1401
|
Shi L, Hou J, Wang L, Fu H, Zhang Y, Song Y, Wang X. Regulatory roles of osteopontin in human lung cancer cell epithelial-to-mesenchymal transitions and responses. Clin Transl Med 2021; 11:e486. [PMID: 34323425 PMCID: PMC8265167 DOI: 10.1002/ctm2.486] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Revised: 06/14/2021] [Accepted: 06/16/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Lung cancer is still the main cause of death in patients with cancer, due to poor understanding of intracellular regulations. Of those, osteopontin (OPN) may induce the epithelial-to-mesenchymal transition (EMT) to promote tumor cell metastasis. The present study aims to evaluate the regulatory mechanism of internal and external OPN in the development of lung cancer. METHODS We evaluated genetic variations and different bioinformatics of genes in chromosome 4 among subtypes of lung cancer using global databases. We validated the expression of OPN and EMT-related proteins (e.g., E-cadherin, vimentin) in 208 non-small-cell lung cancer (NSCLC) tumors and the adjacent nontumorous tissues, further to explore the function of OPN in the progression of lung cancer, with a focus on a potential communication between OPN and EMT in the lung cancer. RESULTS We found that OPN might act as a target molecule in lung cancer, which is associated with lymph node metastasis, postresection recurrence/metastasis, and prognosis of patients with lung cancer. Biological behaviors and pathological responses of OPN varied among diseases, challenges, and severities. Overexpression of OPN was correlated with the existence of EMT in lung cancer tissues. Internal and external OPN plays the decisive roles in lung cancer cell movement, proliferation, and EMT formation, through the upregulation of OPN-PI3K and OPN-MEK pathways. PI3K and MEK inhibitors downregulated the process of EMT and biological behaviors of lung cancer cells, probably through altering vimentin-associated cytoskeletons. CONCLUSION OPN can be a metastasis-associated or specific biomarker for lung cancer and a potential target for antimetastatic treatment.
Collapse
Affiliation(s)
- Lin Shi
- Department of Pulmonary and Critical Care MedicineZhongshan HospitalShanghaiChina
- Shanghai Engineering Research for AI Technology for Cardiopulmonary DiseasesShanghaiChina
- Fudan University Shanghai Medical CollegeShanghaiChina
| | - Jiayun Hou
- Institute for Clinical ScienceShanghaiChina
- Shanghai Institute of Clinical BioinformaticsShanghaiChina
- Shanghai Engineering Research for AI Technology for Cardiopulmonary DiseasesShanghaiChina
- Jinshan Hospital Centre for Tumor Diagnosis and TherapyShanghaiChina
- Fudan University Shanghai Medical CollegeShanghaiChina
| | - Lin Wang
- Department of Pulmonary and Critical Care MedicineZhongshan HospitalShanghaiChina
- Shanghai Engineering Research for AI Technology for Cardiopulmonary DiseasesShanghaiChina
- Fudan University Shanghai Medical CollegeShanghaiChina
| | - Huirong Fu
- Institute for Clinical ScienceShanghaiChina
- Shanghai Institute of Clinical BioinformaticsShanghaiChina
- Jinshan Hospital Centre for Tumor Diagnosis and TherapyShanghaiChina
- Fudan University Shanghai Medical CollegeShanghaiChina
| | - Yiwen Zhang
- Institute for Clinical ScienceShanghaiChina
- Shanghai Institute of Clinical BioinformaticsShanghaiChina
- Shanghai Engineering Research for AI Technology for Cardiopulmonary DiseasesShanghaiChina
- Jinshan Hospital Centre for Tumor Diagnosis and TherapyShanghaiChina
- Fudan University Shanghai Medical CollegeShanghaiChina
| | - Yuanlin Song
- Department of Pulmonary and Critical Care MedicineZhongshan HospitalShanghaiChina
- Shanghai Engineering Research for AI Technology for Cardiopulmonary DiseasesShanghaiChina
- Fudan University Shanghai Medical CollegeShanghaiChina
| | - Xiangdong Wang
- Department of Pulmonary and Critical Care MedicineZhongshan HospitalShanghaiChina
- Institute for Clinical ScienceShanghaiChina
- Shanghai Institute of Clinical BioinformaticsShanghaiChina
- Shanghai Engineering Research for AI Technology for Cardiopulmonary DiseasesShanghaiChina
- Jinshan Hospital Centre for Tumor Diagnosis and TherapyShanghaiChina
- Fudan University Shanghai Medical CollegeShanghaiChina
| |
Collapse
|
1402
|
Rozova VS, Anwer AG, Guller AE, Es HA, Khabir Z, Sokolova AI, Gavrilov MU, Goldys EM, Warkiani ME, Thiery JP, Zvyagin AV. Machine learning reveals mesenchymal breast carcinoma cell adaptation in response to matrix stiffness. PLoS Comput Biol 2021; 17:e1009193. [PMID: 34297718 PMCID: PMC8336795 DOI: 10.1371/journal.pcbi.1009193] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 08/04/2021] [Accepted: 06/17/2021] [Indexed: 12/31/2022] Open
Abstract
Epithelial-mesenchymal transition (EMT) and its reverse process, mesenchymal-epithelial transition (MET), are believed to play key roles in facilitating the metastatic cascade. Metastatic lesions often exhibit a similar epithelial-like state to that of the primary tumour, in particular, by forming carcinoma cell clusters via E-cadherin-mediated junctional complexes. However, the factors enabling mesenchymal-like micrometastatic cells to resume growth and reacquire an epithelial phenotype in the target organ microenvironment remain elusive. In this study, we developed a workflow using image-based cell profiling and machine learning to examine morphological, contextual and molecular states of individual breast carcinoma cells (MDA-MB-231). MDA-MB-231 heterogeneous response to the host organ microenvironment was modelled by substrates with controllable stiffness varying from 0.2kPa (soft tissues) to 64kPa (bone tissues). We identified 3 distinct morphological cell types (morphs) varying from compact round-shaped to flattened irregular-shaped cells with lamellipodia, predominantly populating 2-kPa and >16kPa substrates, respectively. These observations were accompanied by significant changes in E-cadherin and vimentin expression. Furthermore, we demonstrate that the bone-mimicking substrate (64kPa) induced multicellular cluster formation accompanied by E-cadherin cell surface localisation. MDA-MB-231 cells responded to different substrate stiffness by morphological adaptation, changes in proliferation rate and cytoskeleton markers, and cluster formation on bone-mimicking substrate. Our results suggest that the stiffest microenvironment can induce MET.
Collapse
Affiliation(s)
- Vlada S. Rozova
- ARC Centre of Excellence for Nanoscale Biophotonics, Macquarie University, Sydney, Australia
- Institute for Biology and Biomedicine, Lobachevsky State University, Nizhny Novgorod, Russia
| | - Ayad G. Anwer
- ARC Centre of Excellence for Nanoscale Biophotonics, Macquarie University, Sydney, Australia
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, Australia
| | - Anna E. Guller
- ARC Centre of Excellence for Nanoscale Biophotonics, Macquarie University, Sydney, Australia
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, Australia
- Institute for Regenerative Medicine, Sechenov University, Moscow, Russia
| | | | - Zahra Khabir
- ARC Centre of Excellence for Nanoscale Biophotonics, Macquarie University, Sydney, Australia
| | - Anastasiya I. Sokolova
- Centre of Biomedical Engineering, Sechenov University, Moscow, Russia
- Laboratory of Medical Nanotechnologies, Federal Biomedical Agency, Moscow, Russia
| | - Maxim U. Gavrilov
- Centre of Biomedical Engineering, Sechenov University, Moscow, Russia
| | - Ewa M. Goldys
- ARC Centre of Excellence for Nanoscale Biophotonics, Macquarie University, Sydney, Australia
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, Australia
| | | | - Jean Paul Thiery
- Centre of Biomedical Engineering, Sechenov University, Moscow, Russia
- Bioland Laboratory, Guangzhou Regenerative Medicine and Health, Guangdong Laboratory, Guangzhou, China
| | - Andrei V. Zvyagin
- ARC Centre of Excellence for Nanoscale Biophotonics, Macquarie University, Sydney, Australia
- Centre of Biomedical Engineering, Sechenov University, Moscow, Russia
- Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
1403
|
Treps L, Faure S, Clere N. Vasculogenic mimicry, a complex and devious process favoring tumorigenesis – Interest in making it a therapeutic target. Pharmacol Ther 2021; 223:107805. [DOI: 10.1016/j.pharmthera.2021.107805] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
1404
|
Teixeira MP, Passos EF, Haddad NF, Andrade MN, Rumjanek VM, Miranda-Alves L, de Carvalho DP, de Paiva LS. In vitro antitumoral effects of the steroid ouabain on human thyroid papillary carcinoma cell lines. ENVIRONMENTAL TOXICOLOGY 2021; 36:1338-1348. [PMID: 33760381 DOI: 10.1002/tox.23130] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 03/03/2021] [Accepted: 03/07/2021] [Indexed: 06/12/2023]
Abstract
Ouabain is a steroid described as a compound extracted from plants that is capable of binding to Na+ , K+ -ATPase, inhibiting ion transport and triggering cell signaling pathways. Due to its positive ionotropic effect, ouabain was used for more than 200 years for the treatment of cardiac dysfunctions. Numerous antitumor effects of ouabain have been described so far; however, its role on thyroid cancer is still poorly understood. Therefore, the aim of the present work was to evaluate the effect of ouabain on the biology of human papillary thyroid cancer cells. For this, three human thyroid cell lines were used: NTHY-ori, a non-tumor lineage, BCPAP and TPC-1, both derived from papillary carcinomas. Cells were cultured in the presence or absence of ouabain. Subsequently, we evaluated its effects on the viability, cell death, cell cycle, and migratory ability of these cell lines. We also investigated the impact of ouabain in IL-6/IL-6R and epithelial to mesenchymal transition markers expression. Our results indicate that ouabain (10-7 M), decreased the number of NTHY-ori, TPC-1 and BCPAP viable cells and induced cell cycle arrest after in vitro culture, but did not appear to promote cell death. In TPC-1 cells ouabain also inhibited cell migration; increased IL-6/IL-6R expression and IL-6 secretion; and diminished vimentin and SNAIL-1 expression. Collectively, our results indicate that ouabain has an antitumoral role on human papillary thyroid carcinomas in vitro. Even though additional studies are necessary, our work contributes to the discussion of the possibility of new clinical trials of ouabain.
Collapse
Affiliation(s)
- Mariana Pires Teixeira
- Laboratório de Imunorregulação, Departamento de Imunobiologia, Instituto de Biologia, Universidade Federal Fluminense, Niterói, Brazil
- Programa de Pós-Graduação em Patologia, Universidade Federal Fluminense, Niterói, Brazil
- Laboratório de Endocrinologia Experimental-LEEx, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Programa de Pós-Graduação em Endocrinologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Eliza Freitas Passos
- Laboratório de Fisiologia Endócrina Doris Rosenthal, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Natalia Ferreira Haddad
- Programa de Pós-Graduação em Endocrinologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Laboratório de Fisiologia Endócrina Doris Rosenthal, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marcelle Novaes Andrade
- Laboratório de Endocrinologia Experimental-LEEx, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Programa de Pós-graduação em Farmacologia e Química Medicinal, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Vivian Mary Rumjanek
- Laboratório de Imunologia Tumoral, Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Leandro Miranda-Alves
- Laboratório de Endocrinologia Experimental-LEEx, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Programa de Pós-Graduação em Endocrinologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Programa de Pós-graduação em Farmacologia e Química Medicinal, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Denise Pires de Carvalho
- Programa de Pós-Graduação em Endocrinologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Laboratório de Fisiologia Endócrina Doris Rosenthal, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Luciana Souza de Paiva
- Laboratório de Imunorregulação, Departamento de Imunobiologia, Instituto de Biologia, Universidade Federal Fluminense, Niterói, Brazil
- Programa de Pós-Graduação em Patologia, Universidade Federal Fluminense, Niterói, Brazil
| |
Collapse
|
1405
|
Liu X, Ma R, Yi B, Riker AI, Xi Y. MicroRNAs are involved in the development and progression of gastric cancer. Acta Pharmacol Sin 2021; 42:1018-1026. [PMID: 33037405 PMCID: PMC8208993 DOI: 10.1038/s41401-020-00540-0] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 09/14/2020] [Indexed: 02/08/2023]
Abstract
MicroRNAs (miRNAs) are recognized as an essential component of the RNA family, exerting multiple and intricate biological functions, particularly in the process of tumorigenesis, proliferation, and metastatic progression. MiRNAs are altered in gastric cancer (GC), showing activity as both tumor suppressors and oncogenes, although their true roles have not been fully understood. This review will focus upon the recent advances of miRNA studies related to the regulatory mechanisms of gastric tumor cell proliferation, apoptosis, and cell cycle. We hope to provide an in-depth insight into the mechanistic role of miRNAs in GC development and progression. In particular, we summarize the latest studies relevant to miRNAs' impact upon the epithelial-mesenchymal transition, tumor microenvironment, and chemoresistance in GC cells. We expect to elucidate the molecular mechanisms involving miRNAs for better understanding the etiology of GC, and facilitating the development of new treatment regimens for the treatment of GC.
Collapse
Affiliation(s)
- Xiaolin Liu
- Department of Genetics and Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center, New Orleans, LA, USA
- Department of Oncology, the First Affiliated Hospital of Shandong First Medical University, Jinan, 250014, China
| | - Ruixia Ma
- Department of Genetics and Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center, New Orleans, LA, USA
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, 221000, China
| | - Bin Yi
- Department of Genetics and Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - Adam I Riker
- Geaton and JoAnn DeCesaris Cancer Institute, Department of Surgery, Anne Arundel Medical Center, Cancer Service Line, Luminis Health, Annapolis, MD, USA.
| | - Yaguang Xi
- Department of Genetics and Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center, New Orleans, LA, USA.
| |
Collapse
|
1406
|
Lu R, Zhou Q, Ju L, Chen L, Wang F, Shao J. Upregulation of TRIP13 promotes the malignant progression of lung cancer via the EMT pathway. Oncol Rep 2021; 46:172. [PMID: 34184074 PMCID: PMC8261194 DOI: 10.3892/or.2021.8123] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Accepted: 05/26/2021] [Indexed: 12/12/2022] Open
Abstract
Lung cancer is the most common malignant tumor type and it is associated with poor prognosis. The identification of potential biomarkers is of great significance for the early diagnosis and treatment of lung cancer. Non-small cell lung cancer (NSCLC) is the most common pathological type of lung cancer. The present study aimed to investigate the mechanism via which thyroid hormone receptor-interacting protein 13 (TRIP13) participates in the malignant progression of NSCLC. Immunohistochemistry, reverse transcription-quantitative PCR and western blotting were used to assess the expression level of TRIP13. According to The Cancer Genome Atlas database, TRIP13 was upregulated in NSCLC tissues compared with adjacent normal tissues. Moreover, TRIP13 knockdown increased apoptosis, induced cell cycle arrest in the S phase and inhibited the proliferation, invasion and migration of H1299 cells in vitro. Furthermore, TRIP13 upregulation was closely associated with tumor metastasis via epithelial-mesenchymal transformation. In conclusion, TRIP13 could promote the malignant progression of lung cancer, and TRIP13 may be a potential biomarker for the early diagnosis and treatment of NSCLC.
