor, regulates the expression of ANKHD1-BP3 to promote metastasis in UCEC.
Collapse
|
5
|
Alberti G, Vergilio G, Paladino L, Barone R, Cappello F, Conway de Macario E, Macario AJL, Bucchieri F, Rappa F. The Chaperone System in Breast Cancer: Roles and Therapeutic Prospects of the Molecular Chaperones Hsp27, Hsp60, Hsp70, and Hsp90. Int J Mol Sci 2022; 23:ijms23147792. [PMID: 35887137 PMCID: PMC9324353 DOI: 10.3390/ijms23147792] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 06/30/2022] [Accepted: 07/10/2022] [Indexed: 12/26/2022] Open
Abstract
Breast cancer (BC) is a major public health problem, with key pieces of information needed for developing preventive and curative measures still missing. For example, the participation of the chaperone system (CS) in carcinogenesis and anti-cancer responses is poorly understood, although it can be predicted to be a crucial factor in these mechanisms. The chief components of the CS are the molecular chaperones, and here we discuss four of them, Hsp27, Hsp60, Hsp70, and Hsp90, focusing on their pro-carcinogenic roles in BC and potential for developing anti-BC therapies. These chaperones can be targets of negative chaperonotherapy, namely the elimination/blocking/inhibition of the chaperone(s) functioning in favor of BC, using, for instance, Hsp inhibitors. The chaperones can also be employed in immunotherapy against BC as adjuvants, together with BC antigens. Extracellular vesicles (EVs) in BC diagnosis and management are also briefly discussed, considering their potential as easily accessible carriers of biomarkers and as shippers of anti-cancer agents amenable to manipulation and controlled delivery. The data surveyed from many laboratories reveal that, to enhance the understanding of the role of the CS in BS pathogenesis, one must consider the CS as a physiological system, encompassing diverse members throughout the body and interacting with the ubiquitin–proteasome system, the chaperone-mediated autophagy machinery, and the immune system (IS). An integrated view of the CS, including its functional partners and considering its highly dynamic nature with EVs transporting CS components to reach all the cell compartments in which they are needed, opens as yet unexplored pathways leading to carcinogenesis that are amenable to interference by anti-cancer treatments centered on CS components, such as the molecular chaperones.
Collapse
Affiliation(s)
- Giusi Alberti
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BiND), University of Palermo, 90127 Palermo, Italy; (G.A.); (G.V.); (R.B.); (F.C.); (F.B.); (F.R.)
| | - Giuseppe Vergilio
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BiND), University of Palermo, 90127 Palermo, Italy; (G.A.); (G.V.); (R.B.); (F.C.); (F.B.); (F.R.)
- Euro-Mediterranean Institute of Science and Technology (IEMEST), 90139 Palermo, Italy;
| | - Letizia Paladino
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BiND), University of Palermo, 90127 Palermo, Italy; (G.A.); (G.V.); (R.B.); (F.C.); (F.B.); (F.R.)
- Euro-Mediterranean Institute of Science and Technology (IEMEST), 90139 Palermo, Italy;
- Correspondence:
| | - Rosario Barone
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BiND), University of Palermo, 90127 Palermo, Italy; (G.A.); (G.V.); (R.B.); (F.C.); (F.B.); (F.R.)
| | - Francesco Cappello
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BiND), University of Palermo, 90127 Palermo, Italy; (G.A.); (G.V.); (R.B.); (F.C.); (F.B.); (F.R.)
- Euro-Mediterranean Institute of Science and Technology (IEMEST), 90139 Palermo, Italy;
| | - Everly Conway de Macario
- Department of Microbiology and Immunology, School of Medicine, University of Maryland at Baltimore-Institute of Marine and Environmental Technology (IMET), Baltimore, MD 21202, USA;
| | - Alberto J. L. Macario
- Euro-Mediterranean Institute of Science and Technology (IEMEST), 90139 Palermo, Italy;
- Department of Microbiology and Immunology, School of Medicine, University of Maryland at Baltimore-Institute of Marine and Environmental Technology (IMET), Baltimore, MD 21202, USA;
| | - Fabio Bucchieri
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BiND), University of Palermo, 90127 Palermo, Italy; (G.A.); (G.V.); (R.B.); (F.C.); (F.B.); (F.R.)
| | - Francesca Rappa
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BiND), University of Palermo, 90127 Palermo, Italy; (G.A.); (G.V.); (R.B.); (F.C.); (F.B.); (F.R.)
| |
Collapse
|
6
|
Mohammadi Ghahhari N, Sznurkowska MK, Hulo N, Bernasconi L, Aceto N, Picard D. Cooperative interaction between ERα and the EMT-inducer ZEB1 reprograms breast cancer cells for bone metastasis. Nat Commun 2022; 13:2104. [PMID: 35440541 PMCID: PMC9018728 DOI: 10.1038/s41467-022-29723-5] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 03/30/2022] [Indexed: 02/08/2023] Open
Abstract
The epithelial to mesenchymal transition (EMT) has been proposed to contribute to the metastatic spread of breast cancer cells. EMT-promoting transcription factors determine a continuum of different EMT states. In contrast, estrogen receptor α (ERα) helps to maintain the epithelial phenotype of breast cancer cells and its expression is crucial for effective endocrine therapies. Determining whether and how EMT-associated transcription factors such as ZEB1 modulate ERα signaling during early stages of EMT could promote the discovery of therapeutic approaches to suppress metastasis. Here we show that, shortly after induction of EMT and while cells are still epithelial, ZEB1 modulates ERα-mediated transcription induced by estrogen or cAMP signaling in breast cancer cells. Based on these findings and our ex vivo and xenograft results, we suggest that the functional interaction between ZEB1 and ERα may alter the tissue tropism of metastatic breast cancer cells towards bone. The epithelial mesenchymal transition (EMT) is important in the metastatic spread of cancer cells. Here, the authors show that the EMT transcription factor, ZEB1, can modify estrogen receptor α during EMT and facilitate the migration of breast cancer cells to the bone
Collapse
Affiliation(s)
| | - Magdalena K Sznurkowska
- Department of Biology, Institute of Molecular Health Sciences, ETH Zurich, 8093, Zürich, Switzerland
| | - Nicolas Hulo
- Institute of Genetics and Genomics of Geneva, Université de Genève, 1211, Genève 4, Switzerland
| | - Lilia Bernasconi
- Département de Biologie Cellulaire, Université de Genève, Sciences III, 1211, Genève 4, Switzerland
| | - Nicola Aceto
- Department of Biology, Institute of Molecular Health Sciences, ETH Zurich, 8093, Zürich, Switzerland
| | - Didier Picard
- Département de Biologie Cellulaire, Université de Genève, Sciences III, 1211, Genève 4, Switzerland.
| |
Collapse
|
7
|
Du J, Tao Q, Liu Y, Huang Z, Jin H, Lin W, Huang X, Zeng J, Zhao Y, Liu L, Xu Q, Han X, Chen L, Chen XL, Wen Y. Assessment of the targeted effect of Sijunzi decoction on the colorectal cancer microenvironment via the ESTIMATE algorithm. PLoS One 2022; 17:e0264720. [PMID: 35303006 PMCID: PMC8932555 DOI: 10.1371/journal.pone.0264720] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 02/15/2022] [Indexed: 11/18/2022] Open
Abstract
Objective Sijunzi decoction (SJZD) was used to treat patients with colorectal cancer (CRC) as an adjuvant method. The aim of the study was to investigate the therapeutic targets and pathways of SJZD towards the tumor microenvironment of CRC via network pharmacology and the ESTIMATE algorithm. Methods The ESTIMATE algorithm was used to calculate immune and stromal scores to predict the level of infiltrating immune and stromal cells. The active targets of SJZD were searched in the Traditional Chinese Medicine Systems Pharmacology Database and Analysis Platform (TCMSP) and UniProt database. The core targets were obtained by matching the differentially expressed genes in CRC tissues and the targets of SJZD. Then, GO, KEGG and validation in TCGA were carried out. Results According to the ESTIMATE algorithm and survival analysis, the median survival time of the low stromal score group was significantly higher than that of the high stromal score group (P = 0.018), while the patients showed no significant difference of OS between different immune groups (P = 0.19). A total of 929 genes were upregulated and 115 genes were downregulated between the stromal score groups (|logFC| > 2, adjusted P < 0.05); 357 genes were upregulated and 472 genes were downregulated between the immune score groups. The component-target network included 139 active components and 52 related targets. The core targets were HSPB1, SPP1, IGFBP3, and TGFB1, which were significantly associated with poor prognosis in TCGA validation. GO terms included the response to hypoxia, the extracellular space, protein binding and the TNF signaling pathway. Immunoreaction was the main enriched pathway identified by KEGG analysis. Conclusion The core genes (HSPB1, SPP1, IGFBP3 and TGFB1) affected CRC development and prognosis by regulating hypoxia, protein binding and epithelial-mesenchymal transition in the extracellular matrix.
Collapse
Affiliation(s)
- Jiaxin Du
- School of Basic Medical Science, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Quyuan Tao
- School of Basic Medical Science, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ying Liu
- School of Basic Medical Science, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhanming Huang
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - He Jin
- School of Basic Medical Science, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Wenjia Lin
- School of Basic Medical Science, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xinying Huang
- School of Basic Medical Science, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jingyan Zeng
- Shenzhen Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yongchang Zhao
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lingyu Liu
- School of Basic Medical Science, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qian Xu
- School of Basic Medical Science, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xue Han
- School of Basic Medical Science, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lixia Chen
- School of Basic Medical Science, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xin-lin Chen
- School of Basic Medical Science, Guangzhou University of Chinese Medicine, Guangzhou, China
- * E-mail: (XC); (YW)
| | - Yi Wen
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
- * E-mail: (XC); (YW)
| |
Collapse
|
8
|
Proteomic Signatures of Diffuse and Intestinal Subtypes of Gastric Cancer. Cancers (Basel) 2021; 13:cancers13235930. [PMID: 34885041 PMCID: PMC8656738 DOI: 10.3390/cancers13235930] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 11/20/2021] [Accepted: 11/23/2021] [Indexed: 12/14/2022] Open
Abstract
Gastric cancer is a leading cause of death from cancer globally. Gastric cancer is classified into intestinal, diffuse and indeterminate subtypes based on histology according to the Laurén classification. The intestinal and diffuse subtypes, although different in histology, demographics and outcomes, are still treated in the same fashion. This study was designed to discover proteomic signatures of diffuse and intestinal subtypes. Mass spectrometry-based proteomics using tandem mass tags (TMT)-based multiplexed analysis was used to identify proteins in tumor tissues from patients with diffuse or intestinal gastric cancer with adjacent normal tissue control. A total of 7448 or 4846 proteins were identified from intestinal or diffuse subtype, respectively. This quantitative mass spectrometric analysis defined a proteomic signature of differential expression across the two subtypes, which included gremlin1 (GREM1), bcl-2-associated athanogene 2 (BAG2), olfactomedin 4 (OLFM4), thyroid hormone receptor interacting protein 6 (TRIP6) and melanoma-associated antigen 9 (MAGE-A9) proteins. Although GREM1, BAG2, OLFM4, TRIP6 and MAGE-A9 have all been previously implicated in tumor progression and metastasis, they have not been linked to intestinal or diffuse subtypes of gastric cancer. Using immunohistochemical labelling of a tissue microarray comprising of 124 cases of gastric cancer, we validated the proteomic signature obtained by mass spectrometry in the discovery cohort. Our findings should help investigate the pathogenesis of these gastric cancer subtypes and potentially lead to strategies for early diagnosis and treatment.
Collapse
|
9
|
Wang S, Zhang X, Wang H, Wang Y, Chen P, Wang L. Heat Shock Protein 27 Enhances SUMOylation of Heat Shock Protein B8 to Accelerate the Progression of Breast Cancer. THE AMERICAN JOURNAL OF PATHOLOGY 2021; 190:2464-2477. [PMID: 33222991 DOI: 10.1016/j.ajpath.2020.04.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 03/19/2020] [Accepted: 04/08/2020] [Indexed: 02/07/2023]
Abstract
Heat shock proteins (HSPs) are emerging as valuable potential molecular targets in breast cancer therapy owing to their diverse functions in cancer cells. This study investigated the potential role of heat shock protein 27 (HSP27, also known as HSPB1) in breast cancer through heat shock protein B8 (HSPB8). The correlation between HSP27 and HSPB8 was identified by using co-immunoprecipitation, immunoprecipitation, and SUMOylation assays. Through gain- and loss-of-function approaches in MCF-7 cells, the effect of HSP27 on HSPB8 expression, SUMOylation level, and protein stability of HSPB8, as well as on cell proliferation, migration, and stemness, was elucidated. A mouse xenograft model of breast cancer cells was established to verify the function of HSP27 in vivo. Results indicate that HSP27 and HSPB8 were highly expressed in breast cancer tissues and MCF-7 cells. HSP27 was also found to induce the SUMOylation of HSPB8 at the 106 locus and subsequently increased its protein stability, which resulted in accelerated proliferation, migration, and stemness of breast cancer cells in vitro along with increased tumor metastasis of breast cancer in vivo. However, these results could be reversed by the knockdown of HSPB8. Overall, HSP27 induces SUMOylation of HSPB8 to promote HSPB8 expression, thereby endorsing proliferation and metastasis of breast cancer cells. This study may provide insight for the development of new targets for breast cancer.
