1
|
Chen Y, Li M, Wu Y. Heat shock protein 22: A new direction for cardiovascular disease (Review). Mol Med Rep 2025; 31:82. [PMID: 39886946 PMCID: PMC11800183 DOI: 10.3892/mmr.2025.13447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 01/15/2025] [Indexed: 02/01/2025] Open
Abstract
Small heat shock proteins (sHSPs) are common molecular chaperone proteins that function in various biological processes, and serve indispensable roles in maintaining cellular protein homeostasis and regulating the hydrolysis of unfolded proteins. HSP22 is a member of the sHSP family that is primarily expressed in the heart and skeletal muscle, as well as in various types of cancer. There have been important findings concerning the role of HSP22 in cardiovascular diseases. The aim of the present study was to provide insights into the various molecular mechanisms by which HSP22 functions in the heart, including oxidative stress, autophagy, apoptosis, the subcellular distribution of proteins and the promoting effect of proteasomes. In addition, drugs and cytokines, including geranylgeranylacetone, can exert protective effects on the heart by regulating the expression of HSP22. Based on increasingly abundant research, HSP22 may be considered a potential therapeutic target in cardiovascular diseases.
Collapse
Affiliation(s)
- Yi Chen
- Department of Cardiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Meng Li
- Department of Cardiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Yanqing Wu
- Department of Cardiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
2
|
Shu B, Wen Y, Lin R, He C, Luo C, Li F. HSPB8-BAG3 chaperone complex modulates cell invasion in intrahepatic cholangiocarcinoma by regulating CASA-mediated Filamin A degradation. Cancer Biol Ther 2024; 25:2396694. [PMID: 39215616 PMCID: PMC11370900 DOI: 10.1080/15384047.2024.2396694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 04/16/2024] [Accepted: 08/13/2024] [Indexed: 09/04/2024] Open
Abstract
The incidence of intrahepatic cholangiocarcinoma (ICC) is steadily rising, and it is associated with a high mortality rate. Clinical samples were collected to detect the expression of HSPB8 and BAG3 in ICC tissues. ICC cells were cultured and transfected with plasmids that overexpressed or silenced specific genes to investigate the impact of gene expression alterations on cell function. qPCR and Western blot techniques were utilized to measure gene and protein expression levels. A wound healing assay was conducted to assess cell migration ability. The Transwell assay was used to assess cell invasion ability. Co-IP was used to verify the binding relationship between HSPB8 and BAG3. The effects of HSPB8 and BAG3 on lung metastasis of tumors in vivo were verified by constructing a metastatic tumor model. Through the above experiments, we discovered that the expressions of HSPB8 and BAG3 were up-regulated in ICC tissues and cells, and their expressions were positively correlated. The metastatic ability of ICC cells could be promoted or inhibited by upregulating or downregulating the expression of BAG3. Furthermore, the HSPB8-BAG3 chaperone complex resulted in the abnormal degradation of Filamin A by activating autophagy. Increased expression of Filamin A inhibits the migration and invasion of ICC cells. Overexpression of HSPB8 and BAG3 in vivo promoted the lung metastasis ability of ICC cells. The HSPB8-BAG3 chaperone complex promotes ICC cell migration and invasion by regulating CASA-mediated degradation of Filamin A, offering insights for enhancing ICC therapeutic strategies.
Collapse
Affiliation(s)
- Bo Shu
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Yu Wen
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Ronghua Lin
- Department of General Surgery, Huichang County People’s Hospital, Huichang, Jiangxi Province, China
| | - Chao He
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Cailan Luo
- Department of Hospital Nursing, Huichang County People’s Hospital, Huichang, Jiangxi Province, China
| | - Fazhao Li
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| |
Collapse
|
3
|
Wang Q, Zhang B, Wang H, Hu M, Feng H, Gao W, Lu H, Tan Y, Dong Y, Xu M, Guo T, Ji X. Identification of a six-gene signature to predict survival and immunotherapy effectiveness of gastric cancer. Front Oncol 2023; 13:1210994. [PMID: 37404760 PMCID: PMC10316024 DOI: 10.3389/fonc.2023.1210994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 06/05/2023] [Indexed: 07/06/2023] Open
Abstract
Background Gastric cancer (GC) ranks as the fifth most prevalent malignancy and the second leading cause of oncologic mortality globally. Despite staging guidelines and standard treatment protocols, significant heterogeneity exists in patient survival and response to therapy for GC. Thus, an increasing number of research have examined prognostic models recently for screening high-risk GC patients. Methods We studied DEGs between GC tissues and adjacent non-tumor tissues in GEO and TCGA datasets. Then the candidate DEGs were further screened in TCGA cohort through univariate Cox regression analyses. Following this, LASSO regression was utilized to generate prognostic model of DEGs. We used the ROC curve, Kaplan-Meier curve, and risk score plot to evaluate the signature's performance and prognostic power. ESTIMATE, xCell, and TIDE algorithm were used to explore the relationship between the risk score and immune landscape relationship. As a final step, nomogram was developed in this study, utilizing both clinical characteristics and a prognostic model. Results There were 3211 DEGs in TCGA, 2371 DEGs in GSE54129, 627 DEGs in GSE66229, and 329 DEGs in GSE64951 selected as candidate genes and intersected with to obtain DEGs. In total, the 208 DEGs were further screened in TCGA cohort through univariate Cox regression analyses. Following this, LASSO regression was utilized to generate prognostic model of 6 DEGs. External validation showed favorable predictive efficacy. We studied interaction between risk models, immunoscores, and immune cell infiltrate based on six-gene signature. The high-risk group exhibited significantly elevated ESTIMATE score, immunescore, and stromal score relative to low-risk group. The proportions of CD4+ memory T cells, CD8+ naive T cells, common lymphoid progenitor, plasmacytoid dentritic cell, gamma delta T cell, and B cell plasma were significantly enriched in low-risk group. According to TIDE, the TIDE scores, exclusion scores and dysfunction scores for low-risk group were lower than those for high-risk group. As a final step, nomogram was developed in this study, utilizing both clinical characteristics and a prognostic model. Conclusion In conclusion, we discovered a 6 gene signature to forecast GC patients' OS. This risk signature proves to be a valuable clinical predictive tool for guiding clinical practice.
