1
|
Le TNQ, Le MK, Dang MX, Kondo T. CXCL5 expression is associated with active signals of macrophages in the microenvironment of papillary thyroid carcinoma. Pathol Res Pract 2024; 261:155474. [PMID: 39067172 DOI: 10.1016/j.prp.2024.155474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 07/15/2024] [Indexed: 07/30/2024]
Abstract
BACKGROUND C-X-C motif chemokine ligand 5 (CXCL5) is a chemokine molecule that is secreted by immune cells in attracting granulocytes. Studies showed that CXCL5 was related to the progression of papillary thyroid carcinoma (PTC) tumor cells. However, the in vivo effects of CXCL5 on PTC tumor cells and their microenvironment have not been elucidated. The present study aimed to investigate the biological effects of CXCL5 on tumor cells, microenvironment, and clinical progression of PTC. MATERIALS AND METHODS The PTC patients from The Human Cancer Genome Atlas (TCGA) - thyroid carcinoma (THCA) were retrieved. There were a total of 500 patients who met the criteria of our study. Differential expression (DEA) and pathway analyses were used to explore the biological effects of CXCL5 gene expression. RESULTS In DEA, we found that CXCL5 was mostly associated with PBPP, SLC11A1, and MRC1 (adjusted p<0.001). Samples with CXCL5 FPKM≥1 were related to a different immune profile (p<0.001). In pathway analyses, samples with higher CXCL5 expression possessed higher activities of RAS-RAF, NF-kB, PRC2, IL2, IL5, and Wnt pathways (adjusted p<0.001). In microenvironment analysis, CXCL5 was highly correlated with the activity of macrophage (Rho=0.76; adjusted p<0.001). Clinically, high level of CXCL5 expression was an indicator of tumor stages (p<0.001), nodal metastasis (AUC=0.68), and prognosis (p=0.001). CONCLUSION CXCL5 was a significant biomarker of PTC. CXCL5 was highly associated with tumor immunology and microenvironment. Samples with higher CXCL5 expression had more advanced disease status and worse prognosis. CXCL5 target therapy is potentially helpful in advanced PTC.
Collapse
Affiliation(s)
- Tho Ngoc-Quynh Le
- Department of Pathology, University Medical Center, Ho Chi Minh City 70000, Viet Nam.
| | - Minh-Khang Le
- Department of Pathology, University of Yamanashi, Chuo 409-3821, Japan.
| | - Minh-Xuan Dang
- Department of Pathology, University Medical Center, Ho Chi Minh City 70000, Viet Nam.
| | - Tetsuo Kondo
- Department of Pathology, University of Yamanashi, Chuo 409-3821, Japan.
| |
Collapse
|
2
|
Zheng J, Lu W, Wang C, Chen S, Zhang Q, Su C. Unfolding the mysteries of heterogeneity from a high-resolution perspective: integration analysis of single-cell multi-omics and spatial omics revealed functionally heterogeneous cancer cells in ccRCC. Aging (Albany NY) 2024; 16:10943-10971. [PMID: 38944814 PMCID: PMC11272124 DOI: 10.18632/aging.205974] [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: 09/19/2023] [Accepted: 05/16/2024] [Indexed: 07/01/2024]
Abstract
The genomic landscape of clear cell renal cell carcinoma (ccRCC) has a considerable intra-tumor heterogeneity, which is a significant obstacle in the field of precision oncology and plays a pivotal role in metastasis, recurrence, and therapeutic resistance of cancer. The mechanisms of intra-tumor heterogeneity in ccRCC have yet to be fully established. We integrated single-cell RNA sequencing (scRNA-seq) and transposase-accessible chromatin sequencing (scATAC-seq) data from a single-cell multi-omics perspective. Based on consensus non-negative matrix factorization (cNMF) algorithm, functionally heterogeneous cancer cells were classified into metabolism, inflammatory, and EMT meta programs, with spatial transcriptomics sequencing (stRNA-seq) providing spatial information of such disparate meta programs of cancer cells. The bulk RNA sequencing (RNA-seq) data revealed high clinical prognostic values of functionally heterogeneous cancer cells of three meta programs, with transcription factor regulatory network and motif activities revealing the key transcription factors that regulate functionally heterogeneous ccRCC cells. The interactions between varying meta programs and other cell subpopulations in the microenvironment were investigated. Finally, we assessed the sensitivity of cancer cells of disparate meta programs to different anti-cancer agents. Our findings inform on the intra-tumor heterogeneity of ccRCC and its regulatory networks and offers new perspectives to facilitate the designs of rational therapeutic strategies.
Collapse
Affiliation(s)
- Jie Zheng
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
- Center for Genomic and Personalized Medicine, Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, China
| | - Wenhao Lu
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
- Center for Genomic and Personalized Medicine, Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, China
| | - Chengbang Wang
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
- Center for Genomic and Personalized Medicine, Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, China
| | - Shaohua Chen
- Department of Urology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China
- Center for Genomic and Personalized Medicine, Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, China
| | - Qingyun Zhang
- Department of Urology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China
- Center for Genomic and Personalized Medicine, Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, China
| | - Cheng Su
- Department of Pediatric Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
- Center for Genomic and Personalized Medicine, Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, China
| |
Collapse
|
3
|
Guo H, Wang T, Yu J, Shi Z, Liang M, Chen S, He T, Yan H. Vitreous Olink proteomics reveals inflammatory biomarkers for diagnosis and prognosis of traumatic proliferative vitreoretinopathy. Front Immunol 2024; 15:1355314. [PMID: 38455059 PMCID: PMC10917961 DOI: 10.3389/fimmu.2024.1355314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 02/05/2024] [Indexed: 03/09/2024] Open
Abstract
Background The aim of this study was to identify inflammatory biomarkers in traumatic proliferative vitreoretinopathy (TPVR) patients and further validate the expression curve of particular biomarkers in the rabbit TPVR model. Methods The Olink Inflammation Panel was used to compare the differentially expressed proteins (DEPs) in the vitreous of TPVR patients 7-14 days after open globe injury (OGI) (N = 19) and macular hole patients (N = 22), followed by correlation analysis between DEPs and clinical signs, protein-protein interaction (PPI) analysis, area under the receiver operating characteristic curve (AUC) analysis, and function enrichment analysis. A TPVR rabbit model was established and expression levels of candidate interleukin family members (IL-6, IL-7, and IL-33) were measured by enzyme-linked immunosorbent assay (ELISA) at 0, 1, 3, 7, 10, 14, and 28 days after OGI. Results Forty-eight DEPs were detected between the two groups. Correlation analysis showed that CXCL5, EN-RAGE, IL-7, ADA, CD5, CCL25, CASP8, TWEAK, and IL-33 were significantly correlated with clinical signs including ocular wound characteristics, PVR scoring, PVR recurrence, and final visual acuity (R = 0.467-0.699, p < 0.05), and all with optimal AUC values (0.7344-1). Correlations between DEP analysis and PPI analysis further verified that IL-6, IL-7, IL-8, IL-33, HGF, and CXCL5 were highly interactive (combined score: 0.669-0.983). These DEPs were enriched in novel pathways such as cancer signaling pathway (N = 14, p < 0.000). Vitreous levels of IL-6, IL-7, and IL-33 in the rabbit TPVR model displayed consistency with the trend in Olink data, all exhibiting marked differential expression 1 day following the OGI. Conclusion IL-7, IL-33, EN-RAGE, TWEAK, CXCL5, and CD5 may be potential biomarkers for TPVR pathogenesis and prognosis, and early post-injury may be an ideal time for TPVR intervention targeting interleukin family biomarkers.
Collapse
Affiliation(s)
- Haixia Guo
- Department of Ophthalmology, Tianjin Medical University General Hospital, Tianjin, China
| | - Tian Wang
- Shaanxi Eye Hospital, Xi’an People’s Hospital (Xi’an Fourth Hospital), Affiliated People’s Hospital of Northwest University, Xi’an, Shaanxi, China
- Institute of Medical Research, Northwestern Polytechnical University, Xi’an, Shaanxi, China
| | - Jinguo Yu
- Department of Ophthalmology, Tianjin Medical University General Hospital, Tianjin, China
| | - Zhemin Shi
- Department of Histology and Developmental Biology, Tianjin Medical University, Tianjin, China
| | - Minghui Liang
- Department of Ophthalmology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Ocular Trauma, Laboratory of Molecular Ophthalmology, Tianjin Medical University, Tianjin, China
- School of Medicine, Nankai University, Tianjin, China
| | - Siyue Chen
- Department of Ophthalmology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Ocular Trauma, Laboratory of Molecular Ophthalmology, Tianjin Medical University, Tianjin, China
| | - Tiangeng He
- Department of Ophthalmology, Tianjin Medical University General Hospital, Tianjin, China
| | - Hua Yan
- Department of Ophthalmology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Ocular Trauma, Laboratory of Molecular Ophthalmology, Tianjin Medical University, Tianjin, China
- School of Medicine, Nankai University, Tianjin, China
| |
Collapse
|
4
|
Maharati A, Tolue Ghasaban F, Akhlaghipour I, Taghehchian N, Zangouei AS, Moghbeli M. MicroRNA-495: a therapeutic and diagnostic tumor marker. J Mol Histol 2023; 54:559-578. [PMID: 37759132 DOI: 10.1007/s10735-023-10159-0] [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: 10/11/2022] [Accepted: 09/18/2023] [Indexed: 09/29/2023]
Abstract
Therapeutic and diagnostic progresses have significantly reduced the mortality rate among cancer patients during the last decade. However, there is still a high rate of mortality among cancer patients. One of the important reasons involved in the high mortality rate is the late diagnosis in advanced tumor stages that causes the failure of therapeutic strategies in these patients. Therefore, investigating the molecular mechanisms involved in tumor progression has an important role in introducing the efficient early detection markers. MicroRNAs (miRNAs) as stable factors in body fluids are always considered as non-invasive diagnostic and prognostic markers. In the present review, we investigated the role of miR-495 in tumor progression. It has been reported that miR-495 has mainly a tumor suppressor function through the regulation of transcription factors and tyrosine kinases as well as cellular processes such as multidrug resistance, chromatin remodeling, and signaling pathways. This review can be an effective step towards introducing the miR-495 as a non-invasive diagnostic/prognostic marker as well as a suitable target in tumor therapy.
Collapse
Affiliation(s)
- Amirhosein Maharati
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Faezeh Tolue Ghasaban
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Iman Akhlaghipour
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Negin Taghehchian
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Sadra Zangouei
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Meysam Moghbeli
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
5
|
Karan D, Wick J, Dubey S, Kumar-Sinha C, Siddiqui J, Kunju LP, Iczkowski KA, Chinnaiyan AM. Racial differences in serum chemokines in prostate cancer patients. Cancer 2023; 129:3783-3789. [PMID: 37698493 DOI: 10.1002/cncr.35012] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 06/15/2023] [Accepted: 06/20/2023] [Indexed: 09/13/2023]
Abstract
BACKGROUND This study aimed to understand the differential levels of inflammatory chemokines in association with higher prostate cancer incidence and mortality in African American (AA) men than in Caucasians (CA). METHODS The authors used a chemokine assay to simultaneously measure 40 chemokines and cytokines levels in the serum of preoperative prostate cancer patients and healthy controls of AA and CA races. Selected chemokines (CXCL2, CXCL5, and CCL23) serum level was validated in 211 serum samples from prostate cancer patients and healthy controls. Differential expression of CXCL5 and CCL23 was analyzed using immunohistochemistry in a representative cohort of prostate tumor tissues of AA and CA races. RESULTS Race-specific comparisons from 211 serum samples showed significantly higher levels of CXCL2 (control: 3104.0 pg/mL vs. cancer: 2451.0 pg/mL) and CXCL5 (control: 5189.0 pg/mL vs. cancer: 5459.0 pg/mL) in AA men than in CAs (CXCL2; control: 1155.0 pg/mL vs. cancer: 889.3 pg/mL, and CXCL5; control: 1183.0 pg/mL vs. cancer: 977.5 pg/mL). CCL23 differed significantly within and between the races with a lower level in AA cancer cases (454.5 vs. 966.6 pg/mL) than healthy controls (740.5 vs. 1263.0 pg/mL). Patient age, prostate-specific antigen, or Gleason scores were not significantly associated with these chemokines. Immunostaining for CXCL5 and CCL23 in a representative cohort of archival prostate tissues displayed significantly higher CXCL5 in prostate tumors than in adjacent benign tissues, whereas CCL23 was nondetectable in most of the analyzed tumor tissues. CONCLUSION Lower levels of CCL23 in AA prostate cancer patient sera and tumor tissues and high CXCL2 and CXCL5 may contribute to aggressive prostate cancer, as often seen in AA men. The disproportionate levels of serum chemokines associated with race warrant further exploration to improve equitability in precision oncology to benefit prostate cancer patients.
Collapse
Affiliation(s)
- Dev Karan
- Department of Pathology, MCW Cancer Center and Prostate Cancer Center of Excellence, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Jo Wick
- Department of Biostatistics and Data Science, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Seema Dubey
- Department of Pathology, MCW Cancer Center and Prostate Cancer Center of Excellence, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Chandan Kumar-Sinha
- Michigan Center for Translational Pathology, Department of Pathology, University of Michigan, Ann Arbor, Michigan, USA
| | - Javed Siddiqui
- Michigan Center for Translational Pathology, Department of Pathology, University of Michigan, Ann Arbor, Michigan, USA
| | - Lakshmi P Kunju
- Michigan Center for Translational Pathology, Department of Pathology, University of Michigan, Ann Arbor, Michigan, USA
| | - Kenneth A Iczkowski
- Department of Pathology, MCW Cancer Center and Prostate Cancer Center of Excellence, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Arul M Chinnaiyan
- Michigan Center for Translational Pathology, Department of Pathology, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
6
|
Saha A, Kolonin MG, DiGiovanni J. Obesity and prostate cancer - microenvironmental roles of adipose tissue. Nat Rev Urol 2023; 20:579-596. [PMID: 37198266 DOI: 10.1038/s41585-023-00764-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/24/2023] [Indexed: 05/19/2023]
Abstract
Obesity is known to have important roles in driving prostate cancer aggressiveness and increased mortality. Multiple mechanisms have been postulated for these clinical observations, including effects of diet and lifestyle, systemic changes in energy balance and hormonal regulation and activation of signalling by growth factors and cytokines and other components of the immune system. Over the past decade, research on obesity has shifted towards investigating the role of peri-prostatic white adipose tissue as an important source of locally produced factors that stimulate prostate cancer progression. Cells that comprise white adipose tissue, the adipocytes and their progenitor adipose stromal cells (ASCs), which proliferate to accommodate white adipose tissue expansion in obesity, have been identified as important drivers of obesity-associated cancer progression. Accumulating evidence suggests that adipocytes are a source of lipids that are used by adjacent prostate cancer cells. However, results of preclinical studies indicate that ASCs promote tumour growth by remodelling extracellular matrix and supporting neovascularization, contributing to the recruitment of immunosuppressive cells, and inducing epithelial-mesenchymal transition through paracrine signalling. Because epithelial-mesenchymal transition is associated with cancer chemotherapy resistance and metastasis, ASCs are considered to be potential targets of therapies that could be developed to suppress cancer aggressiveness in patients with obesity.
Collapse
Affiliation(s)
- Achinto Saha
- Division of Pharmacology and Toxicology and Dell Paediatric Research Institute, The University of Texas at Austin, Austin, TX, USA
- Center for Molecular Carcinogenesis and Toxicology, The University of Texas at Austin, Austin, TX, USA
- Livestrong Cancer Institutes, Dell Medical School, The University of Texas at Austin, Austin, TX, USA
| | - Mikhail G Kolonin
- The Brown Foundation Institute of Molecular Medicine for the Prevention of Disease, The University of Texas Health Sciences Center at Houston, Houston, Texas, USA.
| | - John DiGiovanni
- Division of Pharmacology and Toxicology and Dell Paediatric Research Institute, The University of Texas at Austin, Austin, TX, USA.
- Center for Molecular Carcinogenesis and Toxicology, The University of Texas at Austin, Austin, TX, USA.