Collapse
Affiliation(s)
- Shuai Wang
- School of Medical Imaging, Weifang Medical University, Weifang, China; Department of Oncology, Weifang Traditional Chinese Medicine Hospital, Weifang, China; Qingdao Cancer Institute, Qingdao, China
| | - Xinyan Zhang
- Department of Intervention, The Affiliated Weihai Second Municipal Hospital of Qingdao University, Weihai, China
| | - Haiwei Wang
- Department of Oncology, Weifang Traditional Chinese Medicine Hospital, Weifang, China
| | - Yang Wang
- Department of Oncology, Weifang Traditional Chinese Medicine Hospital, Weifang, China
| | - Peng Chen
- Department of Oncology, Weifang Traditional Chinese Medicine Hospital, Weifang, China
| | - Longgang Wang
- Department of General Surgery, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China.
| |
Collapse
|
10
|
Yıldız M, Terzi H, Yıldız SH, Varol N, Özdemİr Erdoğan M, Kasap M, Akçalı N, Solak M. Proteomic analysis of the anticancer effect of various extracts of endemicThermopsisturcica in human cervical cancer cells. Turk J Med Sci 2020; 50:1993-2004. [PMID: 32682359 PMCID: PMC7775707 DOI: 10.3906/sag-2005-321] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 07/18/2020] [Indexed: 12/30/2022] Open
Abstract
Background/aim Thermopsisturcica is a perennial species endemic to Turkey and different extracts of T. turcica have an antiproliferative effect on cancer cells, but there has not been any report on HeLa (human cervical cancer) cells. Materials and methods To get a better understanding of the molecular mechanism of anticancer activity of methanolic extracts of leaves (LE) and flowers (FE) of T. turcica, we employed 2-DE-based proteomics to explore the proteins involved in anticancer activity in HeLa cells. Results T. turcica extracts showed a potent cytotoxic effect on HeLa cells with the IC50 values of 1.75 mg/mL for LE and 3.25 mg/mL for FE. The induction of apoptosis by LE and FE was also consistent with increased expression of caspase mRNAs and DNA fragmentation. In terms of the proteomic approach, 27 differentially expressed proteins were detected and identified through MALDI-TOF/TOF mass spectrometry. These altered proteins were involved in cytoskeleton organization and movement, protein folding, proteolysis and translation, cell cycle and proliferation, signal transduction, cell redox homeostasis, and metabolism. Conclusion Up-regulation of protein disulfide isomerases and down-regulation of Rho GDP-dissociation inhibitor, heterogeneous nuclear ribonucleoproteins, and heat shock proteins may contribute to the induction of apoptosis and arresting of the cell cycle in HeLa cells.
Collapse
Affiliation(s)
- Mustafa Yıldız
- Department of Molecular Biology and Genetics, Faculty of Science and Literature, Afyon Kocatepe University, Afyonkarahisar, Turkey
| | - Hakan Terzi
- Department of Molecular Biology and Genetics, Faculty of Science and Literature, Afyon Kocatepe University, Afyonkarahisar, Turkey
| | - Saliha Handan Yıldız
- Department of Medical Genetics, Faculty of Medicine, Afyonkarahisar University of Health Sciences, Afyonkarahisar, Turkey
| | - Nuray Varol
- Department of Medical Genetics, Faculty of Medicine, Afyonkarahisar University of Health Sciences, Afyonkarahisar, Turkey
| | - Müjgan Özdemİr Erdoğan
- Department of Medical Genetics, Faculty of Medicine, Afyonkarahisar University of Health Sciences, Afyonkarahisar, Turkey
| | - Murat Kasap
- Department of Medical Biology, Faculty of Medicine, Kocaeli University, Kocaeli, Turkey
| | - Nermin Akçalı
- Department of Medical Genetics, Faculty of Medicine, Afyonkarahisar University of Health Sciences, Afyonkarahisar, Turkey
| | - Mustafa Solak
- Department of Medical Genetics, Faculty of Medicine, Afyonkarahisar University of Health Sciences, Afyonkarahisar, Turkey
| |
Collapse
|
11
|
Takeshita T, Yan L, Peng X, Kimbung S, Hatschek T, Hedenfalk IA, Rashid OM, Takabe K. Transcriptomic and functional pathway features were associated with survival after pathological complete response to neoadjuvant chemotherapy in breast cancer. Am J Cancer Res 2020; 10:2555-2569. [PMID: 32905537 PMCID: PMC7471342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Accepted: 07/13/2020] [Indexed: 06/11/2023] Open
Abstract
Pathological complete response (pCR) after neoadjuvant chemotherapy (NAC) has been proposed as a surrogate endpoint for the prediction of long-term survival in breast cancer (BC); however, an increased pCR rate has not clearly correlated with improved survival. We hypothesized that some transcriptomic and functional pathway features correlate with survival after pCR in BC. We utilized 2 published NAC cohorts, 105 women with gene expression data before, "Baseline", and that changed during NAC, "Delta", and TCGA database with 1068 BC patients to investigate the relationship between the efficacy of NAC and survival utilizing differentially expressed-mRNAs, construction and analysis of the mRNA-hub gene network, and functional pathway analysis. In mRNA expression profiling, S100A8 was a gene involved in survival after pCR in Baseline and NDP was a gene involved in recurrence after pCR in Delta. In functional pathway analysis, we found multiple pathways involved in survival after pCR. In mRNA-hub gene analysis, HSP90AA1, EEF1A1, APP, and HSPA4 were related to recurrence in BC patients with pCR due to NAC. TP53, EGFR, CTNNB1, ERBB2, and HSPB1 may play a significant role in survival for patients with pCR. Interestingly, high HSP90AA1, HSPA4, S100A8, and TP53, and low EEF1A1, EGFR, and CTNNB1 expressing tumors have significantly worse overall survival in TCGA BC cohort. We demonstrated the genes and functional pathway features associated with pCR and survival utilizing the bioinformatics approach to public BC cohorts. Some genes involved in recurrence after pCR due to NAC also served as prognostic factors in primary BC.
Collapse
Affiliation(s)
- Takashi Takeshita
- Breast Surgery, Department of Surgical Oncology, Roswell Park Comprehensive Cancer CenterBuffalo, NY, USA
| | - Li Yan
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer CenterBuffalo, NY, USA
| | - Xuan Peng
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer CenterBuffalo, NY, USA
| | - Siker Kimbung
- Division of Oncology, Department of Clinical Sciences and Lund University Cancer Center, Lund UniversityLund, Sweden
| | - Thomas Hatschek
- Breast Center, Karolinska University Hospital and Department of Oncology-Pathology, Karolinska InstitutetSolna, Sweden
| | - Ingrid A Hedenfalk
- Division of Oncology, Department of Clinical Sciences and Lund University Cancer Center, Lund UniversityLund, Sweden
| | - Omar M Rashid
- Holy Cross Hospital Michael and Dianne Bienes Comprehensive Cancer CenterFort Lauderdale, Florida, USA
- Department of Surgery, Massachusetts General HospitalBoston, Massachusetts, USA
- Department of Surgery, University of Miami Miller School of MedicineMiami, Florida, USA
- Department of Surgery, Nova Southeastern University School of MedicineFort Lauderdale, Florida, USA
| | - Kazuaki Takabe
- Breast Surgery, Department of Surgical Oncology, Roswell Park Comprehensive Cancer CenterBuffalo, NY, USA
- Department of Surgery, University at Buffalo Jacobs School of Medicine and Biomedical Sciences, The State University of New YorkBuffalo, NY, USA
- Department of Breast Surgery and Oncology, Tokyo Medical UniversityTokyo, Japan
- Department of Surgery, Yokohama City UniversityYokohama, Japan
- Department of Surgery, Niigata University Graduate School of Medical and Dental SciencesNiigata, Japan
- Department of Breast Surgery, Fukushima Medical UniversityFukushima, Japan
| |
Collapse
|
12
|
Netsirisawan P, Chokchaichamnankit D, Saharat K, Srisomsap C, Svasti J, Champattanachai V. Quantitative proteomic analysis of the association between decreasing O‑GlcNAcylation and metastasis in MCF‑7 breast cancer cells. Int J Oncol 2020; 56:1387-1404. [PMID: 32236627 PMCID: PMC7170043 DOI: 10.3892/ijo.2020.5022] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 02/06/2020] [Indexed: 12/11/2022] Open
Abstract
Breast cancer is the most common type of cancer and leading cause of cancer-associated mortality in women worldwide. O-linked N-acetyl glucosaminylation (O-GlcNAcylation) is a dynamic post-translational modification of nuclear, cytoplasmic and mitochondrial proteins. Mounting evidence suggests that abnormal O-GlcNAcylation status is associated with cancer malignancy. In our previous study, it was reported that O-GlcNAc and O-GlcNAc transferase (OGT; an enzyme responsible for the addition of O-GlcNAc) were upregulated in breast cancer tissues and cells. Moreover, O-GlcNAcylation was required for resistance to anoikis and the anchorage-independent growth of breast cancer cells. However, the precise roles of this modification on the development of malignancy are yet to be elucidated. Therefore, in the present study, the effects of inhibiting O-GlcNAc on the malignant transformation of MCF-7 breast cancer cells under different culture conditions were determined, using monolayer (primary growth), anoikis resistance (spheroid growth) and reseeding (secondary growth) to mimic the metastatic process. Decreasing O-GlcNAc levels using small interfering (si)RNA targeting OGT resulted in a reduction in cell viability and invasiveness in anoikis resistant and reseeding conditions. Furthermore, gel-free quantitative proteomics was performed to identify the proteins affected by a reduction of O-GlcNAc. A total of 317 proteins were identified and compared, and the expression of 162 proteins was altered >1.5 fold in the siOGT treated cells compared with the siScamble (siSC) treated cells. Notably, 100 proteins involved in cellular metabolism, cellular localization, stress responses and gene expression were significantly altered in the reseeding condition. Among these differentially expressed proteins, the levels of small nuclear ribonucleoprotein Sm D1 exhibited the largest decrease in expression following knockdown of OGT, and this reduction in expression was associated with a significant decrease in the levels of mTOR expression, a protein which promotes tumor growth and progression. Taken together, the results of the present study demonstrate that decreasing O-GlcNAcylation altered protein expression, and ultimately influenced the metastatic processes, particulary regarding the invasion and reattached growth of MCF-7 breast cancer cells.
Collapse
Affiliation(s)
| | | | - Kittirat Saharat
- Laboratory of Biochemistry, Chulabhorn Research Institue, Bangkok 10210, Thailand
| | - Chantragan Srisomsap
- Laboratory of Biochemistry, Chulabhorn Research Institue, Bangkok 10210, Thailand
| | - Jisnuson Svasti
- Laboratory of Biochemistry, Chulabhorn Research Institue, Bangkok 10210, Thailand
| | | |
Collapse
|
13
|
Chanteloup G, Cordonnier M, Isambert N, Bertaut A, Marcion G, Garrido C, Gobbo J. Membrane-bound exosomal HSP70 as a biomarker for detection and monitoring of malignant solid tumours: a pilot study. Pilot Feasibility Stud 2020; 6:35. [PMID: 32161659 PMCID: PMC7053097 DOI: 10.1186/s40814-020-00577-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 02/24/2020] [Indexed: 12/24/2022] Open
Abstract
Background Cancer is the second leading cause of death globally. Early detection and disease management lead to a better survival rate. Consequently, discovery of novel methods in cancer early diagnosis is a field of active research. Minimally invasive liquid biopsies are generating growing interest. Circulating tumour cells (CTCs) have been identified in patients' blood; nevertheless, these cells are rare and heterogeneous. Exosomes are extracellular nanovesicles released into the extracellular environment via the endosomal vesicle pathway and found in different body fluids. Exosomes deliver bioactive cargo such as proteins, mRNA and miRNA to recipient cells in the tumour environment. We have recently shown that heat shock protein 70 (HSP70) is detected in the membrane of tumour-derived exosomes, in contrast to normal cells. One single cancer cell can release thousands of HSP70-exosomes, facilitating detection. The aim of the pilot study ExoDiag is to determine whether it is possible to detect and quantify HSP70-exosomes in blood in patients with solid cancers. Methods Bicentric pilot study that will include 60 adult patients with metastatic and non-metastatic solid tumours and 20 healthy volunteers. Exosomes will be isolated from blood and urine samples, and HSP70 concentration will be determined. Patients will be followed for 1 year. The study is sponsored by Georges-François Leclerc Centre and is currently ongoing. Discussion We expect to demonstrate that HSP70-exosomes could be a powerful tool to diagnose cancer and to guide clinicians in therapeutic decision-making, improving patient's care. Trial Registration ClinicalTrials.gov identifier NCT02662621. Registered 20 January 2016, https://clinicaltrials.gov/ct2/show/study/NCT02662621?term=NCT02662621&rank=1.