Collapse
|
4
|
Zhang K, Yin W, Ma L, Liu Z, Li Q. HSPB8 facilitates prostate cancer progression via activating the JAK/STAT3 signaling pathway. Biochem Cell Biol 2023; 101:1-11. [PMID: 36318825 DOI: 10.1139/bcb-2022-0205] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Prostate cancer (PC) is a clinically and biologically heterogeneous disease that lacks effective treatment. Heat shock protein B8 (HSPB8) is an important factor in the progression of various types of cancer. However, the clinical significance and biological role of HSPB8 in PC are still unclear. In this study, we determined HSPB8 expression in PC tissues by immunohistochemical staining and explored the in vitro functions of HSPB8 using HSPB8 knockdown DU145 and LNcap PC cell lines. The in vivo effect of HSPB8 was explored by a subcutaneous xenograft mice model. The human phospho-kinase array and signal transducer and activator of transcription (STAT) 3 activator were utilized to explore the potential mechanism of HSPB8-induced PC progression. As a result, we found that HSPB8 was abundantly expressed in PC tissues and cell lines. HSPB8 knockdown inhibited cell proliferation and migration, promoted apoptosis and cycle repression, as well as weakened tumorigenesis ability. Mechanistically, we demonstrated that HSPB8 facilitates the malignant phenotypes of PC by activating the Janus kinase/STAT3 signaling pathway. These results proposed that HSPB8 seems to be an attractive therapeutic target for PC patients.
Collapse
Affiliation(s)
- Kan Zhang
- Department of urinary surgery, Ningbo First Hospital, No.59, Liuting Street, Haishu District, Ningbo, Zhejiang Province 315000, China
| | - Weiqi Yin
- Department of urinary surgery, Ningbo First Hospital, No.59, Liuting Street, Haishu District, Ningbo, Zhejiang Province 315000, China
| | - Luping Ma
- Department of urinary surgery, First Affiliated Hospital School of Medicine, Shihezi University, No.107, North 2nd Road, Shihezi, Xinjiang Province 832008, China
| | - Zhili Liu
- Department of urinary surgery, First Affiliated Hospital School of Medicine, Shihezi University, No.107, North 2nd Road, Shihezi, Xinjiang Province 832008, China
| | - Qiang Li
- Department of urinary surgery, First Affiliated Hospital School of Medicine, Shihezi University, No.107, North 2nd Road, Shihezi, Xinjiang Province 832008, China
| |
Collapse
|
5
|
Sun X, Siri S, Hurst A, Qiu H. Heat Shock Protein 22 in Physiological and Pathological Hearts: Small Molecule, Large Potentials. Cells 2021; 11:cells11010114. [PMID: 35011676 PMCID: PMC8750610 DOI: 10.3390/cells11010114] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 12/22/2021] [Accepted: 12/27/2021] [Indexed: 12/22/2022] Open
Abstract
Small heat shock protein 22 (HSP22) belongs to the superfamily of heat shock proteins and is predominantly expressed in the heart, brain, skeletal muscle, and different types of cancers. It has been found that HSP22 is involved in variant cellular functions in cardiomyocytes and plays a vital role in cardiac protection against cardiomyocyte injury under diverse stress. This review summarizes the multiple functions of HSP22 in the heart and the underlying molecular mechanisms through modulating gene transcription, post-translational modification, subcellular translocation of its interacting proteins, and protein degradation, facilitating mitochondrial function, cardiac metabolism, autophagy, and ROS production and antiapoptotic effect. We also discuss the association of HSP22 in cardiac pathologies, including human dilated cardiomyopathy, pressure overload-induced heart failure, ischemic heart diseases, and aging-related cardiac metabolism disorder. The collected information would provide insights into the understanding of the HSP22 in heart diseases and lead to discovering the therapeutic targets.
Collapse
|
6
|
Li X, Sun C, Chen J, Ma JF, Pan YH. ERK-CREB pathway is involved in HSPB8-mediated glioma cell growth and metastatic properties. Exp Mol Pathol 2021:104653. [PMID: 34043982 DOI: 10.1016/j.yexmp.2021.104653] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 04/14/2021] [Accepted: 05/21/2021] [Indexed: 11/29/2022]
Abstract
OBJECTIVE To investigate the mechanism of HSPB8 (heat shock protein beta-8) in the growth and metastatic properties of glioma cells. METHODS HSPB8 expression in glioma tissue and cell was detected via Western blotting. Then, glioma U87 and U251 cell lines were divided into Mock group, Control siRNA group, HSPB8 siRNA-1 group and HSPB8 siRNA-2 group. Cell proliferation was detected using MTT assay, while its invasion, migration and apoptosis were determined by Transwell, wound-healing and flow cytometry, respectively. The expression of HSPB8 and ERK-CREB pathway-related molecules were also measured by Western blotting. Xenograft models were constructed on nude mice, and accordingly, the growth curve of subcutaneous xenograft was prepared. RESULTS In glioma tissues, HSPB8 expression was upregulated with the increasing grade of glioma. Besides, glioma cells in the HSPB8 siRNA-1 group and HSPB8 siRNA-2 group manifested the significant enhancement in apoptotic rates and reductions in its proliferation, migration and invasion compared to those in the Mock group, meanwhile, the expression of HSPB8, p-ERK1/2/ERK1/2 and p-CREB/CREB were downregulated. On the other hand, the tumor growth in the nude mice of Ad-HSPB8 shRNA-1 group and Ad-HSPB8 shRNA-2 group was retarded significantly, with an acute decrease in the tumor weight. CONCLUSION Silencing HSPB8 can inhibit the malignant features, while facilitate the apoptosis of glioma cells, with inactivation of ERK-CREB pathway.
