1
|
Dalpati N, Rai SK, Sharma P, Sarangi PP. Integrins and integrin-driven secretory pathways as multi-dimensional regulators of tumor-associated macrophage recruitment and reprogramming in tumor microenvironment. Matrix Biol 2025; 135:55-69. [PMID: 39645091 DOI: 10.1016/j.matbio.2024.12.003] [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: 06/30/2024] [Revised: 11/27/2024] [Accepted: 12/04/2024] [Indexed: 12/09/2024]
Abstract
Integrins, a group of transmembrane receptors, play a crucial role in mediating the interactions between cells and extracellular matrix (ECM) proteins. The intracellular signaling initiated by these cell-matrix interactions in leukocytes mediates many essential cellular processes such as survival, migration, metabolism, and other immunological functions. Macrophages, as phagocytes, participate in both proinflammatory and anti-inflammatory processes, including progression. Numerous reports have shown that the integrin-regulated secretome, comprising cytokines, chemokines, growth factors, proteases, and other bioactive molecules, is a crucial modulator of macrophage functions in tumors, significantly influencing macrophage programming and reprogramming within the tumor microenvironment (TME) in addition to driving their step-by-step entry process into tumor tissue spaces. Importantly, studies have demonstrated a pivotal role for integrin receptor-mediated secretome and associated signaling pathways in functional reprogramming from anti-tumorigenic to pro-tumorigenic phenotype in tumor-associated macrophages (TAMs). In this comprehensive review, we have provided an in-depth analysis of the latest findings of various key pathways, mediators, and signaling cascades associated with integrin-driven polarization of macrophages in tumors. This manuscript will provide an updated understanding of the modulation of inflammatory monocytes/ macrophages and TAMs by integrin-driven secretory pathways in various functions such as migration, differentiation, and their role in tumor progression, angiogenesis, and metastasis.
Collapse
Affiliation(s)
- Nibedita Dalpati
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee 247667, Uttarakhand, India
| | - Shubham Kumar Rai
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee 247667, Uttarakhand, India
| | - Prerna Sharma
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee 247667, Uttarakhand, India
| | - Pranita P Sarangi
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee 247667, Uttarakhand, India.
| |
Collapse
|
2
|
Cheng YY, Worley BL, Javed Z, Elhaw AT, Tang PW, Al-Saad S, Kamlapurkar S, White S, Uboveja A, Mythreye K, Aird KM, Czyzyk TA, Hempel N. Loss of the predicted cell adhesion molecule MPZL3 promotes EMT and chemoresistance in ovarian cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.14.623672. [PMID: 39605523 PMCID: PMC11601277 DOI: 10.1101/2024.11.14.623672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Myelin protein zero-like 3 (MPZL3) is an Immunoglobulin-containing transmembrane protein with predicted cell adhesion molecule function. Loss of 11q23, where the MPZL3 gene resides, is frequently observed in cancer, and MPZL3 copy number alterations are frequently detected in tumor specimens. Yet the role and consequences of altered MPZL3 expression have not been explored in tumor development and progression. We addressed this in ovarian cancer, where both MPZL3 amplification and deletions are observed in respective subsets of high-grade serous specimens. While high and low MPZL3 expressing populations were similarly observed in primary ovarian tumors from an independent patient cohort, metastatic omental tumors largely displayed decreased MPZL3 expression, suggesting that MPZL3 loss is associated with metastatic progression. MPZL3 knock-down leads to strong upregulation of vimentin and an EMT gene signature that is associated with poor patient outcomes. Moreover, MPZL3 is necessary for homotypic cancer cell adhesion, and decreasing MPZL3 expression enhances invasion and clearance of mesothelial cell monolayers. In addition, MPZL3 loss abrogated cell cycle progression and proliferation. This was associated with increased resistance to Cisplatin and Olaparib and reduced DNA damage and apoptosis in response to these agents. Enhanced Cisplatin resistance was further validated in vivo . These data demonstrate for the first time that MPZL3 acts as an adhesion molecule and that MPZL3 loss results in EMT, decreased proliferation, and drug resistance in ovarian cancer. Our study suggests that decreased MPZL3 expression is a phenotype of ovarian cancer tumor progression and metastasis and may contribute to treatment failure in advanced-stage patients.
Collapse
|
3
|
Zhang Y, Wu D, Yu T, Liu Y, Zhao C, Xue R. Prognostic value of TMTC1 in pan-cancer analysis. Heliyon 2024; 10:e38308. [PMID: 39397950 PMCID: PMC11471174 DOI: 10.1016/j.heliyon.2024.e38308] [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] [Received: 11/24/2023] [Revised: 09/20/2024] [Accepted: 09/22/2024] [Indexed: 10/15/2024] Open
Abstract
Background Transmembrane and tetratricopeptide repeat containing 1 (TMTC1) is a recently discovered enzyme involved in the O-mannosylation of cadherins and protocadherins. It has been implicated in various types of cancer, but the overall prognostic significance of TMTC1 in pan-cancer and its potential as an immunotherapeutic target remain unclear. Methods We applied various bioinformatics methods to investigate the potential oncogenic roles of TMTC1 using public databases. This analysis involved examining the expression, prognosis, genetic alterations, immune infiltration, immunotherapy response, drug sensitivity, and regulatory mechanisms of the TMTC1 gene in diverse cancer types. Results In this study, we observed that TMTC1 expression is reduced in 19 types of cancer (ACC, BLCA, BRCA, CESC, COAD, ESCA, GBM, KICH, KIRC, KIRP, LAML, LUAD, LUSC, PRAD, READ, STAD, THCA, UCEC, and UCS) compared to normal tissues. Conversely, TMTC1 expression is elevated in OV and PAAD relative to normal tissues. Moreover, our analysis revealed that high expression of TMTC1 was associated with worse overall survival (OS) outcomes in patients with ACC, BLCA, COAD, GBM, KIRP, OV, STAD, and UCEC, but better OS outcomes in patients with CESC, KIRC, LUSC, and PAAD. Notably, patients with TMTC1 mutations or deep deletions demonstrated longer OS, while those with TMTC1 amplification showed shorter OS. There was a significant correlation between the expression level of TMTC1 and the infiltration of cancer-associated fibroblasts (CAFs) and endothelial cells. Using data from six real-world immunotherapy cohorts of BLCA, SKCM and RCC, we discovered that high TMTC1 expression was associated with better OS or progression-free survival (PFS). Lastly, through TMTC1-related gene enrichment analysis, some biological processes and pathways were found to be significantly enriched, such as vascular endothelial growth factor receptor signaling pathway and ECM-receptor interaction. Conclusions Our study demonstrates the prognostic significance of TMTC1 in pan-cancer and highlights its potential as an immunotherapeutic target.
Collapse
Affiliation(s)
- Ying Zhang
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Labouratory of Embryo Original Diseases, 200030, Shanghai, China
- Institute of Birth Defects and Rare Diseases, School of Medicine, Shanghai Jiao Tong University, 200030, Shanghai, China
| | - Dan Wu
- Department of Obstrics and Gynecology, The First People's Hospital of Jiande, Hangzhou, China
| | - Tiantian Yu
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Labouratory of Embryo Original Diseases, 200030, Shanghai, China
- Institute of Birth Defects and Rare Diseases, School of Medicine, Shanghai Jiao Tong University, 200030, Shanghai, China
| | - Yao Liu
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Labouratory of Embryo Original Diseases, 200030, Shanghai, China
- Institute of Birth Defects and Rare Diseases, School of Medicine, Shanghai Jiao Tong University, 200030, Shanghai, China
| | - Chunbo Zhao
- Department of Obstrics and Gynecology, The First People's Hospital of Jiande, Hangzhou, China
| | - Ruihong Xue
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Labouratory of Embryo Original Diseases, 200030, Shanghai, China
- Institute of Birth Defects and Rare Diseases, School of Medicine, Shanghai Jiao Tong University, 200030, Shanghai, China
| |
Collapse
|
4
|
Raj A, Chandran C S, Dua K, Kamath V, Alex AT. Targeting overexpressed surface proteins: A new strategy to manage the recalcitrant triple-negative breast cancer. Eur J Pharmacol 2024; 981:176914. [PMID: 39154820 DOI: 10.1016/j.ejphar.2024.176914] [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: 03/25/2024] [Revised: 08/08/2024] [Accepted: 08/15/2024] [Indexed: 08/20/2024]
Abstract
Triple-negative breast cancer (TNBC) is an aggressive and heterogeneous cancer that lacks all three molecular markers, Estrogen, Progesterone, and Human Epidermal Growth Factor Receptor 2 (HER2). This unique characteristic of TNBC makes it more resistant to hormonal therapy; hence, chemotherapy and surgery are preferred. Active targeting with nanoparticles is more effective in managing TNBC than a passive approach. The surface of TNBC cells overexpresses several cell-specific proteins, which can be explored for diagnostic and therapeutic purposes. Immunohistochemical analysis has revealed that TNBC cells overexpress αVβ3 integrin, Intercellular Adhesion Molecule 1 (ICAM-1), Glucose Transporter 5 (GLUT5), Transmembrane Glycoprotein Mucin 1 (MUC-1), and Epidermal Growth Factor Receptor (EGFR). These surface proteins can be targeted using ligands, such as aptamers, antibodies, and sugar molecules. Targeting the surface proteins of TNBC with ligands helps harmonize treatment and improve patient compliance. In this review, we discuss the proteins expressed, which are limited to αVβ3 integrin proteins, ICAM-1, GLUT-5, MUC1, and EGFR, on the surface of TNBC, the challenges associated with the preclinical setup of breast cancer for targeted nanoformulations, internalization techniques and their challenges, suggestions to overcome the limitations of successful translation of nanoparticles, and the possibility of ligand-conjugated nanoparticles targeting these surface receptors for a better therapeutic outcome.
Collapse
Affiliation(s)
- Alan Raj
- Department of Pharmaceutical Biotechnology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Udupi, Karnataka state, India, 576104.
| | - Sarath Chandran C
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Government Medical College Kannur, Pariyaram, Kerala, India, 670 503; Kerala University of Health Sciences, Thrissur, Kerala, India - 680 596.
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, Faculty of Health, University of Technology Sydney, Sydney, Australia-2007; Australian Research Centre in Complementary and Integrative Medicine, Faculty of Health, University of Technology Sydney, Sydney, Australia-2007.
| | - Venkatesh Kamath
- Department of Pharmaceutical Biotechnology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Udupi, Karnataka state, India, 576104.
| | - Angel Treasa Alex
- Department of Pharmaceutical Biotechnology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Udupi, Karnataka state, India, 576104.
| |
Collapse
|
5
|
Tian Z, Zhou D, Jiang R, Zhou B. Role of AMIGO2 in cancer progression: Novel insights (Review). Oncol Lett 2024; 28:434. [PMID: 39049987 PMCID: PMC11268087 DOI: 10.3892/ol.2024.14567] [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: 04/16/2024] [Accepted: 06/21/2024] [Indexed: 07/27/2024] Open
Abstract
Adhesion molecule with IgG-like domain 2 (AMIGO2) is a novel scaffold protein initially identified in cerebellar granule neurons, and inhibits apoptosis of neurons. It is also widely expressed in various malignant tumors, including gastric cancer, colorectal carcinoma, ovarian cancer, prostate cancer and melanoma. During the past decades, it has been revealed that AMIGO2 can act as an oncogene, participating in tumor occurrence and development, for example by inhibiting apoptosis, accelerating cell proliferation, migration and adhesion, and promoting tumor metastasis and drug resistance. The present review discusses the recent advancements regarding AMIGO2 in the field of cancer, emphasizing its related molecular mechanisms to identify novel therapeutic strategies targeting AMIGO2 for cancer treatment in the future.
Collapse
Affiliation(s)
- Zhen Tian
- Department of Oncology, Huishan Third People's Hospital of Wuxi, Wuxi, Jiangsu 214183, P.R. China
| | - Dongsheng Zhou
- Department of Oncology, Huishan Third People's Hospital of Wuxi, Wuxi, Jiangsu 214183, P.R. China
| | - Rui Jiang
- Department of Oncology, Huishan Third People's Hospital of Wuxi, Wuxi, Jiangsu 214183, P.R. China
| | - Bin Zhou
- Department of Oncology, Huishan Third People's Hospital of Wuxi, Wuxi, Jiangsu 214183, P.R. China
| |
Collapse
|
6
|
Pracharova J, Cyrikova T, Berecka M, Biersack B, Kasparkova J, Brabec V. Antimetastatic activity of (arene)ruthenium(II) complex of 4-aryl-4H-naphthopyran. Chem Biol Interact 2024; 400:111180. [PMID: 39089413 DOI: 10.1016/j.cbi.2024.111180] [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: 06/06/2024] [Revised: 07/21/2024] [Accepted: 07/30/2024] [Indexed: 08/04/2024]
Abstract
Metastatic cancer remains a formidable challenge in anticancer therapy. Despite efforts to develop effective antimetastasis drugs over the past half-century, currently approved treatments fall short of expectations. This report highlights the promising antiproliferative activity of a ruthenium-based therapeutic agent, namely dichlorido(p-cymene)[2-amino-4-(pyridin-3-yl)-4H-benzo[h]-chromene-3-carbonitrile]ruthenium(II) (complex 1) against metastatic cell lines. Complex 1 shows significant efficacy in metastatic LoVo and Du-145 cell lines at nanomolar concentrations, being markedly more active than clinically used anticancer cisplatin. Studies on the MDA-MB-231 cell line, which displays invasive characteristics, demonstrated that 1 significantly reduces cell invasion. This efficacy was confirmed by its impact on matrix metalloproteinase production in MDA-MB-231 cells. Given that cell migration drives cancer invasion and metastasis, complex 1's effect on MDA-MB-231 cell migration was evaluated via wound healing assay and vimentin network analysis. Results indicated a strong reduction in migration. A re-adhesion assay further demonstrated that 1 significantly lowers the re-adhesion ability of MDA-MB-231 cells compared to cisplatin. To better simulate the human body environment, a 3D spheroid invasion assay was used. This method showed that 1 effectively inhibits tumor spheroids from infiltrating the surrounding extracellular matrix. This study underscores the potential of (arene)ruthenium(II) complexes with naphthopyran ligands as potent antimetastatic agents for chemotherapy.
Collapse
Affiliation(s)
- Jitka Pracharova
- Department of Biophysics, Faculty of Science, Palacky University, CZ-77900, Olomouc, Czech Republic
| | - Tereza Cyrikova
- Department of Biophysics, Faculty of Science, Palacky University, CZ-77900, Olomouc, Czech Republic
| | - Michal Berecka
- Department of Biophysics, Faculty of Science, Palacky University, CZ-77900, Olomouc, Czech Republic
| | - Bernhard Biersack
- Organic Chemistry Laboratory, University Bayreuth, 95440, Bayreuth, Germany
| | - Jana Kasparkova
- Department of Biophysics, Faculty of Science, Palacky University, CZ-77900, Olomouc, Czech Republic; Czech Academy of Sciences, Institute of Biophysics, CZ-61200, Brno, Czech Republic
| | - Viktor Brabec
- Department of Biophysics, Faculty of Science, Palacky University, CZ-77900, Olomouc, Czech Republic; Czech Academy of Sciences, Institute of Biophysics, CZ-61200, Brno, Czech Republic.
| |
Collapse
|
7
|
Dietrich N, Castellanos-Martinez R, Kemmling J, Heuser A, Schnoor M, Schinner C, Spindler V. Adhesion of pancreatic tumor cell clusters by desmosomal molecules enhances early liver metastases formation. Sci Rep 2024; 14:18189. [PMID: 39107343 PMCID: PMC11303515 DOI: 10.1038/s41598-024-68493-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 07/24/2024] [Indexed: 08/10/2024] Open
Abstract
Desmosomes are intercellular adhesion complexes providing mechanical coupling and tissue integrity. Previously, a correlation of desmosomal molecule expression with invasion and metastasis formation in several tumor entities was described together with a relevance for circulating tumor cell cluster formation. Here, we investigated the contribution of the desmosomal core adhesion molecule desmoglein-2 (DSG2) to the initial steps of liver metastasis formation by pancreatic cancer cells using a novel ex vivo liver perfusion mouse model. We applied the pancreatic ductal adenocarcinoma cell line AsPC-1 with and without a knockout (KO) of DSG2 and generated mouse lines with a hepatocyte-specific KO of the known interacting partners of DSG2 (DSG2 and desmocollin-2). Liver perfusion with DSG2 KO AsPC-1 cells led to smaller circulating cell clusters and a reduced number of cells adhering to murine livers compared to control cells. While this was independent of the expression levels of desmosomal adhesion molecules in hepatocytes, we show that increased cluster size of cancer cells, which correlates with stronger cell-cell adhesion and expression of desmosomal molecules, is a major factor contributing to the early phase of metastatic spreading. In conclusion, impaired desmosomal adhesion results in reduced circulating cell cluster size, which is relevant for seeding and attachment of metastatic cells to the liver.
