1
|
Kamal R, Awasthi A, Paul P, Mir MS, Singh SK, Dua K. Novel drug delivery systems in colorectal cancer: Advances and future prospects. Pathol Res Pract 2024; 262:155546. [PMID: 39191194 DOI: 10.1016/j.prp.2024.155546] [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: 02/20/2024] [Revised: 08/10/2024] [Accepted: 08/14/2024] [Indexed: 08/29/2024]
Abstract
Colorectal cancer (CRC) is an abnormal proliferation of cells within the colon and rectum, leading to the formation of polyps and disruption of mucosal functions. The disease development is influenced by a combination of factors, including inflammation, exposure to environmental mutagens, genetic alterations, and impairment in signaling pathways. Traditional treatments such as surgery, radiation, and chemotherapy are often used but have limitations, including poor solubility and permeability, treatment resistance, side effects, and post-surgery issues. Novel Drug Delivery Systems (NDDS) have emerged as a superior alternative, offering enhanced drug solubility, precision in targeting cancer cells, and regulated drug release. Thereby addressing the shortcomings of conventional therapies and showing promise for more effective CRC management. The present review sheds light on the pathogenesis, signaling pathways, biomarkers, conventional treatments, need for NDDS, and application of NDDS against CRC. Additionally, clinical trials, ongoing clinical trials, marketed formulations, and patents on CRC are also covered in the present review.
Collapse
Affiliation(s)
- Raj Kamal
- Department of Quality Assurance, ISF College of Pharmacy, Moga, Punjab 142001, India; School of Pharmacy, Desh Bhagat University, Mandi Gobindgarh, Punjab 147301, India
| | - Ankit Awasthi
- Department of Pharmaceutics, ISF College of Pharmacy, Moga, Punjab 142001, India; Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India.
| | - Priyanka Paul
- Department of Pharmaceutical Science, PCTE Group of Institute, Ludhiana, Punjab, India
| | - Mohammad Shabab Mir
- School of Pharmacy, Desh Bhagat University, Mandi Gobindgarh, Punjab 147301, India
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144411, India; Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - Kamal Dua
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW 2007, Australia; Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW 2007, Australia
| |
Collapse
|
2
|
Chong Z, Huang F, McLeod M, Irwin R, Smithson M, Yue Z, Gao M, Hardiman K. Molecular differentiation between complete and incomplete responders to neoadjuvant therapy in rectal cancer. RESEARCH SQUARE 2024:rs.3.rs-4456000. [PMID: 39011117 PMCID: PMC11247942 DOI: 10.21203/rs.3.rs-4456000/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/17/2024]
Abstract
Background Neoadjuvant chemoradiotherapy (nCRT) is the standard treatment for locally advanced rectal cancer, but only 20-40% of patients completely respond to this treatment. Methods To define the molecular features that are associated with response to nCRT, we generated and collected genomic and transcriptomic data from 712 cancers prior to treatment from our own data and from publicly available data. Results We found that patients with a complete response have decreased risk of both local recurrence and future metastasis. We identified multiple differences in DNA mutations and transcripts between complete and incomplete responders. Complete responder tumors have a higher tumor mutation burden and more significant co-occurring mutations than the incomplete responder tumors. In addition, mutations in DNA repair genes (across multiple mechanisms of repair) were enriched in complete responders and they also had lower expression of these genes indicating that defective DNA repair is associated with complete response to nCRT. Using logistic regression, we identified three significant predictors of complete response: tumor size, mutations within specific network genes, and the existence of three or more specific co-occurrent mutations. In incompletely responder tumors, abnormal cell-cell interaction and increased cancer associated fibroblasts were associated with recurrence. Additionally, gene expression analysis identified a subset of immune hot tumors with worse outcomes and upregulated of immune checkpoint proteins. Conclusions Overall, our study provides a comprehensive understanding of the molecular features associated with response to nCRT and the molecular differences in non-responder tumors that later reoccur. This knowledge may provide critical insight for the development of precision therapy for rectal cancer.
Collapse
Affiliation(s)
| | | | - M McLeod
- University of Alabama at Birmingham
| | | | | | | | - Min Gao
- University of Alabama at Birmingham
| | | |
Collapse
|
3
|
Zhao Z, Chen J, Jiang Y, Lu Y. The E156K mutation in the CRYAA gene affects the epithelial-mesenchymal transition and migration of human lens epithelial cells. Heliyon 2024; 10:e23690. [PMID: 38187316 PMCID: PMC10770500 DOI: 10.1016/j.heliyon.2023.e23690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 11/30/2023] [Accepted: 12/09/2023] [Indexed: 01/09/2024] Open
Abstract
Purpose To investigated the biological effects of E156K-mutated αA-crystallin (CRYAA) in human lens epithelial cells (HLECs). Methods FLAG-tagged, human, full-length, wild-type (WT), or E156K-mutated CRYAA was expressed in HLECs under CRYAA knockdown. CRYAA expression was determined by quantitative reverse transcription polymerase chain reaction and western blotting (WB). Rhodamine cytoskeleton staining was used to observe the changes in cell morphology following transfection with WT or E156K-mutated CRYAA plasmids. WB was performed to assess the expression of markers related to epithelial-mesenchymal transition (EMT) and migration. Results Rhodamine cytoskeleton staining revealed changes in the morphology of cells transfected with E156K-mutated CRYAA and opposite responses occurred after treatment with a β-catenin inhibitor. Cells transfected with E156K-mutated CRYAA expressed remarkably higher levels of the mesenchymal biomarkers N-cadherin and vimentin but decreased levels of the epithelial biomarker E-cadherin, whereas opposite trends were observed in cells treated with the β-catenin inhibitor, ICG001. The migratory capability of E156K-mutated CRYAA cells was significantly greater than that of WT cells (P < 0.001). This effect was accompanied by significantly increased expression levels of phosphorylated (p)-focal adhesion kinase (FAK) and p-Src. These changes were decreased significantly by treatment with FAK and Src inhibitors. Conclusion E156K-mutated CRYAA induced EMT, in which the HLECs lost cell polarity, and acquired a mesenchymal phenotype with greater migratory capability. These biological effects may be associated with activation of the Wnt/β-Catenin and FAK/Src signaling pathways.