Collapse
Affiliation(s)
- Xia Li
- Center for Diagnosis and Treatment of Neuro-oncology Diseases, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, Hubei, China
| | - Cui Sun
- Center for Diagnosis and Treatment of Neuro-oncology Diseases, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, Hubei, China
| | - Jing Chen
- Center for Diagnosis and Treatment of Neuro-oncology Diseases, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, Hubei, China
| | - Ji-Fen Ma
- Center for Diagnosis and Treatment of Neuro-oncology Diseases, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, Hubei, China
| | - Yi-Heng Pan
- Center for Diagnosis and Treatment of Neuro-oncology Diseases, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, Hubei, China.
| |
Collapse
|
7
|
Jafari E, Safinejad S, Dabiri S, Naghibzadeh-Tahami A. Study of the Relationship between MMP-2 and MMP-9 and Her2/neu Overexpression in Gastric Cancer: Clinico- Pathological Correlations. Asian Pac J Cancer Prev 2021; 22:811-817. [PMID: 33773545 PMCID: PMC8286688 DOI: 10.31557/apjcp.2021.22.3.811] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Indexed: 12/09/2022] Open
Abstract
Background: The relationship between the expressions of matrix metalloproteinases with clinico-pathological data on gastric cancer has been investigated in many countries, but this relationship remains unexplored in Iranian patients. Also, the correlation of the MMPs and the HER-2/neu proto-oncogene with other clinic-pathological variables has been evaluated for several other malignancies, but little effort has been made to shed light on the relationship with gastric cancer. Methods: We investigated MMP-2 and MMP-9 expression and HERE-2/neu overexpression in 48 gastric cancer patients referred to Afzalipour Hospital, associated with Kerman Medical University. Immunohistochemistry staining with rabbit polyclonal antibodies was used. Data statistical analysis was done by SPSS software (Version 20.0). Results: The mean age was 59, most of the patients were male (79.2%), and the average tumor size was larger than 5 centimeters in its greatest diameter. The majority of tumors were of the intestinal subtype and were located in the pyloric and antrum regions (43.8%). Invasion to muscularis properia was seen in 87.5% of the tumors (T3). MMP-2 and MMP-9 were highly expressed in 58.3% and 50% of cases, respectively, and Her-2/neu positivity was 10.4%. MMP-2, MMP-9 and HER-2 were found to have no relation with any clinicopathological parameters. Conclusion: According to the results of this study, MMP-2 and MMP-9 were highly expressed in gastric cancer, but there was no significant association with other clinicopathological variables.
Collapse
Affiliation(s)
- Elham Jafari
- Pathology and Stem Cells Research Center, Department of Pathology, Kerman University of Medical Science, Kerman, Iran
| | - Somaye Safinejad
- Pathology and Stem Cells Research Center, Department of Pathology, Kerman University of Medical Science, Kerman, Iran
| | - Shahriar Dabiri
- Pathology and Stem Cells Research Center, Department of Pathology, Kerman University of Medical Science, Kerman, Iran
| | - Ahmad Naghibzadeh-Tahami
- Health Services Management Research Center, Institute for Futures Studies in Health, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
8
|
Cristofani R, Piccolella M, Crippa V, Tedesco B, Montagnani Marelli M, Poletti A, Moretti RM. The Role of HSPB8, a Component of the Chaperone-Assisted Selective Autophagy Machinery, in Cancer. Cells 2021; 10:335. [PMID: 33562660 PMCID: PMC7915307 DOI: 10.3390/cells10020335] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 01/27/2021] [Accepted: 02/02/2021] [Indexed: 02/07/2023] Open
Abstract
The cellular response to cancer-induced stress is one of the major aspects regulating cancer development and progression. The Heat Shock Protein B8 (HSPB8) is a small chaperone involved in chaperone-assisted selective autophagy (CASA). CASA promotes the selective degradation of proteins to counteract cell stress such as tumor-induced stress. HSPB8 is also involved in (i) the cell division machinery regulating chromosome segregation and cell cycle arrest in the G0/G1 phase and (ii) inflammation regulating dendritic cell maturation and cytokine production. HSPB8 expression and role are tumor-specific, showing a dual and opposite role. Interestingly, HSPB8 may be involved in the acquisition of chemoresistance to drugs. Despite the fact the mechanisms of HSPB8-mediated CASA activation in tumors need further studies, HSPB8 could represent an important factor in cancer induction and progression and it may be a potential target for anticancer treatment in specific types of cancer. In this review, we will discuss the molecular mechanism underlying HSPB8 roles in normal and cancer conditions. The basic mechanisms involved in anti- and pro-tumoral activities of HSPB8 are deeply discussed together with the pathways that modulate HSPB8 expression, in order to outline molecules with a beneficial effect for cancer cell growth, migration, and death.
