1
|
Werawatganone P, Werawatganon D, Noonak N, Chayanupatkul M, Chatsuwan T, Klaikeaw N, Muangsiri W, Siriviriyakul P. Unveiling the Potency of Gardenia Extract Against H. pylori: Insights from In Vitro and In Vivo Studies. Biomedicines 2025; 13:92. [PMID: 39857676 PMCID: PMC11760463 DOI: 10.3390/biomedicines13010092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Revised: 12/24/2024] [Accepted: 12/31/2024] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND AND AIM Gardenia jasminoides (G. jasminoides) could treat various inflammatory diseases. This study aimed to investigate the effects of G. jasminoides fruit extract on gastric inflammation and protective mechanisms in Helicobacter pylori (H. pylori)-induced gastritis. Experimental procedure: G. jasminoides fruit extract was prepared and analyzed for geniposide content. The inhibitory effect of the extract on H. pylori growth was investigated using the disk diffusion method. The in vitro anti-inflammatory property of the extract was evaluated using the erythrocyte membrane stabilization method. Thirty-five male Sprague-Dawley rats were inoculated with H. pylori (108-1010 colony-forming unit/mL) and divided into five groups. Each group was treated with various doses of the extract (98-395 mg/kg). The serum and stomach tissue of the rats were evaluated using enzyme-linked immunosorbent assay, histopathology, and immunohistochemistry. RESULTS AND CONCLUSIONS The geniposide content in the dried extract was 8.12% ± 0.79% by dry weight. The inhibition zone was observed at the extract ≥ 1.97 mg/disk, and the extract presented anti-inflammatory potential. The H. pylori-inoculated rats had a significant increase in serum interleukin (IL)-17, IL-33, and gastric epidermal growth factor (EGF) levels and a significant decrease in serum prostaglandin E2 level (p < 0.05) in conjunction with the development of gastric inflammation on histopathology. The treatment of the extract could significantly decrease the serum IL-17, IL-33, and gastric EGF levels, significantly increase the serum PGE2 level (p < 0.05), and improve gastric histopathology. Thus, G. jasminoides fruit extract attenuated H. pylori-induced gastritis by inhibiting bacterial growth, reducing inflammation, and enhancing protective mechanisms.
Collapse
Affiliation(s)
- Pornpen Werawatganone
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand; (P.W.); (W.M.)
| | - Duangporn Werawatganon
- Center of Excellence in Alternative and Complementary Medicine for Gastrointestinal and Liver Diseases, Department of Physiology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand (M.C.)
| | - Nattida Noonak
- Center of Excellence in Alternative and Complementary Medicine for Gastrointestinal and Liver Diseases, Department of Physiology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand (M.C.)
| | - Maneerat Chayanupatkul
- Center of Excellence in Alternative and Complementary Medicine for Gastrointestinal and Liver Diseases, Department of Physiology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand (M.C.)
| | - Tanittha Chatsuwan
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| | - Naruemon Klaikeaw
- Department of Pathology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand;
| | - Walaisiri Muangsiri
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand; (P.W.); (W.M.)
| | - Prasong Siriviriyakul
- Center of Excellence in Alternative and Complementary Medicine for Gastrointestinal and Liver Diseases, Department of Physiology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand (M.C.)
| |
Collapse
|
2
|
Chayanupatkul M, Somanawat K, Chuaypen N, Klaikeaw N, Wanpiyarat N, Siriviriyakul P, Tumwasorn S, Werawatganon D. Probiotics and their beneficial effects on alcohol-induced liver injury in a rat model: the role of fecal microbiota. BMC Complement Med Ther 2022; 22:168. [PMID: 35733194 PMCID: PMC9215017 DOI: 10.1186/s12906-022-03643-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 06/09/2022] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Current therapies for alcohol-induced liver injury are of limited efficacy and associated with significant side effects. With the proposed pathophysiology of alcohol-induced liver injury to be related to deranged gut microbiota, we hypothesized that probiotics would have beneficial effects in attenuating alcohol-induced liver injury.
Methods
Twenty-four male Sprague-Dawley rats were divided into 4 groups: control group, alcohol group, Lactobacillus plantarum group, and mixed-strain probiotics group. After 4 weeks, all rats were sacrificed, and blood samples were analyzed for ALT, lipopolysaccharide level (LPS), interleukin 6 (IL-6), and tumor necrosis factor-alpha (TNF-α). Liver tissues were processed for histopathology, malondialdehyde (MDA) level and immunohistochemistry for toll-like receptors 4 (TLR-4). Stool samples were collected, and 16S rRNA sequencing was used to analyze the fecal microbiota.
