1
|
Wu Y, Guo Z, Li Z, Cai C, Liu J, Tang X, Que L. Effects of Integrin-Linked Kinase Silencing Combined With Trichostatin A on Cancer Stem Cells. Oral Dis 2025. [PMID: 40364491 DOI: 10.1111/odi.15377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 02/13/2025] [Accepted: 04/26/2025] [Indexed: 05/15/2025]
Abstract
OBJECTIVES Oral squamous cell carcinoma (OSCC) is characterized by high invasiveness and metastasis, with cancer stem cells (CSCs) playing a central role in tumor progression. This study investigates the effects of integrin-linked kinase (ILK) silencing and trichostatin A (TSA) treatment on CSCs, assessing their potential to diminish CSC properties and inhibit OSCC progression. METHODS AND MATERIALS CSCs were enriched and isolated from primary OSCC samples and Tca8113 cell line and MOC1 cell line using side population (SP) analysis, with their characteristics and the therapeutic impact of treatments assessed through assays such as MTT, wound healing, cell invasion, cell cycle, and apoptosis. RESULTS Higher SP cell content correlated significantly with poor pathological classification, metastasis, and recurrence. Treated CSCs showed reduced proliferation, migration, and invasion, along with increased apoptosis. In vivo experiments demonstrated that the combined treatment substantially reduced tumor growth. CONCLUSION The study confirms the efficacy of targeting CSCs with ILK silencing and TSA treatment in OSCC, suggesting a promising strategy for CSC-directed therapies that merit further investigation.
Collapse
Affiliation(s)
- Yulu Wu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Zhiyong Guo
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Zhangao Li
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Chenchen Cai
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Jiyuan Liu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Xiufa Tang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Lin Que
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
2
|
Ahmed H, Ali H, Ahmed KM. Expression Profiles of Integrin-Linked Kinase, Vascular Endothelial Growth Factor A, and Ephrin Type-A Receptor 2 in Colorectal Cancer Lymph Nodes. Cureus 2024; 16:e71242. [PMID: 39525153 PMCID: PMC11550453 DOI: 10.7759/cureus.71242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/11/2024] [Indexed: 11/16/2024] Open
Abstract
Background Colorectal cancer (CRC) is the third most frequently diagnosed cancer globally, with a high incidence of morbidity and mortality. Early diagnosis of CRC is crucial for determining appropriate treatment regimens, thereby prolonging overall survival rates and improving prognostic outcomes. Objectives This study aimed to investigate the expression profiles of specific proteins in the lymph nodes (LNs) of CRC patients, including integrin-linked kinase (ILK), vascular endothelial growth factor A (VEGFA), and ephrin type-A receptor 2 (EphA2), through immunohistochemical studies. Methods The study involved a sample of 76 patients clinically diagnosed with CRC who were referred by their specialist oncologist. Tumor staging was determined based on the TNM classification. Immunohistochemical studies were conducted to measure the expression patterns of the candidate markers (ILK, EphA2, and VEGFA). Expression levels were scored as negative, low, or high. Results The highest percentage of CRC patients were diagnosed with conventional adenocarcinoma, predominantly in stages II and III. Of the 76 CRC tissue specimens, the majority (46, 60.52%) tested negative for lymphatic invasion, while the remaining 30 (39.47%) tested positive. According to the TNM classification, 14 samples had N1 (one invaded LN), and 16 had N2 (two or more invaded LNs). Furthermore, the percentage of patients with low and high EphA2 expression was significantly higher (p<0.0001) compared to the negative expression controls. Regarding ILK, 15 cases (50.0%) showed negative expression, while an equal number displayed positive expression. Additionally, the group with low VEGFA expression was statistically significantly higher (p=0.01) compared to the negative control. Conclusion The expression levels of EphA2, ILK, and VEGFA were found to be higher in LNs with lymphatic invasion compared to those with negative expression, highlighting the role of these proteins in CRC progression.
Collapse
Affiliation(s)
- Hayat Ahmed
- College of Medicine, University of Duhok, Duhok, IRQ
| | - Hazhmat Ali
- College of Medicine, University of Duhok, Duhok, IRQ
| | | |
Collapse
|
3
|
Wu J, Li W, Su J, Zheng J, Liang Y, Lin J, Xu B, Liu Y. Integration of single-cell sequencing and bulk RNA-seq to identify and develop a prognostic signature related to colorectal cancer stem cells. Sci Rep 2024; 14:12270. [PMID: 38806611 PMCID: PMC11133358 DOI: 10.1038/s41598-024-62913-3] [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: 01/08/2024] [Accepted: 05/22/2024] [Indexed: 05/30/2024] Open
Abstract
The prognosis for patients with colorectal cancer (CRC) remains worse than expected due to metastasis, recurrence, and resistance to chemotherapy. Colorectal cancer stem cells (CRCSCs) play a vital role in tumor metastasis, recurrence, and chemotherapy resistance. However, there are currently no prognostic markers based on CRCSCs-related genes available for clinical use. In this study, single-cell transcriptome sequencing was employed to distinguish cancer stem cells (CSCs) in the CRC microenvironment and analyze their properties at the single-cell level. Subsequently, data from TCGA and GEO databases were utilized to develop a prognostic risk model for CRCSCs-related genes and validate its diagnostic performance. Additionally, functional enrichment, immune response, and chemotherapeutic drug sensitivity of the relevant genes in the risk model were investigated. Lastly, the key gene RPS17 in the risk model was identified as a potential prognostic marker and therapeutic target for further comprehensive studies. Our findings provide new insights into the prognostic treatment of CRC and offer novel perspectives for a systematic and comprehensive understanding of CRC development.
Collapse
Affiliation(s)
- Jiale Wu
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, School of Pharmacy, Guangdong Medical University, Zhanjiang, 524023, Guangdong, China
| | - Wanyu Li
- Well Lead Medical Co., Ltd., Guangzhou, 511434, Guangdong, China
| | - Junyu Su
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, School of Pharmacy, Guangdong Medical University, Zhanjiang, 524023, Guangdong, China
| | - Jiamin Zheng
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, School of Pharmacy, Guangdong Medical University, Zhanjiang, 524023, Guangdong, China
| | - Yanwen Liang
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, School of Pharmacy, Guangdong Medical University, Zhanjiang, 524023, Guangdong, China
| | - Jiansuo Lin
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Guangdong Medical University, Dongguan, 523808, Guangdong, China
| | - Bilian Xu
- School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, 524023, Guangdong, China.
| | - Yi Liu
- School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, 524023, Guangdong, China.
| |
Collapse
|
4
|
Murase H, Matsuo Y, Denda Y, Nonoyama K, Kato T, Aoyama Y, Hayashi Y, Imafuji H, Saito K, Morimoto M, Ogawa R, Takahashi H, Mitsui A, Kimura M, Takiguchi S. Upregulation of integrin‑linked kinase enhances tumor progression in gemcitabine‑resistant pancreatic cancer. Oncol Rep 2023; 50:164. [PMID: 37477162 PMCID: PMC10394728 DOI: 10.3892/or.2023.8601] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 05/18/2023] [Indexed: 07/22/2023] Open
Abstract
Pancreatic cancer (PaCa) tends to be resistant to chemotherapy and is associated with a very poor prognosis. It has been previously reported by the authors that integrin‑linked kinase (ILK) is a prognostic factor in PaCa. ILK expression was examined in a newly established gemcitabine (Gem)‑resistant (Gem‑R) PaCa cell line and it was demonstrated that ILK expression was upregulated compared with that in Gem‑sensitive (Gem‑S) cells. In the present study, the effects of increased ILK expression in Gem‑R PaCa cells were evaluated and it was examined whether compound 22 (Cpd22), an ILK inhibitor, exerted antitumor effects not only in Gem‑S cells but also in Gem‑R cells. Reverse transcription‑quantitative polymerase chain reaction and western blotting revealed that ILK expression was higher in Gem‑R PaCa cells than in Gem‑S PaCa cells. Cpd22 inhibited the growth of PaCa cells in a concentration‑dependent manner. Cpd22 also inhibited the growth of Gem‑R PaCa cells. The invasive and angiogenic potential of Gem‑R PaCa cells was enhanced compared with that in Gem‑S cells; however, ILK small interfering RNA and Cpd22 treatment suppressed this enhancement of invasive potential compared with that in Gem‑S cells. The addition of Cpd22 to Gem also improved the sensitivity of Gem‑R cell lines to Gem. Furthermore, enhanced Akt signaling was associated with increased malignancy in Gem‑R cell lines. In conclusion, ILK was upregulated with resistance and may be involved in tumor angiogenesis, invasive potential, and chemotherapy resistance, which were all suppressed by Cpd22 treatment. Thus, Cpd22 may be a novel therapeutic agent for the treatment of PaCa.
Collapse
Affiliation(s)
- Hiromichi Murase
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Mizuho-cho, Mizuho-ku, Nagoya, Aichi 4678601, Japan
| | - Yoichi Matsuo
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Mizuho-cho, Mizuho-ku, Nagoya, Aichi 4678601, Japan
| | - Yuki Denda
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Mizuho-cho, Mizuho-ku, Nagoya, Aichi 4678601, Japan
| | - Keisuke Nonoyama
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Mizuho-cho, Mizuho-ku, Nagoya, Aichi 4678601, Japan
| | - Tomokatsu Kato
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Mizuho-cho, Mizuho-ku, Nagoya, Aichi 4678601, Japan
| | - Yoshinaga Aoyama
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Mizuho-cho, Mizuho-ku, Nagoya, Aichi 4678601, Japan
| | - Yuichi Hayashi
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Mizuho-cho, Mizuho-ku, Nagoya, Aichi 4678601, Japan
| | - Hiroyuki Imafuji
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Mizuho-cho, Mizuho-ku, Nagoya, Aichi 4678601, Japan
| | - Kenta Saito
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Mizuho-cho, Mizuho-ku, Nagoya, Aichi 4678601, Japan
| | - Mamoru Morimoto
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Mizuho-cho, Mizuho-ku, Nagoya, Aichi 4678601, Japan
| | - Ryo Ogawa
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Mizuho-cho, Mizuho-ku, Nagoya, Aichi 4678601, Japan
| | - Hiroki Takahashi
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Mizuho-cho, Mizuho-ku, Nagoya, Aichi 4678601, Japan
| | - Akira Mitsui
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Mizuho-cho, Mizuho-ku, Nagoya, Aichi 4678601, Japan
| | - Masahiro Kimura
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Mizuho-cho, Mizuho-ku, Nagoya, Aichi 4678601, Japan
| | - Shuji Takiguchi
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Mizuho-cho, Mizuho-ku, Nagoya, Aichi 4678601, Japan
| |
Collapse
|
5
|
Xu M, Yu J, Liu X, Jia W, Duan Y, Ma D, Ma J, Lei W, Tai W. METTL3 regulatory TROAP can regulate the progression of non-small cell lung cancer through PI3K/AKT and EMT signaling pathway. Med Oncol 2023; 40:274. [PMID: 37608033 DOI: 10.1007/s12032-023-02143-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 07/29/2023] [Indexed: 08/24/2023]
Abstract
TROAP, interacts with trophinin and bystin, polys a key role in embryo implantation. TROAP is required for spindle assembly and centrosome integrity during the mitosis. TROAP has been described to promote tumorigenesis in a diverse range of cancer. We performed this study to assess the biological and clinical significance of TROAP in Non-small cell lung cancer. Forty-eight pairs of lung adenocarcinoma (LUAD) tissues and paraneoplastic tissues were collected. RT-qPCR, western bolt and immunohistochemistry assay was used to test TROAP RNA and protein expression not in LUAD tissues and paraneoplastic tissues but in LUAD cell lines and control cell lines. TROAP knockdown and overexpression vector were constructed and transfected into lung cancer cells. CCK-8, transwell, and wound healing assays were used to assess cell viability, migration, and invasion. The expression of PI3K/AKT and EMT signaling proteins and METTL3 were determined by western blot. We found the TROAP was enriched in NSCLC tissues and cell lines. TROAP knockdown inhibited cell proliferation, migration, invasion compared with control group in NSCLC. Mechanism analysis revealed that TROAP activated PI3K/AKT and EMT signaling pathway. To a certain extent, TROAP was regulated by METTL3. In a word, TROAP accelerated the progression of NSCLC through PI3K/AKT and EMT pathway, and TROAP might be considered as a novel target for NSCLC therapy.
Collapse
Affiliation(s)
- Muli Xu
- Department of Clinical Laboratory, Yunnan Molecular Diagnostic Center, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Jiankun Yu
- Chinese Academy of Medical Sciences and Institute of Medical Biology, Peking Union Medical College, Kunming, China
| | - Xiaoxiao Liu
- Department of Clinical Laboratory, Yunnan Molecular Diagnostic Center, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Wanting Jia
- Department of Clinical Laboratory, Yunnan Molecular Diagnostic Center, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Yu Duan
- Department of Clinical Laboratory, Yunnan Molecular Diagnostic Center, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Di Ma
- Department of Clinical Laboratory, Yunnan Molecular Diagnostic Center, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Jiaxuan Ma
- Department of Clinical Laboratory, Yunnan Molecular Diagnostic Center, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Wanyang Lei
- Department of Clinical Laboratory, Yunnan Molecular Diagnostic Center, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Wenlin Tai
- Department of Clinical Laboratory, Yunnan Molecular Diagnostic Center, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China.
| |
Collapse
|
6
|
Jones J, Shi Q, Nath RR, Brito IL. Keystone pathobionts associated with colorectal cancer promote oncogenic reprograming. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.03.535410. [PMID: 37066368 PMCID: PMC10103987 DOI: 10.1101/2023.04.03.535410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Fusobacterium nucleatum (Fn) and enterotoxigenic Bacteroides fragilis (ETBF) are two pathobionts consistently enriched in the gut microbiomes of patients with colorectal cancer (CRC) compared to healthy counterparts and frequently observed for their direct association within tumors. Although several molecular mechanisms have been identified that directly link these organisms to features of CRC in specific cell types, their specific effects on the epithelium and local immune compartment are not well-understood. To fill this gap, we leveraged single-cell RNA sequencing (scRNA-seq) on wildtype mice and mouse model of CRC. We find that Fn and ETBF exacerbate cancer-like transcriptional phenotypes in transit-amplifying and mature enterocytes in a mouse model of CRC. We also observed increased T cells in the pathobiont-exposed mice, but these pathobiont-specific differences observed in wildtype mice were abrogated in the mouse model of CRC. Although there are similarities in the responses provoked by each organism, we find pathobiont-specific effects in Myc-signaling and fatty acid metabolism. These findings support a role for Fn and ETBF in potentiating tumorigenesis via the induction of a cancer stem cell-like transit-amplifying and enterocyte population and the disruption of CTL cytotoxic function.
Collapse
Affiliation(s)
- Josh Jones
- Meinig School for Biomedical Engineering, Cornell University, Ithaca, NY
| | - Qiaojuan Shi
- Meinig School for Biomedical Engineering, Cornell University, Ithaca, NY
| | - Rahul R. Nath
- Meinig School for Biomedical Engineering, Cornell University, Ithaca, NY
| | - Ilana L. Brito
- Meinig School for Biomedical Engineering, Cornell University, Ithaca, NY
| |
Collapse
|
7
|
Liu J, Ma X, Cao L, Wei Y, Gao Y, Qu C, Maimaitiming N, Zhang L. Computational Drug Repurposing Approach to Identify Novel Inhibitors of ILK Protein for Treatment of Esophageal Squamous Cell Carcinoma. JOURNAL OF ONCOLOGY 2022; 2022:3658334. [PMID: 36618074 PMCID: PMC9815933 DOI: 10.1155/2022/3658334] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Revised: 11/19/2022] [Accepted: 12/03/2022] [Indexed: 12/31/2022]
Abstract
Purpose Esophageal squamous cell cancer (ESCC) is a deadly malignant tumor characterized by an overall 5-year survival rate below 20%, with China accounting for approximately 50% of all cases worldwide. Our previous studies have demonstrated that high integrin-linked kinase (ILK) expression plays a key role in development and progression of ESCC both in vitro and in vivo. Here, we employed the drug repurposing approach to identify a novel FDA-approved anticancer inhibitor against ILK-induced tumorigenesis and progression. Methods We screened the ZINC15 database and predicted the molecular docking ability among FDA-approved and publicly available drugs to ILK and then performed computational docking and visual inspection analyses of the top 10 ranked drugs. Two computer-based virtual screened drugs were evaluated in vitro for their ability to directly bind purified ILK by surface plasmon resonance. Cytotoxicity of the two candidate drugs was validated in vitro using CCK-8 and LDH assays. Results We initially selected the top 10 compounds, based on their minimum binding energy to the ILK crystal, after molecular docking and subjected them to further screening. Taking the binding energy of -10 kcal/mol as the threshold, we selected two drugs, namely, nilotinib and teniposide, for the wet-lab experiment. Surface plasmon resonance (SPR) revealed that nilotinib and teniposide had equilibrium dissociation constant (KD) values of 6.410E - 6 and 1.793E - 6, respectively, which were lower than 2.643E - 6 observed in ILK-IN-3 used as the positive control. The IC50 values for nilotinib and teniposide in ESCC cell lines were 40 μM and 200-400 nM, respectively. Results of the CCK-8 assay demonstrated that both nilotinib and teniposide significantly inhibited proliferation of cells (P < 0.01). LDH results revealed that both drugs significantly suppressed the rate of cell death (P < 0.01). Conclusion The drug repositioning procedure can effectively identify new therapeutic tools for ESCC. Our findings suggest that nilotinib and teniposide are efficacious inhibitors of ILK and thus have potential to target ILK-mediated signaling pathways for management of ESCC.