Collapse
|
9
|
Insulin receptor substrate 1 gene expression is strongly up-regulated by HSPB8 silencing in U87 glioma cells. Endocr Regul 2020; 54:231-243. [PMID: 33885248 DOI: 10.2478/enr-2020-0026] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Objective. The aim of the present investigation was to study the expression of genes encoding IRS1 (insulin receptor substrate 1) and some other functionally active proteins in U87 glioma cells under silencing of polyfunctional chaperone HSPB8 for evaluation of the possible significance of this protein in intergenic interactions.Methods. Silencing of HSPB8 mRNA was introduced by HSPB8 specific siRNA. The expression level of HSPB8, IRS1, HK2, GLO1, HOMER3, MYL9, NAMPT, PER2, PERP, GADD45A, and DEK genes was studied in U87 glioma cells by quantitative polymerase chain reaction.Results. It was shown that silencing of HSPB8 mRNA by specific to HSPB8 siRNA led to a strong down-regulation of this mRNA and significant modification of the expression of IRS1 and many other genes in glioma cells: strong up-regulated of HOMER3, GLO1, and PERP and down-regulated of MYL9, NAMPT, PER2, GADD45A, and DEK gene expressions. At the same time, no significant changes were detected in the expression of HK2 gene in glioma cells treated by siRNA, specific to HSPB8. Moreover, the silencing of HSPB8 mRNA enhanced the glioma cells proliferation rate.Conclusions. Results of this investigation demonstrated that silencing of HSPB8 mRNA affected the expression of IRS1 gene as well as many other genes encoding tumor growth related proteins. It is possible that the dysregulation of most of the studied genes in glioma cells after silencing of HSPB8 is reflected by a complex of intergenic interactions and that this polyfunctional chaperone is an important factor for the stability of genome function and regulatory mechanisms contributing to the tumorigenesis control.
Collapse
|
10
|
Heat shock protein B8 promotes proliferation and migration in lung adenocarcinoma A549 cells by maintaining mitochondrial function. Mol Cell Biochem 2020; 476:187-197. [PMID: 32926297 DOI: 10.1007/s11010-020-03896-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 09/02/2020] [Indexed: 12/24/2022]
Abstract
Heat shock protein B8 (HSPB8) impacts on tumor proliferation and migration of malignancy. However, the role of HSPB8 in lung adenocarcinoma (LUAC) remains unclear. The aim of this study, therefore, was to clarify whether HSPB8 could bring benefits to proliferation and migration of LUAC and its underlying mechanisms. The expression of HSPB8 was first evaluated by immunohistochemistry in 35 LUAC samples. Then, A549 lung adenocarcinoma cells were transfected with pcDNA-HSPB8 or si-HSPB8 to induce HSPB8 overexpression and silence. Cellular activity was evaluated with a Cell Counting Kit-8 (CCK-8) assay. Cell proliferation and migration were observed by EdU assay and scratch assay. Mitochondria-specific reactive oxygen species (mtROS) and membrane potential were measured using MitoSOX Red probe and JC-1 staining. Superoxide dismutase (SOD) activities and malondialdehyde (MDA) level were measured using commercial kits, respectively. HSPB8 protein, mitochondrial fusion protein MFN2 and mitochondrial fission protein p-Drp1/Drp1 were measured using western blot. Compared with the normal tissues, the expression of HSPB8 protein was higher in LUAC tissues and upregulation of HSPB8 protein was related to tumor size and tumor location. Furthermore, HSPB8 overexpression aggravated cell proliferation and migration of A549 cells. Mechanistically, HSPB8 suppressed mitochondrial impairment, leading to promoting the progress of A549 lung adenocarcinoma cells. These data demonstrate that HSPB8 plays an important role in progression of LUAC and may be a new target to treat LUAC.
Collapse
|
11
|
Small Heat Shock Proteins in Cancers: Functions and Therapeutic Potential for Cancer Therapy. Int J Mol Sci 2020; 21:ijms21186611. [PMID: 32927696 PMCID: PMC7555140 DOI: 10.3390/ijms21186611] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Revised: 09/06/2020] [Accepted: 09/08/2020] [Indexed: 02/07/2023] Open
Abstract
Small heat shock proteins (sHSPs) are ubiquitous ATP-independent chaperones that play essential roles in response to cellular stresses and protein homeostasis. Investigations of sHSPs reveal that sHSPs are ubiquitously expressed in numerous types of tumors, and their expression is closely associated with cancer progression. sHSPs have been suggested to control a diverse range of cancer functions, including tumorigenesis, cell growth, apoptosis, metastasis, and chemoresistance, as well as regulation of cancer stem cell properties. Recent advances in the field indicate that some sHSPs have been validated as a powerful target in cancer therapy. In this review, we present and highlight current understanding, recent progress, and future challenges of sHSPs in cancer development and therapy.
Collapse
|
12
|
Wu W, Lai L, Xie M, Qiu H. Insights of heat shock protein 22 in the cardiac protection against ischemic oxidative stress. Redox Biol 2020; 34:101555. [PMID: 32388268 PMCID: PMC7215242 DOI: 10.1016/j.redox.2020.101555] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 04/16/2020] [Accepted: 04/23/2020] [Indexed: 12/21/2022] Open
Abstract
the acute and chronic myocardial ischemia results in oxidative stress that impairs myocardial contractility and eventually leads to heart failure. However, the underlying regulatory molecular mechanisms are not fully understood. The heat shock protein 22 (Hsp22), a small-molecular-weight protein preferentially expressed in the heart, was found to be dramatically increased in the cardiac oxidative stress conditions in both human and animal models after the acute and chronic ischemia. Overexpression of Hsp22 largely protects the heart against ischemic damage. Mechanistically, overexpression of Hsp22 attenuates hypoxia-induced oxidative phosphorylation in mitochondrial and the high rate of superoxide production. Short term gene delivery of Hsp22 reduces the infarct size caused by the ischemia/reperfusion, providing a clinical therapeutic potential. This review discusses the new progress of the studies on Hsp22 by focusing on its protective effect against the excessive cardiac oxidative stress, including its adaptive induction in myocardium upon the oxidative stress, its protective role in myocardial ischemia/reperfusion, its regulation in mitochondrial oxidative phosphorylation and the underlying molecular signaling pathways promoting cell survival. This information will increase our understanding of the molecular regulation of cardiac adaption under the oxidative stress and the potential therapeutic relevance.