Collapse
Affiliation(s)
- Rujian Lu
- Department of Cardiothoracic Surgery, Nantong Third People's Hospital, Nantong University, Nantong, Jiangsu 226006, P.R. China
| | - Qian Zhou
- Department of Internal Medicine, Medical School of Nantong University, Nantong, Jiangsu 226006, P.R. China
| | - Linling Ju
- Nantong Institute of Liver Disease, Nantong Third People's Hospital, Nantong University, Nantong, Jiangsu 226006, P.R. China
| | - Lin Chen
- Nantong Institute of Liver Disease, Nantong Third People's Hospital, Nantong University, Nantong, Jiangsu 226006, P.R. China
| | - Feng Wang
- Department of Laboratory Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226006, P.R. China
| | - Jianguo Shao
- Nantong Institute of Liver Disease, Nantong Third People's Hospital, Nantong University, Nantong, Jiangsu 226006, P.R. China
| |
Collapse
|
1407
|
NOX4-Derived ROS Mediates TGF- β1-Induced Metabolic Reprogramming during Epithelial-Mesenchymal Transition through the PI3K/AKT/HIF-1 α Pathway in Glioblastoma. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:5549047. [PMID: 34257808 PMCID: PMC8257383 DOI: 10.1155/2021/5549047] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 04/08/2021] [Accepted: 05/30/2021] [Indexed: 12/22/2022]
Abstract
Current studies on tumor progression focus on the roles of cytokines in the tumor microenvironment (TME), and recent research shows that transforming growth factor-β1 (TGF-β1) released from TME plays a pivotal role in tumor development and malignant transformation. The alteration in cellular metabolism is a hallmark of cancer, which not only provides cancer cells with ATP for fuel cellular reactions, but also generates metabolic intermediates for the synthesis of essential cellular ingredients, to support cell proliferation, migration, and invasion. Interestingly, we found a distinct metabolic change during TGF-β1-induced epithelial-mesenchymal transition (EMT) in glioblastoma cells. Indeed, TGF-β1 participates in metabolic reprogramming, and the molecular basis is still not well understood. NADPH oxidases 4 (NOX4), a member of the Nox family, also plays a key role in the biological effects of glioblastoma. However, the relationship between NOX4, TGF-β1, and cellular metabolic changes during EMT in glioblastoma remains obscure. Here, our findings demonstrated that TGF-β1 upregulated NOX4 expression accompanied by reactive oxygen species (ROS) through Smad-dependent signaling and then induced hypoxia-inducible factor 1α (HIF-1α) overexpression and nuclear accumulation resulting in metabolic reprogramming and promoting EMT. Besides, inhibition of glycolysis reversed EMT suggesting a causal relationship between TGF-β1-induced metabolic changes and tumorigenesis. Moreover, TGF-β1-induced metabolic reprogramming and EMT which modulated by NOX4/ROS were blocked when the phosphoinositide3-kinase (PI3K)/AKT/HIF-1α signaling pathways were inhibited. In conclusion, these suggest that NOX4/ROS induction by TGF-β1 can be one of the main mechanisms mediating the metabolic reprogramming during EMT of glioblastoma cells and provide promising strategies for cancer therapy.
Collapse
|
1408
|
Li M, Zhang X, Ding X, Zheng Y, Du H, Li H, Ji H, Wang Z, Jiao P, Song X, Zhong Y, Wu H. Long Noncoding RNA LINC00460 Promotes Cell Progression by Sponging miR-4443 in Head and Neck Squamous Cell Carcinoma. Cell Transplant 2021; 29:963689720927405. [PMID: 32478564 PMCID: PMC7563806 DOI: 10.1177/0963689720927405] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) is one of the most common cancers worldwide. Long noncoding RNAs were proved to be associated with the development and progression in HNSCC. However, the mechanism of LINC00460 in HNSCC needs to be further investigated. The study used quantitative real-time polymerase chain reaction assay to detect the expression of LINC00460 in cancer tissues and cell lines. Gain and loss of function experiments were conducted to analyze the effects of LINC00460 and miR-4443 on cell proliferation, invasion, and apoptosis of HNSCC cells in vitro. The interactions among miR-4443 and LINC00460 were detected by dual-luciferase reporter assay. Here, the study showed that LINC00460 was highly expressed in HNSCC tissues and cell lines. Functionally, knockdown of LINC00460 inhibited HNSCC cell proliferation and migration in vitro. Besides, LINC00460 promoted cell progression by sponging miR-4443, and miR-4443 inhibitor could reverse the effects of si-LINC00460 on cell proliferation and migration. In summary, LINC00460 could potentially promote cell progression and epithelial mesenchymal transition by sponging miR-4443 in HNSCC. LINC00460 could be used as a potential therapeutic target for HNSCCs.
Collapse
Affiliation(s)
- Meng Li
- Department of Oral and Maxillofacial Surgery, Affiliated Stomatological Hospital of Nanjing Medical University, Jiangsu, China.,Jiangsu Key Laboratory of Oral Disease, Nanjing Medical University, Jiangsu, China.,Both the authors contributed equally to this article
| | - Xiaomin Zhang
- Paediatric Dentistry, Affiliated Hospital of Stomatology, Nanjing Medical University, Jiangsu, China.,Both the authors contributed equally to this article
| | - Xu Ding
- Department of Oral and Maxillofacial Surgery, Affiliated Stomatological Hospital of Nanjing Medical University, Jiangsu, China
| | - Yang Zheng
- Department of Oral and Maxillofacial Surgery, Affiliated Stomatological Hospital of Nanjing Medical University, Jiangsu, China
| | - Hongming Du
- Department of Oral and Maxillofacial Surgery, Affiliated Stomatological Hospital of Nanjing Medical University, Jiangsu, China
| | - Huaiqi Li
- Department of Oral and Maxillofacial Surgery, Affiliated Stomatological Hospital of Nanjing Medical University, Jiangsu, China
| | - Huan Ji
- Department of Oral and Maxillofacial Surgery, Affiliated Stomatological Hospital of Nanjing Medical University, Jiangsu, China.,Jiangsu Key Laboratory of Oral Disease, Nanjing Medical University, Jiangsu, China
| | - Zeyu Wang
- Department of Oral and Maxillofacial Surgery, Affiliated Stomatological Hospital of Nanjing Medical University, Jiangsu, China
| | - Pengfei Jiao
- Department of Oral and Maxillofacial Surgery, Affiliated Stomatological Hospital of Nanjing Medical University, Jiangsu, China
| | - Xiaomeng Song
- Department of Oral and Maxillofacial Surgery, Affiliated Stomatological Hospital of Nanjing Medical University, Jiangsu, China
| | - Yi Zhong
- Department of Oral Pathology, Institute of Stomatology, Nanjing Medical University, Jiangsu, China
| | - HeMing Wu
- Department of Oral and Maxillofacial Surgery, Affiliated Stomatological Hospital of Nanjing Medical University, Jiangsu, China
| |
Collapse
|
1409
|
Fu T, Dai LJ, Wu SY, Xiao Y, Ma D, Jiang YZ, Shao ZM. Spatial architecture of the immune microenvironment orchestrates tumor immunity and therapeutic response. J Hematol Oncol 2021; 14:98. [PMID: 34172088 PMCID: PMC8234625 DOI: 10.1186/s13045-021-01103-4] [Citation(s) in RCA: 266] [Impact Index Per Article: 66.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Accepted: 06/03/2021] [Indexed: 02/08/2023] Open
Abstract
Tumors are not only aggregates of malignant cells but also well-organized complex ecosystems. The immunological components within tumors, termed the tumor immune microenvironment (TIME), have long been shown to be strongly related to tumor development, recurrence and metastasis. However, conventional studies that underestimate the potential value of the spatial architecture of the TIME are unable to completely elucidate its complexity. As innovative high-flux and high-dimensional technologies emerge, researchers can more feasibly and accurately detect and depict the spatial architecture of the TIME. These findings have improved our understanding of the complexity and role of the TIME in tumor biology. In this review, we first epitomized some representative emerging technologies in the study of the spatial architecture of the TIME and categorized the description methods used to characterize these structures. Then, we determined the functions of the spatial architecture of the TIME in tumor biology and the effects of the gradient of extracellular nonspecific chemicals (ENSCs) on the TIME. We also discussed the potential clinical value of our understanding of the spatial architectures of the TIME, as well as current limitations and future prospects in this novel field. This review will bring spatial architectures of the TIME, an emerging dimension of tumor ecosystem research, to the attention of more researchers and promote its application in tumor research and clinical practice.
Collapse
Affiliation(s)
- Tong Fu
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Key Laboratory of Breast Cancer in Shanghai, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Lei-Jie Dai
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Key Laboratory of Breast Cancer in Shanghai, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Song-Yang Wu
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Key Laboratory of Breast Cancer in Shanghai, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Yi Xiao
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Key Laboratory of Breast Cancer in Shanghai, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Ding Ma
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Key Laboratory of Breast Cancer in Shanghai, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| | - Yi-Zhou Jiang
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Key Laboratory of Breast Cancer in Shanghai, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| | - Zhi-Ming Shao
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Key Laboratory of Breast Cancer in Shanghai, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
1410
|
Zhang H, Qin G, Zhang C, Yang H, Liu J, Hu H, Wu P, Liu S, Yang L, Chen X, Zhao X, Wang L, Zhang Y. TRAIL promotes epithelial-to-mesenchymal transition by inducing PD-L1 expression in esophageal squamous cell carcinomas. J Exp Clin Cancer Res 2021; 40:209. [PMID: 34167551 PMCID: PMC8223376 DOI: 10.1186/s13046-021-01972-0] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Accepted: 05/05/2021] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Tumor necrosis factor-associated apoptosis-inducing ligand (TRAIL) was initially considered an immunity guard; however, its function remains controversial. Besides immune cells, lung and colon cancer cells have also been reported to express TRAIL, which can promote tumor invasion and metastasis. However, the biological function and underlying mechanism of action of TRAIL in esophageal squamous cell carcinoma (ESCC) remain poorly elucidated. METHODS The ESCC cells stemness, migration, and proliferation ability was assessed by sphere formation, Transwell, and CCK8 assay. The stemness- and epithelial-mesenchymal transition (EMT)- related genes expression levels were analyzed by Western blot and RT-qPCR. The signal activation was conducted by Western blot. The xenograft mouse experiments and lung metastasis model were performed to confirm our findings in vitro. RESULTS Herein, we found that TRAIL is a negative predictor in patients with ESCC. To further investigate the biological function of TRAIL, we established TRAIL knockdown and overexpression ESCC cell lines and found that TRAIL induced EMT and promoted tumor aggressiveness. Furthermore, we demonstrated that TRAIL- overexpressing cells upregulated PD-L1 expression, which was dependent on the p-ERK/STAT3 signaling pathway. We obtained similar results when using recombinant human TRAIL. Finally, we validated the biological role and mechanism of action of TRAIL in vivo. CONCLUSIONS These findings demonstrate that TRAIL promotes ESCC progression by enhancing PD-L1 expression, which induces EMT. This may explain the failure of TRAIL preclinical trials.
Collapse
Affiliation(s)
- Huanyu Zhang
- Biotherapy Center & Cancer Center, the First Affiliated Hospital, Zhengzhou University, 1 Jianshe East Road, Henan, 450052, Zhengzhou, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Henan, 450052, Zhengzhou, China
| | - Guohui Qin
- Biotherapy Center & Cancer Center, the First Affiliated Hospital, Zhengzhou University, 1 Jianshe East Road, Henan, 450052, Zhengzhou, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Henan, 450052, Zhengzhou, China
| | - Chaoqi Zhang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China
| | - Huiyun Yang
- School of Life Sciences, Zhengzhou University, 450052, Zhengzhou, China
| | - Jinyan Liu
- Biotherapy Center & Cancer Center, the First Affiliated Hospital, Zhengzhou University, 1 Jianshe East Road, Henan, 450052, Zhengzhou, China
| | - Hongwei Hu
- Biotherapy Center & Cancer Center, the First Affiliated Hospital, Zhengzhou University, 1 Jianshe East Road, Henan, 450052, Zhengzhou, China
| | - Peng Wu
- Biotherapy Center & Cancer Center, the First Affiliated Hospital, Zhengzhou University, 1 Jianshe East Road, Henan, 450052, Zhengzhou, China
| | - Shasha Liu
- Biotherapy Center & Cancer Center, the First Affiliated Hospital, Zhengzhou University, 1 Jianshe East Road, Henan, 450052, Zhengzhou, China
| | - Li Yang
- Biotherapy Center & Cancer Center, the First Affiliated Hospital, Zhengzhou University, 1 Jianshe East Road, Henan, 450052, Zhengzhou, China
| | - Xinfeng Chen
- Biotherapy Center & Cancer Center, the First Affiliated Hospital, Zhengzhou University, 1 Jianshe East Road, Henan, 450052, Zhengzhou, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Henan, 450052, Zhengzhou, China
| | - Xueke Zhao
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Henan, 450052, Zhengzhou, China
| | - Lidong Wang
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Henan, 450052, Zhengzhou, China
| | - Yi Zhang
- Biotherapy Center & Cancer Center, the First Affiliated Hospital, Zhengzhou University, 1 Jianshe East Road, Henan, 450052, Zhengzhou, China.
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Henan, 450052, Zhengzhou, China.
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China.
- Henan Key Laboratory for Tumor Immunology and Biotherapy, 450052, Zhengzhou, China.
| |
Collapse
|
1411
|
Zhao Y, Wang Z, Gao M, Wang X, Feng H, Cui Y, Tian X. lncRNA MALAT1 regulated ATAD2 to facilitate retinoblastoma progression via miR-655-3p. Open Med (Wars) 2021; 16:931-943. [PMID: 34222668 PMCID: PMC8231467 DOI: 10.1515/med-2021-0290] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 04/03/2021] [Accepted: 04/22/2021] [Indexed: 02/06/2023] Open
Abstract
Long noncoding RNA (lncRNA) metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) was reported as an oncogene in many tumors including retinoblastoma (RB). This research mainly focused on the functions and mechanism of MALAT1 in RB. MALAT1 was upregulated in RB tissues and cells, and it served as a competing endogenous RNA (ceRNA) and inhibited miRNA-655-3p (miR-655-3p) expression, which eventually regulated the expression of miR-655-3p downstream target ATPase Family AAA Domain Containing 2 (ATAD2). The level of ATAD2 significantly increased, while that of miR-655-3p remarkably decreased in RB tissues and cells. MALAT1 depletion inhibited cell proliferation, metastasis, and epithelial-mesenchymal transition (EMT), but promoted apoptosis in vitro and blocked xenograft tumor growth in vivo. MALAT1 exerted its oncogenic functions in RB by regulating miR-655-3p/ATAD2 axis.
Collapse
Affiliation(s)
- Yuxin Zhao
- Department of Ophthalmology, Weihai Central Hospital, No. 3, Mishandongluxi, Wendeng District, Weihai, 264400, Shandong, China
| | - Zhaoxia Wang
- Department of Pediatric, Weihai Central Hospital, Weihai, Shandong, China
| | - Meili Gao
- Department of Ophthalmology, Weihai Central Hospital, No. 3, Mishandongluxi, Wendeng District, Weihai, 264400, Shandong, China
| | - Xuehong Wang
- Department of Ophthalmology, Weihai Central Hospital, No. 3, Mishandongluxi, Wendeng District, Weihai, 264400, Shandong, China
| | - Hui Feng
- Department of Ophthalmology, Weihai Central Hospital, No. 3, Mishandongluxi, Wendeng District, Weihai, 264400, Shandong, China
| | - Yuanyuan Cui
- Department of Ophthalmology, Weihai Central Hospital, No. 3, Mishandongluxi, Wendeng District, Weihai, 264400, Shandong, China
| | - Xia Tian
- Department of Ophthalmology, Weihai Central Hospital, No. 3, Mishandongluxi, Wendeng District, Weihai, 264400, Shandong, China
| |
Collapse
|
1412
|
Li JP, Zeng SH, Zhang YH, Liu YJ. Bioinformatics-based analysis of the association between the A1-chimaerin ( CHN1) gene and gastric cancer. Bioengineered 2021; 12:2874-2889. [PMID: 34152250 PMCID: PMC8806512 DOI: 10.1080/21655979.2021.1940621] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Gastric cancer (GC) is one of the most common causes of cancer-related deaths worldwide and the identification of additional therapeutic targets and biomarkers has become vital. The A1-chimaerin (CHN1) gene encodes a ras-related protein that can be activated or inactivated by binding to GTP or GDP. The present study aimed to assess the expression of CHN1 in GC tissue and cells, to explore its relationship with GC progression, and to discover the potential mechanisms underlying these associations. The ONCOMINE database and The Cancer Genome Atlas (TCGA) were used to determine the transcriptional levels of CHN1 in GC. Western blot and immunohistochemistry were used for detecting protein expression. Correlations between CHN1 levels and the clinical outcomes of GC patients were examined using Kaplan–Meier and Cox regression analyses. Moreover, the CIBERSORT algorithm was used to estimate immune cell infiltration. In GC patients, CHN1 transcription and CHN1 protein expression were upregulated, and a high expression of CHN1 was remarkably linked to poor survival in GC patients. CHN1 expression was associated with immune infiltrates and this gene showed potential involvement in multiple cancer-related pathways. Furthermore, the expression of CHN1 was correlated with the immunotherapeutic response. Finally, our results indicated that the pro-carcinogenic role of CHN1 may involve DNA methylation. To our knowledge, this is the first report characterizing CHN1 expression in GC. Our results show that high CHN1 levels could be used as a clinical biomarker for poor prognosis and that CHN1 inhibitors may have potential as anti-cancer drugs.