Collapse
Affiliation(s)
- Juan Liu
- Department of General Medicine of Healthy Care Center for Cadres, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region, China
| | - Xiaoli Ma
- Department of General Medicine of Healthy Care Center for Cadres, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region, China
| | - Leiyu Cao
- Department of General Medicine of Healthy Care Center for Cadres, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region, China
| | - Yu Wei
- Department of General Medicine of Healthy Care Center for Cadres, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region, China
| | - Yan Gao
- Department of General Medicine of Healthy Care Center for Cadres, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region, China
| | - Chengcheng Qu
- Department of General Medicine of Healthy Care Center for Cadres, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region, China
| | - Nuersimanguli Maimaitiming
- Department of General Medicine of Healthy Care Center for Cadres, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region, China
| | - Li Zhang
- Department of General Medicine of Healthy Care Center for Cadres, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region, China
| |
Collapse
|
8
|
Di Z, Zhou S, Xu G, Ren L, Li C, Ding Z, Huang K, Liang L, Yuan Y. Single-cell and WGCNA uncover a prognostic model and potential oncogenes in colorectal cancer. Biol Proced Online 2022; 24:13. [PMID: 36117173 PMCID: PMC9484253 DOI: 10.1186/s12575-022-00175-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Accepted: 08/30/2022] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is one of the leading causes of cancer-related death worldwide. Single-cell transcriptome sequencing (scRNA-seq) can provide accurate gene expression data for individual cells. In this study, a new prognostic model was constructed by scRNA-seq and bulk transcriptome sequencing (bulk RNA-seq) data of CRC samples to develop a new understanding of CRC. METHODS CRC scRNA-seq data were downloaded from the GSE161277 database, and CRC bulk RNA-seq data were downloaded from the TCGA and GSE17537 databases. The cells were clustered by the FindNeighbors and FindClusters functions in scRNA-seq data. CIBERSORTx was applied to detect the abundance of cell clusters in the bulk RNA-seq expression matrix. WGCNA was performed with the expression profiles to construct the gene coexpression networks of TCGA-CRC. Next, we used a tenfold cross test to construct the model and a nomogram to assess the independence of the model for clinical application. Finally, we examined the expression of the unreported model genes by qPCR and immunohistochemistry. A clone formation assay and orthotopic colorectal tumour model were applied to detect the regulatory roles of unreported model genes. RESULTS A total of 43,851 cells were included after quality control, and 20 cell clusters were classified by the FindCluster () function. We found that the abundances of C1, C2, C4, C5, C15, C16 and C19 were high and the abundances of C7, C10, C11, C13, C14 and C17 were low in CRC tumour tissues. Meanwhile, the results of survival analysis showed that high abundances of C4, C11 and C13 and low abundances of C5 and C14 were associated with better survival. The WGCNA results showed that the red module was most related to the tumour and the C14 cluster, which contains 615 genes. Lasso Cox regression analysis revealed 8 genes (PBXIP1, MPMZ, SCARA3, INA, ILK, MPP2, L1CAM and FLNA), which were chosen to construct a risk model. In the model, the risk score features had the greatest impact on survival prediction, indicating that the 8-gene risk model can better predict prognosis. qPCR and immunohistochemistry analysis showed that the expression levels of MPZ, SCARA3, MPP2 and PBXIP1 were high in CRC tissues. The functional experiment results indicated that MPZ, SCARA3, MPP2 and PBXIP1 could promote the colony formation ability of CRC cells in vitro and tumorigenicity in vivo. CONCLUSIONS We constructed a risk model to predict the prognosis of CRC patients based on scRNA-seq and bulk RNA-seq data, which could be used for clinical application. We also identified 4 previously unreported model genes (MPZ, SCARA3, MPP2 and PBXIP1) as novel oncogenes in CRC. These results suggest that this model could potentially be used to evaluate the prognostic risk and provide potential therapeutic targets for CRC patients.
Collapse
Affiliation(s)
- Ziyang Di
- Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Sicheng Zhou
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Gaoran Xu
- Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Lian Ren
- Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Chengxin Li
- Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Zheyu Ding
- Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Kaixin Huang
- Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Leilei Liang
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yihang Yuan
- Department of General Surgery, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, 1111 XianXia Road, Shanghai, 200336 China
| |
Collapse
|
9
|
Novoa Díaz MB, Carriere P, Gigola G, Zwenger AO, Calvo N, Gentili C. Involvement of Met receptor pathway in aggressive behavior of colorectal cancer cells induced by parathyroid hormone-related peptide. World J Gastroenterol 2022; 28:3177-3200. [PMID: 36051345 PMCID: PMC9331538 DOI: 10.3748/wjg.v28.i26.3177] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 03/21/2022] [Accepted: 05/28/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Parathyroid hormone-related peptide (PTHrP) plays a key role in the development and progression of many tumors. We found that in colorectal cancer (CRC) HCT116 cells, the binding of PTHrP to its receptor PTHR type 1 (PTHR1) activates events associated with an aggressive phenotype. In HCT116 cell xenografts, PTHrP modulates the expression of molecular markers linked to tumor progression. Empirical evidence suggests that the Met receptor is involved in the development and evolution of CRC. Based on these data, we hypothesized that the signaling pathway trigged by PTHrP could be involved in the transactivation of Met and consequently in the aggressive behavior of CRC cells.
AIM To elucidate the relationship among PTHR1, PTHrP, and Met in CRC models.
METHODS For in vitro assays, HCT116 and Caco-2 cells derived from human CRC were incubated in the absence or presence of PTHrP (1-34) (10-8 M). Where indicated, cells were pre-incubated with specific kinase inhibitors or dimethylsulfoxide, the vehicle of the inhibitors. The protein levels were evaluated by Western blot technique. Real-time polymerase chain reaction (RT-qPCR) was carried out to determine the changes in gene expression. Wound healing assay and morphological monitoring were performed to evaluate cell migration and changes related to the epithelial-mesenchymal transition (EMT), respectively. The number of viable HCT116 cells was counted by trypan blue dye exclusion test to evaluate the effects of irinotecan (CPT-11), oxaliplatin (OXA), or doxorubicin (DOXO) with or without PTHrP. For in vivo tests, HCT116 cell xenografts on 6-wk-old male N:NIH (S)_nu mice received daily intratumoral injections of PTHrP (40 μg/kg) in 100 μL phosphate-buffered saline (PBS) or the vehicle (PBS) as a control during 20 d. Humanitarian slaughter was carried out and the tumors were removed, weighed, and fixed in a 4% formaldehyde solution for subsequent treatment by immunoassays. To evaluate the expression of molecular markers in human tumor samples, we studied 23 specimens obtained from CRC patients which were treated at the Hospital Interzonal de Graves y Agudos Dr. José Penna (Bahía Blanca, Buenos Aires, Argentina) and the Hospital Provincial de Neuquén (Neuquén, Neuquén, Argentina) from January 1990 to December 2007. Seven cases with normal colorectal tissues were assigned to the control group. Tumor tissue samples and clinical histories of patients were analyzed. Paraffin-embedded blocks from primary tumors were reviewed by hematoxylin-eosin staining technique; subsequently, representative histological samples were selected from each patient. From each paraffin block, tumor sections were stained for immunohistochemical detection. The statistical significance of differences was analyzed using proper statistical analysis. The results were considered statistically significant at P < 0.05.
RESULTS By Western blot analysis and using total Met antibody, we found that PTHrP regulated Met expression in HCT116 cells but not in Caco-2 cells. In HCT116 cells, Met protein levels increased at 30 min (P < 0.01) and at 20 h (P < 0.01) whereas the levels diminished at 3 min (P < 0.05), 10 min (P < 0.01), and 1 h to 5 h (P < 0.01) of PTHrP treatment. Using an active Met antibody, we found that where the protein levels of total Met decreased (3 min, 10 min, and 60 min of PTHrP exposure), the status of phosphorylated/activated Met increased (P < 0.01) at the same time, suggesting that Met undergoes proteasomal degradation after its phosphorylation/activation by PTHrP. The increment of its protein level after these decreases (at 30 min and 20 h) suggests a modulation of Met expression by PTHrP in order to improve Met levels and this idea is supported by our observation that the cytokine increased Met mRNA levels at least at 15 min in HCT116 cells as revealed by RT-qPCR analysis (P < 0.05). We then proceeded to evaluate the signaling pathways that mediate the phosphorylation/ activation of Met induced by PTHrP in HCT116 cells. By Western blot technique, we observed that PP1, a specific inhibitor of the activation of the proto-oncogene protein tyrosine kinase Src, blocked the effect of PTHrP on Met phosphorylation (P < 0.05). Furthermore, the selective inhibition of the ERK 1/2 mitogen-activated protein kinase (ERK 1/2 MAPK) using PD98059 and the p38 MAPK using SB203580 diminished the effect of PTHrP on Met phosphorylation/activation (P < 0.05). Using SU11274, the specific inhibitor of Met activation, and trypan blue dye exclusion test, Western blot, wound healing assay, and morphological analysis with a microscope, we observed the reversal of cell events induced by PTHrP such as cell proliferation (P < 0.05), migration (P < 0.05), and the EMT program (P < 0.01) in HCT116 cells. Also, PTHrP favored the chemoresistance to CPT-11 (P < 0.001), OXA (P < 0.01), and DOXO (P < 0.01) through the Met pathway. Taken together, these findings suggest that Met activated by PTHrP participates in events associated with the aggressive phenotype of CRC cells. By immunohistochemical analysis, we found that PTHrP in HCT116 cell xenografts enhanced the protein expression of Met (0.190 ± 0.014) compared to tumors from control mice (0.110 ± 0.012; P < 0.05) and of its own receptor (2.27 ± 0.20) compared to tumors from control mice (1.98 ± 0.14; P < 0.01). Finally, assuming that the changes in the expression of PTHrP and its receptor are directly correlated, we investigated the expression of both Met and PTHR1 in biopsies of CRC patients by immunohistochemical analysis. Comparing histologically differentiated tumors with respect to those less differentiated, we found that the labeling intensity for Met and PTHR1 increased and diminished in a gradual manner, respectively (P < 0.05).
CONCLUSION PTHrP acts through the Met pathway in CRC cells and regulates Met expression in a CRC animal model. More basic and clinical studies are needed to further evaluate the PTHrP/Met relationship.
Collapse
Affiliation(s)
- María Belén Novoa Díaz
- Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur (UNS)- INBIOSUR (CONICET-UNS), Bahía Blanca 8000, Buenos Aires, Argentina
| | - Pedro Carriere
- Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur (UNS)- INBIOSUR (CONICET-UNS), Bahía Blanca 8000, Buenos Aires, Argentina
| | - Graciela Gigola
- Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur (UNS)- INBIOSUR (CONICET-UNS), Bahía Blanca 8000, Buenos Aires, Argentina
| | | | - Natalia Calvo
- Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur (UNS)- INBIOSUR (CONICET-UNS), Bahía Blanca 8000, Buenos Aires, Argentina
| | - Claudia Gentili
- Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur (UNS)- INBIOSUR (CONICET-UNS), Bahía Blanca 8000, Buenos Aires, Argentina
| |
Collapse
|
10
|
Koyande N, Gangopadhyay M, Thatikonda S, Rengan AK. The role of gut microbiota in the development of colorectal cancer: a review. Int J Colorectal Dis 2022; 37:1509-1523. [PMID: 35704091 DOI: 10.1007/s00384-022-04192-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/21/2022] [Indexed: 02/04/2023]
Abstract
PURPOSE Colorectal cancer (CRC) is the cancer of the colon and rectum. Recent research has found a link between CRC and human gut microbiota. This review explores the effect of gut microbiota on colorectal carcinogenesis and the development of chemoresistance. METHODS A literature overview was performed to identify the gut microbiota species that showed altered abundance in CRC patients and the mechanisms by which some of them aid in the development of chemoresistance. RESULTS Types of gut microbiota present and methods of analyzing them were discussed. We observed that numerous microbiota showed altered abundance in CRC patients and could act as a biomarker for CRC diagnosis and treatment. Further, it was demonstrated that microbes also have a role in the development of chemoresistance by mechanisms like immune system activation, drug modification, and autophagy modulation. Finally, the key issue of the growing global problem of antimicrobial resistance and its relationship with CRC was highlighted. CONCLUSION This review discussed the role of gut microbiota dysbiosis on colorectal cancer progression and the development of chemoresistance.
Collapse
Affiliation(s)
- Navami Koyande
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi, Sangareddy- 502284, India
| | - Madhusree Gangopadhyay
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi, Sangareddy- 502284, India
| | - Shashidhar Thatikonda
- Department of Civil Engineering, Indian Institute of Technology Hyderabad, Kandi, Sangareddy- 502284, India
| | - Aravind Kumar Rengan
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi, Sangareddy- 502284, India.
| |
Collapse
|
11
|
McDonald PC, Dedhar S. New Perspectives on the Role of Integrin-Linked Kinase (ILK) Signaling in Cancer Metastasis. Cancers (Basel) 2022; 14:cancers14133209. [PMID: 35804980 PMCID: PMC9264971 DOI: 10.3390/cancers14133209] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 06/22/2022] [Accepted: 06/27/2022] [Indexed: 12/14/2022] Open
Abstract
Simple Summary Today, the vast majority of deaths from cancer are due to cancer metastasis. Metastasis requires that cancer cells escape from the initial tumor, travel through blood vessels, and form new tumors in distant host tissues. Integrin-linked kinase (ILK) is overexpressed by many types of cancer cells and provides both structural and signaling functions that are important for successful metastasis. Here, we discuss recent findings that show how ILK is involved in promoting physical changes important for cell motility and invasion, and how ILK relays signals to other machinery components during metastasis, including interactions with components of the immune system and communication between cancer cells and normal cells, to affect the process of metastasis. We also discuss the contribution of ILK to therapeutic resistance and examine efforts to target ILK for the treatment of metastatic disease. Abstract Cancer metastasis is a major barrier to the long-term survival of cancer patients. In cancer cells, integrin engagement downstream of cell-extracellular matrix (ECM) interactions results in the recruitment of cytoskeletal and signaling molecules to form multi-protein complexes to promote processes critical for metastasis. One of the major functional components of these complexes is Integrin Linked Kinase (ILK). Here, we discuss recent advances in our understanding of the importance of ILK as a signaling effector in processes linked to tumor progression and metastasis. New mechanistic insights as to the role of ILK in cellular plasticity, epithelial mesenchymal transition (EMT), migration, and invasion, including the impact of ILK on the formation of invadopodia, filopodia-like protrusions (FLPs), and Neutrophil Extracellular Trap (NET)-induced motility are highlighted. Recent findings detailing the contribution of ILK to therapeutic resistance and the importance of ILK as a potentially therapeutically tractable vulnerability in both solid tumors and hematologic malignancies are discussed. Indeed, pharmacologic inhibition of ILK activity using specific small molecule inhibitors is effective in curtailing the contribution of ILK to these processes, potentially offering a novel therapeutic avenue for inhibiting critical steps in the metastatic cascade leading to reduced drug resistance and increased therapeutic efficacy.