Collapse
Affiliation(s)
- Wenqian Wu
- Center of Molecular and Translational Medicine, Institution of Biomedical Science, Georgia State University, Atlanta, GA, 30303, USA; Department of Ultrasound, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Lo Lai
- Center of Molecular and Translational Medicine, Institution of Biomedical Science, Georgia State University, Atlanta, GA, 30303, USA
| | - Mingxing Xie
- Department of Ultrasound, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Hongyu Qiu
- Center of Molecular and Translational Medicine, Institution of Biomedical Science, Georgia State University, Atlanta, GA, 30303, USA.
| |
Collapse
|
13
|
Zhou Y, Cui J, Hu H, Wen Y, Du Z, Du H. Identification of a novel anti‑heat shock cognate 71 kDa protein antibody in patients with Kawasaki disease. Mol Med Rep 2020; 21:1771-1778. [PMID: 32319608 PMCID: PMC7057768 DOI: 10.3892/mmr.2020.10973] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 12/18/2020] [Indexed: 12/19/2022] Open
Abstract
Kawasaki disease (KD) is an idiopathic form of acute systemic vasculitis, which clinically mimics febrile diseases. Although it has been hypothesized that immune system malfunction is associated with KD, its etiology remains unclear. The aim of the present study was to identify a KD‑associated antibody. Immunoproteomic methods were used to identify KD‑associated antigens that could be recognized in the sera of patients with KD. HeLa cells were used as an antigen source and KD sera were used as probe antibodies to determine the binding of the antibodies using an indirect immunofluorescence assay. Western blotting was performed to identify KD‑associated antigens in HeLa whole cell lysates. Eight out of 12 serum samples obtained from patients with KD demonstrated immunoreactive bands at ~70 kDa, which was later determined to be heat shock cognate 71 kDa protein (HSP7C) by mass spectrometry. The diagnostic value of serum anti‑HSP7C antibodies for KD was assessed using ELISA. Using a cut‑off value of 0.267, anti‑HSP7C antibodies were observed to be present in the sera of 60.00% (30/50) of patients with KD, in 21.05% (8/38) of non‑KD febrile controls, and in 5.26% (2/38) of healthy controls. High serum levels of anti‑HSP7C antibodies were detected in the peripheral circulation of patients with KD. To the best of our knowledge, the present study is the first to observe the high expression levels of anti‑HSP7C antibodies in patients with KD. Therefore, anti‑HSP7C antibodies may be used as a diagnostic marker to detect KD.
Collapse
Affiliation(s)
- Yabin Zhou
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, P.R. China
| | - Jiawen Cui
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, P.R. China
| | - Huimin Hu
- Department of Pediatrics, Beijing Children's Hospital, Capital Medical University, Beijing 100045, P.R. China
| | - Yongqiang Wen
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, P.R. China
| | - Zhongdong Du
- Department of Pediatrics, Beijing Children's Hospital, Capital Medical University, Beijing 100045, P.R. China
| | - Hongwu Du
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, P.R. China
| |
Collapse
|
14
|
Chen Q, Xiang J, Gong R, Fang HY, Xu CC, Zhang HZ, Wu YQ. Atorvastatin downregulates HSP22 expression in an atherosclerotic model in vitro and in vivo. Int J Mol Med 2018; 43:821-829. [PMID: 30535427 PMCID: PMC6317682 DOI: 10.3892/ijmm.2018.4015] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Accepted: 06/07/2018] [Indexed: 12/20/2022] Open
Abstract
One of the pathological functions of heat shock protein 22 (HSP22) is the association with inflammatory diseases and atherosclerosis. However, the effects of a high-fat diet (HFD) or oxidized low-density lipoprotein (ox-LDL) combined with atorvastatin (ATV) on HSP22 expression are entirely unknown. The present study investigated the effects of ATV on HSP22 expression in HFD-induced atherosclerotic apolipoprotein E-deficient (ApoE−/−) mice and in ox-LDL-induced human umbilical vein endothelial cells (HUVECs). Furthermore, the influence of HSP22-knockdown on the HFD- or ox-LDL-induced atherosclerotic model was also examined. It was found that HFD or ox-LDL treatment significantly increased HSP22 expression in the serum and aorta, accompanied by decreased phosphorylated (p)-endothelial nitric oxide synthase (p-eNOS) activity and activated p38 mitogen-activated protein kinase (MAPK). However, these effects were suppressed by treatment with ATV. Furthermore, HSP22-knockdown showed reduced ox-LDL-induced lesions, evidenced by increased p-eNOS activity and inactivated p38 MAPK, while suppression of cell proliferation inhibition and cell cycle arrest were also observed. Taken together, the results of this study suggest that HFD or ox-LDL increased the expression of HSP22 and p-p38 MAPK, and decreased the p-eNOS activity in vitro and in vivo, and ATV could reduce the effects by downregulating HSP22 expression.
