1
|
Zou R, Lu J, Bai X, Yang Y, Zhang S, Wu S, Tang Z, Li K, Hua X. The epigenetic-modified downregulation of LOXL1 protein mediates EMT in bladder epithelial cells exposed to benzo[a]pyrene and its metabolite BPDE. Int Immunopharmacol 2024; 142:113232. [PMID: 39340995 DOI: 10.1016/j.intimp.2024.113232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 08/30/2024] [Accepted: 09/19/2024] [Indexed: 09/30/2024]
Abstract
Benzo[a]pyrene (B[a]P) is a well-known polycyclic aromatic hydrocarbon (PAH) pollutant with high carcinogenicity, widespread environmental presence, and significant threat to public health. Epidemiological studies have linked exposure to B[a]P and its metabolite 7,8-dihydroxy-9,10-epoxybenzo[a]pyrene (BPDE) to the development and progression of various cancers, including bladder cancer. However, its underlying mechanism remains unclear. Our study revealed that B[a]P and BPDE induced epithelial-mesenchymal transition (EMT), a critical early event in cell malignant transformation, involving a decrease in E-Cadherin and upregulation of N-Cadherin protein levels, leading to increased cell motility and migration in bladder epithelial cells. Further studies have indicated that LOXL1 DNA undergoes methylation and modification influenced by methyltransferase 3a (DNMT3a) and DNMT3b, resulting in decreased LOXL1 protein levels. The decreased LOXL1 promotes the zinc finger transcription factor SLUG, which then inhibits E-Cadherin protein levels and initiates the EMT process. Moreover, DNMT3a/3b expression appears to be influenced by intracellular oxidative stress levels. These findings suggest that exposure to B[a]P/BPDE promotes the EMT process through the pivotal factor LOXL1, thereby contributing to bladder carcinogenesis. Our study provides a theoretical basis for considering LOXL1 as a potential biomarker for early diagnosis and a novel target for the precise diagnosis and treatment of bladder cancer.
Collapse
Affiliation(s)
- Ronghao Zou
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, Hefei, Anhui, China
| | - Juan Lu
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, Hefei, Anhui, China
| | - Xiaoyue Bai
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, Hefei, Anhui, China
| | - Yuyao Yang
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, Hefei, Anhui, China
| | - Shouyue Zhang
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, Hefei, Anhui, China
| | - Shuai Wu
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, Hefei, Anhui, China
| | - Zhixin Tang
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, Hefei, Anhui, China
| | - Kang Li
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, Hefei, Anhui, China
| | - Xiaohui Hua
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, Hefei, Anhui, China.
| |
Collapse
|
2
|
Khan M, Huang X, Ye X, Zhang D, Wang B, Xu A, Li R, Ren A, Chen C, Song J, Zheng R, Yuan Y, Lin J. Necroptosis-based glioblastoma prognostic subtypes: implications for TME remodeling and therapy response. Ann Med 2024; 56:2405079. [PMID: 39387496 PMCID: PMC11469424 DOI: 10.1080/07853890.2024.2405079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 08/14/2024] [Accepted: 08/28/2024] [Indexed: 10/15/2024] Open
Abstract
BACKGROUND Glioblastoma (GBM) is an aggressive primary brain tumor with a high recurrence rate and poor prognosis. Necroptosis, a pathological hallmark of GBM, is poorly understood in terms of its role in prognosis, tumor microenvironment (TME) alteration, and immunotherapy. METHODS & RESULTS We assessed the expression of 55 necroptosis-related genes in GBM and normal brain tissues. We identified necroptosis-stratified clusters using Uni-Cox and Least Absolute Shrinkage and Selection Operator (LASSO) regression to establish the 10-gene Glioblastoma Necroptosis Index (GNI). GNI demonstrated significant prognostic efficacy in the TCGA dataset (n = 160) and internal validation dataset (n = 345) and in external validation cohorts (n = 591). The GNI-high subgroup displayed a mesenchymal phenotype, lacking the IDH1 mutation, and MGMT methylation. This subgroup was characterized by significant enrichment in inflammatory and humoral immune pathways with prominent cell adhesion molecules (CD44 and ICAM1), inflammatory cytokines (TGFB1, IL1B, and IL10), and chemokines (CX3CL1, CXCL9, and CCL5). The TME in this subgroup showed elevated infiltration of M0 macrophages, neutrophils, mast cells, and regulatory T cells. GNI-related genes appeared to limit macrophage polarization, as confirmed by immunohistochemistry and flow cytometry. The top 30% high-risk score subset exhibited increased CD8 T cell infiltration and enhanced cytolytic activity. GNI showed promise in predicting responses to immunotherapy and targeted treatment. CONCLUSIONS Our study highlights the role of necroptosis-related genes in glioblastoma (GBM) and their effects on the tumor microenvironment and patient prognosis. TheGNI demonstrates potential as a prognostic marker and provides insights into immune characteristics and treatment responsiveness.