Collapse
Affiliation(s)
- Niclas Dietrich
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Ramon Castellanos-Martinez
- Department of Biomedicine, University of Basel, Basel, Switzerland
- Department of Molecular Biomedicine, CINVESTAV-IPN, Mexico, Mexico
| | - Julia Kemmling
- Institute of Anatomy and Experimental Morphology, University Medical Center Hamburg Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - Arnd Heuser
- Animal Phenotyping Platform, Max-Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Michael Schnoor
- Department of Molecular Biomedicine, CINVESTAV-IPN, Mexico, Mexico
| | - Camilla Schinner
- Department of Biomedicine, University of Basel, Basel, Switzerland.
- Institute of Anatomy and Experimental Morphology, University Medical Center Hamburg Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany.
- Hannover Medical School, Institute of Applied and Functional Anatomy, Carl-Neuberg-Straße 1, 30625, Hannover, Germany.
| | - Volker Spindler
- Department of Biomedicine, University of Basel, Basel, Switzerland.
- Institute of Anatomy and Experimental Morphology, University Medical Center Hamburg Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany.
| |
Collapse
|
8
|
He M, Zhou X, Wang X. Glycosylation: mechanisms, biological functions and clinical implications. Signal Transduct Target Ther 2024; 9:194. [PMID: 39098853 PMCID: PMC11298558 DOI: 10.1038/s41392-024-01886-1] [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: 10/21/2023] [Revised: 05/25/2024] [Accepted: 06/07/2024] [Indexed: 08/06/2024] Open
Abstract
Protein post-translational modification (PTM) is a covalent process that occurs in proteins during or after translation through the addition or removal of one or more functional groups, and has a profound effect on protein function. Glycosylation is one of the most common PTMs, in which polysaccharides are transferred to specific amino acid residues in proteins by glycosyltransferases. A growing body of evidence suggests that glycosylation is essential for the unfolding of various functional activities in organisms, such as playing a key role in the regulation of protein function, cell adhesion and immune escape. Aberrant glycosylation is also closely associated with the development of various diseases. Abnormal glycosylation patterns are closely linked to the emergence of various health conditions, including cancer, inflammation, autoimmune disorders, and several other diseases. However, the underlying composition and structure of the glycosylated residues have not been determined. It is imperative to fully understand the internal structure and differential expression of glycosylation, and to incorporate advanced detection technologies to keep the knowledge advancing. Investigations on the clinical applications of glycosylation focused on sensitive and promising biomarkers, development of more effective small molecule targeted drugs and emerging vaccines. These studies provide a new area for novel therapeutic strategies based on glycosylation.
Collapse
Affiliation(s)
- Mengyuan He
- Department of Hematology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China
| | - Xiangxiang Zhou
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China.
- National Clinical Research Center for Hematologic Diseases, the First Affiliated Hospital of Soochow University, Suzhou, 251006, China.
| | - Xin Wang
- Department of Hematology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China.
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China.
- National Clinical Research Center for Hematologic Diseases, the First Affiliated Hospital of Soochow University, Suzhou, 251006, China.
- Taishan Scholars Program of Shandong Province, Jinan, Shandong, 250021, China.
- Branch of National Clinical Research Center for Hematologic Diseases, Jinan, Shandong, 250021, China.
| |
Collapse
|
9
|
Wiseglass G, Rubinstein R. Following the Evolutionary Paths of Dscam1 Proteins toward Highly Specific Homophilic Interactions. Mol Biol Evol 2024; 41:msae141. [PMID: 38989909 PMCID: PMC11272049 DOI: 10.1093/molbev/msae141] [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: 03/20/2024] [Revised: 06/05/2024] [Accepted: 07/05/2024] [Indexed: 07/12/2024] Open
Abstract
Many adhesion proteins, evolutionarily related through gene duplication, exhibit distinct and precise interaction preferences and affinities crucial for cell patterning. Yet, the evolutionary paths by which these proteins acquire new specificities and prevent cross-interactions within their family members remain unknown. To bridge this gap, this study focuses on Drosophila Down syndrome cell adhesion molecule-1 (Dscam1) proteins, which are cell adhesion proteins that have undergone extensive gene duplication. Dscam1 evolved under strong selective pressure to achieve strict homophilic recognition, essential for neuronal self-avoidance and patterning. Through a combination of phylogenetic analyses, ancestral sequence reconstruction, and cell aggregation assays, we studied the evolutionary trajectory of Dscam1 exon 4 across various insect lineages. We demonstrated that recent Dscam1 duplications in the mosquito lineage bind with strict homophilic specificities without any cross-interactions. We found that ancestral and intermediate Dscam1 isoforms maintained their homophilic binding capabilities, with some intermediate isoforms also engaging in promiscuous interactions with other paralogs. Our results highlight the robust selective pressure for homophilic specificity integral to the Dscam1 function within the process of neuronal self-avoidance. Importantly, our study suggests that the path to achieving such selective specificity does not introduce disruptive mutations that prevent self-binding but includes evolutionary intermediates that demonstrate promiscuous heterophilic interactions. Overall, these results offer insights into evolutionary strategies that underlie adhesion protein interaction specificities.
Collapse
Affiliation(s)
- Gil Wiseglass
- School of Neurobiology, Biochemistry and Biophysics, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Rotem Rubinstein
- School of Neurobiology, Biochemistry and Biophysics, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
10
|
Yang W, Wu Z, Cai S, Li Z, Wang W, Wu J, Luo H, Ye X. Tumor lymphangiogenesis index reveals the immune landscape and immunotherapy response in lung adenocarcinoma. Front Immunol 2024; 15:1354339. [PMID: 38638428 PMCID: PMC11024352 DOI: 10.3389/fimmu.2024.1354339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 03/25/2024] [Indexed: 04/20/2024] Open
Abstract
Background Lymphangiogenesis (LYM) has an important role in tumor progression and is strongly associated with tumor metastasis. However, the clinical application of LYM has not progressed as expected. The potential value of LYM needs to be further developed in lung adenocarcinoma (LUAD) patients. Methods The Sequencing data and clinical characteristics of LUAD patients were downloaded from The Cancer Genome Atlas and GEO databases. Multiple machine learning algorithms were used to screen feature genes and develop the LYM index. Immune cell infiltration, immune checkpoint expression, Tumor Immune Dysfunction and Exclusion (TIDE) algorithm and drug sensitivity analysis were used to explore the correlation of LYM index with immune profile and anti-tumor therapy. Results We screened four lymphangiogenic feature genes (PECAM1, TIMP1, CXCL5 and PDGFB) to construct LYM index based on multiple machine learning algorithms. We divided LUAD patients into the high LYM index group and the low LYM index group based on the median LYM index. LYM index is a risk factor for the prognosis of LUAD patients. In addition, there was a significant difference in immune profile between high LYM index and low LYM index groups. LUAD patients in the low LYM index group seemed to benefit more from immunotherapy based on the results of TIDE algorithm. Conclusion Overall, we confirmed that the LYM index is a prognostic risk factor and a valuable predictor of immunotherapy response in LUAD patients, which provides new evidence for the potential application of LYM.
Collapse
Affiliation(s)
- Weichang Yang
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
- Jiangxi Key Laboratory of Molecular Medicine, Nanchang, Jiangxi, China
| | - Zhijian Wu
- Department of Cardiology, The Second Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Shanshan Cai
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Zhouhua Li
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Wenjun Wang
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Juan Wu
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Hongdan Luo
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Xiaoqun Ye
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
11
|
Massey A, Stewart J, Smith C, Parvini C, McCormick M, Do K, Cartagena-Rivera AX. Mechanical properties of human tumour tissues and their implications for cancer development. NATURE REVIEWS. PHYSICS 2024; 6:269-282. [PMID: 38706694 PMCID: PMC11066734 DOI: 10.1038/s42254-024-00707-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 02/13/2024] [Indexed: 05/07/2024]
Abstract
The mechanical properties of cells and tissues help determine their architecture, composition and function. Alterations to these properties are associated with many diseases, including cancer. Tensional, compressive, adhesive, elastic and viscous properties of individual cells and multicellular tissues are mostly regulated by reorganization of the actomyosin and microtubule cytoskeletons and extracellular glycocalyx, which in turn drive many pathophysiological processes, including cancer progression. This Review provides an in-depth collection of quantitative data on diverse mechanical properties of living human cancer cells and tissues. Additionally, the implications of mechanical property changes for cancer development are discussed. An increased knowledge of the mechanical properties of the tumour microenvironment, as collected using biomechanical approaches capable of multi-timescale and multiparametric analyses, will provide a better understanding of the complex mechanical determinants of cancer organization and progression. This information can lead to a further understanding of resistance mechanisms to chemotherapies and immunotherapies and the metastatic cascade.
Collapse
Affiliation(s)
- Andrew Massey
- Section on Mechanobiology, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
| | - Jamie Stewart
- Section on Mechanobiology, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
- These authors contributed equally: Jamie Stewart, Chynna Smith
| | - Chynna Smith
- Section on Mechanobiology, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
- These authors contributed equally: Jamie Stewart, Chynna Smith
| | - Cameron Parvini
- Section on Mechanobiology, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
| | - Moira McCormick
- Section on Mechanobiology, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
| | - Kun Do
- Section on Mechanobiology, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
| | - Alexander X. Cartagena-Rivera
- Section on Mechanobiology, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
12
|
Khaodee W, Xiyuan G, Han MTT, Tayapiwatana C, Chiampanichayakul S, Anuchapreeda S, Cressey R. Transcriptomic analysis of glucosidase II beta subunit (GluIIß) knockout A549 cells reveals its roles in regulation of cell adhesion molecules (CAMs) and anti-tumor immunity. BMC Genomics 2024; 25:82. [PMID: 38245670 PMCID: PMC10799456 DOI: 10.1186/s12864-023-09888-z] [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: 06/01/2023] [Accepted: 12/09/2023] [Indexed: 01/22/2024] Open
Abstract
Glucosidase II beta subunit (GluIIß), encoded from PRKCSH, is a subunit of the glucosidase II enzyme responsible for quality control of N-linked glycoprotein folding and suppression of GluIIß led to inhibitory effect of the receptor tyrosine kinase (RTKs) activities known to be critical for survival and development of cancer. In this study, we investigated the effect of GluIIß knockout on the global gene expression of cancer cells and its impact on functions of immune cells. GluIIß knockout lung adenocarcinoma A549 cell line was generated using CRISPR/Cas9-based genome editing system and subjected to transcriptomic analysis. Among 23,502 expressed transcripts, 1068 genes were significantly up-regulated and 807 genes greatly down-regulated. The KEGG enrichment analysis showed significant down-regulation of genes related extracellular matrix (ECM), ECM-receptor interaction, cytokine-cytokine receptor interaction and cell adhesion molecules (CAMs) in GluIIß knockout cells. Of 9 CAMs encoded DEG identified by KEGG enrichment analysis, real time RT-PCR confirmed 8 genes to be significantly down-regulated in all 3 different GluIIß knockout clones, which includes cadherin 4 (CDH4), cadherin 2 (CDH2), versican (VCAN), integrin subunit alpha 4 (ITGA4), endothelial cell-selective adhesion molecule (ESAM), CD274 (program death ligand-1 (PD-L1)), Cell Adhesion Molecule 1 (CADM1), and Nectin Cell Adhesion Molecule 3 (NECTIN3). Whereas PTPRF (Protein Tyrosine Phosphatase Receptor Type F) was significantly decreased only in 1 out of 3 knockout clones. Microscopic analysis revealed distinctively different cell morphology of GluIIβ knockout cells with lesser cytoplasmic and cell surface area compared to parental A549 cells and non-targeted transfected cells.Further investigations revealed that Jurkat E6.1 T cells or human peripheral blood mononuclear cells (PBMCs) co-cultured with GluIIß knockout A549 exhibited significantly increased viability and tumor cell killing activity compared to those co-cultured with non-target transfected cells. Analysis of cytokine released from Jurkat E6.1 T cells co-cultured with GluIIß knockout A549 cells showed significant increased level of angiogenin and significant decreased level of ENA-78. In conclusion, knockout of GluIIß from cancer cells induced altered gene expression profile that improved anti-tumor activities of co-cultured T lymphocytes and PBMCs thus suppression of GluIIß may represent a novel approach of boosting anti-tumor immunity.
Collapse
Affiliation(s)
- Worapong Khaodee
- Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Guo Xiyuan
- Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
- Public Experimental Technology Center School of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, China
| | - Moe Thi Thi Han
- Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Chatchai Tayapiwatana
- Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Sawitree Chiampanichayakul
- Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
- Cancer Research Unit, Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Songyot Anuchapreeda
- Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
- Cancer Research Unit, Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Ratchada Cressey
- Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand.
- Cancer Research Unit, Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand.
| |
Collapse
|
13
|
Mandal T, Biswas A, Ghosh T, Manikandan S, Kundu A, Banerjee A, Mitra D, Sinha B. Mechano-regulation by clathrin pit-formation and passive cholesterol-dependent tubules during de-adhesion. Cell Mol Life Sci 2024; 81:43. [PMID: 38217571 PMCID: PMC10787898 DOI: 10.1007/s00018-023-05072-4] [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: 07/08/2023] [Revised: 11/10/2023] [Accepted: 11/30/2023] [Indexed: 01/15/2024]
Abstract
Adherent cells ensure membrane homeostasis during de-adhesion by various mechanisms, including endocytosis. Although mechano-chemical feedbacks involved in this process have been studied, the step-by-step build-up and resolution of the mechanical changes by endocytosis are poorly understood. To investigate this, we studied the de-adhesion of HeLa cells using a combination of interference reflection microscopy, optical trapping and fluorescence experiments. We found that de-adhesion enhanced membrane height fluctuations of the basal membrane in the presence of an intact cortex. A reduction in the tether force was also noted at the apical side. However, membrane fluctuations reveal phases of an initial drop in effective tension followed by saturation. The area fractions of early (Rab5-labelled) and recycling (Rab4-labelled) endosomes, as well as transferrin-labelled pits close to the basal plasma membrane, also transiently increased. On blocking dynamin-dependent scission of endocytic pits, the regulation of fluctuations was not blocked, but knocking down AP2-dependent pit formation stopped the tension recovery. Interestingly, the regulation could not be suppressed by ATP or cholesterol depletion individually but was arrested by depleting both. The data strongly supports Clathrin and AP2-dependent pit-formation to be central to the reduction in fluctuations confirmed by super-resolution microscopy. Furthermore, we propose that cholesterol-dependent pits spontaneously regulate tension under ATP-depleted conditions.
Collapse
Affiliation(s)
- Tithi Mandal
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Nadia, Mohanpur, 741246, India
| | - Arikta Biswas
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Nadia, Mohanpur, 741246, India
- Mechanobiology Institute, National University of Singapore, 5A Engineering Drive 1, Singapore, 117411, Singapore
| | - Tanmoy Ghosh
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Nadia, Mohanpur, 741246, India
| | - Sreekanth Manikandan
- NORDITA, KTH Royal Institute of Technology and Stockholm University, Roslagstullsbacken 23, 10691, Stockholm, Sweden
| | - Avijit Kundu
- Department of Physical Sciences, Indian Institute of Science Education and Research Kolkata, Nadia, Mohanpur, 741246, India
- Experimental Physics I, Universität Bayreuth, Universitätsstraße 30, 95447, Bayreuth, Germany
| | - Ayan Banerjee
- Department of Physical Sciences, Indian Institute of Science Education and Research Kolkata, Nadia, Mohanpur, 741246, India
| | - Dhrubaditya Mitra
- NORDITA, KTH Royal Institute of Technology and Stockholm University, Roslagstullsbacken 23, 10691, Stockholm, Sweden
| | - Bidisha Sinha
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Nadia, Mohanpur, 741246, India.
| |
Collapse
|
14
|
Ersuz R, Karapınar OS, Doğan S. Comparison of serum levels of cell adhesion molecules (E-selectin, P-selectin, ICAM-1, VCAM-1, LRG-1) in placental invasion and adhesion anomalies with patients with vaginal delivery and former cesarean. Arch Gynecol Obstet 2024; 309:167-173. [PMID: 36625879 DOI: 10.1007/s00404-023-06911-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 01/02/2023] [Indexed: 01/11/2023]
Abstract
OBJECTIVE It is aimed to be a technique that can be used for diagnosis and to prevent maternal deaths in cases where the serum levels of cell adhesion molecules are different in patients with abnormal placentation compared to healthy pregnant women. MATERIALS AND METHODS Patients between March 2020 and September 2021 were included in the study. While 56 patients, out of 153 cases formed the placental adhesion and/or localization anomaly group, 55 cases without placental adhesion anomaly (placental invasion anomaly and/or previa pathology) constituted the cesarean section group and 42 cases constituted the vaginal birth control group. Demographic characteristics and histories of 153 patients were questioned. I-CAM-1, V-CAM-1, E-Selectin, P-Selectin, LRG-1 levels were studied. The parameters measured by the ELISA method were studied in the Thermo Fisher Scientific Multiscan Go (Finland) device at the Hatay Mustafa Kemal University Medical Faculty Medical Biochemistry USA ELISA Laboratory. Wholehouse and One Way Anova analysis methods were used to compare the results. RESULTS There were significant differences in E-Selectin, P-Selectin, ICAM-1 and LRG-1 values between the groups (p < 0.05). There was a significant difference between the vaginal birth (VB) and previa/percreata (PP) groups in terms of E-Selectin (p = 0.038). In terms of P-Selectin, there was a significant difference between the C/S and previa/percreata (PP) groups (p < 001). P-Selectin was higher in the previa/percreata (PP) group. There was a significant difference between the Vaginally birth (VB), C/S group (p = 0.041) and the vaginal birth (VB), previa/percreata (PP) group (p = 0.013) in terms of ICAM-1, but there was no significant difference between the C/S and previa/percreata (PP) groups. In terms of LRG-1, there was a significant difference between all 3 groups (p < 0.05). DISCUSSION A recent study investigated the potential modulatory effects of trans-resveratrol (RSV), arginase and endothelial dysfunction biomarkers in patients with PE. Another reflection of endothelial dysfunction in PE is increased endothelial activation biomarkers such as intercellular adhesion molecule-1 (ICAM-1), von Willebrand factor (vWF), and Caspase-3 (CASP-3). The study, regarding vWF expression, the preeclampsia (PE) group showed higher levels compared to endothelial cells incubated with healty pregnant (HP) plasma [Bueno-Pereira et al 2022 Antioxidants 2111]. From this and similar studies, the hypothesis that the role of cell adhesion molecules in endothelial damage may be the underlying cause of invasion and location anomalies emerges. This hypothesis is the starting point of our study. CONCLUSIONS In our study, all adhesion molecules except V-CAM-1 were found to be significantly higher in the previa/percreata (PP) group. E-Selectin and LRG-1 adhesion molecules were found to be significantly higher even in C/S patients compared to normal delivery. As a result; these adhesion molecules can be studied as a marker in previa/percreata (PP) patients.