Collapse
Affiliation(s)
- Qi Chen
- Department of Cardiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Jian Xiang
- Department of Cardiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Ren Gong
- Department of Cardiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Hai-Yang Fang
- Department of Cardiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Cong-Cong Xu
- Department of Cardiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Hong-Zhou Zhang
- Department of Cardiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Yan-Qing Wu
- Department of Cardiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
15
|
The mTOR inhibitor AZD8055 overcomes tamoxifen resistance in breast cancer cells by down-regulating HSPB8. Acta Pharmacol Sin 2018; 39:1338-1346. [PMID: 29345254 DOI: 10.1038/aps.2017.181] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2017] [Accepted: 10/31/2017] [Indexed: 12/22/2022]
Abstract
Tamoxifen, an important endocrine therapeutic agent, is widely used for the treatment of estrogen receptor positive (ER+) breast cancer. However, de novo or acquired resistance prevents patients from benefitting from endocrine approaches and necessitates alternative treatments. In this study, we report that small heat protein beta-8 (HSPB8) may serve as an important molecule in tamoxifen resistance. HSPB8 expression is enhanced in MCF-7 cells resistant to tamoxifen (MCF-7/R) compared to parent cells. Moreover, high expression of HSPB8 associates with poor prognosis in ER+ breast cancer patients but not in patients without classification. Stimulating ER signaling by heterogeneous expression of ERa or 17β-estradiol promotes HSPB8 expression and reduces the cell population in G1 phase. In contrast, blockage of ER signaling by tamoxifen down-regulates the expression of HSPB8. In addition, knocking down HSPB8 by specific siRNAs induces significant cell cycle arrest at G1 phase. AZD8055 was found to be more potent against the proliferation of MCF-7/R cells than that of parent cells, which was associated with down-regulation of HSPB8. We found that the anti-proliferative activity of AZD8055 was positively correlated with the HSPB8 expression level in ER+ breast cancer cells. Thus, AZD8055 was able to overcome tamoxifen resistance in breast cancer cells, and the expression of HSPB8 may predict the efficacy of AZD8055 in ER+ breast cancer. This hypothesis deserves further investigation.
Collapse
|
16
|
Minchenko OH, Kharkova AP, Hnatiuk OS. ERN1 modifies the effect of glutamine deprivation on tumor growth related factors expression in U87 glioma cells. UKRAINIAN BIOCHEMICAL JOURNAL 2018. [DOI: 10.15407/ubj90.03.049] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
|
17
|
Exploring the multifaceted roles of heat shock protein B8 (HSPB8) in diseases. Eur J Cell Biol 2018; 97:216-229. [PMID: 29555102 DOI: 10.1016/j.ejcb.2018.03.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 03/09/2018] [Accepted: 03/12/2018] [Indexed: 02/06/2023] Open
Abstract
HSPB8 is a member of ubiquitous small heat shock protein (sHSP) family, whose expression is induced in response to a wide variety of unfavorable physiological and environmental conditions. Investigation of HSPB8 structure indicated that HSPB8 belongs to the group of so-called intrinsically disordered proteins and possesses a highly flexible structure. Unlike most other sHSPs, HSPB8 tends to form small-molecular-mass oligomers and exhibits substrate-dependent chaperone activity. In cooperation with BAG3, the chaperone activity of HSPB8 was reported to be involved in the delivery of misfolded proteins to the autophagy machinery. Through this way, HSPB8 interferes with pathological processes leading to neurodegenerative diseases. Accordingly, published studies have identified genetic links between mutations of HSPB8 and some kind of neuromuscular diseases, further supporting its important role in neurodegenerative disorders. In addition to their anti-aggregation properties, HSPB8 is indicated to interact with a wide range of client proteins, modulating their maturations and activities, and therefore, regulates a large repertoire of cellular functions, including apoptosis, proliferation, inflammation and etc. As a result, HSPB8 has key roles in cancer biology, autoimmune diseases, cardiac diseases and cerebral vascular diseases.
Collapse
|
18
|
Ge H, Yan Y, Guo L, Tian F, Wu D. Prognostic role of HSPs in human gastrointestinal cancer: a systematic review and meta-analysis. Onco Targets Ther 2018; 11:351-359. [PMID: 29391812 PMCID: PMC5774472 DOI: 10.2147/ott.s155816] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Background Heat shock proteins (HSPs) have been reported to be overexpressed in a wide range of human tumors. It has been shown that HSPs act as an oncogenic regulator and are involved in tumorigenesis. The clinical and prognostic significance of HSPs in gastrointestinal cancers (GICs) remains controversial. The aim of this study was to conduct a meta-analysis to assess the prognostic value of HSPs in GICs. Materials and methods A literature search was performed in PubMed, Cochrane Library, Web of Science, and Embase databases. Data on the relationship between expression of HSPs and survival outcomes were extracted. Pooled hazard ratios (HRs) with 95% CI were calculated. Results The expression of HSPs was not associated with the overall survival (OS) of GIC patients; however, it was significantly associated with worse OS for gastric cancer (GC) and colorectal cancer (CRC) patients. Conclusion Current evidence suggests that a high level of HSPs may not be a potential marker to predict the survival rate for every type of GICs. However, the expression of HSPs may predict a poor prognosis for GC and CRC patients.