Collapse
Affiliation(s)
- Zhennan Zhao
- Department of Ophthalmology and Vision Science, Eye Ear Nose and Throat Hospital of Fudan University, Shanghai, China
- National Health Commission Key Laboratory of Myopia (Fudan University), Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, China
- Key Laboratory of Visual Impairment and Restoration of Shanghai, Shanghai, China
| | - Jiahui Chen
- Department of Ophthalmology and Vision Science, Eye Ear Nose and Throat Hospital of Fudan University, Shanghai, China
- National Health Commission Key Laboratory of Myopia (Fudan University), Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, China
- Key Laboratory of Visual Impairment and Restoration of Shanghai, Shanghai, China
| | - Yongxiang Jiang
- Department of Ophthalmology and Vision Science, Eye Ear Nose and Throat Hospital of Fudan University, Shanghai, China
- National Health Commission Key Laboratory of Myopia (Fudan University), Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, China
- Key Laboratory of Visual Impairment and Restoration of Shanghai, Shanghai, China
| | - Yi Lu
- Department of Ophthalmology and Vision Science, Eye Ear Nose and Throat Hospital of Fudan University, Shanghai, China
- National Health Commission Key Laboratory of Myopia (Fudan University), Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, China
- Key Laboratory of Visual Impairment and Restoration of Shanghai, Shanghai, China
| |
Collapse
|
4
|
Jerin S, Harvey AJ, Lewis A. Therapeutic Potential of Protein Tyrosine Kinase 6 in Colorectal Cancer. Cancers (Basel) 2023; 15:3703. [PMID: 37509364 PMCID: PMC10377740 DOI: 10.3390/cancers15143703] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 07/10/2023] [Accepted: 07/19/2023] [Indexed: 07/30/2023] Open
Abstract
PTK6, a non-receptor tyrosine kinase, modulates the pathogenesis of breast and prostate cancers and is recognized as a biomarker of breast cancer prognosis. There are over 30 known substrates of PTK6, including signal transducers, transcription factors, and RNA-binding proteins. Many of these substrates are known drivers of other cancer types, such as colorectal cancer. Colon and rectal tumors also express higher levels of PTK6 than the normal intestine suggesting a potential role in tumorigenesis. However, the importance of PTK6 in colorectal cancer remains unclear. PTK6 inhibitors such as XMU-MP-2 and Tilfrinib have demonstrated potency and selectivity in breast cancer cells when used in combination with chemotherapy, indicating the potential for PTK6 targeted therapy in cancer. However, most of these inhibitors are yet to be tested in other cancer types. Here, we discuss the current understanding of the function of PTK6 in normal intestinal cells compared with colorectal cancer cells. We review existing PTK6 targeting therapeutics and explore the possibility of PTK6 inhibitory therapy for colorectal cancer.
Collapse
Affiliation(s)
- Samanta Jerin
- Centre for Genome Engineering and Maintenance, Division of Biosciences, Department of Life Sciences, College of Health and Life Sciences, Brunel University London, Uxbridge UB8 3PH, UK
| | - Amanda J Harvey
- Centre for Genome Engineering and Maintenance, Institute for Health Medicine and Environments, Brunel University London, Uxbridge UB8 3PH, UK
| | - Annabelle Lewis
- Centre for Genome Engineering and Maintenance, Division of Biosciences, Department of Life Sciences, College of Health and Life Sciences, Brunel University London, Uxbridge UB8 3PH, UK
| |
Collapse
|
5
|
Hu J, He Y, Liao K, Yang Q, Xu Y, Cao G, Wang X. Identification of inflammatory factor-related genes associated with the prognostic and immune cell infiltration in colorectal cancer patients. Genes Dis 2022. [PMID: 37492736 PMCID: PMC10363590 DOI: 10.1016/j.gendis.2022.07.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
This study aims to identify the inflammatory factor-related genes which help to predict the prognosis of patients with colorectal cancer. GSEA (Gene Set Enrichment Analysis) was used to acquire inflammation-related genes and the corresponding expression information was collected from TCGA database to determine the DEGs (differentially-expressed genes) in CRC patients. We conducted enrichment analysis and PPI (protein-protein interaction) of these DEGs. Besides, key genes that are both differentially-expressed and prognosis-related were screened out, which were used to establish the prognostic model. We obtained 79 DEGs and 19 prognostic genes, 10 prognostic-related differential genes were eventually screened. These genes were used to construct the prognostic model. We also identified that the immune infiltration score of macrophages between different risk groups was significantly different and similar distinction was witnessed in immune function score of APC (antigen-presenting cell) co-stimulation and type I IFN (interferon) response.
Collapse
|
6
|
Rahiminejad S, Maurya MR, Mukund K, Subramaniam S. Modular and mechanistic changes across stages of colorectal cancer. BMC Cancer 2022; 22:436. [PMID: 35448980 PMCID: PMC9022252 DOI: 10.1186/s12885-022-09479-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Accepted: 03/23/2022] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND While mechanisms contributing to the progression and metastasis of colorectal cancer (CRC) are well studied, cancer stage-specific mechanisms have been less comprehensively explored. This is the focus of this manuscript. METHODS Using previously published data for CRC (Gene Expression Omnibus ID GSE21510), we identified differentially expressed genes (DEGs) across four stages of the disease. We then generated unweighted and weighted correlation networks for each of the stages. Communities within these networks were detected using the Louvain algorithm and topologically and functionally compared across stages using the normalized mutual information (NMI) metric and pathway enrichment analysis, respectively. We also used Short Time-series Expression Miner (STEM) algorithm to detect potential biomarkers having a role in CRC. RESULTS Sixteen Thousand Sixty Two DEGs were identified between various stages (p-value ≤ 0.05). Comparing communities of different stages revealed that neighboring stages were more similar to each other than non-neighboring stages, at both topological and functional levels. A functional analysis of 24 cancer-related pathways indicated that several signaling pathways were enriched across all stages. However, the stage-unique networks were distinctly enriched only for a subset of these 24 pathways (e.g., MAPK signaling pathway in stages I-III and Notch signaling pathway in stages III and IV). We identified potential biomarkers, including HOXB8 and WNT2 with increasing, and MTUS1 and SFRP2 with decreasing trends from stages I to IV. Extracting subnetworks of 10 cancer-relevant genes and their interacting first neighbors (162 genes in total) revealed that the connectivity patterns for these genes were different across stages. For example, BRAF and CDK4, members of the Ser/Thr kinase, up-regulated in cancer, displayed changing connectivity patterns from stages I to IV. CONCLUSIONS Here, we report molecular and modular networks for various stages of CRC, providing a pseudo-temporal view of the mechanistic changes associated with the disease. Our analysis highlighted similarities at both functional and topological levels, across stages. We further identified stage-specific mechanisms and biomarkers potentially contributing to the progression of CRC.
Collapse
Affiliation(s)
- Sara Rahiminejad
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA
- Department of Mechanical and Aerospace Engineering, University of California, San Diego, La Jolla, CA, USA
| | - Mano R Maurya
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA
| | - Kavitha Mukund
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA
| | - Shankar Subramaniam
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA.
- San Diego Supercomputer Center, University of California, San Diego, La Jolla, CA, USA.
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, USA.