Collapse
Affiliation(s)
- Rutkay Ersuz
- Hatay Mustafa Kemal Universitesi, Antakya, Turkey.
| | | | - Serdar Doğan
- Hatay Mustafa Kemal Universitesi, Antakya, Turkey
| |
Collapse
|
15
|
Soares CLR, Wilairatana P, Silva LR, Moreira PS, Vilar Barbosa NMM, da Silva PR, Coutinho HDM, de Menezes IRA, Felipe CFB. Biochemical aspects of the inflammatory process: A narrative review. Biomed Pharmacother 2023; 168:115764. [PMID: 37897973 DOI: 10.1016/j.biopha.2023.115764] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 10/11/2023] [Accepted: 10/17/2023] [Indexed: 10/30/2023] Open
Abstract
Inflammation is a protective response of the body potentially caused by microbial, viral, or fungal infections, tissue damage, or even autoimmune reactions. The cardinal signs of inflammation are consequences of immunological, biochemical, and physiological changes that trigger the release of pro-inflammatory chemical mediators at the local of the injured site thus, increasing blood flow, vascular permeability, and leukocyte recruitment. The aim of this study is to give an overview of the inflammatory process, focusing on chemical mediators. The literature review was based on a search of journals published between the years 2009 and 2023, regarding the role of major chemical mediators in the inflammatory process and current studies in pathogenesis, diagnosis, and therapy. Some of the recent contributions in the study of inflammatory pathologies and their mediators, including cytokines and chemokines, the kinin system, free radicals, nitric oxide, histamine, cell adhesion molecules, leukotrienes, prostaglandins and the complement system and their role in human health and chronic diseases.
Collapse
Affiliation(s)
- Caroline Leal Rodrigues Soares
- Departamento de Biologia Molecular - DBM. Universidade Federal da Paraíba - UFPB, Campus I - Jardim Cidade Universitária, CEP 58059-900 João Pessoa, Brazil
| | - Polrat Wilairatana
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand.
| | - Larissa Rodrigues Silva
- Departamento de Biologia Molecular - DBM. Universidade Federal da Paraíba - UFPB, Campus I - Jardim Cidade Universitária, CEP 58059-900 João Pessoa, Brazil
| | - Polyanna Silva Moreira
- Departamento de Biologia Molecular - DBM. Universidade Federal da Paraíba - UFPB, Campus I - Jardim Cidade Universitária, CEP 58059-900 João Pessoa, Brazil
| | - Nayana Maria Medeiros Vilar Barbosa
- Departamento de Biologia Molecular - DBM. Universidade Federal da Paraíba - UFPB, Campus I - Jardim Cidade Universitária, CEP 58059-900 João Pessoa, Brazil
| | - Pablo Rayff da Silva
- Departamento de Biologia Molecular - DBM. Universidade Federal da Paraíba - UFPB, Campus I - Jardim Cidade Universitária, CEP 58059-900 João Pessoa, Brazil
| | - Henrique Douglas Melo Coutinho
- Laboratório de Microbiologia e Biologia Molecular - LMBM. Universidade Regional do Cariri - URCA, Rua Cel Antônio Luiz, 1161, Oimenta, CEP 63105-000 Crato, Brazil.
| | - Irwin Rose Alencar de Menezes
- Laboratório de Farmacologia e Química Molecular - LFQM. Universidade Regional do Cariri - URCA, Rua Cel Antônio Luiz, 1161, Pimenta, CEP 63105-000 Crato, Brazil
| | - Cícero Francisco Bezerra Felipe
- Departamento de Biologia Molecular - DBM. Universidade Federal da Paraíba - UFPB, Campus I - Jardim Cidade Universitária, CEP 58059-900 João Pessoa, Brazil.
| |
Collapse
|
16
|
Chen SN, Mai ZY, Mai JN, Liang W, Dong ZX, Ju FE, Chan SH, Fang Z, Xu Y, Uziel O, He C, Zhang XD, Zheng Y. E2F2 modulates cell adhesion through the transcriptional regulation of PECAM1 in multiple myeloma. Br J Haematol 2023; 202:840-855. [PMID: 37365680 DOI: 10.1111/bjh.18958] [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: 12/08/2022] [Revised: 05/26/2023] [Accepted: 06/17/2023] [Indexed: 06/28/2023]
Abstract
Multiple myeloma (MM) is the second most common haematological malignancy. Despite the development of new drugs and treatments in recent years, the therapeutic outcomes of patients are not satisfactory. It is necessary to further investigate the molecular mechanism underlying MM progression. Herein, we found that high E2F2 expression was correlated with poor overall survival and advanced clinical stages in MM patients. Gain- and loss-of-function studies showed that E2F2 inhibited cell adhesion and consequently activated cell epithelial-to-mesenchymal transition (EMT) and migration. Further experiments revealed that E2F2 interacted with the PECAM1 promoter to suppress its transcriptional activity. The E2F2-knockdown-mediated promotion of cell adhesion was significantly reversed by the repression of PECAM1 expression. Finally, we observed that silencing E2F2 significantly inhibited viability and tumour progression in MM cell models and xenograft mouse models respectively. This study demonstrates that E2F2 plays a vital role as a tumour accelerator by inhibiting PECAM1-dependent cell adhesion and accelerating MM cell proliferation. Therefore, E2F2 may serve as a potential independent prognostic marker and therapeutic target for MM.
Collapse
Affiliation(s)
- Shu-Na Chen
- Department of Hematology, Institute of Hematology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
- Key Laboratory for Efficacy and Safety Evaluation of Hematological Malignancy Targeted Medicine of Guangdong Provincial Drug Administration, School of Medicine, Sun Yat-Sen University, Shenzhen, China
| | - Zhi-Ying Mai
- Department of Hematology, Institute of Hematology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
- Key Laboratory for Efficacy and Safety Evaluation of Hematological Malignancy Targeted Medicine of Guangdong Provincial Drug Administration, School of Medicine, Sun Yat-Sen University, Shenzhen, China
| | - Jun-Na Mai
- Department of Hematology, Institute of Hematology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Weiyao Liang
- Key Laboratory for Efficacy and Safety Evaluation of Hematological Malignancy Targeted Medicine of Guangdong Provincial Drug Administration, School of Medicine, Sun Yat-Sen University, Shenzhen, China
| | - Zhao-Xia Dong
- Key Laboratory for Efficacy and Safety Evaluation of Hematological Malignancy Targeted Medicine of Guangdong Provincial Drug Administration, School of Medicine, Sun Yat-Sen University, Shenzhen, China
| | - Fei-Er Ju
- Key Laboratory for Efficacy and Safety Evaluation of Hematological Malignancy Targeted Medicine of Guangdong Provincial Drug Administration, School of Medicine, Sun Yat-Sen University, Shenzhen, China
| | - Sze-Hoi Chan
- Key Laboratory for Efficacy and Safety Evaluation of Hematological Malignancy Targeted Medicine of Guangdong Provincial Drug Administration, School of Medicine, Sun Yat-Sen University, Shenzhen, China
| | - Zhigang Fang
- Department of Hematology, Institute of Hematology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Yichuan Xu
- Department of Hematology, Institute of Hematology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Orit Uziel
- The Felsenstein Medical Research Center, Institute of Hematology Rabin Medical Center and Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Chengwei He
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau SAR, China
| | - Xing-Ding Zhang
- Key Laboratory for Efficacy and Safety Evaluation of Hematological Malignancy Targeted Medicine of Guangdong Provincial Drug Administration, School of Medicine, Sun Yat-Sen University, Shenzhen, China
| | - Yongjiang Zheng
- Department of Hematology, Institute of Hematology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
17
|
Kostrhunova H, McGhie BS, Markova L, Novakova O, Kasparkova J, Aldrich-Wright JR, Brabec V. Platinum(IV) Derivatives of [Pt(1 S,2 S-diaminocyclohexane)(5,6-dimethyl-1,10-phenanthroline)] with Diclofenac Ligands in the Axial Positions: A New Class of Potent Multi-action Agents Exhibiting Selectivity to Cancer Cells. J Med Chem 2023. [PMID: 37285472 DOI: 10.1021/acs.jmedchem.3c00269] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
The platinum(II) complex [Pt(1S,2S-diaminocyclohexane)(5,6-dimethyl-1,10-phenanthroline)]2+ (PtII56MeSS, 1) exhibits high potency across numerous cancer cell lines acting by a multimodal mechanism. However, 1 also displays side toxicity and in vivo activity; all details of its mechanism of action are not entirely clear. Here, we describe the synthesis and biological properties of new platinum(IV) prodrugs that combine 1 with one or two axially coordinated molecules of diclofenac (DCF), a non-steroidal anti-inflammatory cancer-selective drug. The results suggest that these Pt(IV) complexes exhibit mechanisms of action typical for Pt(II) complex 1 and DCF, simultaneously. The presence of DCF ligand(s) in the Pt(IV) complexes promotes the antiproliferative activity and selectivity of 1 by inhibiting lactate transporters, resulting in blockage of the glycolytic process and impairment of mitochondrial potential. Additionally, the investigated Pt(IV) complexes selectively induce cell death in cancer cells, and the Pt(IV) complexes containing DCF ligands induce hallmarks of immunogenic cell death in cancer cells.
Collapse
Affiliation(s)
- Hana Kostrhunova
- Institute of Biophysics, Czech Academy of Sciences, Kralovopolska 135, CZ-61200 Brno, Czech Republic
| | - Brondwyn S McGhie
- School of Science, Western Sydney University, Penrith South DC 1797, New South Wales, Australia
| | - Lenka Markova
- Institute of Biophysics, Czech Academy of Sciences, Kralovopolska 135, CZ-61200 Brno, Czech Republic
| | - Olga Novakova
- Institute of Biophysics, Czech Academy of Sciences, Kralovopolska 135, CZ-61200 Brno, Czech Republic
| | - Jana Kasparkova
- Institute of Biophysics, Czech Academy of Sciences, Kralovopolska 135, CZ-61200 Brno, Czech Republic
- Department of Biophysics, Faculty of Science, Palacky University, Slechtitelu 27, 783 71 Olomouc, Czech Republic
| | - Janice R Aldrich-Wright
- School of Science, Western Sydney University, Penrith South DC 1797, New South Wales, Australia
| | - Viktor Brabec
- Institute of Biophysics, Czech Academy of Sciences, Kralovopolska 135, CZ-61200 Brno, Czech Republic
| |
Collapse
|
18
|
Sato Y, Elbadawy M, Suzuki K, Tsunedomi R, Nagano H, Ishihara Y, Yamamoto H, Azakami D, Uchide T, Nabeta R, Fukushima R, Abugomaa A, Kaneda M, Yamawaki H, Shinohara Y, Usui T, Sasaki K. Establishment of an experimental model of canine malignant mesothelioma organoid culture using a three-dimensional culture method. Biomed Pharmacother 2023; 162:114651. [PMID: 37030135 DOI: 10.1016/j.biopha.2023.114651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 03/21/2023] [Accepted: 03/31/2023] [Indexed: 04/09/2023] Open
Abstract
Canine malignant mesothelioma (cMM) is a rare and drug-resistant malignant tumor. Due to few patients and experimental models, there have not been enough studies to demonstrate the pathogenesis of the disease and novel effective treatment for cMM. Since cMM resembles human MM (hMM) in histopathological characteristics, it is also considered a promising research model of hMM. Compared with conventional 2-dimensional (2D) culture methods, 3-dimensional (3D) organoid culture can recapitulate the properties of original tumor tissues. However, cMM organoids have never been developed. In the present study, we for the first time generated cMM organoids using the pleural effusion samples. Organoids from individual MM dogs were successfully generated. They exhibited the characteristics of MM and expressed mesothelial cell markers, such as WT-1 and mesothelin. The sensitivity to anti-cancer drugs was different in each strain of cMM organoids. RNA sequencing analysis showed cell adhesion molecule pathways were specifically upregulated in cMM organoids compared with their corresponding 2D cultured cells. Among these genes, the expression level of E-cadherin was drastically higher in the organoids than that in the 2D cells. In conclusion, our established cMM organoids might become a new experimental tool to provide new insights into canine and human MM therapy.
Collapse
Affiliation(s)
- Yomogi Sato
- Laboratory of Veterinary Pharmacology, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu, Tokyo 183-8509, Japan
| | - Mohamed Elbadawy
- Laboratory of Veterinary Pharmacology, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu, Tokyo 183-8509, Japan; Department of Pharmacology, Faculty of Veterinary Medicine, Benha University, 13736, Moshtohor, Toukh, Elqaliobiya, Egypt.
| | - Kazuhiko Suzuki
- Laboratory of Veterinary Toxicology, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu, Tokyo 183-8509, Japan
| | - Ryouichi Tsunedomi
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi 755-8505, Japan
| | - Hiroaki Nagano
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi 755-8505, Japan
| | - Yusuke Ishihara
- Laboratory of Veterinary Pharmacology, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu, Tokyo 183-8509, Japan
| | - Haru Yamamoto
- Laboratory of Veterinary Pharmacology, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu, Tokyo 183-8509, Japan
| | - Daigo Azakami
- Laboratory of Veterinary Clinical Oncology, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu, Tokyo 183-8509, Japan
| | - Tsuyoshi Uchide
- Laboratory of Veterinary Molecular Pathology and Therapeutics, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu, Tokyo 183-8509, Japan
| | - Rina Nabeta
- Laboratory of Veterinary Molecular Pathology and Therapeutics, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu, Tokyo 183-8509, Japan
| | - Ryuji Fukushima
- Animal Medical Emergency Center, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 2-24-16 Nakamachi, Koganei, Tokyo 184-8588, Japan
| | - Amira Abugomaa
- Laboratory of Veterinary Pharmacology, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu, Tokyo 183-8509, Japan; Faculty of Veterinary Medicine, Mansoura University, 35516 Mansoura, Egypt
| | - Masahiro Kaneda
- Laboratory of Veterinary Anatomy, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu, Tokyo 183-8509, Japan
| | - Hideyuki Yamawaki
- Laboratory of Veterinary Pharmacology, School of Veterinary Medicine, Kitasato University, 35-1, Higashi 23 ban-cho, Towada, Aomori 034-8628, Japan
| | - Yuta Shinohara
- Pet Health & Food Division, Iskara Industry CO., LTD, 1-14-2, Nihonbashi, Chuo-ku, Tokyo, 103-0027, Japan
| | - Tatsuya Usui
- Laboratory of Veterinary Pharmacology, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu, Tokyo 183-8509, Japan.
| | - Kazuaki Sasaki
- Laboratory of Veterinary Pharmacology, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu, Tokyo 183-8509, Japan
| |
Collapse
|
19
|
Wang H, Xu X, Wang Y, Xue X, Guo W, Guo S, Qiu S, Cui J, Qiao Y. NMT1 sustains ICAM-1 to modulate adhesion and migration of tumor cells. Cell Signal 2023:110739. [PMID: 37269961 DOI: 10.1016/j.cellsig.2023.110739] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 04/17/2023] [Accepted: 05/27/2023] [Indexed: 06/05/2023]
Abstract
Protein modifications have significant effects on tumorigenesis. N-Myristoylation is one of the most important lipidation modifications, and N-myristoyltransferase 1 (NMT1) is the main enzyme required for this process. However, the mechanism underlying how NMT1 modulates tumorigenesis remains largely unclear. Here, we found that NMT1 sustains cell adhesion and suppresses tumor cell migration. Intracellular adhesion molecule 1 (ICAM-1) was a potential functional downstream effector of NMT1, and its N-terminus could be N-myristoylated. NMT1 prevented ubiquitination and proteasome degradation of ICAM-1 by inhibiting Ub E3 ligase F-box protein 4, which prolonged the half-life of ICAM1 protein. Correlations between NMT1 and ICAM-1 were observed in liver and lung cancers, which were associated with metastasis and overall survival. Therefore, carefully designed strategies focusing on NMT1 and its downstream effectors might be helpful to treat tumors.