Results
Liver histopathology showed the presence of significant hepatocyte ballooning in the alcohol group as compared with the control group, and the treatment with L. plantarum or mixed-strain probiotics alleviated these changes. Significant elevation of serum ALT, LPS, IL-6, and TNF-α, hepatic MDA levels, and hepatic TLR-4 expression were observed in alcohol-fed rats as compared with control rats. The administration of L. plantarum or mixed-strain probiotics restored these changes to the levels of control rats. The relative abundance of fecal bacteria at genus level showed a significant reduction in Allobaculum, Romboutsia, Bifidobacterium, and Akkermansia in the alcohol group as compared with the control group. In probiotics-treated rats, significant increases in Allobaculum and Bifidobacterium were observed, while the relative abundance of Romboutsia and Akkermansia was unchanged compared to the alcohol group. A reduction in alpha diversity was observed in alcohol-treated rats, whereas the improvement was noted after probiotic treatment.
Conclusions
The treatment with Lactobacillus, whether as single-, or mixed-strain probiotics, was beneficial in reducing the severity of alcohol-induced liver injury likely through the increase in beneficial bacteria, and the reduction of inflammatory responses, and oxidative stress.
Collapse
|
3
|
Ning Z, Zhu X, Jiang Y, Gao A, Zou S, Gu C, He C, Chen Y, Ding WQ, Zhou J. Integrin-Linked Kinase Is Involved In the Proliferation and Invasion of Esophageal Squamous Cell Carcinoma. J Cancer 2020; 11:324-333. [PMID: 31897228 PMCID: PMC6930430 DOI: 10.7150/jca.33737] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 09/18/2019] [Indexed: 11/09/2022] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) is an aggressive type of cancer with high mortality rate in China, largely due to its high invasive and metastatic potential. The purposes of this study are to investigate the potential molecular mechanisms behind the aggressive nature of ESCC and search for new prognostic biomarkers. By employing the quantitative proteomic based strategy, we compared the proteomic profile between three ESCC samples and paired adjacent tissues. After bioinformatics analysis, four candidate proteins were validated in thirteen paired patient samples. Further validation of the key candidate, integrin-linked kinase (ILK), was carried out in one hundred patient samples. The specific inhibitor compound 22 (cpd22) was used to assess the influence of ILK to ESCC cell motility and invasiveness by applying wound-healing and transwell assay. Western blot analysis was performed to elucidate the signaling pathways involved in ILK-mediated ESCC invasion. Total 236 proteins were identified by proteomic analysis. Bioinformatics analysis suggested a key role of the collagen/integrin/ILK signaling pathway during ESCC progression. Further validation indicated that ILK is overexpressed in ESCC tissues and is correlated with poor patient prognosis. Inhibition of ILK kinase activity suppresses proliferation and blocks invasion and migration of ESCC cells. Signaling pathway analysis revealed that ILK regulates AKT phosphorylation on Ser473 but not GSK-3β on Ser9 to promote proliferation and motility of ESCC cells. In conclusion, our results indicated that ILK may play a crucial role in ESCC invasion and metastasis and may serve as a prognostic biomarker and therapeutic target for ESCC.
Collapse
Affiliation(s)
- Zhonghua Ning
- Department of Radiation Oncology, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, P.R. China
| | - Xiaozhong Zhu
- Department of Thoracic Surgery, the Affiliated Hospital of the Jiangsu University, Zhenjiang, Jiangsu, P.R. China
| | - Youqin Jiang
- Department of Radiation Oncology, The Third People's Hospital of Yancheng, Yancheng, Jiangsu, P.R. China
| | - Aidi Gao
- Suzhou Cancer Center Core Laboratory, Nanjing Medical University Affiliated Suzhou Hospital, Suzhou, Jiangsu, P.R. China
| | - Shitao Zou
- Suzhou Cancer Center Core Laboratory, Nanjing Medical University Affiliated Suzhou Hospital, Suzhou, Jiangsu, P.R. China
| | - Chao Gu
- Department of Gastrointestinal surgery, Nanjing Medical University Affiliated Suzhou Hospital, Suzhou, Jiangsu, P.R. China
| | - Chao He
- Suzhou Cancer Center Core Laboratory, Nanjing Medical University Affiliated Suzhou Hospital, Suzhou, Jiangsu, P.R. China
| | - Yihong Chen
- Department of Radio-Oncology, The First Affiliated Hospital of Wannan Medical College, Wuhu, Anhui, P.R. China
| | - Wei-Qun Ding
- Department of Pathology, University of Oklahoma Health Science Center, Oklahoma City, OK, USA
| | - Jundong Zhou