- Livestrong Cancer Institutes, Dell Medical School, The University of Texas at Austin, Austin, TX, USA.
| |
Collapse
|
7
|
Arebro J, Towle R, Lee CM, Bennewith KL, Garnis C. Extracellular vesicles promote activation of pro-inflammatory cancer-associated fibroblasts in oral cancer. Front Cell Dev Biol 2023; 11:1240159. [PMID: 37745296 PMCID: PMC10513103 DOI: 10.3389/fcell.2023.1240159] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 08/23/2023] [Indexed: 09/26/2023] Open
Abstract
Introduction: Oral squamous cell carcinoma (OSCC) is the most common form of head and neck cancer and has a survival rate of ∼50% over 5 years. New treatment strategies are sorely needed to improve survival rates-and a better understanding of the mechanisms underlying tumorigenesis is needed to develop these strategies. The role of the tumor microenvironment (TME) has increasingly been identified as crucial in tumor progression and metastasis. One of the main constituents of the TME, cancer-associated fibroblasts (CAFs), plays a key role in influencing the biological behavior of tumors. Multiple mechanisms contribute to CAF activation, such as TGFβ signaling, but the role of extracellular vesicles (EVs) in CAF activation in OSCC is poorly understood. Assessing the impact of oral cancer-derived EVs on CAF activation will help to better illuminate OSCC pathophysiology and may drive development of novel treatments options. Methods: EVs were isolated from OSCC cell lines (Cal 27, SCC-9, SCC-25) using differential centrifugation. Nanoparticle tracking analysis was used for EV characterization, and Western blot to confirm the presence of EV protein markers. Oral fibroblasts were co-cultured with enriched EVs, TGFβ, or PBS over 72 h to assess activation. Flow cytometry was used to evaluate CAF markers. RNA collected from fibroblasts was extracted and the transcriptome was sequenced. Conditioned media from the co-cultures was evaluated with cytokine array profiling. Results: OSCC-derived EVs can activate oral fibroblasts into CAFs that are different from those activated by TGFβ, suggesting different mechanisms of activation and different functional properties. Gene set enrichment analysis showed several upregulated inflammatory pathways in those CAFs exposed to OSCC-derived EVs. Marker genes for inflammatory CAF subtypes were also upregulated, but not in CAFs activated by TGFβ. Finally, cytokine array analysis on secreted proteins revealed elevated levels of several pro-inflammatory cytokines from EV-activated CAFs, for instance IL-8 and CXCL5. Discussion: Our results reveal the ability of OSCC-derived EVs to activate fibroblasts into CAFs. These CAFs seem to have unique properties, differing from TGFβ-activated CAFs. Gaining an understanding of the interplay between EVs and stromal cells such as CAFs could lead to further insights into OSCC tumorigenesis and potential novel therapeutics.
Collapse
Affiliation(s)
- Julia Arebro
- Department of Integrative Oncology, British Columbia Cancer Research Center, Vancouver, BC, Canada
- Division of ENT Diseases, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
- Department of ENT Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Rebecca Towle
- Department of Integrative Oncology, British Columbia Cancer Research Center, Vancouver, BC, Canada
| | - Che-Min Lee
- Department of Integrative Oncology, British Columbia Cancer Research Center, Vancouver, BC, Canada
- Interdisciplinary Oncology Program, University of British Columbia, Vancouver, BC, Canada
| | - Kevin L. Bennewith
- Department of Integrative Oncology, British Columbia Cancer Research Center, Vancouver, BC, Canada
- Interdisciplinary Oncology Program, University of British Columbia, Vancouver, BC, Canada
- Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Cathie Garnis
- Department of Integrative Oncology, British Columbia Cancer Research Center, Vancouver, BC, Canada
- Interdisciplinary Oncology Program, University of British Columbia, Vancouver, BC, Canada
- Division of Otolaryngology, Department of Surgery, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
8
|
Wang ZZ, Li XT, Li QJ, Zhou JX. Targeting CXCL5 in Pancreatic Cancer Cells Inhibits Cancer Xenograft Growth by Reducing Proliferation and Inhibiting EMT Progression. Dig Dis Sci 2023; 68:841-851. [PMID: 35650416 DOI: 10.1007/s10620-022-07529-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 04/18/2022] [Indexed: 12/09/2022]
Abstract
BACKGROUND Pancreatic cancer (PC) is the most lethal malignant tumor, with average survival period of about 10 months. C-X-C ligand 5 (CXCL5), an important chemokine for immune cell accumulation in tumor tissues, has been reported to be involved in a variety of human cancers. However, the exact role of CXCL5 in PC progression has not been well defined. METHODS The expression of CXCL5 in PC was analyzed based on online databases and clinical specimens immunohistochemical staining, and Western blotting of CXCL5 in PC cell lines and patient samples. The correlation between CXCL5 expression and prognosis in PC was explored. The role of CXCL5 in PC was investigated through in vitro and in vivo experiments. RESULTS The expression of CXCL5 was significantly increased in PC tissues compared with that in pancreas tissues, and CXCL5 high expression predicts poor prognosis in PC patients. Further analyses demonstrated that overexpression of CXCL5 in PC cells was positively related to higher proliferation rate, higher migration ability, and higher EMT markers including SNAI2 and TWIST1 of tumor cells in vitro. Consistently, the knockdown of CXCL5 in PC cells harmed the proliferation rate, migration ability, and expression of EMT indexes of tumor cells in vitro. Importantly, knockdown of CXCL5 inhibited the growth of xenograft tumors in vivo. CONCLUSION CXCL5 high expression predicts poor prognosis in PC patients. CXCL5 promotes PC cell growth and EMT process. Inhibition of CXCL5 may be a potential therapeutic approach for PC.
Collapse
Affiliation(s)
- Zheng-Zheng Wang
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, 450008, China
| | - Xiao-Ting Li
- Department of Obstetrics and Gynecology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, 450003, China
| | - Qing-Jun Li
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, 450008, China
| | - Jin-Xue Zhou
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, 450008, China.
| |
Collapse
|
9
|
Yin L, Zhang J, Sun Y. Early growth response-1 is a new substrate of the GSK3β-FBXW7 axis. Neoplasia 2022; 34:100839. [PMID: 36240645 PMCID: PMC9573921 DOI: 10.1016/j.neo.2022.100839] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 09/25/2022] [Accepted: 09/26/2022] [Indexed: 11/06/2022]
Abstract
EGR1, a short-lived transcription factor, regulates several biological processes, including cell proliferation and tumor progression. Whether and how EGR1 is regulated by Cullin-RING ligases (CRLs) remains elusive. Here, we report that MLN4924, a small molecule inhibitor of neddylation, causes EGR1 accumulation by inactivating SCFFBXW7 (CRL1), which is a new E3 ligase for EGR1. Specifically, FBXW7 binds to EGR1 via its consensus binding motif/degron, whereas cancer-derived FBXW7 mutants showed a much reduced EGR1 binding. SiRNA-mediated FBXW7 knockdown caused EGR1 accumulation, whereas FBXW7 overexpression reduced EGR1 levels. Likewise, FBXW7 knockdown significantly extended EGR1 protein half-life, while FBXW7 overexpression promotes polyubiquitylation of wild-type EGR1, but not EGR1-S2A mutant with the binding site abrogated. GSK3β kinase is required for the FBXW7-EGR1 binding, and for enhanced EGR1 degradation by wild type FBXW7, but not by FBXW7 mutants. Likewise, GSK3β knockdown or treatment with GSK3β inhibitor significantly increased the EGR1 levels and extended EGR1 protein half-life, while reducing EGR1 polyubiquitylation. Hypoxia exposure reduces the EGR1 levels via enhancing the FBXW7-EGR1 binding, and FBXW7-induced EGR1 polyubiquitylation. Biologically, EGR1 knockdown suppressed cancer cell growth, whereas growth stimulation by FBXW7 knockdown is partially rescued by EGR1 knockdown. Thus, EGR1 is a new substrate of the GSK3β-FBXW7 axis, and the FBXW7-EGR1 axis coordinately regulates growth of cancer cells.
Collapse
Affiliation(s)
- Lu Yin
- Cancer Institute of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China; Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310029, China
| | - Jiagui Zhang
- Cancer Institute of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China; Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310029, China
| | - Yi Sun
- Cancer Institute of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China; Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310029, China; Cancer Center, Zhejiang University, Hangzhou 310058, China; Research Center for Life Science and Human Health, Binjiang Institute of Zhejiang University, Hangzhou 310053, China.
| |
Collapse
|
10
|
Xu J, Li J. Construction of a three commitment points for S phase entry cell cycle model and immune-related ceRNA network to explore novel therapeutic options for psoriasis. MATHEMATICAL BIOSCIENCES AND ENGINEERING : MBE 2022; 19:13483-13525. [PMID: 36654055 DOI: 10.3934/mbe.2022630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
While competing endogenous RNAs (ceRNAs) play pivotal roles in various diseases, the proliferation and differentiation of keratinocytes are becoming a research focus in psoriasis. Therefore, the three commitment points for S phase entry (CP1-3) cell cycle model has pointed to a new research direction in these areas. However, it is unclear what role ceRNA regulatory mechanisms play in the interaction between keratinocytes and the immune system in psoriasis. In addition, the ceRNA network-based screening of potential therapeutic agents for psoriasis has not been explored. Therefore, we used multiple bioinformatics approaches to construct a ceRNA network for psoriasis, identified CTGF as the hub gene, and constructed a ceRNA subnetwork, after which validation datasets authenticated the results' accuracy. Subsequently, we used multiple online databases and the single-sample gene-set enrichment analysis algorithm, including the CP1-3 cell cycle model, to explore the mechanisms accounting for the increased proliferation and differentiation of keratinocytes and the possible roles of the ceRNA subnetwork in psoriasis. Next, we performed cell cycle and cell trajectory analyses based on a single-cell RNA-seq dataset of psoriatic skin biopsies. We also used weighted gene co-expression network analysis and single-gene batch correlation analysis-based gene set enrichment analysis to explore the functions of CTGF. Finally, we used the Connectivity Map to identify MS-275 (entinostat) as a novel treatment for psoriasis, SwissTargetPrediction to predict drug targets, and molecular docking to investigate the minimum binding energy and binding sites of the drug to target proteins.
Collapse
Affiliation(s)
- Jingxi Xu
- North Sichuan Medical College, Nanchong 637000, China
- Department of Rheumatology and Immunology, The First People's Hospital of Yibin, Yibin 644000, China
| | - Jiangtao Li
- Department of Rheumatology and Immunology, The First People's Hospital of Yibin, Yibin 644000, China
| |
Collapse
|
11
|
Deng J, Jiang R, Meng E, Wu H. CXCL5: A coachman to drive cancer progression. Front Oncol 2022; 12:944494. [PMID: 35978824 PMCID: PMC9376318 DOI: 10.3389/fonc.2022.944494] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Accepted: 07/08/2022] [Indexed: 11/13/2022] Open
Abstract
Chemokines are a class of pro-inflammatory cytokines that can recruit and activate chemotactic cells. C‐X‐C motif chemokine ligand 5 (CXCL5) is a member of the chemokine family binding CXCR2 (C-X-C Motif Chemokine Receptor 2), a G-protein coupled receptor. Accumulated evidence has shown that dysregulated CXCL5 participates in tumor metastasis and angiogenesis in human malignant tumors. In this review, we summarized the advances in research on CXCL5, including its dysregulation in different tumors and the mechanism associated with tumor behavior (formation of the immunosuppressive microenvironment, promotion of tumor angiogenesis, and metastasis). We also summarized and discussed the perspective about the potential application of CXCL5 in tumor therapy targeting the tumor inflammatory microenvironment.
Collapse
|
12
|
Balázs K, Kocsis ZS, Ágoston P, Jorgo K, Gesztesi L, Farkas G, Székely G, Takácsi-Nagy Z, Polgár C, Sáfrány G, Jurányi Z, Lumniczky K. Prostate Cancer Survivors Present Long-Term, Residual Systemic Immune Alterations. Cancers (Basel) 2022; 14:3058. [PMID: 35804830 PMCID: PMC9264868 DOI: 10.3390/cancers14133058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 06/10/2022] [Accepted: 06/16/2022] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND The development of cancer and anti-tumor therapies can lead to systemic immune alterations but little is known about how long immune dysfunction persists in cancer survivors. METHODS We followed changes in the cellular immune parameters of prostate cancer patients with good prognostic criteria treated with low dose rate brachytherapy before and up to 3 years after the initiation of therapy. RESULTS Patients before therapy had a reduced CD4+ T cell pool and increased regulatory T cell fraction and these alterations persisted or got amplified during the 36-month follow-up. A significant decrease in the total NK cell number and a redistribution of the circulating NK cells in favor of a less functional anergic subpopulation was seen in patients before therapy but tumor regression led to the regeneration of the NK cell pool and functional integrity. The fraction of lymphoid DCs was increased in patients both before therapy and throughout the whole follow-up. Increased PDGF-AA, BB, CCL5 and CXCL5 levels were measured in patients before treatment but protein levels rapidly normalized. CONCLUSIONS while NK cell dysfunction recovered, long-term, residual alterations persisted in the adaptive and partly in the innate immune system.
Collapse
Affiliation(s)
- Katalin Balázs
- National Public Health Center, Unit of Radiation Medicine, Department of Radiobiology and Radiohygiene, 1221 Budapest, Hungary; (K.B.); (G.S.)
- Doctoral School of Pathological Sciences, Semmelweis University, 1085 Budapest, Hungary
| | - Zsuzsa S. Kocsis
- Department of Radiobiology and Diagnostic Onco-Cytogenetics and The National Tumorbiology Laboratory, Centre of Radiotherapy, National Institute of Oncology, 1122 Budapest, Hungary; (Z.S.K.); (G.F.); (G.S.); (Z.J.)
| | - Péter Ágoston
- Centre of Radiotherapy and The National Tumorbiology Laboratory, National Institute of Oncology, 1122 Budapest, Hungary; (P.Á.); (K.J.); (L.G.); (Z.T.-N.); (C.P.)
- Department of Oncology, Semmelweis University, 1122 Budapest, Hungary
| | - Kliton Jorgo
- Centre of Radiotherapy and The National Tumorbiology Laboratory, National Institute of Oncology, 1122 Budapest, Hungary; (P.Á.); (K.J.); (L.G.); (Z.T.-N.); (C.P.)
- Department of Oncology, Semmelweis University, 1122 Budapest, Hungary
| | - László Gesztesi
- Centre of Radiotherapy and The National Tumorbiology Laboratory, National Institute of Oncology, 1122 Budapest, Hungary; (P.Á.); (K.J.); (L.G.); (Z.T.-N.); (C.P.)
| | - Gyöngyi Farkas
- Department of Radiobiology and Diagnostic Onco-Cytogenetics and The National Tumorbiology Laboratory, Centre of Radiotherapy, National Institute of Oncology, 1122 Budapest, Hungary; (Z.S.K.); (G.F.); (G.S.); (Z.J.)
| | - Gábor Székely
- Department of Radiobiology and Diagnostic Onco-Cytogenetics and The National Tumorbiology Laboratory, Centre of Radiotherapy, National Institute of Oncology, 1122 Budapest, Hungary; (Z.S.K.); (G.F.); (G.S.); (Z.J.)
| | - Zoltán Takácsi-Nagy
- Centre of Radiotherapy and The National Tumorbiology Laboratory, National Institute of Oncology, 1122 Budapest, Hungary; (P.Á.); (K.J.); (L.G.); (Z.T.-N.); (C.P.)
- Department of Oncology, Semmelweis University, 1122 Budapest, Hungary
| | - Csaba Polgár
- Centre of Radiotherapy and The National Tumorbiology Laboratory, National Institute of Oncology, 1122 Budapest, Hungary; (P.Á.); (K.J.); (L.G.); (Z.T.-N.); (C.P.)