- Department of Computer Science and Engineering, University of California, San Diego, La Jolla, CA, USA.
| |
Collapse
|
7
|
Predicted miRNAs suppressed cell proliferation and migration via FAK/VASP axis; Systems biology approach. INFORMATICS IN MEDICINE UNLOCKED 2022. [DOI: 10.1016/j.imu.2022.100890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
8
|
Deng L, Jiang N, Zeng J, Wang Y, Cui H. The Versatile Roles of Cancer-Associated Fibroblasts in Colorectal Cancer and Therapeutic Implications. Front Cell Dev Biol 2021; 9:733270. [PMID: 34660589 PMCID: PMC8517274 DOI: 10.3389/fcell.2021.733270] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 09/14/2021] [Indexed: 01/07/2023] Open
Abstract
The tumor microenvironment (TME) is populated by abundant cancer-associated fibroblasts (CAFs) that radically influence the disease progression across many cancers, including the colorectal cancer (CRC). In theory, targeting CAFs holds great potential in optimizing CRC treatment. However, attempts to translate the therapeutic benefit of CAFs into clinic practice face many obstacles, largely due to our limited understanding of the heterogeneity in their origins, functions, and mechanisms. In recent years, accumulating evidence has uncovered some cellular precursors and molecular markers of CAFs and also revealed their versatility in impacting various hallmarks of CRC, together helping us to better define the population of CAFs and also paving the way toward their future therapeutic targeting for CRC treatment. In this review, we outline the emerging concept of CAFs in CRC, with an emphasis on their origins, biomarkers, prognostic significance, as well as their functional roles and underlying mechanisms in CRC biology. At last, we discuss the prospect of harnessing CAFs as promising therapeutic targets for the treatment of patients with CRC.
Collapse
Affiliation(s)
- Longfei Deng
- Cancer Center, Medical Research Institute, Southwest University, Chongqing, China
| | - Nianfen Jiang
- Health Management Center, Southwest University Hospital, Chongqing, China
| | - Jun Zeng
- Department of Genetics and Cell Biology, College of Life Sciences, Chongqing Normal University, Chongqing, China
| | - Yi Wang
- Department of General Surgery, The Ninth People's Hospital of Chongqing, Affiliated Hospital of Southwest University, Chongqing, China
| | - Hongjuan Cui
- Cancer Center, Medical Research Institute, Southwest University, Chongqing, China.,Department of General Surgery, The Ninth People's Hospital of Chongqing, Affiliated Hospital of Southwest University, Chongqing, China.,State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, China
| |
Collapse
|
9
|
Patelli G, Tosi F, Amatu A, Mauri G, Curaba A, Patanè DA, Pani A, Scaglione F, Siena S, Sartore-Bianchi A. Strategies to tackle RAS-mutated metastatic colorectal cancer. ESMO Open 2021; 6:100156. [PMID: 34044286 PMCID: PMC8167159 DOI: 10.1016/j.esmoop.2021.100156] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 04/22/2021] [Accepted: 04/23/2021] [Indexed: 12/18/2022] Open
Abstract
The RAS oncogene is among the most commonly mutated in cancer. RAS mutations are identified in about half of patients diagnosed with metastatic colorectal cancer (mCRC), conferring poor prognosis and lack of response to anti-epidermal growth factor receptor (EGFR) antibodies. In the last decades, several investigational attempts failed in directly targeting RAS mutations, thus RAS was historically regarded as 'undruggable'. Recently, novel specific KRASG12C inhibitors showed promising results in different solid tumors, including mCRC, renewing interest in this biomarker as a target. In this review, we discuss different strategies of RAS targeting in mCRC, according to literature data in both clinical and preclinical settings. We recognized five main strategies focusing on those more promising: direct RAS targeting, targeting the mitogen-activated protein kinase (MAPK) pathway, harnessing RAS through immunotherapy combinations, RAS targeting through metabolic pathways, and finally other miscellaneous approaches. Direct KRASG12C inhibition is emerging as the most promising strategy in mCRC as well as in other solid malignancies. However, despite good disease control rates, tumor response and duration of response are still limited in mCRC. At this regard, combinational approaches with anti-epidermal growth factor receptor drugs or checkpoint inhibitors have been proposed to enhance treatment efficacy, based on encouraging results achieved in preclinical studies. Besides, concomitant therapies increasing metabolic stress are currently under evaluation and expected to also provide remarkable results in RAS codon mutations apart from KRASG12C. In conclusion, based on hereby reported efforts of translational research, RAS mutations should no longer be regarded as 'undruggable' and future avenues are now opening for translation in the clinic in mCRC.
Collapse
Affiliation(s)
- G Patelli
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy; Department of Oncology and Hemato-Oncology, Università degli Studi di Milano (La Statale), Milan, Italy
| | - F Tosi
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - A Amatu
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - G Mauri
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy; Department of Oncology and Hemato-Oncology, Università degli Studi di Milano (La Statale), Milan, Italy
| | - A Curaba
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy; Department of Oncology and Hemato-Oncology, Università degli Studi di Milano (La Statale), Milan, Italy
| | - D A Patanè
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy; Department of Oncology and Hemato-Oncology, Università degli Studi di Milano (La Statale), Milan, Italy
| | - A Pani
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano (La Statale), Milan, Italy
| | - F Scaglione
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano (La Statale), Milan, Italy; Clinical Pharmacology Unit, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - S Siena
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy; Department of Oncology and Hemato-Oncology, Università degli Studi di Milano (La Statale), Milan, Italy
| | - A Sartore-Bianchi
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy; Department of Oncology and Hemato-Oncology, Università degli Studi di Milano (La Statale), Milan, Italy.
| |
Collapse
|
10
|
Jiang H, Tang J, Qiu L, Zhang Z, Shi S, Xue L, Kui L, Huang T, Nan W, Zhou B, Zhao C, Yu M, Sun Q. Semaphorin 4D is a potential biomarker in pediatric leukemia and promotes leukemogenesis by activating PI3K/AKT and ERK signaling pathways. Oncol Rep 2021. [PMID: 33649851 DOI: 10.3892/or.2021.8021/html] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/23/2023] Open
Abstract
Semaphorin 4D (Sema4D) is highly expressed in a variety of tumors and is associated with high invasion, poor prognosis and poor therapeutic response. However, the expression and role of Sema4D in leukemia remains unclear. The present study investigated the expression of Sema4D in pediatric leukemia and its effects in leukemia cells. The results demonstrated that Sema4D protein was highly expressed in peripheral blood mononuclear cells of patients with pediatric leukemia, and high levels of soluble Sema4D were also observed in the plasma of these patients. Sema4D knockdown induced cell cycle arrest in G0/G1 phase, inhibited proliferation and promoted apoptosis in BALL‑1 cells, while Sema4D overexpression exhibited the opposite effect. In Jurkat cells, Sema4D knockdown inhibited proliferation and promoted apoptosis, while Sema4D overexpression decreased the abundance of the cells in the G0/G1 phase of the cell cycle and promoted proliferation. Sema4D overexpression also increased the migratory capacity of Jurkat cells and the invasive capacity of BALL‑1 cells. The phosphorylation level of PI3K was decreased in both Sema4D knocked‑down Jurkat and BALL‑1 cells, and the phosphorylation level of ERK was decreased in Sema4D knocked‑down BALL‑1 cells. The phosphorylation levels of PI3K, ERK and AKT were elevated in patients with pediatric leukemia, and were correlated to the increased Sema4D expression. Sema4D overexpression was associated with a shorter overall survival in patients with acute myeloid leukemia. Overall, the results of the present study indicated that Sema4D serves an important role in leukemia development by activating PI3K/AKT and ERK signaling, and it may be used as a potential target for the diagnosis and treatment of leukemia.