Collapse
Affiliation(s)
- Gaétan Chanteloup
- 1Laboratoire d'Excellence LipSTIC, UMR 1231, INSERM, Dijon, France.,2Faculty of Medicine and Pharmacy, University of Burgundy, Dijon, France
| | - Marine Cordonnier
- 1Laboratoire d'Excellence LipSTIC, UMR 1231, INSERM, Dijon, France.,2Faculty of Medicine and Pharmacy, University of Burgundy, Dijon, France
| | - Nicolas Isambert
- 3Pôle Régional de Cancérologie, CHU de Poitiers Poitiers, INSERM U-1084, University de Poitiers, Poitiers, France
| | - Aurélie Bertaut
- Unit of Methodology, Biostatistics and Data Management, Georges-François Leclerc Centre, Dijon, France
| | - Guillaume Marcion
- 1Laboratoire d'Excellence LipSTIC, UMR 1231, INSERM, Dijon, France.,2Faculty of Medicine and Pharmacy, University of Burgundy, Dijon, France
| | - Carmen Garrido
- 1Laboratoire d'Excellence LipSTIC, UMR 1231, INSERM, Dijon, France.,2Faculty of Medicine and Pharmacy, University of Burgundy, Dijon, France.,Department of Medical Oncology, Early Phase Unit, Georges-François Leclerc Centre, 1, Rue du Professeur Marion, 21079 Dijon, France
| | - Jessica Gobbo
- 1Laboratoire d'Excellence LipSTIC, UMR 1231, INSERM, Dijon, France.,2Faculty of Medicine and Pharmacy, University of Burgundy, Dijon, France.,5CIC-1432, INSERM, Dijon, France.,Department of Medical Oncology, Early Phase Unit, Georges-François Leclerc Centre, 1, Rue du Professeur Marion, 21079 Dijon, France
| |
Collapse
|
14
|
Heat Shock Proteins Are Essential Components in Transformation and Tumor Progression: Cancer Cell Intrinsic Pathways and Beyond. Int J Mol Sci 2019; 20:ijms20184507. [PMID: 31514477 PMCID: PMC6769451 DOI: 10.3390/ijms20184507] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 09/06/2019] [Accepted: 09/07/2019] [Indexed: 02/08/2023] Open
Abstract
Heat shock protein (HSP) synthesis is switched on in a remarkably wide range of tumor cells, in both experimental animal systems and in human cancer, in which these proteins accumulate in high levels. In each case, elevated HSP concentrations bode ill for the patient, and are associated with a poor outlook in terms of survival in most cancer types. The significance of elevated HSPs is underpinned by their essential roles in mediating tumor cell intrinsic traits such as unscheduled cell division, escape from programmed cell death and senescence, de novo angiogenesis, and increased invasion and metastasis. An increased HSP expression thus seems essential for tumorigenesis. Perhaps of equal significance is the pronounced interplay between cancer cells and the tumor milieu, with essential roles for intracellular HSPs in the properties of the stromal cells, and their roles in programming malignant cells and in the release of HSPs from cancer cells to influence the behavior of the adjacent tumor and infiltrating the normal cells. These findings of a triple role for elevated HSP expression in tumorigenesis strongly support the targeting of HSPs in cancer, especially given the role of such stress proteins in resistance to conventional therapies.
Collapse
|
15
|
Hollern DP, Swiatnicki MR, Rennhack JP, Misek SA, Matson BC, McAuliff A, Gallo KA, Caron KM, Andrechek ER. E2F1 Drives Breast Cancer Metastasis by Regulating the Target Gene FGF13 and Altering Cell Migration. Sci Rep 2019; 9:10718. [PMID: 31341204 PMCID: PMC6656723 DOI: 10.1038/s41598-019-47218-0] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 07/12/2019] [Indexed: 12/27/2022] Open
Abstract
In prior work we demonstrated that loss of E2F transcription factors inhibits metastasis. Here we address the mechanisms for this phenotype and identify the E2F regulated genes that coordinate tumor cell metastasis. Transcriptomic profiling of E2F1 knockout tumors identified a role for E2F1 as a master regulator of a suite of pro-metastatic genes, but also uncovered E2F1 target genes with an unknown role in pulmonary metastasis. High expression of one of these genes, Fgf13, is associated with early human breast cancer metastasis in a clinical dataset. Together these data led to the hypothesis that Fgf13 is critical for breast cancer metastasis, and that upregulation of Fgf13 may partially explain how E2F1 promotes breast cancer metastasis. To test this hypothesis we ablated Fgf13 via CRISPR. Deletion of Fgf13 in a MMTV-PyMT breast cancer cell line reduces colonization of the lungs in a tail vein injection. In addition, loss of Fgf13 reduced in vitro cell migration, suggesting that Fgf13 may be critical for tumor cells to escape the primary tumor and to colonize the distal sites. The significance of this work is twofold: we have both uncovered genomic features by which E2F1 regulates metastasis and we have identified new pro-metastatic functions for the E2F1 target gene Fgf13.
Collapse
Affiliation(s)
- Daniel P Hollern
- Lineberger Comprehensive Cancer Center University of North Carolina, Chapel Hill, United States
| | - Matthew R Swiatnicki
- Department of Physiology, Michigan State University, East Lansing, United States
| | - Jonathan P Rennhack
- Department of Physiology, Michigan State University, East Lansing, United States
| | - Sean A Misek
- Department of Physiology, Michigan State University, East Lansing, United States
| | - Brooke C Matson
- University of North Carolina Department of Cell Biology, Chapel Hill, United States
| | - Andrew McAuliff
- Department of Physiology, Michigan State University, East Lansing, United States
| | - Kathleen A Gallo
- Department of Physiology, Michigan State University, East Lansing, United States
| | - Kathleen M Caron
- Lineberger Comprehensive Cancer Center University of North Carolina, Chapel Hill, United States
| | - Eran R Andrechek
- Department of Physiology, Michigan State University, East Lansing, United States.
| |
Collapse
|
16
|
Xie YH, Li LY, He JZ, Xu XE, Liao LD, Zhang Q, Xie JJ, Xu LY, Li EM. Heat shock protein family B member 1 facilitates ezrin activation to control cell migration in esophageal squamous cell carcinoma. Int J Biochem Cell Biol 2019; 112:79-87. [PMID: 31082616 DOI: 10.1016/j.biocel.2019.05.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 05/06/2019] [Accepted: 05/08/2019] [Indexed: 02/05/2023]
Abstract
Ezrin plays an important role in the development and progression of human esophageal squamous cell carcinoma (ESCC), providing a link between the cortical actin cytoskeleton and the plasma membrane to govern membrane structure and protrusions. However, the mechanism by which ezrin is activated still remains unknown in ESCC. Here, we identify a novel interaction between ezrin and heat shock protein family B (small) member 1 (HSPB1) in ESCC cells by mass spectroscopy and co-immunoprecipitation. HSPB1 only interacts with inactive ezrin and binds to the α-helical coiled coil region of ezrin. Knockdown of HSPB1 resulted to the decline of phosphorylation at ezrin Thr567, markedly suppressing the ability of ezrin to bind to the actin cytoskeleton and migration of ESCC cells. Furthermore, neither the constitutively active phosphomimetic ezrin T567D, nor inactivated ezrin T567A could restore cell migration following HSPB1 knockdown. Low HSPB1 expression was associated with favorable overall survival of ESCC patients. Taken together, HSPB1, as an important partner, participates in the activation of ezrin and merits further evaluation as a novel therapeutic target against human ESCC.
Collapse
Affiliation(s)
- Ying-Hua Xie
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou 515041, Guangdong, PR China; Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, Guangdong, PR China
| | - Li-Yan Li
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou 515041, Guangdong, PR China; Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, Guangdong, PR China
| | - Jian-Zhong He
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou 515041, Guangdong, PR China; Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, Guangdong, PR China
| | - Xiu-E Xu
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou 515041, Guangdong, PR China; Institute of Oncologic Pathology, Shantou University Medical College, Shantou 515041, Guangdong, PR China
| | - Lian-Di Liao
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou 515041, Guangdong, PR China; Institute of Oncologic Pathology, Shantou University Medical College, Shantou 515041, Guangdong, PR China
| | - Qiang Zhang
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou 515041, Guangdong, PR China; Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, Guangdong, PR China
| | - Jian-Jun Xie
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou 515041, Guangdong, PR China; Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, Guangdong, PR China
| | - Li-Yan Xu
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou 515041, Guangdong, PR China; Institute of Oncologic Pathology, Shantou University Medical College, Shantou 515041, Guangdong, PR China.
| | - En-Min Li
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou 515041, Guangdong, PR China; Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, Guangdong, PR China.
| |
Collapse
|
17
|
Narayanankutty V, Narayanankutty A, Nair A. Heat Shock Proteins (HSPs): A Novel Target for Cancer Metastasis Prevention. Curr Drug Targets 2019; 20:727-737. [DOI: 10.2174/1389450120666181211111815] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 11/11/2018] [Accepted: 11/27/2018] [Indexed: 02/08/2023]
Abstract
Background:
Heat shock proteins (HSPs) are predominant molecular chaperones which are
actively involved in the protein folding; which is essential in protecting the structure and functioning
of proteins during various stress conditions. Though HSPs have important physiological roles, they
have been well known for their roles in various pathogenic conditions such as carcinogenesis; however,
limited literature has consolidated its potential as an anti-metastatic drug target.
Objectives:
The present review outlines the role of different HSPs on cancer progression and metastasis;
possible role of HSP inhibitors as anti-neoplastic agents is also discussed.
Methods:
The data were collected from PubMed/Medline and other reputed journal databases. The literature
that was too old and had no significant role to the review was then omitted.
Results:
Despite their strong physiological functions, HSPs are considered as good markers for cancer
prognosis and diagnosis. They have control over survival, proliferation and progression events of cancer
including drug resistance, metastasis, and angiogenesis. Since, neoplastic cells are more dependent
on HSPs for survival and proliferation, the selectivity and specificity of HSP-targeted cancer drugs
remain high. This has made various HSPs potential clinical and experimental targets for cancer prevention.
An array of HSP inhibitors has been in trials and many others are in experimental conditions
as anticancer and anti-metastatic agents. Several natural products are also being investigated for their
efficacy for anticancer and anti-metastatic agents by modulating HSPs.
Conclusion:
Apart from their role as an anticancer drug target, HSPs have shown to be promising targets
for the prevention of cancer progression. Extensive studies are required for the use of these molecules
as anti-metastatic agents. Further studies in this line may yield specific and effective antimetastatic
agents.
Collapse
Affiliation(s)
| | - Arunaksharan Narayanankutty
- Postgraduate & Research Department of Zoology, St. Joseph’s College, Devagiri (Autonomous), Calicut, Kerala- 673 008, India
| | - Anusree Nair
- Cell and Tissue Culture Department, Micro labs, Bangalore, India
| |
Collapse
|
18
|
Bolhassani A, Shahbazi S, Milani A, Nadji SA. Small Heat Shock Proteins B1 and B6: Which One is the Most Effective Adjuvant in Therapeutic HPV Vaccine? IUBMB Life 2018; 70:1002-1011. [DOI: 10.1002/iub.1892] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 06/05/2018] [Accepted: 06/06/2018] [Indexed: 12/25/2022]
Affiliation(s)
- Azam Bolhassani
- Department of Hepatitis and AIDs; Pasteur Institute of Iran; Tehran Iran
| | - Sepideh Shahbazi
- Department of Hepatitis and AIDs; Pasteur Institute of Iran; Tehran Iran
| | - Alireza Milani
- Department of Hepatitis and AIDs; Pasteur Institute of Iran; Tehran Iran
| | - Seyed Alireza Nadji
- Virology Research Center (VRC); National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences; Tehran Iran
| |
Collapse
|
19
|
Han L, Jiang Y, Han D, Tan W. Hsp27 regulates epithelial mesenchymal transition, metastasis and proliferation in colorectal carcinoma. Oncol Lett 2018; 16:5309-5316. [PMID: 30250600 DOI: 10.3892/ol.2018.9286] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Accepted: 01/05/2018] [Indexed: 12/20/2022] Open
Abstract
The primary factor associated with poor survival rate in patients with colorectal carcinoma (CRC) is the presence of metastasis. The underlying molecular mechanisms of CRC metastasis are yet to be fully elucidated. The present study investigated the function of heat shock protein 27 (Hsp27) on the invasion and proliferation of CRC cells. The clinical significance of Hsp27 was evaluated using tissue microarray analysis (n=81). Invasion and metastasis assays were used to determine the function of Hsp27 in CRC metastasis in vitro and in vivo using RNA interference and the ectopic expression of Hsp27. The upregulation of Hsp27 has been frequently identified in CRC tissues. Patients with CRC and a high expression level of Hsp27 have a reduced overall survival rate. Silencing Hsp27 inhibited the growth and invasion of CRC cells in vitro and in vivo, whereas ectopic overexpression of Hsp27 promoted the proliferation and invasion of CRC cells in vitro. Furthermore, depletion of Hsp27 expression inhibited the epithelial-to-mesenchymal transition (EMT), whilst ectopic overexpression of Hsp27 induced EMT. The results of the present study indicated that Hsp27 serves an important function in the aggressiveness of CRC through inducing EMT. Hsp27 suppression may represent a potential therapeutic option for the suppression of CRC progression.