Collapse
Affiliation(s)
- Paul C. McDonald
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada;
| | - Shoukat Dedhar
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada;
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
- Correspondence:
| |
Collapse
|
12
|
Nikou S, Arbi M, Dimitrakopoulos FID, Kalogeropoulou A, Geramoutsou C, Zolota V, Kalofonos HP, Taraviras S, Lygerou Z, Bravou V. Ras suppressor-1 (RSU1) exerts a tumor suppressive role with prognostic significance in lung adenocarcinoma. Clin Exp Med 2022:10.1007/s10238-022-00847-8. [PMID: 35729367 DOI: 10.1007/s10238-022-00847-8] [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: 12/18/2021] [Accepted: 05/25/2022] [Indexed: 11/03/2022]
Abstract
Ras suppressor-1 (RSU1), originally described as a suppressor of Ras oncogenic transformation, localizes to focal adhesions interacting with the ILK-PINCH-PARVIN (IPP) complex that exerts a well-established oncogenic role in cancer. However, RSU1 implication in lung cancer is currently unknown. Our study aims to address the role of RSU1 in lung adenocarcinoma (LUADC). We here show that RSU1 protein expression by immunohistochemistry is downregulated in LUADC human tissue samples and represents a significant prognostic indicator. In silico analysis of gene chip and RNA seq data validated our findings. Depletion of RSU1 by siRNA in lung cancer cells promotes anchorage-independent cell growth, cell motility and epithelial to mesenchymal transition (EMT). Silencing of RSU1 also alters IPP complex expression in lung cancer cells. The p29 RSU1 truncated isoform is detected in lung cancer cells, and its expression is downregulated upon RSU1 silencing, whereas it is overexpressed upon ILK overexpression. These findings suggest that RSU1 exerts a tumor suppressive role with prognostic significance in LUADC.
Collapse
Affiliation(s)
- Sofia Nikou
- Department of Anatomy-Histology-Embryology, Medical School, University of Patras, 26504, Patras, Greece
| | - Marina Arbi
- Department of General Biology, Medical School, University of Patras, 26504, Patras, Greece
| | - Foteinos-Ioannis D Dimitrakopoulos
- Molecular Oncology Laboratory, Division of Oncology, Department of Medicine, Medical School, University of Patras, 26504, Patras, Greece
| | - Argiro Kalogeropoulou
- Department of Physiology, School of Medicine, University of Patras, 26504, Rio, Patras, Greece
| | - Christina Geramoutsou
- Department of Anatomy-Histology-Embryology, Medical School, University of Patras, 26504, Patras, Greece
| | - Vasiliki Zolota
- Department of Anatomy-Histology-Embryology, Medical School, University of Patras, 26504, Patras, Greece.,Department of Pathology, University Hospital of Patras, 26504, Patras, Greece
| | - Haralabos P Kalofonos
- Molecular Oncology Laboratory, Division of Oncology, Department of Medicine, Medical School, University of Patras, 26504, Patras, Greece.,Division of Oncology, Department of Internal Medicine, University Hospital of Patras, 26504, Rio Patras, Greece
| | - Stavros Taraviras
- Department of Physiology, School of Medicine, University of Patras, 26504, Rio, Patras, Greece
| | - Zoi Lygerou
- Department of General Biology, Medical School, University of Patras, 26504, Patras, Greece
| | - Vasiliki Bravou
- Department of Anatomy-Histology-Embryology, Medical School, University of Patras, 26504, Patras, Greece.
| |
Collapse
|
13
|
Almasabi S, Boyd R, Ahmed AU, Williams BRG. Integrin-Linked Kinase Expression Characterizes the Immunosuppressive Tumor Microenvironment in Colorectal Cancer and Regulates PD-L1 Expression and Immune Cell Cytotoxicity. Front Oncol 2022; 12:836005. [PMID: 35692780 PMCID: PMC9174997 DOI: 10.3389/fonc.2022.836005] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 04/22/2022] [Indexed: 11/13/2022] Open
Abstract
Integrin-linked kinase (ILK) has been implicated as a molecular driver and mediator in both inflammation and tumorigenesis of the colon. However, a role for ILK in the tumor microenvironment (TME) and immune evasion has not been investigated. Here, we show a correlation of ILK expression with the immunosuppressive TME and cancer prognosis. We also uncover a role for ILK in the regulation of programmed death-ligand 1 (PD-L1) expression and immune cell cytotoxicity. Interrogation of web-based data-mining platforms, showed upregulation of ILK expression in tumors and adjacent-non tumor tissue of colorectal cancer (CRC) associated with poor survival and advanced stages. ILK expression was correlated with cancer-associated fibroblast (CAFs) and immunosuppressive cell infiltration including regulatory T cells (Treg) and M2 macrophages (M2) in addition to their gene markers. ILK expression was also significantly correlated with the expression of different cytokines and chemokines. ILK expression showed pronounced association with different important immune checkpoints including PD-L1. Deletion of the ILK gene in PD-L1 positive CRC cell lines using a doxycycline inducible-CRISPR/Cas9, resulted in suppression of both the basal and IFNγ-induced PD-L1 expression via downregulating NF-κB p65. This subsequently sensitized the CRC cells to NK92 immune cell cytotoxicity. These findings suggest that ILK can be used as a biomarker for prognosis and immune cell infiltration in colon cancer. Moreover, ILK could provide a therapeutic target to prevent immune evasion mediated by the expression of PD-L1.
Collapse
Affiliation(s)
- Saleh Almasabi
- Cancer and Innate Immunity, Centre for Cancer Research, Hudson Institute of Medical Research, Monash University, Clayton, VIC, Australia.,Cartherics, Hudson Institute of Medical Research, Monash University, Clayton, VIC, Australia.,Clinical Laboratory Sciences, Applied Medical Sciences, Najran University, Najran, Saudi Arabia
| | - Richard Boyd
- Cartherics, Hudson Institute of Medical Research, Monash University, Clayton, VIC, Australia
| | - Afsar U Ahmed
- Cancer and Innate Immunity, Centre for Cancer Research, Hudson Institute of Medical Research, Monash University, Clayton, VIC, Australia
| | - Bryan R G Williams
- Cancer and Innate Immunity, Centre for Cancer Research, Hudson Institute of Medical Research, Monash University, Clayton, VIC, Australia
| |
Collapse
|
14
|
Paul M, Gope TK, Das P, Ain R. Nitric-Oxide Synthase trafficking inducer (NOSTRIN) is an emerging negative regulator of colon cancer progression. BMC Cancer 2022; 22:594. [PMID: 35642021 PMCID: PMC9158178 DOI: 10.1186/s12885-022-09670-6] [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: 07/25/2021] [Accepted: 05/17/2022] [Indexed: 11/10/2022] Open
Abstract
Background NOSTRIN, abundantly expressed in colon, was reported to be anti-angiogenic, anti-invasive and anti-inflammatory. NOSTRIN expression was inversely related to survival of pancreatic ductal adeno-carcinoma patients. Yet its function and regulatory mechanism in CRC remains elusive. Methods NOSTRIN’s influence on EMT of CRC cells were analysed using realtime PCR array containing the functional EMT-transcriptome followed by western blotting. Regulation of oncogenic potential of CRC cells by NOSTRIN was elucidated using soft agar colony formation, trans-well invasion, wound healing and colonosphere formation assays. Biochemical assays were used to reveal mechanism of NOSTRIN function. Human CRC tissue array was used to test NOSTRIN mark in control and CRC disease stages. Results We showed here that CRC cell lines with less NOSTRIN expression has more invasive and migratory potential. NOSTRIN affected EMT-associated transcriptome of CRC cells by down regulating 33 genes that were functionally annotated to transcription factors, genes important for cell growth, proliferation, migration, cell adhesion and cytoskeleton regulators in CRC cells. NOSTRIN over-expression significantly reduced soft agar colony formation, wound healing and cell invasion. In line with this, RNA interference of Nostrin enhanced metastatic potential of CRC cells. Furthermore, stable overexpression of NOSTRIN in CRC cell line not only curtailed its ability to form colonosphere but also decreased expression of stemness markers CD133, CD44 and EpCAM. NOSTRIN’s role in inhibiting self-renewal was further confirmed using BrdU incorporation assay. Interestingly, NOSTRIN formed immune-complex with Cdk1 in CRC cells and aided in increase of inhibitory Y15 and T14 phosphorylation of Cdk1 that halts cytokinesis. These ex vivo findings were substantiated using human colon cancer tissue array containing cDNAs from patients’ samples with various stages of disease progression. Significant decrease in NOSTRIN expression was found with initiation and progression of advanced colon cancer disease stages. Conclusion We illustrate function of a novel molecule, NOSTRIN in curtailing EMT and maintenance of CRC cell stemness. Our data validates importance of NOSTRIN mark during onset and disease progression of CRC indicating its diagnostic potential. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-022-09670-6.
Collapse
Affiliation(s)
- Madhurima Paul
- Division of Cell Biology and Physiology, CSIR-Indian Institute of Chemical Biology, 4, Raja S.C. Mullick Road, Jadavpur, Kolkata, West Bengal, 700032, India
| | - Tamal Kanti Gope
- Division of Cell Biology and Physiology, CSIR-Indian Institute of Chemical Biology, 4, Raja S.C. Mullick Road, Jadavpur, Kolkata, West Bengal, 700032, India
| | - Priyanka Das
- Division of Cell Biology and Physiology, CSIR-Indian Institute of Chemical Biology, 4, Raja S.C. Mullick Road, Jadavpur, Kolkata, West Bengal, 700032, India
| | - Rupasri Ain
- Division of Cell Biology and Physiology, CSIR-Indian Institute of Chemical Biology, 4, Raja S.C. Mullick Road, Jadavpur, Kolkata, West Bengal, 700032, India.
| |
Collapse
|
15
|
Zhang H, Wang J, Yu T, Wang J, Lu J, Yu Z. Silencing LncRNA CASC9 inhibits proliferation and invasion of colorectal cancer cells by MiR-542-3p/ILK. PLoS One 2022; 17:e0265901. [PMID: 35427373 PMCID: PMC9012350 DOI: 10.1371/journal.pone.0265901] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 03/09/2022] [Indexed: 12/12/2022] Open
Abstract
Colorectal cancer (CRC) ranks the third in cancers and the second in the reasons of cancer-related death. More evidence indicates that long non-coding RNA participates in tumor initiation and progression. It’s known that cancer susceptibility candidate 9 is an oncogenic long non-coding RNA in CRC. miR-542-3p is a negative regulator of CRC, while integrin-linked kinase could contribute to tumor progression and chemoresistance. However, the correlation among long non-coding RNA cancer susceptibility candidate 9, miR-542-3p and integrin-linked kinase in CRC is still unclear. We demonstrated long non-coding RNA cancer susceptibility candidate 9 in CRC specimens and cell lines overexpressed via real-time quantitative polymerase chain reaction. Once long non-coding RNA cancer susceptibility candidate 9 was knocked down, it significantly inhibited proliferation, invasion, and migration of CRC cells in real-time quantitative polymerase chain reaction, cell counting kit-8, 5-ethynyl-2’-deoxyuridine, and transwell assays, which also was validated in vivo. Long non-coding RNA cancer susceptibility candidate 9 negatively regulates miR-542-3p in a targeted manner, and the function of up-regulated miR-542-3p was confirmed similarly. While miR-542-3p negatively regulates integrin-linked kinase. Thus, we further verified that overexpression of integrin-linked kinase on down-regulated long non-coding RNA cancer susceptibility candidate 9 or up-regulated miR-542-3p significantly restored CRC cell proliferation via bioinformatic analysis, dual-luciferase report assay, real-time quantitative polymerase chain reaction, RNA immunoprecipitation, and western blot. This study testified that silencing long non-coding RNA cancer susceptibility candidate 9 could inhibit proliferation and invasion of CRC cells by miR-542-3p/integrin-linked kinase.
Collapse
Affiliation(s)
- Heping Zhang
- Department of Anorectal, People’s Hospital of Jiaozuo, Jiaozuo, Henan Province, China
| | - Jingfang Wang
- Medical College of Rehabilitation, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian Province, China
| | - Taoyuan Yu
- Institute of International Education, Beijing University of Chemical Technology, Beijing, China
| | - Jingmin Wang
- Infertility Clinic, People’s Hospital of Jiaozuo, Jiaozuo, Henan Province, China
| | - Jun Lu
- Basic Medical Laboratory, 900th Hospital of the Joint Logistics Team, Fuzhou, Fujian Province, China
| | - Zongyang Yu
- Pulmonary and Critical Care Medicine, 900th Hospital of the Joint Logistics Team, Fuzhou, Fujian Province, China
- * E-mail:
| |
Collapse
|
16
|
Aliabedi B, Mousavi SH, Ebrahimi M, Alizadeh S, Hedayati Asl AA, Mohammad M, Samieyan Dehkordi S. Hsa-miR-625 Upregulation Promotes Apoptosis in Acute Myeloid Leukemia Cell Line by Targeting Integrin-linked Kinase Pathway. Asian Pac J Cancer Prev 2022; 23:1159-1167. [PMID: 35485671 PMCID: PMC9375600 DOI: 10.31557/apjcp.2022.23.4.1159] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 04/21/2022] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND Growing evidence has demonstrated that microRNAs have a major effect on development of different types of cancer including AML. The overexpression of miR-625 could decrease tumorgenesis of acute myeloid leukemia cell lines through Integrin-linked kinase signaling pathway and reducing the associated oncogenes. The aim of this study is to evaluate the effect of hsa-miR-625 upregulation on apoptosis and proliferation of KG1 cell line via ILK signaling pathway. METHODS The KG-1 cell line was transfected with pLenti-III-premir625-GFP through viral method. Then, expression of miR-625 and genes were analyzed by quantitative PCR. Western blotting was used to evaluate for the protein level. Apoptosis was investigated by flow cytometry. Cell cycle analysis with PI and CCK-8 assay were performed to evaluate proliferation. RESULTS KG-1 cells transfected with pLenti-III-pre mir625-GFP construct showed a significant increase in cell apoptosis. Gene expression of ILK and NF-κB were downregulated and AKT, c-fos, Caspase3, cyclin D1, KLF-4, OCT-4 and Nanog were upregulated but no alteration in GSK3 expression profile was observed. Downregulation of NF-κB and upregulation of Caspase 3 and p-β-catenin protein levels were indicated (p<0.05). CONCLUSION MiR-625 can be a promising approach to aid in the treatment of AML. However, further studies are required in this respect.
Collapse
Affiliation(s)
- Bahareh Aliabedi
- Department of Hematology, School of Allied Medical Sciences, Tehran University of Medical Sciences, Tehran, Iran.
| | - Seyed Hadi Mousavi
- Department of Hematology, School of Allied Medical Sciences, Tehran University of Medical Sciences, Tehran, Iran.
| | - Marzieh Ebrahimi
- Department of Regenerative Biomedicine, Cell Science Research Center, Royan Institute for Stem Cell Biology & Technology, ACECR, Tehran, Iran.
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology & Technology, ACECR, Tehran, Iran.
| | - Shaban Alizadeh
- Department of Hematology, School of Allied Medical Sciences, Tehran University of Medical Sciences, Tehran, Iran.
| | - Amir Abbas Hedayati Asl
- Department of Regenerative Biomedicine, Cell Science Research Center, Royan Institute for Stem Cell Biology & Technology, ACECR, Tehran, Iran.
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology & Technology, ACECR, Tehran, Iran.
| | - Monireh Mohammad
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology & Technology, ACECR, Tehran, Iran.
| | - Sahar Samieyan Dehkordi
- Department of Hematology, School of Allied Medical Sciences, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
17
|
Comparison of Colorectal Cancer Stem Cells and Oxaliplatin-Resistant Cells Unveils Functional Similarities. Cells 2022; 11:cells11030511. [PMID: 35159320 PMCID: PMC8833894 DOI: 10.3390/cells11030511] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 01/27/2022] [Accepted: 01/29/2022] [Indexed: 02/01/2023] Open
Abstract
Colorectal cancer is the second most common cancer in women, the third in men, and an important cause of cancer-related mortality. Recurrence and the development of chemotherapy resistance are major hindrances for patients’ treatment. The presence of cancer stem cells with chemotherapy resistance able to generate proliferating tumor cells contributes to tumor recurrence and resistance. In addition, tumor cells can develop chemoresistance through adaptation mechanisms. In this article, cancer stem cells were isolated from HT29 and SW620 colorectal cancer cell lines. Oxaliplatin resistance was induced by a single drug treatment simulating the usual guidelines of patient treatment. A comparison of these two populations showed similarities since cancer stem cells presented increased oxaliplatin resistance, and resistant cells contained an increased number of cancer stem cells. Cancer stem cells isolated from resistant cells showed increased oxaliplatin resistance. Cell invasion capacity and epithelial-mesenchymal transition were increased both in cancer stem cells and oxaliplatin-resistant cells. mRNA expression analysis showed that both cell types shared a significant proportion of commonly regulated genes. In summary, the data presented indicate that colorectal cancer stem cells and oxaliplatin-resistant cells are highly related cell populations that might have interesting implications in the development of tumor recurrence and resistance to chemotherapy.