Collapse
MESH Headings
- Adolescent
- Antigens, CD/biosynthesis
- Antigens, CD/blood
- Biomarkers, Tumor/biosynthesis
- Biomarkers, Tumor/blood
- Case-Control Studies
- Cell Line, Tumor
- Cell Proliferation/physiology
- Child
- Child, Preschool
- Female
- Humans
- Infant
- Jurkat Cells
- Leukemia, Myeloid, Acute/blood
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- Leukocytes, Mononuclear/metabolism
- MAP Kinase Signaling System
- Male
- Phosphatidylinositol 3-Kinases/metabolism
- Phosphorylation
- Precursor Cell Lymphoblastic Leukemia-Lymphoma/blood
- Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism
- Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology
- Proto-Oncogene Proteins c-akt/metabolism
- Semaphorins/biosynthesis
- Semaphorins/blood
Collapse
Affiliation(s)
- Hongchao Jiang
- Institute of Pediatrics, The Kunming Children's Hospital, Kunming, Yunnan 650228, P.R. China
| | - Jiaolian Tang
- Institute of Pediatrics, Children's Hospital Affiliated to Kunming Medical University, Kunming, Yunnan 650228, P.R. China
| | - Lijuan Qiu
- Institute of Pediatrics, The Kunming Children's Hospital, Kunming, Yunnan 650228, P.R. China
| | - Zhen Zhang
- Institute of Pediatrics, The Kunming Children's Hospital, Kunming, Yunnan 650228, P.R. China
| | - Shulan Shi
- Institute of Pediatrics, The Kunming Children's Hospital, Kunming, Yunnan 650228, P.R. China
| | - Li Xue
- Institute of Medicine, Dali University, Dali, Yunnan 671000, P.R. China
| | - Liyue Kui
- Institute of Pediatrics, The Kunming Children's Hospital, Kunming, Yunnan 650228, P.R. China
| | - Tilong Huang
- Institute of Pediatrics, The Kunming Children's Hospital, Kunming, Yunnan 650228, P.R. China
| | - Weiwei Nan
- Institute of Medicine, Dali University, Dali, Yunnan 671000, P.R. China
| | - Bailing Zhou
- Institute of Pediatrics, The Kunming Children's Hospital, Kunming, Yunnan 650228, P.R. China
| | - Canchun Zhao
- Institute of Pediatrics, The Kunming Children's Hospital, Kunming, Yunnan 650228, P.R. China
| | - Ming Yu
- Yunnan Key Laboratory of Stem Cell and Regenerative Medicine, Biomedical Engineering Research Center, Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| | - Qiangming Sun
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, Yunnan 650118, P.R. China
| |
Collapse
|
11
|
Jiang H, Tang J, Qiu L, Zhang Z, Shi S, Xue L, Kui L, Huang T, Nan W, Zhou B, Zhao C, Yu M, Sun Q. Semaphorin 4D is a potential biomarker in pediatric leukemia and promotes leukemogenesis by activating PI3K/AKT and ERK signaling pathways. Oncol Rep 2021; 45:1. [PMID: 33649851 PMCID: PMC7877000 DOI: 10.3892/or.2021.7952] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Accepted: 12/18/2020] [Indexed: 12/18/2022] Open
Abstract
Semaphorin 4D (Sema4D) is highly expressed in a variety of tumors and is associated with high invasion, poor prognosis and poor therapeutic response. However, the expression and role of Sema4D in leukemia remains unclear. The present study investigated the expression of Sema4D in pediatric leukemia and its effects in leukemia cells. The results demonstrated that Sema4D protein was highly expressed in peripheral blood mononuclear cells of patients with pediatric leukemia, and high levels of soluble Sema4D were also observed in the plasma of these patients. Sema4D knockdown induced cell cycle arrest in G0/G1 phase, inhibited proliferation and promoted apoptosis in BALL-1 cells, while Sema4D overexpression exhibited the opposite effect. In Jurkat cells, Sema4D knockdown inhibited proliferation and promoted apoptosis, while Sema4D overexpression decreased the abundance of the cells in the G0/G1 phase of the cell cycle and promoted proliferation. Sema4D overexpression also increased the migratory capacity of Jurkat cells and the invasive capacity of BALL-1 cells. The phosphorylation level of PI3K was decreased in both Sema4D knocked-down Jurkat and BALL-1 cells, and the phosphorylation level of ERK was decreased in Sema4D knocked-down BALL-1 cells. The phosphorylation levels of PI3K, ERK and AKT were elevated in patients with pediatric leukemia, and were correlated to the increased Sema4D expression. Sema4D overexpression was associated with a shorter overall survival in patients with acute myeloid leukemia. Overall, the results of the present study indicated that Sema4D serves an important role in leukemia development by activating PI3K/AKT and ERK signaling, and it may be used as a potential target for the diagnosis and treatment of leukemia.