- Suzhou Cancer Center Core Laboratory, Nanjing Medical University Affiliated Suzhou Hospital, Suzhou, Jiangsu, P.R. China
| |
Collapse
|
4
|
Brown C, Sekhavati F, Cardenes R, Windmueller C, Dacosta K, Rodriguez-Canales J, Steele KE. CTLA-4 Immunohistochemistry and Quantitative Image Analysis for Profiling of Human Cancers. J Histochem Cytochem 2019; 67:901-918. [PMID: 31609157 DOI: 10.1369/0022155419882292] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
There is an important need in immuno-oncology to develop reliable immunohistochemistry (IHC) to assess the expression of CTLA-4+ tumor-infiltrating lymphocytes in human cancers and quantify them with image analysis (IA). We used commercial polyclonal and monoclonal antibodies and characterized three chromogenic cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) assays with suitable specificity and sensitivity for use in formalin-fixed, paraffin-embedded (FFPE) tissues. We found variable numbers of CTLA-4+ lymphocytes in multiple types of cancer and secondary lymphoid organs (SLOs) and other normal human tissues. Combining CTLA-4 with CD3, CD4, or CD8 by immunofluorescence showed that CTLA-4+ lymphocytes in SLOs and tumors were typically CD3+ and CD4+, but not CD8+. Individual lymphocytes expressed CTLA-4 either as primarily granular cytoplasmic staining or as excentric globular deposits. The CTLA-4/FoxP3 (forkhead box P3 protein) duplex IHC demonstrated that CTLA-4+/FoxP3- lymphocytes predominated in the germinal centers of SLOs and tumor tertiary lymphoid structures (TLSs), whereas CTLA-4+/FoxP3+ lymphocytes populated the T-cell zone of SLOs and TLSs, plus tumor stroma. IA scoring was highly comparable with pathologist scoring for CTLA-4 and CTLA-4/FoxP3 assays and a FoxP3 single IHC. Our findings show that CTLA-4 IHC can be used to reliably label lymphocytes in FFPE human tissues, making it possible to investigate the role of CTLA-4 in the tumor microenvironment.
Collapse
Affiliation(s)
- Charles Brown
- Department of Pathology, AstraZeneca, Gaithersburg, Maryland
| | | | | | | | - Karma Dacosta
- Department of Pathology, AstraZeneca, Gaithersburg, Maryland
| | | | - Keith E Steele
- Department of Pathology, AstraZeneca, Gaithersburg, Maryland
| |
Collapse
|
5
|
Hanson PJ, Hossain AR, Qiu Y, Zhang HM, Zhao G, Li C, Lin V, Sulaimon S, Vlok M, Fung G, Chen VH, Jan E, McManus BM, Granville DJ, Yang D. Cleavage and Sub-Cellular Redistribution of Nuclear Pore Protein 98 by Coxsackievirus B3 Protease 2A Impairs Cardioprotection. Front Cell Infect Microbiol 2019; 9:265. [PMID: 31396490 PMCID: PMC6667557 DOI: 10.3389/fcimb.2019.00265] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 07/08/2019] [Indexed: 01/15/2023] Open
Abstract
Myocarditis, inflammation of the heart muscle, affects all demographics and is a major cause of sudden and unexpected death in young people. It is most commonly caused by viral infections of the heart, with coxsackievirus B3 (CVB3) being among the most prevalent pathogens. To understand the molecular pathogenesis of CVB3 infection and provide strategies for developing treatments, we examined the role of a key nuclear pore protein 98 (NUP98) in the setting of viral myocarditis. NUP98 was cleaved as early as 2 h post-CVB3 infection. This cleavage was further verified through both the ectopic expression of viral proteases and in vitro using purified recombinant CVB3 proteases (2A and 3C), which demonstrated that CVB3 2A but not 3C is responsible for this cleavage. By immunostaining and confocal imaging, we observed that cleavage resulted in the redistribution of NUP98 to punctate structures in the cytoplasm. Targeted siRNA knockdown of NUP98 during infection further increased viral protein expression and viral titer, and reduced cell viability, suggesting a potential antiviral role of NUP98. Moreover, we discovered that expression levels of neuregulin-1 (NRG1), a cardioprotective gene, and presenilin-1 (PSEN1), a cellular protease processing the tyrosine kinase receptor ERBB4 of NRG1, were reliant upon NUP98 and were downregulated during CVB3 infection. In addition, expression of these NUP98 target genes in myocardium tissue not only occurred at an earlier phase of infection, but also appeared in areas away from the initial inflammatory regions. Collectively, CVB3-induced cleavage of NUP98 and subsequent impairment of the cardioprotective NRG1-ERBB4/PSEN1 signaling cascade may contribute to increased myocardial damage in the context of CVB3-induced myocarditis. To our knowledge, this is the first study to demonstrate the link between NUP98 and the NRG1 signaling pathway in viral myocarditis.