Collapse
Affiliation(s)
- Lu Han
- Department of Surgery, Shanghai Jiangong Hospital, Shanghai 200083, P.R. China
| | - Yong Jiang
- Department of Surgery, Shanghai Jiangong Hospital, Shanghai 200083, P.R. China
| | - Dongxing Han
- Department of Surgery, Shanghai Jiangong Hospital, Shanghai 200083, P.R. China
| | - Weilin Tan
- Department of Surgery, Shanghai Jiangong Hospital, Shanghai 200083, P.R. China
| |
Collapse
|
20
|
Calderwood SK. Heat shock proteins and cancer: intracellular chaperones or extracellular signalling ligands? Philos Trans R Soc Lond B Biol Sci 2018; 373:rstb.2016.0524. [PMID: 29203709 DOI: 10.1098/rstb.2016.0524] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/01/2017] [Indexed: 12/11/2022] Open
Abstract
Heat shock proteins (HSPs) are found at elevated concentrations in tumour cells, and this increase reflects the proteotoxic stress experienced by the cells due to expanding levels of the mutated oncoproteins that drive tumorigenesis. The protection of oncogenic proteins by HSPs offers a window of vulnerability in tumour metabolism that has been exploited using Hsp90-targeting drugs. Such compounds have been shown to cause inhibition and degradation of a wide range of proteins essential for oncogenesis. Recently, Hsp90 has also been shown to be secreted by tumour cells and to interact in autocrine or paracrine manners with the surfaces of adjacent cells, leading to increased growth and metastasis. Future studies will address a number of key questions associated with these findings, including the relative importance of intracellular versus extracellular HSPs in tumorigenesis, as well as overcoming potential problems with normal tissue toxicity associated with Hsp90 drugs. Targeting individual members of HSP families and inactivating extracellular HSPs may be desirable future approaches that offer increased selectivity in targeting HSPs in cancer.This article is part of the theme issue 'Heat shock proteins as modulators and therapeutic targets of chronic disease: an integrated perspective'.
Collapse
Affiliation(s)
- Stuart K Calderwood
- Molecular and Cellular Radiation Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Center for Life Sciences 610, Boston, MA 02115, USA
| |
Collapse
|
21
|
Wiegmans AP, Miranda M, Wen SW, Al-Ejeh F, Möller A. RAD51 inhibition in triple negative breast cancer cells is challenged by compensatory survival signaling and requires rational combination therapy. Oncotarget 2018; 7:60087-60100. [PMID: 27507046 PMCID: PMC5312370 DOI: 10.18632/oncotarget.11065] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Accepted: 07/18/2016] [Indexed: 02/07/2023] Open
Abstract
The molecular rationale to induce synthetic lethality, by targeting defective homologous recombination repair in triple negative breast cancer (TNBC), has proven to have several shortcomings. Not meeting the expected minimal outcomes in clinical trials has highlighted common clinical resistance mechanisms including; increased expression of the target gene PARP1, increased expression or reversion mutation of BRCA1, or up-regulation of the compensatory homologous recombination protein RAD51. Indeed, RAD51 has been demonstrated to be an alternative synthetic lethal target in BRCA1-mutated cancers. To overcome selective pressure on DNA repair pathways, we examined new potential targets within TNBC that demonstrate synthetic lethality in association with RAD51 depletion. We confirmed complementary targets of PARP1/2 and DNA-PK as well as a new synthetic lethality combination with p38. p38 is considered a relevant target in breast cancer, as it has been implicated in resistance to chemotherapy, including tamoxifen. We show that the combination of targeting RAD51 and p38 inhibits cell proliferation both in vitro and in vivo, which was further enhanced by targeting of PARP1. Analysis of the molecular mechanisms revealed that depletion of RAD51 increased ERK1/2 and p38 signaling. Our results highlight a potential compensatory mechanism via p38 that limits DNA targeted therapy.
Collapse
Affiliation(s)
- Adrian P Wiegmans
- Tumor Microenvironment Laboratory, QIMR Berghofer, Herston Rd, Herston QLD 4006, Australia
| | - Mariska Miranda
- Personalized Medicine Laboratory, QIMR Berghofer, Herston Rd, Herston QLD 4006, Australia
| | - Shu Wen Wen
- Tumor Microenvironment Laboratory, QIMR Berghofer, Herston Rd, Herston QLD 4006, Australia
| | - Fares Al-Ejeh
- Personalized Medicine Laboratory, QIMR Berghofer, Herston Rd, Herston QLD 4006, Australia
| | - Andreas Möller
- Tumor Microenvironment Laboratory, QIMR Berghofer, Herston Rd, Herston QLD 4006, Australia.,School of Medicine, University of Queensland, Brisbane, QLD 4072, Australia
| |
Collapse
|
22
|
Analysis of HspB1 (Hsp27) Oligomerization and Phosphorylation Patterns and Its Interaction with Specific Client Polypeptides. Methods Mol Biol 2018; 1709:163-178. [PMID: 29177658 DOI: 10.1007/978-1-4939-7477-1_12] [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: 02/06/2023]
Abstract
Human HspB1 (also denoted as Hsp27) belongs to the family of small (or stress) proteins (sHsps). The family, which contains ten members including αA,B-crystallin polypeptides, is characterized by a conserved C-terminal α-crystallin domain and molecular weights ranging from 20 to 40 kDa. Here, procedures are described for analyzing the dynamic oligomerization and phosphorylation patterns of HspB1 in cells exposed to different environments. Changes in the structural organization of HspB1 can reprogram its interaction with specific partner/client polypeptides. Methods are presented to analyze these interactions using tissue culture cells genetically modified to express different levels of this protein. In addition, the laboratory approaches presented here could be used to test the nine other human sHsp members as well as sHsps from other species.
Collapse
|
23
|
A randomized phase 2 study of a HSP27 targeting antisense, apatorsen with prednisone versus prednisone alone, in patients with metastatic castration resistant prostate cancer. Invest New Drugs 2017; 36:278-287. [PMID: 29250742 DOI: 10.1007/s10637-017-0553-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 12/12/2017] [Indexed: 01/04/2023]
Abstract
Purpose Heat shock protein 27 (Hsp27) is implicated in prostate cancer progression. Apatorsen is a second generation phosphorothioate antisense inhibitor of Hsp27 expression. We evaluated apatorsen in patients with metastatic castration resistant prostate cancer (mCRPC). Experimental design Eligible patients were randomized 1:1 to receive intravenous apatorsen (3 loading doses of 600 mg within 5-9 days followed by weekly doses of 1000 mg) with oral prednisone 5 mg twice daily or prednisone alone. The primary endpoint was disease progression at 12 weeks. Crossover from prednisone alone was allowed after radiographic progression. Results 74 patients received apatorsen + prednisone (n = 36) or prednisone alone (n = 38). Twenty-five patients crossed-over to receive apatorsen + prednisone. Apatorsen treated patients received a median of 19 infusions. 50% of apatorsen + prednisone patients (95% CI: 32.9%, 67.1%) compared with 42% of prednisone patients (95% CI: 26.3%, 59.2%) did not have disease progression at week 12 (P = 0.33). A PSA decline of ≥50% was observed in 47% of apatorsen + prednisone and 24% of prednisone patients (P = 0.04), with a median duration of response of 24.1 weeks (95% CI: 12.0, 52) and 14.0 weeks (95% CI: 4.0, 44.4), respectively. A PSA decline of ≥50% was observed in 5 patients (20%) that received cross-over apatorsen. Infusion reactions were the most commonly reported adverse event occurring in 77% of apatorsen-treated patients. Conclusions Apatorsen + prednisone did not change the proportion of CRPC patients without disease progression at 12 weeks compared to prednisone but was associated with significant PSA declines. Further evaluation of Hsp27 targeting in prostate cancer is warranted.
Collapse
|
24
|
Charmpilas N, Kyriakakis E, Tavernarakis N. Small heat shock proteins in ageing and age-related diseases. Cell Stress Chaperones 2017; 22:481-492. [PMID: 28074336 PMCID: PMC5465026 DOI: 10.1007/s12192-016-0761-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2016] [Revised: 12/19/2016] [Accepted: 12/23/2016] [Indexed: 12/19/2022] Open
Abstract
Small heat shock proteins (sHSPs) are gatekeepers of cellular homeostasis across species, preserving proteome integrity under stressful conditions. Nonetheless, recent evidence suggests that sHSPs are more than molecular chaperones with merely auxiliary role. In contrast, sHSPs have emerged as central lifespan determinants, and their malfunction has been associated with the manifestation of neurological disorders, cardiovascular disease and cancer malignancies. In this review, we focus on the role of sHSPs in ageing and age-associated diseases and highlight the most prominent paradigms, where impairment of sHSP function has been implicated in human pathology.
Collapse
Affiliation(s)
- Nikolaos Charmpilas
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, 70013, Heraklion, Crete, Greece
- Department of Biology, University of Crete, 70013, Heraklion, Crete, Greece
| | - Emmanouil Kyriakakis
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, 70013, Heraklion, Crete, Greece
- Department of Biomedicine, Laboratory for Signal Transduction, Basel University Hospital and University of Basel, Basel, Switzerland
| | - Nektarios Tavernarakis
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, 70013, Heraklion, Crete, Greece.
- Department of Basic Sciences, Faculty of Medicine, University of Crete, 70013, Heraklion, Crete, Greece.
| |
Collapse
|
25
|
Arrigo AP. Mammalian HspB1 (Hsp27) is a molecular sensor linked to the physiology and environment of the cell. Cell Stress Chaperones 2017; 22:517-529. [PMID: 28144778 PMCID: PMC5465029 DOI: 10.1007/s12192-017-0765-1] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Revised: 01/11/2017] [Accepted: 01/14/2017] [Indexed: 12/11/2022] Open
Abstract
Constitutively expressed small heat shock protein HspB1 regulates many fundamental cellular processes and plays major roles in many human pathological diseases. In that regard, this chaperone has a huge number of apparently unrelated functions that appear linked to its ability to recognize many client polypeptides that are subsequently modified in their activity and/or half-life. A major parameter to understand how HspB1 is dedicated to interact with particular clients in defined cellular conditions relates to its complex oligomerization and phosphorylation properties. Indeed, HspB1 structural organization displays dynamic and complex rearrangements in response to changes in the cellular environment or when the cell physiology is modified. These structural modifications probably reflect the formation of structural platforms aimed at recognizing specific client polypeptides. Here, I have reviewed data from the literature and re-analyzed my own studies to describe and discuss these fascinating changes in HspB1 structural organization.