Collapse
Affiliation(s)
- Hongchao Jiang
- Institute of Pediatrics, The Kunming Children's Hospital, Kunming, Yunnan 650228, P.R. China
| | - Jiaolian Tang
- Institute of Pediatrics, Children's Hospital Affiliated to Kunming Medical University, Kunming, Yunnan 650228, P.R. China
| | - Lijuan Qiu
- Institute of Pediatrics, The Kunming Children's Hospital, Kunming, Yunnan 650228, P.R. China
| | - Zhen Zhang
- Institute of Pediatrics, The Kunming Children's Hospital, Kunming, Yunnan 650228, P.R. China
| | - Shulan Shi
- Institute of Pediatrics, The Kunming Children's Hospital, Kunming, Yunnan 650228, P.R. China
| | - Li Xue
- Institute of Medicine, Dali University, Dali, Yunnan 671000, P.R. China
| | - Liyue Kui
- Institute of Pediatrics, The Kunming Children's Hospital, Kunming, Yunnan 650228, P.R. China
| | - Tilong Huang
- Institute of Pediatrics, The Kunming Children's Hospital, Kunming, Yunnan 650228, P.R. China
| | - Weiwei Nan
- Institute of Medicine, Dali University, Dali, Yunnan 671000, P.R. China
| | - Bailing Zhou
- Institute of Pediatrics, The Kunming Children's Hospital, Kunming, Yunnan 650228, P.R. China
| | - Canchun Zhao
- Institute of Pediatrics, The Kunming Children's Hospital, Kunming, Yunnan 650228, P.R. China
| | - Ming Yu
- Yunnan Key Laboratory of Stem Cell and Regenerative Medicine, Biomedical Engineering Research Center, Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| | - Qiangming Sun
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, Yunnan 650118, P.R. China
| |
Collapse
|
12
|
Jeong KY, Park M, Sim JJ, Kim HM. Combination Antitumor Effect of Sorafenib via Calcium-Dependent Deactivation of Focal Adhesion Kinase Targeting Colorectal Cancer Cells. Molecules 2020; 25:5299. [PMID: 33202899 PMCID: PMC7697278 DOI: 10.3390/molecules25225299] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 10/28/2020] [Accepted: 11/06/2020] [Indexed: 12/31/2022] Open
Abstract
Sorafenib has been recently used for the treatment of patients with advanced colorectal cancer (CRC) and is recognized for its therapeutic value. However, the continuous use of sorafenib may cause resistance in the treatment of cancer patients. In this study, we investigated whether sorafenib exerts an enhanced anticancer effect on CRC cells via the calcium-mediated deactivation of the focal adhesion kinase (FAK) signaling pathways. The appropriate dose of sorafenib and lactate calcium salt (CaLa) for a combination treatment were determined using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assays. Then, cell cycle analysis was performed following treatment with 2.5 μM sorafenib and/or 2.5 mM CaLa. CRC cells were found to be in the G1 phase by sorafenib treatment, and they accumulated in the sub-G1 phase with CaLa treatment. Western blots and enzyme-linked immunosorbent assays were performed to analyze the elements of the recombinant activated factor (RAF) and focal adhesion kinase (FAK) signaling cascades. Sorafenib-inhibited RAF-dependent signaling in CRC cells, however, either did not affect the expression of Akt or increased it. As the upstream signaling of FAK was suppressed by CaLa, we observed that the expression of the sub-signaling phospho (p) AKT and p-mammalian target of rapamycin was also suppressed. Treatment with a combination of sorafenib and CaLa enhanced the antitumor activity of CRC cells. The % viability of CRC cells was significantly decreased compared to the single treatment with sorafenib or CaLa, and the accumulation of Sub G1 of CRC cells was clearly confirmed. The migration ability of CRC cells was significantly reduced. The findings of this study indicate that sorafenib will show further improved antitumor efficacy against CRC due to overcoming resistance through the use of CaLa.
Collapse
Affiliation(s)
- Keun-Yeong Jeong
- R&D Center, Metimedi Pharmaceuticals, 263, Central-ro, Yeonsu-gu, Incheon 22006, Korea; (M.P.); (J.-J.S.)
| | - Minhee Park
- R&D Center, Metimedi Pharmaceuticals, 263, Central-ro, Yeonsu-gu, Incheon 22006, Korea; (M.P.); (J.-J.S.)
| | - Jae-Jun Sim
- R&D Center, Metimedi Pharmaceuticals, 263, Central-ro, Yeonsu-gu, Incheon 22006, Korea; (M.P.); (J.-J.S.)
| | - Hwan Mook Kim
- Gachon Institute of Pharmaceutical Science, Gachon University, 191, Hambangmoe-ro, Yeonsu-gu, Incheon 21936, Korea
| |
Collapse
|
13
|
Combination Antitumor Effect of Sorafenib via Calcium-Dependent Deactivation of Focal Adhesion Kinase Targeting Colorectal Cancer Cells. MOLECULES (BASEL, SWITZERLAND) 2020. [PMID: 33202899 DOI: 10.3390/molecules25225299.] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Sorafenib has been recently used for the treatment of patients with advanced colorectal cancer (CRC) and is recognized for its therapeutic value. However, the continuous use of sorafenib may cause resistance in the treatment of cancer patients. In this study, we investigated whether sorafenib exerts an enhanced anticancer effect on CRC cells via the calcium-mediated deactivation of the focal adhesion kinase (FAK) signaling pathways. The appropriate dose of sorafenib and lactate calcium salt (CaLa) for a combination treatment were determined using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assays. Then, cell cycle analysis was performed following treatment with 2.5 μM sorafenib and/or 2.5 mM CaLa. CRC cells were found to be in the G1 phase by sorafenib treatment, and they accumulated in the sub-G1 phase with CaLa treatment. Western blots and enzyme-linked immunosorbent assays were performed to analyze the elements of the recombinant activated factor (RAF) and focal adhesion kinase (FAK) signaling cascades. Sorafenib-inhibited RAF-dependent signaling in CRC cells, however, either did not affect the expression of Akt or increased it. As the upstream signaling of FAK was suppressed by CaLa, we observed that the expression of the sub-signaling phospho (p) AKT and p-mammalian target of rapamycin was also suppressed. Treatment with a combination of sorafenib and CaLa enhanced the antitumor activity of CRC cells. The % viability of CRC cells was significantly decreased compared to the single treatment with sorafenib or CaLa, and the accumulation of Sub G1 of CRC cells was clearly confirmed. The migration ability of CRC cells was significantly reduced. The findings of this study indicate that sorafenib will show further improved antitumor efficacy against CRC due to overcoming resistance through the use of CaLa.
Collapse
|
14
|
Barbirou M, Woldu HG, Sghaier I, Bedoui SA, Mokrani A, Aami R, Mezlini A, Yacoubi-Loueslati B, Tonellato PJ, Bouhaouala-Zahar B. Western influenced lifestyle and Kv2.1 association as predicted biomarkers for Tunisian colorectal cancer. BMC Cancer 2020; 20:1086. [PMID: 33172410 PMCID: PMC7656678 DOI: 10.1186/s12885-020-07605-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 10/31/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is the third most diagnosed malignancy worldwide. The global burden is expected to increase along with ongoing westernized behaviors and lifestyle. The etiology of CRC remains elusive and most likely combines environmental and genetic factors. The Kv2.1 potassium channel encoded by KCNB1 plays a collection of roles in malignancy of cancer and may be a key factor of CRC susceptibility. Our study provides baseline association between Tunisian CRC and interactions between KCNB1 variants and lifestyle factors. METHODS A case-control study involving 300 CRC patients, and 300 controls was conducted Patients were carefully phenotyped and followed till the end of study. KCNB1 genotyping was confirmed by Sanger sequencing. Bivariate and multivariable logistic regression analyses were used to assess the clinical status, lifestyle and study polymorphisms association with CRC. RESULTS We noted significant gender association with CRC occurrence. Moreover, CRC risk increases with high meat and fat consumption, alcohol use and physical activity (PA). Carriage of rs1051296 A/G and both rs11468831 ins/del and del/del genotypes (p < 0.001) were significantly associated with CRC risk. Analysis according to gender reveals correlation of rs1051295 A/G, rs11468831 non ins/ins (p = 0.01) with CRC susceptibility regardless of patients' gender while rs3331 T/C (p = 0.012) was associated with females. Stratification study according to malignancy site; Rectal Cancer (RC) and Colon Cancer (CC), reveals increasing RC risk by gender and high meat and fat consumption, alcohol use and PA. However, additional association with high brine consumption was noted for CC. The rs1051295 A/G (p = 0.01) was associated with RC risk. Increased CC risk was associated with carriage of rs1051295 A/G, rs11168831 (del/del) and (ins/del) genotypes. CONCLUSION The risk of CRC increases with modifiable factors by Western influences on Tunisian lifestyle such as alcohol use, high fat consumption and possibly inadequate intake of vegetables. In addition, KCNB1 polymorphisms also markedly influence CRC susceptibility. Our study establishes key elements of a baseline characterization of clinical state, Western influenced lifestyle and KCNB1 variants associated with Tunisian CRC.