Collapse
Affiliation(s)
- Hong Wang
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xin Xu
- Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, 200030, China
| | - Yikun Wang
- Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, 200030, China
| | - Xiangfei Xue
- Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, 200030, China
| | - Wanxin Guo
- Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, 200030, China
| | - Susu Guo
- Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, 200030, China
| | - Shiyu Qiu
- Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, 200030, China
| | - Jiangtao Cui
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, 200030, China
| | - Yongxia Qiao
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China.
| |
Collapse
|
20
|
Long C, Li G, Meng Y, Huang X, Chen J, Liu J. Weighted gene co-expression network analysis identifies the prognosis-related models of left- and right-sided colon cancer. Medicine (Baltimore) 2023; 102:e33390. [PMID: 37144998 PMCID: PMC10158920 DOI: 10.1097/md.0000000000033390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 03/08/2023] [Indexed: 05/06/2023] Open
Abstract
Left-sided colon cancer (LC) and right-sided colon cancer (RC) are 2 essentially different diseases, and the potential mechanisms regulating them remain unidentified. In this study, we applied weighted gene co-expression network analysis (WGCNA) to confirm a yellow module, mainly enriched in metabolism-related signaling pathways related to LC and RC. Based on the RNA-seq data of colon cancer in The Cancer Genome Atlas (TCGA) and GSE41258 dataset with their corresponding clinical information, a training set (TCGA: LC: n = 171; RC: n = 260) and a validation set (GSE41258: LC: n = 94; RC: n = 77) were divided. Least absolute shrinkage and selection operator (LASSO) penalized COX regression analysis identified 20 prognosis-related genes (PRGs) and helped constructed 2 risk (LC-R and RC-R) models in LC and RC, respectively. The model-based risk scores accurately performed in risk stratification for colon cancer patients. The high-risk group of the LC-R model showed associations with ECM-receptor interaction, focal adhesion, and PI3K-AKT signaling pathway. Interestingly, the low-risk group of the LC-R model showed associations with immune-related signaling pathways like antigen processing and presentation. On the other hand, the high-risk group of the RC-R model showed enrichment for cell adhesion molecules and axon guidance signaling pathways. Furthermore, we identified 20 differentially expressed PRGs between LC and RC. Our findings provide new insights into the difference between LC and RC, and uncover the potential biomarkers for the treatment of LC and RC.
Collapse
Affiliation(s)
- Chenyan Long
- Division of Colorectal & Anal Surgery, Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, Guangxi Zhuang Autonomous Region, The People’s Republic of China
- Guangxi Clinical Research Center for Colorectal Cancer, Nanning, Guangxi Zhuang Autonomous Region, The People’s Republic of China
| | - Gang Li
- School of Public Health, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, The People’s Republic of China
| | - Yongsheng Meng
- Division of Colorectal & Anal Surgery, Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, Guangxi Zhuang Autonomous Region, The People’s Republic of China
- Guangxi Clinical Research Center for Colorectal Cancer, Nanning, Guangxi Zhuang Autonomous Region, The People’s Republic of China
| | - Xiaoliang Huang
- Division of Colorectal & Anal Surgery, Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, Guangxi Zhuang Autonomous Region, The People’s Republic of China
- Guangxi Clinical Research Center for Colorectal Cancer, Nanning, Guangxi Zhuang Autonomous Region, The People’s Republic of China
| | - Jianhong Chen
- Division of Colorectal & Anal Surgery, Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, Guangxi Zhuang Autonomous Region, The People’s Republic of China
- Guangxi Clinical Research Center for Colorectal Cancer, Nanning, Guangxi Zhuang Autonomous Region, The People’s Republic of China
| | - Jungang Liu
- Division of Colorectal & Anal Surgery, Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, Guangxi Zhuang Autonomous Region, The People’s Republic of China
- Guangxi Clinical Research Center for Colorectal Cancer, Nanning, Guangxi Zhuang Autonomous Region, The People’s Republic of China
| |
Collapse
|
21
|
Faria L, Canato S, Jesus TT, Gonçalves M, Guerreiro PS, Lopes CS, Meireles I, Morais-de-Sá E, Paredes J, Janody F. Activation of an actin signaling pathway in pre-malignant mammary epithelial cells by P-cadherin is essential for transformation. Dis Model Mech 2023; 16:dmm049652. [PMID: 36808468 PMCID: PMC9983776 DOI: 10.1242/dmm.049652] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 01/19/2023] [Indexed: 02/23/2023] Open
Abstract
Alterations in the expression or function of cell adhesion molecules have been implicated in all steps of tumor progression. Among those, P-cadherin is highly enriched in basal-like breast carcinomas, playing a central role in cancer cell self-renewal, collective cell migration and invasion. To establish a clinically relevant platform for functional exploration of P-cadherin effectors in vivo, we generated a humanized P-cadherin Drosophila model. We report that actin nucleators, Mrtf and Srf, are main P-cadherin effectors in fly. We validated these findings in a human mammary epithelial cell line with conditional activation of the SRC oncogene. We show that, prior to promoting malignant phenotypes, SRC induces a transient increase in P-cadherin expression, which correlates with MRTF-A accumulation, its nuclear translocation and the upregulation of SRF target genes. Moreover, knocking down P-cadherin, or preventing F-actin polymerization, impairs SRF transcriptional activity. Furthermore, blocking MRTF-A nuclear translocation hampers proliferation, self-renewal and invasion. Thus, in addition to sustaining malignant phenotypes, P-cadherin can also play a major role in the early stages of breast carcinogenesis by promoting a transient boost of MRTF-A-SRF signaling through actin regulation.
Collapse
Affiliation(s)
- Lídia Faria
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- Instituto de Patologia e Imunologia Molecular da Universidade do Porto (Ipatimup), Rua Júlio Amaral de Carvalho, n 45, 4200-135 Porto, Portugal
- Master Programme in Oncology, School of Medicine and Biomedical Sciences, University of Porto (ICBAS-UP), Rua Jorge Viterbo Ferreira 228, 4050-513 Porto, Portugal
| | - Sara Canato
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- Instituto de Patologia e Imunologia Molecular da Universidade do Porto (Ipatimup), Rua Júlio Amaral de Carvalho, n 45, 4200-135 Porto, Portugal
- Physiology and Cancer Program, Champalimaud Foundation, Avenida de Brasília, 1400-038 Lisboa, Portugal
| | - Tito T. Jesus
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- Instituto de Patologia e Imunologia Molecular da Universidade do Porto (Ipatimup), Rua Júlio Amaral de Carvalho, n 45, 4200-135 Porto, Portugal
| | - Margarida Gonçalves
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
| | - Patrícia S. Guerreiro
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- Instituto de Patologia e Imunologia Molecular da Universidade do Porto (Ipatimup), Rua Júlio Amaral de Carvalho, n 45, 4200-135 Porto, Portugal
- Vector B2B - Drug Developing - Associação Para Investigação em Biotecnologia, Av. Prof. Egas Moniz, Edifício Egas Moniz, 1649-028 Lisboa, Portugal
| | - Carla S. Lopes
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
| | - Isabel Meireles
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- Instituto de Patologia e Imunologia Molecular da Universidade do Porto (Ipatimup), Rua Júlio Amaral de Carvalho, n 45, 4200-135 Porto, Portugal
| | - Eurico Morais-de-Sá
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
| | - Joana Paredes
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- Instituto de Patologia e Imunologia Molecular da Universidade do Porto (Ipatimup), Rua Júlio Amaral de Carvalho, n 45, 4200-135 Porto, Portugal
- FMUP, Medical Faculty of University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Florence Janody
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- Instituto de Patologia e Imunologia Molecular da Universidade do Porto (Ipatimup), Rua Júlio Amaral de Carvalho, n 45, 4200-135 Porto, Portugal
- Instituto Gulbenkian de Ciência, Rua da Quinta Grande 6, P-2780-156 Oeiras, Portugal
| |
Collapse
|
22
|
Mostafa H, Behrendt I, Meroño T, González-Domínguez R, Fasshauer M, Rudloff S, Andres-Lacueva C, Kuntz S. Plasma anthocyanins and their metabolites reduce in vitro migration of pancreatic cancer cells, PANC-1, in a FAK- and NF-kB dependent manner: Results from the ATTACH-study a randomized, controlled, crossover trial in healthy subjects. Biomed Pharmacother 2023; 158:114076. [PMID: 36516693 DOI: 10.1016/j.biopha.2022.114076] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 11/25/2022] [Accepted: 12/02/2022] [Indexed: 12/15/2022] Open
Abstract
Pancreatic cancer is primarily considered to be a metastatic disease with a low 5-year survival rate. We aimed to detect if plasma-isolated anthocyanins and their metabolites (PAMs) modulate pancreatic cancer cells migration and to describe molecular targets of PAMs in this process. Plasma metabolites were isolated by solid-phase extraction before and after a 28-days intervention trial involving 35 healthy subjects comparing effects of a daily anthocyanin-rich juice intake vs. placebo. Plasma extracts were used for migration and mechanistic in vitro studies as well as for metabolomic analysis. Pancreatic PANC-1 and AsPC-1 were used for migration studies in a Boyden chamber co-cultured with endothelial cells. Expression of adhesion molecules on cancer and endothelial cells were determined by flow cytometry and NF-kB (nuclear factor-kappa B) p65 and focal adhesion kinase activation were measured by immunoassays. UHPLC-MS/MS metabolomics was done in plasma and urine samples. Plasma extracts isolated after the intake of the anthocyanin-rich juice significantly reduced PANC-1 migration, but not AsPC-1 migration. In PANC-1, and to a lower extent in endothelial cells, plasma extracts after juice intake decreased the expression of ß1- and ß4-integrins and intercellular adhesion molecule-1. Pooled plasma from volunteers with the highest inhibition of PANC-1 migration (n = 10) induced a reduction of NF-kB-p65 and FAK-phosphorylation in cancer and in endothelial cells. Concerning metabolites, 14 were significantly altered by juice intervention and PANC-1 migration was inversely associated with the increase of o-coumaric acid and peonidin-3-galactoside. PAMs were associated with lower PANC-1 cell migration opening new strategies for metastatic pancreatic cancer treatment.
Collapse
Affiliation(s)
- Hamza Mostafa
- Biomarkers and Nutrimetabolomics Laboratory, Department of Nutrition, Food Sciences and Gastronomy, Food Innovation Network (XIA), Nutrition and Food Safety Research Institute (INSA), Facultat de Farmàcia i Ciències de l'Alimentació, Universitat de Barcelona (UB), 08028 Barcelona, Spain; Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Inken Behrendt
- Department of Nutritional Science, Human Nutrition, Justus-Liebig-University, 35390 Giessen, Germany.
| | - Tomás Meroño
- Biomarkers and Nutrimetabolomics Laboratory, Department of Nutrition, Food Sciences and Gastronomy, Food Innovation Network (XIA), Nutrition and Food Safety Research Institute (INSA), Facultat de Farmàcia i Ciències de l'Alimentació, Universitat de Barcelona (UB), 08028 Barcelona, Spain; Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid 28029, Spain.
| | - Raúl González-Domínguez
- Biomarkers and Nutrimetabolomics Laboratory, Department of Nutrition, Food Sciences and Gastronomy, Food Innovation Network (XIA), Nutrition and Food Safety Research Institute (INSA), Facultat de Farmàcia i Ciències de l'Alimentació, Universitat de Barcelona (UB), 08028 Barcelona, Spain; Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Mathias Fasshauer
- Department of Nutritional Science, Human Nutrition, Justus-Liebig-University, 35390 Giessen, Germany
| | - Silvia Rudloff
- Department of Nutritional Science, Human Nutrition, Justus-Liebig-University, 35390 Giessen, Germany; Department of Nutritional Science and Department of Pediatrics, Justus-Liebig-University, 35392 Giessen, Germany
| | - Cristina Andres-Lacueva
- Biomarkers and Nutrimetabolomics Laboratory, Department of Nutrition, Food Sciences and Gastronomy, Food Innovation Network (XIA), Nutrition and Food Safety Research Institute (INSA), Facultat de Farmàcia i Ciències de l'Alimentació, Universitat de Barcelona (UB), 08028 Barcelona, Spain; Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Sabine Kuntz
- Department of Nutritional Science, Human Nutrition, Justus-Liebig-University, 35390 Giessen, Germany
| |
Collapse
|
23
|
Fereshteh Z, Dang MN, Wenck C, Day ES, Slater JH. E-Selectin Targeted Gold Nanoshells to Inhibit Breast Cancer Cell Binding to Lung Endothelial Cells. ACS APPLIED NANO MATERIALS 2023; 6:1315-1324. [PMID: 37789828 PMCID: PMC10544796 DOI: 10.1021/acsanm.2c04967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/05/2023]
Abstract
Extravasation of circulating tumor cells (CTCs) from the vasculature is a key step in cancer metastasis. CTCs bind to cell adhesion molecules (CAMs) expressed by endothelial cells (ECs) for flow arrest prior to extravasation. While a number of EC-expressed CAMs have been implicated in facilitating CTC binding, this work investigated the efficacy of inhibiting cancer cell binding to human lung microvascular ECs via antibody blocking of E-selectin using antibody-functionalized gold nanoshells (NS). The antibody-functionalized gold NS were synthesized using both directional and non-directional antibody conjugation techniques with variations in synthesis parameters (linker length, amount of passivating agents, and ratio of antibodies to NS) to gain a better understanding of these properties on the resultant hydrodynamic diameter, zeta potential, and antibody loading density. We quantified the ability of E-selectin antibody-functionalized NS to bind human lung microvascular endothelial cells (HMVEC-Ls) under non-inflamed and inflamed (TNF-α) conditions to inhibit binding of triple-negative MDA-MB-231s. E-selectin-targeted NS prepared using non-directional conjugation had higher antibody loading than those prepared via directional conjugation, resulting in the conjugates having similar overall binding to HMVEC-Ls at a given antibody concentration. E-selectin-targeted NS reduced MDA-MB-231 binding to HMVEC-Ls by up to 41% as determined using an in vitro binding assay. These results provide useful insights into the characteristics of antibody-functionalized NS prepared under different conditions while also demonstrating proof of concept that these conjugates hold potential to inhibit CTC binding to ECs, a critical step in extravasation during metastasis.
Collapse
Affiliation(s)
- Z Fereshteh
- Department of Biomedical Engineering, University of Delaware, Newark, Delaware 19713, United States
| | - M N Dang
- Department of Biomedical Engineering, University of Delaware, Newark, Delaware 19713, United States
| | - C Wenck
- Department of Biomedical Engineering, University of Delaware, Newark, Delaware 19713, United States
| | - E S Day
- Department of Biomedical Engineering, University of Delaware, Newark, Delaware 19713, United States
| | - J H Slater
- Department of Biomedical Engineering, University of Delaware, Newark, Delaware 19713, United States
| |
Collapse
|
24
|
de-Souza-Ferreira M, Ferreira ÉE, de-Freitas-Junior JCM. Aberrant N-glycosylation in cancer: MGAT5 and β1,6-GlcNAc branched N-glycans as critical regulators of tumor development and progression. Cell Oncol 2023; 46:481-501. [PMID: 36689079 DOI: 10.1007/s13402-023-00770-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/03/2023] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND Changes in protein glycosylation are widely observed in tumor cells. N-glycan branching through adding β1,6-linked N-acetylglucosamine (β1,6-GlcNAc) to an α1,6-linked mannose, which is catalyzed by the N-acetylglucosaminyltransferase V (MGAT5 or GnT-V), is one of the most frequently observed tumor-associated glycan structure formed. Increased levels of this branching structure play a pro-tumoral role in various ways, for example, through the stabilization of growth factor receptors, the destabilization of intercellular adhesion, or the acquisition of a migratory phenotype. CONCLUSION In this review, we provide an updated and comprehensive summary of the physiological and pathophysiological roles of MGAT5 and β1,6-GlcNAc branched N-glycans, including their regulatory mechanisms. Specific emphasis is given to the role of MGAT5 and β1,6-GlcNAc branched N-glycans in cellular mechanisms that contribute to the development and progression of solid tumors. We also provide insight into possible future clinical implications, such as the use of MGAT5 as a prognostic biomarker.