Collapse
Affiliation(s)
- André-Patrick Arrigo
- Apoptosis, Cancer and Development Laboratory, Lyon Cancer Research Center, INSERM U1052-CNRS UMR5286, Centre Léon Bérard, 28 rue Laennec, Lyon, 69008, France.
| |
Collapse
|
26
|
Cordonnier M, Chanteloup G, Isambert N, Seigneuric R, Fumoleau P, Garrido C, Gobbo J. Exosomes in cancer theranostic: Diamonds in the rough. Cell Adh Migr 2017; 11:151-163. [PMID: 28166442 DOI: 10.1080/19336918.2016.1250999] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
During the last 10 years, exosomes, which are small vesicles of 50-200 nm diameter of endosomal origin, have aroused a great interest in the scientific and clinical community for their roles in intercellular communication in almost all physiological and pathological processes. Most cells can potentially release these nanovesicles that share with the parent cell a similar lipid bilayer with transmembrane proteins and a panel of enclosed soluble proteins such as heat shock proteins and genetic material, thus acting as potential nanoshuttles of biomarkers. Exosomes surface proteins allow their targeting and capture by recipient cells, while the exosomes' content can modify the physiological state of recipient cells. Tumor derived exosomes by interacting with other cells of the tumor microenvironment modulate tumor progression, angiogenic switch, metastasis, and immune escape. Targeting tumor-derived exosomes might be an interesting approach in cancer therapy. Furthermore, because a key issue to improve cancer patients' outcome relies on earlier cancer diagnosis (metastases, as opposed to the primary tumor, are responsible for most cancer deaths) exosomes have been put forward as promising biomarker candidates for cancer diagnosis and prognosis. This review summarizes the roles of exosomes in cancer and clinical interest, focusing on the importance of exosomal heat shock proteins (HSP). The challenges of clinical translation of HSP-exosomes as therapeutic targets and biomarkers for early cancer detection are also discussed.
Collapse
Affiliation(s)
- Marine Cordonnier
- a INSERM, UMR 866, Laboratoire d'Excellence LipSTIC , Dijon , France.,b University of Burgundy, Faculty of Medicine and Pharmacy , Dijon , France
| | - Gaëtan Chanteloup
- a INSERM, UMR 866, Laboratoire d'Excellence LipSTIC , Dijon , France.,b University of Burgundy, Faculty of Medicine and Pharmacy , Dijon , France
| | - Nicolas Isambert
- a INSERM, UMR 866, Laboratoire d'Excellence LipSTIC , Dijon , France.,b University of Burgundy, Faculty of Medicine and Pharmacy , Dijon , France.,c Department of Medical Oncology , Georges-François Leclerc Centre , Dijon , France
| | - Renaud Seigneuric
- a INSERM, UMR 866, Laboratoire d'Excellence LipSTIC , Dijon , France.,b University of Burgundy, Faculty of Medicine and Pharmacy , Dijon , France
| | - Pierre Fumoleau
- c Department of Medical Oncology , Georges-François Leclerc Centre , Dijon , France
| | - Carmen Garrido
- a INSERM, UMR 866, Laboratoire d'Excellence LipSTIC , Dijon , France.,b University of Burgundy, Faculty of Medicine and Pharmacy , Dijon , France.,c Department of Medical Oncology , Georges-François Leclerc Centre , Dijon , France.,d Equipe Labellisée par la Ligue Nationale Contre le Cancer , Paris , France
| | - Jessica Gobbo
- a INSERM, UMR 866, Laboratoire d'Excellence LipSTIC , Dijon , France.,b University of Burgundy, Faculty of Medicine and Pharmacy , Dijon , France.,c Department of Medical Oncology , Georges-François Leclerc Centre , Dijon , France
| |
Collapse
|
27
|
Kaigorodova EV, Zavyalova MV, Bychkov VA, Perelmuter VM, Choynzonov EL. Functional state of the Hsp27 chaperone as a molecular marker of an unfavorable course of larynx cancer. Cancer Biomark 2017; 17:145-53. [PMID: 27540972 DOI: 10.3233/cbm-160625] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
BACKGROUND The small heat shock protein 27 kDA (Hsp27) acts as an ATP-independent chaperone in protein folding, but is also implicated in architecture of the cytoskeleton, cell migration, metabolism, cell survival, growth/differentiation, mRNA stabilization, and tumor progression. OBJECTIVE To study the intracellular localization of phosphorylated and non-phosphorylated forms of Hsp27 in squamous cell carcinoma of the larynx (SCCL) and to evaluate their relationship with regional lymphatic metastasis and overall five-year survival. METHODS Tumor biopsies of larynx tissue were collected from 50 patients who were between the ages of 30 to 80 years and had a confirmed diagnosis of squamous cell carcinoma of the larynx. Immunohistochemistry was used to determine the intracellular localization of the phosphorylated and non-phosphorylated forms of Hsp27. RESULTS The study revealed that the Hsp27 chaperone was expressed in both the cytoplasm and the nucleus of tumor cells in SCCL. The biopsies of patients with lymph node metastases showed significantly higher expression of the phosphorylated and unphosphorylated forms of Hsp27 in the nucleus compared to those of patients without lymph node metastases. At the same time, the cytoplasmic expression of Hsp27 in these patients did not differ statistically. Analysis of the overall five-year survival rates showed that negative Hsp27 expression in the nucleus of tumor cells is associated with the survival rate of patients with SCCL. CONCLUSION The nuclear expression of phosphorylated and unphosphorylated forms of Hsp27 is a molecular marker of unfavorable squamous cell carcinoma of the larynx associated with lymphogenous metastasis and decreased total five-year survival.
Collapse
Affiliation(s)
- Evgeniya V Kaigorodova
- Tomsk Cancer Research Institute, Tomsk, Russian Federation.,Laboratory for Translational Cellular and Molecular Biomedicine, Tomsk State University, Tomsk, Russian Federation.,Siberian State Medical University, Tomsk, Russian Federation
| | - Marina V Zavyalova
- Tomsk Cancer Research Institute, Tomsk, Russian Federation.,Laboratory for Translational Cellular and Molecular Biomedicine, Tomsk State University, Tomsk, Russian Federation.,Siberian State Medical University, Tomsk, Russian Federation
| | | | - Vladimir M Perelmuter
- Tomsk Cancer Research Institute, Tomsk, Russian Federation.,Siberian State Medical University, Tomsk, Russian Federation
| | - Evgenii L Choynzonov
- Tomsk Cancer Research Institute, Tomsk, Russian Federation.,Siberian State Medical University, Tomsk, Russian Federation
| |
Collapse
|
28
|
Owen S, Zhao H, Dart A, Wang Y, Ruge F, Gao Y, Wei C, Wu Y, Jiang WG. Heat shock protein 27 is a potential indicator for response to YangZheng XiaoJi and chemotherapy agents in cancer cells. Int J Oncol 2016; 49:1839-1847. [PMID: 27600495 PMCID: PMC5063420 DOI: 10.3892/ijo.2016.3685] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Accepted: 04/22/2016] [Indexed: 12/14/2022] Open
Abstract
Heat shock protein 27 (HSP27) is a member of the heat shock protein family which has been linked to tumour progression and, most interestingly, to chemotherapy resistance in cancer patients. The present study examined the potential interplay between HSP27 and YangZheng XiaoJi, a traditional Chinese medicine used in cancer treatment. A range of cell lines from different tumour types including pancreatic, lung, gastric, colorectal, breast, prostate and ovarian cancer (both wild-type and resistant) were used. Levels and activation of HSP27 and its potential associated signalling pathways were evaluated by protein array and western blotting. Knockdown of HSP27 in cancer cells was achieved using siRNA. Localisation and co-localisation of HSP27 and other proteins were carried out by immunofluorescence. Cell growth and migration were evaluated in their response to a range of chemotherapeutic agents. The present study first identified, by way of protein array, that YangZheng XiaoJi was able to inhibit the phosphorylation of HSP27 protein in cancer cells. We further demonstrated that HSP27, which is co-localised with caspase-9, can be blocked from localising in focal adhesions and co-localising with caspase-9 by YangZheng XiaoJi. The study also demonstrated that YangZheng XiaoJi was able to sensitise cancer cells including those cells that were resistant to chemotherapy, to chemotherapeutic agents. Finally, knocking down HSP27 markedly reduced the migration of cancer cells and increased the sensitivity of cancer cells to the inhibitory effect on cellular migration by YangZheng XiaoJi. YangZheng XiaoJi can act as an agent in first sensitising cancer cells to chemotherapy and secondly to overcome, to some degree, chemoresistance when used in an appropriate fashion in patients who have active HSP27.
Collapse
Affiliation(s)
- Sioned Owen
- Cardiff China Medical Research Collaborative, Cardiff University School of Medicine, Cardiff, UK
| | - Huishan Zhao
- Cardiff China Medical Research Collaborative, Cardiff University School of Medicine, Cardiff, UK
| | - Alwyn Dart
- Cardiff China Medical Research Collaborative, Cardiff University School of Medicine, Cardiff, UK
| | - Yamei Wang
- Cardiff China Medical Research Collaborative, Cardiff University School of Medicine, Cardiff, UK
| | - Fiona Ruge
- Cardiff China Medical Research Collaborative, Cardiff University School of Medicine, Cardiff, UK
| | - Yong Gao
- Yiling Medical Research Institute, Shijiazhuang, Hebei, P.R. China
| | - Cong Wei
- Yiling Medical Research Institute, Shijiazhuang, Hebei, P.R. China
| | - Yiling Wu
- Yiling Medical Research Institute, Shijiazhuang, Hebei, P.R. China
| | - Wen G Jiang
- Cardiff China Medical Research Collaborative, Cardiff University School of Medicine, Cardiff, UK
| |
Collapse
|
29
|
Wang S, Yang S, Vlantis AC, Liu SY, Ng EK, Chan AB, Wu J, Du J, Wei W, Liu X, Liu Z, Ng SK, van Hasselt CA, Tong MC, Chen GG. Expression of Antioxidant Molecules and Heat Shock Protein 27 in Thyroid Tumors. J Cell Biochem 2016; 117:2473-81. [PMID: 26970173 DOI: 10.1002/jcb.25539] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 03/09/2016] [Indexed: 01/18/2023]
Affiliation(s)
- Shanshan Wang
- Department of Otorhinolaryngology; Head and Neck Surgery, The Chinese University of Hong Kong; Shatin, NT Hong Kong China
| | - Shucui Yang
- Department of Surgery; The Chinese University of Hong Kong; Shatin, NT Hong Kong China
| | - Alexander C. Vlantis
- Department of Otorhinolaryngology; Head and Neck Surgery, The Chinese University of Hong Kong; Shatin, NT Hong Kong China
| | - Shirley Y.W. Liu
- Department of Surgery; The Chinese University of Hong Kong; Shatin, NT Hong Kong China
| | - Enders K.W. Ng
- Department of Surgery; The Chinese University of Hong Kong; Shatin, NT Hong Kong China
| | - Amy B.W. Chan
- Department of Anatomical and Cellular Pathology; The Chinese University of Hong Kong, Prince of Wales Hospital; Shatin, NT Hong Kong China
| | - Juekun Wu
- Department of Thyroid and Breast Surgery; The Third Affiliated Hospital of Sun Yat-Sen University; Guangzhou Guangdong 510665 China
| | - Jing Du
- Peking University Shenzhen Hospital; Shenzhen Guangdong 518036 China
| | - Wei Wei
- Peking University Shenzhen Hospital; Shenzhen Guangdong 518036 China
| | - Xiaoling Liu
- Peking University Shenzhen Hospital; Shenzhen Guangdong 518036 China
| | - Zhimin Liu
- Faculty of Basic Medical Sciences; Department of Biochemistry and Molecular Biology, Chongqing Medical University; Chongqing 40016 China
| | - Siu Kwan Ng
- Department of Otorhinolaryngology; Head and Neck Surgery, The Chinese University of Hong Kong; Shatin, NT Hong Kong China
| | - C. Andrew van Hasselt
- Department of Otorhinolaryngology; Head and Neck Surgery, The Chinese University of Hong Kong; Shatin, NT Hong Kong China
| | - Michael C.F. Tong
- Department of Otorhinolaryngology; Head and Neck Surgery, The Chinese University of Hong Kong; Shatin, NT Hong Kong China
| | - George G. Chen
- Department of Surgery; The Chinese University of Hong Kong; Shatin, NT Hong Kong China
| |
Collapse
|
30
|
Batulan Z, Pulakazhi Venu VK, Li Y, Koumbadinga G, Alvarez-Olmedo DG, Shi C, O'Brien ER. Extracellular Release and Signaling by Heat Shock Protein 27: Role in Modifying Vascular Inflammation. Front Immunol 2016; 7:285. [PMID: 27507972 PMCID: PMC4960997 DOI: 10.3389/fimmu.2016.00285] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 07/14/2016] [Indexed: 12/31/2022] Open
Abstract
Heat shock protein 27 (HSP27) is traditionally viewed as an intracellular chaperone protein with anti-apoptotic properties. However, recent data indicate that a number of heat shock proteins, including HSP27, are also found in the extracellular space where they may signal via membrane receptors to alter gene transcription and cellular function. Therefore, there is increasing interest in better understanding how HSP27 is released from cells, its levels and composition in the extracellular space, and the cognate cell membrane receptors involved in effecting cell signaling. In this paper, the knowledge to date, as well as some emerging paradigms about the extracellular function of HSP27 is presented. Of particular interest is the role of HSP27 in attenuating atherogenesis by modifying lipid uptake and inflammation in the plaque. Moreover, the abundance of HSP27 in serum is an emerging new biomarker for ischemic events. Finally, HSP27 replacement therapy may represent a novel therapeutic opportunity for chronic inflammatory disorders, such as atherosclerosis.