Collapse
Affiliation(s)
- Mouadh Barbirou
- Laboratory of Venoms and Therapeutic Biomolecules, LR16IPT08 Institute Pasteur of Tunis, University of Tunis El Manar, 13 Place Pasteur, BP74, 1002, Tunis, Belvédère, Tunisia.,Center for Biomedical Informatics, School of Medicine, University of Missouri, Columbia, MO, USA
| | - Henok G Woldu
- Department of Health Management and Informatics, Biostatistics & Research Design Unit School of Medicine, University of Missouri-Columbia, Columbia, MO, USA
| | - Ikram Sghaier
- Center for Biomedical Informatics, School of Medicine, University of Missouri, Columbia, MO, USA.,University of Tunis El Manar, Tunis, Tunisia
| | - Sinda A Bedoui
- Laboratory of Mycology Pathologies and Biomarkers Faculty of Sciences of Tunis, University of Tunis El Manar, Tunis, Tunisia.,Medical Oncology Division, Salah Azeiz Oncology Institute, University of Tunis El Manar, Tunis, Tunisia
| | - Amina Mokrani
- Medical School of Tunis, University of Tunis El Manar, Tunis, Tunisia
| | - Radhia Aami
- Laboratory of Venoms and Therapeutic Biomolecules, LR16IPT08 Institute Pasteur of Tunis, University of Tunis El Manar, 13 Place Pasteur, BP74, 1002, Tunis, Belvédère, Tunisia
| | - Amel Mezlini
- Medical School of Tunis, University of Tunis El Manar, Tunis, Tunisia
| | - Besma Yacoubi-Loueslati
- Laboratory of Mycology Pathologies and Biomarkers Faculty of Sciences of Tunis, University of Tunis El Manar, Tunis, Tunisia.,Medical Oncology Division, Salah Azeiz Oncology Institute, University of Tunis El Manar, Tunis, Tunisia
| | - Peter J Tonellato
- Center for Biomedical Informatics, School of Medicine, University of Missouri, Columbia, MO, USA
| | - Balkiss Bouhaouala-Zahar
- Laboratory of Venoms and Therapeutic Biomolecules, LR16IPT08 Institute Pasteur of Tunis, University of Tunis El Manar, 13 Place Pasteur, BP74, 1002, Tunis, Belvédère, Tunisia. .,Medical School of Tunis, University of Tunis El Manar, Tunis, Tunisia.
| |
Collapse
|
15
|
Han Y, Wang L, Wang Y. Integrated Analysis of Three Publicly Available Gene Expression Profiles Identified Genes and Pathways Associated with Clear Cell Renal Cell Carcinoma. Med Sci Monit 2020; 26:e919965. [PMID: 32712616 PMCID: PMC7405617 DOI: 10.12659/msm.919965] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Although advances have been achieved in the therapy of clear cell renal cell carcinoma (ccRCC), the pathogenesis of ccRCC is not yet fully understood. This study aimed to explore the critical genes and pathways associated with ccRCC by meta-analysis. MATERIAL AND METHODS We performed an integrated analysis of 3 publicly available microarray datasets developed from ccRCC tumor samples and normal tissues. A list of overlapped differentially expressed genes (DEGs) with the consistent expression trend in ccRCC tumor samples were identified, for which the protein-protein interaction (PPI) network was constructed, followed by topology structure and module analysis. The microRNA (miRNA) regulatory network and ccRCC associated pathway network were reconstructed. RESULTS A total of 504 genes were found to be consistently and differentially regulated based on 3 microarray datasets. The overrepresented pathways for DEGs included citric acid cycle (TCA cycle) and peroxisome proliferator-activated receptor (PPAR) signaling pathway and cell cycle. The PPI network was clustered into 6 modules that were closely related with the M phase, desmosome assembly, and response to hormone stimulus. The hsa04110: cell cycle and hsa04510: focal adhesion were the significant pathways associated with ccRCC overlapped with enrichment analysis. KDR and ITGB4 were focal-adhesion-associated genes, which were regulated by has-miR-424 and has-miR-204, respectively. CCND2 and CCNA2 were cell-cycle-associated genes, which were regulated by hsa-miR-324-3p, hsa-miR-146a and hsa-miR-145. CONCLUSIONS Cell cycle and focal adhesion were dysregulated in ccRCC, which were associated with the expression of CCND2, ITGB4, KDR, and CCNA2 genes. The deregulation of pathways and associated genes may provide insights to ccRCC research and therapy.
Collapse
Affiliation(s)
- YuPing Han
- Department of Urology, The Third Hospital of Jilin University, Changchun, Jilin, China (mainland)
| | - LinLin Wang
- Department of Ultrasound, The Third Hospital of Jilin University, Changchun, Jilin, China (mainland)
| | - Ye Wang
- Departmen of Pediatrics, The Third Hospital of Jilin University, Changchun, Jilin, China (mainland)
| |
Collapse
|
16
|
Palma S, Raffa CI, Garcia-Fabiani MB, Ferretti VA, Zwenger A, Perez Verdera PV, Llontop A, Rojas Bilbao E, Cuartero V, Abba MC, Lacunza E. RHBDD2 overexpression promotes a chemoresistant and invasive phenotype to rectal cancer tumors via modulating UPR and focal adhesion genes. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165810. [PMID: 32339641 DOI: 10.1016/j.bbadis.2020.165810] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2019] [Revised: 04/07/2020] [Accepted: 04/19/2020] [Indexed: 12/29/2022]
Abstract
The current standard of care for locally advanced rectal cancer (RC) is neoadjuvant radio-chemotherapy (NRC) with 5-fluorouracil (5Fu) as the main drug, followed by surgery and adjuvant chemotherapy. While a group of patients will achieve a pathological complete response, a significant percentage will not respond to the treatment. The Unfolding Protein Response (UPR) pathway is generally activated in tumors and results in resistance to radio-chemotherapy. We previously showed that RHBDD2 gene is overexpressed in the advanced stages of colorectal cancer (CRC) and that it could modulate the UPR pathway. Moreover, RHBDD2 expression is induced by 5Fu. In this study, we demonstrate that the overexpression of RHBDD2 in CACO2 cell line confers resistance to 5Fu, favors cell migration, adhesion and proliferation and has a profound impact on the expression of both, the UPR genes BiP, PERK and CHOP, and on the cell adhesion genes FAK and PXN. We also determined that RHBDD2 binds to BiP protein, the master UPR regulator. Finally, we confirmed that a high expression of RHBDD2 in RC tumors after NRC treatment is associated with the development of local or distant metastases. The collected evidence positions RHBDD2 as a promising prognostic biomarker to predict the response to neoadjuvant therapy in patients with RC.