Collapse
Affiliation(s)
- Jie-Pin Li
- Department of Oncology, Zhangjiagang TCM Hospital Affiliated to Nanjing, University of Chinese Medicine, Zhangjiagang, Jiangsu, China.,No. 1 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Shu-Hong Zeng
- No. 1 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China.,Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Yong-Hua Zhang
- Department of Oncology, Zhangjiagang TCM Hospital Affiliated to Nanjing, University of Chinese Medicine, Zhangjiagang, Jiangsu, China
| | - Yuan-Jie Liu
- No. 1 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China.,Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| |
Collapse
|
1413
|
Man S, Wu Z, Sun R, Guan Q, Li Z, Zuo D, Zhang W, Wu Y. W436, a novel SMART derivative, exhibits anti-hepatocarcinoma activity by inducing apoptosis and G2/M cell cycle arrest in vitro and in vivo and induces protective autophagy. J Biochem Mol Toxicol 2021; 35:e22831. [PMID: 34155709 DOI: 10.1002/jbt.22831] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 02/25/2021] [Accepted: 05/18/2021] [Indexed: 01/19/2023]
Abstract
Hepatocellular carcinoma (HCC) is considered one of the most common primary liver cancers and the second leading cause of cancer-associated mortality around the world annually. Therefore, it is urgent to develop novel drugs for HCC therapy. We synthesized a novel 4-substituted-methoxybenzoyl-aryl-thiazole (SMART) analog, (5-(4-aminopiperidin-1-yl)-2-phenyl-2H-1,2,3-triazol-4-yl) (3,4,5-trimethoxyphenyl) methanone (W436), with higher solubility, stability, and antitumor activity than SMART against HCC cells in vivo. The purpose of this study was to investigate the mechanisms by which W436 inhibited cell growth in HCC cells. We observed that W436 inhibited the proliferation of HepG2 and Hep3B cells in a dose-dependent manner. Importantly, the anticancer activity of W436 against HCC cells was even higher than that of SMART in vivo. In addition, the antiproliferative effects of W436 on HCC cells were associated with G2/M cell cycle arrest and apoptosis via the activation of reactive oxygen species-mediated mitochondrial apoptotic pathway. W436 also induced protective autophagy by inhibiting the protein kinase B/mammalian target of rapamycin pathway. At the same time, W436 treatment inhibited the cell adhesion and invasion as well as the process of epithelial-to-mesenchymal transition Taken together, our results showed that W436 had the promising potential for the therapeutic treatment of HCC with improved solubility, stability, and bioavailability.
Collapse
Affiliation(s)
- Shuai Man
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, Liaoning, China
| | - Zhuzhu Wu
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, Liaoning, China
| | - Rui Sun
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, Liaoning, China
| | - Qi Guan
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, Liaoning, China
| | - Zengqiang Li
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, Liaoning, China
| | - Daiying Zuo
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, Liaoning, China
| | - Weige Zhang
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, Liaoning, China
| | - Yingliang Wu
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, Liaoning, China
| |
Collapse
|
1414
|
Wen Z, Liang W, Zhong Y, Sun F, Zhang Q. [Expression of nicotinamide-N-methyltransferase in gastric cancer and its biological and clinicopathological significance]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2021; 41:828-838. [PMID: 34238734 PMCID: PMC8267982 DOI: 10.12122/j.issn.1673-4254.2021.06.04] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
OBJECTIVE To investigate the expression of nicotinamide-N-methyltransferase (NNMT) in gastric cancer (GC) and explore its biological and clinicopathological significance. OBJECTIVE We screened the candidate genes associated with the classification and prognosis of gastric cancer by analyzing GEO, Oncomine and TCGA datasets. The molecular pathways and protein interaction network involving these candidate genes were analyzed using STRING, GSEA, David and Cytoscape software. The expressions of the candidate genes in 28 pairs of gastric cancer and adjacent tissues were detected with qRTPCR, and CCK-8 assay, clone formation assay, wound healing assay and Transwell assay were carried out to analyze the effects of modulation of NNMT expression on proliferation, invasion and migration of different gastric cancer cell lines. OBJECTIVE NNMT was highly expressed in gastric cancer tissues and was negatively correlated with the prognosis of patients with gastric cancer. Pathway analysis showed that the high expression of NNMT was associated with adhesion-related pathway molecules such as extracellular matrix receptors, cell adhesion molecules, and cytokine receptors, while its low expression was associated with base mismatch repair and riboflavin metabolism. Protein interaction analysis showed that NNMT interacted with 16 differentially expressed proteins such as AURKA and was co-expressed with TAGLN, PTRF, AKAP12 and IGF2BP2. In clinical tissue specimens, qRT-PCR results showed that the expression of NNMT mRNA was significantly higher in gastric cancer tissues than in the adjacent tissues (P < 0.05). In gastric cancer cell lines, overexpression of NNMT was found to significantly promote cell proliferation, invasion and migration, while NNMT knockdown produced obvious inhibitory effects on cell proliferation, invasion and migration. OBJECTIVE NNMT is highly expressed in gastric cancer and negatively correlated with the prognosis of gastric cancer patients. The high expression of NNMT promotes the proliferation, invasion and metastasis of gastric cancer cells, suggesting the potential of NNMT as prognostic marker of gastric cancer.
Collapse
Affiliation(s)
- Z Wen
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - W Liang
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
- First Clinical Medical School, Southern Medical University, Guangzhou 510515, China
| | - Y Zhong
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
- First Clinical Medical School, Southern Medical University, Guangzhou 510515, China
| | - F Sun
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
- First Clinical Medical School, Southern Medical University, Guangzhou 510515, China
| | - Q Zhang
- Department of Pathology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
1415
|
Zhao X, Jiang M, Teng Y, Li J, Li Z, Hao W, Zhao H, Yin C, Yue W. Cytoplasmic Localization Isoform of Cyclin Y Enhanced the Metastatic Ability of Lung Cancer via Regulating Tropomyosin 4. Front Cell Dev Biol 2021; 9:684819. [PMID: 34222253 PMCID: PMC8250429 DOI: 10.3389/fcell.2021.684819] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 05/26/2021] [Indexed: 11/13/2022] Open
Abstract
Cyclin Y (CCNY) is a novel cyclin and highly conserved in metazoan species. Previous studies from our and other laboratory indicate that CCNY play a crucial role in tumor progression. There are two CCNY isoform which has different subcellular distributions, with cytoplasmic isoform (CCNYc) and membrane distribution isoform (CCNYm). However, the expression and function of CCNY isoforms is still unclear. We firstly found CCNYc was expressed in natural lung cancer tissue and cells through the subcellular distribution. Co-IP and immunofluorescence showed that both CCNYm and CCNYc could interact with PFTK1. Further studies illustrated that CCNYc but not CCNYm enhanced cell migration and invasion activity both in vivo and vitro. The function of CCNYc could be inhibited by suppression of PFTK1 expression. In addition, our data indicated that tropomyosin 4 (TPM4), a kind of actin-binding proteins, was down-regulated by suppression of CCNY. F-actin assembly could be controlled by CCNYc as well as PFTK1 and TPM4. As a result, CCNY was mainly expressed in lung cancer. CCNYc could promote cell motility and invasion. It indicated that CCNYc/PFTK1 complex could promote cell metastasis by regulating the formation of F-actin via TPM4.
Collapse
Affiliation(s)
- Xiaoting Zhao
- Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, China
| | - Mei Jiang
- Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, China
| | - Yu Teng
- Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, China
| | - Jie Li
- Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, China
| | - Zhefeng Li
- Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, China
| | - Wende Hao
- Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, China
| | - Hongyu Zhao
- Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, China
| | - Chenghong Yin
- Departments of Internal Medicine, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, China
| | - Wentao Yue
- Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
1416
|
Li W, Song Y, Pan C, Yu J, Zhang J, Zhu X. Aquaporin-8 is a novel marker for progression of human cervical cancer cells. Cancer Biomark 2021; 32:391-400. [PMID: 34151838 PMCID: PMC8673491 DOI: 10.3233/cbm-203251] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND: Role of aquaporin-8 (AQP8) in cervical cancer has not been fully elucidated. OBJECTIVE: We aim to explore the impacts of AQP8 on viability, apoptosis and metastasis in cervical cancer cells. METHODS: AQP8 protein expression in cervical carcinoma specimens and cell lines was detected by IHC and western blot analysis. Lentivirus-mediated transfection was used to upregulate and knockdown AQP8 in cells. Cell viability and apoptosis were assessed by CCK-8 and flow cytometry assays, respectively. Transwell experiments were conducted to investigate cell invasive and migratory capabilities. EMT-related markers were detected by western blot analysis. RESULTS: A strong positive of AQP8 protein expression was observed in cervical cancer tissues. Western blot analysis confirmed overexpression and knockdown of AQP8 in SiHa cells. AQP8-overexpressed SiHa cells displayed an enhanced viability, reduced apoptotic rate, increased invasive and migratory abilities. Knockdown of AQP8 inhibited the viability, promoted the apoptosis, and suppressed invasion and migration. Furthermore, AQP8 overexpression significantly upregulated vimentin and N-cadherin, and downregulated E-cadherin, which were reversed by AQP8 knockdown. CONCLUSIONS: AQP8 increases viability, inhibits apoptosis, and facilitates metastasis in SiHa cells. This may be associated with EMT-related markers regulated by AQP8. AQP8 could serve as a potential marker for cervical cancer progression.
Collapse
Affiliation(s)
- Weibo Li
- Center of Uterine Cancer Diagnosis and Therapy Research of Zhejiang Province, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.,Department of Obstetrics and Gynecology, Affiliated Taizhou Hospital of Wenzhou Medical University, Taizhou, Zhejiang, China.,Center of Uterine Cancer Diagnosis and Therapy Research of Zhejiang Province, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yizuo Song
- Center of Uterine Cancer Diagnosis and Therapy Research of Zhejiang Province, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.,Center of Uterine Cancer Diagnosis and Therapy Research of Zhejiang Province, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Chunyu Pan
- Center of Uterine Cancer Diagnosis and Therapy Research of Zhejiang Province, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Junhui Yu
- Center of Uterine Cancer Diagnosis and Therapy Research of Zhejiang Province, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jianan Zhang
- Center of Uterine Cancer Diagnosis and Therapy Research of Zhejiang Province, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xueqiong Zhu
- Center of Uterine Cancer Diagnosis and Therapy Research of Zhejiang Province, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
1417
|
González-González R, Ortiz-Sarabia G, Molina-Frechero N, Salas-Pacheco JM, Salas-Pacheco SM, Lavalle-Carrasco J, López-Verdín S, Tremillo-Maldonado O, Bologna-Molina R. Epithelial-Mesenchymal Transition Associated with Head and Neck Squamous Cell Carcinomas: A Review. Cancers (Basel) 2021; 13:3027. [PMID: 34204259 PMCID: PMC8234594 DOI: 10.3390/cancers13123027] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 06/09/2021] [Accepted: 06/15/2021] [Indexed: 02/07/2023] Open
Abstract
Head and neck squamous cell carcinomas (HNSCCs) are aggressive, recurrent, and metastatic neoplasms with a high occurrence around the world and can lead to death when not treated appropriately. Several molecules and signaling pathways are involved in the malignant conversion process. Epithelial-mesenchymal transition (EMT) has been described in HNSCCs, a major type of aggressive carcinoma. EMT describes the development of epithelial cells into mesenchymal cells, which depends on several molecular interactions and signaling pathways that facilitate mesenchymal conversion. This is related to interactions with the microenvironment of the tumor, hypoxia, growth factors, matrix metalloproteinases, and the presence of viral infections. In this review, we focus on the main molecules related to EMT, their interactions with the tumor microenvironment, plasticity phenomena, epigenetic regulation, hypoxia, inflammation, their relationship with immune cells, and the inhibition of EMT in the context of HNSCCs.
Collapse
Affiliation(s)
- Rogelio González-González
- Department of Research, School of Dentistry, Universidad Juárez del Estado de Durango, Durango 34000, Mexico; (R.G.-G.); (G.O.-S.); (O.T.-M.)
| | - Gamaliel Ortiz-Sarabia
- Department of Research, School of Dentistry, Universidad Juárez del Estado de Durango, Durango 34000, Mexico; (R.G.-G.); (G.O.-S.); (O.T.-M.)
| | - Nelly Molina-Frechero
- Xochimilco Unit, Department of Health Care, Universidad Autónoma Metropolitana (UAM) Xochimilco, Mexico City 04960, Mexico; (N.M.-F.); (J.L.-C.)
| | - José Manuel Salas-Pacheco
- Scientific Research Institute, Universidad Juárez del Estado de Durango, Avenida Universidad S/N, Durango 34000, Mexico; (J.M.S.-P.); (S.M.S.-P.)
| | - Sergio Manuel Salas-Pacheco
- Scientific Research Institute, Universidad Juárez del Estado de Durango, Avenida Universidad S/N, Durango 34000, Mexico; (J.M.S.-P.); (S.M.S.-P.)
| | - Jesús Lavalle-Carrasco
- Xochimilco Unit, Department of Health Care, Universidad Autónoma Metropolitana (UAM) Xochimilco, Mexico City 04960, Mexico; (N.M.-F.); (J.L.-C.)
| | - Sandra López-Verdín
- Health Science Center, Dentistry Research Institute, Universidad de Guadalajara, Guadalajara 4430, Mexico;
| | - Omar Tremillo-Maldonado
- Department of Research, School of Dentistry, Universidad Juárez del Estado de Durango, Durango 34000, Mexico; (R.G.-G.); (G.O.-S.); (O.T.-M.)
| | - Ronell Bologna-Molina
- Department of Research, School of Dentistry, Universidad Juárez del Estado de Durango, Durango 34000, Mexico; (R.G.-G.); (G.O.-S.); (O.T.-M.)
- Molecular Pathology Area, School of Dentistry, Universidad de la República, Montevideo 11600, Uruguay
| |
Collapse
|
1418
|
Kähkönen TE, Halleen JM, Bernoulli J. Osteoimmuno-Oncology: Therapeutic Opportunities for Targeting Immune Cells in Bone Metastasis. Cells 2021; 10:1529. [PMID: 34204474 PMCID: PMC8233913 DOI: 10.3390/cells10061529] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 06/11/2021] [Accepted: 06/14/2021] [Indexed: 12/11/2022] Open
Abstract
Immunotherapies provide a potential treatment option for currently incurable bone metastases. Bone marrow is an important secondary lymphoid organ with a unique immune contexture. Even at non-disease state immune cells and bone cells interact with each other, bone cells supporting the development of immune cells and immune cells regulating bone turnover. In cancer, tumor cells interfere with this homeostatic process starting from formation of pre-metastatic niche and later supporting growth of bone metastases. In this review, we introduce a novel concept osteoimmuno-oncology (OIO), which refers to interactions between bone, immune and tumor cells in bone metastatic microenvironment. We also discuss therapeutic opportunities of targeting immune cells in bone metastases, and associated efficacy and safety concerns.
Collapse
Affiliation(s)
| | | | - Jenni Bernoulli
- Institute of Biomedicine, University of Turku, 20500 Turku, Finland;
| |
Collapse
|
1419
|
Miao C, Liang C, Li P, Liu B, Qin C, Yuan H, Liu Y, Zhu J, Cui Y, Xu A, Wang S, Su S, Li J, Shao P, Wang Z. TRIM37 orchestrates renal cell carcinoma progression via histone H2A ubiquitination-dependent manner. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2021; 40:195. [PMID: 34130705 PMCID: PMC8204444 DOI: 10.1186/s13046-021-01980-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Accepted: 05/12/2021] [Indexed: 01/02/2023]
Abstract
Background Ubiquitylation modification is one of the multiple post-transcriptional process to regulate cellular physiology, including cell signaling, cycle regulation, DNA repair and transcriptional regulation. Members of TRIM family proteins could be defined as E3 ubiquitin ligases as they contain a RING-finger domain, and alterations of TRIM proteins are involved into a broad range of diverse disorders including cancer. TRIM37 is a novel discovered E3 ubiquitin ligase and acts as a oncoprotein in multiple human neoplasms, however its biological role in RCC still remains elusive. Methods RCC microarray chips and public datasets were screened to identify novel TRIMs member as TRIM37, which was dysregulated in RCC. Gain or loss of functional cancer cell models were constructed, and in vitro and in vivo assays were performed to elucidate its tumorigenic phenotypes. Interactive network analyses were utilized to define intrinsic mechanism. Results We identified TRIM37 was upregulated in RCC tumors, and its aberrant function predicted aggressive neoplastic phenotypes, poorer survival endings. TRIM37 promoted RCC cells EMT and malignant progression via TGF-β1 signaling activation, as a consequence of directly mediated by ubiquitinating-H2A modifications. Conclusions Our findings identified a previously unappreciated role of TRIM37 in RCC progression and prognostic prediction. Importantly, we declared a novel ubiquitination-dependent link between TRIM ubiquitin ligases and TGF-β1 signaling in regulating cancerous malignancies. Supplementary Information The online version contains supplementary material available at 10.1186/s13046-021-01980-0.