Collapse
Affiliation(s)
- Paul J Hanson
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada.,UBC Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, BC, Canada
| | - Al Rohet Hossain
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada.,UBC Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, BC, Canada
| | - Ye Qiu
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada.,UBC Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, BC, Canada
| | - Huifang M Zhang
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada.,UBC Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, BC, Canada
| | - Guangze Zhao
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada.,UBC Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, BC, Canada
| | - Cheng Li
- UBC Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, BC, Canada
| | - Veena Lin
- UBC Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, BC, Canada
| | - Saheedat Sulaimon
- UBC Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, BC, Canada.,Jefferson College of Population Health, Thomas Jefferson University, Philadelphia, PA, United States
| | - Marli Vlok
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
| | - Gabriel Fung
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada.,UBC Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, BC, Canada
| | - Victoria H Chen
- UBC Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, BC, Canada.,Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Eric Jan
- Jefferson College of Population Health, Thomas Jefferson University, Philadelphia, PA, United States
| | - Bruce M McManus
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada.,UBC Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, BC, Canada.,Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - David J Granville
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada.,UBC Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, BC, Canada
| | - Decheng Yang
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada.,UBC Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, BC, Canada
| |
Collapse
|
6
|
Abraham V, Cao G, Parambath A, Lawal F, Handumrongkul C, Debs R, DeLisser HM. Involvement of TIMP-1 in PECAM-1-mediated tumor dissemination. Int J Oncol 2018; 53:488-502. [PMID: 29845213 PMCID: PMC6017270 DOI: 10.3892/ijo.2018.4422] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 05/17/2018] [Indexed: 02/07/2023] Open
Abstract
Platelet endothelial cell adhesion molecule-1 (PECAM-1) is expressed on the vascular endothelium and has been implicated in the late progression of metastatic tumors. The activity of PECAM-1 appears to be mediated by modulation of the tumor microenvironment (TME) and promotion of tumor cell proliferation, rather than through the stimulation of tumor angiogenesis. The present study aimed to extend those initial findings by indicating that the presence of functional PECAM-1 on the endothelium promotes a proliferative tumor cell phenotype in vivo, as well as in tumor cell (B16-F10 melanoma and 4T1 breast cancer cell lines) co-culture assays with mouse endothelial cells (ECs) or a surrogate EC line (REN-MP). The pro-proliferative effects were mediated by soluble endothelial-derived factors that were dependent on PECAM-1 homophilic ligand interactions, but which were independent of PECAM-1-dependent signaling. Further analysis of the conditioned media obtained from tumor/EC and tumor/REN-MP co-cultures identified TIMP metallopeptidase inhibitor-1 (TIMP-1) as a PECAM-1-regulated factor, the targeting of which in the tumor cell/REN-MP system inhibited tumor cell proliferation. In addition, TIMP-1 expression was decreased in metastatic tumors from the lungs of PECAM-1-null mice, thus providing evidence of the in vivo significance of co-culture studies. Taken together, these studies indicated that endothelial PECAM-1, through PECAM-1-dependent homophilic binding interactions, may induce release of TIMP-1 from the endothelium into the TME, thus leading to increased tumor cell proliferation.
Collapse
Affiliation(s)
- Valsamma Abraham
- Pulmonary, Allergy and Critical Care Division, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Gaoyuan Cao
- Rutgers Institute for Translational Medicine and Science, Child Health Institute of New Jersey, Rutgers University, New Brunswick, NJ 08901, USA
| | - Andrew Parambath
- Pulmonary, Allergy and Critical Care Division, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Fareedah Lawal
- Pulmonary, Allergy and Critical Care Division, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | - Robert Debs
- California Pacific Medical Center Research Institute, San Francisco, CA 94107, USA
| | - Horace M DeLisser
- Pulmonary, Allergy and Critical Care Division, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
7
|
Kalra J, Baker J. Using Digital Quantification of Stained Tissue Microarrays as a Medium-Throughput, Quantitative Method for Measuring the Kinetics of Signal Transduction. Methods Mol Biol 2017; 1554:107-125. [PMID: 28185185 DOI: 10.1007/978-1-4939-6759-9_5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Determining how a molecular targeting agent affects signaling pathways is challenging. To facilitate this exploration, we use a combination of medium-throughput methodologies such as tissue microarrays (TMA) and digital image analysis for quantification of immunohistochemistry (IHC). Because TMAs enable the simultaneous analysis of up to 1000 tissues, this tool can be used to comprehensively assess changes in signal transduction pathways over time. TMAs can be digitized and analyzed using the Aperio ScanScope imaging system and Aperio ImageScope software to deliver semiquantitative data. This chapter describes the methodologies for tissue collection and for the construction, sectioning, staining, and digital analysis of TMAs.