Collapse
Affiliation(s)
- S Palma
- Centro de Investigaciones Inmunológicas Básicas y Aplicadas, Facultad de Ciencias Médicas, Universidad Nacional de La Plata, La Plata, Buenos Aires, Argentina
| | - C I Raffa
- Gastroenterology and Proctology Department, Instituto de Oncología Angel H. Roffo, University of Buenos Aires, Buenos Aires, Argentina
| | - M B Garcia-Fabiani
- Instituto de Investigaciones Bioquímicas de La Plata Rodolfo R. Brenner, CONICET, Facultad de Ciencias Médicas, Universidad Nacional de La Plata, La Plata, Buenos Aires, Argentina
| | - V A Ferretti
- Centro de Investigaciones Inmunológicas Básicas y Aplicadas, Facultad de Ciencias Médicas, Universidad Nacional de La Plata, La Plata, Buenos Aires, Argentina
| | - A Zwenger
- Grupo Oncológico Cooperativo del Sur (GOCS), Neuquén, Argentina
| | | | - A Llontop
- Pathology Department, Instituto de Oncología Angel H. Roffo, University of Buenos Aires, Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - E Rojas Bilbao
- Pathology Department, Instituto de Oncología Angel H. Roffo, University of Buenos Aires, Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - V Cuartero
- Clinic Oncology Department, Functional Unit of Digestive Tumors, Instituto de Oncología Angel H. Roffo, University of Buenos Aires, Buenos Aires, Argentina
| | - M C Abba
- Centro de Investigaciones Inmunológicas Básicas y Aplicadas, Facultad de Ciencias Médicas, Universidad Nacional de La Plata, La Plata, Buenos Aires, Argentina
| | - E Lacunza
- Centro de Investigaciones Inmunológicas Básicas y Aplicadas, Facultad de Ciencias Médicas, Universidad Nacional de La Plata, La Plata, Buenos Aires, Argentina.
| |
Collapse
|
17
|
Sun C, Hu A, Wang S, Tian B, Jiang L, Liang Y, Wang H, Dong J. ADAM17-regulated CX3CL1 expression produced by bone marrow endothelial cells promotes spinal metastasis from hepatocellular carcinoma. Int J Oncol 2020; 57:249-263. [PMID: 32319605 PMCID: PMC7252465 DOI: 10.3892/ijo.2020.5045] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 03/30/2020] [Indexed: 12/13/2022] Open
Abstract
Spinal metastasis occurs in 50-75% of bone metastases caused by hepatocellular carcinoma (HCC), and HCC-derived spinal metastasis can lead to a less favorable prognosis. Recently, several studies have demonstrated that C-X3-C motif chemokine ligand 1 (CX3CL1) is closely associated with cancer metastasis, and its secretion is modulated by a disintegrin and metalloproteinase 17 (ADAM17). Bone marrow endothelial cells (BMECs) are an essential component of bone marrow. However, little is known about the roles in and effects of BMECs on HCC spinal metastasis. The present study demonstrated that CX3CL1 and C-X-C motif chemokine receptor 3 (CXCR3) expression was upregulated in HCC spinal metastases, and that CX3CL1 promoted the migration and invasion of HCC cells to the spine. Western blot analysis revealed that the Src/protein tyrosine kinase 2 (PTK2) axis participated in CX3CL1-induced HCC cell invasion and migration. CX3CL1 also increased the expression of M2 macrophage markers in THP-1 monocytes. BMECs promoted the migration and invasion of Hep3B and MHCC97H cells by secreting soluble CX3CL1, whereas the neutralization of CX3CL1 inhibited this enhancement. CX3CL1 enhanced the activation of the phosphatidylinositol-4,5-bisphos-phate 3-kinase catalytic subunit alpha (PIK3CA)/AKT serine/threonine kinase 1 (AKT1) and Ras homolog family member A (RHOA)/Rho associated coiled-coil containing protein kinase 2 (ROCK2) signaling pathways through the Src/PTK2 signaling pathway. Furthermore, ADAM17 was activated by mitogen-activated protein kinase (MAPK) z14 in BMECs and significantly promoted the secretion of CX3CL1. HCC cells enhanced the recruitment and proliferation of BMECs. The overexpression of CX3CR1 facilitated the spinal metastasis of HCC in a mouse model in vivo. In addition, in vivo experiments revealed that BMECs promoted the growth of HCC in the spine. The present study demonstrated that CX3CL1 participates in HCC spinal metastasis, and that BMECs play an important role in the regulation of CX3CL1 in the spinal metastatic environment.
Collapse
Affiliation(s)
- Chi Sun
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| | - Annan Hu
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| | - Shengxing Wang
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| | - Bo Tian
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| | - Libo Jiang
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| | - Yun Liang
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| | - Houlei Wang
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| | - Jian Dong
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| |
Collapse
|
18
|
Yang ZD, Kang H. Exploring prognostic potential of long noncoding RNAs in colorectal cancer based on a competing endogenous RNA network. World J Gastroenterol 2020; 26:1298-1316. [PMID: 32256018 PMCID: PMC7109275 DOI: 10.3748/wjg.v26.i12.1298] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Revised: 01/08/2020] [Accepted: 03/09/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is one of the most prevalent tumors worldwide. Recently, long noncoding RNAs (lncRNAs) have been shown to influence tumorigenesis and tumor progression by acting as competing endogenous RNAs (ceRNAs). It is difficult to extract prognostic lncRNAs and useful bioinformation from most ceRNA networks constructed previously.
AIM To construct a prognostic related ceRNA regulatory network and lncRNA related signature based on risk score in CRC.
METHODS RNA transcriptome profile and clinical information of 506 CRC patients were downloaded from the Cancer Genome Atlas database. R packages and Perl program were used for data processing. Cox regression analysis was used for prognostic model construction. Quantitative real-time polymerase chain reaction was used to detect the expression of lncRNAs.