Collapse
Affiliation(s)
- Zarah Batulan
- Vascular Biology Laboratory, Health Research Innovation Centre, Libin Cardiovascular Institute of Alberta, University of Calgary Cumming School of Medicine , Calgary, AB , Canada
| | - Vivek Krishna Pulakazhi Venu
- Vascular Biology Laboratory, Health Research Innovation Centre, Libin Cardiovascular Institute of Alberta, University of Calgary Cumming School of Medicine , Calgary, AB , Canada
| | - Yumei Li
- Vascular Biology Laboratory, Health Research Innovation Centre, Libin Cardiovascular Institute of Alberta, University of Calgary Cumming School of Medicine , Calgary, AB , Canada
| | - Geremy Koumbadinga
- Vascular Biology Laboratory, Health Research Innovation Centre, Libin Cardiovascular Institute of Alberta, University of Calgary Cumming School of Medicine , Calgary, AB , Canada
| | - Daiana Gisela Alvarez-Olmedo
- Oncology Laboratory, Institute for Experimental Medicine and Biology of Cuyo (IMBECU), CCT CONICET , Mendoza , Argentina
| | - Chunhua Shi
- Vascular Biology Laboratory, Health Research Innovation Centre, Libin Cardiovascular Institute of Alberta, University of Calgary Cumming School of Medicine , Calgary, AB , Canada
| | - Edward R O'Brien
- Vascular Biology Laboratory, Health Research Innovation Centre, Libin Cardiovascular Institute of Alberta, University of Calgary Cumming School of Medicine , Calgary, AB , Canada
| |
Collapse
|
31
|
Chi KN, Yu EY, Jacobs C, Bazov J, Kollmannsberger C, Higano CS, Mukherjee SD, Gleave ME, Stewart PS, Hotte SJ. A phase I dose-escalation study of apatorsen (OGX-427), an antisense inhibitor targeting heat shock protein 27 (Hsp27), in patients with castration-resistant prostate cancer and other advanced cancers. Ann Oncol 2016; 27:1116-1122. [PMID: 27022067 DOI: 10.1093/annonc/mdw068] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Accepted: 02/15/2016] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Heat shock protein 27 (Hsp27) is a chaperone protein that regulates cell survival via androgen receptor and other signaling pathways, thereby mediating cancer progression. Apatorsen (OGX-427) is a 2'-methoxyethyl-modified antisense oligonucleotide that inhibits Hsp27 expression. This study evaluated the safety profile and recommended phase II dosing of apatorsen in patients with advanced cancer. PATIENTS AND METHODS Patients with castration-resistant prostate (CRPC), breast, ovary, lung, or bladder cancer were enrolled to this phase I dose-escalation study. Apatorsen was administered i.v. weekly in 21-day cycles following 3 loading doses and over 5 dose levels (200-1000 mg). Apatorsen plasma concentrations, circulating tumor cells (CTCs) and CTC Hsp27 expression, and serum Hsp27 levels were evaluated. RESULTS Forty-two patients were accrued, of which 52% had CRPC. Patients were heavily pretreated, with 57% having had ≥3 prior chemotherapy regimens. During the loading dose/cycle 1 and overall study period, 93% and 100% of patients (N = 42) experienced treatment-related adverse events, respectively; most were grade 1-2 and included chills, pruritus, flushing, prolonged aPTT, lymphopenia, and anemia. One patient experienced a dose-limiting toxicity at the 600 mg dose level (intracranial hemorrhage in a previously undiagnosed brain metastasis). A maximum tolerated dose was not defined. Apatorsen Cmax increased proportionally with dose. Decreases in tumor markers and declines in CTCs were observed, with a prostate-specific antigen decline >%50% occurring in 10% of patients with CRPC; 29/39 assessable patients (74%) had reductions from ≥5 CTC/7.5 ml at baseline to <5 CTC/7.5 ml post-treatment. Twelve patients had stable measurable disease as best response. CONCLUSIONS Apatorsen was tolerated at the highest dose evaluated (1000 mg). Single-agent activity was suggested by changes in tumor markers, CTC, and stable measurable disease. Phase II studies evaluating apatorsen are underway. CLINICALTRIALSGOV ID NCT00487786.
Collapse
Affiliation(s)
- K N Chi
- Department of Medical Oncology, British Columbia Cancer Agency, Vancouver; Department of Urologic Sciences, Vancouver Prostate Center, University of British Columbia, Vancouver, Canada.
| | - E Y Yu
- University of Washington, Fred Hutchinson Cancer Research Center, Seattle
| | - C Jacobs
- Clinical Development, OncoGenex Pharmaceuticals, Inc., Bothell, USA
| | - J Bazov
- Department of Urologic Sciences, Vancouver Prostate Center, University of British Columbia, Vancouver, Canada
| | - C Kollmannsberger
- Department of Medical Oncology, British Columbia Cancer Agency, Vancouver
| | - C S Higano
- University of Washington, Fred Hutchinson Cancer Research Center, Seattle
| | - S D Mukherjee
- Department of Medical Oncology, Juravinski Cancer Centre, Hamilton, Canada
| | - M E Gleave
- Department of Urologic Sciences, Vancouver Prostate Center, University of British Columbia, Vancouver, Canada
| | - P S Stewart
- Clinical Development, OncoGenex Pharmaceuticals, Inc., Bothell, USA
| | - S J Hotte
- Department of Medical Oncology, Juravinski Cancer Centre, Hamilton, Canada
| |
Collapse
|
32
|
Zhang Y, Tao X, Jin G, Jin H, Wang N, Hu F, Luo Q, Shu H, Zhao F, Yao M, Fang J, Cong W, Qin W, Wang C. A Targetable Molecular Chaperone Hsp27 Confers Aggressiveness in Hepatocellular Carcinoma. Am J Cancer Res 2016; 6:558-70. [PMID: 26941848 PMCID: PMC4775865 DOI: 10.7150/thno.14693] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Accepted: 01/11/2016] [Indexed: 12/31/2022] Open
Abstract
Heat shock protein 27 (Hsp27) is an ATP-independent molecular chaperone and confers survival advantages and resistance to cancer cells under stress conditions. The effects and molecular mechanisms of Hsp27 in HCC invasion and metastasis are still unclear. In this study, hepatocellular carcinoma (HCC) tissue array (n = 167) was used to investigate the expression and prognostic relevance of Hsp27 in HCC patients. HCC patients with high expression of Hsp27 exhibited poor prognosis. Overexpression of Hsp27 led to the forced invasion of HCC cells, whereas silencing Hsp27 attenuated invasion and metastasis of HCC cells in vitro and in vivo. We revealed that Hsp27 activated Akt signaling, which in turn promoted MMP2 and ITGA7 expression and HCC metastasis. We further observed that targeting Hsp27 using OGX-427 obviously suppressed HCC metastasis in two metastatic models. These findings indicate that Hsp27 is a useful predictive factor for prognosis of HCC and it facilitates HCC metastasis through Akt signaling. Targeting Hsp27 with OGX-427 may represent an attractive therapeutic option for suppressing HCC metastasis.
Collapse
|
33
|
Calderwood SK, Gong J. Heat Shock Proteins Promote Cancer: It's a Protection Racket. Trends Biochem Sci 2016; 41:311-323. [PMID: 26874923 DOI: 10.1016/j.tibs.2016.01.003] [Citation(s) in RCA: 275] [Impact Index Per Article: 30.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Revised: 01/06/2016] [Accepted: 01/19/2016] [Indexed: 12/20/2022]
Abstract
Heat shock proteins (HSP) are expressed at high levels in cancer and form a fostering environment that is essential for tumor development. Here, we review the recent data in this area, concentrating mainly on Hsp27, Hsp70, and Hsp90. The overriding role of HSPs in cancer is to stabilize the active functions of overexpressed and mutated cancer genes. Thus, elevated HSPs are required for many of the traits that underlie the morbidity of cancer, including increased growth, survival, and formation of secondary cancers. In addition, HSPs participate in the evolution of cancer treatment resistance. HSPs are also released from cancer cells and influence malignant properties by receptor-mediated signaling. Current data strongly support efforts to target HSPs in cancer treatment.
Collapse
Affiliation(s)
- Stuart K Calderwood
- Department of Radiation Oncology, Harvard Medical School at Beth Israel Deaconess Medical Center. CLS610, 300 Brookline Avenue, Boston, MA 02215, USA.
| | - Jianlin Gong
- Department of Medicine, Boston University, Boston, MA 02118, USA
| |
Collapse
|
34
|
Pendharkar N, Gajbhiye A, Taunk K, RoyChoudhury S, Dhali S, Seal S, Mane A, Abhang S, Santra MK, Chaudhury K, Rapole S. Quantitative tissue proteomic investigation of invasive ductal carcinoma of breast with luminal B HER2 positive and HER2 enriched subtypes towards potential diagnostic and therapeutic biomarkers. J Proteomics 2015; 132:112-30. [PMID: 26642762 DOI: 10.1016/j.jprot.2015.11.024] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Revised: 10/16/2015] [Accepted: 11/26/2015] [Indexed: 02/09/2023]
Abstract
Worldwide, breast cancer is one of the frequently diagnosed cancers in women with high mortality if not diagnosed at early stage. Although biomarker discoveries through various proteomic approaches have been studied in breast cancer, a limited number of studies have explored the invasive ductal carcinoma with Luminal B HER2 positive (LB) and HER2 enriched (HE) subtypes. The present study employed the complementary quantitative proteomic approaches to find a panel of markers that could discriminate LB and HE subtypes as well as early (ES) and late stages (LS) of these subtypes. A total of 67 and 68 differentially expressed proteins were identified by DIGE for the subtype and stage wise categories, respectively. Multivariate statistical analysis was employed to identify the set of most significant proteins, which could discriminate between these two subtypes and also early and late stages under study. Immunoblotting and MRM based validation in a separate cohort of samples confirmed that panel of biosignatures for LB are APOA1, GELS, HS90B, EF1A1, NHRF1 and PRDX3 and for HE are PRDX1, CATD, CALR, ATPB and CH60. For the diagnosis of early and late stages the potential markers are TPM4, CATD, PRDX3, ANXA3, HSPB1 and CALR, TRFE, GELS, CH60, CAPG, NHRF1, 1433G, GRP78 respectively.
Collapse
Affiliation(s)
- Namita Pendharkar
- Proteomics Lab, National Centre for Cell Science, Ganeshkhind, Pune 411007, MH, India; B. J. Medical College, Sassoon Hospital, Pune 411001, MH, India
| | - Akshada Gajbhiye
- Proteomics Lab, National Centre for Cell Science, Ganeshkhind, Pune 411007, MH, India
| | - Khushman Taunk
- Proteomics Lab, National Centre for Cell Science, Ganeshkhind, Pune 411007, MH, India
| | - Sourav RoyChoudhury
- School of Medical Science and Technology, Indian Institute of Technology, Kharagpur 721302, WB, India
| | - Snigdha Dhali
- Proteomics Lab, National Centre for Cell Science, Ganeshkhind, Pune 411007, MH, India
| | | | - Anupama Mane
- Grant Medical Foundation, Ruby Hall Clinic, Pune 411001, MH, India
| | | | - Manas K Santra
- Proteomics Lab, National Centre for Cell Science, Ganeshkhind, Pune 411007, MH, India
| | - Koel Chaudhury
- School of Medical Science and Technology, Indian Institute of Technology, Kharagpur 721302, WB, India
| | - Srikanth Rapole
- Proteomics Lab, National Centre for Cell Science, Ganeshkhind, Pune 411007, MH, India.
| |
Collapse
|
35
|
Introducing differential expression of human heat shock protein 27 in hepatocellular carcinoma: moving toward identification of cancer biomarker. Tumour Biol 2015; 37:715-21. [PMID: 26242269 DOI: 10.1007/s13277-015-3858-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Accepted: 07/28/2015] [Indexed: 10/23/2022] Open
Abstract
Previously, it has to be acknowledged that overexpressed heat shock protein B27 (HSPB27) have been implicated in the etiology of wide range of human cancers. However, the molecular mechanism leading to the disease initiation to progression in liver cancer is still unknown. Present work was undertaken to investigate the differentially expressed HSPB27 in association with those damages that lead to liver cancer development. For the identification of liver cancer biomarker, samples were subjected to comparative proteomic analysis using two-dimensional gel electrophoresis (2-DE) and were further validated by Western blot and immunohistochemical analysis. After validation, in silico studies were applied to demonstrate the significantly induced phosphorylated and S-nitrosylated signals. The later included the interacting partner of HSPB27, i.e., mitogen-activated protein kinase-3 and 5 (MAPK3 and 5), ubiquitin C (UBC), v-akt murine thymoma viral oncogene homolog 1 (AKT1), mitogen-activated protein kinase 14 (MAPK14), and tumor protein p53 (TP53), which bestowed with critical capabilities, namely, apoptosis, cell cycling, stress activation, tumor suppression, cell survival, angiogenesis, proliferation, and stress resistance. Taking together, these results shed new light on the potential biomarker HSPB27 that overexpression of HSPB27 did lead to upregulation of their interacting partner that together demonstrate their possible role as a novel tumor progressive agent for the treatment of metastasis in liver cancer. HSPB27 is a promising diagnostic marker for liver cancer although further large-scale studies are required. Also, molecular profiling may help pave the road to the discovery of new therapies.