Collapse
Affiliation(s)
- Chenkui Miao
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Chao Liang
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Pu Li
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Bianjiang Liu
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Chao Qin
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Han Yuan
- Center for Quantitative Medicine, Duke-NUS Medical School, National University of Singapore, Singapore, SG, 169857, Singapore
| | - Yiyang Liu
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Jundong Zhu
- Department of Urology, The First People's Hospital of Changzhou, Changzhou, 213003, China
| | - Yankang Cui
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Aiming Xu
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Shangqian Wang
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Shifeng Su
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Jie Li
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Pengfei Shao
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.
| | - Zengjun Wang
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.
| |
Collapse
|
1420
|
Zhang C, Zhang Y, Pan H, Tan Y, Wei Q, Dai X, Wei J, Chen Y. Combination of Ferulic Acid, Ligustrazine and Tetrahydropalmatine attenuates Epithelial-mesenchymal Transformation via Wnt/β-catenin Pathway in Endometriosis. Int J Biol Sci 2021; 17:2449-2460. [PMID: 34326686 PMCID: PMC8315018 DOI: 10.7150/ijbs.60167] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Accepted: 05/22/2021] [Indexed: 12/22/2022] Open
Abstract
Previously the potential therapeutic action of ferulic acid, ligustrazine and tetrahydropalmatine (FLT) are discovered with unclear mechanism in rat autograft endometriosis. However, the effect of FLT on endometrial cells and allograft endometriosis is still unclear. This study is designed to elucidate the influence of FLT on epithelial-mesenchymal transformation in allograft endometriosis and endometrium cells. In vivo, fluorescent xenogeneic endometriosis model was established. In vitro, invasion and metastasis were analyzed after treating FLT. Epithelial-mesenchymal transformation and Wnt/β-catenin pathway were inspected in vitro and in vivo. Activator or inhibitor of Wnt/β-catenin signaling was performed to inspect mechanism of epithelial-mesenchymal transformation. In vivo, FLT not only decreased fluorescent intensity and volume of ectopic lesion, but also ameliorated pathological morphology. E2 and PROG levels in serum were reduced by FLT. In endometrial cells, FLT significantly inhibited the invasion and metastasis. Meantime, epithelial-mesenchymal transformation was reversed, accompanied by suppression of Wnt/β-catenin pathway. In-depth study, activation of Wnt/β-catenin pathway lead to promotion of epithelial-mesenchymal transformation, which was reversed by FLT. FLT prevented fluorescent allograft endometriosis and endometrium cells, which was related to suppress epithelial-mesenchymal transformation through inactivating Wnt/β-catenin pathway. The findings disclose molecular mechanism of epithelial-mesenchymal transformation in endometriosis by FLT, and contribute to further application.
Collapse
Affiliation(s)
- Chengling Zhang
- College of Pharmaceutical Sciences & Chinese Medicine, Southwest University, Chongqing, China.,Chongqing Key Laboratory of New Drug Screening from Traditional Chinese Medicine, Chongqing, China.,Pharmacology of Chinese Materia Medica - the Key Discipline Constructed by the state Administration of Traditional Chinese Medicine, Chongqing, China.,National Demonstration Center for Experimental Pharmacy Education (Southwest University), Chongqing, China
| | - Ying Zhang
- College of Pharmaceutical Sciences & Chinese Medicine, Southwest University, Chongqing, China.,Chongqing Key Laboratory of New Drug Screening from Traditional Chinese Medicine, Chongqing, China.,Pharmacology of Chinese Materia Medica - the Key Discipline Constructed by the state Administration of Traditional Chinese Medicine, Chongqing, China.,National Demonstration Center for Experimental Pharmacy Education (Southwest University), Chongqing, China
| | - Haiying Pan
- Sichuan Jinxin Women & Children Hospital, Chengdu 610066, China
| | - Yi Tan
- College of Pharmaceutical Sciences & Chinese Medicine, Southwest University, Chongqing, China.,Chongqing Key Laboratory of New Drug Screening from Traditional Chinese Medicine, Chongqing, China.,Pharmacology of Chinese Materia Medica - the Key Discipline Constructed by the state Administration of Traditional Chinese Medicine, Chongqing, China.,National Demonstration Center for Experimental Pharmacy Education (Southwest University), Chongqing, China
| | - Qinghua Wei
- College of Pharmaceutical Sciences & Chinese Medicine, Southwest University, Chongqing, China.,Chongqing Key Laboratory of New Drug Screening from Traditional Chinese Medicine, Chongqing, China.,Pharmacology of Chinese Materia Medica - the Key Discipline Constructed by the state Administration of Traditional Chinese Medicine, Chongqing, China.,National Demonstration Center for Experimental Pharmacy Education (Southwest University), Chongqing, China
| | - Xueshan Dai
- College of Pharmaceutical Sciences & Chinese Medicine, Southwest University, Chongqing, China.,Chongqing Key Laboratory of New Drug Screening from Traditional Chinese Medicine, Chongqing, China.,Pharmacology of Chinese Materia Medica - the Key Discipline Constructed by the state Administration of Traditional Chinese Medicine, Chongqing, China.,National Demonstration Center for Experimental Pharmacy Education (Southwest University), Chongqing, China
| | - Jiahui Wei
- College of Pharmaceutical Sciences & Chinese Medicine, Southwest University, Chongqing, China.,Chongqing Key Laboratory of New Drug Screening from Traditional Chinese Medicine, Chongqing, China.,Pharmacology of Chinese Materia Medica - the Key Discipline Constructed by the state Administration of Traditional Chinese Medicine, Chongqing, China.,National Demonstration Center for Experimental Pharmacy Education (Southwest University), Chongqing, China
| | - Yi Chen
- College of Pharmaceutical Sciences & Chinese Medicine, Southwest University, Chongqing, China.,Chongqing Key Laboratory of New Drug Screening from Traditional Chinese Medicine, Chongqing, China.,Pharmacology of Chinese Materia Medica - the Key Discipline Constructed by the state Administration of Traditional Chinese Medicine, Chongqing, China.,National Demonstration Center for Experimental Pharmacy Education (Southwest University), Chongqing, China
| |
Collapse
|
1421
|
Gómez-Valenzuela F, Escobar E, Pérez-Tomás R, Montecinos VP. The Inflammatory Profile of the Tumor Microenvironment, Orchestrated by Cyclooxygenase-2, Promotes Epithelial-Mesenchymal Transition. Front Oncol 2021; 11:686792. [PMID: 34178680 PMCID: PMC8222670 DOI: 10.3389/fonc.2021.686792] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Accepted: 05/13/2021] [Indexed: 12/11/2022] Open
Abstract
The tumor microenvironment (TME) corresponds to a complex and dynamic interconnection between the extracellular matrix and malignant cells and their surrounding stroma composed of immune and mesenchymal cells. The TME has constant cellular communication through cytokines that sustain an inflammatory profile, which favors tumor progression, angiogenesis, cell invasion, and metastasis. Although the epithelial-mesenchymal transition (EMT) represents a relevant metastasis-initiating event that promotes an invasive phenotype in malignant epithelial cells, its relationship with the inflammatory profile of the TME is poorly understood. Previous evidence strongly suggests that cyclooxygenase-2 (COX-2) overexpression, a pro-inflammatory enzyme related to chronic unresolved inflammation, is associated with common EMT-signaling pathways. This review article summarizes how COX-2 overexpression, within the context of the TME, orchestrates the EMT process and promotes initial metastatic-related events.
Collapse
Affiliation(s)
- Fernán Gómez-Valenzuela
- Department of Hematology-Oncology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Enrico Escobar
- Department of Oral Pathology and Medicine, Faculty of Dentistry, University of Chile, Santiago, Chile
| | - Ricardo Pérez-Tomás
- Department of Pathology and Experimental Therapy - Bellvitge, Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
| | - Viviana P Montecinos
- Department of Hematology-Oncology, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
1422
|
Tata A, Chow RD, Tata PR. Epithelial cell plasticity: breaking boundaries and changing landscapes. EMBO Rep 2021; 22:e51921. [PMID: 34096150 DOI: 10.15252/embr.202051921] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 05/08/2021] [Accepted: 05/18/2021] [Indexed: 12/17/2022] Open
Abstract
Epithelial tissues respond to a wide variety of environmental and genotoxic stresses. As an adaptive mechanism, cells can deviate from their natural paths to acquire new identities, both within and across lineages. Under extreme conditions, epithelial tissues can utilize "shape-shifting" mechanisms whereby they alter their form and function at a tissue-wide scale. Mounting evidence suggests that in order to acquire these alternate tissue identities, cells follow a core set of "tissue logic" principles based on developmental paradigms. Here, we review the terminology and the concepts that have been put forward to describe cell plasticity. We also provide insights into various cell intrinsic and extrinsic factors, including genetic mutations, inflammation, microbiota, and therapeutic agents that contribute to cell plasticity. Additionally, we discuss recent studies that have sought to decode the "syntax" of plasticity-i.e., the cellular and molecular principles through which cells acquire new identities in both homeostatic and malignant epithelial tissues-and how these processes can be manipulated for developing novel cancer therapeutics.
Collapse
Affiliation(s)
- Aleksandra Tata
- Department of Cell Biology, Duke University School of Medicine, Durham, NC, USA
| | - Ryan D Chow
- Department of Genetics, Systems Biology Institute, Medical Scientist Training Program, Yale University School of Medicine, New Haven, CT, USA
| | - Purushothama Rao Tata
- Department of Cell Biology, Duke University School of Medicine, Durham, NC, USA.,Duke Cancer Institute, Duke University School of Medicine, Durham, NC, USA.,Regeneration Next, Duke University, Durham, NC, USA.,Center for Advanced Genomic Technologies, Duke University, Durham, NC, USA
| |
Collapse
|
1423
|
Kim D, Kim KI, Baek SH. Roles of lysine-specific demethylase 1 (LSD1) in homeostasis and diseases. J Biomed Sci 2021; 28:41. [PMID: 34082769 PMCID: PMC8175190 DOI: 10.1186/s12929-021-00737-3] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 06/01/2021] [Indexed: 12/14/2022] Open
Abstract
Lysine-specific demethylase 1 (LSD1) targets mono- or di-methylated histone H3K4 and H3K9 as well as non-histone substrates and functions in the regulation of gene expression as a transcriptional repressor or activator. This enzyme plays a pivotal role in various physiological processes, including development, differentiation, inflammation, thermogenesis, neuronal and cerebral physiology, and the maintenance of stemness in stem cells. LSD1 also participates in pathological processes, including cancer as the most representative disease. It promotes oncogenesis by facilitating the survival of cancer cells and by generating a pro-cancer microenvironment. In this review, we discuss the role of LSD1 in several aspects of cancer, such as hypoxia, epithelial-to-mesenchymal transition, stemness versus differentiation of cancer stem cells, as well as anti-tumor immunity. Additionally, the current understanding of the involvement of LSD1 in various other pathological processes is discussed.
Collapse
Affiliation(s)
- Dongha Kim
- Department of Anatomy, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Keun Il Kim
- Department of Biological Sciences, Sookmyung Women's University, Seoul, 04310, Republic of Korea.
| | - Sung Hee Baek
- Creative Research Initiatives Center for Epigenetic Code and Diseases, Department of Biological Sciences, Seoul National University, Seoul, 08826, Republic of Korea.
| |
Collapse
|
1424
|
Zhu B, Qian W, Han C, Bai T, Hou X. Piezo 1 activation facilitates cholangiocarcinoma metastasis via Hippo/YAP signaling axis. MOLECULAR THERAPY. NUCLEIC ACIDS 2021; 24:241-252. [PMID: 33767919 PMCID: PMC7973248 DOI: 10.1016/j.omtn.2021.02.026] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Accepted: 02/22/2021] [Indexed: 12/12/2022]
Abstract
Tumor metastasis is one of the major factors for the high mortality in cholangiocarcinoma (CCA), but its underlying mechanisms are not fully understood. Here, we report that Piezo-type mechanosensitive ion channel component 1 (Piezo 1) is detected to be significantly upregulated in CCA tissues, which is linked to a poor prognosis in patients, suggesting that Piezo 1 may act in a pro-metastatic role in CCA development. Piezo 1 is activated through 20% simulated physiological stretch, and deleting Piezo 1 impedes epithelial-to-mesenchymal transition (EMT) of CCA cells, as well as impairing their metastatic capacity in vitro and in vivo. Mechanistically, the activation of Piezo 1 results in large amounts of Yes-associated protein 1 (YAP) translocated into the nucleus from the cytoplasm, and thus the motility of CCA cells is significantly increased. These findings indicate that mechanical stimulation induces Piezo 1 activation, which might be involved in CCA metastasis via the Hippo/YAP signaling axis. Therefore, Piezo 1 and its downstream effectors may be a novel therapeutic target for CCA treatment.
Collapse
Affiliation(s)
- Biqiang Zhu
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Wei Qian
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Chaoqun Han
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Tao Bai
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Xiaohua Hou
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| |
Collapse
|
1425
|
Huang W, Zhu J, Shi H, Wu Q, Zhang C. ITGA2 Overexpression Promotes Esophageal Squamous Cell Carcinoma Aggression via FAK/AKT Signaling Pathway. Onco Targets Ther 2021; 14:3583-3596. [PMID: 34113124 PMCID: PMC8185252 DOI: 10.2147/ott.s302028] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 05/10/2021] [Indexed: 12/13/2022] Open
Abstract
Background Integrin alpha 2 (ITGA2) is highly expressed in various cancers. ITGA2 up regulation promotes tumor proliferation, invasion, migration, and angiogenesis and ITGA2 is a poor prognostic factor in many tumors. However, the mechanism underlying its role in esophageal squamous cell carcinoma (ESCC) is unknown. Methods The expression profile of ITGA2 in ESCC was analyzed using the Gene expression profiling interactive analysis (GEPIA). ESCC tissues were analyzed by real time PCR (RT-qPCR) and immunohistochemistry to verify ITGA2 expression. The impact of ITGA2 on the clinicopathological characteristics was explored using a chi-square test. Apoptosis, Transwell, colony formation, and wound healing assays were conducted to characterize the roles of ITGA2 in ESCC. Its impact on tumorigenesis was further examined using a tumor xenograft model. The expression of proteins associated with the epithelial-mesenchymal Transition (EMT) and focal adhesion kinase (FAK)/AKT pathway and regulated by ITGA2 was evaluated with Western blot analysis. The Akt inhibitor MK-2206 was used to explore the interaction of ITGA2 with the FAK/Akt pathway. Results ITGA2 was upregulated in ESCC tissues and related to lymph node metastasis as well as TNM stage. In vitro experimental models revealed that ITGA2 promotes proliferation, invasion, and migration, and inhibits apoptosis. In vivo experiments show that ITGA2 promotes ESCC proliferation. Additionally, Western blot analysis revealed that ITGA2 silencing inhibits FAK/AKT signaling and suppresses EMT, while its overexpression activates FAK/AKT signaling and promotes EMT. Moreover, treatment with the AKT inhibitor MK-2206 successfully repressed the progression of ESCC caused by ITGA2 overexpression. Conclusion Our findings indicated that in ESCC, ITGA2 promotes proliferation, invasion and migration, while inhibiting apoptosis and promoting EMT in ESCC, possibly via FAK/AKT phosphorylation. These findings highlight the therapeutic value of ITGA2 in ESCC.