RESULTS A prognostic-related ceRNA network was constructed, including 9 lncRNAs, 44 mRNAs, and 30 miRNAs. In addition, a four-lncRNA model was constructed using multivariate Cox regression analysis, which could be an independent prognostic model in CRC. The risk score for each patient was calculated, and the 506 patients were divided into high and low-risk groups (253 for each group) based on the median risk score. The results of the survival analysis showed that patients with a high-risk score had a poor survival rate. Furthermore, the predictive value of the four-lncRNA model was evaluated in GSE38832. Patient survival probabilities could be better predicted when combing the risk score and clinical features. Gene Set Enrichment Analysis results verified that a number of cancer-related signaling pathways were enriched with a high-risk score in CRC. Finally, we validated a novel lncRNA (LINC00488) using quantitative real-time polymerase chain reaction in 22 paired CRC patient tumor tissues compared to adjacent non-tumor tissues.
CONCLUSION The four-lncRNA model could give better predictive value for CRC patients. Our understanding of the lncRNA-related ceRNA regulatory mechanism could provide a potential diagnostic indicator for CRC patients.
Collapse
Affiliation(s)
- Zhi-Dong Yang
- Department of Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang 110001, Liaoning Province, China
| | - Hui Kang
- Department of Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang 110001, Liaoning Province, China
| |
Collapse
|
19
|
Meng NN, Zhang RR, Liu C, Wang Q, Wang XK, Guo X, Wang PP, Sun JY. PDB-1 from Potentilla discolor Bunge suppresses lung cancer cell migration and invasion via FAK/Src and MAPK signaling pathways. Med Chem Res 2020. [DOI: 10.1007/s00044-020-02527-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
20
|
Extracellular matrix-cell interactions: Focus on therapeutic applications. Cell Signal 2019; 66:109487. [PMID: 31778739 DOI: 10.1016/j.cellsig.2019.109487] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 11/25/2019] [Accepted: 11/25/2019] [Indexed: 02/06/2023]
Abstract
Extracellular matrix (ECM) macromolecules together with a multitude of different molecules residing in the extracellular space play a vital role in the regulation of cellular phenotype and behavior. This is achieved via constant reciprocal interactions between the molecules of the ECM and the cells. The ECM-cell interactions are mediated via cell surface receptors either directly or indirectly with co-operative molecules. The ECM is also under perpetual remodeling process influencing cell-signaling pathways on its part. The fragmentation of ECM macromolecules provides even further complexity for the intricate environment of the cells. However, as long as the interactions between the ECM and the cells are in balance, the health of the body is retained. Alternatively, any dysregulation in these interactions can lead to pathological processes and finally to various diseases. Thus, therapeutic applications that are based on retaining normal ECM-cell interactions are highly rationale. Moreover, in the light of the current knowledge, also concurrent multi-targeting of the complex ECM-cell interactions is required for potent pharmacotherapies to be developed in the future.
Collapse
|
21
|
Yao Y, Liu R, Gao C, Zhang T, Qi L, Liu G, Zhang W, Wang X, Li J, Li J, Sun C. Identification of prognostic biomarkers for breast cancer based on miRNA and mRNA co-expression network. J Cell Biochem 2019; 120:15378-15388. [PMID: 31037764 DOI: 10.1002/jcb.28805] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 01/28/2019] [Accepted: 01/30/2019] [Indexed: 12/31/2022]
Abstract
PURPOSE Breast cancer (BC) remains a serious health threat for women due to its high incidence and the trend of rejuvenation. Accumulating evidence has highlighted that microRNAs (miRNAs) and messenger RNAs (mRNAs) could play important roles in various biological processes involved in the pathogenesis of BC. The present study aimed to identify potential prognostic biomarkers associated with BC. METHODS Here, original gene expression profiles of patients with BC was downloaded from The Cancer Genome Atlas (TCGA) database. TargetScan, miRDB, and miRTarBase databases were used to predict the target genes of prognostic-related differentially expressed miRNAs (DEMs). Subsequently, functional enrichment analysis and topological analysis were performed on the overlaps of target genes and differentially expressed mRNAs (DEGs), and Kaplan-Meier analysis was used to predict prognosis-related target genes to identify prognostic biomarkers. RESULTS A total of 218 DEMs and 2222 DEGs were extracted in which eight miRNAs were associated with prognosis, and 278 target DEGs were screened out incorporated into functional enrichment analysis and protein-protein interaction network visualization studies. Additionally, five hub genes (CXCL12, IGF1, LEF1, MMP1, and RACGAP1) were observed as potential biomarkers for BC prognosis through survival analysis. CONCLUSION We performed a distinctive correlation analysis of miRNA-mRNA in BC patients, and identified eight miRNAs and five hub genes may be effective biomarkers for the prognosis of BC patients.
Collapse
Affiliation(s)
- Yan Yao
- Clinical Medical Colleges, Weifang Medical University, Weifang, Shandong, People's Republic of China
| | - Ruijuan Liu
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, Shandong, People's Republic of China
| | - Chundi Gao
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, People's Republic of China
| | - Tingting Zhang
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, People's Republic of China
| | - Lingyu Qi
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, People's Republic of China
| | - Gongxi Liu
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, Shandong, People's Republic of China
| | - Wenfeng Zhang
- Clinical Medical Colleges, Weifang Medical University, Weifang, Shandong, People's Republic of China
| | - Xue Wang
- Medical Colleges, Qingdao University, Qingdao, Shandong, People's Republic of China
| | - Jie Li
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, People's Republic of China
| | - Jia Li
- Clinical Medical Colleges, Weifang Medical University, Weifang, Shandong, People's Republic of China
| | - Changgang Sun
- Department of Oncology, Affiliated Hospital of Weifang Medical University, Weifang, Shandong, People's Republic of China.,Department of Oncology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, People's Republic of China
| |
Collapse
|
22
|
Gargalionis AN, Basdra EK, Papavassiliou AG. Polycystins in Colorectal Cancer. Int J Mol Sci 2018; 20:104. [PMID: 30597875 PMCID: PMC6337659 DOI: 10.3390/ijms20010104] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 12/20/2018] [Accepted: 12/25/2018] [Indexed: 12/18/2022] Open
Abstract
Cell and extracellular matrix (ECM) biomechanics emerge as a distinct feature during the development and progression of colorectal cancer (CRC). Polycystins are core mechanosensitive protein molecules that mediate mechanotransduction in a variety of epithelial cells. Polycystin-1 (PC1) and polycystin-2 (PC2) are engaged in signal transduction mechanisms and during alterations in calcium influx, which regulate cellular functions such as proliferation, differentiation, orientation, and migration in cancer cells. Recent findings implicate polycystins in the deregulation of such functions and the formation of CRC invasive phenotypes. Polycystins participate in all aspects of the cell's biomechanical network, from the perception of extracellular mechanical cues to focal adhesion protein and nuclear transcriptional complexes. Therefore, polycystins could be employed as novel biomarkers and putative targets of selective treatment in CRC.
Collapse
Affiliation(s)
- Antonios N Gargalionis
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece.
| | - Efthimia K Basdra
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece.
| | - Athanasios G Papavassiliou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece.
| |
Collapse
|