- Department of Oncology, Semmelweis University, 1122 Budapest, Hungary
| | - Géza Sáfrány
- National Public Health Center, Unit of Radiation Medicine, Department of Radiobiology and Radiohygiene, 1221 Budapest, Hungary; (K.B.); (G.S.)
| | - Zsolt Jurányi
- Department of Radiobiology and Diagnostic Onco-Cytogenetics and The National Tumorbiology Laboratory, Centre of Radiotherapy, National Institute of Oncology, 1122 Budapest, Hungary; (Z.S.K.); (G.F.); (G.S.); (Z.J.)
| | - Katalin Lumniczky
- National Public Health Center, Unit of Radiation Medicine, Department of Radiobiology and Radiohygiene, 1221 Budapest, Hungary; (K.B.); (G.S.)
| |
Collapse
|
13
|
Ren Z, Chen Y, Shi L, Shao F, Sun Y, Ge J, Zhang J, Zang Y. Sox9/CXCL5 axis facilitates tumour cell growth and invasion in hepatocellular carcinoma. FEBS J 2022; 289:3535-3549. [PMID: 35038357 DOI: 10.1111/febs.16357] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Revised: 12/20/2021] [Accepted: 01/13/2022] [Indexed: 12/11/2022]
Abstract
High rates of metastasis and postsurgical recurrence contribute to the higher mortality of hepatocellular carcinoma (HCC), partly due to cancer stem cell (CSC)-dependent tumorigenesis and metastasis. Sex-determining region Y-box 9 (Sox9) has been previously characterized as a candidate CSC marker of HCC. Here, we observed that the increase of Sox9 significantly promoted HCC cell growth and invasion in cell cultures, whereas knockdown of Sox9 showed the opposite effects, suggesting that Sox9 may regulate the proliferation and invasion of hepatoma cells in an autocrine manner. RNA sequencing, together with functional assays and clinical analyses, identified CXCL5 as a key mediator downstream of Sox9 in HCC cells. Mechanistic studies revealed that Sox9 induced CXCL5 expression by directly binding to a promoter region. Using gain- and loss-of-function approaches, we demonstrated that the intrinsic effective role of Sox9 in hepatoma cell growth and invasion depended on CXCL5, and that blockade of CXCL5/CXCR2 signalling abolished Sox9-triggered HCC cell proliferation and migration. Furthermore, the Sox9/CXCL5 axis activated PI3K-AKT and ERK1/2 signalling which are implicated in regulating HCC cell proliferation and invasion. Finally, the Sox9/CXCL5 axis contributed to the infiltration of neutrophils and macrophages in both tumour and peritumoral tissues from the orthotopic xenograft model. In summary, our data identify the Sox9/CXCL5 axis as an endogenous factor in controlling HCC cell growth and invasion, thereby raising the possibility of pharmacologic intervention with CXCL5/CXCR2 pathway inhibitors in therapy for HCC patients with higher Sox9 expression.
Collapse
Affiliation(s)
- Zhengrong Ren
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Science, Nanjing University, China
| | - Yu Chen
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Science, Nanjing University, China
| | - Lei Shi
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Science, Nanjing University, China
| | - Fang Shao
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Science, Nanjing University, China
| | - Yanyan Sun
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Science, Nanjing University, China
| | - Jia Ge
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Science, Nanjing University, China
| | - Junfeng Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Science, Nanjing University, China
| | - Yuhui Zang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Science, Nanjing University, China
| |
Collapse
|
14
|
Murashka DI, Tahanovich AD, Kauhanka MM, Gotko OV, Prokhorova VI. On the issue of diagnostic value of determining the level of receptors and their ligands in blood in non-small cell lung cancer. Klin Lab Diagn 2022; 67:277-285. [PMID: 35613346 DOI: 10.51620/0869-2084-2022-67-5-277-285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Non-small cell lung cancer (NSCLC) occupies the first place in the structure of mortality due to oncological diseases. Late diagnosis worsens the effectiveness of its treatment. There are no informative biomarkers that allow us to judge the prevalence of the tumor process, especially in the early stages of NSCLC. To determine the level of CXCL5, CXCL8, CXCR1 and CXCR2 in the peripheral blood of patients with NSCLC to assess the possibility of their use in the diagnosis of the disease. The material was the blood of 218 patients with NSCLC, 19 patients with lung hamartoma and 42 healthy people. The concentration of CXCL5, CXCL8, and SCC in blood serum was determined by enzyme immunoassay, the CYFRA 21-1 level was determined by immunochemiluminescence analysis. The proportion of leukocytes equipped with CXCR1 and CXCR2 receptors and the fluorescence intensity of receptor complexes with antibodies (MFI) in them were measured by flow cytometry. MFI CXCR1 in granulocytes and the proportion of lymphocytes supplied CXCR2, increased in the blood already at stage I of NSCLC and showed an even more significant increase in subsequent stages. The level of these indicators was correlatively related to the stages and characteristics of NSCLC. Measuring the level of MFI CXCR1 in the blood serum makes it possible to diagnose the early stages of NSCLC with a sensitivity of 87.4% (specificity - 73.8%). Determination of the proportion of lymphocytes equipped with CXCR2 demonstrates comparable diagnostic sensitivity (87.2%) and specificity of 66.7% in the detection of stages I-II of NSCLC. MFI CXCR1 in granulocytes can also be used to differentiate stages I and II of NSCLC (diagnostic sensitivity - 75,3%, specificity - 69,6%). The sensitivity of determining for this purpose the proportion of lymphocytes equipped with CXCR2 is 75.0% with a specificity of 71.7%. In 89.7% of patients with stages III-IV NSCLC, the MFI CXCR1 in granulocytes exceeds the threshold value of 47.8 (specificity - 74.8%). Diagnostic sensitivity of determining the proportion of lymphocytes for this purpose was 90.7%.
Collapse
Affiliation(s)
- D I Murashka
- Belarusian stage medical university, Belarusian Ministry of Health
| | - A D Tahanovich
- Belarusian stage medical university, Belarusian Ministry of Health
| | - M M Kauhanka
- Belarusian stage medical university, Belarusian Ministry of Health
| | - O V Gotko
- N.N. Alexandrov Republican Scientific-Practical Centre of Oncology and Medical Radiology, Belarusian Ministry of Health
| | - V I Prokhorova
- N.N. Alexandrov Republican Scientific-Practical Centre of Oncology and Medical Radiology, Belarusian Ministry of Health
| |
Collapse
|
15
|
A highly branched α-D-glucan facilitates antitumor immunity by reducing cancer cell CXCL5 expression. Int J Biol Macromol 2022; 209:166-179. [PMID: 35390399 DOI: 10.1016/j.ijbiomac.2022.03.217] [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: 10/25/2021] [Revised: 03/22/2022] [Accepted: 03/31/2022] [Indexed: 11/22/2022]
Abstract
Tumor immunotherapy has emerged as a major pillar of anticancer therapeutic strategies. Natural polysaccharides, known for their strong immunomodulatory activities with relatively low cost and toxicity, are becoming promising prospects for cancer immunotherapy. In this study, we investigated the antitumor mechanism of JNY2PW, a highly branched α-D-glucan previously purified from the traditional marine Chinese medicine Arca inflata. JNY2PW was shown to enhance the sensitivity of tumor cells to co-culture macrophage supernatants by decreasing cancer cell CXCL5 expression. Furthermore, JNY2PW exerted antitumor effects without obvious toxic side effects in tumor-bearing mice by triggering the Akt/mTOR and ERK/GSK3β/β-catenin pathways and attenuating expression of CXCL5 in cancer cells. Remarkably, JNY2PW reduced tumor proliferation and dampened CXCL5 expression in tumor cells overexpressing CXCL5 both in vitro and in vivo. Additionally, JNY2PW blocked epithelial-mesenchymal transition (EMT) in both CXCL5-overexpressing and wild type tumor cells. Our data therefore uncovered a previously unrecognized antitumor mechanism for JNY2PW, suggesting that JNY2PW is a promising adjuvant as an immunomodulator for cancer immunotherapy.
Collapse
|
16
|
Wang W, Zhang M, Huang Z, Wang L, Yue Y, Wang X, Lu S, Fan J. Knockdown of CXCL5 inhibits the invasion, metastasis and stemness of bladder cancer lung metastatic cells by downregulating CD44. Anticancer Drugs 2022; 33:e103-e112. [PMID: 34407043 PMCID: PMC8670357 DOI: 10.1097/cad.0000000000001153] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 06/04/2021] [Indexed: 12/09/2022]
Abstract
In our previous studies, we found that T24 lung metastatic cancer cells showed high invasion and metastasis abilities and cancer stem cell characteristics compared with T24 primary cancer cells. By screening for the expression of CXC chemokines in both cell lines, we found that CXCL5 is highly expressed in T24-L cells. The aim of this study is to shed light on the relationship of CXCL5 with epithelial-mesenchymal transition (EMT) and cancer stem cells (CSCs). RNAi technology was used to decrease CXCL5 expression in the T24-L cell line, and the EMT and CSCs of the shCXCL5 group and the control group were compared. The CXCR2 inhibitor SB225002 was used to inhibit the receptor of CXCL5 to determine the effect of the CXCL5/CXCR2 axis. The knockdown of CXCL5 expression in T24-L cells reduced their EMT and CSC characteristics. RT-PCR and Western blot analyses revealed the downregulation of N-cadherin, Vimentin and CD44. In addition, when CD44 expression was knocked down, the EMT ability of the cells was also inhibited. This phenomenon was most pronounced when both CXCL5 and CD44 were knocked down. CXCL5 and CD44 can affect the EMT and stem cell capacity of T24-L cells through some interaction.
Collapse
Affiliation(s)
- Weiyi Wang
- Department of Urology
- Department of Vascular Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an
| | | | - Zhixin Huang
- Department of Urology
- Department of Urology, Yulin First Hospital, Shaanxi Province
| | | | - Yangyang Yue
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Xi’an Jiaotong University
| | - Xinyang Wang
- Oncology Research Lab, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi’an, China
| | - Shaoying Lu
- Department of Vascular Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an
| | - Jinhai Fan
- Department of Urology
- Oncology Research Lab, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi’an, China
| |
Collapse
|
17
|
Wang B, Guo H, Yu H, Chen Y, Xu H, Zhao G. The Role of the Transcription Factor EGR1 in Cancer. Front Oncol 2021; 11:642547. [PMID: 33842351 PMCID: PMC8024650 DOI: 10.3389/fonc.2021.642547] [Citation(s) in RCA: 102] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 02/25/2021] [Indexed: 12/12/2022] Open
Abstract
Early growth response factor 1 (EGR1) is a transcription factor that is mainly involved in the processes of tissue injury, immune responses, and fibrosis. Recent studies have shown that EGR1 is closely related to the initiation and progression of cancer and may participate in tumor cell proliferation, invasion, and metastasis and in tumor angiogenesis. Nonetheless, the specific mechanism whereby EGR1 modulates these processes remains to be elucidated. This review article summarizes possible mechanisms of action of EGR1 in tumorigenesis and tumor progression and may serve as a reference for clinical efficacy predictions and for the discovery of new therapeutic targets.
Collapse
Affiliation(s)
- Bin Wang
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, China
| | - Hanfei Guo
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Hongquan Yu
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, China
| | - Yong Chen
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, China
| | - Haiyang Xu
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, China
| | - Gang Zhao
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
18
|
Tsui KH, Chang KS, Sung HC, Hsu SY, Lin YH, Hou CP, Yang PS, Chen CL, Feng TH, Juang HH. Mucosa-Associated Lymphoid Tissue 1 Is an Oncogene Inducing Cell Proliferation, Invasion, and Tumor Growth via the Upregulation of NF-κB Activity in Human Prostate Carcinoma Cells. Biomedicines 2021; 9:biomedicines9030250. [PMID: 33802402 PMCID: PMC8000469 DOI: 10.3390/biomedicines9030250] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 02/21/2021] [Accepted: 02/26/2021] [Indexed: 11/16/2022] Open
Abstract
Prostate cancer is one of the most common seen malignancies and the leading cause of cancer-related death among men. Given the importance of early diagnosis and treatment, it is worth to identify a potential novel therapeutic target for prostate cancer. Mucosa-associated lymphoid tissue 1 (MALT1) is a novel gene involved in nuclear factor κB (NF-κB) signal transduction by acting as an adaptor protein and paracaspase, with an essential role in inflammation and tumorigenesis in many cancers. This study investigated the functions and the potential regulatory mechanisms of MALT1 in the human prostate cancer cells. We found that MALT1 is abundant in prostate cancer tissues. MALT1 facilitated NF-κB subunits (p50 and p65) nuclear translocation to induce gene expression of interleukin 6 (IL-6) and C-X-C motif chemokine 5 (CXCL5) in prostate carcinoma cells. MALT1 promoted cell proliferation, invasion, and tumor growth in vitro and in vivo. MALT1 enhanced NF-κB activity in prostate carcinoma cells; moreover, NF-κB induced MALT1 expression determined by reporter and immunoblot assays, implying there is a positive feedback loop between MALT1 and NF-κB. In conclusion, MALT1 is a NF-κB-induced oncogene in the human prostate carcinoma cells.
Collapse
Affiliation(s)
- Ke-Hung Tsui
- Department of Urology, Chang Gung Memorial Hospital-Linkou, Kwei-Shan, Tao-Yuan 33302, Taiwan; (K.-H.T.); (Y.-H.L.); (C.-P.H.); (P.-S.Y.); (C.-L.C.)
| | - Kang-Shuo Chang
- Department of Anatomy, College of Medicine, Chang Gung University, Kwei-Shan, Tao-Yuan 33302, Taiwan; (K.-S.C.); (H.-C.S.); (S.-Y.H.)
| | - Hsin-Ching Sung
- Department of Anatomy, College of Medicine, Chang Gung University, Kwei-Shan, Tao-Yuan 33302, Taiwan; (K.-S.C.); (H.-C.S.); (S.-Y.H.)
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Kwei-Shan, Tao-Yuan 33302, Taiwan
| | - Shu-Yuan Hsu
- Department of Anatomy, College of Medicine, Chang Gung University, Kwei-Shan, Tao-Yuan 33302, Taiwan; (K.-S.C.); (H.-C.S.); (S.-Y.H.)
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Kwei-Shan, Tao-Yuan 33302, Taiwan
| | - Yu-Hsiang Lin
- Department of Urology, Chang Gung Memorial Hospital-Linkou, Kwei-Shan, Tao-Yuan 33302, Taiwan; (K.-H.T.); (Y.-H.L.); (C.-P.H.); (P.-S.Y.); (C.-L.C.)
| | - Chen-Pang Hou
- Department of Urology, Chang Gung Memorial Hospital-Linkou, Kwei-Shan, Tao-Yuan 33302, Taiwan; (K.-H.T.); (Y.-H.L.); (C.-P.H.); (P.-S.Y.); (C.-L.C.)
| | - Pei-Shan Yang
- Department of Urology, Chang Gung Memorial Hospital-Linkou, Kwei-Shan, Tao-Yuan 33302, Taiwan; (K.-H.T.); (Y.-H.L.); (C.-P.H.); (P.-S.Y.); (C.-L.C.)
| | - Chien-Lun Chen
- Department of Urology, Chang Gung Memorial Hospital-Linkou, Kwei-Shan, Tao-Yuan 33302, Taiwan; (K.-H.T.); (Y.-H.L.); (C.-P.H.); (P.-S.Y.); (C.-L.C.)
| | - Tsui-Hsia Feng
- School of Nursing, College of Medicine, Chang Gung University, Kwei-Shan, Tao-Yuan 33302, Taiwan;
| | - Horng-Heng Juang
- Department of Urology, Chang Gung Memorial Hospital-Linkou, Kwei-Shan, Tao-Yuan 33302, Taiwan; (K.-H.T.); (Y.-H.L.); (C.-P.H.); (P.-S.Y.); (C.-L.C.)
- Department of Anatomy, College of Medicine, Chang Gung University, Kwei-Shan, Tao-Yuan 33302, Taiwan; (K.-S.C.); (H.-C.S.); (S.-Y.H.)