Collapse
|
18
|
Górska A, Mazur AJ. Integrin-linked kinase (ILK): the known vs. the unknown and perspectives. Cell Mol Life Sci 2022; 79:100. [PMID: 35089438 PMCID: PMC8799556 DOI: 10.1007/s00018-021-04104-1] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 11/29/2021] [Accepted: 12/17/2021] [Indexed: 02/08/2023]
Abstract
Integrin-linked kinase (ILK) is a multifunctional molecular actor in cell-matrix interactions, cell adhesion, and anchorage-dependent cell growth. It combines functions of a signal transductor and a scaffold protein through its interaction with integrins, then facilitating further protein recruitment within the ILK-PINCH-Parvin complex. ILK is involved in crucial cellular processes including proliferation, survival, differentiation, migration, invasion, and angiogenesis, which reflects on systemic changes in the kidney, heart, muscle, skin, and vascular system, also during the embryonal development. Dysfunction of ILK underlies the pathogenesis of various diseases, including the pro-oncogenic activity in tumorigenesis. ILK localizes mostly to the cell membrane and remains an important component of focal adhesion. We do know much about ILK but a lot still remains either uncovered or unclear. Although it was initially classified as a serine/threonine-protein kinase, its catalytical activity is now questioned due to structural and functional issues, leaving the exact molecular mechanism of signal transduction by ILK unsolved. While it is known that the three isoforms of ILK vary in length, the presence of crucial domains, and modification sites, most of the research tends to focus on the main isoform of this protein while the issue of functional differences of ILK2 and ILK3 still awaits clarification. The activity of ILK is regulated on the transcriptional, protein, and post-transcriptional levels. The crucial role of phosphorylation and ubiquitylation has been investigated, but the functions of the vast majority of modifications are still unknown. In the light of all those open issues, here we present an extensive literature survey covering a wide spectrum of latest findings as well as a past-to-present view on controversies regarding ILK, finishing with pointing out some open questions to be resolved by further research.
Collapse
Affiliation(s)
- Agata Górska
- Department of Cell Pathology, Faculty of Biotechnology, University of Wroclaw, ul. Joliot-Curie 14a, 50-383, Wrocław, Poland.
| | - Antonina Joanna Mazur
- Department of Cell Pathology, Faculty of Biotechnology, University of Wroclaw, ul. Joliot-Curie 14a, 50-383, Wrocław, Poland.
| |
Collapse
|
19
|
Tanabe S. Epithelial-Mesenchymal Transition and Cancer Stem Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1393:1-49. [PMID: 36587300 DOI: 10.1007/978-3-031-12974-2_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Epithelial-mesenchymal transition (EMT), a cellular phenotypic change from epithelial to mesenchymal-like features, is related to the resistance and metastasis of cancer stem cells (CSCs). Several signal transduction mechanisms induce EMT, which causes the gene expression alteration to induce the acquisition of resistance and metastasis in cancer. EMT is characterized with high gene expression of cadherin 2 (N-cadherin) and vimentin, and sparse cell-cell junction. The cells with EMT-phenotype have migration, metastasis and drug-resistance capacity, which are main characteristics of CSCs. It seems that the main population of CSCs exhibits EMT phenotype, whereas some populations consist of phenotypes other than EMT. In this chapter, EMT mechanism, phenotypic features of EMT and CSCs, signal transduction in EMT and CSCs, differences between EMT and CSCs, and the role of EMT in CSCs are described.
Collapse
Affiliation(s)
- Shihori Tanabe
- Division of Risk Assessment, Center for Biological Safety and Research, National Institute of Health Sciences, Kawasaki, 210-9501, Japan.
| |
Collapse
|
20
|
Novoa Díaz MB, Carriere PM, Martín MJ, Calvo N, Gentili C. Involvement of parathyroid hormone-related peptide in the aggressive phenotype of colorectal cancer cells. World J Gastroenterol 2021; 27:7025-7040. [PMID: 34887626 PMCID: PMC8613645 DOI: 10.3748/wjg.v27.i41.7025] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 06/26/2021] [Accepted: 08/23/2021] [Indexed: 02/06/2023] Open
Abstract
Colorectal cancer (CRC) remains one of the leading causes of mortality from malignant diseases worldwide. In general terms, CRC presents high heterogeneity due to the influence of different genetic and environmental factors; also, the neoplastic cells are strongly influenced by the extracellular matrix and several surrounding cells, known together as the tumor microenvironment (TME). Bidirectional communication takes place between the tumor and the TME through the release of autocrine and paracrine factors. Parathyroid hormone-related peptide (PTHrP) is a cytokine secreted by a wide variety of tissues and is able to regulate several cellular functions both in physiological as well as in pathological processes. It exerts its effects as a paracrine/autocrine factor, although its mode of action is mainly paracrine. It has been shown that this peptide is expressed by several tumors and that the tumor secretion of PTHrP is responsible for the malignant humoral hypercalcemia. Eight years ago, when our research group started studying PTHrP effects in the experimental models derived from intestinal tumors, the literature available at the time addressing the effects of PTHrP on colorectal tumors was limited, and no articles had been published regarding to the paracrine action of PTHrP in CRC cells. Based on this and on our previous findings regarding the role of PTH in CRC cells, our purpose in recent years has been to explore the role of PTHrP in CRC. We analyzed the behavior of CRC cells treated with exogenous PTHrP, focalizing in the study of the following events: Survival, cell cycle progression and proliferation, migration, chemoresistance, tumor-associated angiogenesis, epithelial to mesenchymal transition program and other events also associated with invasion, such us the induction of cancer stem cells features. This work summarizes the major findings obtained by our investigation group using in vitro and in vivo CRC models that evidence the participation of PTHrP in the acquisition of an aggressive phenotype of CRC cells and the molecular mechanisms involved in these processes. Recently, we found that this cytokine induces this malignant behavior not only by its direct action on these intestinal cells but also through its influence on cells derived from TME, promoting a communication between CRC cells and surrounding cells that contributes to the molecular and morphological changes observed in CRC cells. These investigations establish the basis for our next studies in order to address the clinical applicability of our findings. Recognizing the factors and mechanisms that promote invasion in CRC cells, evasion to the cytotoxic effects of current CRC therapies and thus metastasis is decisive for the identification of new markers with the potential to improve early diagnosis and/or to predict prognosis, to predetermine drug resistance and to provide treatment guidelines that include targeted therapies for this disease.
Collapse
Affiliation(s)
- María Belén Novoa Díaz
- Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur (UNS)- INBIOSUR (CONICET-UNS), Bahía Blanca 8000, Buenos Aires, Argentina
| | - Pedro Matías Carriere
- Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur (UNS)- INBIOSUR (CONICET-UNS), Bahía Blanca 8000, Buenos Aires, Argentina
| | - María Julia Martín
- Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur (UNS)- INBIOSUR (CONICET-UNS), Bahía Blanca 8000, Buenos Aires, Argentina
- Departamento de Química, Universidad Nacional del Sur (UNS)- INQUISUR (CONICET-UNS), Bahía Blanca 8000, Buenos Aires, Argentina
| | - Natalia Calvo
- Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur (UNS)- INBIOSUR (CONICET-UNS), Bahía Blanca 8000, Buenos Aires, Argentina
| | - Claudia Gentili
- Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur (UNS)- INBIOSUR (CONICET-UNS), Bahía Blanca 8000, Buenos Aires, Argentina
| |
Collapse
|
21
|
Greco L, Rubbino F, Morelli A, Gaiani F, Grizzi F, de’Angelis GL, Malesci A, Laghi L. Epithelial to Mesenchymal Transition: A Challenging Playground for Translational Research. Current Models and Focus on TWIST1 Relevance and Gastrointestinal Cancers. Int J Mol Sci 2021; 22:11469. [PMID: 34768901 PMCID: PMC8584071 DOI: 10.3390/ijms222111469] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Revised: 10/18/2021] [Accepted: 10/21/2021] [Indexed: 12/15/2022] Open
Abstract
Resembling the development of cancer by multistep carcinogenesis, the evolution towards metastasis involves several passages, from local invasion and intravasation, encompassing surviving anoikis into the circulation, landing at distant sites and therein establishing colonization, possibly followed by the outgrowth of macroscopic lesions. Within this cascade, epithelial to mesenchymal transition (EMT) works as a pleiotropic program enabling cancer cells to overcome local, systemic, and distant barriers against diffusion by replacing traits and functions of the epithelial signature with mesenchymal-like ones. Along the transition, a full-blown mesenchymal phenotype may not be accomplished. Rather, the plasticity of the program and its dependency on heterotopic signals implies a pendulum with oscillations towards its reversal, that is mesenchymal to epithelial transition. Cells in intermixed E⇔M states can also display stemness, enabling their replication together with the epithelial reversion next to successful distant colonization. If we aim to include the EMT among the hallmarks of cancer that could modify clinical practice, the gap between the results pursued in basic research by animal models and those achieved in translational research by surrogate biomarkers needs to be filled. We review the knowledge on EMT, derived from models and mechanistic studies as well as from translational studies, with an emphasis on gastrointestinal cancers (GI).
Collapse
Affiliation(s)
- Luana Greco
- Laboratory of Molecular Gastroenterology, IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Rozzano, Italy; (L.G.); (F.R.); (A.M.)
| | - Federica Rubbino
- Laboratory of Molecular Gastroenterology, IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Rozzano, Italy; (L.G.); (F.R.); (A.M.)
| | - Alessandra Morelli
- Laboratory of Molecular Gastroenterology, IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Rozzano, Italy; (L.G.); (F.R.); (A.M.)
| | - Federica Gaiani
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy; (F.G.); (G.L.d.)
- Gastroenterology and Endoscopy Unit, University-Hospital of Parma, Via Gramsci 14, 43126 Parma, Italy
| | - Fabio Grizzi
- Department of Immunology and Inflammation, IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Rozzano, Italy;
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20072 Pieve Emanuele, Italy;
| | - Gian Luigi de’Angelis
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy; (F.G.); (G.L.d.)
- Gastroenterology and Endoscopy Unit, University-Hospital of Parma, Via Gramsci 14, 43126 Parma, Italy
| | - Alberto Malesci
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20072 Pieve Emanuele, Italy;
- IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Rozzano, Italy
| | - Luigi Laghi
- Laboratory of Molecular Gastroenterology, IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Rozzano, Italy; (L.G.); (F.R.); (A.M.)
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy; (F.G.); (G.L.d.)
| |
Collapse
|
22
|
Common and mutation specific phenotypes of KRAS and BRAF mutations in colorectal cancer cells revealed by integrative -omics analysis. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2021; 40:225. [PMID: 34233735 PMCID: PMC8265010 DOI: 10.1186/s13046-021-02025-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 06/18/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND Genes in the Ras pathway have somatic mutations in at least 60 % of colorectal cancers. Despite activating the same pathway, the BRAF V600E mutation and the prevalent mutations in codon 12 and 13 of KRAS have all been linked to different clinical outcomes, but the molecular mechanisms behind these differences largely remain to be clarified. METHODS To characterize the similarities and differences between common activating KRAS mutations and between KRAS and BRAF mutations, we used genome editing to engineer KRAS G12C/D/V and G13D mutations in colorectal cancer cells that had their mutant BRAF V600E allele removed and subjected them to transcriptome sequencing, global proteomics and metabolomics analyses. RESULTS By intersecting differentially expressed genes, proteins and metabolites, we uncovered (i) two-fold more regulated genes and proteins when comparing KRAS to BRAF mutant cells to those lacking Ras pathway mutation, (ii) five differentially expressed proteins in KRAS mutants compared to cells lacking Ras pathway mutation (IFI16, S100A10, CD44, GLRX and AHNAK2) and 6 (CRABP2, FLNA, NXN, LCP1, S100A10 and S100A2) compared to BRAF mutant cells, (iii) 19 proteins expressed differentially in a KRAS mutation specific manner versus BRAF V600E cells, (iv) regulation of the Integrin Linked Kinase pathway by KRAS but not BRAF mutation, (v) regulation of amino acid metabolism, particularly of the tyrosine, histidine, arginine and proline pathways, the urea cycle and purine metabolism by Ras pathway mutations, (vi) increased free carnitine in KRAS and BRAF mutant RKO cells. CONCLUSIONS This comprehensive integrative -omics analysis confirms known and adds novel genes, proteins and metabolic pathways regulated by mutant KRAS and BRAF signaling in colorectal cancer. The results from the new model systems presented here can inform future development of diagnostic and therapeutic approaches targeting tumors with KRAS and BRAF mutations.
Collapse
|
23
|
Ghazi A, Le Corre D, Pilati C, Taieb J, Aparicio T, Didelot A, Dedhar S, Mulot C, Le Malicot K, Djouadi F, de Reynies A, Launay JM, Laurent-Puig P, Mouillet-Richard S. Prognostic value of the PrP C-ILK-IDO1 axis in the mesenchymal colorectal cancer subtype. Oncoimmunology 2021; 10:1940674. [PMID: 34249475 PMCID: PMC8244775 DOI: 10.1080/2162402x.2021.1940674] [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: 10/24/2022] Open
Abstract
The CMS4 mesenchymal subtype of colorectal cancer (CRC) is associated with poor prognosis and resistance to treatment. The cellular prion protein PrPC is overexpressed in CMS4 tumors and controls the expression of a panel of CMS4-specific genes in CRC cell lines. Here, we sought to investigate PrPC downstream pathways that may underlie its role in CMS4 CRC. By combining gene set enrichment analyses and gain and loss of function approaches in CRC cell lines, we identify the integrin-linked kinase ILK as a proximal effector of PrPC that mediates its control on the CMS4 phenotype. We further leveraged three independent large CRC cohorts to assess correlations in gene expression pattern with patient outcomes and found that ILK is overexpressed in CMS4 mesenchymal tumors and confers a poor prognosis, especially when combined with high expression of the PrPC encoding gene PRNP. Of note, we discovered that the PrPC-ILK signaling axis controls the expression and activity of the tryptophan metabolizing enzyme indoleamine 2,3 dioxygenase IDO1, a key player in immune tolerance. In addition, we monitored alterations in the levels of tryptophan and its metabolites of the kynurenine pathway in the plasma of metastatic CRC patients (n = 325) and we highlight their prognostic value in combination with plasma PrPC levels. Thus, the PrPC-ILK-IDO1 axis plays a key role in the mesenchymal subtype of CRC. PrPC and IDO1-targeted strategies may represent new avenues for patient stratification and treatment in CRC.