Collapse
Affiliation(s)
- Michelle de-Souza-Ferreira
- Cellular and Molecular Oncobiology Program, Cancer Glycobiology Group, Brazilian National Cancer Institute (INCA), 37 André Cavalcanti Street, Rio de Janeiro, RJ, 20231-050, Brazil
| | - Érika Elias Ferreira
- Cellular and Molecular Oncobiology Program, Cancer Glycobiology Group, Brazilian National Cancer Institute (INCA), 37 André Cavalcanti Street, Rio de Janeiro, RJ, 20231-050, Brazil
| | - Julio Cesar Madureira de-Freitas-Junior
- Cellular and Molecular Oncobiology Program, Cancer Glycobiology Group, Brazilian National Cancer Institute (INCA), 37 André Cavalcanti Street, Rio de Janeiro, RJ, 20231-050, Brazil.
| |
Collapse
|
25
|
Qiu Z, Wang Y, Zhang Z, Qin R, Peng Y, Tang W, Xi Y, Tian G, Zhang Y. Roles of intercellular cell adhesion molecule-1 (ICAM-1) in colorectal cancer: expression, functions, prognosis, tumorigenesis, polymorphisms and therapeutic implications. Front Oncol 2022; 12:1052672. [PMID: 36505809 PMCID: PMC9728583 DOI: 10.3389/fonc.2022.1052672] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 11/09/2022] [Indexed: 11/24/2022] Open
Abstract
Colorectal cancer (CRC) is a major global health problem and one of the major causes of cancer-related death worldwide. It is very important to understand the pathogenesis of CRC for early diagnosis, prevention strategies and identification of new therapeutic targets. Intercellular adhesion molecule-1 (ICAM-1, CD54) displays an important role in the the pathogenesis of CRC. It is a cell surface glycoprotein of the immunoglobulin (Ig) superfamily and plays an essential role in cell-cell, cell-extracellular matrix interaction, cell signaling and immune process. It is also expressed by tumor cells and modulates their functions, including apoptosis, cell motility, invasion and angiogenesis. The interaction between ICAM-1 and its ligand may facilitate adhesion of tumor cells to the vascular endothelium and subsequently in the promotion of metastasis. ICAM-1 expression determines malignant potential of cancer. In this review, we will discuss the expression, function, prognosis, tumorigenesis, polymorphisms and therapeutic implications of ICAM-1 in CRC.
Collapse
Affiliation(s)
- Zhiyuan Qiu
- Department of Oncology, the Affiliated People’s Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Yan Wang
- Department of Oncology, the Affiliated People’s Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Zhao Zhang
- Department of Oncology, the Affiliated People’s Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Rong Qin
- Department of Oncology, the Affiliated People’s Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Yong Peng
- Department of Oncology, the Affiliated People’s Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Weifeng Tang
- Department of Cardiothoracic Surgery, Nanjing Drum Tower Hospital, Nanjing, Jiangsu, China
| | - Yan Xi
- Department of Geriatrics, the Affiliated People’s Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Guangyu Tian
- Department of Oncology, Jiangdu People’s Hospital Affiliated to Medical College of Yangzhou University, Yangzhou, Jiangsu, China
| | - Yeqing Zhang
- Department of Vascular Surgery, the Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
26
|
Sivakumar N, Warner HV, Peirce SM, Lazzara MJ. A computational modeling approach for predicting multicell spheroid patterns based on signaling-induced differential adhesion. PLoS Comput Biol 2022; 18:e1010701. [PMID: 36441822 PMCID: PMC9747056 DOI: 10.1371/journal.pcbi.1010701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 12/13/2022] [Accepted: 11/01/2022] [Indexed: 11/29/2022] Open
Abstract
Physiological and pathological processes including embryogenesis and tumorigenesis rely on the ability of individual cells to work collectively to form multicell patterns. In these heterogeneous multicell systems, cell-cell signaling induces differential adhesion between cells that leads to tissue-level patterning. However, the sensitivity of pattern formation to changes in the strengths of signaling or cell adhesion processes is not well understood. Prior work has explored these issues using synthetically engineered heterogeneous multicell spheroid systems, in which cell subpopulations engage in bidirectional intercellular signaling to regulate the expression of different cadherins. While engineered cell systems provide excellent experimental tools to observe pattern formation in cell populations, computational models of these systems may be leveraged to explore more systematically how specific combinations of signaling and adhesion parameters can drive the emergence of unique patterns. We developed and validated two- and three-dimensional agent-based models (ABMs) of spheroid patterning for previously described cells engineered with a bidirectional signaling circuit that regulates N- and P-cadherin expression. Systematic exploration of model predictions, some of which were experimentally validated, revealed how cell seeding parameters, the order of signaling events, probabilities of induced cadherin expression, and homotypic adhesion strengths affect pattern formation. Unsupervised clustering was also used to map combinations of signaling and adhesion parameters to these unique spheroid patterns predicted by the ABM. Finally, we demonstrated how the model may be deployed to design new synthetic cell signaling circuits based on a desired final multicell pattern.
Collapse
Affiliation(s)
- Nikita Sivakumar
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, United States of America
| | - Helen V. Warner
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, United States of America
| | - Shayn M. Peirce
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, United States of America
| | - Matthew J. Lazzara
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, United States of America
- Department of Chemical Engineering, University of Virginia, Charlottesville, Virginia, United States of America
- * E-mail:
| |
Collapse
|
27
|
Narasimhan H, Ferraro F, Bleilevens A, Weiskirchen R, Stickeler E, Maurer J. Tumor Necrosis Factor-α (TNFα) Stimulate Triple-Negative Breast Cancer Stem Cells to Promote Intratumoral Invasion and Neovasculogenesis in the Liver of a Xenograft Model. BIOLOGY 2022; 11:biology11101481. [PMID: 36290384 PMCID: PMC9598572 DOI: 10.3390/biology11101481] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 09/28/2022] [Accepted: 10/01/2022] [Indexed: 11/06/2022]
Abstract
TNBC represents the most aggressive breast cancer subtype. Although cancer stem cells (CSCs) are a minor fraction of all cancer cells, they are highly cancerous when compared to their non-stem counterparts, playing a major role in tumor recurrence and metastasis. Angiogenic stimuli and the tumor environment response are vital factors in cancer metastasis. However, the causes and effects of tumor angiogenesis are still poorly understood. In this study, we demonstrate TNFα effects on primary triple-negative breast cancer stem cells (BCSCs). TNFα stimulation increased the mesenchymality of BCSCs in an intermediate epithelial-to-mesenchymal transition (EMT) state, enhanced proliferation, self-renewal, and invasive capacity. TNFα-treatment elicited BCSC signaling on endothelial networks in vitro and increased the network forming capacity of the endothelial cells. Our findings further demonstrate that TNFα stimulation in BCSCs has the ability to instigate distinct cellular communication within the tumor microenvironment, inducing intra-tumoral stromal invasion. Further, TNFα-treatment in BCSCs induced a pre-metastatic niche through breast-liver organ crosstalk by inducing vascular cell adhesion molecule-1 (VCAM-1) enriched neovasculogenesis in the liver of tumor-bearing mice. Overall, TNFα is an important angiogenic target to be considered in breast cancer progression to attenuate any angiogenic response in the tumor environment that could lead to secondary organ metastasis.
Collapse
Affiliation(s)
- Harini Narasimhan
- Department of Obstetrics and Gynecology, University Hospital Aachen, D-52074 Aachen, Germany
| | - Francesca Ferraro
- Department of Obstetrics and Gynecology, University Hospital Aachen, D-52074 Aachen, Germany
| | - Andreas Bleilevens
- Department of Obstetrics and Gynecology, University Hospital Aachen, D-52074 Aachen, Germany
| | - Ralf Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC) RWTH University Hospital Aachen, D-52074 Aachen, Germany
| | - Elmar Stickeler
- Department of Obstetrics and Gynecology, University Hospital Aachen, D-52074 Aachen, Germany
- Center for Integrated Oncology (CIO), Aachen, Bonn, Cologne, Düsseldorf (ABCD), Venusberg-Campus 1, 53127 Bonn, Germany
| | - Jochen Maurer
- Department of Obstetrics and Gynecology, University Hospital Aachen, D-52074 Aachen, Germany
- Center for Integrated Oncology (CIO), Aachen, Bonn, Cologne, Düsseldorf (ABCD), Venusberg-Campus 1, 53127 Bonn, Germany
| |
Collapse
|
28
|
Zhang X, Ding C, Zhao Z. Exploring a 7-gene prognostic model based on ferroptosis for efficiently guiding immunotherapy in melanoma patients. Adv Med Sci 2022; 67:364-378. [PMID: 36155341 DOI: 10.1016/j.advms.2022.09.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Revised: 09/09/2022] [Accepted: 09/10/2022] [Indexed: 11/01/2022]
Abstract
PURPOSE Although skin cutaneous melanoma (SKCM) is a relatively immunotherapy-sensitive tumor type, there is still a certain fraction that benefits less from treatment. Ferroptosis has been demonstrated to modulate tumor progression in many cancer types. This study focused on ferroptosis-related genes to construct a prognostic model for SKCM patients. MATERIALS AND METHODS Gene expression profiles of SKCM samples were obtained from public databases. Unsupervised consensus clustering was used to determine molecular subtypes related to ferroptosis. Least absolute shrinkage and selection operator (LASSO) and stepwise Akaike information criterion (stepAIC) were applied to construct a prognostic model based on differentially expressed genes between two molecular subtypes. RESULTS C1 and C2 subtypes were identified with differential prognosis and immune infiltration. A 7-gene prognostic model was constructed to classify samples into high-FPRS and low-FPRS groups. Low-FPRS group with favorable prognosis had higher immune infiltration and more enriched immune-related pathways than the high-FPRS group. The two groups showed distinct sensitivity to immunotherapy, with the low-FPRS group predicted to have more positive response to immunotherapy than the high-FPRS group. A nomogram based on the FPRS score and clinical features was built for more convenient use. CONCLUSIONS The critical role of ferroptosis involved in SKCM development was further validated in this study. The prognostic model was efficient and stable to be applied in clinical conditions to support clinicians in determining personalized therapy for SKCM patients especially those with metastasis.
Collapse
Affiliation(s)
- Xin Zhang
- Department of Dermatology, The First Medical Center of PLA General Hospital, Beijing, China
| | - Changrui Ding
- Department of Dermatology, The First Affiliated Hospital of Qiqihar Medical College, Qiqihar City, Heilongjiang Province, China
| | - Zigang Zhao
- Department of Dermatology, Hainan Hospital of PLA General Hospital, Sanya City, Hainan Province, China.
| |
Collapse
|
29
|
Immunoglobulin superfamily 9 (IGSF9) is trans-activated by p53, inhibits breast cancer metastasis via FAK. Oncogene 2022; 41:4658-4672. [PMID: 36088502 PMCID: PMC9546770 DOI: 10.1038/s41388-022-02459-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Revised: 08/23/2022] [Accepted: 08/26/2022] [Indexed: 11/08/2022]
Abstract
AbstractMetastasis of breast cancer represents the major reason for its poor prognosis, leading to high mortality. In breast cancer, a tumor suppressor gene TP53 is commonly mutated. TP53 mutation leads to an altered expression of various genes, an event that is associated with aggressive tumor and is a strong independent marker for survival. In this study, we identified a novel p53 target gene, immunoglobulin superfamily 9 (IGSF9). IGSF9 is generally down-regulated in breast cancer tissues. Loss of IGSF9 is associated with frequent metastasis and poor prognosis of breast cancer patients. Wild-type p53, but not R175H mutant, trans-activates the transcription of IGSF9 via binding to its promoter (−137 to −131 bp), inhibits epithelial-mesenchymal transition (EMT), consequently the inhibition of breast cancer cells migration and invasion. IGSF9 interacts with focal adhesion kinase (FAK) and inhibits FAK/AKT signaling activity. PND1186, FAK inhibitor, inhibits breast cancer metastasis induced by IGSF9 knockdown in vitro and in vivo. Taken together, IGSF9 is trans-activated by p53 and inhibits breast cancer metastasis by modulating FAK/AKT signaling pathway. IGSF9 could serve as a prognostic marker and potential therapeutic target for breast cancer.
Collapse
|
30
|
Alonso-Diez A, Affolter V, Sevane N, Dunner S, Valdivia G, Clemente M, De Andrés P, Illera J, Pérez-Alenza M, Peña L. Cell adhesion molecules E-cadherin and CADM1 are differently expressed in canine inflammatory mammary cancer. Res Vet Sci 2022; 152:307-313. [DOI: 10.1016/j.rvsc.2022.08.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 07/27/2022] [Accepted: 08/23/2022] [Indexed: 10/14/2022]
|
31
|
Ma H, Liu G, Yu B, Wang J, Qi Y, Kou Y, Hu Y, Wang S, Wang F, Chen D. RNA-binding protein CELF6 modulates transcription and splicing levels of genes associated with tumorigenesis in lung cancer A549 cells. PeerJ 2022; 10:e13800. [PMID: 35910766 PMCID: PMC9336609 DOI: 10.7717/peerj.13800] [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: 12/13/2021] [Accepted: 07/06/2022] [Indexed: 01/18/2023] Open
Abstract
CELF6 (CUGBP Elav-Like Family Member 6), a canonical RNA binding protein (RBP), plays important roles in post-transcriptional regulation of pre-mRNAs. However, the underlying mechanism of lower expressed CELF6 in lung cancer tissues is still unclear. In this study, we increased CELF6 manually in lung cancer cell line (A549) and utilized transcriptome sequencing (RNA-seq) technology to screen out differentially expressed genes (DEGs) and alternative splicing events (ASEs) after CELF6 over-expression (CELF6-OE). We found that CELF6-OE induced 417 up-regulated and 1,351 down-regulated DEGs. Functional analysis of down-regulated DEGs showed that they were highly enriched in immune/inflammation response- related pathways and cell adhesion molecules (CAMs). We also found that CELF6 inhibited the expression of many immune-related genes, including TNFSF10, CCL5, JUNB, BIRC3, MLKL, PIK3R2, CCL20, STAT1, MYD88, and CFS1, which mainly promote tumorigenesis in lung cancer. The dysregulated DEGs were also validated by reverse transcriptase quantitative polymerase chain reaction (RT-qPCR) experiment. In addition, CELF6 regulates the splicing pattern of large number of genes that are enriched in p53 signaling pathway and apoptosis, including TP53 and CD44. In summary, we made an extensive analysis of the transcriptome profile of gene expression and alternative splicing by CELF6-OE, providing a global understanding of the target genes and underlying regulation mechanisms mediated by CELF6 in the pathogenesis and development of lung cancer.
Collapse
Affiliation(s)
- HuSai Ma
- Department of thoracic Surgery, Qinghai Provincial People’s Hospital, Xining, Qinghai Province, China,Department of Thoracic Surgery, Qinghai Red Cross Hospital, Xining, Qinghai Province, P. R. China
| | - GuoWei Liu
- Department of thoracic Surgery, Qinghai Provincial People’s Hospital, Xining, Qinghai Province, China
| | - Bin Yu
- Department of thoracic Surgery, Qinghai Provincial People’s Hospital, Xining, Qinghai Province, China
| | - Joshua Wang
- Wuhan Ruixing Biotechnology Co. Ltd., Wuhan, Hubei Province, China
| | - YaLi Qi
- Department of Respiratory Medicine, Qinghai Provincial People’s Hospital, Xining, Qinghai Province, China
| | - YiYing Kou
- Qinghai University School of Medicine, Qinghai University School of Medicine, Xining, Qinghai Province, China
| | - Ying Hu
- Qinghai University School of Medicine, Qinghai University School of Medicine, Xining, Qinghai Province, China
| | - ShunJun Wang
- Department of thoracic Surgery, Qinghai Provincial People’s Hospital, Xining, Qinghai Province, China
| | - Fei Wang
- Qinghai University, Xining, Qinghai Province, China
| | - Dong Chen
- Wuhan Ruixing Biotechnology Co. Ltd., Wuhan, Hubei Province, China
| |
Collapse
|
32
|
Serum and Pleural Soluble Cell Adhesion Molecules in Mesothelioma Patients: A Retrospective Cohort Study. Cancers (Basel) 2022; 14:cancers14122825. [PMID: 35740491 PMCID: PMC9221497 DOI: 10.3390/cancers14122825] [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: 04/19/2022] [Revised: 05/29/2022] [Accepted: 06/06/2022] [Indexed: 12/04/2022] Open
Abstract
Mesothelioma, a malignant neoplasm of mesothelial cells, has overall poor prognosis. Cell adhesion molecules (CAMs) are proteins that contribute to the immune response. In this study the clinical utility and prognostic significance of serum and pleural fluid soluble CAM (sCAM) levels were assessed in patients with mesothelioma. Mesothelioma patients were retrospectively recruited (2016-2020). Clinical characteristics, serum and pleural sCAM levels (sE-cadherin, sE-selectin, intercellular adhesion molecule 1 (sICAM-1) and vascular cell adhesion molecule 1 (sVCAM-1)) and histopathological characteristics were gathered. A total of 51 healthy controls were also recruited for a secondary cross-sectional analysis. 92 mesothelioma patients were analyzed (mean age 64.5 years, 87% males, performance status 0-2). Patients with increased pleural sE-cadherin had higher risk for disease progression (adjusted HR 1.11 (1.02, 1.20), p = 0.013). Serum and pleural sE-selectin were decreased in patients with high-grade mesothelioma. Patients with increased serum or pleural sE-selectin levels had lower risk for death (adjusted HR 0.88 (0.81, 0.96), p = 0.003; 0.90 (0.82, 0.99), p = 0.039, respectively). Serum sE-cadherin, sE-selectin and sICAM-1 levels were significantly increased in mesothelioma patients compared to healthy controls. Further studies are needed to indicate the clinical utility of serum and pleural sCAMs in mesothelioma patients.