Collapse
|
36
|
Tormos AM, Taléns-Visconti R, Sastre J. Regulation of cytokinesis and its clinical significance. Crit Rev Clin Lab Sci 2015; 52:159-67. [DOI: 10.3109/10408363.2015.1012191] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
37
|
Abstract
Heat shock proteins are molecular chaperones with a central role in protein folding and cellular protein homeostasis. They also play major roles in the development of cancer and in recent years have emerged as promising therapeutic targets. In this review, we discuss the known molecular mechanisms of various heat shock protein families and their involvement in cancer and in particular, multiple myeloma. In addition, we address the current progress and challenges in pharmacologically targeting these proteins as anti-cancer therapeutic strategies.
Collapse
|
38
|
Arrigo AP, Ducarouge B, Lavial F, Gibert B. Immense Cellular Implications Associated to Small Stress Proteins Expression: Impacts on Human Pathologies. HEAT SHOCK PROTEINS 2015. [DOI: 10.1007/978-3-319-16077-1_2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
39
|
Bakthisaran R, Tangirala R, Rao CM. Small heat shock proteins: Role in cellular functions and pathology. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2014; 1854:291-319. [PMID: 25556000 DOI: 10.1016/j.bbapap.2014.12.019] [Citation(s) in RCA: 312] [Impact Index Per Article: 28.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2014] [Revised: 12/16/2014] [Accepted: 12/18/2014] [Indexed: 01/18/2023]
Abstract
Small heat shock proteins (sHsps) are conserved across species and are important in stress tolerance. Many sHsps exhibit chaperone-like activity in preventing aggregation of target proteins, keeping them in a folding-competent state and refolding them by themselves or in concert with other ATP-dependent chaperones. Mutations in human sHsps result in myopathies, neuropathies and cataract. Their expression is modulated in diseases such as Alzheimer's, Parkinson's and cancer. Their ability to bind Cu2+, and suppress generation of reactive oxygen species (ROS) may have implications in Cu2+-homeostasis and neurodegenerative diseases. Circulating αB-crystallin and Hsp27 in the plasma may exhibit immunomodulatory and anti-inflammatory functions. αB-crystallin and Hsp20 exhitbit anti-platelet aggregation: these beneficial effects indicate their use as potential therapeutic agents. sHsps have roles in differentiation, proteasomal degradation, autophagy and development. sHsps exhibit a robust anti-apoptotic property, involving several stages of mitochondrial-mediated, extrinsic apoptotic as well as pro-survival pathways. Dynamic N- and C-termini and oligomeric assemblies of αB-crystallin and Hsp27 are important factors for their functions. We propose a "dynamic partitioning hypothesis" for the promiscuous interactions and pleotropic functions exhibited by sHsps. Stress tolerance and anti-apoptotic properties of sHsps have both beneficial and deleterious consequences in human health and diseases. Conditional and targeted modulation of their expression and/or activity could be used as strategies in treating several human disorders. The review attempts to provide a critical overview of sHsps and their divergent roles in cellular processes particularly in the context of human health and disease.
Collapse
Affiliation(s)
- Raman Bakthisaran
- CSIR-Centre for Cellular and Molecular Biology, Uppal Road, Hyderabad 500 007, India
| | - Ramakrishna Tangirala
- CSIR-Centre for Cellular and Molecular Biology, Uppal Road, Hyderabad 500 007, India
| | - Ch Mohan Rao
- CSIR-Centre for Cellular and Molecular Biology, Uppal Road, Hyderabad 500 007, India.
| |
Collapse
|
40
|
Research on the efficacy of Celastrus Orbiculatus in suppressing TGF-β1-induced epithelial-mesenchymal transition by inhibiting HSP27 and TNF-α-induced NF-κ B/Snail signaling pathway in human gastric adenocarcinoma. Altern Ther Health Med 2014; 14:433. [PMID: 25370696 PMCID: PMC4232669 DOI: 10.1186/1472-6882-14-433] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2014] [Accepted: 10/27/2014] [Indexed: 01/13/2023]
Abstract
BACKGROUND Celastrus orbiculatus has been used as a folk medicine in China for the treatment of many diseases. In the laboratory, the ethyl acetate extract of Celastrus orbiculatus (COE) displays a wide range of anticancer functions. However, the inhibition of the metastasis mechanism of COE in gastric cancer cells has not been investigated so far. METHODS The present study was undertaken to determine if the anti-metastasis effect of COE was involved in inhibiting of epithelial-mesenchymal transition (EMT) of human gastric adenocarcinoma SGC-7901 cells. In vitro, a well-established experimental EMT model involving transforming growth factor β1 (TGF-β1) was applied. Viability, invasion and migration, protein and mRNA expression of tumor cells were analyzed by MTT assay, transwell assay, western blot and real-time PCR, respectively. The molecular targets of COE in SGC-7901 cells were investigated by two-dimensional gel electrophoresis (2-DE) and MALDI-TOF-TOF mass spectrometer. Overexpression of heat shock protein 27 (HSP27) was performed by transfected with the recombinant retroviral expression plasmid. In vivo, the anti-metastasis mechanisms of COE in the peritoneal gastric cancer xenograft model was explored and the effect was tested. RESULTS The non-cytostatic concentrations of COE effectively inhibited TGF-β1 induced EMT process in SGC-7901 cells, which is characterized by prevented morphological changes, increased E-cadherin expression and decreased Vimentin, N-cadherin expression. Moreover, COE inhibited invasion and migration induced by TGF-β1. Using a comparative proteomics approach, four proteins were identified as differently expressed, with HSP27 protein being one of the most significantly down-regulated proteins induced by COE. Moreover, the activation of nuclear factor κB (NF-κB)/Snail signaling pathway induced by tumor necrosis factor-α (TNF-α) was also attenuated under the pretreatment of COE. Interestingly, overexpression of HSP27 significantly decreases the inhibitory effect of COE on EMT and the NF-κB/Snail pathway. Furthermore, COE significantly reduced the number of peritoneal metastatic nodules in the peritoneal gastric cancer xenograft model. CONCLUSIONS Taken together, these results suggest that COE inhibits the EMT by suppressing the expression of HSP27, correlating with inhibition of NF-κB/Snail signal pathways in SGC-7901 cells. Based on these results, COE may be considered a novel anti-cancer agent for the treatment of metastasis in gastric cancer.
Collapse
|
41
|
The forkhead transcription factor FOXM1 promotes endocrine resistance and invasiveness in estrogen receptor-positive breast cancer by expansion of stem-like cancer cells. Breast Cancer Res 2014; 16:436. [PMID: 25213081 PMCID: PMC4303117 DOI: 10.1186/s13058-014-0436-4] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2014] [Accepted: 08/28/2014] [Indexed: 02/06/2023] Open
Abstract
INTRODUCTION The forkhead transcription factor FOXM1 coordinates expression of cell cycle-related genes and plays a pivotal role in tumorigenesis and cancer progression. We previously showed that FOXM1 acts downstream of 14-3-3ζ signaling, the elevation of which correlates with a more aggressive tumor phenotype. However, the role that FOXM1 might play in engendering resistance to endocrine treatments in estrogen receptor-positive (ER+) patients when tumor FOXM1 is high has not been clearly defined yet. METHODS We analyzed FOXM1 protein expression by immunohistochemistry in 501 ER-positive breast cancers. We also mapped genome-wide FOXM1, extracellular signal-regulated kinase 2 and ERα binding events by chromatin immunoprecipitation followed by high-throughput sequencing (ChIP-seq) in hormone-sensitive and resistant breast cancer cells after tamoxifen treatment. These binding profiles were integrated with gene expression data derived from cells before and after FOXM1 knockdown to highlight specific FOXM1 transcriptional networks. We also modulated the levels of FOXM1 and newly discovered FOXM1-regulated genes and examined their impact on the cancer stem-like cell population and on cell invasiveness and resistance to endocrine treatments. RESULTS FOXM1 protein expression was high in 20% of the tumors, which correlated with significantly reduced survival in these patients (P = 0.003 by logrank Mantel-Cox test). ChIP-seq analyses revealed that FOXM1 binding sites were enriched at the transcription start site of genes involved in cell-cycle progression, maintenance of stem cell properties, and invasion and metastasis, all of which are associated with a poor prognosis in ERα-positive patients treated with tamoxifen. Integration of binding profiles with gene expression highlighted FOXM1 transcriptional networks controlling cell proliferation, stem cell properties, invasion and metastasis. Increased expression of FOXM1 was associated with an expansion of the cancer stem-like cell population and with increased cell invasiveness and resistance to endocrine treatments. Use of a selective FOXM1 inhibitor proved very effective in restoring endocrine therapy sensitivity and decreasing breast cancer aggressiveness. CONCLUSIONS Collectively, our findings uncover novel roles for FOXM1 and FOXM1-regulated genes in promoting cancer stem-like cell properties and therapy resistance. They highlight the relevance of FOXM1 as a therapeutic target to be considered for reducing invasiveness and enhancing breast cancer response to endocrine treatments.
Collapse
|
42
|
Singh S, Suri A. Targeting the testis-specific heat-shock protein 70-2 (HSP70-2) reduces cellular growth, migration, and invasion in renal cell carcinoma cells. Tumour Biol 2014; 35:12695-706. [PMID: 25213699 DOI: 10.1007/s13277-014-2594-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Accepted: 09/03/2014] [Indexed: 01/24/2023] Open
Abstract
Renal cell carcinoma (RCC) represents one of the most resistant tumors to radiotherapy and chemotherapy. Current therapies for the RCC patients are limited owing to lack of diagnosis and therapeutic treatments. Testis-specific heat-shock protein 70-2 (HSP70-2), a member of HSP70 chaperone family, has been shown to be associated with various cancers. In the present study, we investigated the putative role of HSP70-2 in various malignant properties of the RCC cells. HSP70-2 messenger RNA (mRNA) and protein expression was investigated in A704, ACHN, and Caki-1 cells derived from the RCC patients. We assessed the expression of HSP70-2 gene and protein by reverse transcription-polymerase chain reaction (RT-PCR) and Western blotting, respectively. The expression of HSP70-2 protein was further validated by performing indirect immunofluorescence (IIF) and flow cytometry. The malignant properties of high-grade invasive A704 and Caki-1 cells, such as cellular proliferation, colony formation, migration, invasion, and wound healing, were evaluated by silencing the expression of HSP70-2 gene in these cells. Statistical significance was defined using Student's t test. Our RT-PCR and Western blotting data showed the expression of HSP70-2 in all RCC cells. Our results showed that HSP70-2 was predominantly expressed in cytoplasm and found to be colocalized with endoplasmic reticulum, mitochondria, Golgi body, and plasma membrane but not the nuclear envelope. Knockdown of HSP70-2 expression with specific short hairpin RNA (shRNA) demonstrated significant reduction in cell growth and colony formation. Further, a marked reduction in cell migration and invasion was also observed, indicating the potential role of HSP70-2 in metastasis. Collectively, our data suggest that HSP70-2 plays a key role in cancerous growth and invasive potential of RCC cells. Thus, HSP70-2 could serve as a novel potential therapeutic target for the RCC.