Collapse
Affiliation(s)
- Alexandre Ghazi
- Centre de Recherche Des Cordeliers, INSERM, Sorbonne Université, Université de Paris, Paris, France
| | - Delphine Le Corre
- Centre de Recherche Des Cordeliers, INSERM, Sorbonne Université, Université de Paris, Paris, France
| | - Camilla Pilati
- Centre de Recherche Des Cordeliers, INSERM, Sorbonne Université, Université de Paris, Paris, France
| | - Julien Taieb
- Centre de Recherche Des Cordeliers, INSERM, Sorbonne Université, Université de Paris, Paris, France.,Department of Gastroenterology and GI Oncology, AP-HP, Hôpital Européen Georges Pompidou, Paris, France
| | - Thomas Aparicio
- Department of Gastroenterology and Digestive Oncology, AP-HP, Hôpital Saint-Louis, Université de Paris, Université Paris Diderot, Paris, France
| | - Audrey Didelot
- Centre de Recherche Des Cordeliers, INSERM, Sorbonne Université, Université de Paris, Paris, France
| | - Shoukat Dedhar
- Genetics Unit, Integrative Oncology, BC Cancer, Vancouver, Canada
| | - Claire Mulot
- Centre de Recherche Des Cordeliers, INSERM, Sorbonne Université, Université de Paris, Paris, France
| | - Karine Le Malicot
- Fédération Francophone de Cancérologie Digestive, Epicad Inserm, Université de Bourgogne et and Franche Comté, Dijon, France
| | - Fatima Djouadi
- Centre de Recherche Des Cordeliers, INSERM, Sorbonne Université, Université de Paris, Paris, France
| | - Aurélien de Reynies
- Programme carte d'identité des tumeurs, Ligue Nationale Contre Le Cancer, Paris, France
| | - Jean-Marie Launay
- AP-HP Service de Biochimie, INSERM U942 Lariboisière Hospital, Paris, France.,Pharma Research Department, F. Hoffmann-La-Roche Ltd., Basel, Switzerland
| | - Pierre Laurent-Puig
- Centre de Recherche Des Cordeliers, INSERM, Sorbonne Université, Université de Paris, Paris, France.,Department of Biology, AP-HP, Hôpital Européen Georges Pompidou, Paris, France
| | - Sophie Mouillet-Richard
- Centre de Recherche Des Cordeliers, INSERM, Sorbonne Université, Université de Paris, Paris, France
| |
Collapse
|
24
|
Ding J, Wang X, Gao J, Song T. Silencing of cystatin SN abrogates cancer progression and stem cell properties in papillary thyroid carcinoma. FEBS Open Bio 2021. [PMID: 34102026 PMCID: PMC8329778 DOI: 10.1002/2211-5463.13221] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 03/29/2021] [Accepted: 06/07/2021] [Indexed: 01/03/2023] Open
Abstract
Papillary thyroid carcinoma (PTC) accounts for approximately 80% of total thyroid cancers worldwide. Although the prognosis for early‐stage PTC is favorable, the 5‐year survival rate of patients with late‐stage PTC is still very poor. Cystatin SN (cystatin 1, CST1) facilitates the progression of multiple cancers, but its role in regulating PTC pathogenesis is still largely unknown. In this study, we measured the expression levels of CST1 in PTC clinical tissues and cell lines by real‐time quantitative PCR and western blot analysis, and we performed gain‐ and loss‐of‐function experiments to examine the effects of CST1 on PTC cell growth, invasion, migration, epithelial–mesenchymal transition and stemness. Tumorigenicity was assessed using in vivo tumor‐bearing nude mouse models. As expected, upregulated CST1 was observed in PTC tissues (P < 0.05) and cells, compared with their normal counterparts (P < 0.05); furthermore, patients with PTC with higher levels of CST1 exhibited unfavorable prognosis (P < 0.05). In addition, CST1 ablation inhibited PTC cell growth (P < 0.05) in vivo and in vitro. Silencing of CST1 also inhibited cell motility and epithelial–mesenchymal transition in PTC cells (P < 0.05), whereas CST1 overexpression had the opposite effects on the earlier cellular functions. Notably, up‐regulation of CST1 promoted cell spheroid formation (P < 0.05) and increased the expression levels of stemness signatures (P < 0.05) in PTC cells. Collectively, these findings suggest that CST1 functions as an oncogene to facilitate cancer development and promote cancer stem cell properties in PTC cells, increasing our understanding of PTC pathogenesis mechanisms and possibly aiding in the development of potential therapeutic strategies.
Collapse
Affiliation(s)
- Jiaojiao Ding
- Department of Ultrasound, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Xiaorong Wang
- Department of Ultrasound, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Junxi Gao
- Department of Ultrasound, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Tao Song
- Department of Ultrasound, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| |
Collapse
|
25
|
Almasabi S, Ahmed AU, Boyd R, Williams BRG. A Potential Role for Integrin-Linked Kinase in Colorectal Cancer Growth and Progression via Regulating Senescence and Immunity. Front Genet 2021; 12:638558. [PMID: 34163519 PMCID: PMC8216764 DOI: 10.3389/fgene.2021.638558] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 03/08/2021] [Indexed: 01/10/2023] Open
Abstract
Integrin-linked kinase (ILK) has been implicated as a molecular driver and mediator in both inflammation and tumorigenesis of the colon. ILK functions as an adaptor and mediator protein linking the extracellular matrix with downstream signaling pathways. ILK is broadly expressed in many human tissues and cells. It is also overexpressed in many cancers, including colorectal cancer (CRC). Inflammation, as evidenced by inflammatory bowel disease (IBD), is one of the highest risk factors for initiating CRC. This has led to the hypothesis that targeting ILK therapeutically could have potential in CRC, as it regulates different cellular processes associated with CRC development and progression as well as inflammation in the colon. A number of studies have indicated an ILK function in senescence, a cellular process that arrests the cell cycle while maintaining active metabolism and transcription. Senescent cells produce different secretions collectively known as the senescence-associated secretory phenotype (SASP). The SASP secretions influence infiltration of different immune cells, either positively for clearing senescent cells or negatively for promoting tumor growth, reflecting the dual role of senescence in cancer. However, a role for ILK in senescence and immunity in CRC remains to be determined. In this review, we discuss the possible role for ILK in senescence and immunity, paying particular attention to the relevance of ILK in CRC. We also examine how activating Toll-like receptors (TLRs) and their agonists in CRC could trigger immune responses against cancer, as a combination therapy with ILK inhibition.
Collapse
Affiliation(s)
- Saleh Almasabi
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, VIC, Australia.,Cartherics, Hudson Institute of Medical Research, Clayton, VIC, Australia.,Clinical Laboratory Sciences, Applied Medical Sciences, Najran University, Najran, Saudi Arabia.,Department of Molecular and Translational Sciences, Faculty of Medicine Nursing and Health Sciences, Monash University, Clayton, VIC, Australia
| | - Afsar U Ahmed
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, VIC, Australia
| | - Richard Boyd
- Cartherics, Hudson Institute of Medical Research, Clayton, VIC, Australia
| | - Bryan R G Williams
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, VIC, Australia.,Department of Molecular and Translational Sciences, Faculty of Medicine Nursing and Health Sciences, Monash University, Clayton, VIC, Australia
| |
Collapse
|
26
|
Zheng GL, Liu YL, Yan ZX, Xie XY, Xiang Z, Yin L, Wang QQ, Chong DC, Xue GL, Xu LL, Zhou K, Wang Q. Elevated LOXL2 expression by LINC01347/miR-328-5p axis contributes to 5-FU chemotherapy resistance of colorectal cancer. Am J Cancer Res 2021; 11:1572-1585. [PMID: 33948374 PMCID: PMC8085882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 10/22/2020] [Indexed: 06/12/2023] Open
Abstract
Chemotherapy resistance after curative surgery is a major contributor to the mortality of colorectal cancer (CRC). Detailed mechanism studies of specific molecular alterations are critical to improving the available therapies for long-term disease administration. We explored the functional role of LINC01347 in chemotherapy resistance of CRC. Elevated LINC01347 expression was correlated with CRC disease progression during chemotherapy treatment. However, the functional role of LINC01347 and mechanism remained undefined. In this study, we demonstrated that elevated LINC01347 expression was correlated with late clinical stage and poor prognosis in CRC tumor tissues with TCGA data. Exogenous LINC01347 expression promoted cell proliferation and 5-FU resistance of CRC cells, while LINC01347 knockdown attenuated cell growth and 5-FU resistance in vitro and in vivo. Molecular analysis indicated that LINC01347 participated in the transcriptional regulation of LOXL2 by sponging miR-328-5p. LOXL2 knockdown impaired the LINC01347 overexpression induced 5-FU resistance in CRC cells. The clinical analysis supported miR-328-5p/LOXL2 as a candidate biomarker for chemotherapy resistance of CRC patients. Our study provided a molecular basis for the development of 5-FU based chemotherapy resistance in CRC by LINC01347/miR-328/LOXL2 axis. We identified LINC01347 as a prognostic biomarker and potential therapeutic target against 5-FU based chemotherapy resistance of CRC.
Collapse
Affiliation(s)
- Gui-Li Zheng
- Department of Oncology, 960 Hospital of PLAJinan 250031, China
| | - Yu-Lin Liu
- Clinical Laboratory, Navy 971 Hospital of PLAQingdao 266071, China
| | - Ze-Xuan Yan
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Army Medical UniversityChongqing 400038, China
| | - Xiao-Ye Xie
- Department of Oncology, 960 Hospital of PLAJinan 250031, China
| | - Zhuo Xiang
- Department of Pharmacy, Navy 971 Hospital of PLAQingdao 266071, China
| | - Li Yin
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Army Medical UniversityChongqing 400038, China
| | - Qing-Qing Wang
- Clinical Laboratory, Navy 971 Hospital of PLAQingdao 266071, China
| | - Dao-Chen Chong
- Clinical Laboratory, Navy 971 Hospital of PLAQingdao 266071, China
| | - Guo-Liang Xue
- Department of Oncology, 960 Hospital of PLAJinan 250031, China
| | - Li-Li Xu
- Clinical Laboratory, Navy 971 Hospital of PLAQingdao 266071, China
| | - Kai Zhou
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Army Medical UniversityChongqing 400038, China
| | - Qiang Wang
- Department of Oncology, 960 Hospital of PLAJinan 250031, China
- Clinical Laboratory, Navy 971 Hospital of PLAQingdao 266071, China
| |
Collapse
|
27
|
Shimizu S, Sakai K, Chikugo T, Satou T, Shiraishi N, Mitsudomi T, Nishio K. Integrin-linked kinase pathway in heterogeneous pulmonary sarcomatoid carcinoma. Oncol Lett 2021; 21:320. [PMID: 33692852 DOI: 10.3892/ol.2021.12582] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Accepted: 12/14/2020] [Indexed: 12/23/2022] Open
Abstract
Pulmonary sarcomatoid carcinoma (PSC) is classified as poorly differentiated, and non-small cell lung carcinomas that contained a component of sarcoma or sarcoma-like differentiation are rare. The underlying carcinogenetic mechanism governing PSC remains unclear. The current study investigated the underlying carcinogenetic mechanism of PSC based on the hypothesis that it involves the epithelial-mesenchymal transition (EMT) process. Mutation analysis of PSCs, including carcinosarcoma, pleomorphic carcinoma and epithelial carcinoma specimens, was performed using targeted deep sequencing, whole transcriptome analysis and digital spatial profiling (DSP). PSCs exhibit a distinct mutation profile, with TP53, SYNE1 and APC mutations. Therefore, clustering of the gene expression profiles allowed the PSCs to be distinguished from the epithelial carcinomas. Increased gene expression of fibronectin in PSC was an important contributor to differential profiles. Pathway analysis revealed enhanced activity of the integrin-linked kinase (ILK) signaling pathway in the PSCs. DSP analysis using 56 antibodies of marker proteins confirmed significantly higher expression of fibronectin in PSCs. Intratumor heterogeneity of fibronectin expression was observed in sarcoma components. In conclusion, epithelial-mesenchymal transition process mediated by ILK signaling may be associated with carcinogenetic mechanisms of PSC. Overexpression of fibronectin mediated by ILK signaling appears to serve a role in the EMT involved in the PSC transformation process.
Collapse
Affiliation(s)
- Shigeki Shimizu
- Department of Diagnostic Pathology, Kindai University Hospital, Osaka-Sayama, Osaka 589-8511, Japan
| | - Kazuko Sakai
- Department of Genome Biology, Kindai University Faculty of Medicine, Osaka-Sayama, Osaka 589-8511, Japan
| | - Takaaki Chikugo
- Department of Diagnostic Pathology, Kindai University Hospital, Osaka-Sayama, Osaka 589-8511, Japan
| | - Takao Satou
- Department of Diagnostic Pathology, Kindai University Hospital, Osaka-Sayama, Osaka 589-8511, Japan
| | - Naoki Shiraishi
- Department of Diagnostic Pathology, Kindai University Hospital, Osaka-Sayama, Osaka 589-8511, Japan
| | - Tetsuya Mitsudomi
- Department of Thoracic Surgery, Kindai University Faculty of Medicine, Osaka-Sayama, Osaka 589-8511, Japan
| | - Kazuto Nishio
- Department of Genome Biology, Kindai University Faculty of Medicine, Osaka-Sayama, Osaka 589-8511, Japan
| |
Collapse
|
28
|
Zhang J, Song Q, Wu M, Zheng W. The Emerging Roles of Exosomes in the Chemoresistance of Hepatocellular Carcinoma. Curr Med Chem 2021; 28:93-109. [PMID: 32000636 DOI: 10.2174/0929867327666200130103206] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 10/08/2019] [Accepted: 10/23/2019] [Indexed: 11/22/2022]
Abstract
Hepatocellular carcinoma (HCC) is a common gastrointestinal malignancy with a leading incidence of cancer-related mortality worldwide. Despite the progress of treatment options, there remains low efficacy for patients with intermediate-advanced HCC, due to tumor metastasis, recurrence and chemoresistance. Increasing evidence suggests that exosomes in the tumor microenvironment (TME), along with other extracellular vesicles (EVs) and cytokines, contribute to the drug chemosensitivity of cancer cells. Exosomes, the intercellular communicators in various biological activities, have shown to play important roles in HCC progression. This review summarizes the underlying associations between exosomes and chemoresistance of HCC cells. The exosomes derived from distinct cell types mediate the drug resistance by regulating drug efflux, epithelial-mesenchymal transition (EMT), cancer stem cell (CSC) properties, autophagic phenotypes, as well as the immune response. In summary, TME-related exosomes can be a potential target to reverse chemoresistance and a candidate biomarker of drug efficacy in HCC patients.
Collapse
Affiliation(s)
- Jie Zhang
- Department of Chemotherapy, Affiliated Hospital of Nantong University, 20 Xisi Road, 226001 Nantong, Jiangsu, China
| | - Qianqian Song
- Department of Radiology, Wake Forest School of Medicine, One Medical Center Boulevard, Winston-Salem, 27157 NC, United States
| | - Mengna Wu
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, 20 Xisi Road, 226001 Nantong, Jiangsu, China
| | - Wenjie Zheng
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, 20 Xisi Road, 226001 Nantong, Jiangsu, China
| |
Collapse
|
29
|
Katoch A, Nayak D, Faheem MM, Kumar A, Sahu PK, Gupta AP, Kumar LD, Goswami A. Natural podophyllotoxin analog 4DPG attenuates EMT and colorectal cancer progression via activation of checkpoint kinase 2. Cell Death Discov 2021; 7:25. [PMID: 33500399 PMCID: PMC7838189 DOI: 10.1038/s41420-021-00405-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 10/17/2020] [Accepted: 11/13/2020] [Indexed: 02/07/2023] Open
Abstract
Epithelial–mesenchymal transition (EMT) is critical for the metastatic dissemination of cancer cells and contributes to drug resistance. In this study, we observed that epithelial colorectal cancer (CRC) cells transiently exposed to 5-fluorouracil (5-FU) (a chemotherapeutic drug for CRC) as well as 5-FU-resistant cells (5-FU-R) develop EMT characters as evidenced by activation of Vimentin and augmented invasive properties. On the other hand, 4DPG (4′-demethyl-deoxypodophyllotoxin glucoside), a natural podophyllotoxin analog attenuates EMT and invadopodia formation abilities of HCT-116/5-FU-R and SW-620/5-FU-R cells. Treatment with 4DPG restrains Vimentin phosphorylation (Ser38) in 5-FU-R cells, along with downregulation of mesenchymal markers Twist1 and MMP-2 while augmenting the expression of epithelial markers E-cadherin and TIMP-1. Moreover, 4DPG boosts the tumor-suppressor protein, checkpoint kinase 2 (Chk2) via phosphorylation at Thr68 in a dose-dependent manner in 5-FU-R cells. Mechanistically, SiRNA-mediated silencing of Chk2, as well as treatment with Chk2-specific small-molecule inhibitor (PV1019), divulges that 4DPG represses Vimentin activation in a Chk2-dependent manner. Furthermore, immunoprecipitation analysis unveiled that 4DPG prevents complex formation between Vimentin and p53 resulting in the rescue of p53 and its nuclear localization in aggressive 5-FU-R cells. In addition, 4DPG confers suitable pharmacokinetic properties and strongly abrogates tumor growth, polyps formation, and lung metastasis in an orthotopic rat colorectal carcinoma model. In conclusion, our findings demonstrate 4DPG as a targeted antitumor/anti-metastatic pharmacological lead compound to circumvent EMT-associated drug resistance and suggest its clinical benefits for the treatment of aggressive cancers.