Collapse
Affiliation(s)
- Wei Huang
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China.,Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Ju Zhu
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Haoming Shi
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Qingchen Wu
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Cheng Zhang
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| |
Collapse
|
1426
|
Huo Q, Chen S, Li Z, Wang J, Li J, Xie N. Inhibiting of TACC3 Promotes Cell Proliferation, Cell Invasion and the EMT Pathway in Breast Cancer. Front Genet 2021; 12:640078. [PMID: 34149795 PMCID: PMC8209498 DOI: 10.3389/fgene.2021.640078] [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: 12/10/2020] [Accepted: 04/06/2021] [Indexed: 01/15/2023] Open
Abstract
Accumulating evidences indicate that transforming acidic coiled-coil 3 (TACC3) is a tumor-related gene, was highly expressed in a variety of human cancers, which is involved in cancer development. However, the potential role of TACC3 in breast cancer remains largely unknown. In the present study, we found that TACC3 was highly-expressed in breast cancer tissues, and its level was positively correlated with the clinical features of breast cancer patients. Specifically, TACC3 expression was significantly associated with the estrogen receptor (ER), progesterone receptor (PR), human epidermal growth factor receptor 2 (HER2) status, nodal status, the scarff-bloom-richardson (SBR) grade, nottingham prognostic index (NPI), age, subtypes, and triple-negative and basal-like status, suggesting that TACC3 may be a potential diagnostic indicator of breast cancer. Furthermore, functional studies have shown that inhibition of TACC3 can significantly promote the cell proliferation and viability of breast cancer cells. Moreover, TACC3 knockdown suppressed the expression of E-cadherin, but increased the expression of N-cadherin, Snail, ZEB1, and TWIST, which indicate that TACC3 may impact the migration of breast cancer cells in vitro. Taken together, these findings indicate that TACC3 may serve as a prognostic and therapeutic indicator of breast cancer.
Collapse
Affiliation(s)
- Qin Huo
- Biobank, Institute of Translational medicine, Shenzhen Second People's Hospital, Graduate School of Guangzhou Medical University, Shenzhen, China
| | - Siqi Chen
- Biobank, Institute of Translational medicine, Shenzhen Second People's Hospital, Graduate School of Guangzhou Medical University, Shenzhen, China
| | - Zhenwei Li
- Biobank, Institute of Translational medicine, Shenzhen Second People's Hospital, Graduate School of Guangzhou Medical University, Shenzhen, China
| | - Juan Wang
- Department of Clinical Medicine, University of South China, Hengyang, China
| | - Jiaying Li
- Department of Clinical Medicine, University of South China, Hengyang, China
| | - Ni Xie
- Biobank, Institute of Translational medicine, Shenzhen Second People's Hospital, Graduate School of Guangzhou Medical University, Shenzhen, China
| |
Collapse
|
1427
|
Ishikawa S, Nishida N, Fujino S, Ogino T, Takahashi H, Miyoshi N, Uemura M, Satoh T, Yamamoto H, Mizushima T, Doki Y, Eguchi H. Comprehensive profiling of novel epithelial-mesenchymal transition mediators and their clinical significance in colorectal cancer. Sci Rep 2021; 11:11759. [PMID: 34083586 PMCID: PMC8175715 DOI: 10.1038/s41598-021-91102-9] [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: 10/11/2020] [Accepted: 05/21/2021] [Indexed: 12/27/2022] Open
Abstract
Epithelial-mesenchymal transition (EMT) is a drastic phenotypic change during cancer metastasis and is one of the most important hallmarks of aggressive cancer. Although the overexpression of some specific transcription factors explains the functional alteration of EMT-induced cells, a complete picture of this biological process is yet to be elucidated. To comprehensively profile EMT-related genes in colorectal cancer, we quantified the EMT induction ability of each gene according to its similarity to the cancer stromal gene signature and termed it "mesenchymal score." This bioinformatic approach successfully identified 90 candidate EMT mediators, which are strongly predictive of survival in clinical samples. Among these candidates, we discovered that the neuronal gene ARC, possibly originating from the retrotransposon, unexpectedly plays a crucial role in EMT induction. Profiling of novel EMT mediators we demonstrated here may help understand the complexity of the EMT program and open up new avenues for therapeutic intervention in colorectal cancer.
Collapse
Affiliation(s)
- Satoshi Ishikawa
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Naohiro Nishida
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan.
- Department of Frontier Science for Cancer and Chemotherapy, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| | - Shiki Fujino
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Takayuki Ogino
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Hidekazu Takahashi
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Norikatsu Miyoshi
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Mamoru Uemura
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Taroh Satoh
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
- Department of Frontier Science for Cancer and Chemotherapy, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Hirofumi Yamamoto
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Tsunekazu Mizushima
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Yuichiro Doki
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Hidetoshi Eguchi
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| |
Collapse
|
1428
|
Song X, Jiao X, Yan H, Yu L, Jiang L, Zhang M, Chen L, Ju M, Wang L, Wei Q, Zhao L, Wei M. Overexpression of PTPRN Promotes Metastasis of Lung Adenocarcinoma and Suppresses NK Cell Cytotoxicity. Front Cell Dev Biol 2021; 9:622018. [PMID: 34150744 PMCID: PMC8207963 DOI: 10.3389/fcell.2021.622018] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 04/06/2021] [Indexed: 01/04/2023] Open
Abstract
Background Lung adenocarcinoma (LUAD) is the most common diagnostic histologic subtype of non-small cell lung cancer, but the role of receptor-type tyrosine-protein phosphatase-like N (PTPRN) in LUAD has not been studied. Methods We conducted a bioinformatic analysis to identify the expression of PTPRN on LUAD data from the Cancer Genome Atlas (TCGA) and the relationship between PTPRN and overall survival of LUAD patients. The effects of PTPRN on the migration ability of LUAD cells and the underlying mechanisms were investigated by in vitro and in vivo assays (i.e., wound healing assay, transwell assay, western blotting, xenograft model, and immunohistochemistry). Gene-set enrichment analysis and computational resource were used to analyze the correlation between PTPRN and different tumor-infiltrating immune cells (TIICs). Lactate dehydrogenase assay and Enzyme-linked immunosorbent assay were conducted to examine natural killer (NK) cell cytotoxicity. Results In our study, we found that PTPRN was up-regulated in LUAD and related to metastasis of LUAD patients. Besides, PTPRN was correlated with poor prognosis in the TCGA-LUAD dataset. PTPRN overexpression promoted LUAD cell migration and the expression of EMT markers by influencing MEK/ERK and PI3K/AKT signaling. Moreover, PTPRN expression was significantly associated with TIICs, especially NK cells. A549 and H1299 cells overexpressed PTPRN inhibited NK cell cytotoxicity. Conclusion Taken together, these findings demonstrated that PTPRN might be a potential and novel therapeutic target modulating antitumor immune response in treatment of LUAD.
Collapse
Affiliation(s)
- Xinyue Song
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China.,Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, China Medical University, Shenyang, China.,Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, China Medical University, Shenyang, China.,Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, China
| | - Xue Jiao
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China.,Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, China Medical University, Shenyang, China.,Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, China Medical University, Shenyang, China.,Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, China
| | - Han Yan
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China.,Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, China Medical University, Shenyang, China.,Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, China Medical University, Shenyang, China.,Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, China
| | - Lifeng Yu
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China.,Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, China Medical University, Shenyang, China.,Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, China Medical University, Shenyang, China.,Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, China
| | - Longyang Jiang
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China.,Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, China Medical University, Shenyang, China.,Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, China Medical University, Shenyang, China.,Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, China
| | - Ming Zhang
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China.,Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, China Medical University, Shenyang, China.,Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, China Medical University, Shenyang, China.,Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, China
| | - Lianze Chen
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China.,Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, China Medical University, Shenyang, China.,Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, China Medical University, Shenyang, China.,Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, China
| | - Mingyi Ju
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China.,Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, China Medical University, Shenyang, China.,Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, China Medical University, Shenyang, China.,Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, China
| | - Lin Wang
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China.,Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, China Medical University, Shenyang, China.,Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, China Medical University, Shenyang, China.,Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, China
| | - Qian Wei
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China.,Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, China Medical University, Shenyang, China.,Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, China Medical University, Shenyang, China.,Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, China
| | - Lin Zhao
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China.,Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, China Medical University, Shenyang, China.,Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, China Medical University, Shenyang, China.,Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, China
| | - Minjie Wei
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China.,Shenyang Kangwei Medical Laboratory Analysis Co. LTD., Shenyang, China
| |
Collapse
|
1429
|
Huang W, Xing Y, Zhu L, Zhuo J, Cai M. Sorafenib derivatives-functionalized gold nanoparticles confer protection against tumor angiogenesis and proliferation via suppression of EGFR and VEGFR-2. Exp Cell Res 2021; 406:112633. [PMID: 34089726 DOI: 10.1016/j.yexcr.2021.112633] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 03/25/2021] [Accepted: 04/30/2021] [Indexed: 11/29/2022]
Abstract
Sorafenib is a multi-kinase inhibitor that has been highlighted as a tumor suppressor due to its anti-proliferative and anti-angiogenic properties, whereas the clinical application of Sorafenib is restricted by the side effects it may cause. The past decade has witnessed the development of a series of sorafenib derivatives to improve the clinical performance of sorafenib. Gold nanoparticles (AuNPs) have been widely utilized in drug delivery systems due to their unique properties, including biocompatible nature, simple preparation, and easy surface modification. Herein, this study is aimed to investigate the anti-tumor effects of new sorafenib derivatives-capped gold nanoparticles (AuNPs-New Sor) in tumor formation and metastasis as well as the underlying mechanisms. Initially, new sorafenib derivatives were constructed and combined with AuNPs to form AuNPs-New Sor, and the properties of synthesized AuNPs-New Sor were identified in a mouse model of tumorigenesis. The effect of AuNPs-New Sor on tumor vascular normalization was investigated by assessing vascular permeability and perfusion rate. Next, we evaluated the effect of AuNPs-New Sor on migration and viability of tumor cells and human umbilical vein endothelial cells (HUVECs) as well as on HUVEC angiogenesis in vitro. A melanoma mouse model was further established for in vivo substantiation of the anti-tumor effect of AuNPs-New Sor. According to the results, AuNPs could deliver new sorafenib derivatives into tumor tissues and downregulate the expression of epidermal growth factor receptor (EGFR) and vascular endothelial growth factor receptor-2 (VEGFR-2), thereby suppressing tumor migration, EMT, and angiogenesis in vitro. In addition, AuNPs-New Sor displayed competitive anti-tumor activities in vivo. Taken together, AuNPs-New Sor may attenuate tumor development and angiogenesis through downregulation of EGFR and VEGFR-2.
Collapse
Affiliation(s)
- Wei Huang
- Department of Urology Surgery, Chinese Medical Hospital of Hainan Province, Haikou, 570203, PR China
| | - Yitao Xing
- Department of Urology Surgery, Chinese Medical Hospital of Hainan Province, Haikou, 570203, PR China
| | - Lile Zhu
- Department of Respiratory, Chinese Medical Hospital of Hainan Province, Haikou, 570203, PR China
| | - Jinsheng Zhuo
- Department of Gastroenterology, Chinese Medical Hospital of Hainan Province, Haikou, 570203, PR China
| | - Min Cai
- Department of Urology Surgery, Chinese Medical Hospital of Hainan Province, Haikou, 570203, PR China.
| |
Collapse
|
1430
|
Huang Z, Su Q, Li W, Ren H, Huang H, Wang A. MCTS1 promotes invasion and metastasis of oral cancer by modifying the EMT process. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:997. [PMID: 34277797 PMCID: PMC8267330 DOI: 10.21037/atm-21-2361] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Accepted: 05/28/2021] [Indexed: 12/12/2022]
Abstract
Background The oncogene, malignant T-cell-amplified sequence 1 (MCTS1), has been found to be highly expressed in a variety of cancer cell lines. It has been shown to be involved in cell cycle progression and to confer a growth advantage for lymphomas and breast cancer. Nevertheless, the role of MCTS1 in contributing to the development of oral cancer remains elusive. Methods We analyzed the gene expression profiles of MCTS1 in normal oral keratinocytes and cancerous cells. Cellular proliferation, invasion, and migration experiments were performed to detect the effect of MCTS1 on the biological evolution of oral cancer. The in vitro results were verified by the in vivo lymphatic metastasis test. The underlying mechanism of MCTS1 in promoting oral cancer invasion and metastasis correlated with the epithelial-mesenchymal transition (EMT) process as revealed by western blotting. Results The results showed that MCTS1 was aberrantly expressed in oral cancer cells. MCTS1 overexpression significantly promoted tumor cell growth, proliferation, migration, and invasion. MCTS1-mediated lymphatic metastasis was verified in vivo using an intraplantar tumor model. Biomarkers associated with EMT progression were positively or negatively regulated upon knockdown or overexpression of MCTS1, respectively. Conclusions Higher MCTS1 expression in oral cancer may be connected with an unfavorable prognosis due to involvement of MCTS1. MCTS1 potentiates the growth and proliferation of oral cancer cells and subsequent metastasis by regulating cell cycle and modifying the EMT process. Keywords Oral cancer; oncogene; malignant T-cell-amplified sequence 1 (MCTS1); metastasis; invasion.
Collapse
Affiliation(s)
- Zhexun Huang
- Department of Oral and Maxillofacial Surgery, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Qiao Su
- Animal Experiment Center, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Wuguo Li
- Animal Experiment Center, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Hui Ren
- Department of Oral and Maxillofacial Surgery, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Huiqiang Huang
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China.,State Key Laboratory of Oncology in South China, Guangzhou, China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Anxun Wang
- Department of Oral and Maxillofacial Surgery, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
1431
|
Feng X, Xu ES. Alectinib and lorlatinib function by modulating EMT-related proteins and MMPs in NSCLC metastasis. Bosn J Basic Med Sci 2021; 21:331-338. [PMID: 33091333 PMCID: PMC8112553 DOI: 10.17305/bjbms.2020.5066] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 10/15/2020] [Indexed: 12/25/2022] Open
Abstract
Most advanced non-small cell lung cancer (NSCLC) patients are accompanied by brain metastasis which is the major cause of increased mortality. The fusion rearrangement of anaplastic lymphoma kinase (ALK) gene is an important feature of brain metastasis in lung cancer. The novel ALK inhibitors alectinib and lorlatinib are shown to be effective against NSCLC brain metastasis, while their underlying mechanism of action is unclear. Epithelial–mesenchymal transition (EMT) proteins and matrix metalloproteinases (MMPs) play important roles in brain metastasis by regulating the blood-brain barrier (BBB). To reveal the molecular function of alectinib and lorlatinib, we explored their effects on the cellular levels of EMT markers: VIM and FN1 and the matrix metalloproteinases MMP-9 and MMP-7. The mRNA and protein levels of VIM, FN1, MMP-9, and MMP-7 were elevated in H3122 cells. However, upon alectinib and lorlatinib treatment, the levels were significantly reduced. Similar results were obtained when these experiments were performed either in a dose-dependent or time-dependent manner. Furthermore, alectinib and lorlatinib also inhibited the cell viability and migration of H3122 cells. Interestingly, in comparison to individual drugs, the combination of alectinib and lorlatinib was found to be substantially more effective. Overall, these results suggest that alectinib and lorlatinib possibly function through the downregulation of MMPs and EMT in NSCLC metastasis.
Collapse
Affiliation(s)
- Xu Feng
- Department of Neurosurgery, The First Affiliated Hospital of Jinzhou Medical University 2 Section 5, Liaoning, China
| | - En-Shi Xu
- Department of General Surgery, The First Affiliated Hospital of Jinzhou Medical University 2 Section 5, Liaoning, China
| |
Collapse
|
1432
|
Lee JE, Kang YW, Jung KH, Son MK, Shin SM, Kim JS, Kim SJ, Fang Z, Yan HH, Park JH, Yoon YC, Han B, Cheon MJ, Woo MG, Seo MS, Lim JH, Kim YS, Hong SS. Intracellular KRAS-specific antibody enhances the anti-tumor efficacy of gemcitabine in pancreatic cancer by inducing endosomal escape. Cancer Lett 2021; 507:97-111. [PMID: 33744388 DOI: 10.1016/j.canlet.2021.03.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 02/16/2021] [Accepted: 03/11/2021] [Indexed: 02/07/2023]
Abstract
KRAS mutation is associated with the progression and growth of pancreatic cancer and contributes to chemo-resistance, which poses a significant clinical challenge in pancreatic cancer. Here, we developed a RT22-ep59 antibody (Ab) that directly targets the intracellularly activated GTP-bound form of oncogenic KRAS mutants after it is internalized into cytosol by endocytosis through tumor-associated receptor of extracellular epithelial cell adhesion molecule (EpCAM) and investigated its synergistic anticancer effects in the presence of gemcitabine in pancreatic cancer. We first observed that RT22-ep59 specifically recognized tumor-associated EpCAM and reached the cytosol by endosomal escape. In addition, the anticancer effect of RT22-ep59 was observed in the high-EpCAM-expressing pancreatic cancer cells and gemcitabine-resistant pancreatic cancer cells, but it had little effect on the low-EpCAM-expressing pancreatic cancer cells. Additionally, co-treatment with RT22-ep59 and gemcitabine synergistically inhibited cell viability, migration, and invasion in 3D-cultures and exhibited synergistic anticancer activity by inhibiting the RAF/ERK or PI3K/AKT pathways in cells with high-EpCAM expression. In an orthotopic mouse model, combined administration of RT22-ep59 and gemcitabine significantly inhibited tumor growth. Furthermore, the co-treatment suppressed cancer metastasis by blocking EMT signaling in vitro and in vivo. Our results demonstrated that RT22-ep59 synergistically increased the antitumor activity of gemcitabine by inhibiting RAS signaling by specifically targeting KRAS. This indicates that co-treatment with RT22-ep59 and gemcitabine might be considered a potential therapeutic strategy for pancreatic cancer patients harboring KRAS mutation.