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Kwei-Shan, Tao-Yuan 33302, Taiwan
- Correspondence: ; Tel.: +886-3-2118800; Fax: +886-3-2118112
| |
Collapse
|
19
|
Mutalifu N, Du P, Zhang J, Akbar H, Yan B, Alimu S, Tong L, Luan X. Circ_0000215 Increases the Expression of CXCR2 and Promoted the Progression of Glioma Cells by Sponging miR-495-3p. Technol Cancer Res Treat 2020; 19:1533033820957026. [PMID: 33089764 PMCID: PMC7586024 DOI: 10.1177/1533033820957026] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND In recent years, accumulating studies have found that circular RNA (circRNA) exerts a great effect on tumor progression. Circ_0000215, a novel circRNA, remains largely unknown in terms of its effect and mechanism in glioma. METHOD Quantitative real-time polymerase chain reaction (qRT-PCR) was carried out to detect the expressions of circ_0000215, miR-495-3p and CXCR2 in human glial cell line HEB and glioma cell lines (A172, U251, U87, SHG-44, LN-18), human glioma tissues and adjacent healthy tissues. Gain- and loss-assays of circ_0000215 were conducted. Cell proliferation ability was detected via the CCK8 assay, and cell invasion ability was examined by Transwell assay. CXCR2 expression was evaluated via RT-PCR and Western blot. Moreover, bioinformatics was applied to analyze the targeting molecules of circ_0000215 and CXCR2. Verification of the relationship between these molecules were supported through the dual-luciferase reporter gene and RNA immunocoprecipitation (RIP) assay. RESULTS Circ_0000215 and CXCR2 were remarkably upregulated in glioma tissues and cells. Overexpression of circ_0000215 notably promoted the proliferation, invasion and epithelial-mesenchymal transition (EMT) but inhibited apoptosis of glioma cells, while knocking down circ_0000215 had the opposite effects. Additionally, miR-495-3p, a sponge RNA of circ_0000215, inhibited the growth, invasion and EMT of glioma cells. Mechanistically, miR-495-3p targeted CXCR2 and negatively regulated CXCR2/PI3K/Akt pathway. However, the effects of miR-495-3p were all dampened by overexpression of circ_0000215. CONCLUSION These data demonstrated that circ_0000215 functions as a competitive endogenous RNA by sponging miR-495-3p, thus accelerating glioma progression through CXCR2 axis.
Collapse
Affiliation(s)
- Nurehemaiti Mutalifu
- Department of Neurosurgery, Second Affiliated Hospital, 223527Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Peng Du
- Department of Neurosurgery, Second Affiliated Hospital, 223527Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Jingjing Zhang
- Department of Neurosurgery, Second Affiliated Hospital, 223527Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Halik Akbar
- Department of Neurosurgery, Second Affiliated Hospital, 223527Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Baofeng Yan
- Department of Neurosurgery, Second Affiliated Hospital, 223527Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Sulaiman Alimu
- Department of Neurosurgery, Second Affiliated Hospital, 223527Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Lingxiao Tong
- Department of Neurosurgery, Second Affiliated Hospital, 223527Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Xinping Luan
- Department of Neurosurgery, Second Affiliated Hospital, 223527Xinjiang Medical University, Urumqi, Xinjiang, China
| |
Collapse
|
20
|
Kawagoe Y, Kawashima I, Sato Y, Okamoto N, Matsubara K, Kawamura K. CXCL5-CXCR2 signaling is a senescence-associated secretory phenotype in preimplantation embryos. Aging Cell 2020; 19:e13240. [PMID: 32959976 PMCID: PMC7576282 DOI: 10.1111/acel.13240] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 08/15/2020] [Accepted: 08/22/2020] [Indexed: 12/12/2022] Open
Abstract
Pregnancy rate of women decreases with age due to declining quality of oocytes and embryos. However, there is no established method to improve pregnancy rate in aging women. In this study, we identified a senescence-associated secretory phenotype (SASP) factor partially responsible for the decline in embryo implantation potential. Based on microarray analysis using young and aging human embryos at the same morphological grade, 702 genes showed >fivefold increases in aging human blastocysts. Among these genes, C-X-C motif chemokine 5 (CXCL5) showed 7.7-fold increases in aging human blastocysts. However, no-age-dependent changes in expression of the CXCR2, the cognate receptor for CXCL5, were found. In aging mice, Cxcl5 transcript levels were also increased in oocytes and embryos. Treatment of young mouse embryos with CXCL5 decreased implantation rates, together with increased expression of aging markers (P53, P21, Pai-1, and Il-6). Moreover, CXCL5 treatment suppressed trophoblast outgrowth in young mouse blastocysts. Conversely, suppression of CXCL5-CXCR2 signaling in aging mouse embryos using neutralizing antibodies and a receptor antagonist improved the implantation rate, leading to increases in pregnancy and delivery of normal pups. The gene expression pattern of these embryos was comparable to that in young mouse embryos showing enriched cell proliferation-related pathways. In conclusion, we identified CXCL5 as a SASP factor in human and mouse embryos and suppression of CXCL5-CXCR2 signaling during embryo culture improved pregnancy success in aging mice. Future analysis on CXCL5-CXCR2 signaling suppression in human embryos could be the basis to improve embryo development and pregnancy outcome in middle-aged infertile patients.
Collapse
Affiliation(s)
- Yuta Kawagoe
- Department of Obstetrics and Gynecology Advanced Reproduction Research Center International University of Health and Welfare School of Medicine Narita Japan
- The United Graduate School of Agriculture Sciences Iwate University Morioka Japan
| | - Ikko Kawashima
- Institute of Advanced BioMedical Engineering and Science Tokyo Women's Medical University Shinjuku‐ku Japan
| | - Yorino Sato
- Department of Obstetrics and Gynecology Advanced Reproduction Research Center International University of Health and Welfare School of Medicine Narita Japan
| | - Naoki Okamoto
- Department of Obstetrics and Gynecology Advanced Reproduction Research Center International University of Health and Welfare School of Medicine Narita Japan
| | - Kazuei Matsubara
- The United Graduate School of Agriculture Sciences Iwate University Morioka Japan
| | - Kazuhiro Kawamura
- Department of Obstetrics and Gynecology Advanced Reproduction Research Center International University of Health and Welfare School of Medicine Narita Japan
| |
Collapse
|
21
|
Chemokines and their receptors promoting the recruitment of myeloid-derived suppressor cells into the tumor. Mol Immunol 2019; 117:201-215. [PMID: 31835202 DOI: 10.1016/j.molimm.2019.11.014] [Citation(s) in RCA: 124] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 11/27/2019] [Accepted: 11/30/2019] [Indexed: 02/07/2023]
Abstract
Myeloid-derived suppressor cells (MDSCs) expand in tumor-bearing host. They suppress anti-tumor immune response and promote tumor growth. Chemokines play a vital role in recruiting MDSCs into tumor tissue. They can also induce the generation of MDSCs in the bone marrow, maintain their suppressive activity, and promote their proliferation and differentiation. Here, we review CCL2/CCL12-CCR2, CCL3/4/5-CCR5, CCL15-CCR1, CX3CL1/CCL26-CX3CR1, CXCL5/2/1-CXCR2, CXCL8-CXCR1/2, CCL21-CCR7, CXCL13-CXCR5 signaling pathways, their role in MDSCs recruitment to tumor tissue, and their correlation with tumor development, metastasis and prognosis. Targeting chemokines and their receptors may serve as a promising strategy in immunotherapy, especially combined with other strategies such as chemotherapy, cyclin-dependent kinase or immune checkpoints inhibitors.
Collapse
|
22
|
Lim WK, Chai X, Ghosh S, Ray D, Wang M, Rasheed SAK, Casey PJ. Gα-13 induces C XC motif chemokine ligand 5 expression in prostate cancer cells by transactivating NF-κB. J Biol Chem 2019; 294:18192-18206. [PMID: 31636124 PMCID: PMC6885619 DOI: 10.1074/jbc.ra119.010018] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 10/03/2019] [Indexed: 12/24/2022] Open
Abstract
GNA13, the α subunit of a heterotrimeric G protein, mediates signaling through G-protein-coupled receptors (GPCRs). GNA13 is up-regulated in many solid tumors, including prostate cancer, where it contributes to tumor initiation, drug resistance, and metastasis. To better understand how GNA13 contributes to tumorigenesis and tumor progression, we compared the entire transcriptome of PC3 prostate cancer cells with those cells in which GNA13 expression had been silenced. This analysis revealed that GNA13 levels affected multiple CXC-family chemokines. Further investigation in three different prostate cancer cell lines singled out pro-tumorigenic CXC motif chemokine ligand 5 (CXCL5) as a target of GNA13 signaling. Elevation of GNA13 levels consistently induced CXCL5 RNA and protein expression in all three cell lines. Analysis of the CXCL5 promoter revealed that the -505/+62 region was both highly active and influenced by GNA13, and a single NF-κB site within this region of the promoter was critical for GNA13-dependent promoter activity. ChIP experiments revealed that, upon induction of GNA13 expression, occupancy at the CXCL5 promoter was significantly enriched for the p65 component of NF-κB. GNA13 knockdown suppressed both p65 phosphorylation and the activity of a specific NF-κB reporter, and p65 silencing impaired the GNA13-enhanced expression of CXCL5. Finally, blockade of Rho GTPase activity eliminated the impact of GNA13 on NF-κB transcriptional activity and CXCL5 expression. Together, these findings suggest that GNA13 drives CXCL5 expression by transactivating NF-κB in a Rho-dependent manner in prostate cancer cells.
Collapse
Affiliation(s)
- Wei Kiang Lim
- Programme in Cancer and Stem Cell Biology, Duke-NUS Medical School, 169857 Singapore
| | - Xiaoran Chai
- Programme in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, 169857 Singapore
| | - Sujoy Ghosh
- Programme in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, 169857 Singapore
| | - Debleena Ray
- Programme in Cancer and Stem Cell Biology, Duke-NUS Medical School, 169857 Singapore
| | - Mei Wang
- Programme in Cancer and Stem Cell Biology, Duke-NUS Medical School, 169857 Singapore
| | | | - Patrick J Casey
- Programme in Cancer and Stem Cell Biology, Duke-NUS Medical School, 169857 Singapore; Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina 27710.
| |
Collapse
|
23
|
Li TT, Liu MR, Pei DS. Friend or foe, the role of EGR-1 in cancer. Med Oncol 2019; 37:7. [PMID: 31748910 DOI: 10.1007/s12032-019-1333-6] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Accepted: 11/11/2019] [Indexed: 12/18/2022]
Abstract
Early growth response-1 (EGR-1), also termed NEFI-A and Krox-24, as a multi-domain protein is implicated in several vital physiological processes, including development, metabolism, cell growth and proliferation. Previous studies have implied that EGR-1 was producing in response to the tissue injury, immune response and fibrosis. Meanwhile, emerging studies stressed the pronounced correlation of EGR-1 and human cancers. Nevertheless, the intricate mechanisms of cancer-reduce EGR-1 alteration still poorly characterized. In the review, we evaluated the effects of EGR-1 in tumor cell proliferation, apoptosis, migration, invasion and tumor microenvironment, and then, we dwell on the intricate signaling pathways that EGR-1 involved in. The aberrantly expressed of EGR-1 in cancers are expected to provide a new cancer therapy strategy or a new marker for assessing treatment efficacy.
Collapse
Affiliation(s)
- Tong-Tong Li
- Department of Pathology, Xuzhou Medical University, 209 Tong-shan Road, Xuzhou, 221004, Jiangsu, People's Republic of China
| | - Man-Ru Liu
- Department of Pathology, Xuzhou Medical University, 209 Tong-shan Road, Xuzhou, 221004, Jiangsu, People's Republic of China
| | - Dong-Sheng Pei
- Department of Pathology, Xuzhou Medical University, 209 Tong-shan Road, Xuzhou, 221004, Jiangsu, People's Republic of China.
| |
Collapse
|
24
|
EGR1 regulates angiogenic and osteoclastogenic factors in prostate cancer and promotes metastasis. Oncogene 2019; 38:6241-6255. [PMID: 31312026 PMCID: PMC6715537 DOI: 10.1038/s41388-019-0873-8] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 03/18/2019] [Accepted: 04/07/2019] [Indexed: 12/21/2022]
Abstract
Early growth response-1 (EGR1) is a transcription factor correlated with prostate cancer (PC) progression in a variety of contexts. For example, EGR1 levels increase in response to suppressed androgen receptor signaling or loss of the tumor suppressor, PTEN. EGR1 has been shown to regulate genes influencing proliferation, apoptosis, immune cell activation, and matrix degradation, among others. Despite this, the impact of EGR1 on PC metastatic colonization is unclear. We demonstrate using a PC model (DU145/RasB1) of bone and brain metastasis that EGR1 expression regulates angiogenic and osteoclastogenic properties of metastases. We have shown previously that FN14 (TNFRSF12A) and downstream NF-κB signaling is required for metastasis in this model. Here we demonstrate that FN14 ligation also leads to NF-κB-independent, MEK-dependent EGR1 expression. EGR1-depletion in DU145/RasB1 cells reduced both the number and size of metastases but did not affect primary tumor growth. Decreased EGR1 expression led to reduced blood vessel density in brain and bone metastases as well as decreased osteolytic bone lesion area and reduced numbers of osteoclasts at the bone-tumor interface. TWEAK (TNFSF12) induced several EGR1-dependent angiogenic and osteoclastogenic factors (e.g. PDGFA, TGFB1, SPP1, IL6, IL8, and TGFA, among others). Consistent with this, in clinical samples of PC, the level of several genes encoding angiogenic/osteoclastogenic pathway effectors correlated with EGR1 levels. Thus, we show here that EGR1 has a direct effect on prostate cancer metastases. EGR1 regulates angiogenic and osteoclastogenic factors, informing the underlying signaling networks that impact autonomous and microenvironmental mechanisms of cancer metastases.
Collapse
|
25
|
Xiang QF, Zhan MX, Li Y, Liang H, Hu C, Huang YM, Xiao J, He X, Xin YJ, Chen MS, Lu LG. Activation of MET promotes resistance to sorafenib in hepatocellular carcinoma cells via the AKT/ERK1/2-EGR1 pathway. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2019; 47:83-89. [PMID: 30663411 DOI: 10.1080/21691401.2018.1543195] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Sorafenib is an oral multikinase inhibitor that has become an established therapeutic approach in advanced hepatocellular carcinoma (HCC). However, the benefit of sorafenib in clinical therapy is often affected by drug resistance. Therefore, it is important to explore the mechanisms underlying sorafenib resistance and to develop individualized therapeutic strategies for coping with this problem. In this study, we found that addition of HGF to sorafenib-treated HCC cells activated MET and re-stimulated the downstream AKT and ERK1/2 pathways. Thereby, restored sorafenib-treated HCC cells proliferation, migration and invasion ability, and rescued cells from apoptosis. In addition, we found that HGF treatment of HCC cells induced early growth response protein (EGR1) expression, which is involved in sorafenib resistance. Importantly, the HGF rescued effect in sorafenib-treated HCC cells could be abrogated by inhibiting MET activation with PHA-665752 or by downregulating EGR1 expression with small interfering RNA (siRNA). Therefore, inhibition of the HGF/MET pathway may improve response to sorafenib in HCC, and combination therapy should be further investigated.