Collapse
Affiliation(s)
- Hua Ge
- Department of Gastrointestinal Surgery
| | - Yan Yan
- Quality Control Department, The First People's Hospital of Zunyi, Zunyi Medical University, Huichuan, Zunyi, Guizhou, China
| | | | - Fei Tian
- Department of Gastrointestinal Surgery
| | - Di Wu
- Department of Gastrointestinal Surgery
| |
Collapse
|
19
|
Ge H, He X, Guo L, Yang X. Clinicopathological significance of HSP27 in gastric cancer: a meta-analysis. Onco Targets Ther 2017; 10:4543-4551. [PMID: 28979146 PMCID: PMC5602475 DOI: 10.2147/ott.s146590] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Background Many studies have provided increasing evidence to demonstrate that HSP27 has been involved in the development of gastric cancer; however, they all include few patients and the results remain controversial. Hence, we conducted a meta-analysis to evaluate correlations between HSP27 and the clinicopathological characteristics of gastric cancer. Methods An electronic search for relevant articles was conducted in PubMed, Cochrane Library, Web of Science, EMBASE database, Chinese CNKI, and Wan Fang. Data on the relationship between HSP27 expression and lymph node metastasis, serosal invasion, gender, tumor size, differentiation, and TNM stage were extracted. Pooled odds ratios and 95% confidence intervals were estimated by forest plot. Results The pooled analyses suggested that HSP27 expression was significantly associated with the incidence of gastric cancer. However, HSP27 expression had no significant relationship with lymph node metastasis, serosal invasion, gender, tumor size, differentiation, and TNM stage. Conclusion Our meta-analysis demonstrated that HSP27 may play vital roles in tumorigenesis and deterioration of gastric cancer. However, further high-quality studies are needed to provide more reliable evidence.
Collapse
Affiliation(s)
- Hua Ge
- Department of Gastrointestinal Surgery, The First People's Hospital of Zunyi, Zunyi Medical University, Zunyi, Guizhou, People's Republic of China
| | - Xueyan He
- Department of Gastrointestinal Surgery, The First People's Hospital of Zunyi, Zunyi Medical University, Zunyi, Guizhou, People's Republic of China
| | - Lingfei Guo
- Department of Gastrointestinal Surgery, The First People's Hospital of Zunyi, Zunyi Medical University, Zunyi, Guizhou, People's Republic of China
| | - Xianzhi Yang
- Department of Gastrointestinal Surgery, The First People's Hospital of Zunyi, Zunyi Medical University, Zunyi, Guizhou, People's Republic of China
| |
Collapse
|
20
|
Heat shock protein 22 (HSPB8) reduces the migration of hepatocellular carcinoma cells through the suppression of the phosphoinositide 3-kinase (PI3K)/AKT pathway. Biochim Biophys Acta Mol Basis Dis 2017; 1863:1629-1639. [DOI: 10.1016/j.bbadis.2017.04.021] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Revised: 04/07/2017] [Accepted: 04/25/2017] [Indexed: 12/13/2022]
|
21
|
Shan YQ, Ying RC, Zhou CH, Zhu AK, Ye J, Zhu W, Ju TF, Jin HC. MMP-9 is increased in the pathogenesis of gastric cancer by the mediation of HER2. Cancer Gene Ther 2015; 22:101-7. [PMID: 25633484 DOI: 10.1038/cgt.2014.61] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2014] [Revised: 08/28/2014] [Accepted: 08/30/2014] [Indexed: 12/12/2022]
Abstract
Human epidermal growth factor receptor 2 (HER2) overexpression is not only closely associated with the tumor growth, but is also related to tumor invasion. We here aimed to investigate the mechanism of HER2 mediation in the pathogenesis of gastric cancer. The human gastric cancer cell lines SGC-7901, MKN-45, AGS, the immortalized cell line GES-1 derived from normal gastric mucosa. Cell transfection and selection of stable cell lines and the gene and protein levels of HER2 and Matrix metalloproteinase-9 (MMP-9) were examined to determine the molecular relationship between them in the pathogenesis of gastric cancer. The human gastric cancer cell lines SGC-7901, MKN-45, AGS, the immortalized cell line GES-1 derived from normal gastric mucosa. Cell transfection and selection of stable cell lines and the gene and protein levels of HER2 and MMP-9 were examined to determine the molecular relationship between them in the pathogenesis of gastric cancer. We demonstrated that vector-based shRNA significantly knocked down the expression of HER2 and considerably inhibited both the migration and invasion of gastric cancer cells. HER2 knockdown resulted in the downregulation of the expression of MMP-9, whereas HER2 overexpression improved the transcription of MMP-9 through the activation of an MMP-9 promoter. The promoter region of MMP-9 between -2500 and -2000 bp was found to be crucial for the upregulation of HER2-mediated transcription. Furthermore, a truncated promoter (-70 to +63) did not display any transcriptional activity. Cell invasion activity was almost completely inhibited when MMP-9 was knocked down. Conversely, the overexpression of MMP-9 partly rescued the invasion ability of cell strains with knockdown HER2. These findings help further understanding of the molecular mechanisms through which HER2 promotes malignancy, and suggest that targeting both HER2 and MMP-9 may be required to effectively block HER2 signaling in gastric cancer therapy.