Collapse
Affiliation(s)
- Archana Katoch
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, 201002, India.,Cancer Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu, Jammu and Kashmir, 180001, India
| | - Debasis Nayak
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, 43210, USA
| | - Mir Mohd Faheem
- Cancer Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu, Jammu and Kashmir, 180001, India.,School of Biotechnology, University of Jammu, Jammu, Jammu and Kashmir, 180006, India
| | - Aviral Kumar
- Cancer Biology, CSIR-Centre for Cellular & Molecular Biology, Hyderabad, Telangana, 500007, India
| | - Promod Kumar Sahu
- Cancer Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu, Jammu and Kashmir, 180001, India
| | - Ajai Prakash Gupta
- Quality Control and Quality Assurance Division, CSIR-Indian Institute of Integrative Medicine, Jammu, Jammu and Kashmir, 180001, India
| | - Lekha Dinesh Kumar
- Cancer Biology, CSIR-Centre for Cellular & Molecular Biology, Hyderabad, Telangana, 500007, India
| | - Anindya Goswami
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, 201002, India. .,Cancer Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu, Jammu and Kashmir, 180001, India.
| |
Collapse
|
30
|
Zang Y, Dong Q, Lu Y, Dong K, Wang R, Liang Z. Lumican inhibits immune escape and carcinogenic pathways in colorectal adenocarcinoma. Aging (Albany NY) 2021; 13:4388-4408. [PMID: 33493133 PMCID: PMC7906189 DOI: 10.18632/aging.202401] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 11/23/2020] [Indexed: 12/18/2022]
Abstract
Lumican (LUM), a small leucine-rich proteoglycan, is a component of the extracellular matrix. Abnormal LUM expression is potentially associated with cancer progression. In the present study, we confirmed high LUM mRNA expression in colorectal adenocarcinoma (COAD) through the UALCAN database. The Kaplan-Meier method, univariate, and multivariate COX analysis showed that high LUM expression is an independent determinant of poor prognosis in COAD. A COX regression model was constructed based on clinical information and LUM expression. The receiver operating characteristic (ROC) curve indicated that this model was highly accurate in monitoring COAD prognosis. The co-expression network of LUM was determined by LinkedOmics, which showed that LUM expression was closely related to immune escape and the miR200 family. Furthermore, we studied the co-expression network of LUM and found that LUM could promote tumor metastasis and invasion. The Tumor Immune Estimation Resource website showed that LUM was closely related to immune infiltration and correlated with regulatory T cells, tumour-associated macrophages, and dendritic cells. We found that LUM cultivated cancer progression by targeting the miR200 family to promote epithelial-to-mesenchymal transition. These findings suggest that LUM is a potential target for inhibiting immune escape and carcinogenic pathways.
Collapse
Affiliation(s)
- Yiqing Zang
- Department of Otorhinolaryngology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Qiuping Dong
- Department of Cancer Cell Biology, Tianjin's Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China
| | - Yi Lu
- Department of Otorhinolaryngology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Kaiti Dong
- Department of Otorhinolaryngology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Rong Wang
- Department of Laboratory Medicine, Tianjin Medical University, Tianjin 300060, China
| | - Zheng Liang
- Department of Otorhinolaryngology, Tianjin Medical University General Hospital, Tianjin 300052, China
| |
Collapse
|
31
|
Jokela TA, LaBarge MA. Integration of mechanical and ECM microenvironment signals in the determination of cancer stem cell states. CURRENT STEM CELL REPORTS 2020; 7:39-47. [PMID: 33777660 DOI: 10.1007/s40778-020-00182-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Purpose of review Cancer stem cells (CSCs) are increasingly understood to play a central role in tumor progression. Growing evidence implicates tumor microenvironments as a source of signals that regulate or even impose CSC states on tumor cells. This review explores points of integration for microenvironment-derived signals that are thought to regulate CSCs in carcinomas. Recent findings CSC states are directly regulated by the mechanical properties and extra cellular matrix (ECM) composition of tumor microenvironments that promote CSC growth and survival, which may explain some modes of therapeutic resistance. CSCs sense mechanical forces and ECM composition through integrins and other cell surface receptors, which then activate a number of intracellular signaling pathways. The relevant signaling events are dynamic and context-dependent. Summary CSCs are thought to drive cancer metastases and therapeutic resistance. Cells that are in CSC states and more differentiated states appear to be reversible and conditional upon the components of the tumor microenvironment. Signals imposed by tumor microenvironment are of a combinatorial nature, ultimately representing the integration of multiple physical and chemical signals. Comprehensive understanding of the tumor microenvironment-imposed signaling that maintains cells in CSC states may guide future therapeutic interventions.
Collapse
Affiliation(s)
- Tiina A Jokela
- Department of Population Sciences, Beckman Research Institute, City of Hope, 1500 E. Duarte Rd, Duarte CA 91010
| | - Mark A LaBarge
- Department of Population Sciences, Beckman Research Institute, City of Hope, 1500 E. Duarte Rd, Duarte CA 91010
| |
Collapse
|
32
|
Yu X, Zhu L, Liu J, Xie M, Chen J, Li J. Emerging Role of Immunotherapy for Colorectal Cancer with Liver Metastasis. Onco Targets Ther 2020; 13:11645-11658. [PMID: 33223838 PMCID: PMC7671511 DOI: 10.2147/ott.s271955] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 10/29/2020] [Indexed: 02/05/2023] Open
Abstract
Colorectal cancer (CRC) is the third most common malignant tumor in the world and the second leading cause of cancer-related deaths, with the liver as the most common site of distant metastasis. The prognosis of CRC with liver metastasis is poor, and most patients cannot undergo surgery. In addition, conventional antitumor approaches such as chemotherapy, radiotherapy, targeted therapy, and surgery result in unsatisfactory outcomes. In recent years, immunotherapy has shown good prospects in the treatment of assorted tumors by enhancing the host's antitumor immune function, and it may become a new effective treatment for liver metastasis of CRC. However, challenges remain in applying immunotherapy to CRC with liver metastasis. This review examines how the microenvironment and immunosuppressive landscape of the liver favor tumor progression. It also highlights the latest research advances in immunotherapy for colorectal liver metastasis and identifies immunotherapy as a treatment regimen with a promising future in clinical applications.
Collapse
Affiliation(s)
- Xianzhe Yu
- Gastrointestinal Department, The Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou Province, People’s Republic of China
| | - Lingling Zhu
- Lung Cancer Center, West China Hospital of Sichuan University, Chengdu, Sichuan Province, People’s Republic of China
| | - Jiewei Liu
- Lung Cancer Center, West China Hospital of Sichuan University, Chengdu, Sichuan Province, People’s Republic of China
| | - Ming Xie
- Gastrointestinal Department, The Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou Province, People’s Republic of China
| | - Jiang Chen
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, Zhejiang Province, People’s Republic of China
| | - Jianguo Li
- Gastrointestinal Department, The Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou Province, People’s Republic of China
| |
Collapse
|
33
|
Abuderman AA, Harb OA, Gertallah LM. Prognostic and clinic-pathological significances of HOXB8, ILK and FAT4 expression in colorectal cancer. Contemp Oncol (Pozn) 2020; 24:183-192. [PMID: 33235545 PMCID: PMC7670183 DOI: 10.5114/wo.2020.100281] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Accepted: 10/11/2020] [Indexed: 12/12/2022] Open
Abstract
INTRODUCTION HOXB8 is a protein that was found to promote cancer proliferation and invasion. ILK is a protein kinase which has a role in carcinogenesis. FAT4 is a tumor homologue that has a role in EMT and autophagy regulation. AIM OF THE STUDY To identify expression of Human HOXB8, Integrin-linked kinase (ILK1) and FAT homolog 4 (FAT4) in colorectal cancer (CRC) correlating their expression with pathological, prognostic and clinical parameters of CRC. MATERIAL AND METHODS We assessed the expression of HOXB8, ILK and FAT4 in fifty CRC patients and ten samples from nearby non-neoplastic colonic mucosa using immunohistochemistry. RESULTS The expression of HOXB8 and ILK in CRC was positively associated with high tumor grade, advanced tumor stage, lymph node involvement (p < 0.001), occurrence of distant metastases (p = 0.003 and 0.024 respectively), higher incidence of tumor recurrence (p = 0.03, p < 0.001 respectively), worse survival rates (p = 0.038 and 0.003 respectively). The expression of FAT4 in CRC was correlated with lower grade, early stage of the tumor, absence of lymph node involvement (p < 0.001) and lack of distant metastases (p = 0.011). High FAT4 expression was associated with absence of tumor recurrence (p < 0.001) and favorable survival rates (p < 0.001 and 0.003). CONCLUSIONS High immunohistochemical expression of HOXB8 and ILK in addition to low immunohistochemical expression of FAT4 was associated with unfavorable prognostic and pathological parameters of CRC.
Collapse
Affiliation(s)
- Abdulwahab A. Abuderman
- Department of Basic Medical Sciences, College of Medicine, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Ola A. Harb
- Department of Pathology, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Loay M. Gertallah
- Department of General Surgery, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| |
Collapse
|
34
|
Zheng Q, Wang J, Li W, Chen X, Chen S, Chen L. Emodin Reverses the Epithelial-Mesenchymal Transition of Human Endometrial Stromal Cells by Inhibiting ILK/GSK-3β Pathway. DRUG DESIGN DEVELOPMENT AND THERAPY 2020; 14:3663-3672. [PMID: 32982173 PMCID: PMC7490435 DOI: 10.2147/dddt.s262816] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 08/20/2020] [Indexed: 12/12/2022]
Abstract
Purpose To explore the exact mechanism through which emodin down-regulates the migration and invasion abilities of endometrial stromal cells. Moreover, to explore the theoretical basis of emodin in the treatment of endometriosis. Patients and Methods Endometriosis endometrial stromal cells (EESs) were cultured from 15 women with endometriosis and control endometrial stromal cells (CESs) were cultured from 12 women without endometriosis. The levels of proteins were evaluated by Western blot. The migration and invasion abilities of cells were detected by transwell assays. Results The abilities of migration and invasion of EESs were much stronger than those of CESs. After treated with emodin, the migration and invasion abilities of EESs and CESs were significantly down-regulated, and the levels of integrin-linked kinase (ILK) and p-GSK-3β were statistically down-regulated in EESs. Besides that, the expression of keratin was up-regulated while the expression of vimentin, β-catenin and slug were all down-regulated by emodin in a dose- and time-dependent manner. Silencing of ILK gene in EESs also achieved the above effects, which were strengthened by emodin. Conversely, exogenous expression of ILK in CESs increased the expression of p-GSK-3β, which were abrogated by emodin. Furthermore, SB216763 increased migration and invasion abilities of CESs by facilitating the epithelial-mesenchymal transition (EMT) through up-regulating levels of p-GSK-3β, β-catenin and slug, which were also abrogated by emodin. Conclusion Emodin inhibits the migration and invasion abilities of human endometrial stromal cells by reversing the EMT via ILK/GSK-3β pathway. So, emodin may be considered as a promising targeted therapy for endometriosis.
Collapse
Affiliation(s)
- Qiaomei Zheng
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Fujian Medical University, Fujian Medical University, Fuzhou, Fujian 350001, People's Republic of China
| | - Jinhua Wang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Fujian Medical University, Fujian Medical University, Fuzhou, Fujian 350001, People's Republic of China
| | - Wenwen Li
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Fujian Medical University, Fujian Medical University, Fuzhou, Fujian 350001, People's Republic of China
| | - Xiaoyun Chen
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Fujian Medical University, Fujian Medical University, Fuzhou, Fujian 350001, People's Republic of China
| | - Shaozhan Chen
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Fujian Medical University, Fujian Medical University, Fuzhou, Fujian 350001, People's Republic of China
| | - Lihong Chen
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Fujian Medical University, Fujian Medical University, Fuzhou, Fujian 350001, People's Republic of China
| |
Collapse
|
35
|
Zhang J, Miller Z, Musich PR, Thomas AE, Yao ZQ, Xie Q, Howe PH, Jiang Y. DSTYK Promotes Metastasis and Chemoresistance via EMT in Colorectal Cancer. Front Pharmacol 2020; 11:1250. [PMID: 32982725 PMCID: PMC7493073 DOI: 10.3389/fphar.2020.01250] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 07/30/2020] [Indexed: 12/11/2022] Open
Abstract
Objective Tumor metastasis and resistance to chemotherapy are two critical factors that contribute to the high death rate of colorectal cancer (CRC) patients. Metastasis is facilitated by the epithelial-mesenchymal transition (EMT) of tumor cells, which has emerged not only as a fundamental process during metastasis, but is also a key process leading to chemoresistance of cancer cells. However, the underlying mechanisms of EMT in CRC cell remain unknown. Here, we aim to assess the role of dual serine/threonine and tyrosine protein kinase (DSTYK) in CRC metastasis and chemoresistance. Methods To study the role of DSTYK in TGF-β-induced EMT, we employed techniques including Crispr/Cas9 knockout (KO) to generate DSTYK KO cell lines, RT-PCR to detect the mRNA expression, immunofluorescence analyses, and western blots to detect protein levels of DSTYK in the following 4 cell lines: control LS411N-TβRII and LS411N-TβRII/DSTYK KO, control LS513 and LS513/DSTYK KO cells, treated with/without TGF-β. The effects of DSTYK on apoptosis were investigated by MTT assays, flow cytometry assays, and TUNEL assays. The expression of DSTYK in CRC patients and its correlation with EMT markers were determined by bioinformatics analysis. For in vivo analysis, both xenograft and orthotopic tumor mouse models were employed to investigate the function of DSTYK in chemoresistance and metastasis of tumors. Results In this study, we demonstrate that the novel kinase DSTYK promotes both TGF-β-induced EMT and the subsequent chemoresistance in CRC cells. DSTYK KO significantly attenuates TGF-β–induced EMT and chemoresistance in CRC cells. According to the Gene Expression Omnibus (GEO) database, the expression of DSTYK is not only positively correlated to the expression of TGF-β, but proportional to the death rate of CRC patients as well. Evidently, the expression of DSTYK in the metastatic colorectal cancer samples from patients was significantly higher than that of primary colorectal cancer samples. Further, we demonstrate in mouse models that chemotherapeutic drug treatment suppresses the growth of DSTYK KO tumors more effectively than control tumors. Conclusion Our findings identify DSTYK as a novel protein kinase in regulating TGF-β–mediated EMT and chemoresistance in CRC cells, which defines DSTYK as a potential therapeutic target for CRC therapy.
Collapse
Affiliation(s)
- Jinyu Zhang
- Department of Biomedical Sciences, J. H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States.,Division of Infectious, Inflammatory and Immunologic Diseases, Department of Internal Medicine, Quillen College of Medicine, ETSU, Johnson City, TN, United States
| | - Zachary Miller
- Department of Biomedical Sciences, J. H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
| | - Phillip R Musich
- Department of Biomedical Sciences, J. H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
| | - Ashlin E Thomas
- Department of Biomedical Sciences, J. H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
| | - Zhi Q Yao
- Division of Infectious, Inflammatory and Immunologic Diseases, Department of Internal Medicine, Quillen College of Medicine, ETSU, Johnson City, TN, United States
| | - Qian Xie
- Department of Biomedical Sciences, J. H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
| | - Philip H Howe
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, United States
| | - Yong Jiang
- Department of Biomedical Sciences, J. H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
| |
Collapse
|
36
|
Zhan Y, Wang H, Ning Y, Zheng H, Liu S, Yang Y, Zhou M, Fan S. Understanding the roles of stress granule during chemotherapy for patients with malignant tumors. Am J Cancer Res 2020; 10:2226-2241. [PMID: 32905441 PMCID: PMC7471355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Accepted: 07/19/2020] [Indexed: 06/11/2023] Open
Abstract
The assembly of stress granules (SGs) is a conserved mechanism to regulate protein synthesis under cell stress, where the translation of global protein is silenced and selective protein synthesis for survival maintains. SG formation confers survival advantages and chemotherapeutic resistance to malignant cells. Targeting SG assembly may represent a potential treatment strategy to overcome the primary and acquired chemotherapeutic resistance and enhance curative effect. We conduct a comprehensive review of the published literatures focusing on the drugs that potentially induce SGs and the related mechanism, retrospect the relationship between SGs and drug resistance related proteins, illuminate the regulated pathways and potential targets for SG assembly, and discuss future directions of overcoming the resistance to chemotherapy.