Collapse
Affiliation(s)
- Qing-Feng Xiang
- a Department of Hepatobiliary Surgery , Sun Yat-Sen University Cancer Center , Guangzhou , China.,b Department of General Surgery , Zhuhai People's Hospital, Zhuhai Hospital of Jinan University , Zhuhai , China
| | - Mei-Xiao Zhan
- c Zhuhai Interventional Medical Center, Zhuhai Precision Medicine Center , Zhuhai People's Hospital , Zhuhai Hospital of Jinan University , Zhuhai , China
| | - Yong Li
- c Zhuhai Interventional Medical Center, Zhuhai Precision Medicine Center , Zhuhai People's Hospital , Zhuhai Hospital of Jinan University , Zhuhai , China
| | - Hui Liang
- b Department of General Surgery , Zhuhai People's Hospital, Zhuhai Hospital of Jinan University , Zhuhai , China
| | - Cong Hu
- b Department of General Surgery , Zhuhai People's Hospital, Zhuhai Hospital of Jinan University , Zhuhai , China
| | - Yao-Ming Huang
- b Department of General Surgery , Zhuhai People's Hospital, Zhuhai Hospital of Jinan University , Zhuhai , China
| | - Jing Xiao
- c Zhuhai Interventional Medical Center, Zhuhai Precision Medicine Center , Zhuhai People's Hospital , Zhuhai Hospital of Jinan University , Zhuhai , China
| | - Xu He
- c Zhuhai Interventional Medical Center, Zhuhai Precision Medicine Center , Zhuhai People's Hospital , Zhuhai Hospital of Jinan University , Zhuhai , China
| | - Yong-Jie Xin
- c Zhuhai Interventional Medical Center, Zhuhai Precision Medicine Center , Zhuhai People's Hospital , Zhuhai Hospital of Jinan University , Zhuhai , China
| | - Min-Shan Chen
- a Department of Hepatobiliary Surgery , Sun Yat-Sen University Cancer Center , Guangzhou , China
| | - Li-Gong Lu
- c Zhuhai Interventional Medical Center, Zhuhai Precision Medicine Center , Zhuhai People's Hospital , Zhuhai Hospital of Jinan University , Zhuhai , China
| |
Collapse
|
26
|
Cheng Y, Ma XL, Wei YQ, Wei XW. Potential roles and targeted therapy of the CXCLs/CXCR2 axis in cancer and inflammatory diseases. Biochim Biophys Acta Rev Cancer 2019; 1871:289-312. [DOI: 10.1016/j.bbcan.2019.01.005] [Citation(s) in RCA: 127] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2018] [Revised: 11/19/2018] [Accepted: 01/09/2019] [Indexed: 12/16/2022]
|
27
|
Cui D, Zhao Y, Xu J. Activation of CXCL5-CXCR2 axis promotes proliferation and accelerates G1 to S phase transition of papillary thyroid carcinoma cells and activates JNK and p38 pathways. Cancer Biol Ther 2018; 20:608-616. [PMID: 30404567 DOI: 10.1080/15384047.2018.1539289] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
C-X-C motif chemokine ligand 5 (CXCL5) is initially identified to recruit neutrophils by interacting with its receptor, C-X-C motif chemokine receptor 2 (CXCR2). Our prior work demonstrated that the expression levels of CXCL5 and CXCR2 were higher in the papillary thyroid carcinoma (PTC) tumors than that in the non-tumors. This study was performed to further investigate how this axis regulates the growth of PTC cells. B-CPAP cells (BRAFV600E) and TPC-1 cells (RET/PTC rearrangement) expressing CXCR-2 were used as in vitro cell models. Our results showed that the recombinant human CXCL5 (rhCXCL5) promoted the proliferation of PTC cells. rhCXCL5 accelerated the G1/S transition, upregulated the expression of a group of S (DNA synthesis) or M (mitosis)-promoting cyclins and cyclin-dependent kinases (CDKs), and downregulated CDK inhibitors in PTC cells. The CDS region of homo sapiens CXCL5 gene was inserted into an eukaryotic expression vector to mediate the overexpression of CXCL5 in PTC cells. The phosphorylation of c-Jun N-terminal kinases (JNK) and p38, and the nuclear translocation of c-Jun were enhanced by CXCL5 overexpression, whereas attenuated by CXCR2 antagonist SB225002. Additionally, CXCL5/CXCR2 axis, JNK and p38 pathway inhibitors, SB225002, SP600125 and SB203580, suppressed the growth of PTC cells overexpressing CXCL5 in nude mice, respectively. Collectively, our study demonstrates a growth-promoting effect of CXCL5-CXCR2 axis in PTC cells in vitro and in vivo.
Collapse
Affiliation(s)
- Dong Cui
- a Department of Thyroid Surgery , The Second Affiliated Hospital of Dalian Medical University , Dalian , People's Republic of China
| | - Yongfu Zhao
- a Department of Thyroid Surgery , The Second Affiliated Hospital of Dalian Medical University , Dalian , People's Republic of China
| | - Jingchao Xu
- a Department of Thyroid Surgery , The Second Affiliated Hospital of Dalian Medical University , Dalian , People's Republic of China
| |
Collapse
|
28
|
Hu B, Fan H, Lv X, Chen S, Shao Z. Prognostic significance of CXCL5 expression in cancer patients: a meta-analysis. Cancer Cell Int 2018; 18:68. [PMID: 29743818 PMCID: PMC5930840 DOI: 10.1186/s12935-018-0562-7] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 04/21/2018] [Indexed: 12/21/2022] Open
Abstract
Background CXCL5 is a member of the CXC-type chemokine family, which has been found to play important roles in tumorigenesis and cancer progression. Recent studies have demonstrated that CXCL5 could serve as a potential prognostic biomarker for cancer patients. However, the prognostic value of CXCL5 is still controversial. Methods We systematically searched PubMed, Embase and Web of Science to obtain all relevant articles investigating the prognostic significance of CXCL5 expression in cancer patients. Hazards ratios (HR) with corresponding 95% confidence intervals (CI) were pooled to estimate the association between CXCL5 expression levels with survival of cancer patients. Results A total of 15 eligible studies including 19 cohorts and 5070 patients were enrolled in the current meta-analysis. Our results demonstrated that elevated expression level of CXCL5 was significantly associated with poor overall survival (OS) (pooled HR 1.70; 95% CI 1.36–2.12), progression-free survival (pooled HR 1.65; 95% CI 1.09–2.49) and recurrence-free survival (pooled HR 1.49; 95% CI 1.15–1.93) in cancer patients. However, high or low expression of CXCL5 made no difference in predicting the disease-free survival (pooled HR 0.63; 95% CI 0.11–3.49) of cancer patients. Furthermore, we found that high CXCL5 expression was associated with reduced OS in intrahepatic cholangiocarcinoma (HR 1.91; 95% CI 1.31–2.78) and hepatocellular carcinoma (HR 1.87; 95% CI 1.55–2.27). However, there was no significant association between expression level of CXCL5 with the OS in lung cancer (HR 1.25; 95% CI 0.79–1.99) and colorectal cancer (HR 1.16; 95% CI 0.32–4.22, p = 0.826) in current meta-analysis. Conclusions In conclusion, our meta-analysis suggested that elevated CXCL5 expression might be an adverse prognostic marker for cancer patients, which could help the clinical decision making process.
Collapse
Affiliation(s)
- Binwu Hu
- 1Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 China
| | - Huiqian Fan
- 2Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 China
| | - Xiao Lv
- 1Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 China
| | - Songfeng Chen
- 3Department of Orthopaedic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zengwu Shao
- 1Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 China
| |
Collapse
|
29
|
Qiu WZ, Zhang HB, Xia WX, Ke LR, Yang J, Yu YH, Liang H, Huang XJ, Liu GY, Li WZ, Xiang YQ, Kang TB, Guo X, Lv X. The CXCL5/CXCR2 axis contributes to the epithelial-mesenchymal transition of nasopharyngeal carcinoma cells by activating ERK/GSK-3β/snail signalling. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2018; 37:85. [PMID: 29665837 PMCID: PMC5905166 DOI: 10.1186/s13046-018-0722-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Accepted: 02/26/2018] [Indexed: 12/22/2022]
Abstract
Background Distant metastasis is the major cause of treatment failure in patients with nasopharyngeal carcinoma (NPC). Although several biomarkers correlate with metastasis and prognosis, the molecular mechanisms of NPC development and progression remain unclear. Methods Quantitative RT-PCR (qRT-PCR), western blotting, cell growth, foci formation, migration and invasion assays, and xenograft mouse models were utilized to examine the expression levels and functions of the CXCL5/CXCR2 axis in NPC. A luciferase reporter assay, western blotting, immunofluorescence, and migration and invasion assays were used to identify and verify the ERK/GSK-3β/Snail signalling pathway. Results CXCL5 was significantly increased in the sera of NPC patients, and high expression levels of CXCL5/CXCR2 in NPC primary tissues indicated poor survival. CXCL5 and CXCR2 were upregulated in NPC cell lines. Ectopic expression of the CXCL5/CXCR2 axis promoted NPC cell migration and invasion in vitro and the formation of lung metastases in vivo. Mechanistically, the dual overexpression of CXCL5 and CXCR2 promoted cell spreading by inducing the epithelial-mesenchymal transition (EMT) through the activation of the ERK/GSK-3β/Snail signalling pathway. Conclusion The CXCL5/CXCR2 axis contributes to the EMT of NPC cells by activating ERK/GSK-3β/Snail signalling, and this axis may be a potential diagnostic marker and therapeutic target for patients with NPC. Electronic supplementary material The online version of this article (10.1186/s13046-018-0722-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Wen-Ze Qiu
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, People's Republic of China.,Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, People's Republic of China
| | - Hai-Bo Zhang
- Department of Radiation Oncology, Shandong Cancer Hospital Affiliated to Shandong University, Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Wei-Xiong Xia
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, People's Republic of China.,Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, People's Republic of China
| | - Liang-Ru Ke
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, People's Republic of China.,Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, People's Republic of China
| | - Jing Yang
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, 4365 Kangxin Road, Shanghai, 201321, People's Republic of China
| | - Ya-Hui Yu
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, People's Republic of China.,Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, People's Republic of China
| | - Hu Liang
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, People's Republic of China.,Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, People's Republic of China
| | - Xin-Jun Huang
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, People's Republic of China.,Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, People's Republic of China
| | - Guo-Ying Liu
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, People's Republic of China.,Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, People's Republic of China
| | - Wang-Zhong Li
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, People's Republic of China.,Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, People's Republic of China
| | - Yan-Qun Xiang
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, People's Republic of China.,Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, People's Republic of China
| | - Tie-Bang Kang
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, People's Republic of China.
| | - Xiang Guo
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, People's Republic of China. .,Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, People's Republic of China.
| | - Xing Lv
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, People's Republic of China. .,Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, People's Republic of China.
| |
Collapse
|
30
|
Yu D, Geng H, Liu Z, Zhao L, Liang Z, Zhang Z, Xie D, Wang Y, Zhang T, Min J, Zhong C. Cigarette smoke induced urocystic epithelial mesenchymal transition via MAPK pathways. Oncotarget 2018; 8:8791-8800. [PMID: 28060741 PMCID: PMC5352442 DOI: 10.18632/oncotarget.14456] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2016] [Accepted: 12/06/2016] [Indexed: 02/07/2023] Open
Abstract
Cigarette smoke has been shown to be a major risk factor for bladder cancer. Epithelial-mesenchymal transition (EMT) is a crucial process in cancer development. The role of MAPK pathways in regulating cigarette smoke-triggered urocystic EMT remains to be elucidated. Human normal urothelial cells and BALB/c mice were used as in vitro and in vivo cigarette smoke exposure models. Exposure of human normal urothelial cells to cigarette smoke induced morphological change, enhanced migratory and invasive capacities, reduced epithelial marker expression and increased mesenchymal marker expression, along with the activation of MAPK pathways. Moreover, we revealed that ERK1/2 and p38 inhibitors, but rather JNK inhibitor, effectively attenuated cigarette smoke-induced urocystic EMT. Importantly, the regulatory function of ERK1/2 and p38 pathways in cigarette smoke-triggered urocystic EMT was further confirmed in mice exposed to CS for 12 weeks. These findings could provide new insight into the molecular mechanisms of cigarette smoke-associated bladder cancer development as well as its potential intervention.
Collapse
Affiliation(s)
- Dexin Yu
- Department of Urology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230032, China
| | - Hao Geng
- Department of Urology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230032, China
| | - Zhiqi Liu
- Department of Urology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230032, China
| | - Li Zhao
- Department of Urology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230032, China
| | - Zhaofeng Liang
- Department of Preventive Medicine and Public Health Laboratory Sciences, School of Medicine, Jiangsu University, Jiangsu 212013, China
| | - Zhiqiang Zhang
- Department of Urology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230032, China
| | - Dongdong Xie
- Department of Urology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230032, China
| | - Yi Wang
- Department of Urology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230032, China
| | - Tao Zhang
- Department of Urology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230032, China
| | - Jie Min
- Department of Urology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230032, China
| | - Caiyun Zhong
- Department of Toxicology and Nutritional Science, School of Public Health, Nanjing Medical University, Nanjing 210029, China
| |
Collapse
|
31
|
Wu K, Yu S, Liu Q, Bai X, Zheng X, Wu K. The clinical significance of CXCL5 in non-small cell lung cancer. Onco Targets Ther 2017; 10:5561-5573. [PMID: 29200871 PMCID: PMC5702175 DOI: 10.2147/ott.s148772] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
As a CXC-type chemokine, ENA78/CXCL5 is an important attractant for granulocytes by binding to its receptor CXCR2. Recent studies proved that CXCL5/CXCR2 axis plays an oncogenic role in many human cancers. However, the exact clinical significance of CXCL5 in lung cancer has not been well defined. Here, we found that the serum protein expression of CXCL5 was significantly increased in non-small cell lung cancer (NSCLC) compared with that in healthy volunteers. Immunohistochemistry staining revealed that CXCL5 protein was higher in various lung cancer tissues compared with normal tissues. Moreover, CXCL5 expression correlated with histological grade, tumor size, and TNM stage in NSCLC. Elevated CXCL5 protein abundance predicted poor overall survival in adenocarcinoma patients. Further meta-analysis demonstrated that CXCL5 mRNA expression was also positively associated with tumor stage, lymph node metastasis, and worse survival. Kaplan–Meier plot analyses indicated high CXCL5 was associated with short overall survival and progression-free survival. Together, these results indicated that CXCL5 may be a potential biomarker for NSCLC.
Collapse
Affiliation(s)
- Kongju Wu
- Medical School of Pingdingshan University, Pingdingshan, Henan
| | - Shengnan Yu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Qian Liu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Xianguang Bai
- Medical School of Pingdingshan University, Pingdingshan, Henan
| | - Xinhua Zheng
- Medical School of Pingdingshan University, Pingdingshan, Henan
| | - Kongming Wu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| |
Collapse
|
32
|
Chuang MH, Jan MS, Chang JT, Lu FJ. The Chinese medicine JC-001 enhances the chemosensitivity of Lewis lung tumors to cisplatin by modulating the immune response. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2017; 17:210. [PMID: 28399860 PMCID: PMC5387375 DOI: 10.1186/s12906-017-1728-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/14/2017] [Accepted: 04/05/2017] [Indexed: 01/06/2023]
Abstract
BACKGROUND JC-001 is a Chinese medicine that can modulate the immunity in Hepa 1-6 tumor-bearing mice, and we questioned whether JC-001 can serve as efficient adjuvant chemotherapy. We aimed to identify a novel approach for enhancing cis-diamminedichloroplatinum (II) (CDDP)-based chemotherapy by immunomodulation. METHODS The anti-tumor activity in vitro was determined based on foci formation and a 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. A LLC1 tumor xenograft model was used to analyze the activity of tumor rejection in vivo. The tumors were analyzed through hematoxylin and eosin (H&E) staining, immunohistochemistry (IHC) staining and cytokine arrays. RESULTS JC-001 suppressed foci formation and reduced the viability of Lewis lung carcinoma (LLC1) cells in vitro. JC-001 suppressed LLC1 tumor growth in immunodeficient BALB/c nude mice and in immunocompetent C57BL/6 mice to an even greater extent. Furthermore, JC-001 up-regulated interferon-γ expression in the tumor microenvironment, enhanced the Th1 response in tumor-bearing mice, and increased the chemosensitivity of LLC1 tumors to CDDP chemotherapy. The results of our study suggest that JC-001 is associated with low cytotoxicity and can significantly suppress tumor growth by enhancing the Th1 response. CONCLUSION JC-001 is a Chinese medicine with potential clinical applications in CDDP-based chemotherapeutic regimens.
Collapse
|
33
|
Dang H, Wu W, Wang B, Cui C, Niu J, Chen J, Chen Z, Liu Y. CXCL5 Plays a Promoting Role in Osteosarcoma Cell Migration and Invasion in Autocrine- and Paracrine-Dependent Manners. Oncol Res 2017; 25:177-186. [PMID: 28277189 PMCID: PMC7840695 DOI: 10.3727/096504016x14732772150343] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
CXCL5, a CXC-type chemokine, is an important attractant for granulocytic immune cells by binding to its receptor CXCR2. Recently, CXCL5/CXCR2 has been found to play an oncogenic role in many human cancers. However, the exact role of CXCL5 in osteosarcoma cell migration and invasion has not been revealed. Here we found that the protein expression of CXCL5 was significantly increased in osteosarcoma tissues compared with that in matched adjacent nontumor tissues. Moreover, the expression of CXCL5 was significantly associated with advanced clinical stage and metastasis. Further investigation showed that the CXCL5 expression levels were also significantly increased in osteosarcoma cell lines, including Saos-2, MG63, U2OS, and SW1353, when compared with those in normal osteoblast hFoB1.19 cells. U2OS cells were further transfected with CXCL5-specific siRNA or overexpression plasmid. Knockdown of CXCL5 significantly suppressed U2OS cell migration and invasion. On the contrary, overexpression of CXLC5 remarkably promoted the migration and invasion of U2OS cells. Interestingly, both exogenous CXCL5 treatment and the conditioned medium of CXCL5-overexpressing hFoB1.19 cells could also enhance the migration and invasion of U2OS cells, suggesting that the promoting role of CXCL5 in U2OS cell migration and invasion is also in a paracrine-dependent manner. According to these data, our study demonstrates that CXCL5 is upregulated in osteosarcoma and may play an oncogenic role in osteosarcoma metastasis. Therefore, CXCL5 may become a potential therapeutic target for osteosarcoma treatment.