Collapse
Affiliation(s)
- Swarnendra Singh
- Cancer Microarray, Genes and Proteins Laboratory, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi, 110 067, India
| | | |
Collapse
|
43
|
Xue L, Yang L, Jin ZA, Gao F, Kang JQ, Xu GH, Liu B, Li H, Wang XJ, Liu LJ, Wang BL, Liang SH, Ding J. Increased expression of HSP27 inhibits invasion and metastasis in human esophageal squamous cell carcinoma. Tumour Biol 2014; 35:6999-7007. [PMID: 24748206 DOI: 10.1007/s13277-014-1946-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2014] [Accepted: 04/03/2014] [Indexed: 12/19/2022] Open
Abstract
Previous studies have indicated that heat shock protein 27 (HSP27) had high correlation with the development and progression in several tumors. However, the roles of HSP27 in esophageal squamous cell carcinoma (ESCC) were uncertain. The aim in this study is to investigate the potential roles of HSP27 in the metastasis of ESCC. The expression of HSP27 in ESCC tissues and four human esophageal cancer cell lines were examined by immunohistochemistry and Western blotting, respectively. Wound healing assays, transwell assays, and in vivo assays were used to identify the differences of metastasis potential between normal and HSP27 overexpressed cells. HSP27 expression was downregulated in cancer tissue compared to the matched normal tissue. And the positive staining was mainly located in the cytoplasm. Statistical analyses showed that the expression of HSP27 in ESCC was significantly correlated with the tumor differentiation (P = 0.023), the patient's TNM stage (P = 0.013), lymph metastasis (P = 0.020), and distant metastasis (P = 0.017). HSP27 expression was significantly lower in highly metastatic cells than the less ones. The metastatic potentials of EC9706-H and EC109-H cells were higher than EC9706-L and EC109-L cells. In vitro and in vivo assays showed that overexpression of HSP27 in highly metastatic cells dramatically decreased their metastatic capacity. This study indicated that the expression level of HSP27 may be inversely correlated with the metastasis behavior of ESCC, and HSP27 may play an important role in this progression. HSP27 may be a potential molecular target for the therapy and prognosis of patients with ESCC.
Collapse
Affiliation(s)
- Lin Xue
- State Key Laboratory of Cancer Biology and Xijing Hospital of Digestive Diseases, Xijing Hospital, Fourth Military Medical University, 127 Changle Western Road, Xi'an, 710032, China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Pavan S, Musiani D, Torchiaro E, Migliardi G, Gai M, Di Cunto F, Erriquez J, Olivero M, Di Renzo MF. HSP27 is required for invasion and metastasis triggered by hepatocyte growth factor. Int J Cancer 2014; 134:1289-1299. [DOI: 10.1002/ijc.28464] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
Abstract
The hepatocyte growth factor (HGF) also known as scatter factor activates cancer cell invasion and metastasis. We show that in ovarian cancer cells HGF induced the phosphorylation of the small heat shock protein of 27 kDa (HSP27) by activating the p38MAPK. HSP27 is increased in many cancers at advanced stage including ovarian cancer and associated with cancer resistance to therapy and poor patients' survival. The phosphorylation of HSP27 regulates both its chaperone activity and its control of cytoskeletal stability. We show that HSP27 was necessary for the remodeling of actin filaments induced by HGF and that motility in vitro depended on the p38MAPK‐MK2 axis. In vivo, HSP27 silencing impaired the ability of the highly metastatic, HGF‐secreting ovarian cancer cells to give rise to spontaneous metastases. This was due to defective motility across the vessel wall and reduced growth. Indeed, HSP27 silencing impaired the ability of circulating ovarian cancer cells to home to the lungs and to form experimental hematogenous metastases and the capability of cancer cells to grow as subcutaneous xenografts. Moreover, HSP27 suppression resulted in the sensitization of xenografts to low doses of the chemotherapeutic paclitaxel, likely because HSP27 protected microtubules from bundling caused by the drug. Altogether, these data show that the HSP27 is required for the proinvasive and prometastatic activity of HGF and suggest that HSP27 might be not only a marker of progression of ovarian cancer, but also a suitable target for therapy.
Collapse
Affiliation(s)
- Simona Pavan
- Department of Oncology University of Torino, School of Medicine Torino Italy
- Laboratory of Cancer Genetics Institute for Cancer Research at Candiolo Torino Italy
| | - Daniele Musiani
- Department of Oncology University of Torino, School of Medicine Torino Italy
- Laboratory of Cancer Genetics Institute for Cancer Research at Candiolo Torino Italy
| | - Erica Torchiaro
- Department of Oncology University of Torino, School of Medicine Torino Italy
- Laboratory of Cancer Genetics Institute for Cancer Research at Candiolo Torino Italy
| | - Giorgia Migliardi
- Department of Oncology University of Torino, School of Medicine Torino Italy
- Laboratory of Molecular Pharmacology Institute for Cancer Research at Candiolo Torino Italy
| | - Marta Gai
- Department of Molecular Biotechnology and Health Sciences Molecular Biotechnology Center University of Torino Torino Italy
| | - Ferdinando Di Cunto
- Department of Molecular Biotechnology and Health Sciences Molecular Biotechnology Center University of Torino Torino Italy
| | - Jessica Erriquez
- Laboratory of Cancer Genetics Institute for Cancer Research at Candiolo Torino Italy
| | - Martina Olivero
- Department of Oncology University of Torino, School of Medicine Torino Italy
- Laboratory of Cancer Genetics Institute for Cancer Research at Candiolo Torino Italy
| | - Maria Flavia Di Renzo
- Department of Oncology University of Torino, School of Medicine Torino Italy
- Laboratory of Cancer Genetics Institute for Cancer Research at Candiolo Torino Italy
| |
Collapse
|
45
|
Arrigo AP, Gibert B. HspB1, HspB5 and HspB4 in Human Cancers: Potent Oncogenic Role of Some of Their Client Proteins. Cancers (Basel) 2014; 6:333-65. [PMID: 24514166 PMCID: PMC3980596 DOI: 10.3390/cancers6010333] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2013] [Revised: 01/03/2014] [Accepted: 01/17/2014] [Indexed: 12/20/2022] Open
Abstract
Human small heat shock proteins are molecular chaperones that regulate fundamental cellular processes in normal unstressed cells as well as in many cancer cells where they are over-expressed. These proteins are characterized by cell physiology dependent changes in their oligomerization and phosphorylation status. These structural changes allow them to interact with many different client proteins that subsequently display modified activity and/or half-life. Nowdays, the protein interactomes of small Hsps are under intense investigations and will represent, when completed, key parameters to elaborate therapeutic strategies aimed at modulating the functions of these chaperones. Here, we have analyzed the potential pro-cancerous roles of several client proteins that have been described so far to interact with HspB1 (Hsp27) and its close members HspB5 (αB-crystallin) and HspB4 (αA-crystallin).
Collapse
Affiliation(s)
- André-Patrick Arrigo
- Apoptosis, Cancer and Development Laboratory, Lyon Cancer Research Center, INSERM U1052-CNRS UMR5286, Claude Bernard University Lyon 1, Lyon 69008, France.
| | - Benjamin Gibert
- Apoptosis, Cancer and Development Laboratory, Lyon Cancer Research Center, INSERM U1052-CNRS UMR5286, Claude Bernard University Lyon 1, Lyon 69008, France.
| |
Collapse
|
46
|
Kuo PL, Huang MS, Hung JY, Chou SH, Chiang SY, Huang YF, Yang CJ, Tsai MJ, Chang WA, Hsu YL. Synergistic effect of lung tumor-associated dendritic cell-derived HB-EGF and CXCL5 on cancer progression. Int J Cancer 2014; 135:96-108. [PMID: 24346967 DOI: 10.1002/ijc.28673] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2013] [Revised: 11/26/2013] [Accepted: 12/02/2013] [Indexed: 01/19/2023]
Abstract
The interaction between cancer cells and their microenvironment is a paradoxical cycle that exacerbates cancer progression and results in metastasis. Our study investigated the mechanism underlying the synergistic enhancement of release of soluble factors from tumor-associated dendritic cells and its effect on cancer development. The combination of HB-EGF (heparin-binding EGF-like growth factor) and CXCL5 (CXCL5/epithelial neutrophil-activating peptide-78) produced a strong synergistic effect on cancer proliferation, epithelial-mesenchymal transition, migration and invasion. CXCL5 not only potentiated the classical EGFR pathway and the AKT and ERK/RSK1/2 signaling pathways but also increased the phosphorylation of heat shock protein 27 (HSP27), which was slightly increased in A549 cells treated with either HB-EGF or CXCL5 only. Phosphorylated HSP27 stabilized sustained AKT activity by direct interaction, leading to enhanced tumor spheroid formation. Knockdown of HSP27 by shRNA decreased HB-EGF plus CXCL5-mediated tumor spheroid formation in a three-dimensional culture system, suggesting that AKT/HSP27 was required for HB-EGF/CXCL5-mediated cancer progression. Inhibiting RSK also reduces the modulation of c-Fos phosphorylation, Snail upregulation and cell migration by HB-EGF plus CXCL5, suggesting a synergistic effect of ERK/RSK and HB-EGF plus CXCL5 on cell migration. In mice, CXCL5 antibody synergistically enhances the efficiency of the tyrosine kinase inhibitor, gefitinib, without increasing its toxicity. These results provide evidence that elucidates potential cross-points between extracellular signals affecting lung cancer progression. Targeting CXCL5 may provide therapeutic benefits for lung cancer chemotherapy or immunotherapy.
Collapse
Affiliation(s)
- Po-Lin Kuo
- Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan; Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan; Cancer Center, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Song IS, Kang SS, Kim ES, Park HM, Choi CY, Tchah H, Kim JY. Heat shock protein 27 phosphorylation is involved in epithelial cell apoptosis as well as epithelial migration during corneal epithelial wound healing. Exp Eye Res 2014; 118:36-41. [DOI: 10.1016/j.exer.2013.11.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2013] [Revised: 10/18/2013] [Accepted: 11/05/2013] [Indexed: 12/27/2022]
|
48
|
Gurgis FMS, Ziaziaris W, Munoz L. Mitogen-Activated Protein Kinase–Activated Protein Kinase 2 in Neuroinflammation, Heat Shock Protein 27 Phosphorylation, and Cell Cycle: Role and Targeting. Mol Pharmacol 2013; 85:345-56. [DOI: 10.1124/mol.113.090365] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
|
49
|
Prevalence and risk factors of bone metastasis and skeletal related events in patients with primary breast cancer in Japan. Int J Clin Oncol 2013; 19:852-62. [PMID: 24292334 DOI: 10.1007/s10147-013-0643-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2013] [Accepted: 11/06/2013] [Indexed: 12/31/2022]
Abstract
BACKGROUND Bone metastasis (BM) is important for studying systemic spread of breast cancer. It often causes skeletal-related events (SREs) that worsen quality of life. We investigated the prevalence and risk factors for BM and SRE using a dataset from the Breast Oncology Research Network (BORN) in Japan. PATIENTS AND METHODS We collected data on primary breast cancer patients with node-positive or node-negative disease at intermediate to high risk of recurrence. The risk factors affecting the BM-free rate, SRE-free rate and overall survival were analyzed by using the Cox proportional hazard model. RESULTS Data of 1,779 patients who were diagnosed with breast cancer during 2003-2005 were collected from the BORN and 1,708 cases were used for analysis. The median follow-up duration was 5.71 years. BM developed in 193 cases (11.3 %) and the BM-free rate at 5 years was 89.2 %. The annual hazard ratio of BM development differs remarkably according to the tumor subtype. SREs occurred in 133 (68.9 %) out of 193 patients and the SRE-free rate at 5 years was 92.6 %. In the multivariate analysis, clinical stage (P < 0.0001), number of lymph node (LN) metastases (P = 0.0029), tumor subtype (P = 0.034) and progesterone receptor status (P = 0.038) were independently significant risk factors for BM-free rate, but only clinical stage (P < 0.0001) and number of LN metastases (P = 0.0004) significantly correlated with SRE-free rate. CONCLUSIONS This retrospective study clarifies the prevalence and risk factors for BM and SRE in Japanese breast cancer patients. Our results show the importance of considering subtype in the care of BM and SRE.
Collapse
|
50
|
Ma W, Teng Y, Hua H, Hou J, Luo T, Jiang Y. Upregulation of heat shock protein 27 confers resistance to actinomycin D-induced apoptosis in cancer cells. FEBS J 2013; 280:4612-24. [PMID: 23848600 DOI: 10.1111/febs.12432] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2013] [Accepted: 07/04/2013] [Indexed: 02/05/2023]
Affiliation(s)
- Wenbo Ma
- State Key Laboratory of Biotherapy; Section of Signal Transduction and Molecular Targeted Therapy; West China Hospital; Sichuan University; Chengdu China
| | - Yan Teng
- State Key Laboratory of Biotherapy; Section of Signal Transduction and Molecular Targeted Therapy; West China Hospital; Sichuan University; Chengdu China
| | - Hui Hua
- Laboratory of Stem Cell Biology; West China Hospital; Sichuan University; Chengdu China
| | - Jinlin Hou
- State Key Laboratory of Biotherapy; Section of Signal Transduction and Molecular Targeted Therapy; West China Hospital; Sichuan University; Chengdu China
| | - Ting Luo
- Cancer Center; West China Hospital; Sichuan University; Chengdu China
| | - Yangfu Jiang
- State Key Laboratory of Biotherapy; Section of Signal Transduction and Molecular Targeted Therapy; West China Hospital; Sichuan University; Chengdu China
| |
Collapse
|