Collapse
Affiliation(s)
- Ji Eun Lee
- Department of Medicine, College of Medicine and Program in Biomedical Science & Engineering, Inha University, 3-ga, Sinheung-dong, Jung-gu, Incheon, 400-712, Republic of Korea
| | - Yeo Wool Kang
- Department of Medicine, College of Medicine and Program in Biomedical Science & Engineering, Inha University, 3-ga, Sinheung-dong, Jung-gu, Incheon, 400-712, Republic of Korea
| | - Kyung Hee Jung
- Department of Medicine, College of Medicine and Program in Biomedical Science & Engineering, Inha University, 3-ga, Sinheung-dong, Jung-gu, Incheon, 400-712, Republic of Korea
| | - Mi Kwon Son
- Department of Medicine, College of Medicine and Program in Biomedical Science & Engineering, Inha University, 3-ga, Sinheung-dong, Jung-gu, Incheon, 400-712, Republic of Korea
| | - Seung-Min Shin
- Department of Molecular Science and Technology, Ajou University, Suwon, 16499, Republic of Korea
| | - Ji-Sun Kim
- Department of Molecular Science and Technology, Ajou University, Suwon, 16499, Republic of Korea
| | - Soo Jung Kim
- Department of Medicine, College of Medicine and Program in Biomedical Science & Engineering, Inha University, 3-ga, Sinheung-dong, Jung-gu, Incheon, 400-712, Republic of Korea
| | - Zhenghuan Fang
- Department of Medicine, College of Medicine and Program in Biomedical Science & Engineering, Inha University, 3-ga, Sinheung-dong, Jung-gu, Incheon, 400-712, Republic of Korea
| | - Hong Hua Yan
- Department of Medicine, College of Medicine and Program in Biomedical Science & Engineering, Inha University, 3-ga, Sinheung-dong, Jung-gu, Incheon, 400-712, Republic of Korea
| | - Jung Hee Park
- Department of Medicine, College of Medicine and Program in Biomedical Science & Engineering, Inha University, 3-ga, Sinheung-dong, Jung-gu, Incheon, 400-712, Republic of Korea
| | - Young-Chan Yoon
- Department of Medicine, College of Medicine and Program in Biomedical Science & Engineering, Inha University, 3-ga, Sinheung-dong, Jung-gu, Incheon, 400-712, Republic of Korea
| | - Boreum Han
- Department of Medicine, College of Medicine and Program in Biomedical Science & Engineering, Inha University, 3-ga, Sinheung-dong, Jung-gu, Incheon, 400-712, Republic of Korea
| | - Min Ji Cheon
- Department of Medicine, College of Medicine and Program in Biomedical Science & Engineering, Inha University, 3-ga, Sinheung-dong, Jung-gu, Incheon, 400-712, Republic of Korea
| | - Min Gyu Woo
- Department of Medicine, College of Medicine and Program in Biomedical Science & Engineering, Inha University, 3-ga, Sinheung-dong, Jung-gu, Incheon, 400-712, Republic of Korea
| | - Myung Sung Seo
- Department of Medicine, College of Medicine and Program in Biomedical Science & Engineering, Inha University, 3-ga, Sinheung-dong, Jung-gu, Incheon, 400-712, Republic of Korea
| | - Joo Han Lim
- Department of Medicine, College of Medicine and Program in Biomedical Science & Engineering, Inha University, 3-ga, Sinheung-dong, Jung-gu, Incheon, 400-712, Republic of Korea
| | - Yong-Sung Kim
- Department of Molecular Science and Technology, Ajou University, Suwon, 16499, Republic of Korea.
| | - Soon-Sun Hong
- Department of Medicine, College of Medicine and Program in Biomedical Science & Engineering, Inha University, 3-ga, Sinheung-dong, Jung-gu, Incheon, 400-712, Republic of Korea.
| |
Collapse
|
1433
|
Gonçalves CFL, Hecht F, Cazarin J, Fortunato RS, Vaisman M, Carvalho DPD, Ferreira ACF. The flavonoid quercetin reduces cell migration and increases NIS and E-cadherin mRNA in the human thyroid cancer cell line BCPAP. Mol Cell Endocrinol 2021; 529:111266. [PMID: 33831503 DOI: 10.1016/j.mce.2021.111266] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2020] [Revised: 03/26/2021] [Accepted: 03/26/2021] [Indexed: 01/06/2023]
Abstract
Thyroid cancer is the most frequent cancer of the endocrine system. Most patients are treated with thyroidectomy followed by radioiodine therapy. However, in part of the patients, a reduction of the sodium-iodide symporter (NIS) occurs, rendering radioiodine therapy ineffective. Moreover, epithelial-mesenchymal transition (EMT) may occur, leading to more aggressive and invasive features. Herein, we evaluated the effect of the flavonoid quercetin on EMT and NIS expression in BCPAP, a papillary thyroid carcinoma cell line. BCPAP was treated with 100 μM quercetin for 24 h and cell viability, apoptosis, EMT markers and NIS were evaluated. Quercetin decreased cell viability by enhancing apoptosis. The flavonoid also reduced matrix metalloproteinase 9 and increased E-cadherin mRNA levels, inhibiting BCPAP adhesion and migration. Additionally, quercetin increased NIS expression and function. Thus, our results suggest that quercetin could be useful as adjuvant in thyroid cancer therapy, inducing apoptosis, reducing invasion and increasing the efficacy of radioiodine therapy.
Collapse
Affiliation(s)
- Carlos Frederico Lima Gonçalves
- Laboratório de Fisiologia Endócrina Doris Rosenthal, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, 21941-902, Brazil.
| | - Fabio Hecht
- Laboratório de Fisiologia Endócrina Doris Rosenthal, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, 21941-902, Brazil.
| | - Juliana Cazarin
- Laboratório de Fisiologia Endócrina Doris Rosenthal, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, 21941-902, Brazil.
| | - Rodrigo Soares Fortunato
- Laboratório de Fisiologia e Sinalização Redox, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, 21941-902, Brazil.
| | - Mario Vaisman
- Serviço de Endocrinologia do Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro (HUCFF/UFRJ), Rio de Janeiro, RJ, Brazil.
| | - Denise Pires de Carvalho
- Laboratório de Fisiologia Endócrina Doris Rosenthal, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, 21941-902, Brazil.
| | - Andrea Claudia Freitas Ferreira
- Laboratório de Fisiologia Endócrina Doris Rosenthal, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, 21941-902, Brazil; NUMPEX, Campus Duque de Caxias, Universidade Federal do Rio de Janeiro, Brazil.
| |
Collapse
|
1434
|
Bannerman D, Pascual-Gil S, Floryan M, Radisic M. Bioengineering strategies to control epithelial-to-mesenchymal transition for studies of cardiac development and disease. APL Bioeng 2021; 5:021504. [PMID: 33948525 PMCID: PMC8068500 DOI: 10.1063/5.0033710] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Accepted: 03/15/2021] [Indexed: 12/24/2022] Open
Abstract
Epithelial-to-mesenchymal transition (EMT) is a process that occurs in a wide range of tissues and environments, in response to numerous factors and conditions, and plays a critical role in development, disease, and regeneration. The process involves epithelia transitioning into a mobile state and becoming mesenchymal cells. The investigation of EMT processes has been important for understanding developmental biology and disease progression, enabling the advancement of treatment approaches for a variety of disorders such as cancer and myocardial infarction. More recently, tissue engineering efforts have also recognized the importance of controlling the EMT process. In this review, we provide an overview of the EMT process and the signaling pathways and factors that control it, followed by a discussion of bioengineering strategies to control EMT. Important biological, biomaterial, biochemical, and physical factors and properties that have been utilized to control EMT are described, as well as the studies that have investigated the modulation of EMT in tissue engineering and regenerative approaches in vivo, with a specific focus on the heart. Novel tools that can be used to characterize and assess EMT are discussed and finally, we close with a perspective on new bioengineering methods that have the potential to transform our ability to control EMT, ultimately leading to new therapies.
Collapse
|
1435
|
Xiao B, Liu L, Chen Z, Li A, Wang P, Xiang C, Zeng Y, Li H, Xiao T. Identification of Epithelial-Mesenchymal Transition-Related Prognostic lncRNAs Biomarkers Associated With Melanoma Microenvironment. Front Cell Dev Biol 2021; 9:679133. [PMID: 34136488 PMCID: PMC8201078 DOI: 10.3389/fcell.2021.679133] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 05/07/2021] [Indexed: 12/11/2022] Open
Abstract
Melanoma is the most common cancer of the skin, associated with a worse prognosis and distant metastasis. Epithelial–mesenchymal transition (EMT) is a reversible cellular biological process that plays significant roles in diverse tumor functions, and it is modulated by specific genes and transcription factors. The relevance of EMT-related lncRNAs in melanoma has not been determined. Therefore, RNA expression data and clinical features were collected from the TCGA database (N = 447). Melanoma samples were randomly assigned into the training (315) and testing sets (132). An EMT-related lncRNA signature was constructed via comprehensive analyses of lncRNA expression level and corresponding clinical data. The Kaplan-Meier analysis showed significant differences in overall survival in patients with melanoma in the low and high-risk groups in two sets. Receiver operating characteristic (ROC) curves were used to measure the performance of the model. Cox regression analysis indicated that the risk score was an independent prognostic factor in two sets. Besides, a nomogram was constructed based on the independent variables. Gene Set Enrichment Analysis (GSEA) was applied to evaluate the potential biological functions in the two risk groups. Furthermore, the melanoma microenvironment was evaluated using ESTIMATE and CIBERSORT algorithms in the risk groups. This study indicates that EMT-related lncRNAs can function as potential independent prognostic biomarkers for melanoma survival.
Collapse
Affiliation(s)
- Bo Xiao
- Department of Orthopedics, Second Xiangya Hospital, Central South University, Changsha, China.,Orthopedic Biomedical Materials Engineering Laboratory of Hunan Province, Changsha, China
| | - Liyan Liu
- Department of Orthopedics, Second Xiangya Hospital, Central South University, Changsha, China.,Orthopedic Biomedical Materials Engineering Laboratory of Hunan Province, Changsha, China
| | - Zhuoyuan Chen
- Department of Orthopedics, Second Xiangya Hospital, Central South University, Changsha, China.,Orthopedic Biomedical Materials Engineering Laboratory of Hunan Province, Changsha, China
| | - Aoyu Li
- Department of Orthopedics, Second Xiangya Hospital, Central South University, Changsha, China.,Orthopedic Biomedical Materials Engineering Laboratory of Hunan Province, Changsha, China
| | - Pingxiao Wang
- Department of Orthopedics, Second Xiangya Hospital, Central South University, Changsha, China.,Orthopedic Biomedical Materials Engineering Laboratory of Hunan Province, Changsha, China
| | - Cheng Xiang
- Department of Orthopedics, Second Xiangya Hospital, Central South University, Changsha, China.,Orthopedic Biomedical Materials Engineering Laboratory of Hunan Province, Changsha, China
| | - Yi Zeng
- Department of Orthopedics, Second Xiangya Hospital, Central South University, Changsha, China.,Orthopedic Biomedical Materials Engineering Laboratory of Hunan Province, Changsha, China
| | - Hui Li
- Department of Orthopedics, Second Xiangya Hospital, Central South University, Changsha, China.,Orthopedic Biomedical Materials Engineering Laboratory of Hunan Province, Changsha, China
| | - Tao Xiao
- Department of Orthopedics, Second Xiangya Hospital, Central South University, Changsha, China.,Orthopedic Biomedical Materials Engineering Laboratory of Hunan Province, Changsha, China
| |
Collapse
|
1436
|
Han B, Ge Y, Cui J, Liu B. Down-regulation of lncRNA DNAJC3-AS1 inhibits colon cancer via regulating miR-214-3p/LIVIN axis. Bioengineered 2021; 11:524-535. [PMID: 32352854 PMCID: PMC7202691 DOI: 10.1080/21655979.2020.1757224] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Long non-coding RNAs (lncRNAs) play a key role in the development and metastasis of cancer. However, the biological role and clinical significance of lncRNA DNAJC3-AS1 in the development of colon cancer is still unknown. In this study, the effects of DNAJC3-AS1 on cell proliferation, migration, and invasion were evaluated by MTT assay, wound-healing assay, and transwell assay, respectively. The relationship between DNAJC3-AS1, miR-214-3p and LIVIN was predicted by the online software and confirmed by dual-luciferase reporter assay. We found that the down-regulation of DNAJC3-AS1 inhibited the proliferation of colon cancer cells and induced growth arrest. Down-regulation of DNAJC3-AS1 also inhibited the migration, invasion, and epithelial-mesenchymal transition (EMT) of colon cancer cells. Moreover, miR-214-3p can bind to DNAJC3-AS1, and knockdown of DNAJC3-AS1 increased miR-214-3p expression in colon cancer cells. LIVIN was identified as a target of miR-214-3p. The up-regulation of miR-214-3p inhibited the protein expression of LIVIN and suppressed the activation of the NF-κB signaling pathway. Besides, down-regulation of DNAJC3-AS1 reduced cell viability, invasion, and EMT of colon cancer cells, while miR-214-3p inhibitor could reverse these effects. The expression of LIVIN and the activation of the NF-κB signaling pathway were suppressed by down-regulating DNAJC3-AS1, while these effects could be restored by miR-214-3p inhibitor. These findings suggested that DNAJC3-AS1 may promote colon cancer progression by regulating the miR-214-3p/LIVIN axis. DNAJC3-AS1 may serve as a new biomarker and therapeutic target for colon cancer, stimulating new research directions and treatment options.
Collapse
Affiliation(s)
- Bing Han
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Yang Ge
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Junpeng Cui
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Baolin Liu
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| |
Collapse
|
1437
|
Jia D, Xing Y, Zhan Y, Cao M, Tian F, Fan W, Huang J, Cui Y, Gu R, Cui Y, Liu Y, Zhang S, Cai L, Li X. LINC02678 as a Novel Prognostic Marker Promotes Aggressive Non-small-cell Lung Cancer. Front Cell Dev Biol 2021; 9:686975. [PMID: 34124072 PMCID: PMC8194704 DOI: 10.3389/fcell.2021.686975] [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: 03/28/2021] [Accepted: 05/07/2021] [Indexed: 12/24/2022] Open
Abstract
Non-small-cell lung carcinoma (NSCLC) is considered to be a fatal disease and characterized by a poor prognosis. Long non-coding RNAs (lncRNAs) have been reported to act as biomarkers and therapeutic targets in solid tumors. However, the expression of lncRNAs and their clinical relevance in NSCLC remain undetermined. The gene expression data profiled in The Cancer Genome Atlas and Gene Expression Omnibus (GSE81089) were employed to screen differentially expressed lncRNAs in NSCLC. LINC02678 was found to be upregulated in NSCLC and exhibited hypomethylation of the promoter region in NSCLC tissues. LINC02678 (also called RP11-336A10.5) was associated with poorer overall survival and relapse-free survival in NSCLC patients. In vitro models of gain- and loss-of-function demonstrated that LINC02678 promotes NSCLC progression by promoting NSCLC cell proliferation and cell cycle progression, as well as inducing NSCLC cell migration, invasion and epithelial-mesenchymal transition. LINC02678 was primarily located in the nucleus and could bind with the enhancer of zeste homolog 2 (EZH2). Moreover, we found that LINC02678 knockdown impaired the occupancy capacity of EZH2 and trimethylation of lysine 27 on histone 3 (H3K27me3) at the promoter region of cyclin dependent kinase inhibitor 1B (CDKN1B) and E-cadherin, as confirmed by ChIP-qPCR. A mouse transplantation model further demonstrated that LINC02678 could promote the tumorigenic and metastatic capacities of NSCLC cells. We identified LINC02678 as a tumor promoter in NSCLC, which enhanced the growth and metastasis of NSCLC cells by binding with EZH2, indicating that LINC02678 may serve as a potential biomarker for cancer diagnosis and treatment.