Collapse
Affiliation(s)
- Hongsheng Dang
- Department of Orthopaedics, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, P.R. China
| | | | | | | | | | | | | | | |
Collapse
|
34
|
RAS-Mitogen-Activated Protein Kinase Signal Is Required for Enhanced PD-L1 Expression in Human Lung Cancers. PLoS One 2016. [PMID: 27846317 DOI: 10.1371/journal.pone.0166626.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Ectopic programmed cell death ligand 1 (PD-L1) expression in non-small cell lung cancers (NSCLCs) is related to immune evasion by cancer, and it is a molecular target of immune checkpoint therapies. Although some altered signals in NSCLCs are responsible for ectopic PD-L1 expression, the precise mechanisms remain obscure. Because we found a higher frequency of EGFR/KRAS mutations in NSCLC cell lines with high PD-L1 expression (p < 0.001), we evaluated the relationships between downstream signals and PD-L1 expression, particularly in three KRAS-mutant adenocarcinoma cell lines. The MEK inhibitor U0126 (20 μM) significantly decreased the surface PD-L1 levels by 50-60% compared with dimethyl sulfoxide (p < 0.0001). Phorbol 12-myristate 13-acetate stimulation (100 nM, 15 min) increased (p < 0.05) and two ERK2 siRNAs as well as KRAS siRNAs decreased (p < 0.05) PD-L1 expression. The transcriptional activity of the potential AP-1 site (+4785 to +5056 from the transcription start site) in the PD-L1 gene was demonstrated by luciferase assays, which was inhibited by U0126. The chromatin immunoprecipitation assay demonstrated the binding of cJUN to the AP-1 site. Two STAT3 siRNAs decreased PD-L1 expression by 10-32% in two of the three KRAS-mutant lung adenocarcinoma cell lines (p < 0.05), while the PI3K inhibitor LY294002 (40 μM) did not change the expression level. Supervised cluster analysis and gene set enrichment analysis between the PD-L1-high and -low NSCLCs revealed a correlation between PD-L1 expression and genes/pathways related to cell motility/adhesion. These results indicate that MAPK signaling is the dominant downstream signal responsible for ectopic PD-L1 expression, in which STAT3 is also involved to some extent. Furthermore, MAPK signaling may control the expression of PD-L1 and several genes related to enhanced cell motility. Our findings suggest that MAPK, along with STAT3, is important for determining PD-L1 expression, which could be useful for targeted therapies against lung cancers.
Collapse
|
35
|
Sumimoto H, Takano A, Teramoto K, Daigo Y. RAS-Mitogen-Activated Protein Kinase Signal Is Required for Enhanced PD-L1 Expression in Human Lung Cancers. PLoS One 2016; 11:e0166626. [PMID: 27846317 PMCID: PMC5112979 DOI: 10.1371/journal.pone.0166626] [Citation(s) in RCA: 138] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 11/01/2016] [Indexed: 01/09/2023] Open
Abstract
Ectopic programmed cell death ligand 1 (PD-L1) expression in non-small cell lung cancers (NSCLCs) is related to immune evasion by cancer, and it is a molecular target of immune checkpoint therapies. Although some altered signals in NSCLCs are responsible for ectopic PD-L1 expression, the precise mechanisms remain obscure. Because we found a higher frequency of EGFR/KRAS mutations in NSCLC cell lines with high PD-L1 expression (p < 0.001), we evaluated the relationships between downstream signals and PD-L1 expression, particularly in three KRAS-mutant adenocarcinoma cell lines. The MEK inhibitor U0126 (20 μM) significantly decreased the surface PD-L1 levels by 50–60% compared with dimethyl sulfoxide (p < 0.0001). Phorbol 12-myristate 13-acetate stimulation (100 nM, 15 min) increased (p < 0.05) and two ERK2 siRNAs as well as KRAS siRNAs decreased (p < 0.05) PD-L1 expression. The transcriptional activity of the potential AP-1 site (+4785 to +5056 from the transcription start site) in the PD-L1 gene was demonstrated by luciferase assays, which was inhibited by U0126. The chromatin immunoprecipitation assay demonstrated the binding of cJUN to the AP-1 site. Two STAT3 siRNAs decreased PD-L1 expression by 10–32% in two of the three KRAS-mutant lung adenocarcinoma cell lines (p < 0.05), while the PI3K inhibitor LY294002 (40 μM) did not change the expression level. Supervised cluster analysis and gene set enrichment analysis between the PD-L1-high and -low NSCLCs revealed a correlation between PD-L1 expression and genes/pathways related to cell motility/adhesion. These results indicate that MAPK signaling is the dominant downstream signal responsible for ectopic PD-L1 expression, in which STAT3 is also involved to some extent. Furthermore, MAPK signaling may control the expression of PD-L1 and several genes related to enhanced cell motility. Our findings suggest that MAPK, along with STAT3, is important for determining PD-L1 expression, which could be useful for targeted therapies against lung cancers.
Collapse
Affiliation(s)
- Hidetoshi Sumimoto
- Department of Medical Oncology and Cancer Center, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Atsushi Takano
- Department of Medical Oncology and Cancer Center, Shiga University of Medical Science, Otsu, Shiga, Japan
- Center for Antibody and Vaccine Therapy, Research Hospital, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Koji Teramoto
- Department of Medical Oncology and Cancer Center, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Yataro Daigo
- Department of Medical Oncology and Cancer Center, Shiga University of Medical Science, Otsu, Shiga, Japan
- Center for Antibody and Vaccine Therapy, Research Hospital, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- * E-mail:
| |
Collapse
|
36
|
Bennett CG, Riemondy K, Chapnick DA, Bunker E, Liu X, Kuersten S, Yi R. Genome-wide analysis of Musashi-2 targets reveals novel functions in governing epithelial cell migration. Nucleic Acids Res 2016; 44:3788-800. [PMID: 27034466 PMCID: PMC4857000 DOI: 10.1093/nar/gkw207] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Accepted: 03/15/2016] [Indexed: 12/19/2022] Open
Abstract
The Musashi-2 (Msi2) RNA-binding protein maintains stem cell self-renewal and promotes oncogenesis by enhancing cell proliferation in hematopoietic and gastrointestinal tissues. However, it is unclear how Msi2 recognizes and regulates mRNA targets in vivo and whether Msi2 primarily controls cell growth in all cell types. Here we identified Msi2 targets with HITS-CLIP and revealed that Msi2 primarily recognizes mRNA 3′UTRs at sites enriched in multiple copies of UAG motifs in epithelial progenitor cells. RNA-seq and ribosome profiling demonstrated that Msi2 promotes targeted mRNA decay without affecting translation efficiency. Unexpectedly, the most prominent Msi2 targets identified are key regulators that govern cell motility with a high enrichment in focal adhesion and extracellular matrix-receptor interaction, in addition to regulators of cell growth and survival. Loss of Msi2 stimulates epithelial cell migration, increases the number of focal adhesions and also compromises cell growth. These findings provide new insights into the molecular mechanisms of Msi2's recognition and repression of targets and uncover a key function of Msi2 in restricting epithelial cell migration.
Collapse
Affiliation(s)
- Christopher G Bennett
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado, Boulder, CO 80309, USA
| | - Kent Riemondy
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado, Boulder, CO 80309, USA
| | - Douglas A Chapnick
- Department of Chemistry and Biochemistry, University of Colorado, Boulder, CO 80309, USA
| | - Eric Bunker
- Department of Chemistry and Biochemistry, University of Colorado, Boulder, CO 80309, USA
| | - Xuedong Liu
- Department of Chemistry and Biochemistry, University of Colorado, Boulder, CO 80309, USA
| | - Scott Kuersten
- Illumina Inc., 5602 Research Park Blvd. Suite 200, Madison, WI 53719, USA
| | - Rui Yi
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado, Boulder, CO 80309, USA
| |
Collapse
|
37
|
Choi SH, Ruggiero D, Sorice R, Song C, Nutile T, Vernon Smith A, Concas MP, Traglia M, Barbieri C, Ndiaye NC, Stathopoulou MG, Lagou V, Maestrale GB, Sala C, Debette S, Kovacs P, Lind L, Lamont J, Fitzgerald P, Tönjes A, Gudnason V, Toniolo D, Pirastu M, Bellenguez C, Vasan RS, Ingelsson E, Leutenegger AL, Johnson AD, DeStefano AL, Visvikis-Siest S, Seshadri S, Ciullo M. Six Novel Loci Associated with Circulating VEGF Levels Identified by a Meta-analysis of Genome-Wide Association Studies. PLoS Genet 2016; 12:e1005874. [PMID: 26910538 PMCID: PMC4766012 DOI: 10.1371/journal.pgen.1005874] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 01/26/2016] [Indexed: 12/31/2022] Open
Abstract
Vascular endothelial growth factor (VEGF) is an angiogenic and neurotrophic factor, secreted by endothelial cells, known to impact various physiological and disease processes from cancer to cardiovascular disease and to be pharmacologically modifiable. We sought to identify novel loci associated with circulating VEGF levels through a genome-wide association meta-analysis combining data from European-ancestry individuals and using a dense variant map from 1000 genomes imputation panel. Six discovery cohorts including 13,312 samples were analyzed, followed by in-silico and de-novo replication studies including an additional 2,800 individuals. A total of 10 genome-wide significant variants were identified at 7 loci. Four were novel loci (5q14.3, 10q21.3, 16q24.2 and 18q22.3) and the leading variants at these loci were rs114694170 (MEF2C, P = 6.79x10-13), rs74506613 (JMJD1C, P = 1.17x10-19), rs4782371 (ZFPM1, P = 1.59x10-9) and rs2639990 (ZADH2, P = 1.72x10-8), respectively. We also identified two new independent variants (rs34528081, VEGFA, P = 1.52x10-18; rs7043199, VLDLR-AS1, P = 5.12x10-14) at the 3 previously identified loci and strengthened the evidence for the four previously identified SNPs (rs6921438, LOC100132354, P = 7.39x10-1467; rs1740073, C6orf223, P = 2.34x10-17; rs6993770, ZFPM2, P = 2.44x10-60; rs2375981, KCNV2, P = 1.48x10-100). These variants collectively explained up to 52% of the VEGF phenotypic variance. We explored biological links between genes in the associated loci using Ingenuity Pathway Analysis that emphasized their roles in embryonic development and function. Gene set enrichment analysis identified the ERK5 pathway as enriched in genes containing VEGF associated variants. eQTL analysis showed, in three of the identified regions, variants acting as both cis and trans eQTLs for multiple genes. Most of these genes, as well as some of those in the associated loci, were involved in platelet biogenesis and functionality, suggesting the importance of this process in regulation of VEGF levels. This work also provided new insights into the involvement of genes implicated in various angiogenesis related pathologies in determining circulating VEGF levels. The understanding of the molecular mechanisms by which the identified genes affect circulating VEGF levels could be important in the development of novel VEGF-related therapies for such diseases. Vascular Endothelial Growth Factor (VEGF) is a protein with a fundamental role in development of vascular system. The protein, produced by many types of cells, is released in the blood. High levels of VEGF have been observed in different pathological conditions especially in cancer, cardiovascular, and inflammatory diseases. Therefore, identifying the genetic factors influencing VEGF levels is important for predicting and treating such pathologies. The number of genetic variants associated with VEGF levels has been limited. To identify new loci, we have performed a Genome Wide Association Study meta-analysis on a sample of more than 16,000 individuals from 10 cohorts, using a high-density genetic map. This analysis revealed 10 variants associated with VEGF circulating levels, 6 of these being novel associations. The 10 variants cumulatively explain more than 50% of the variability of VEGF serum levels. Our analyses have identified genes known to be involved in angiogenesis related diseases and genes implicated in platelet metabolism, suggesting the importance of links between this process and VEGF regulation. Overall, these data have improved our understanding of the genetic variation underlying circulating VEGF levels. This in turn could guide our response to the challenge posed by various VEGF-related pathologies.
Collapse
Affiliation(s)
- Seung Hoan Choi
- Department of Neurology, Boston University School of Medicine, Boston, Massachusetts, United States of America
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts, United States of America
- National Heart, Lung and Blood Institute’s Framingham Heart Study, Framingham, Massachusetts, United States of America
| | - Daniela Ruggiero
- Institute of Genetics and Biophysics, National Research Council of Italy, Naples, Italy
| | - Rossella Sorice
- Institute of Genetics and Biophysics, National Research Council of Italy, Naples, Italy
| | - Ci Song
- Population Sciences Branch, National Heart, Lung and Blood Institute’s Framingham Heart Study, Framingham, Massachusetts, United States of America
- Department of Medical Sciences, Molecular Epidemiology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Teresa Nutile
- Institute of Genetics and Biophysics, National Research Council of Italy, Naples, Italy
| | - Albert Vernon Smith
- Icelandic Heart Association, Kopavogur, Iceland
- University of Iceland, Reykjavik, Iceland
| | - Maria Pina Concas
- Institute of Population Genetics, National Research Council of Italy, Sassari, Italy
| | - Michela Traglia
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milano, Italy
| | - Caterina Barbieri
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milano, Italy
| | - Ndeye Coumba Ndiaye
- UMR INSERM U1122, IGE-PCV “Interactions Gène-Environnement en Physiopathologie Cardio-Vasculaire”, Faculté de Pharmacie, Université de Lorraine, Nancy, France
| | - Maria G. Stathopoulou
- UMR INSERM U1122, IGE-PCV “Interactions Gène-Environnement en Physiopathologie Cardio-Vasculaire”, Faculté de Pharmacie, Université de Lorraine, Nancy, France
| | - Vasiliki Lagou
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | | | - Cinzia Sala
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milano, Italy
| | - Stephanie Debette
- Department of Neurology, Boston University School of Medicine, Boston, Massachusetts, United States of America
- Department of Neurology, Bordeaux University Hospital, Bordeaux, France
- INSERM U897, Bordeaux, France
| | - Peter Kovacs
- University of Leipzig, IFB Adiposity Diseases, Leipzig, Germany
| | - Lars Lind
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - John Lamont
- Randox Laboratories, Crumlin, United Kingdom
| | | | - Anke Tönjes
- University of Leipzig, Department of Medicine, Leipzig, Germany
| | - Vilmundur Gudnason
- Icelandic Heart Association, Kopavogur, Iceland
- University of Iceland, Reykjavik, Iceland
| | - Daniela Toniolo
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milano, Italy
| | - Mario Pirastu
- Institute of Population Genetics, National Research Council of Italy, Sassari, Italy
| | - Celine Bellenguez
- Institut Pasteur de Lille, Lille, France
- INSEM U744, Lille, France
- Université Lille-Nord de France, Lille, France
| | - Ramachandran S. Vasan
- National Heart, Lung and Blood Institute’s Framingham Heart Study, Framingham, Massachusetts, United States of America
- Section of Preventive Medicine and Epidemiology, Department of Medicine, Boston University Schools of Medicine and Public Health, Boston, Massachusetts, United States of America
| | - Erik Ingelsson
- Department of Medical Sciences, Molecular Epidemiology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Anne-Louise Leutenegger
- INSERM U946, Paris, France
- Université Paris Diderot, Sorbonne Paris Cité, IUH, UMR-S 946, Paris, France
| | - Andrew D. Johnson
- Population Sciences Branch, National Heart, Lung and Blood Institute’s Framingham Heart Study, Framingham, Massachusetts, United States of America
| | - Anita L. DeStefano
- Department of Neurology, Boston University School of Medicine, Boston, Massachusetts, United States of America
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts, United States of America
- National Heart, Lung and Blood Institute’s Framingham Heart Study, Framingham, Massachusetts, United States of America
| | - Sophie Visvikis-Siest
- UMR INSERM U1122, IGE-PCV “Interactions Gène-Environnement en Physiopathologie Cardio-Vasculaire”, Faculté de Pharmacie, Université de Lorraine, Nancy, France
| | - Sudha Seshadri
- Department of Neurology, Boston University School of Medicine, Boston, Massachusetts, United States of America
- National Heart, Lung and Blood Institute’s Framingham Heart Study, Framingham, Massachusetts, United States of America
- * E-mail: (SS); (MC)
| | - Marina Ciullo
- Institute of Genetics and Biophysics, National Research Council of Italy, Naples, Italy
- * E-mail: (SS); (MC)
| |
Collapse
|
38
|
Langie SAS, Koppen G, Desaulniers D, Al-Mulla F, Al-Temaimi R, Amedei A, Azqueta A, Bisson WH, Brown DG, Brunborg G, Charles AK, Chen T, Colacci A, Darroudi F, Forte S, Gonzalez L, Hamid RA, Knudsen LE, Leyns L, Lopez de Cerain Salsamendi A, Memeo L, Mondello C, Mothersill C, Olsen AK, Pavanello S, Raju J, Rojas E, Roy R, Ryan EP, Ostrosky-Wegman P, Salem HK, Scovassi AI, Singh N, Vaccari M, Van Schooten FJ, Valverde M, Woodrick J, Zhang L, van Larebeke N, Kirsch-Volders M, Collins AR. Causes of genome instability: the effect of low dose chemical exposures in modern society. Carcinogenesis 2015; 36 Suppl 1:S61-88. [PMID: 26106144 DOI: 10.1093/carcin/bgv031] [Citation(s) in RCA: 128] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Genome instability is a prerequisite for the development of cancer. It occurs when genome maintenance systems fail to safeguard the genome's integrity, whether as a consequence of inherited defects or induced via exposure to environmental agents (chemicals, biological agents and radiation). Thus, genome instability can be defined as an enhanced tendency for the genome to acquire mutations; ranging from changes to the nucleotide sequence to chromosomal gain, rearrangements or loss. This review raises the hypothesis that in addition to known human carcinogens, exposure to low dose of other chemicals present in our modern society could contribute to carcinogenesis by indirectly affecting genome stability. The selected chemicals with their mechanisms of action proposed to indirectly contribute to genome instability are: heavy metals (DNA repair, epigenetic modification, DNA damage signaling, telomere length), acrylamide (DNA repair, chromosome segregation), bisphenol A (epigenetic modification, DNA damage signaling, mitochondrial function, chromosome segregation), benomyl (chromosome segregation), quinones (epigenetic modification) and nano-sized particles (epigenetic pathways, mitochondrial function, chromosome segregation, telomere length). The purpose of this review is to describe the crucial aspects of genome instability, to outline the ways in which environmental chemicals can affect this cancer hallmark and to identify candidate chemicals for further study. The overall aim is to make scientists aware of the increasing need to unravel the underlying mechanisms via which chemicals at low doses can induce genome instability and thus promote carcinogenesis.