Collapse
|
33
|
Choi HS, Kim YK, Yun PY. Assessing Gene Expression Related to Cisplatin Resistance in Human Oral Squamous Cell Carcinoma Cell Lines. Pharmaceuticals (Basel) 2022; 15:ph15060704. [PMID: 35745623 PMCID: PMC9228236 DOI: 10.3390/ph15060704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 05/26/2022] [Accepted: 06/01/2022] [Indexed: 11/16/2022] Open
Abstract
Cisplatin-based chemotherapy has been effectively used to treat oral cancer, but treatment often fails owing to the development of drug resistance. However, the important gene expression alterations associated with these resistances remain unclear. In this study, we aimed to identify the gene expressions related to cisplatin resistance in oral squamous cell carcinoma (OSCC) cell lines. RNA samples were obtained from three cisplatin-resistant (YD-8/CIS, YD-9/CIS, and YD-38/CIS) and -sensitive (YD-8, YD-9, and YD-38) cell lines. Global gene expression was analyzed using RNA sequencing (RNA-Seq). Differentially expressed genes were determined. Based on the gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) databases, functional enrichment and signaling pathways analyses were performed. Candidate genes selected from RNA-Seq analysis were validated by quantitative real-time polymerase chain reaction (qRT-PCR) analysis. The YD-8/CIS and YD-9/CIS samples had very similar expression patterns. qRT-PCR analysis was performed on selected genes commonly expressed between the two samples. The expression levels of 11 genes were changed in cisplatin-resistant samples compared with their parental samples; several of these genes were related to cell adhesion molecules and proteoglycans in cancer pathways. Our data provide candidate genes associated with cisplatin resistance in OSCC, but further study is required to determine which genes have an important role. Nevertheless, these results may provide new ideas to improve the clinical therapeutic outcomes of OSCC.
Collapse
Affiliation(s)
- Hyeong Sim Choi
- Department of Oral and Maxillofacial Surgery, Section of Dentistry, Seoul National University Bundang Hospital, 82 Gumi-ro 173 beon-gil, Bundang-gu, Seongnam 13620, Korea; (H.S.C.); (Y.-K.K.)
| | - Young-Kyun Kim
- Department of Oral and Maxillofacial Surgery, Section of Dentistry, Seoul National University Bundang Hospital, 82 Gumi-ro 173 beon-gil, Bundang-gu, Seongnam 13620, Korea; (H.S.C.); (Y.-K.K.)
| | - Pil-Young Yun
- Department of Oral and Maxillofacial Surgery, Section of Dentistry, Seoul National University Bundang Hospital, 82 Gumi-ro 173 beon-gil, Bundang-gu, Seongnam 13620, Korea; (H.S.C.); (Y.-K.K.)
- Department of Dentistry and Dental Research Institute, School of Dentistry, Seoul National University, 101 Daehak-ro, Jongno-gu, Seoul 03080, Korea
- Correspondence: ; Tel.: +82-31-787-7545; Fax: +82-31-787-4068
| |
Collapse
|
34
|
Zhang Y, Zhang L, Zheng S, Li M, Xu C, Jia D, Qi Y, Hou T, Wang L, Wang B, Li A, Chen S, Si J, Zhuo W. Fusobacterium nucleatum promotes colorectal cancer cells adhesion to endothelial cells and facilitates extravasation and metastasis by inducing ALPK1/NF-κB/ICAM1 axis. Gut Microbes 2022; 14:2038852. [PMID: 35220887 PMCID: PMC8890384 DOI: 10.1080/19490976.2022.2038852] [Citation(s) in RCA: 81] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Metastasis is the leading cause of death for colorectal cancer (CRC) patients, and the spreading tumor cells adhesion to endothelial cells is a critical step for extravasation and further distant metastasis. Previous studies have documented the important roles of gut microbiota-host interactions in the CRC malignancy, and Fusobacterium nucleatum (F. nucleatum) was reported to increase proliferation and invasive activities of CRC cells. However, the potential functions and underlying mechanisms of F. nucleatum in the interactions between CRC cells and endothelial cells and subsequent extravasation remain unclear. Here, we uncovered that F. nucleatum enhanced the adhesion of CRC cells to endothelial cells, promoted extravasation and metastasis by inducing ICAM1 expression. Mechanistically, we identified that F. nucleatum induced a new pattern recognition receptor ALPK1 to activate NF-κB pathway, resulting in the upregulation of ICAM1. Interestingly, the abundance of F. nucleatum in tumor tissues of CRC patients was positively associated with the expression levels of ALPK1 and ICAM1. Moreover, high expression of ALPK1 or ICAM1 was significantly associated with a shorter overall survival time of CRC patients. This study provides a new insight into the role of gut microbiota in engaging into the distant metastasis of CRC cells.
Collapse
Affiliation(s)
- Ying Zhang
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China,Cancer Center, Zhejiang University, Hangzhou, China,Institute of Gastroenterology, Zhejiang University, Hangzhou, China
| | - Lu Zhang
- Department of Cell Biology and Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China,Cancer Center, Zhejiang University, Hangzhou, China,Institute of Gastroenterology, Zhejiang University, Hangzhou, China
| | - Sheng Zheng
- Cancer Center, Zhejiang University, Hangzhou, China,Institute of Gastroenterology, Zhejiang University, Hangzhou, China,Department of Gastroenterology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Mengjie Li
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China,Cancer Center, Zhejiang University, Hangzhou, China,Institute of Gastroenterology, Zhejiang University, Hangzhou, China
| | - Chaochao Xu
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China,Cancer Center, Zhejiang University, Hangzhou, China,Institute of Gastroenterology, Zhejiang University, Hangzhou, China
| | - Dingjiacheng Jia
- Cancer Center, Zhejiang University, Hangzhou, China,Institute of Gastroenterology, Zhejiang University, Hangzhou, China,Department of Gastroenterology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Yadong Qi
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China,Cancer Center, Zhejiang University, Hangzhou, China,Institute of Gastroenterology, Zhejiang University, Hangzhou, China
| | - Tongyao Hou
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China,Cancer Center, Zhejiang University, Hangzhou, China,Institute of Gastroenterology, Zhejiang University, Hangzhou, China
| | - Lan Wang
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China,Cancer Center, Zhejiang University, Hangzhou, China,Institute of Gastroenterology, Zhejiang University, Hangzhou, China
| | - Boya Wang
- Institute of Gastroenterology, Zhejiang University, Hangzhou, China,Department of Pharmacy, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Aiqing Li
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China,Cancer Center, Zhejiang University, Hangzhou, China,Institute of Gastroenterology, Zhejiang University, Hangzhou, China
| | - Shujie Chen
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China,Cancer Center, Zhejiang University, Hangzhou, China,Institute of Gastroenterology, Zhejiang University, Hangzhou, China,Shujie Chen Department of Gastroenterology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, Zhejiang, China
| | - Jianmin Si
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China,Cancer Center, Zhejiang University, Hangzhou, China,Institute of Gastroenterology, Zhejiang University, Hangzhou, China,Jianmin Si Department of Gastroenterology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, Zhejiang, China
| | - Wei Zhuo
- Department of Cell Biology and Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China,Cancer Center, Zhejiang University, Hangzhou, China,Institute of Gastroenterology, Zhejiang University, Hangzhou, China,CONTACT Wei Zhuo Department of Cell Biology and Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou310058, Zhejiang, China
| |
Collapse
|
35
|
Sun JL, Cao ZQ, Sun SW, Sun ZH, Sun SH, Ye JF, Leng P. Effects of 2,2',4'‑trihydroxychalcone on the proliferation, metastasis and apoptosis of A549 human lung cancer cells. Oncol Lett 2022; 23:116. [PMID: 35261630 PMCID: PMC8855167 DOI: 10.3892/ol.2022.13236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Accepted: 01/13/2022] [Indexed: 11/21/2022] Open
Abstract
The aim of the present study was to evaluate the antitumor effects of 2,2′,4′-trihydroxychalcone (7a) on the A549 human lung cancer cell line. A549 cells were treated with different concentrations of 7a for different time periods. Cells without 7a were used as the negative control group. Cell proliferation, invasion, vasculogenic mimicry (VM) formation, heterogeneous adhesion and apoptosis were measured using Cell Counting Kit-8, Transwell invasion, VM, adhesion and flow cytometric assays, respectively. In addition, the expression of related proteins was determined using western blot analysis or ELISA. The present study found that 7a had a significant inhibitory effect on the survival rate of the A549 lung cancer cells but almost no effect on BEAS-2B human lung epithelial cells or human venous endothelial cells. The migration rate, VM length, invasion rate and heterogeneous adhesion number of cells treated with 7a significantly decreased as the concentration increased, while the apoptosis rate increased. Western blot analysis showed that 7a treatment significantly increased the expression levels of E-cadherin, cleaved poly (ADP-ribose) polymerase, Bax and caspase-3 and simultaneously decreased the expression levels of metalloproteinase-2/9, Bcl-2, phosphorylated (p)-PI3K, p-AKT, p-mTOR, vascular endothelial growth factor (VEGF), E-selectin and N-cadherin. At the same time, the ELISA results showed that the level of the pro-angiogenic factor VEGF in the culture media was reduced in the presence of 7a. In addition, 7a could also reduce the nuclear NF-κB protein expression, which could inhibit the gene transcription of tumor apoptosis and metastasis-related proteins. Therefore, 7a may exert inhibitory effects on A549 cells by inhibiting cell proliferation, migration, VM formation and heterogeneous adhesion, as well as by inducing apoptosis through the suppression of the PI3K/AKT/NF-κB signaling pathway; these findings suggested that 7a may be a promising agent for the treatment of lung cancer.
Collapse
Affiliation(s)
- Jia-Lin Sun
- Department of Pharmacy, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, P.R. China
| | - Zhan-Qi Cao
- Department of Pharmacy, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, P.R. China
| | - Shi-Wei Sun
- Department of Natural Medicine and Pharmacognosy, School of Pharmacy, Qingdao University, Qingdao, Shandong 266071, P.R. China
| | - Zhong-Hua Sun
- Department of Pharmacy, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, P.R. China
| | - Shu-Hong Sun
- Department of Pharmacy, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, P.R. China
| | - Jin-Feng Ye
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, Shandong 266100, P.R. China
| | - Ping Leng
- Department of Pharmacy, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, P.R. China
| |
Collapse
|
36
|
Ceci C, Lacal PM, Graziani G. Antibody-drug conjugates: Resurgent anticancer agents with multi-targeted therapeutic potential. Pharmacol Ther 2022; 236:108106. [PMID: 34990642 DOI: 10.1016/j.pharmthera.2021.108106] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 12/23/2021] [Accepted: 12/29/2021] [Indexed: 12/18/2022]
Abstract
Antibody-drug conjugates (ADCs) constitute a relatively new group of anticancer agents, whose first appearance took place about two decades ago, but a renewed interest occurred in recent years, following the success of anti-cancer immunotherapy with monoclonal antibodies. Indeed, an ADC combines the selectivity of a monoclonal antibody with the cell killing properties of a chemotherapeutic agent (payload), joined together through an appropriate linker. The antibody moiety targets a specific cell surface antigen expressed by tumor cells and/or cells of the tumor microenvironment and acts as a carrier that delivers the cytotoxic payload within the tumor mass. Despite advantages in terms of selectivity and potency, the development of ADCs is not devoid of challenges, due to: i) low tumor selectivity when the target antigens are not exclusively expressed by cancer cells; ii) premature release of the cytotoxic drug into the bloodstream as a consequence of linker instability; iii) development of tumor resistance mechanisms to the payload. All these factors may result in lack of efficacy and/or in no safety improvement compared to unconjugated cytotoxic agents. Nevertheless, the development of antibodies engineered to remain inert until activated in the tumor (e.g., antibodies activated proteolytically after internalization or by the acidic conditions of the tumor microenvironment) together with the discovery of innovative targets and cytotoxic or immunomodulatory payloads, have allowed the design of next-generation ADCs that are expected to possess improved therapeutic properties. This review provides an overview of approved ADCs, with related advantages and limitations, and of novel targets exploited by ADCs that are presently under clinical investigation.
Collapse
Affiliation(s)
- Claudia Ceci
- Department of Systems Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| | | | - Grazia Graziani
- Department of Systems Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; IDI-IRCCS, Via Monti di Creta 104, 00167 Rome, Italy.
| |
Collapse
|
37
|
Ruan Y, Chen L, Xie D, Luo T, Xu Y, Ye T, Chen X, Feng X, Wu X. Mechanisms of Cell Adhesion Molecules in Endocrine-Related Cancers: A Concise Outlook. Front Endocrinol (Lausanne) 2022; 13:865436. [PMID: 35464064 PMCID: PMC9021432 DOI: 10.3389/fendo.2022.865436] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Accepted: 03/09/2022] [Indexed: 11/13/2022] Open
Abstract
Chemotherapy is a critical treatment for endocrine-related cancers; however, chemoresistance and disease recurrence remain a challenge. The interplay between cancer cells and the tumor microenvironment via cell adhesion molecules (CAMs) promotes drug resistance, known as cell adhesion-mediated drug resistance (CAM-DR). CAMs are cell surface molecules that facilitate cell-to-cell or cell-to-extracellular matrix binding. CAMs exert an adhesion effect and trigger intracellular signaling that regulates cancer cell stemness maintenance, survival, proliferation, metastasis, epithelial-mesenchymal transition, and drug resistance. To understand these mechanisms, this review focuses on the role of CD44, cadherins, selectins, and integrins in CAM-DR in endocrine-related cancers.
Collapse
Affiliation(s)
- Yongsheng Ruan
- Department of Pediatrics, Nanfang Hospital, Southern Medical University, Guangzhou, China
- *Correspondence: Yongsheng Ruan, ; Xuedong Wu,
| | - Libai Chen
- Department of Pediatrics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Danfeng Xie
- Department of Pediatrics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Tingting Luo
- Department of Pediatrics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yiqi Xu
- Department of Pediatrics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Tao Ye
- Department of Endocrinology, Affiliated Baoan Hospital of Shenzhen, Southern Medical University, Shenzhen, China
| | - Xiaona Chen
- Department of Pediatrics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaoqin Feng
- Department of Pediatrics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xuedong Wu
- Department of Pediatrics, Nanfang Hospital, Southern Medical University, Guangzhou, China
- *Correspondence: Yongsheng Ruan, ; Xuedong Wu,
| |
Collapse
|
38
|
Guo K, Xiao N, Liu Y, Wang Z, Tóth J, Gyenis J, Thakur VK, Oyane A, Shubhra QT. Engineering polymer nanoparticles using cell membrane coating technology and their application in cancer treatments: Opportunities and challenges. NANO MATERIALS SCIENCE 2021. [DOI: 10.1016/j.nanoms.2021.12.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
39
|
Mackie GM, Copland A, Takahashi M, Nakanishi Y, Everard I, Kato T, Oda H, Kanaya T, Ohno H, Maslowski KM. Bacterial cancer therapy in autochthonous colorectal cancer affects tumor growth and metabolic landscape. JCI Insight 2021; 6:e139900. [PMID: 34710062 PMCID: PMC8675204 DOI: 10.1172/jci.insight.139900] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 10/27/2021] [Indexed: 12/21/2022] Open
Abstract
Bacterial cancer therapy (BCT) shows great promise for treatment of solid tumors, yet basic mechanisms of bacterial-induced tumor suppression remain undefined. Attenuated strains of Salmonella enterica serovar Typhimurium (STm) have commonly been used in mouse models of BCT in xenograft and orthotopic transplant cancer models. We aimed to better understand the tumor epithelium-targeted mechanisms of BCT by using autochthonous mouse models of intestinal cancer and tumor organoid cultures to assess the effectiveness and consequences of oral treatment with aromatase A-deficient STm (STmΔaroA). STmΔaroA delivered by oral gavage significantly reduced tumor burden and tumor load in both a colitis-associated colorectal cancer (CAC) model and in a spontaneous Apcmin/+ intestinal cancer model. STmΔaroA colonization of tumors caused alterations in transcription of mRNAs associated with tumor stemness, epithelial-mesenchymal transition, and cell cycle. Metabolomic analysis of tumors demonstrated alteration in the metabolic environment of STmΔaroA-treated tumors, suggesting that STmΔaroA imposes metabolic competition on the tumor. Use of tumor organoid cultures in vitro recapitulated effects seen on tumor stemness, mesenchymal markers, and altered metabolome. Furthermore, live STmΔaroA was required, demonstrating active mechanisms including metabolite usage. We have demonstrated that oral BCT is efficacious in autochthonous intestinal cancer models, that BCT imposes metabolic competition, and that BCT has direct effects on the tumor epithelium affecting tumor stem cells.