Collapse
Affiliation(s)
- Y-Q Shan
- 1] Department of General Surgery, Hangzhou First People's Hospital, Hangzhou, China [2] Affiliated Hangzhou Hospital, Nanjing Medical University, Hangzhou, China
| | - R-C Ying
- 1] Department of General Surgery, Hangzhou First People's Hospital, Hangzhou, China [2] Affiliated Hangzhou Hospital, Nanjing Medical University, Hangzhou, China
| | - C-H Zhou
- 1] Department of General Surgery, Hangzhou First People's Hospital, Hangzhou, China [2] Affiliated Hangzhou Hospital, Nanjing Medical University, Hangzhou, China
| | - A-K Zhu
- 1] Department of General Surgery, Hangzhou First People's Hospital, Hangzhou, China [2] Affiliated Hangzhou Hospital, Nanjing Medical University, Hangzhou, China
| | - J Ye
- 1] Department of General Surgery, Hangzhou First People's Hospital, Hangzhou, China [2] Affiliated Hangzhou Hospital, Nanjing Medical University, Hangzhou, China
| | - W Zhu
- 1] Department of General Surgery, Hangzhou First People's Hospital, Hangzhou, China [2] Affiliated Hangzhou Hospital, Nanjing Medical University, Hangzhou, China
| | - T-F Ju
- 1] Department of General Surgery, Hangzhou First People's Hospital, Hangzhou, China [2] Affiliated Hangzhou Hospital, Nanjing Medical University, Hangzhou, China
| | - H-C Jin
- 1] Department of General Surgery, Hangzhou First People's Hospital, Hangzhou, China [2] Affiliated Hangzhou Hospital, Nanjing Medical University, Hangzhou, China
| |
Collapse
|
22
|
Fu YF, Gui R, Liu J. HER-2-induced PI3K signaling pathway was involved in the pathogenesis of gastric cancer. Cancer Gene Ther 2015; 22:145-53. [PMID: 25613482 DOI: 10.1038/cgt.2014.80] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Revised: 12/09/2014] [Accepted: 12/09/2014] [Indexed: 01/02/2023]
Abstract
Human epidermal growth factor receptor-2 (HER-2) overexpression was closely associated with the tumor growth and invasion, we here aimed to investigate the mechanism of HER-2 mediation in the pathogenesis of gastric cancer (GC). We first detected the expression of HER-2 in GC cell line SGC-7901 and then examined the levels of nuclear factor-κB (NF-κB), matrix metalloproteinase-9 (MMP-9) and intercellular adhesion molecule-1 (ICAM-1) and the association between them by molecular methods. Statistical analysis was used to compare the significance. We further detected the possible molecular mechanism involved in their relationship in the SGC-7901 genesis. The MMP-9, NF-κB and secretory type (s-ICAM-1) levels were significantly greater in peripheral blood serum from SGC-7901 than healthy control GES-1 (P<0.01). ICAM-1, MMP-9 and NF-κB mRNA and protein levels were more highly expressed in SGC-7901 than healthy control GES-1. The expression levels of NF-κB, MMP-9 and ICAM-1 were positively related in GC cell line SGC-7901, which was HER-2 positive. The HER-2 positive SGC-7901 secreted more transforming growth factor beta 1 (TGF-β1) and resultantly activated MMP-9 to enhance s-ICAM-1 secretion and further studies showed that phosphatidylinositol-3 kinase (PI3K)/Akt/NF-κB signaling pathway was involved in GC pathogenesis. The GC cells that express the HER-2 oncogene spur the activation of NF-κB that can upregulate the expression of ICAM-1 and induce the expression of MMP-9, which hydrolyzes ICAM-1 into s-ICAM-1 to promote tumor immune escape. TGF-β1-induced PI3K/Akt/NF-κB signaling pathway was involved in the pathogenesis of GC and they could be a new target for cancer therapy. The GC cells that express the HER-2 oncogene spur the activation of NF-κB that can upregulate the expression of ICAM-1 and induce the expression of MMP-9, which hydrolyzes ICAM-1 into s-ICAM-1 to promote tumor immune escape. TGF-β1-induced PI3K/Akt/NF-κB signaling pathway was involved in the pathogenesis of GC and they could be a new target for cancer therapy.
Collapse
Affiliation(s)
- Y F Fu
- The Third Xiang-Ya Hospital, Central South University, Changsha, China
| | - R Gui
- The Third Xiang-Ya Hospital, Central South University, Changsha, China
| | - J Liu
- The Third Xiang-Ya Hospital, Central South University, Changsha, China
| |
Collapse
|
23
|
MicroRNA let-7b suppresses human gastric cancer malignancy by targeting ING1. Cancer Gene Ther 2015; 22:122-9. [PMID: 25613480 DOI: 10.1038/cgt.2014.75] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Revised: 11/23/2014] [Accepted: 11/24/2014] [Indexed: 12/21/2022]
Abstract
MicroRNAs (miRNAs) are important regulators that play key roles in tumorigenesis and tumor progression. In this study, we investigate whether let-7b acts as a tumor suppressor to inhibit invasion and metastasis in gastric cancers. We analyzed the expression of let-7b in 60 pair-matched gastric neoplastic and adjacent non-neoplastic tissues by quantitative real-time polymerase chain reaction. Functional analysis of let-7b expression was assessed in vitro in gastric cancer cell lines with let-7b precursor and inhibitor. The roles of let-7b in tumorigenesis and tumor metastasis were analyzed using a stable let-7b expression plasmid in nude mice. A luciferase reporter assay was used to assess the effect of let-7b on inhibitor of growth family, member 1 (ING1) expression. Real-time PCR showed decreased levels of let-7b expression in metastatic gastric cancer tissues and cell lines that are potentially highly metastatic. Cell invasion and migration were significantly impaired in GC9811-P and SGC7901-M cell lines after transfection with let-7b mimics. Nude mice with xenograft models of gastric cancer confirmed that let-7b could inhibit gastric cancer metastasis in vivo after transfection by the lentivirus pGCsil-GFP- let-7b. Luciferase reporter assays demonstrated that let-7b directly binds to the 3'-UTR of ING1, and real-time PCR and western blotting further indicated that let-7b downregulated the expression of ING1 at the mRNA and protein levels. Our study demonstrates that overexpression of let-7b in gastric cancer can inhibit invasion and migration of gastric cancer cells through directly targeting the tumor metastasis-associated gene ING1. These findings help clarify the molecular mechanisms involved in gastric cancer metastasis and indicate that let-7b modulation may be a bona fide treatment of gastric cancer.
Collapse
|