Collapse
Affiliation(s)
- Sabine A S Langie
- Environmental Risk and Health Unit, Flemish Institute for Technological Research (VITO), Boeretang 200, 2400 Mol, Belgium, Health Canada, Environmental Health Sciences and Research Bureau, Environmental Health Centre, Ottawa, Ontario K1A0K9, Canada, Department of Pathology, Kuwait University, Safat 13110, Kuwait, Department of Experimental and Clinical Medicine, University of Firenze, Florence 50134, Italy, Department of Pharmacology and Toxicology, Faculty of Pharmacy, University of Navarra, Pamplona 31009, Spain, Environmental and Molecular Toxicology, Environmental Health Sciences Center, Oregon State University, Corvallis, OR 97331, USA, Department of Environmental and Radiological Health Sciences/Food Science and Human Nutrition, College of Veterinary Medicine and Biomedical Sciences, Colorado State University/Colorado School of Public Health, Fort Collins, CO 80523-1680, USA, Department of Chemicals and Radiation, Division of Environmental Medicine, Norwegian Institute of Public Health, PO Box 4404, N-0403 Oslo, Norway, Hopkins Building, School of Biological Sciences, University of Reading, Reading, Berkshire RG6 6UB, UK, Division of Genetic and Molecular Toxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR 72079, USA, Center for Environmental Carcinogenesis and Risk Assessment, Environmental Protection and Health Prevention Agency, Bologna 40126, Italy, Human and Environmental Safety Research, Department of Health Sciences, College of North Atlantic, Doha, State of Qatar, Mediterranean Institute of Oncology, 95029 Viagrande, Italy, Laboratory for Cell Genetics, Vrije Universiteit Brussel, Brussels 1050, Belgium, Department of Biomedical Science, Faculty of Medicine and Health Sciences, University Putra, Serdang 43400, Selangor, Malaysia, University of Copenhagen, Department of Public Health, Copenhagen 1353, Denmark, Institute of Molecular Genetics, National Research Council, Pavia 27100, Italy, Medical Phys
| | - Gudrun Koppen
- Environmental Risk and Health Unit, Flemish Institute for Technological Research (VITO), Boeretang 200, 2400 Mol, Belgium, Health Canada, Environmental Health Sciences and Research Bureau, Environmental Health Centre, Ottawa, Ontario K1A0K9, Canada, Department of Pathology, Kuwait University, Safat 13110, Kuwait, Department of Experimental and Clinical Medicine, University of Firenze, Florence 50134, Italy, Department of Pharmacology and Toxicology, Faculty of Pharmacy, University of Navarra, Pamplona 31009, Spain, Environmental and Molecular Toxicology, Environmental Health Sciences Center, Oregon State University, Corvallis, OR 97331, USA, Department of Environmental and Radiological Health Sciences/Food Science and Human Nutrition, College of Veterinary Medicine and Biomedical Sciences, Colorado State University/Colorado School of Public Health, Fort Collins, CO 80523-1680, USA, Department of Chemicals and Radiation, Division of Environmental Medicine, Norwegian Institute of Public Health, PO Box 4404, N-0403 Oslo, Norway, Hopkins Building, School of Biological Sciences, University of Reading, Reading, Berkshire RG6 6UB, UK, Division of Genetic and Molecular Toxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR 72079, USA, Center for Environmental Carcinogenesis and Risk Assessment, Environmental Protection and Health Prevention Agency, Bologna 40126, Italy, Human and Environmental Safety Research, Department of Health Sciences, College of North Atlantic, Doha, State of Qatar, Mediterranean Institute of Oncology, 95029 Viagrande, Italy, Laboratory for Cell Genetics, Vrije Universiteit Brussel, Brussels 1050, Belgium, Department of Biomedical Science, Faculty of Medicine and Health Sciences, University Putra, Serdang 43400, Selangor, Malaysia, University of Copenhagen, Department of Public Health, Copenhagen 1353, Denmark, Institute of Molecular Genetics, National Research Council, Pavia 27100, Italy, Medical Phys
| | - Daniel Desaulniers
- Health Canada, Environmental Health Sciences and Research Bureau, Environmental Health Centre, Ottawa, Ontario K1A0K9, Canada
| | - Fahd Al-Mulla
- Department of Pathology, Kuwait University, Safat 13110, Kuwait
| | | | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, University of Firenze, Florence 50134, Italy
| | - Amaya Azqueta
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, University of Navarra, Pamplona 31009, Spain
| | - William H Bisson
- Environmental and Molecular Toxicology, Environmental Health Sciences Center, Oregon State University, Corvallis, OR 97331, USA
| | - Dustin G Brown
- Department of Environmental and Radiological Health Sciences/Food Science and Human Nutrition, College of Veterinary Medicine and Biomedical Sciences, Colorado State University/Colorado School of Public Health, Fort Collins, CO 80523-1680, USA
| | - Gunnar Brunborg
- Department of Chemicals and Radiation, Division of Environmental Medicine, Norwegian Institute of Public Health, PO Box 4404, N-0403 Oslo, Norway
| | - Amelia K Charles
- Hopkins Building, School of Biological Sciences, University of Reading, Reading, Berkshire RG6 6UB, UK
| | - Tao Chen
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR 72079, USA
| | - Annamaria Colacci
- Center for Environmental Carcinogenesis and Risk Assessment, Environmental Protection and Health Prevention Agency, Bologna 40126, Italy
| | - Firouz Darroudi
- Human and Environmental Safety Research, Department of Health Sciences, College of North Atlantic, Doha, State of Qatar
| | - Stefano Forte
- Mediterranean Institute of Oncology, 95029 Viagrande, Italy
| | - Laetitia Gonzalez
- Laboratory for Cell Genetics, Vrije Universiteit Brussel, Brussels 1050, Belgium
| | - Roslida A Hamid
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, University Putra, Serdang 43400, Selangor, Malaysia
| | - Lisbeth E Knudsen
- University of Copenhagen, Department of Public Health, Copenhagen 1353, Denmark
| | - Luc Leyns
- Laboratory for Cell Genetics, Vrije Universiteit Brussel, Brussels 1050, Belgium
| | | | - Lorenzo Memeo
- Mediterranean Institute of Oncology, 95029 Viagrande, Italy
| | - Chiara Mondello
- Institute of Molecular Genetics, National Research Council, Pavia 27100, Italy
| | - Carmel Mothersill
- Medical Physics & Applied Radiation Sciences, McMaster University, Hamilton, Ontario L8S4L8, Canada
| | - Ann-Karin Olsen
- Department of Chemicals and Radiation, Division of Environmental Medicine, Norwegian Institute of Public Health, PO Box 4404, N-0403 Oslo, Norway
| | - Sofia Pavanello
- Department of Cardiac, Thoracic and Vascular Sciences, Unit of Occupational Medicine, University of Padova, Padova 35128, Italy
| | - Jayadev Raju
- Toxicology Research Division, Bureau of Chemical Safety Food Directorate, Health Products and Food Branch Health Canada, Ottawa, Ontario K1A0K9, Canada
| | - Emilio Rojas
- Departamento de Medicina Genomica y Toxicologia Ambiental, Instituto de Investigaciones Biomedicas, Universidad Nacional Autonoma de México, México CP 04510, México
| | - Rabindra Roy
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Elizabeth P Ryan
- Department of Environmental and Radiological Health Sciences/Food Science and Human Nutrition, College of Veterinary Medicine and Biomedical Sciences, Colorado State University/Colorado School of Public Health, Fort Collins, CO 80523-1680, USA
| | - Patricia Ostrosky-Wegman
- Departamento de Medicina Genomica y Toxicologia Ambiental, Instituto de Investigaciones Biomedicas, Universidad Nacional Autonoma de México, México CP 04510, México
| | - Hosni K Salem
- Urology Department, kasr Al-Ainy School of Medicine, Cairo University, El Manial, Cairo 12515, Egypt
| | - A Ivana Scovassi
- Institute of Molecular Genetics, National Research Council, Pavia 27100, Italy
| | - Neetu Singh
- Centre for Advanced Research, King George's Medical University, Chowk, Lucknow 226003, Uttar Pradesh, India
| | - Monica Vaccari
- Center for Environmental Carcinogenesis and Risk Assessment, Environmental Protection and Health Prevention Agency, Bologna 40126, Italy
| | - Frederik J Van Schooten
- Department of Toxicology, NUTRIM School for Nutrition, Toxicology and Metabolism, Maastricht University, 6200MD, PO Box 61, Maastricht, The Netherlands
| | - Mahara Valverde
- Departamento de Medicina Genomica y Toxicologia Ambiental, Instituto de Investigaciones Biomedicas, Universidad Nacional Autonoma de México, México CP 04510, México
| | - Jordan Woodrick
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Luoping Zhang
- Division of Environmental Health Sciences, School of Public Health, University of California, Berkeley, CA 94720-7360, USA
| | - Nik van Larebeke
- Laboratory for Analytical and Environmental Chemistry, Vrije Universiteit Brussel, Brussels 1050, Belgium, Study Centre for Carcinogenesis and Primary Prevention of Cancer, Ghent University, Ghent 9000, Belgium
| | | | | |
Collapse
|
39
|
GAO YE, GUAN ZHENFENG, CHEN JIAQI, XIE HONGJUN, YANG ZHAO, FAN JINHAI, WANG XINYANG, LI LEI. CXCL5/CXCR2 axis promotes bladder cancer cell migration and invasion by activating PI3K/AKT-induced upregulation of MMP2/MMP9. Int J Oncol 2015; 47:690-700. [DOI: 10.3892/ijo.2015.3041] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Accepted: 05/15/2015] [Indexed: 11/05/2022] Open
|
40
|
Lim JB, Chung HW. Serum ENA78/CXCL5, SDF-1/CXCL12, and their combinations as potential biomarkers for prediction of the presence and distant metastasis of primary gastric cancer. Cytokine 2015; 73:16-22. [PMID: 25689618 DOI: 10.1016/j.cyto.2015.01.010] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Revised: 12/29/2014] [Accepted: 01/13/2015] [Indexed: 02/08/2023]
Abstract
BACKGROUND Chemokines play important roles in cancer development and progression. Epithelial-derived neutrophil-activating peptide-78 (ENA78/CXCL5) and stromal cell-derived factor (SDF-1/CXCL12) supposedly contribute to gastric cancer (GC) development and progression. This study aims to evaluate serum levels of ENA78/CXCL5 and SDF-1/CXCL12 along the GC carcinogenesis, and analyze their clinical significance, and diagnostic potentials through human serum samples. METHODS A total of 300 subjects were enrolled in this study. Serum levels of ENA78/CXCL5 and SDF-1/CXCL12, measured by chemiluminescent immunoassay, were compared among 4 disease groups; normal, high-risk (intestinal metaplasia and adenoma), early GC (EGC), and advanced GC (AGC) groups in both training (n=25 per group) and validation dataset (n=70, 30, 50, 50, respectively) by ANOVA test (post hoc Bonferroni). Correlations between serum ENA78/CXCL5 or SDF-1/CXCL12 levels and clinicopathological parameters of GC patients were evaluated (Spearman's correlation; γs). To validate the diagnostic accuracy, receiver operating characteristic (ROC) curve and logistic regression analysis was performed. RESULTS Serum ENA78/CXCL5 and SDF-1/CXCL12 levels were significantly higher in AGC groups than EGC, high-risk and normal groups in both training and validation dataset (Bonferroni, from p<0.01 to p<0.001). Clinicopathologically, serum ENA78/CXCL5 was correlated with T-stage (γs=0.231, p=0.021) and distant metastasis (γs=0.357, p<0.001), while serum SDF-1/CXCL12 was correlated with lymph node (γs=0.220, p=0.029) and distant (γs=0.425, p<0.001) metastasis. ROC curve and logistic regression demonstrated that serum ENA78/CXCL5 and SDF-1/CXCL12 showed higher diagnostic accuracy compared with carcinoembryonic antigen (CEA) in predicting GC. Serum ENA78/CXCL5 could predict both the presence of GC and distant metastasis, while serum SDF-1/CXCL12 could mainly predict its distant metastasis. All combination of serum ENA78/CXCL5, SDF-1/CXCL12, and CEA achieved 92.8% specificity at 75.0% sensitivity to predict distant metastasis of GC. CONCLUSIONS Combinations of initial serum ENA78/CXCL5, SDF-1/CXCL12, and CEA before any treatment for GC can produce valuable serum biomarker panels to predict the presence and distant metastasis of GC.
Collapse
Affiliation(s)
- Jong-Baeck Lim
- Department of Laboratory Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hye Won Chung
- Department of Laboratory Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea; Department of Internal Medicine, Division of Gastroenterology, International St. Mary's Hospital, Incheon Metropolitan City, Republic of Korea.
| |
Collapse
|
41
|
Xia J, Xu X, Huang P, He M, Wang X. The potential of CXCL5 as a target for liver cancer - what do we know so far? Expert Opin Ther Targets 2014; 19:141-6. [PMID: 25495348 DOI: 10.1517/14728222.2014.993317] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
CXCL5, epithelial cell derived neutrophil attractant 78, is a CXC chemokine predominantly expressed on epithelial cells. It has specificity for CXCR2 receptors and is involved in the recruitment and activation of neutrophils. CXCL5 is considered a therapeutic target in liver cancer, since treatment with small-interfering RNAs or antibodies against CXCL5 can suppress tumor growth, proliferation, migration and invasion. Experimental evidence demonstrated that CXCL5 antibodies could reduce the tumor growth and synergistically increase the efficiency of the tyrosine kinase inhibitor, Gefitinib, without the addition of toxicity. A number of challenges are encountered and should be considered during the development and clinical application of CXCL5 target-specific drugs. The specificity of CXCL5 as a therapeutic target for certain types and duration of cancer should be more carefully clarified, since it seems that CXCL5 is involved in many molecular pathways and crosstalk between targeted chemokines/receptors. The concept that CXCL5 serves as the therapeutic target for liver cancer was evidenced by preclinical studies, and is the beginning of CXCL5-based drug discovery and development.