Collapse
Affiliation(s)
- Jessica Kalra
- Department of Experimental Therapeutics, British Columbia Cancer Agency, 675 West 10th Avenue, 5th Floor, Vancouver, BC, V5Z 1L3, Canada.
- Langara College, Vancouver, BC, Canada.
- Department of Anesthesia Pharmacology, University of British Columbia, Vancouver, BC, Canada.
| | - Jennifer Baker
- Integrative Oncology, BC Cancer Agency, Vancouver, BC, Canada
| |
Collapse
|
8
|
Zhuang X, Lv M, Zhong Z, Zhang L, Jiang R, Chen J. Interplay between intergrin-linked kinase and ribonuclease inhibitor affects growth and metastasis of bladder cancer through signaling ILK pathways. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2016; 35:130. [PMID: 27576342 PMCID: PMC5006283 DOI: 10.1186/s13046-016-0408-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Accepted: 08/17/2016] [Indexed: 12/29/2022]
Abstract
Background Integrin-linked kinase (ILK) is a multifunctional adaptor protein which is involved with protein signalling within cells to modulate malignant (cancer) cell movement, cell cycle, metastasis and epithelial–mesenchymal transition (EMT). Our previous experiment demonstrated that ILK siRNA inhibited the growth and induced apoptosis of bladder cancer cells as well as increased the expression of Ribonuclease inhibitor (RI), an important cytoplasmic protein with many functions. We also reported that RI overexpression inhibited ILK and phosphorylation of AKT and GSK3β. ILK and RI gene both locate on chromosome 11p15 and the two genes are always at the adjacent position of same chromosome during evolution, which suggest that ILK and RI could have some relationship. However, underlying interacting mechanisms remain unclear between them. Here, we postulate that RI might regulate ILK signaling pathway via interacting with ILK. Methods Co-immunoprecipitation, GST pull-down and co-localization under laser confocal microscope assay were used to determine the interaction between ILK and RI exogenously and endogenously. Furthermore, we further verified that there is a direct binding between the two proteins by fluorescence resonance energy transfer (FRET) in cells. Next, The effects of interplay between ILK and RI on the key target protein expressions of PI3K/AKT/mTOR signaling pathway were determined by western blot, immunohistochemistry and immunofluorescence assay in vivo and in vitro. Finally, the interaction was assessed using nude mice xenograft model. Results We first found that ILK could combine with RI both in vivo and in vitro by GST pull-down, co-immunoprecipitation (Co-IP) and FRET. The protein levels of ILK and RI revealed a significant inverse correlation in vivo and in vitro. Subsequently, The results showed that up-regulating ILK could increase cell proliferation, change cell morphology and regulate cell cycle. We also demonstrated that the overexpression of ILK remarkably promoted EMT and expressions of target molecules of ILK signaling pathways in vitro and in vivo. Finally, we found that ILK overexpression significantly enhanced growth, metastasis and angiogenesis of xenograft tumor; Whereas, RI has a contrary role compared to ILK in vivo and in vitro. Conclusions Our findings, for the first time, directly proved that the interplay between ILK and RI regulated EMT via ILK/PI3K/AKT signaling pathways for bladder cancer, which highlights the possibilities that ILK/RI could be valuable markers together for the therapy and diagnosis of human carcinoma of urinary bladder.
Collapse
Affiliation(s)
- Xiang Zhuang
- Department of Cell Biology and Genetics, Chongqing Medical University, Chongqing, 400016, China
| | - Mengxin Lv
- Department of Cell Biology and Genetics, Chongqing Medical University, Chongqing, 400016, China
| | - Zhenyu Zhong
- The First Clinical College, Chongqing Medical University, Chongqing, 400016, China
| | - Luyu Zhang
- Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, 400016, China
| | - Rong Jiang
- Laboratory of Stem Cells and Tissue Engineering, Chongqing Medical University, Chongqing, China
| | - Junxia Chen
- Department of Cell Biology and Genetics, Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|