Collapse
Affiliation(s)
- Dexin Jia
- The Fourth Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Ying Xing
- The Fourth Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yuning Zhan
- The Fourth Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Mengru Cao
- The Fourth Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Fanglin Tian
- The Fourth Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Weina Fan
- The Fourth Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Jian Huang
- The Fourth Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yimeng Cui
- The Fourth Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Ruixue Gu
- The Fourth Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yaowen Cui
- The Fourth Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yuechao Liu
- The Fourth Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Shuai Zhang
- The Fourth Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Li Cai
- The Fourth Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Xiaomei Li
- Department of Pathology, Harbin Medical University Cancer Hospital, Harbin, China
| |
Collapse
|
1438
|
Zhang S, Li J, Gao H, Tong Y, Li P, Wang Y, Du L, Wang C. lncRNA Profiles Enable Prognosis Prediction and Subtyping for Esophageal Squamous Cell Carcinoma. Front Cell Dev Biol 2021; 9:656554. [PMID: 34127945 PMCID: PMC8196240 DOI: 10.3389/fcell.2021.656554] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 04/08/2021] [Indexed: 12/24/2022] Open
Abstract
Long non-coding RNAs (lncRNAs) have emerged as useful prognostic markers in many tumors. In this study, we investigated the potential application of lncRNA markers for the prognostic prediction of esophageal squamous cell carcinoma (ESCC). We identified ESCC-associated lncRNAs by comparing ESCC tissues with normal tissues. Subsequently, Kaplan–Meier (KM) method in combination with the univariate Cox proportional hazards regression (UniCox) method was used to screen prognostic lncRNAs. By combining the differential and prognostic lncRNAs, we developed a prognostic model using cox stepwise regression analysis. The obtained prognostic prediction model could effectively predict the 3- and 5-year prognosis and survival of ESCC patients by time-dependent receiver operating characteristic (ROC) curves (area under curve = 0.87 and 0.89, respectively). Besides, a lncRNA-based classification of ESCC was generated using k-mean clustering method and we obtained two clusters of ESCC patients with association with race and Barrett’s esophagus (BE) (both P < 0.001). Finally, we found that lncRNA AC007128.1 was upregulated in both ESCC cells and tissues and associated with poor prognosis of ESCC patients. Furthermore, AC007128.1 could promote epithelial-mesenchymal transition (EMT) of ESCC cells by increasing the activation of MAPK/ERK and MAPK/p38 signaling pathways. Collectively, our findings indicated the potentials of lncRNA markers in the prognosis, molecular subtyping, and EMT of ESCC.
Collapse
Affiliation(s)
- Shujun Zhang
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Juan Li
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Huiru Gao
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yao Tong
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Peilong Li
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yunshan Wang
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Lutao Du
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Shandong Engineering & Technology Research Center for Tumor Marker Detection, Jinan, China.,Shandong Provincial Clinical Medicine Research Center for Clinical Laboratory, Jinan, China
| | - Chuanxin Wang
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Shandong Engineering & Technology Research Center for Tumor Marker Detection, Jinan, China.,Shandong Provincial Clinical Medicine Research Center for Clinical Laboratory, Jinan, China
| |
Collapse
|
1439
|
Guan Y, Du Y, Wang G, Gou H, Xue Y, Xu J, Li E, Chan DW, Wu D, Xu P, Ni P, Xu D, Hu Y. Overexpression of PLXDC2 in Stromal Cell-Associated M2 Macrophages Is Related to EMT and the Progression of Gastric Cancer. Front Cell Dev Biol 2021; 9:673295. [PMID: 34124056 PMCID: PMC8194078 DOI: 10.3389/fcell.2021.673295] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Accepted: 05/04/2021] [Indexed: 12/14/2022] Open
Abstract
The tumor microenvironment (TME) comprises distinct cell types, including stromal types such as fibroblast cells and macrophage cells, which have recently become a critical factor in tumor development and progression. Here, we identified the TME-related gene, plexin domain containing 2 (PLXDC2), in a high-stromal-score population. And we revealed that this gene was related to poor survival and advanced (tumor-node-metastasis) stage in gastric cancer (GC) patients from The Cancer Genome Atlas database. An integrated gene profile and functional analysis of the proportions of tumor-infiltrating immune cells revealed that the expression of the M2 macrophages cell marker CD163 was positively correlated with PLXDC2 expression. In addition, the M2 macrophages gene signature and high PLXDC2 expression were associated with the inflammatory signaling pathway and the epithelial-to-mesenchymal transition (EMT)-related gene signature. Single-cell study of GC identified PLXDC2 was enriched specifically in fibroblasts and monocytes/macrophages populations, which supported its important role in the stroma. Furthermore, according to a tissue microarray immunohistochemistry analysis, the expression of PLXDC2 elevated in human GC stromal specimens compared to tumor tissue specimens. Moreover, PLXDC2 overexpression in the stromal compartment was associated with CD163-positive regulatory M2 macrophages, and its functions were related to the pathogenesis of GC. Multiplexed immunohistochemistry verified PLXDC2's correlation with EMT markers. Our data suggested that PLXDC2 was expressed in stromal cells and that its crosstalk with tumor-associated macrophages could contribute to cancer biology by inducing the EMT process.
Collapse
Affiliation(s)
- Yiming Guan
- Faculty of Medical Laboratory Science, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yuzhang Du
- Faculty of Medical Laboratory Science, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Guanzheng Wang
- Faculty of Medical Laboratory Science, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Hongquan Gou
- Faculty of Medical Laboratory Science, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yilun Xue
- Faculty of Medical Laboratory Science, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jingsong Xu
- Faculty of Medical Laboratory Science, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Enhao Li
- Faculty of Medical Laboratory Science, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - David W Chan
- Department of Obstetrics and Gynaecology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Di Wu
- Faculty of Medical Laboratory Science, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Peiqing Xu
- Faculty of Medical Laboratory Science, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Peihua Ni
- Faculty of Medical Laboratory Science, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Dakang Xu
- Faculty of Medical Laboratory Science, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yiqun Hu
- Faculty of Medical Laboratory Science, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
1440
|
Pattarawat P, Hunt JT, Poloway J, Archibald CJ, Wang HCR. A triple combination gemcitabine + romidepsin + cisplatin to effectively control triple-negative breast cancer tumor development, recurrence, and metastasis. Cancer Chemother Pharmacol 2021; 88:415-425. [PMID: 34043046 DOI: 10.1007/s00280-021-04298-y] [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: 12/15/2020] [Accepted: 05/15/2021] [Indexed: 12/14/2022]
Abstract
PURPOSE Triple-negative breast cancer (TNBC) is an aggressive, lethal, heterogeneous type of breast cancer (BC). TNBC tends to have a lower response rate to chemotherapy and a lower 5-year survival rate than other types of BC due to recurrence and metastasis. Our previous study revealed that a combination of gemcitabine, romidepsin, and cisplatin was efficacious in controlling TNBC tumor development. In this study, we extended our investigation of gemcitabine + romidepsin + cisplatin in controlling TNBC tumor recurrence and metastasis. METHODS We investigated the ability of gemcitabine + romidepsin + cisplatin to control cell survival and invasiveness using cell viability, soft agar colony formation, and transwell invasion assays. We determined the efficacy of gemcitabine + romidepsin + cisplatin in controlling tumor recurrence and metastasis using cell-derived xenograft animal models. We used immunoblotting to study signaling modulators regulated by gemcitabine + romidepsin + cisplatin in TNBC cells and tumor tissues. RESULTS Treatment with gemcitabine + romidepsin + cisplatin reduced the TNBC MDA-MB231 and MDA-MB468 cell survival to ~ 50% and ~ 15%, as well as invasiveness to ~ 31% and ~ 13%, respectively. Gemcitabine + romidepsin + cisplatin suppressed modulators involved in epithelial-mesenchymal transition in an ROS-dependent manner. Controlling tumor recurrence, the Gem plus Rom + Cis regimen (~ 112%) was more efficacious than the Gem plus Cis regimen (~ 21%) in tumor growth inhibition. The Gem plus Rom + Cis regimen efficaciously reduced the development of metastatic nodules to 20% in animals. CONCLUSION The gemcitabine plus romidepsin + cisplatin regimen was highly efficacious in controlling TNBC tumor development, recurrence, and metastasis in animals. The combination regimen should be poised for efficient translation into clinical trials for controlling the recurrence and metastasis, ultimately contributing to reducing mortality and improving TNBC patients' quality of life.
Collapse
Affiliation(s)
- Pawat Pattarawat
- Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, 2407 River Drive, Knoxville, TN, 37996, USA
| | - Jessica T Hunt
- Animal Resource Laboratory, College of Veterinary Medicine, University of Tennessee, 2407 River Drive, Knoxville, TN, 37996, USA
| | - Jacob Poloway
- Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, 2407 River Drive, Knoxville, TN, 37996, USA
| | - Collin J Archibald
- Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, 2407 River Drive, Knoxville, TN, 37996, USA
| | - Hwa-Chain Robert Wang
- Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, 2407 River Drive, Knoxville, TN, 37996, USA.
| |
Collapse
|
1441
|
Control of Tumor Progression by Angiocrine Factors. Cancers (Basel) 2021; 13:cancers13112610. [PMID: 34073394 PMCID: PMC8198241 DOI: 10.3390/cancers13112610] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 05/21/2021] [Accepted: 05/21/2021] [Indexed: 12/24/2022] Open
Abstract
Tumor progression, therapy resistance and metastasis are profoundly controlled by the tumor microenvironment. The contribution of endothelial cells to tumor progression was initially only attributed to the formation of new blood vessels (angiogenesis). Research in the last decade has revealed however that endothelial cells control their microenvironment through the expression of membrane-bound and secreted factors. Such angiocrine functions are frequently hijacked by cancer cells, which deregulate the signaling pathways controlling the expression of angiocrine factors. Here, we review the crosstalk between cancer cells and endothelial cells and how this contributes to the cancer stem cell phenotype, epithelial to mesenchymal transition, immunosuppression, remodeling of the extracellular matrix and intravasation of cancer cells into the bloodstream. We also address the long-distance crosstalk of a primary tumor with endothelial cells at the pre-metastatic niche and how this contributes to metastasis.
Collapse
|
1442
|
Abstract
The HOXC10 gene, a member of the HOX genes family, plays crucial roles in mammalian physiological processes, such as limb morphological development, limb regeneration, and lumbar motor neuron differentiation. HOXC10 is also associated with angiogenesis, fat metabolism, and sex regulation. Additional evidence suggests that HOXC10 dysregulation is closely associated with various tumors. HOXC10 is an important transcription factor that can activate several oncogenic pathways by regulating various target molecules such as ERK, AKT, p65, and epithelial mesenchymal transition-related genes. HOXC10 also induces drug resistance in cancers by promoting the DNA repair pathway. In this review, we summarize HOXC10 gene structure and expression as well as the role of HOXC10 in different human cancer processes. This review will provide insight into the status of HOXC10 research and help identify novel targets for cancer therapy.
Collapse
Affiliation(s)
- Jinyong Fang
- Department of Science and Education, Jinhua Guangfu Oncology Hospital, Jinhua, China
| | - Jianjun Wang
- Department of Gastroenterological Surgery, Jinhua Guangfu Oncology Hospital, Jinhua, China
| | - Liangliang Yu
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Medical School of Zhejiang University, Hangzhou, China
| | - Wenxia Xu
- Central Laboratory, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| |
Collapse
|
1443
|
Yao J, Yang Z, Yang J, Wang ZG, Zhang ZY. Long non-coding RNA FEZF1-AS1 promotes the proliferation and metastasis of hepatocellular carcinoma via targeting miR-107/Wnt/β-catenin axis. Aging (Albany NY) 2021; 13:13726-13738. [PMID: 34023817 PMCID: PMC8202841 DOI: 10.18632/aging.202960] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Accepted: 04/06/2021] [Indexed: 12/18/2022]
Abstract
Hepatocellular carcinoma (HCC) is a public health problem around the world, with the molecular mechanisms being still incompletely clear. This study was carried out to explore the role and mechanism of long-noncoding RNA (lncRNA) FEZF1-AS1 in HCC progression. RNA sequencing and quantitative real time polymerase chain reaction (qRT- PCR) were applied to identify differently expressed lncRNAs in HCC tissues and adjacent normal tissues. CCK8 assay was adopted to test cell proliferation and flow cytometry was taken to detect cell apoptosis. Wound healing assay and transwell experiment were performed to determine cell migration and invasion. To validate the function of lncRNA FEZF1-AS1 in vivo, tumor-burdened models were established. The results showed that lncRNA FEZF1-AS1 level was prominently enhanced in HCC tumor specimens and overexpression of FEZF1-AS1 promoted the proliferation, migration and invasion of HCC cells. In mechanism, overexpression of FEZF1-AS1 reduced the expression of miR-107 which inhibited the activation of Wnt/β-catenin signaling. Overexpression of β-catenin promoted cell proliferation, migration and invasion which were inhibited by FEZF1-AS1 downregulation. In conclusion, our study demonstrated that FEZF1-AS1 promoted HCC progression through activating Wnt/β-catenin signaling by targeting miR-107, which provided a novel target for the therapy of HCC.
Collapse
Affiliation(s)
- Jing Yao
- Department of Surgery, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai 200233, China
| | - Zhe Yang
- Department of Surgery, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai 200233, China
| | - Jun Yang
- Department of Surgery, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai 200233, China
| | - Zhi-Gang Wang
- Department of Surgery, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai 200233, China
| | - Zheng-Yun Zhang
- Department of Surgery, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai 200233, China
| |
Collapse
|
1444
|
Lachat C, Peixoto P, Hervouet E. Epithelial to Mesenchymal Transition History: From Embryonic Development to Cancers. Biomolecules 2021; 11:biom11060782. [PMID: 34067395 PMCID: PMC8224685 DOI: 10.3390/biom11060782] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/18/2021] [Accepted: 05/19/2021] [Indexed: 12/11/2022] Open
Abstract
Epithelial to mesenchymal transition (EMT) is a process that allows epithelial cells to progressively acquire a reversible mesenchymal phenotype. Here, we recount the main events in the history of EMT. EMT was first studied during embryonic development. Nowadays, it is an important field in cancer research, studied all around the world by more and more scientists, because it was shown that EMT is involved in cancer aggressiveness in many different ways. The main features of EMT's involvement in embryonic development, fibrosis and cancers are briefly reviewed here.
Collapse
Affiliation(s)
- Camille Lachat
- UMR 1098 RIGHT, University Bourgogne-Franche-Comté, INSERM, EFS-BFC, F-25000 Besançon, France; (P.P.); (E.H.)
- Correspondence:
| | - Paul Peixoto
- UMR 1098 RIGHT, University Bourgogne-Franche-Comté, INSERM, EFS-BFC, F-25000 Besançon, France; (P.P.); (E.H.)
- EPIgenetics and GENe EXPression Technical Platform (EPIGENExp), University Bourgogne Franche-Comté, F-25000 Besançon, France
| | - Eric Hervouet
- UMR 1098 RIGHT, University Bourgogne-Franche-Comté, INSERM, EFS-BFC, F-25000 Besançon, France; (P.P.); (E.H.)
- EPIgenetics and GENe EXPression Technical Platform (EPIGENExp), University Bourgogne Franche-Comté, F-25000 Besançon, France
- DImaCell Platform, University Bourgogne Franche-Comté, F-25000 Besançon, France
| |
Collapse
|
1445
|
Luttman JH, Colemon A, Mayro B, Pendergast AM. Role of the ABL tyrosine kinases in the epithelial-mesenchymal transition and the metastatic cascade. Cell Commun Signal 2021; 19:59. [PMID: 34022881 PMCID: PMC8140471 DOI: 10.1186/s12964-021-00739-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 04/16/2021] [Indexed: 12/20/2022] Open
Abstract
The ABL kinases, ABL1 and ABL2, promote tumor progression and metastasis in various solid tumors. Recent reports have shown that ABL kinases have increased expression and/or activity in solid tumors and that ABL inactivation impairs metastasis. The therapeutic effects of ABL inactivation are due in part to ABL-dependent regulation of diverse cellular processes related to the epithelial to mesenchymal transition and subsequent steps in the metastatic cascade. ABL kinases target multiple signaling pathways required for promoting one or more steps in the metastatic cascade. These findings highlight the potential utility of specific ABL kinase inhibitors as a novel treatment paradigm for patients with advanced metastatic disease. Video abstract.