Collapse
Affiliation(s)
- Jinglin Xia
- Fudan University, Minhang Hospital , Minhang , China +86 21 64041990 Ext: 5420 ; +86 21 54961729 ;
| | | | | | | | | |
Collapse
|
42
|
Xu X, Huang P, Yang B, Wang X, Xia J. Roles of CXCL5 on migration and invasion of liver cancer cells. J Transl Med 2014; 12:193. [PMID: 25011526 PMCID: PMC4097051 DOI: 10.1186/1479-5876-12-193] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Accepted: 07/01/2014] [Indexed: 12/15/2022] Open
Abstract
Inflammatory factors play a vital role in the progression of liver cancer, although exact factors and related mechanisms still remain unclear. The present study aimed at screening inflammatory factors related to liver cancer metastasis and investigating the potential mechanism by which cancer cells are recruited. We screened and validated inflammatory factors by microarray and RT-PCR. Small interfering RNA (siRNA) and recombinant protein were used to assess CXCL5 effects on the movement of liver cancer cells (LCCs). Our screening microarray demonstrated over-expression of CXCL5 in LCCs with high metastatic potentials. CXCL5 increased LCCs migration and invasion, probably through autocrine and paracrine mechanisms. CXCL5-CXCR2 and ERK1/2 pathways could play critical roles in the regulation of LCCs migration. Our data indicates that LCCs per se may act as the producer and receptor of CXCL5 responsible for liver cancer migration and invasion.
Collapse
Affiliation(s)
| | | | | | - Xiangdong Wang
- Liver Cancer Institute, Zhongshan Hospital, Fudan University; Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai 200032, China.
| | | |
Collapse
|
43
|
Sun S, Ning X, Zhai Y, Du R, Lu Y, He L, Li R, Wu W, Sun W, Wang H. Egr-1 mediates chronic hypoxia-induced renal interstitial fibrosis via the PKC/ERK pathway. Am J Nephrol 2014; 39:436-48. [PMID: 24819335 DOI: 10.1159/000362249] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Accepted: 03/03/2014] [Indexed: 12/27/2022]
Abstract
BACKGROUND Chronic hypoxia-induced epithelial-to-mesenchymal transition (EMT) is a crucial process in renal fibrogenesis. Egr-1, as a transcription factor, has been proven to be important in promoting EMT. However, whether it functions in hypoxia-induced renal tubular EMT has not been fully elucidated. METHODS Egr-1 were detected at mRNA and protein levels by qPCR and Western blot analysis respectively after renal epithelial cells were subjected to hypoxia treatment. Meanwhile, EMT phenotype was also observed through identification of relevant EMT-specific markers. siRNA was used to knock down Egr-1 expression and subsequent changes were observed. Specific PKC and MAPK/ERK inhibitors were employed to determine the molecular signaling pathway involved in Egr-1-mediated EMT phenotype. In vivo assays using rat remnant kidney model were used to validate the in vitro results. Furthermore, Egr-1 expression was examined in the samples of CKD patients with the clinical relevance revealed. RESULTS Hypoxia treatment enhanced the mRNA and protein levels of Egr-1 in HK-2 cells, which was accompanied by a reduced expression of the epithelial marker E-cadherin and an enhanced expression of the mesenchymal marker Fsp-1. Downregulation of Egr-1 with siRNA reversed hypoxia-induced EMT. Using the specific inhibitors to protein kinase C (calphostin C) or MAPK/ERK (PD98059), we identified that hypoxia induced Egr-1 expression through the PKC/ERK pathway. In addition, the upregulation of Egr-1 raised endogenous Snail levels, and the downregulation of Snail inhibited Egr-1-mediated EMT in HK-2 cells. Through in vivo assays using rat remnant kidney and CKD patients' kidney tissues, we found that Egr-1 and Snail were overexpressed in tubular epithelial cells with EMT. CONCLUSION Egr-1 may be an important regulator of the development of renal tubular EMT induced by hypoxia through the PKC/ERK pathway and the activation of Snail. Targeting Egr-1 expression or activity might be a novel therapeutic strategy to control renal fibrosis.
Collapse
Affiliation(s)
- Shiren Sun
- Department of Nephrology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Tumor microenvironment: a new treatment target for cancer. ISRN BIOCHEMISTRY 2014; 2014:351959. [PMID: 25937967 PMCID: PMC4392996 DOI: 10.1155/2014/351959] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/12/2014] [Accepted: 03/03/2014] [Indexed: 12/21/2022]
Abstract
Recent advances in cancer therapy encounter a bottleneck. Relapsing/recurrent disease almost always developed eventually with resistance to the initially effective drugs. Tumor microenvironment has been gradually recognized as a key contributor for cancer progression, epithelial-mesenchymal transition of the cancer cells, angiogenesis, cancer metastasis, and development of drug resistance, while dysregulated immune responses and interactions between various components in the microenvironment all play important roles. Future development of anticancer treatment should take tumor microenvironment into consideration. Besides, we also discuss the limitations of current pre-clinical testing models that mainly come from the impossibility in simulating all detailed carcinogenic mechanisms in human, especially failure to create the same tumor microenvironment. With the cumulating knowledge about tumor microenvironment, the design of a novel anticancer therapy may be facilitated and may have better chance for success in cancer eradication.
Collapse
|
45
|
Sepuru KM, Poluri KM, Rajarathnam K. Solution structure of CXCL5--a novel chemokine and adipokine implicated in inflammation and obesity. PLoS One 2014; 9:e93228. [PMID: 24695525 PMCID: PMC3973705 DOI: 10.1371/journal.pone.0093228] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Accepted: 03/01/2014] [Indexed: 11/19/2022] Open
Abstract
The chemokine CXCL5 is selectively expressed in highly specialized cells such as epithelial type II cells in the lung and white adipose tissue macrophages in muscle, where it mediates diverse functions from combating microbial infections by regulating neutrophil trafficking to promoting obesity by inhibiting insulin signaling. Currently very little is known regarding the structural basis of how CXCL5 mediates its novel functions. Towards this missing knowledge, we have solved the solution structure of the CXCL5 dimer by NMR spectroscopy. CXCL5 is a member of a subset of seven CXCR2-activating chemokines (CAC) that are characterized by the highly conserved ELR motif in the N-terminal tail. The structure shows that CXCL5 adopts the typical chemokine fold, but also reveals several distinct differences in the 30 s loop and N-terminal residues; not surprisingly, crosstalk between N-terminal and 30 s loop residues have been implicated as a major determinant of receptor activity. CAC function also involves binding to highly sulfated glycosaminoglycans (GAG), and the CXCL5 structure reveals a distinct distribution of positively charged residues, suggesting that differences in GAG interactions also influence function. The availability of the structure should now facilitate the design of experiments to better understand the molecular basis of various CXCL5 functions, and also serve as a template for the design of inhibitors for use in a clinical setting.
Collapse
Affiliation(s)
- Krishna Mohan Sepuru
- Department of Biochemistry & Molecular Biology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Krishna Mohan Poluri
- Department of Biochemistry & Molecular Biology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Krishna Rajarathnam
- Department of Biochemistry & Molecular Biology, University of Texas Medical Branch, Galveston, Texas, United States of America
- Sealy Center for Structural Biology and Molecular Biophysics, University of Texas Medical Branch, Galveston, Texas, United States of America
- * E-mail:
| |
Collapse
|
46
|
Kim J, Kang HS, Lee YJ, Lee HJ, Yun J, Shin JH, Lee CW, Kwon BM, Hong SH. EGR1-dependent PTEN upregulation by 2-benzoyloxycinnamaldehyde attenuates cell invasion and EMT in colon cancer. Cancer Lett 2014; 349:35-44. [PMID: 24704156 DOI: 10.1016/j.canlet.2014.03.025] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2014] [Revised: 03/17/2014] [Accepted: 03/23/2014] [Indexed: 01/17/2023]
Abstract
There has been little evidence to support EGR1 and PTEN function on the EMT of cancer cells. We tried to evaluate how these genes affect cancer cell invasion and EMT through investigating the molecular mechanism(s) of 2'-benzoyloxycinnamaldehyde (BCA). Matrigel invasion and wound healing assay, and in vivo mice model were used to evaluate the effect of BCA on colon cancer cell migration. The molecular mechanism(s) of BCA were evaluated by knock-down or overexpression of EGR1 and PTEN. BCA at 50 nM increased E-cadherin and EGR1 expression without cytotoxicity. Cell migration was inhibited significantly by BCA both in vitro and in vivo. Moreover, BCA inhibits Snail and Vimentin expression, as well as β-catenin nuclear accumulation. Suppression of EGR1 by siRNA attenuated the inhibition of matrigel invasion by BCA, indicating that EGR1 is responsible for BCA effect. PTEN was upregulated by BCA treatment or EGR1 overexpression. In addition, shPTEN transfection stimulated EMT and cell invasion in vitro. Our data suggest that BCA leads to a remarkable upregulation of EGR1 expression, and that EMT and invasion is decreased via EGR1-dependent PTEN activation. These data showed a critical role of EGR1-PTEN signaling pathway in the EMT of colon cancer, as well as metastasis.
Collapse
Affiliation(s)
- Jinkyung Kim
- Department of Oral Microbiology, School of Dentistry, Kyungpook National University, Daegu 700-412, South Korea.
| | - Hye Suk Kang
- Department of Oral Microbiology, School of Dentistry, Kyungpook National University, Daegu 700-412, South Korea.
| | - Yu-Jin Lee
- Laboratory of Chemical Biology and Genomics, Korea Research Institute of Bioscience and Biotechnology, Daejon 305-806, South Korea.
| | - Heon-Jin Lee
- Department of Oral Microbiology, School of Dentistry, Kyungpook National University, Daegu 700-412, South Korea.
| | - Jieun Yun
- Bioevaluation Center, Korea Research Institute of Bioscience and Biotechnology, Ochang, Cheongwon, Chungbuk 363-883, South Korea.
| | - Jung Hyu Shin
- Bioevaluation Center, Korea Research Institute of Bioscience and Biotechnology, Ochang, Cheongwon, Chungbuk 363-883, South Korea.
| | - Chang Woo Lee
- Bioevaluation Center, Korea Research Institute of Bioscience and Biotechnology, Ochang, Cheongwon, Chungbuk 363-883, South Korea.
| | - Byoung-Mog Kwon
- Laboratory of Chemical Biology and Genomics, Korea Research Institute of Bioscience and Biotechnology, Daejon 305-806, South Korea.
| | - Su-Hyung Hong
- Department of Oral Microbiology, School of Dentistry, Kyungpook National University, Daegu 700-412, South Korea.
| |
Collapse
|
47
|
Zheng J, Zhu X, Zhang J. CXCL5 knockdown expression inhibits human bladder cancer T24 cells proliferation and migration. Biochem Biophys Res Commun 2014; 446:18-24. [PMID: 24583128 DOI: 10.1016/j.bbrc.2014.01.172] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2014] [Accepted: 01/28/2014] [Indexed: 11/17/2022]
Abstract
CXCL5 (epithelial neutrophil activating peptide-78) which acts as a potent chemoattractant and activator of neutrophil function was reported to play a multifaceted role in tumorigenesis. To investigate the role of CXCL5 in bladder cancer progression, we examined the CXCL5 expression in bladder cancer tissues by real-time PCR and Western blot, additionally, we used shRNA-mediated silencing to generate stable CXCL5 silenced bladder cancer T24 cells and defined its biological functions. Our results demonstrated that mRNA and protein of CXCL5 is increased in human bladder tumor tissues and cell lines, down-regulation of CXCL5 in T24 cells resulted in significantly decreased cell proliferation, migration and increased cell apoptosis in vitro through Snail, PI3K-AKT and ERK1/2 signaling pathways. These data suggest that CXCL5 is critical for bladder tumor growth and progression, it may represent a potential application in cancer diagnosis and therapy.
Collapse
Affiliation(s)
- Jiajia Zheng
- Department of Laboratory Medicine, Peking University Third Hospital, Beijing, China
| | - Xi Zhu
- Department of Urology, Beijing Friendship Hospital Affiliated to Capital Medical University, Beijing, China
| | - Jie Zhang
- Department of Laboratory Medicine, Peking University Third Hospital, Beijing, China.
| |
Collapse
|
48
|
Kinehara M, Kawamura S, Mimura S, Suga M, Hamada A, Wakabayashi M, Nikawa H, Furue MK. Protein kinase C-induced early growth response protein-1 binding to SNAIL promoter in epithelial-mesenchymal transition of human embryonic stem cells. Stem Cells Dev 2014; 23:2180-9. [PMID: 24410631 DOI: 10.1089/scd.2013.0424] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Epithelial-mesenchymal transition (EMT) has been thought to occur during early embryogenesis, and also the differentiation process of human embryonic stem (hES) cells. Spontaneous differentiation is sometimes observed at the peripheral of the hES cell colonies in conventional culture conditions, indicating that EMT occurs in hES cell culture. However, the triggering mechanism of EMT is not yet fully understood. The balance between self-renewal and differentiation of human pluripotent stem (hPS) cells is controlled by various signal pathways, including the fibroblast growth factor (FGF)-2. However, FGF-2 has a complex role for self-renewal of hES cells. FGF-2 activates phosphatidylinositol-3 kinase/AKT, mitogen-activated protein kinase/extracellular signal-regulated kinase-1/2 kinase, and also protein kinase C (PKC). Here, we showed that a PKC rapidly induced an early growth response protein-1 (EGR-1) in hES cells, which was followed by upregulation of EMT-related genes. Before the induction of EMT-related genes, EGR-1 was translocated into the nucleus, and then bound directly to the promoter region of SNAIL, which is a master regulator of EMT. SNAIL expression was attenuated by knockdown of EGR-1, but upregulated by ectopic expression of EGR-1. EGR-1 as the downstream signal of PKC might play a key role in EMT initiation during early differentiation of hES cells. This study would lead to a more robust understanding of the mechanisms underlying the balance between self-renewal and initiation of differentiation in hPS cells.
Collapse
Affiliation(s)
- Masaki Kinehara
- 1 Laboratory of Stem Cell Cultures, Department of Disease Bioresources Research, National Institute of Biomedical Innovation , Ibaraki, Japan
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Liggett JL, Zhang X, Eling TE, Baek SJ. Anti-tumor activity of non-steroidal anti-inflammatory drugs: cyclooxygenase-independent targets. Cancer Lett 2014; 346:217-24. [PMID: 24486220 DOI: 10.1016/j.canlet.2014.01.021] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Revised: 01/21/2014] [Accepted: 01/22/2014] [Indexed: 12/27/2022]
Abstract
Non-steroidal anti-inflammatory drugs (NSAIDs) are used extensively for analgesic and antipyretic treatments. In addition, NSAIDs reduce the risk and mortality to several cancers. Their mechanisms in anti-tumorigenesis are not fully understood, but both cyclooxygenase (COX)-dependent and -independent pathways play a role. We and others have been interested in elucidating molecular targets of NSAID-induced apoptosis. In this review, we summarize updated literature regarding cellular and molecular targets modulated by NSAIDs. Among those NSAIDs, sulindac sulfide and tolfenamic acid are emphasized in this review because these two drugs have been well investigated for their anti-tumorigenic activity in many different types of cancer.
Collapse
Affiliation(s)
- Jason L Liggett
- Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN 37996 USA
| | - Xiaobo Zhang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Thomas E Eling
- Laboratory of Molecular Carcinogenesis, National Institutes of Environmental Health Sciences, NIH, Research Triangle Park, NC 27709, USA
| | - Seung Joon Baek
- Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN 37996 USA.
| |
Collapse
|
50
|
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: 28] [Impact Index Per Article: 2.8] [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
|