Collapse
Affiliation(s)
- Yuting Zhan
- Department of Pathology, The Second Xiangya Hospital, Central South UniversityChangsha 410011, Hunan, China
| | - Haihua Wang
- Department of Pathology, The Second Xiangya Hospital, Central South UniversityChangsha 410011, Hunan, China
| | - Yue Ning
- Department of Pathology, The Second Xiangya Hospital, Central South UniversityChangsha 410011, Hunan, China
| | - Hongmei Zheng
- Department of Pathology, The Second Xiangya Hospital, Central South UniversityChangsha 410011, Hunan, China
| | - Sile Liu
- Department of Pathology, The Second Xiangya Hospital, Central South UniversityChangsha 410011, Hunan, China
| | - Yang Yang
- Department of Pathology, The Second Xiangya Hospital, Central South UniversityChangsha 410011, Hunan, China
| | - Ming Zhou
- Cancer Research Institute Xiangya School of Medicine, Central South UniversityChangsha 410078, Hunan, China
| | - Songqing Fan
- Department of Pathology, The Second Xiangya Hospital, Central South UniversityChangsha 410011, Hunan, China
| |
Collapse
|
37
|
Gaponova AV, Rodin S, Mazina AA, Volchkov PV. Epithelial-Mesenchymal Transition: Role in Cancer Progression and the Perspectives of Antitumor Treatment. Acta Naturae 2020; 12:4-23. [PMID: 33173593 PMCID: PMC7604894 DOI: 10.32607/actanaturae.11010] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 05/20/2020] [Indexed: 12/12/2022] Open
Abstract
About 90% of all malignant tumors are of epithelial nature. The epithelial tissue is characterized by a close interconnection between cells through cell-cell interactions, as well as a tight connection with the basement membrane, which is responsible for cell polarity. These interactions strictly determine the location of epithelial cells within the body and are seemingly in conflict with the metastatic potential that many cancers possess (the main criteria for highly malignant tumors). Tumor dissemination into vital organs is one of the primary causes of death in patients with cancer. Tumor dissemination is based on the so-called epithelial-mesenchymal transition (EMT), a process when epithelial cells are transformed into mesenchymal cells possessing high mobility and migration potential. More and more studies elucidating the role of the EMT in metastasis and other aspects of tumor progression are published each year, thus forming a promising field of cancer research. In this review, we examine the most recent data on the intracellular and extracellular molecular mechanisms that activate EMT and the role they play in various aspects of tumor progression, such as metastasis, apoptotic resistance, and immune evasion, aspects that have usually been attributed exclusively to cancer stem cells (CSCs). In conclusion, we provide a detailed review of the approved and promising drugs for cancer therapy that target the components of the EMT signaling pathways.
Collapse
Affiliation(s)
- A. V. Gaponova
- Moscow Institute of Physics and Technology, Dolgoprudny, Moscow Region, 141701 Russia
| | - S. Rodin
- Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, 17177 Sweden
| | - A. A. Mazina
- Moscow Institute of Physics and Technology, Dolgoprudny, Moscow Region, 141701 Russia
| | - P. V. Volchkov
- Moscow Institute of Physics and Technology, Dolgoprudny, Moscow Region, 141701 Russia
| |
Collapse
|
38
|
Nikou S, Arbi M, Dimitrakopoulos FID, Sirinian C, Chadla P, Pappa I, Ntaliarda G, Stathopoulos GT, Papadaki H, Zolota V, Lygerou Z, Kalofonos HP, Bravou V. Integrin-linked kinase (ILK) regulates KRAS, IPP complex and Ras suppressor-1 (RSU1) promoting lung adenocarcinoma progression and poor survival. J Mol Histol 2020; 51:385-400. [PMID: 32592097 DOI: 10.1007/s10735-020-09888-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Accepted: 06/13/2020] [Indexed: 12/11/2022]
Abstract
Integrin-linked kinase (ILK) forms a heterotrimeric protein complex with PINCH and PARVIN (IPP) in Focal Adhesions (FAs) that acts as a signaling platform between the cell and its microenvironment regulating important cancer-related functions. We aimed to elucidate the role of ILK in lung adenocarcinoma (LUADC) focusing on a possible link with KRAS oncogene. We used immunohistochemistry on human tissue samples and KRAS-driven LUADC in mice, analysis of large scale publicly available RNA sequencing data, ILK overexpression and pharmacological inhibition as well as knockdown of KRAS in lung cancer cells. ILK, PINCH1 and PARVB (IPP) proteins are overexpressed in human LUADC and KRAS-driven LUADC in mice representing poor prognostic indicators. Genes implicated in ILK signaling are significantly enriched in KRAS-driven LUADC. Silencing of KRAS, as well as, overexpression and pharmacological inhibition of ILK in lung cancer cells provide evidence of a two-way association between ILK and KRAS. Upregulation of PINCH, PARVB and Ras suppressor-1 (RSU1) expression was demonstrated in ILK overexpressing lung cancer cells in addition to a significant positive correlation between these factors in tissue samples, while KRAS silencing downregulates IPP and RSU1. Pharmacological inhibition of ILK in KRAS mutant lung cancer cells suppresses cell growth, migration, EMT and increases sensitivity to platinum-based chemotherapy. ILK promotes an aggressive lung cancer phenotype with prognostic and therapeutic value through functions that involve KRAS, IPP complex and RSU1, rendering ILK a promising biomarker and therapeutic target in lung adenocarcinoma.
Collapse
Affiliation(s)
- Sofia Nikou
- Department of Anatomy-Histology-Embryology, Medical School, University of Patras, 26500, Patras, Greece
| | - Marina Arbi
- Department of General Biology, Medical School, University of Patras, 26504, Patras, Greece
| | | | - Chaido Sirinian
- Clinical and Molecular Oncology Laboratory, Division of Oncology, Medical School, University of Patras, 26504, Rio, Greece
| | - Panagiota Chadla
- Department of Anatomy-Histology-Embryology, Medical School, University of Patras, 26500, Patras, Greece
| | - Ioanna Pappa
- Department of Anatomy-Histology-Embryology, Medical School, University of Patras, 26500, Patras, Greece
| | - Giannoula Ntaliarda
- Laboratory for Molecular Respiratory Carcinogenesis, Department of Physiology, Faculty of Medicine, University of Patras, 2504, Rio, Achaia, Greece
| | - Georgios T Stathopoulos
- Laboratory for Molecular Respiratory Carcinogenesis, Department of Physiology, Faculty of Medicine, University of Patras, 2504, Rio, Achaia, Greece.,Comprehensive Pneumology Center (CPC) and Institute for Lung Biology and Disease (iLBD), University Hospital, Ludwig-Maximilians University and Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Bavaria, Germany
| | - Helen Papadaki
- Department of Anatomy-Histology-Embryology, Medical School, University of Patras, 26500, Patras, Greece
| | - Vasiliki Zolota
- Department of Pathology, University Hospital of Patras, 26504, Patras, Greece
| | - Zoi Lygerou
- Department of General Biology, Medical School, University of Patras, 26504, Patras, Greece
| | - Haralabos P Kalofonos
- Clinical and Molecular Oncology Laboratory, Division of Oncology, Medical School, University of Patras, 26504, Rio, Greece.,Division of Oncology, Department of Internal Medicine, University Hospital of Patras, 26504, Rio, Greece
| | - Vasiliki Bravou
- Department of Anatomy-Histology-Embryology, Medical School, University of Patras, 26500, Patras, Greece.
| |
Collapse
|
39
|
Li H, Feng Z, He ML. Lipid metabolism alteration contributes to and maintains the properties of cancer stem cells. Theranostics 2020; 10:7053-7069. [PMID: 32641978 PMCID: PMC7330842 DOI: 10.7150/thno.41388] [Citation(s) in RCA: 109] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 04/28/2020] [Indexed: 12/11/2022] Open
Abstract
Lipids, the basic components of the cell membrane, execute fundamental roles in almost all the cell activities including cell-cell recognition, signalling transduction and energy supplies. Lipid metabolism is elementary for life sustentation that balances activity between synthesis and degradation. An accumulating amount of data has indicated abnormal lipid metabolism in cancer stem cells (CSCs), and that the alteration of lipid metabolism exerts a great impact on CSCs' properties such as the capability of self-renewal, differentiation, invasion, metastasis, and drug sensitivity and resistance. CSCs' formation and maintenance cannot do without the regulation of fatty acids and cholesterol. In normal cells and embryonic development, fatty acids and cholesterol metabolism are regulated by some important signalling pathways (such as Hedgehog, Notch, Wnt signalling pathways); these signalling pathways also play crucial roles in initiating and/or maintaining CSCs' properties, and such signalling is shown to be commonly modulated by the abnormal lipid metabolism in CSCs; on the other hand, the altered lipid metabolism in turn modifies the cell signalling and generates additional impacts on CSCs. Metabolic rewiring is considered as an ideal hallmark of CSCs, and metabolic alterations would be promising therapeutic targets of CSCs for aggressive tumors. In this review, we summarize the most updated findings of lipid metabolic abnormalities in CSCs and prospect the potential applications of targeting lipid metabolism for anticancer treatment.
Collapse
|
40
|
Cheng C, Huang Z, Zhou R, An H, Cao G, Ye J, Huang C, Wu D. Numb negatively regulates the epithelial-to-mesenchymal transition in colorectal cancer through the Wnt signaling pathway. Am J Physiol Gastrointest Liver Physiol 2020; 318:G841-G853. [PMID: 32146835 DOI: 10.1152/ajpgi.00178.2019] [Citation(s) in RCA: 15] [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] [Indexed: 01/31/2023]
Abstract
Colorectal cancer (CRC) is one of the most common malignant tumors and is associated with a high mortality rate due to the lack of specific biomarkers available for early diagnosis, targeted therapies, and prognostic surveillance. In the present study, we investigated the function of Numb and its underlying mechanism in CRC. Immunohistochemical staining and clinicopathological analysis were used to assess the expression of Numb and its clinical significance in patients with CRC. Quantitative real-time polymerase chain reaction, cell proliferation, Western blot, wound healing, Transwell, and TOP/FOP flash reporter assays were used to investigate the function of Numb and its underlying mechanism in CRC. Numb expression was downregulated and negatively correlated with the depth of invasion, tumor size, metastasis, TNM stage, and epithelial-to-mesenchymal transition (EMT) markers in CRC specimens. Numb negatively regulates the EMT, proliferation, invasion, migration, and the Wnt signaling pathway in vitro, as well as tumor growth and metastasis in vivo. Furthermore, activation of the Wnt signaling pathway by Wnt-3A negated the effect of Numb overexpression, whereas inhibition of the Wnt signaling pathway by IWR-1 impaired the effect of the Numb knockdown on the EMT. We concluded that Numb downregulation is a common event in patients with CRC and is closely correlated with cancer progression and a poor prognosis. Numb functions as a tumor suppressor in CRC, and its tumor suppressor function is mediated by negative regulation of the EMT through the Wnt signaling pathway.NEW & NOTEWORTHY We investigate the function of Numb and its underlying mechanism in colorectal cancer through quantitative real-time polymerase chain reaction, cell proliferation, Western blot, wound healing, Transwell, and TOP/FOP flash reporter assays. We conclude that Numb can negatively regulate the epithelial-to-mesenchymal transition through the Wnt signaling pathway to inhibit the development of colorectal cancer.
Collapse
Affiliation(s)
- Chi Cheng
- Department of Gastrointestinal Surgery, Ruian People's Hospital, Ruian, Zhejiang, China
| | - Zhenfeng Huang
- Department of Gastrointestinal Surgery, Ruian People's Hospital, Ruian, Zhejiang, China
| | - Ruiyao Zhou
- Department of Gastrointestinal Surgery, Ruian People's Hospital, Ruian, Zhejiang, China
| | - Huimin An
- Department of Pathology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Zhejiang, China
| | - Gaojian Cao
- Department of Gastrointestinal Surgery, Ruian People's Hospital, Ruian, Zhejiang, China
| | - Jun Ye
- Department of Laboratory Medicine, The Second Affiliated Hospital of Guizhou Medical University, Guizhou, China
| | - Chaolin Huang
- Department of Obstetrics and Gynecology, Chengdu First People's Hospital, Chengdu, Sichuan, China
| | - Daoyi Wu
- Department of Gastrointestinal Surgery, Ruian People's Hospital, Ruian, Zhejiang, China
| |
Collapse
|
41
|
Ribatti D, Tamma R, Annese T. Epithelial-Mesenchymal Transition in Cancer: A Historical Overview. Transl Oncol 2020; 13:100773. [PMID: 32334405 PMCID: PMC7182759 DOI: 10.1016/j.tranon.2020.100773] [Citation(s) in RCA: 595] [Impact Index Per Article: 119.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 03/26/2020] [Accepted: 04/01/2020] [Indexed: 12/12/2022] Open
Abstract
Epithelial-mesenchymal transitions (EMTs), the acquisition of mesenchymal features from epithelial cells, occur during some biological processes and are classified into three types: the first type occurs during embryonic development, the second type is associated with adult tissue regeneration, and the third type occurs in cancer progression. EMT occurring during embryonic development in gastrulation, renal development, and the origin and fate of the neural crest is a highly regulated process, while EMT occurring during tumor progression is highly deregulated. EMT allows the solid tumors to become more malignant, increasing their invasiveness and metastatic activity. Secondary tumors frequently maintain the typical histologic characteristics of the primary tumor. These histologic features connecting the secondary metastatic tumors to the primary is due to a process called mesenchymal-epithelial transition (MET). MET has been demonstrated in different mesenchymal tumors and is the expression of the reversibility of EMT. EMT modulation could constitute an approach to avoid metastasis. Some of the targeted small molecules utilized as antiproliferative agents have revealed to inhibit EMT initiation or maintenance because EMT is regulated through signaling pathways for which these molecules have been designed.
Collapse
Affiliation(s)
- Domenico Ribatti
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, Bari, Italy.
| | - Roberto Tamma
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, Bari, Italy
| | - Tiziana Annese
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, Bari, Italy
| |
Collapse
|
42
|
Kottorou AE, Dimitrakopoulos FID, Antonacopoulou AG, Diamantopoulou G, Tsoumas D, Koutras A, Makatsoris T, Stavropoulos M, Thomopoulos KC, Hulbert A, Tzelepi V, Kalofonos HP. Differentially Methylated Ultra-Conserved Regions Uc160 and Uc283 in Adenomas and Adenocarcinomas Are Associated with Overall Survival of Colorectal Cancer Patients. Cancers (Basel) 2020; 12:895. [PMID: 32272654 PMCID: PMC7226527 DOI: 10.3390/cancers12040895] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 03/20/2020] [Accepted: 03/25/2020] [Indexed: 12/18/2022] Open
Abstract
: Deregulation of the transcribed ultra-conserved regions (T-UCRs) Uc160, Uc283, and Uc346 has been reported in colorectal cancer (CRC) recently. Here, we investigated promoter methylation of these T-UCRs during the adenoma-carcinoma sequence and their clinical significance in CRC patients. Methylation levels were assessed in CRC, adenomas, infiltrated lymph nodes, and metastatic tissue specimens. In situ hybridization was performed in representative tissue specimens. T-UCRs expression levels were also evaluated in HT-29 colon cancer cells before and after the acquired resistance to 5-fluorouracil (5-FU) and oxaliplatin. A gradual increase in T-UCRs methylation levels from hyperplastic polyps to adenomas and to in situ carcinomas (ISC) and a gradual decrease from ISC to infiltrative and metastatic carcinomas was observed (p < 0.001 for Uc160 and Uc283, p = 0.018 for Uc346). Uc160 and Uc283 methylation was associated with the grade of dysplasia in adenoma specimens (p = 0.034 and p = 0.019, respectively). Furthermore, higher Uc160 methylation, mainly in stage III and IV patients, was related to improved overall survival (OS) in univariate (p = 0.009; HR, 0.366) and multivariate analysis (p = 0.005; HR, 0.240). Similarly, higher methylation of Uc283 was associated with longer OS (p = 0.030). Finally, T-UCRs expression was significantly reduced in HT-29 cells after resistance to chemotherapy. This study suggests that promoter methylation of Uc160, Uc283, and Uc346 is altered during CRC development and that Uc160 and Uc283 methylation may have prognostic significance for CRC patients.