Collapse
Affiliation(s)
- Gillian M. Mackie
- University of Birmingham, Institute of Immunology and Immunotherapy and Institute of Metabolism and Systems Research, Birmingham, United Kingdom
| | - Alastair Copland
- University of Birmingham, Institute of Immunology and Immunotherapy and Institute of Metabolism and Systems Research, Birmingham, United Kingdom
| | - Masumi Takahashi
- Laboratory for Intestinal Ecosystem, RIKEN Institute for Integrative Medical Science, Yokohama, Japan
| | - Yumiko Nakanishi
- Laboratory for Intestinal Ecosystem, RIKEN Institute for Integrative Medical Science, Yokohama, Japan
- Intestinal Microbiota Project, Kanagawa Institute of Industrial Science and Technology, Kawasaki, Japan
- Immunobiology Laboratory, Graduate School of Medical Life Science, Yokohama City University, Yokohama, Japan
| | - Isabel Everard
- University of Birmingham, Institute of Immunology and Immunotherapy and Institute of Metabolism and Systems Research, Birmingham, United Kingdom
| | - Tamotsu Kato
- Laboratory for Intestinal Ecosystem, RIKEN Institute for Integrative Medical Science, Yokohama, Japan
- Intestinal Microbiota Project, Kanagawa Institute of Industrial Science and Technology, Kawasaki, Japan
- Immunobiology Laboratory, Graduate School of Medical Life Science, Yokohama City University, Yokohama, Japan
| | - Hirotsugu Oda
- Laboratory for Integrative Genomics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Inflammatory Disease Section, National Human Genome Research Institute, NIH, Bethesda, Maryland, USA
| | - Takashi Kanaya
- Laboratory for Intestinal Ecosystem, RIKEN Institute for Integrative Medical Science, Yokohama, Japan
- Immunobiology Laboratory, Graduate School of Medical Life Science, Yokohama City University, Yokohama, Japan
| | - Hiroshi Ohno
- Laboratory for Intestinal Ecosystem, RIKEN Institute for Integrative Medical Science, Yokohama, Japan
- Intestinal Microbiota Project, Kanagawa Institute of Industrial Science and Technology, Kawasaki, Japan
- Immunobiology Laboratory, Graduate School of Medical Life Science, Yokohama City University, Yokohama, Japan
| | - Kendle M. Maslowski
- University of Birmingham, Institute of Immunology and Immunotherapy and Institute of Metabolism and Systems Research, Birmingham, United Kingdom
- Laboratory for Intestinal Ecosystem, RIKEN Institute for Integrative Medical Science, Yokohama, Japan
| |
Collapse
|
40
|
Rose M, Cardon T, Aboulouard S, Hajjaji N, Kobeissy F, Duhamel M, Fournier I, Salzet M. Surfaceome Proteomic of Glioblastoma Revealed Potential Targets for Immunotherapy. Front Immunol 2021; 12:746168. [PMID: 34646273 PMCID: PMC8503648 DOI: 10.3389/fimmu.2021.746168] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 09/08/2021] [Indexed: 12/21/2022] Open
Abstract
Glioblastoma (GBM) is the most common and devastating malignant brain tumor in adults. The mortality rate is very high despite different treatments. New therapeutic targets are therefore highly needed. Cell-surface proteins represent attractive targets due to their accessibility, their involvement in essential signaling pathways, and their dysregulated expression in cancer. Moreover, they are potential targets for CAR-based immunotherapy or mRNA vaccine strategies. In this context, we investigated the GBM-associated surfaceome by comparing it to astrocytes cell line surfaceome to identify new specific targets for GBM. For this purpose, biotinylation of cell surface proteins has been carried out in GBM and astrocytes cell lines. Biotinylated proteins were purified on streptavidin beads and analyzed by shotgun proteomics. Cell surface proteins were identified with Cell Surface Proteins Atlas (CSPA) and Gene Ontology enrichment. Among all the surface proteins identified in the different cell lines we have confirmed the expression of 66 of these in patient’s glioblastoma using spatial proteomic guided by MALDI-mass spectrometry. Moreover, 87 surface proteins overexpressed or exclusive in GBM cell lines have been identified. Among these, we found 11 specific potential targets for GBM including 5 mutated proteins such as RELL1, CYBA, EGFR, and MHC I proteins. Matching with drugs and clinical trials databases revealed that 7 proteins were druggable and under evaluation, 3 proteins have no known drug interaction yet and none of them are the mutated form of the identified proteins. Taken together, we discovered potential targets for immune therapy strategies in GBM.
Collapse
Affiliation(s)
- Mélanie Rose
- Université Lille, Inserm, CHU Lille, U1192, Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse (PRISM), Lille, France
| | - Tristan Cardon
- Université Lille, Inserm, CHU Lille, U1192, Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse (PRISM), Lille, France
| | - Soulaimane Aboulouard
- Université Lille, Inserm, CHU Lille, U1192, Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse (PRISM), Lille, France
| | - Nawale Hajjaji
- Université Lille, Inserm, CHU Lille, U1192, Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse (PRISM), Lille, France.,Breast Cancer Unit, Oscar Lambret Center, Lille, France
| | - Firas Kobeissy
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Marie Duhamel
- Université Lille, Inserm, CHU Lille, U1192, Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse (PRISM), Lille, France
| | - Isabelle Fournier
- Université Lille, Inserm, CHU Lille, U1192, Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse (PRISM), Lille, France.,Institut Universitaire de France, Paris, France
| | - Michel Salzet
- Université Lille, Inserm, CHU Lille, U1192, Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse (PRISM), Lille, France.,Institut Universitaire de France, Paris, France
| |
Collapse
|
41
|
Gao J, Wang Y, Lyu B, Chen J, Chen G. Component Identification of Phenolic Acids in Cell Suspension Cultures of Saussureainvolucrata and Its Mechanism of Anti-Hepatoma Revealed by TMT Quantitative Proteomics. Foods 2021; 10:foods10102466. [PMID: 34681515 PMCID: PMC8535732 DOI: 10.3390/foods10102466] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 10/11/2021] [Accepted: 10/12/2021] [Indexed: 12/13/2022] Open
Abstract
Saussurea involucrata (S. involucrata) had been reported to have anti-hepatoma function. However, the mechanism is complex and unclear. To evaluate the anti-hepatoma mechanism of S. involucrata comprehensively and make a theoretical basis for the mechanical verification of later research, we carried out this work. In this study, the total phenolic acids from S. involucrata determined by a cell suspension culture (ESPI) was mainly composed of 4,5-dicaffeoylquinic acid, according to the LC-MS analysis. BALB/c nude female mice were injected with HepG2 cells to establish an animal model of liver tumor before being divided into a control group, a low-dose group, a middle-dose group, a high-dose group, and a DDP group. Subsequently, EPSI was used as the intervention drug for mice. Biochemical indicators and differences in protein expression determined by TMT quantitative proteomics were used to resolve the mechanism after the low- (100 mg/kg), middle- (200 mg/kg), and high-dose (400 mg/kg) interventions for 24 days. The results showed that EPSI can not only limit the growth of HepG2 cells in vitro, but also can inhibit liver tumors significantly with no toxicity at high doses in vivo. Proteomics analysis revealed that the upregulated differentially expressed proteins (DE proteins) in the high-dose group were over three times that in the control group. ESPI affected the pathways significantly associated with the protein metabolic process, metabolic process, catalytic activity, hydrolase activity, proteolysis, endopeptidase activity, serine-type endopeptidase activity, etc. The treatment group showed significant differences in the pathways associated with the renin-angiotensin system, hematopoietic cell lineage, etc. In conclusion, ESPI has a significant anti-hepatoma effect and the potential mechanism was revealed.
Collapse
Affiliation(s)
- Junpeng Gao
- College of Life Science, Jilin Agricultural University, Changchun 130118, China;
| | - Yi Wang
- College of Food Science and Engineering, Jilin Agricultural University, Changchun 130118, China; (Y.W.); (B.L.); (J.C.)
| | - Bo Lyu
- College of Food Science and Engineering, Jilin Agricultural University, Changchun 130118, China; (Y.W.); (B.L.); (J.C.)
- College of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Jian Chen
- College of Food Science and Engineering, Jilin Agricultural University, Changchun 130118, China; (Y.W.); (B.L.); (J.C.)
| | - Guang Chen
- College of Life Science, Jilin Agricultural University, Changchun 130118, China;
- Correspondence:
| |
Collapse
|
42
|
Association of VLA4, 5, 6 and PSGL1 expression levels with engraftment in autologous HPSC transplantation in multiple myeloma patients. Transfus Apher Sci 2021; 60:103285. [PMID: 34620563 DOI: 10.1016/j.transci.2021.103285] [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: 05/24/2021] [Revised: 09/12/2021] [Accepted: 09/21/2021] [Indexed: 11/21/2022]
Abstract
The most promising therapy for leukemia is hematopoietic stem cell transplantation. Engraftment of HPSCs mainly depends on some factors such as adhesion molecules, including VLAs. This study tried to delineate the relationship between HPSCs engraftment and expression level of PSGL1 and VLA4, 5, and 6 genes in candidate MM patients for autologous bone marrow transplantation. Firstly, the CD 34+ HPSCs were collected from multiple myeloma (MM) patients after five days of G-CSF therapy through apheresis processes. Then, the patients were categorized into two groups of good and bad prognosis depending on engraftment time (Less or more than 18 days). Followingly, the expression of PSGL1 and VLA4, VLA5, and VLA6 genes were assessed by the qRT-PCR technique in each patient. Finally, the correlation between the genes and engraftment time was investigated to determine the prognostic role of each gene on HPSCs transplantation. Our findings demonstrated that there is a significant correlation between VLA4 (P=< 0.0001) and 5 (P = 0.005) levels and HPSCs engraftment time. As the higher levels of VLA4 and 5, the shorter time HPSCs engraftment occurs. In contrast, there was no significant correlation between VLA6 (P = 0.2) and PSGL1 (P = 0.3) genes levels and engraftment time. So that, the patients with a good prognosis had a higher level of VLA4 and VLA5, but no relation was found between VLA6 and PSGL1. It is concluded that VLA4 and VLA5 expression could be considered a significant prognostic factor for HPSC transplantation.
Collapse
|
43
|
Xu R, Li S, Li S, Wong EM, Southey MC, Hopper JL, Abramson MJ, Guo Y. Ambient temperature and genome-wide DNA methylation: A twin and family study in Australia. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2021; 285:117700. [PMID: 34380236 DOI: 10.1016/j.envpol.2021.117700] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 06/29/2021] [Accepted: 06/30/2021] [Indexed: 06/13/2023]
Abstract
Little is known about the association between ambient temperature and DNA methylation, which is a potential biological process through which ambient temperature affects health. This study aimed to evaluate the association between ambient temperature and DNA methylation across human genome. We included 479 Australian women, including 132 twin pairs and 215 sisters of these twins. Blood-derived DNA methylation was measured using the HumanMethylation450 BeadChip array. Data on average ambient temperature during eight different exposure windows [lag0d (the blood draw day), lag0-7d (the current day and previous seven days prior to blood draw), lag0-14d, lag0-21d, lag0-28d, lag0-90d, lag0-180d, and lag0-365d)] was linked to each participant's home address. For each cytosine-guanine dinucleotide (CpG), we evaluated the association between its methylation level and temperature using generalized estimating equations (GEE), adjusting for important covariates. We used comb-p and DMRcate to identify differentially methylated regions (DMRs). We identified 31 CpGs at which blood DNA methylation were significantly associated with ambient temperature with false discovery rate [FDR] < 0.05. There were 82 significant DMRs identified by both comb-p (Sidak p-value < 0.01) and DMRcate (FDR < 0.01). Most of these CpGs and DMRs only showed association with temperature during one specific exposure window. These CpGs and DMRs were mapped to 85 genes. These related genes have been related to many human chronic diseases or phenotypes (e.g., diabetes, arthritis, breast cancer, depression, asthma, body height) in previous studies. The signals of short-term windows (lag0d and lag0-21d) showed enrichment in biological processes related to cell adhesion. In conclusion, short-, medium-, and long-term exposures to ambient temperature were all associated with blood DNA methylation, but the target genomic loci varied by exposure window. These differential methylation signals may serve as potential biomarkers to understand the health impacts of temperature.
Collapse
Affiliation(s)
- Rongbin Xu
- School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC, 3004, Australia
| | - Shuai Li
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, VIC, 3010, Australia; Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, CB1 8RN, UK; Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, Clayton, VIC, 3800, Australia
| | - Shanshan Li
- School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC, 3004, Australia
| | - Ee Ming Wong
- Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, Clayton, VIC, 3800, Australia; Department of Clinical Pathology, Melbourne Medical School, The University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Melissa C Southey
- Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, Clayton, VIC, 3800, Australia; Department of Clinical Pathology, Melbourne Medical School, The University of Melbourne, Melbourne, VIC, 3010, Australia; Cancer Epidemiology Division, Cancer Council Victoria, VIC, 3004, Australia
| | - John L Hopper
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Michael J Abramson
- School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC, 3004, Australia
| | - Yuming Guo
- School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC, 3004, Australia.
| |
Collapse
|
44
|
Sang C, Song Y, Jin TW, Zhang S, Fu L, Zhao Y, Zou X, Wang Z, Gao H, Liu S. Bisphenol A induces ovarian cancer cell proliferation and metastasis through estrogen receptor-α pathways. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2021; 28:36060-36068. [PMID: 33683587 DOI: 10.1007/s11356-021-13267-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 03/01/2021] [Indexed: 06/12/2023]
Abstract
Bisphenol A (BPA) is a widely used raw material that can be detected both in the environment and in the human body. Due to its estrogen-like effects, wide concerns have been raised about the potential role of BPA in the initiation and development of hormone-dependent cancers. Ovarian cancer is the most common reproductive system cancer and has a high mortality rate in women. Despite recent investigations into BPA's carcinogenic effects, studies on its role in ovarian cancer development remain limited. In this study, we aimed to assess the effect of BPA at various environmentally relevant concentrations on proliferation and metastasis of ovarian cancer cells. We discovered that BPA can stimulate proliferation of OVCAR-3 ovarian cancer cells after exposure for up to 5 days. Strikingly, BPA enhanced ovarian cancer cell migration, invasion, and adhesion (to vascular endothelial cells) through upregulation of matrix metalloproteinase-2 (MMP-2), MMP-9, and intercellular cell adhesion molecule-1 (IMAC-1). The stimulatory effects of BPA on cancer cell proliferation and metastasis were reversed by treatment with an ERα inhibitor, but not by treatment with an ERβ inhibitor. Together, these results suggest that BPA induces proliferation and metastasis of ovarian cancer cells through ERα signaling pathways. This study provides new insights into the carcinogenic effects of BPA with regard to ovarian cancer.
Collapse
Affiliation(s)
- Chen Sang
- School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China
| | - Yu Song
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, Heilongjiang Province, China
| | - Tong-Wang Jin
- School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China
| | - Shuo Zhang
- Department of Obstetrics & Gynecology, Beijing Tian Tan Hospital, Capital Medical University, Beijing, 100730, China
| | - Linyan Fu
- Department of Obstetrics & Gynecology, Beijing Tian Tan Hospital, Capital Medical University, Beijing, 100730, China
| | - Yi Zhao
- Department of Obstetrics & Gynecology, Beijing Tian Tan Hospital, Capital Medical University, Beijing, 100730, China
| | - Xinxin Zou
- Department of Obstetrics & Gynecology, Beijing Tian Tan Hospital, Capital Medical University, Beijing, 100730, China
| | - Zhe Wang
- School of Public Health, Xinxiang Medical University, Xinxiang, 453003, Henan Province, China.
| | - Hui Gao
- Department of Obstetrics & Gynecology, Beijing Tian Tan Hospital, Capital Medical University, Beijing, 100730, China.
| | - Sijin Liu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environment Sciences, Chinese Academy of Sciences, Beijing, 100085, China
| |
Collapse
|
45
|
Targeting Telomere Biology in Acute Lymphoblastic Leukemia. Int J Mol Sci 2021; 22:ijms22136653. [PMID: 34206297 PMCID: PMC8268026 DOI: 10.3390/ijms22136653] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 06/02/2021] [Accepted: 06/14/2021] [Indexed: 01/19/2023] Open
Abstract
Increased cell proliferation is a hallmark of acute lymphoblastic leukemia (ALL), and genetic alterations driving clonal proliferation have been identified as prognostic factors. To evaluate replicative history and its potential prognostic value, we determined telomere length (TL) in lymphoblasts, B-, and T-lymphocytes, and measured telomerase activity (TA) in leukocytes of patients with ALL. In addition, we evaluated the potential to suppress the in vitro growth of B-ALL cells by the telomerase inhibitor imetelstat. We found a significantly lower TL in lymphoblasts (4.3 kb in pediatric and 2.3 kb in adult patients with ALL) compared to B- and T-lymphocytes (8.0 kb and 8.2 kb in pediatric, and 6.4 kb and 5.5 kb in adult patients with ALL). TA in leukocytes was 3.2 TA/C for pediatric and 0.7 TA/C for adult patients. Notably, patients with high-risk pediatric ALL had a significantly higher TA of 6.6 TA/C compared to non-high-risk patients with 2.2 TA/C. The inhibition of telomerase with imetelstat ex vivo led to significant dose-dependent apoptosis of B-ALL cells. These results suggest that TL reflects clonal expansion and indicate that elevated TA correlates with high-risk pediatric ALL. In addition, telomerase inhibition induces apoptosis of B-ALL cells cultured in vitro. TL and TA might complement established markers for the identification of patients with high-risk ALL. Moreover, TA seems to be an effective therapeutic target; hence, telomerase inhibitors, such as imetelstat, may augment standard ALL treatment.