Collapse
Affiliation(s)
- Jillian Hattaway Luttman
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, 308 Research Drive, C-233A LSRC Bldg., P.O. Box 3813, Durham, NC 27710 USA
| | - Ashley Colemon
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, 308 Research Drive, C-233A LSRC Bldg., P.O. Box 3813, Durham, NC 27710 USA
| | - Benjamin Mayro
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, 308 Research Drive, C-233A LSRC Bldg., P.O. Box 3813, Durham, NC 27710 USA
| | - Ann Marie Pendergast
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, 308 Research Drive, C-233A LSRC Bldg., P.O. Box 3813, Durham, NC 27710 USA
| |
Collapse
|
1446
|
Pontecorvi P, Megiorni F, Camero S, Ceccarelli S, Bernardini L, Capalbo A, Anastasiadou E, Gerini G, Messina E, Perniola G, Benedetti Panici P, Grammatico P, Pizzuti A, Marchese C. Altered Expression of Candidate Genes in Mayer-Rokitansky-Küster-Hauser Syndrome May Influence Vaginal Keratinocytes Biology: A Focus on Protein Kinase X. BIOLOGY 2021; 10:biology10060450. [PMID: 34063745 PMCID: PMC8223793 DOI: 10.3390/biology10060450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 05/13/2021] [Accepted: 05/20/2021] [Indexed: 11/16/2022]
Abstract
Mayer-Rokitansky-Küster-Hauser (MRKH) syndrome is a rare and complex disease defined by congenital aplasia of the vagina and uterus in 46,XX women, often associated with kidney and urinary tract anomalies. The aetiopathogenesis of MRKH syndrome is still largely unknown. Herein, we investigated the role of selected candidate genes in the aetiopathogenesis of MRKH syndrome, with a focus on PRKX, which encodes for protein kinase X. Through RT-qPCR analyses performed on vaginal dimple samples from patients, and principal component analysis (PCA), we highlighted a phenotype-related expression pattern of PRKX, MUC1, HOXC8 and GREB1L in MRKH patients. By using an in vitro approach, we proved that PRKX ectopic overexpression in a cell model of vaginal keratinocytes promotes cell motility through epithelial-to-mesenchymal transition (EMT) activation, a fundamental process in urogenital tract morphogenesis. Moreover, our findings showed that PRKX upregulation in vaginal keratinocytes is able to affect transcriptional levels of HOX genes, implicated in urinary and genital tract development. Our study identified the dysregulation of PRKX expression as a possible molecular cause for MRKH syndrome. Moreover, we propose the specific role of PRKX in vaginal keratinocyte biology as one of the possible mechanisms underlying this complex disease.
Collapse
Affiliation(s)
- Paola Pontecorvi
- Department of Experimental Medicine, Sapienza University of Rome—Viale Regina Elena 324, 00161 Rome, Italy; (P.P.); (F.M.); (S.C.); (E.A.); (G.G.); (E.M.); (A.P.)
| | - Francesca Megiorni
- Department of Experimental Medicine, Sapienza University of Rome—Viale Regina Elena 324, 00161 Rome, Italy; (P.P.); (F.M.); (S.C.); (E.A.); (G.G.); (E.M.); (A.P.)
| | - Simona Camero
- Department of Maternal and Child Health and Urological Sciences, Sapienza University of Rome—Viale Regina Elena 324, 00161 Rome, Italy; (S.C.); (G.P.); (P.B.P.)
| | - Simona Ceccarelli
- Department of Experimental Medicine, Sapienza University of Rome—Viale Regina Elena 324, 00161 Rome, Italy; (P.P.); (F.M.); (S.C.); (E.A.); (G.G.); (E.M.); (A.P.)
| | - Laura Bernardini
- Division of Medical Genetics, IRCCS Casa Sollievo della Sofferenza Foundation-Viale Cappuccini, 1, 71013 San Giovanni Rotondo (FG), Italy; (L.B.); (A.C.)
| | - Anna Capalbo
- Division of Medical Genetics, IRCCS Casa Sollievo della Sofferenza Foundation-Viale Cappuccini, 1, 71013 San Giovanni Rotondo (FG), Italy; (L.B.); (A.C.)
| | - Eleni Anastasiadou
- Department of Experimental Medicine, Sapienza University of Rome—Viale Regina Elena 324, 00161 Rome, Italy; (P.P.); (F.M.); (S.C.); (E.A.); (G.G.); (E.M.); (A.P.)
| | - Giulia Gerini
- Department of Experimental Medicine, Sapienza University of Rome—Viale Regina Elena 324, 00161 Rome, Italy; (P.P.); (F.M.); (S.C.); (E.A.); (G.G.); (E.M.); (A.P.)
| | - Elena Messina
- Department of Experimental Medicine, Sapienza University of Rome—Viale Regina Elena 324, 00161 Rome, Italy; (P.P.); (F.M.); (S.C.); (E.A.); (G.G.); (E.M.); (A.P.)
| | - Giorgia Perniola
- Department of Maternal and Child Health and Urological Sciences, Sapienza University of Rome—Viale Regina Elena 324, 00161 Rome, Italy; (S.C.); (G.P.); (P.B.P.)
| | - Pierluigi Benedetti Panici
- Department of Maternal and Child Health and Urological Sciences, Sapienza University of Rome—Viale Regina Elena 324, 00161 Rome, Italy; (S.C.); (G.P.); (P.B.P.)
| | - Paola Grammatico
- Division of Medical Genetics, Department of Molecular Medicine, Sapienza University of Rome-San Camillo-Forlanini Hospital, Circonvallazione Gianicolense, 87, 00152 Rome, Italy;
| | - Antonio Pizzuti
- Department of Experimental Medicine, Sapienza University of Rome—Viale Regina Elena 324, 00161 Rome, Italy; (P.P.); (F.M.); (S.C.); (E.A.); (G.G.); (E.M.); (A.P.)
- Division of Medical Genetics, IRCCS Casa Sollievo della Sofferenza Foundation-Viale Cappuccini, 1, 71013 San Giovanni Rotondo (FG), Italy; (L.B.); (A.C.)
| | - Cinzia Marchese
- Department of Experimental Medicine, Sapienza University of Rome—Viale Regina Elena 324, 00161 Rome, Italy; (P.P.); (F.M.); (S.C.); (E.A.); (G.G.); (E.M.); (A.P.)
- Correspondence: ; Tel.: +39-06-4997-2872
| |
Collapse
|
1447
|
Tanaka T, Ohishi T, Asano T, Takei J, Nanamiya R, Hosono H, Sano M, Harada H, Kawada M, Kaneko MK, Kato Y. An anti‑TROP2 monoclonal antibody TrMab‑6 exerts antitumor activity in breast cancer mouse xenograft models. Oncol Rep 2021; 46:132. [PMID: 34013368 PMCID: PMC8144932 DOI: 10.3892/or.2021.8083] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Accepted: 04/21/2021] [Indexed: 12/16/2022] Open
Abstract
Trophoblast cell surface antigen 2 (TROP2), reported to be overexpressed in several types of cancer, is involved in cell proliferation, invasion, metastasis, and poor prognosis of many types of cancer. Previously, a highly sensitive anti-TROP2 monoclonal antibody (clone TrMab-6; mouse IgG2b, κ) was developed using a Cell-Based Immunization and Screening (CBIS) method. TrMab-6 was useful for investigations using flow cytometry, western blot, and immunohistochemistry. The aim of the present study was to investigate whether TrMab-6 possesses in vitro antibody-dependent cellular cytotoxicity (ADCC) or complement-dependent cytotoxicity (CDC) activities or in vivo antitumor activities using mouse xenograft models of TROP2-overexpressed CHO-K1 (CHO/TROP2) and breast cancer cell lines, including MCF7, MDA-MB-231, and MDA-MB-468. In vitro experiments revealed that TrMab-6 strongly induced ADCC and CDC activities against CHO/TROP2 and the three breast cancer cell lines, whereas it did not show those activities against parental CHO-K1 and MCF7/TROP2-knockout cells. Furthermore, in vivo experiments on CHO/TROP2 and MCF7 ×enografts revealed that TrMab-6 significantly reduced tumor growth, whereas it did not show antitumor activities against parental CHO-K1 and MCF7/TROP2-knockout xenografts. The findings suggest that TrMab-6 is a promising treatment option for TROP2-expressing breast cancers.
Collapse
Affiliation(s)
- Tomohiro Tanaka
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Aoba‑ku, Sendai, Miyagi 980‑8575, Japan
| | - Tomokazu Ohishi
- Institute of Microbial Chemistry (BIKAKEN), Numazu, Microbial Chemistry Research Foundation, Numazu‑shi, Shizuoka 410‑0301, Japan
| | - Teizo Asano
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Aoba‑ku, Sendai, Miyagi 980‑8575, Japan
| | - Junko Takei
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Aoba‑ku, Sendai, Miyagi 980‑8575, Japan
| | - Ren Nanamiya
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Aoba‑ku, Sendai, Miyagi 980‑8575, Japan
| | - Hideki Hosono
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Aoba‑ku, Sendai, Miyagi 980‑8575, Japan
| | - Masato Sano
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Aoba‑ku, Sendai, Miyagi 980‑8575, Japan
| | - Hiroyuki Harada
- Department of Oral and Maxillofacial Surgery, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Bunkyo‑ku, Tokyo 113‑8510, Japan
| | - Manabu Kawada
- Institute of Microbial Chemistry (BIKAKEN), Numazu, Microbial Chemistry Research Foundation, Numazu‑shi, Shizuoka 410‑0301, Japan
| | - Mika K Kaneko
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Aoba‑ku, Sendai, Miyagi 980‑8575, Japan
| | - Yukinari Kato
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Aoba‑ku, Sendai, Miyagi 980‑8575, Japan
| |
Collapse
|
1448
|
Liang X, Lu J, Wu Z, Guo Y, Shen S, Liang J, Dong Z, Guo W. LINC00239 Interacts with C-Myc Promoter-Binding Protein-1 (MBP-1) to Promote Expression of C-Myc in Esophageal Squamous Cell Carcinoma. Mol Cancer Res 2021; 19:1465-1475. [PMID: 34016746 DOI: 10.1158/1541-7786.mcr-20-1025] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 03/29/2021] [Accepted: 05/14/2021] [Indexed: 11/16/2022]
Abstract
Increasing evidence demonstrates that long non-coding RNAs (lncRNA) play a vital role in the progression of tumors, containing esophageal squamous cell carcinoma (ESCC). LINC00239 was reported as an oncogene in diverse kinds of cancers, whereas its specific role is still unclear in ESCC. In this study, we detected the expression and functional role of LINC00239 in ESCC specimens and cells, and investigated the molecular mechanisms of it. LINC00239 was highly expressed in ESCC tissues and cells, and was related to poor prognosis of patients with ESCC. The proliferation, metastasis, and invasion ability as well as epithelial-mesenchymal transition (EMT) process were all enhanced in LINC00239-overexpressed ESCC cells. LINC00239 was upregulated in TGF-β1-treated ESCC cells. Furthermore, LINC00239 was found to bind directly to the transcription factor c-Myc promoter-binding protein-1 (MBP-1). MBP-1 was detected to inhibit the transcription of c-Myc in ESCC. Moreover, LINC00239 could activate c-Myc transcription through influencing MBP-1-binding ability to c-Myc promoter. These data suggest that LINC00239 may act as an oncogene to promote the transcription of c-Myc by competitively combining with MBP-1 in ESCC, and may serve as a potential target for antitumor therapy in ESCC. IMPLICATIONS: LINC00239 may function as an oncogenic lncRNA in ESCC through the LINC00239/MBP-1/c-Myc axis to activate EMT process.
Collapse
Affiliation(s)
- Xiaoliang Liang
- Laboratory of Pathology, Hebei Cancer Institute, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Juntao Lu
- Laboratory of Pathology, Hebei Cancer Institute, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Zheng Wu
- Laboratory of Pathology, Hebei Cancer Institute, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yanli Guo
- Laboratory of Pathology, Hebei Cancer Institute, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Supeng Shen
- Laboratory of Pathology, Hebei Cancer Institute, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Jia Liang
- Laboratory of Pathology, Hebei Cancer Institute, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Zhiming Dong
- Laboratory of Pathology, Hebei Cancer Institute, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Wei Guo
- Laboratory of Pathology, Hebei Cancer Institute, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China.
| |
Collapse
|
1449
|
Huang D, Chen J, Hu D, Xie F, Yang T, Li Z, Wang X, Xiao Y, Zhong J, Jiang Y, Zhang X, Zhong T. Advances in Biological Function and Clinical Application of Small Extracellular Vesicle Membrane Proteins. Front Oncol 2021; 11:675940. [PMID: 34094979 PMCID: PMC8172959 DOI: 10.3389/fonc.2021.675940] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Accepted: 04/30/2021] [Indexed: 12/15/2022] Open
Abstract
Small extracellular vesicles are membrane-bound vesicles secreted into extracellular spaces by virtually all types of cells. These carry a large number of membrane proteins on their surface that are incorporated during their biogenesis in cells. The composition of the membrane proteins hence bears the signature of the cells from which they originate. Recent studies have suggested that the proteins on these small extracellular vesicles can serve as biomarkers and target proteins for the diagnosis and treatment of diseases. This article classifies small extracellular vesicle membrane proteins and summarizes their pathophysiological functions in the diagnosis and treatment of diseases.
Collapse
Affiliation(s)
- Defa Huang
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, China.,Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Jie Chen
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, China.,Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Die Hu
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, China.,Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Fangfang Xie
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, China.,Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Tong Yang
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, China.,Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Zhengzhe Li
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, China.,Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Xiaoxing Wang
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, China.,Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Yongwei Xiao
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, China.,Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Jianing Zhong
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, China
| | - Yu Jiang
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Xiaokang Zhang
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, China.,Department of Preventive Medicine, Gannan Medical University, Ganzhou, China
| | - Tianyu Zhong
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, China.,Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China.,Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, China.,Precision Medicine Center, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| |
Collapse
|
1450
|
Zhu T, Zou X, Yang C, Li L, Wang B, Li R, Li H, Xu Z, Huang D, Wu Q. Neutrophil extracellular traps promote gastric cancer metastasis by inducing epithelial‑mesenchymal transition. Int J Mol Med 2021; 48:127. [PMID: 34013374 PMCID: PMC8128417 DOI: 10.3892/ijmm.2021.4960] [Citation(s) in RCA: 88] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 06/19/2020] [Indexed: 12/13/2022] Open
Abstract
The risks of tumor recurrence following the successful resection of the primary tumor have been known for decades; however, the precise mechanisms underlying treatment failures remain unknown. The formation of neutrophil extracellular traps (NETs) has increasingly been demonstrated to be associated with thrombi formation in cancer patients, as well as with the development and metastasis of cancer. The present study demonstrated that the level of peripheral blood NETs in patients with gastric cancer (GC) was associated with tumor progression, and patients with stage III/IV disease exhibited significant differences compared with the healthy controls and patients with stage I/II disease, which may be associated with an increased risk of metastasis. In addition, plasma from patients with stage III/IV GC was more prone to stimulate neutrophils to form NETs; thus, it was hypothesized that the formation of NETs may be affected by the tumor microenvironment. A higher deposition of NETs in GC tissues compared with normal resection margins was also identified. In vitro, following treatment with phorbol myristate acetate, which promotes the formation of NETs, or with DNAse-1/GSK-484, which inhibits the formation of NETs, it was found that the tumor migratory ability was altered; however, no significant changes were observed in cell proliferation and cell cycle progression. Epithelial-mesenchymal transition (EMT) is a key event associated with dissemination and metastasis in GC pathogenesis. Finally, the present study demonstrated that NETs promote a more aggressive mesenchymal phenotype and promote the progression of GC in vitro and in vivo. On the whole, to the best of our knowledge, the present study reports a previously unknown role of NETs in the regulation of GC, which is associated with EMT and migration. Therefore, targeting NETs may prove to be therapeutically beneficial.
Collapse
Affiliation(s)
- Tong Zhu
- The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Xiaoming Zou
- The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Chunfa Yang
- Shuangyashan Shuangkuang Hospital, Shuangyashan, Heilongjiang 155100, P.R. China
| | - Liangliang Li
- Department of Pharmacology, Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Bing Wang
- Department of Pharmacology, Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Rong Li
- The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Hongxuan Li
- The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Zhangxuan Xu
- The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Di Huang
- Department of Pharmacology, Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Qingyun Wu
- The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| |
Collapse
|