Collapse
Affiliation(s)
- Anastasia E. Kottorou
- Clinical and Molecular Oncology Laboratory, Division of Oncology, Medical School, University of Patras, 26504 Rio, Greece; (A.E.K.); (F.-I.D.D.); (A.G.A.); (D.T.); (A.K.); (T.M.)
| | - Foteinos-Ioannis D. Dimitrakopoulos
- Clinical and Molecular Oncology Laboratory, Division of Oncology, Medical School, University of Patras, 26504 Rio, Greece; (A.E.K.); (F.-I.D.D.); (A.G.A.); (D.T.); (A.K.); (T.M.)
| | - Anna G. Antonacopoulou
- Clinical and Molecular Oncology Laboratory, Division of Oncology, Medical School, University of Patras, 26504 Rio, Greece; (A.E.K.); (F.-I.D.D.); (A.G.A.); (D.T.); (A.K.); (T.M.)
| | - Georgia Diamantopoulou
- Division of Gastroenterology, University Hospital of Patras, 26504 Rio, Greece; (G.D.); (K.C.T.)
| | - Dimitrios Tsoumas
- Clinical and Molecular Oncology Laboratory, Division of Oncology, Medical School, University of Patras, 26504 Rio, Greece; (A.E.K.); (F.-I.D.D.); (A.G.A.); (D.T.); (A.K.); (T.M.)
| | - Angelos Koutras
- Clinical and Molecular Oncology Laboratory, Division of Oncology, Medical School, University of Patras, 26504 Rio, Greece; (A.E.K.); (F.-I.D.D.); (A.G.A.); (D.T.); (A.K.); (T.M.)
- Division of Oncology, Department of Internal Medicine, University Hospital of Patras, 26504 Rio, Greece
| | - Thomas Makatsoris
- Clinical and Molecular Oncology Laboratory, Division of Oncology, Medical School, University of Patras, 26504 Rio, Greece; (A.E.K.); (F.-I.D.D.); (A.G.A.); (D.T.); (A.K.); (T.M.)
- Division of Oncology, Department of Internal Medicine, University Hospital of Patras, 26504 Rio, Greece
| | | | | | - Alicia Hulbert
- Cancer Center, University of Illinois at Chicago School of Medicine, Chicago, IL 60612, USA;
- Department of Surgery, University of Illinois at Chicago School of Medicine, Chicago, IL 60612, USA
| | - Vassiliki Tzelepi
- Department of Pathology, Medical School, University of Patras, 26504 Rio, Greece;
| | - Haralabos P. Kalofonos
- Clinical and Molecular Oncology Laboratory, Division of Oncology, Medical School, University of Patras, 26504 Rio, Greece; (A.E.K.); (F.-I.D.D.); (A.G.A.); (D.T.); (A.K.); (T.M.)
- Division of Oncology, Department of Internal Medicine, University Hospital of Patras, 26504 Rio, Greece
| |
Collapse
|
43
|
Downstream Effectors of ILK in Cisplatin-Resistant Ovarian Cancer. Cancers (Basel) 2020; 12:cancers12040880. [PMID: 32260415 PMCID: PMC7226328 DOI: 10.3390/cancers12040880] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 03/31/2020] [Indexed: 12/14/2022] Open
Abstract
Despite good responses to first-line treatment with platinum-based combination chemotherapy, most ovarian cancer patients will relapse and eventually develop platinum-resistant disease with poor prognosis. Although reports suggest that integrin-linked kinase (ILK) is a potential target for ovarian cancer treatment, identification of ILK downstream effectors has not been fully explored. The purpose of this study was to investigate the molecular and biological effects of targeting ILK in cisplatin-resistant ovarian cancer. Western blot analysis showed that phosphorylation levels of ILK were higher in cisplatin-resistant compared with cisplatin-sensitive ovarian cancer cells. Further immunohistochemical analysis of ovarian cancer patient samples showed a significant increase in phosphorylated ILK levels in the tumor tissue when compared to normal ovarian epithelium. Targeting ILK by small-interfering RNA (siRNA) treatment reduced cisplatin-resistant cell growth and invasion ability, and increased apoptosis. Differential gene expression analysis by RNA sequencing (RNA-Seq) upon ILK-siRNA transfection followed by Ingenuity Pathway Analysis (IPA) and survival analysis using the Kaplan-Meier plotter database identified multiple target genes involved in cell growth, apoptosis, invasion, and metastasis, including several non-coding RNAs. Taken together, results from this study support ILK as an attractive target for ovarian cancer and provide potential ILK downstream effectors with prognostic and therapeutic value.
Collapse
|
44
|
Highly expressed CCR7 predicts poor prognosis in locally advanced nasopharyngeal carcinoma. Ir J Med Sci 2019; 189:669-676. [PMID: 31758524 DOI: 10.1007/s11845-019-02141-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 11/14/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND Nasopharyngeal carcinoma (NC) is a malignant human tumor with a high incidence that occurs on the top and lateral wall of the nasopharynx. AIMS To investigate the clinical value of chemokine receptor 7 (CCR7) in locally advanced NC. METHODS We enrolled 114 patients with locally advanced NC admitted to our hospital in the observation group (OBG) and 100 normal healthy subjects who underwent physical examination in the control group (COG). The serum CCR7 expression levels in each group were measured using enzyme-linked immunosorbent assay and were compared between the groups. RESULTS None of the 114 patients or their family members were lost during follow-up. Thirty-five patients died within 3 years and 79 survived (survival rate: 69.29%). The serum CCR7 level was higher in the OBG than in the COG (P < 0.05). The receiver operating characteristic (ROC) curve showed that the area under the ROC curve (AUC) was 0.837 for diagnostic value for locally advanced NC and the AUC was 0.759 for predictive value for 3-year mortality. The CCR7 AUC for diagnosis of locally advanced NC was 0.837 and for prediction of mortality was 0.759. Univariate analysis revealed significant differences in smoking history, long-term consumption of pickled food, family history of NC, primary lesion staging, lymph node staging, distant metastasis, clinical pathological staging, and CCR7 between the two groups (P < 0.05). CONCLUSIONS CCR7 was valuable in the diagnosis of locally advanced NC, and highly expressed CCR7 was predictive of poor prognosis.
Collapse
|
45
|
Proteomic Technology "Lens" for Epithelial-Mesenchymal Transition Process Identification in Oncology. Anal Cell Pathol (Amst) 2019; 2019:3565970. [PMID: 31781477 PMCID: PMC6855076 DOI: 10.1155/2019/3565970] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 08/20/2019] [Accepted: 09/10/2019] [Indexed: 02/08/2023] Open
Abstract
The epithelial-mesenchymal transition (EMT) is a complex transformation process that induces local and distant progression of many malignant tumours. Due to its complex array of proteins that are dynamically over-/underexpressed during this process, proteomic technologies gained their place in the EMT research in the last years. Proteomics has identified new molecular pathways of this process and brought important insights to develop new therapy targets. Various proteomic tools and multiple combinations were developed in this area. Out of the proteomic technology armentarium, mass spectrometry and array technologies are the most used approaches. The main characteristics of the proteomic technology used in this domain are high throughput and detection of minute concentration in small samples. We present herein, using various proteomic technologies, the identification in cancer cell lines and in tumour tissue EMT-related proteins, proteins that are involved in the activation of different cellular pathways. Proteomics has brought besides standard EMT markers (e.g., cell-cell adhesion proteins and transcription factors) other future potential markers for improving diagnosis, monitoring evolution, and developing new therapy targets. Future will increase the proteomic role in clinical investigation and validation of EMT-related biomarkers.
Collapse
|
46
|
A Unique Pattern of Mesothelial-Mesenchymal Transition Induced in the Normal Peritoneal Mesothelium by High-Grade Serous Ovarian Cancer. Cancers (Basel) 2019; 11:cancers11050662. [PMID: 31086083 PMCID: PMC6562987 DOI: 10.3390/cancers11050662] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2019] [Revised: 05/07/2019] [Accepted: 05/13/2019] [Indexed: 01/05/2023] Open
Abstract
The study was designed to establish whether high aggressiveness of high-grade serous ovarian cancer cells (HGSOCs), which display rapid growth, advanced stage at diagnosis and the highest mortality among all epithelial ovarian cancer histotypes, may be linked with a specific pattern of mesothelial-mesenchymal transition (MMT) elicited by these cells in normal peritoneal mesothelial cells (PMCs). Experiments were performed on primary PMCs, stable and primary ovarian cancer cells, tumors from patients with ovarian cancer, and laboratory animals. Results of in vitro and in vivo tests showed that MMT triggered by HGSOCs (primary cells and OVCAR-3 line) is far more pronounced than the process evoked by cells representing less aggressive ovarian cancer histotypes (A2780, SKOV-3). Mechanistically, HGSOCs induce MMT via Smad 2/3, ILK, TGF-β1, HGF, and IGF-1, whereas A2780 and SKOV-3 cells via exclusively Smad 2/3 and HGF. The conditioned medium from PMCs undergoing MMT promoted the progression of cancer cells and the effects exerted by the cells triggered to undergo MMT by the HGSOCs were significantly stronger than those related to the activity of their less aggressive counterparts. Our findings indicate that MMT in PMCs provoked by HGSOCs is stronger, proceeds via different mechanisms and has more procancerous characteristics than MMT provoked by less aggressive cancer histotypes, which may at least partly explain high aggressiveness of HGSOCs.
Collapse
|
47
|
Skarkova V, Kralova V, Vitovcova B, Rudolf E. Selected Aspects of Chemoresistance Mechanisms in Colorectal Carcinoma-A Focus on Epithelial-to-Mesenchymal Transition, Autophagy, and Apoptosis. Cells 2019; 8:cells8030234. [PMID: 30871055 PMCID: PMC6468859 DOI: 10.3390/cells8030234] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 03/08/2019] [Accepted: 03/08/2019] [Indexed: 12/15/2022] Open
Abstract
Chemoresistance has been found in all malignant tumors including colorectal carcinoma (CRC). Nowadays chemoresistance is understood as a major reason for therapy failure, with consequent tumor growth and spreading leading ultimately to the patient's premature death. The chemotherapy-related resistance of malignant colonocytes may be manifested in diverse mechanisms that may exist both prior to the onset of the therapy or after it. The ultimate function of this chemoresistance is to ensure the survival of malignant cells through continuing adaptation within an organism, therefore, the nature and spectrum of cell-survival strategies in CRC represent a highly significant target of scientific inquiry. Among these survival strategies employed by CRC cells, three unique but significantly linked phenomena stand out-epithelial-to-mesenchymal transition (EMT), autophagy, and cell death. In this mini-review, current knowledge concerning all three mechanisms including their emergence, timeline, regulation, and mutual relationships will be presented and discussed.
Collapse
Affiliation(s)
- Veronika Skarkova
- Department of Medical Biology and Genetics, Charles University, Faculty of Medicine in Hradec Králové, Zborovská 2089, 500 03 Hradec Králové, Czech Republic.
| | - Vera Kralova
- Department of Medical Biology and Genetics, Charles University, Faculty of Medicine in Hradec Králové, Zborovská 2089, 500 03 Hradec Králové, Czech Republic.
| | - Barbora Vitovcova
- Department of Medical Biology and Genetics, Charles University, Faculty of Medicine in Hradec Králové, Zborovská 2089, 500 03 Hradec Králové, Czech Republic.
| | - Emil Rudolf
- Department of Medical Biology and Genetics, Charles University, Faculty of Medicine in Hradec Králové, Zborovská 2089, 500 03 Hradec Králové, Czech Republic.
| |
Collapse
|
48
|
Zheng CC, Hu HF, Hong P, Zhang QH, Xu WW, He QY, Li B. Significance of integrin-linked kinase (ILK) in tumorigenesis and its potential implication as a biomarker and therapeutic target for human cancer. Am J Cancer Res 2019; 9:186-197. [PMID: 30755822 PMCID: PMC6356918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Accepted: 12/07/2018] [Indexed: 06/09/2023] Open
Abstract
Integrin-linked kinase (ILK), which is an ankyrin repeat-containing serine/threonine protein kinase, interacts with integrin β1 and the β3 cytoplasmic domain and phosphorylates integrin β1. ILK has multiple functions in cells, such as cell-extracellular matrix interactions, cell cycle, apoptosis, cell proliferation and cell motility, which are associated with the interacting partners of ILK and downstream signaling pathways. Upregulation of ILK is frequently observed in cancer tissues compared to corresponding normal tissues. Emerging evidence has demonstrated that ILK plays an important role in biological processes associated with tumorigenesis, including cancer cell proliferation, angiogenesis, metastasis, and drug resistance. Furthermore, inhibition of ILK expression and activity using siRNA or chemical inhibitors has shown a significant suppressive effect on cancer development and progression, implicating the potential of ILK as a target for cancer treatment. In this review, we summarized the functional role of ILK in tumorigenesis, with the expectation that targeting ILK could provide more evidence for cancer therapy.
Collapse
Affiliation(s)
- Can-Can Zheng
- Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan UniversityGuangzhou 510632, China
| | - Hui-Fang Hu
- Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan UniversityGuangzhou 510632, China
| | - Pan Hong
- Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan UniversityGuangzhou 510632, China
| | - Qi-Hua Zhang
- Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan UniversityGuangzhou 510632, China
| | - Wen Wen Xu
- Institute of Biomedicine, Guangdong Provincial Key Laboratory of Bioengineering Medicine, National Engineering Research Center of Genetic Medicine, Jinan UniversityGuangzhou 510632, China
| | - Qing-Yu He
- Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan UniversityGuangzhou 510632, China
| | - Bin Li
- Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan UniversityGuangzhou 510632, China
| |
Collapse
|
49
|
Basu S, Cheriyamundath S, Ben-Ze'ev A. Cell-cell adhesion: linking Wnt/β-catenin signaling with partial EMT and stemness traits in tumorigenesis. F1000Res 2018; 7. [PMID: 30271576 PMCID: PMC6144947 DOI: 10.12688/f1000research.15782.1] [Citation(s) in RCA: 143] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/13/2018] [Indexed: 12/18/2022] Open
Abstract
Changes in cell adhesion and motility are considered key elements in determining the development of invasive and metastatic tumors. Co-opting the epithelial-to-mesenchymal transition (EMT) process, which is known to occur during embryonic development, and the associated changes in cell adhesion properties in cancer cells are considered major routes for tumor progression. More recent
in vivo studies in tumor tissues and circulating tumor cell clusters suggest a stepwise EMT process rather than an “all-or-none” transition during tumor progression. In this commentary, we addressed the molecular mechanisms underlying the changes in cell adhesion and motility and adhesion-mediated signaling and their relationships to the partial EMT states and the acquisition of stemness traits by cancer cells.
Collapse
Affiliation(s)
- Sayon Basu
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Sanith Cheriyamundath
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Avri Ben-Ze'ev
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
50
|
Shen Y, Wang C, Ren Y, Ye J. A comprehensive look at the role of hyperlipidemia in promoting colorectal cancer liver metastasis. J Cancer 2018; 9:2981-2986. [PMID: 30123367 PMCID: PMC6096362 DOI: 10.7150/jca.25640] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 06/09/2018] [Indexed: 02/06/2023] Open
Abstract
Colorectal cancer (CRC) is one of the most malignant cancers, and it tends to migrate to the liver and has a high mortality rate. Several mechanisms behind the metastasis of CRC have been identified, including hyperlipidemia. For example, hyperlipidemia can lead to enhanced stemness and neutrophil infiltration, which increases CRC metastasis. There are three primary aspects to the relationship between hyperlipidemia and CRC metastasis: hyperlipidemia (1) promotes the initial metastatic properties of CRC, (2) stimulates CRC cells to leave the vasculature, and (3) facilitates the development of CRC metastasis. In this study, we provide a comprehensive overview of the role that hyperlipidemia played in CRC metastasis to help reduce the mortality associated with CRC metastasis from the standpoint of metabolic. We also review cancer metastasis.
Collapse
Affiliation(s)
- Yimin Shen
- 1 Department of Endocrinology and Metabolism, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Caihua Wang
- 2 Department of Gastroenterology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Yuezhong Ren
- 1 Department of Endocrinology and Metabolism, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Jun Ye
- 2 Department of Gastroenterology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| |
Collapse
|