Collapse
|
46
|
Vecchi L, Araújo TG, Azevedo FVPDV, Mota STS, Ávila VDMR, Ribeiro MA, Goulart LR. Phospholipase A 2 Drives Tumorigenesis and Cancer Aggressiveness through Its Interaction with Annexin A1. Cells 2021; 10:cells10061472. [PMID: 34208346 PMCID: PMC8231270 DOI: 10.3390/cells10061472] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 06/03/2021] [Accepted: 06/08/2021] [Indexed: 12/14/2022] Open
Abstract
Phospholipids are suggested to drive tumorigenesis through their essential role in inflammation. Phospholipase A2 (PLA2) is a phospholipid metabolizing enzyme that releases free fatty acids, mostly arachidonic acid, and lysophospholipids, which contribute to the development of the tumor microenvironment (TME), promoting immune evasion, angiogenesis, tumor growth, and invasiveness. The mechanisms mediated by PLA2 are not fully understood, especially because an important inhibitory molecule, Annexin A1, is present in the TME but does not exert its action. Here, we will discuss how Annexin A1 in cancer does not inhibit PLA2 leading to both pro-inflammatory and pro-tumoral signaling pathways. Moreover, Annexin A1 promotes the release of cancer-derived exosomes, which also lead to the enrichment of PLA2 and COX-1 and COX-2 enzymes, contributing to TME formation. In this review, we aim to describe the role of PLA2 in the establishment of TME, focusing on cancer-derived exosomes, and modulatory activities of Annexin A1. Unraveling how these proteins interact in the cancer context can reveal new strategies for the treatment of different tumors. We will also describe the possible strategies to inhibit PLA2 and the approaches that could be used in order to resume the anti-PLA2 function of Annexin A1.
Collapse
Affiliation(s)
- Lara Vecchi
- Laboratory of Nanobiotechnology, Federal University of Uberlandia, Uberlandia 38400-902, MG, Brazil; (L.V.); (T.G.A.); (F.V.P.d.V.A.); (S.T.S.M.)
| | - Thaise Gonçalves Araújo
- Laboratory of Nanobiotechnology, Federal University of Uberlandia, Uberlandia 38400-902, MG, Brazil; (L.V.); (T.G.A.); (F.V.P.d.V.A.); (S.T.S.M.)
- Laboratory of Genetics and Biotechnology, Federal University of Uberlandia, Patos de Minas 387400-128, MG, Brazil;
| | | | - Sara Teixeria Soares Mota
- Laboratory of Nanobiotechnology, Federal University of Uberlandia, Uberlandia 38400-902, MG, Brazil; (L.V.); (T.G.A.); (F.V.P.d.V.A.); (S.T.S.M.)
| | - Veridiana de Melo Rodrigues Ávila
- Laboratory of Biochemistry and Animal Toxins, Institute of Biotechnology, Federal University of Uberlandia, Uberlandia 38400-902, MG, Brazil;
| | - Matheus Alves Ribeiro
- Laboratory of Genetics and Biotechnology, Federal University of Uberlandia, Patos de Minas 387400-128, MG, Brazil;
| | - Luiz Ricardo Goulart
- Laboratory of Nanobiotechnology, Federal University of Uberlandia, Uberlandia 38400-902, MG, Brazil; (L.V.); (T.G.A.); (F.V.P.d.V.A.); (S.T.S.M.)
- Correspondence: ; Tel.: +55-3432258440
| |
Collapse
|
47
|
Danilova NV, Mikhailov IA, Oleynikova NA, Malkov PG. [E-cadherin expression in tumor emboli in gastric cancer]. Arkh Patol 2021; 83:11-19. [PMID: 34041891 DOI: 10.17116/patol20218303111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
OBJECTIVE To determine the level of E-cadherin expression in tumor emboli, to compare it with expression in a tumor, to determine the dependence of E-cadherin expression in tumor emboli on the clinical and morphological characteristics of gastric cancer. MATERIAL AND METHODS We used samples of surgical material from 280 patients with a verified diagnosis of gastric cancer. E-cadherin expression was determined by immunohistochemical method. The results of the reactions were assessed semi-quantitatively and compared with the main clinical and morphological characteristics of gastric cancer (histological type according to the WHO classification 2019, histological type according to the classification of P. Lauren, clinical stage, depth of invasion (T), number of metastases in lymph nodes (N), presence or/absence of distant metastases (M), tumor localization in the stomach). RESULTS Among 280 cases of cancer, emboli were detected only in 67 cases, used for further analysis. The rest of the samples were excluded from the analysis, since emboli did not get into the sections during the cutting of immunohistochemical preparations. The expression of E-cadherin in tumor emboli was significantly higher (p<0.001) than in tumor tissue. At the same time, no cases identified where the level of E-cadherin decreased in emboli compared to the tumor. A significant increase in the expression of E-cadherin in tumor emboli compared to the primary tumor was noted for all histological types according to WHO 2019, for intermediate and diffuse types according to the P. Lauren classification (p<0.001). Comparison of expression in emboli and tumors for neoplasms with different depths of invasion (T), different stages and different localizations did not reveal statistically significant differences. An increase in the expression of E-cadherin in emboli compared to tumors was characterized by a higher level of significance in the presence of metastases (N1, N2, N3a, N3b; p<0.001) than in the absence of metastases (N0; p=0.016). CONCLUSION The study revealed a statistically significant increase in the expression of E-cadherin in tumor emboli compared to the primary tumor, which is evidence of its important role in maintaining the integrity of emboli and tumor dissemination.
Collapse
Affiliation(s)
- N V Danilova
- Lomonosov Moscow State University, Moscow, Russia
| | | | | | - P G Malkov
- Lomonosov Moscow State University, Moscow, Russia.,Russian Medical Academy for Continuous Professional Education, Moscow, Russia
| |
Collapse
|
48
|
Shrestha RL, Rossi A, Wangsa D, Hogan AK, Zaldana KS, Suva E, Chung YJ, Sanders CL, Difilippantonio S, Karpova TS, Karim B, Foltz DR, Fachinetti D, Aplan PD, Ried T, Basrai MA. CENP-A overexpression promotes aneuploidy with karyotypic heterogeneity. J Cell Biol 2021; 220:211820. [PMID: 33620383 PMCID: PMC7905998 DOI: 10.1083/jcb.202007195] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 12/15/2020] [Accepted: 01/26/2021] [Indexed: 02/07/2023] Open
Abstract
Chromosomal instability (CIN) is a hallmark of many cancers. Restricting the localization of centromeric histone H3 variant CENP-A to centromeres prevents CIN. CENP-A overexpression (OE) and mislocalization have been observed in cancers and correlate with poor prognosis; however, the molecular consequences of CENP-A OE on CIN and aneuploidy have not been defined. Here, we show that CENP-A OE leads to its mislocalization and CIN with lagging chromosomes and micronuclei in pseudodiploid DLD1 cells and xenograft mouse model. CIN is due to reduced localization of proteins to the kinetochore, resulting in defects in kinetochore integrity and unstable kinetochore–microtubule attachments. CENP-A OE contributes to reduced expression of cell adhesion genes and higher invasion of DLD1 cells. We show that CENP-A OE contributes to aneuploidy with karyotypic heterogeneity in human cells and xenograft mouse model. In summary, our results provide a molecular link between CENP-A OE and aneuploidy, and suggest that karyotypic heterogeneity may contribute to the aggressive phenotype of CENP-A–overexpressing cancers.
Collapse
Affiliation(s)
- Roshan L Shrestha
- Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Austin Rossi
- Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Darawalee Wangsa
- Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Ann K Hogan
- Department of Biochemistry and Molecular Genetics, Northwestern University, Chicago, IL
| | - Kimberly S Zaldana
- Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Evelyn Suva
- Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Yang Jo Chung
- Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Chelsea L Sanders
- Laboratory Animal Sciences Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, MD
| | - Simone Difilippantonio
- Laboratory Animal Sciences Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, MD
| | - Tatiana S Karpova
- Laboratory of Receptor Biology and Gene Expression, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Baktiar Karim
- Laboratory Animal Sciences Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, MD
| | - Daniel R Foltz
- Department of Biochemistry and Molecular Genetics, Northwestern University, Chicago, IL
| | - Daniele Fachinetti
- Institut Curie, PSL Research University, Centre National de la Recherche Scientifique, Unité Mixte de Recherche 144, Paris, France
| | - Peter D Aplan
- Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Thomas Ried
- Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Munira A Basrai
- Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| |
Collapse
|
49
|
Barad M, Csukasi F, Bosakova M, Martin JH, Zhang W, Paige Taylor S, Lachman RS, Zieba J, Bamshad M, Nickerson D, Chong JX, Cohn DH, Krejci P, Krakow D, Duran I. Biallelic mutations in LAMA5 disrupts a skeletal noncanonical focal adhesion pathway and produces a distinct bent bone dysplasia. EBioMedicine 2020; 62:103075. [PMID: 33242826 PMCID: PMC7695969 DOI: 10.1016/j.ebiom.2020.103075] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 09/30/2020] [Accepted: 10/01/2020] [Indexed: 12/13/2022] Open
Abstract
Background Beyond its structural role in the skeleton, the extracellular matrix (ECM), particularly basement membrane proteins, facilitates communication with intracellular signaling pathways and cell to cell interactions to control differentiation, proliferation, migration and survival. Alterations in extracellular proteins cause a number of skeletal disorders, yet the consequences of an abnormal ECM on cellular communication remains less well understood Methods Clinical and radiographic examinations defined the phenotype in this unappreciated bent bone skeletal disorder. Exome analysis identified the genetic alteration, confirmed by Sanger sequencing. Quantitative PCR, western blot analyses, immunohistochemistry, luciferase assay for WNT signaling were employed to determine RNA, proteins levels and localization, and dissect out the underlying cell signaling abnormalities. Migration and wound healing assays examined cell migration properties. Findings This bent bone dysplasia resulted from biallelic mutations in LAMA5, the gene encoding the alpha-5 laminin basement membrane protein. This finding uncovered a mechanism of disease driven by ECM-cell interactions between alpha-5-containing laminins, and integrin-mediated focal adhesion signaling, particularly in cartilage. Loss of LAMA5 altered β1 integrin signaling through the non-canonical kinase PYK2 and the skeletal enriched SRC kinase, FYN. Loss of LAMA5 negatively impacted the actin cytoskeleton, vinculin localization, and WNT signaling. Interpretation This newly described mechanism revealed a LAMA5-β1 Integrin-PYK2-FYN focal adhesion complex that regulates skeletogenesis, impacted WNT signaling and, when dysregulated, produced a distinct skeletal disorder. Funding Supported by NIH awards R01 AR066124, R01 DE019567, R01 HD070394, and U54HG006493, and Czech Republic grants INTER-ACTION LTAUSA19030, V18-08-00567 and GA19-20123S.
Collapse
Affiliation(s)
- Maya Barad
- Department of Orthopaedic Surgery, University of California-Los Angeles, 615 Charles E. Young Drive South, BSRB 512, Los Angeles, CA 90095, United States
| | - Fabiana Csukasi
- Department of Orthopaedic Surgery, University of California-Los Angeles, 615 Charles E. Young Drive South, BSRB 512, Los Angeles, CA 90095, United States; Laboratory of Bioengineering and Tissue Regeneration-LABRET, Department of Cell Biology, Genetics and Physiology, University of Málaga, IBIMA, Málaga 29071, Spain
| | - Michaela Bosakova
- Department of Biology, Faculty of Medicine, Masaryk University, Brno 62500, Czech Republic; International Clinical Research Center, St. Anne's University Hospital, Brno 65691, Czech Republic
| | - Jorge H Martin
- Department of Orthopaedic Surgery, University of California-Los Angeles, 615 Charles E. Young Drive South, BSRB 512, Los Angeles, CA 90095, United States
| | - Wenjuan Zhang
- Department of Molecular, Cell and Developmental Biology, University of California- Los Angeles, Los Angeles, CA 90095, United States
| | - S Paige Taylor
- Department of Orthopaedic Surgery, University of California-Los Angeles, 615 Charles E. Young Drive South, BSRB 512, Los Angeles, CA 90095, United States
| | - Ralph S Lachman
- International Skeletal Dysplasia Registry, University of California, Los Angeles, CA 90095 United States
| | - Jennifer Zieba
- Department of Orthopaedic Surgery, University of California-Los Angeles, 615 Charles E. Young Drive South, BSRB 512, Los Angeles, CA 90095, United States
| | - Michael Bamshad
- University of Washington Center for Mendelian Genomics, University of Washington, Seattle, WA 98195 United States
| | - Deborah Nickerson
- University of Washington Center for Mendelian Genomics, University of Washington, Seattle, WA 98195 United States
| | - Jessica X Chong
- University of Washington Center for Mendelian Genomics, University of Washington, Seattle, WA 98195 United States
| | - Daniel H Cohn
- Department of Orthopaedic Surgery, University of California-Los Angeles, 615 Charles E. Young Drive South, BSRB 512, Los Angeles, CA 90095, United States; Department of Molecular, Cell and Developmental Biology, University of California- Los Angeles, Los Angeles, CA 90095, United States; Orthopaedic Institute for Children, University of California-Los Angeles, Los Angeles, CA 90095, United States
| | - Pavel Krejci
- Department of Biology, Faculty of Medicine, Masaryk University, Brno 62500, Czech Republic; International Clinical Research Center, St. Anne's University Hospital, Brno 65691, Czech Republic
| | - Deborah Krakow
- Department of Orthopaedic Surgery, University of California-Los Angeles, 615 Charles E. Young Drive South, BSRB 512, Los Angeles, CA 90095, United States; International Skeletal Dysplasia Registry, University of California, Los Angeles, CA 90095 United States; Orthopaedic Institute for Children, University of California-Los Angeles, Los Angeles, CA 90095, United States; Department of Human Genetics, University of California-Los Angeles, Los Angeles, CA 90095, United States; Department of Obstetrics and Gynecology, University of California-Los Angeles, Los Angeles, CA 90095, United States.
| | - Ivan Duran
- Department of Orthopaedic Surgery, University of California-Los Angeles, 615 Charles E. Young Drive South, BSRB 512, Los Angeles, CA 90095, United States; Laboratory of Bioengineering and Tissue Regeneration-LABRET, Department of Cell Biology, Genetics and Physiology, University of Málaga, IBIMA, Málaga 29071, Spain; Networking Biomedical Research Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Andalusian Centre for Nanomedicine and Biotechnology-BIONAND, Severo Ochoa 35, Málaga 29590, Spain
| |
Collapse
|
50
|
Liu JF, Chen PC, Chang TM, Hou CH. Monocyte Chemoattractant Protein-1 promotes cancer cell migration via c-Raf/MAPK/AP-1 pathway and MMP-9 production in osteosarcoma. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2020; 39:254. [PMID: 33228783 PMCID: PMC7684958 DOI: 10.1186/s13046-020-01756-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 10/30/2020] [Indexed: 01/07/2023]
Abstract
Background Osteosarcoma is generally reported among younger individuals and has a very poor prognosis, particularly for the development of metastasis. However, more effective metastatic biomarkers and therapeutic methods are absent. Monocyte chemoattractant protein-1 (MCP-1) is involved in cancer progression and inflammatory recruitment. Although previous studies have reported higher serum MCP-1 levels in patients with osteosarcoma, the role of MCP-1 in osteosarcoma progression remains to be addressed. Methods The osteosarcoma cell migratory ability was assessed by transwell migration assay. The MCP-1 and MMP-9 expression levels were analyzed by Western blot and qPCR. The signal activation was conducted by Western blot. The in vivo mouse experiment and tumor tissue array were performed to confirm our findings in vitro. Results The present study demonstrates that MCP-1 regulates cell mobility through matrix metalloproteinase (MMP)-9 expression in osteosarcoma cells. Moreover, MCP-1 promotes MMP-9 expression, cell migration, and cell invasion by mediating CCR2, c-Raf, MAPK, and AP-1 signal transduction. Using MCP-1 knockdown stable cell lines, we found that MCP-1 knockdown reduces MMP-9 expression and cell mobility. Finally, we found high MCP-1 expression levels in osteosarcoma specimens. Conclusions Our results provide prognostic value of MCP-1 in osteosarcoma by promoting MMP-9 expression. Supplementary Information The online version contains supplementary material available at 10.1186/s13046-020-01756-y.
Collapse
Affiliation(s)
- Ju-Fang Liu
- School of Oral Hygiene, College of Oral Medicine, Taipei Medical University, Taipei, 110, Taiwan
| | - Po-Chun Chen
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, 40447, Taiwan.,Translational medicine center, Shin-Kong Wu Ho-Su Memorial Hospital, Taipei City, 11101, Taiwan.,Department of Biotechnology, College of Medical and Health Science, Asia University, Taichung, 41354, Taiwan
| | - Tsung-Ming Chang
- School of Medicine, Institute of Physiology, National Yang-Ming University, Taipei City, 11221, Taiwan
| | - Chun-Han Hou
- Department of Orthopedic Surgery, National Taiwan University Hospital, 100, NO. 1, Jen-Ai Road, Taipei City, 11102, Taiwan, ROC.
| |
Collapse
|