Collapse
Affiliation(s)
- Muhammad Khan
- Department of Radiation Oncology, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, People’s Republic of China
| | - Xiuting Huang
- Department of Radiation Oncology, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, People’s Republic of China
| | - Xiaoxin Ye
- Department of Radiation Oncology, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, People’s Republic of China
| | - Donghui Zhang
- Department of Pathology, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, People’s Republic of China
| | - Baiyao Wang
- Department of Radiation Oncology, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, People’s Republic of China
| | - Anan Xu
- Department of Radiation Oncology, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, People’s Republic of China
| | - Rong Li
- Department of Radiation Oncology, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, People’s Republic of China
| | - Anbang Ren
- Department of Radiation Oncology, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, People’s Republic of China
| | - Chengcong Chen
- Department of Radiation Oncology, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, People’s Republic of China
| | - Jingjing Song
- Department of Pathology, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, People’s Republic of China
| | - Rong Zheng
- Department of Radiation Oncology, Fujian Medical University Union Hospital, Fuzhou, People’s Republic of China
- Fujian Key Laboratory of Intelligent Imaging and Precision Radiotherapy for Tumors, Fujian Medical University, Fuzhou, People’s Republic of China
- Clinical Research Center for Radiology and Radiotherapy of Fujian Province (Digestive, Hematological and Breast Malignancies), Fuzhou, People’s Republic of China
| | - Yawei Yuan
- Department of Radiation Oncology, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, People’s Republic of China
| | - Jie Lin
- Department of Radiation Oncology, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, People’s Republic of China
| |
Collapse
|
3
|
Xia T, Pan Z, Wan H, Li Y, Mao G, Zhao J, Zhang F, Pan S. Mechanisms of mechanical stimulation in the development of respiratory system diseases. Am J Physiol Lung Cell Mol Physiol 2024; 327:L724-L739. [PMID: 39316681 DOI: 10.1152/ajplung.00122.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 09/06/2024] [Accepted: 09/11/2024] [Indexed: 09/26/2024] Open
Abstract
During respiration, mechanical stress can initiate biological responses that impact the respiratory system. Mechanical stress plays a crucial role in the development of the respiratory system. However, pathological mechanical stress can impact the onset and progression of respiratory diseases by influencing the extracellular matrix and cell transduction processes. In this article, we explore the mechanisms by which mechanical forces communicate with and influence cells. We outline the basic knowledge of respiratory mechanics, elucidating the important role of mechanical stimulation in influencing respiratory system development and differentiation from a microscopic perspective. We also explore the potential mechanisms of mechanical transduction in the pathogenesis and development of respiratory diseases such as asthma, lung injury, pulmonary fibrosis, and lung cancer. Finally, we look forward to new research directions in cellular mechanotransduction, aiming to provide fresh insights for future therapeutic research on respiratory diseases.
Collapse
Affiliation(s)
- Tian Xia
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
- Institute of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Ziyin Pan
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University, Shanghai, People's Republic of China
| | - Haoxin Wan
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
- Institute of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Yongsen Li
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
- Institute of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Guocai Mao
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
- Institute of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Jun Zhao
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
- Institute of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Fangbiao Zhang
- Department of Cardiothoracic Surgery, Lishui Municipal Central Hospital, Lishui, People's Republic of China
| | - Shu Pan
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
- Institute of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| |
Collapse
|
4
|
Mezentsev A, Durymanov M, Makarov VA. A Comprehensive Review of Protein Biomarkers for Invasive Lung Cancer. Curr Oncol 2024; 31:4818-4854. [PMID: 39329988 PMCID: PMC11431409 DOI: 10.3390/curroncol31090360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 08/16/2024] [Accepted: 08/22/2024] [Indexed: 09/28/2024] Open
Abstract
Invasion and metastasis are important hallmarks of lung cancer, and affect patients' survival. Early diagnostics of metastatic potential are important for treatment management. Recent findings suggest that the transition to an invasive phenotype causes changes in the expression of 700-800 genes. In this context, the biomarkers restricted to the specific type of cancer, like lung cancer, are often overlooked. Some well-known protein biomarkers correlate with the progression of the disease and the immunogenicity of the tumor. Most of these biomarkers are not exclusive to lung cancer because of their significant role in tumorigenesis. The dysregulation of others does not necessarily indicate cell invasiveness, as they play an active role in cell division. Clinical studies of lung cancer use protein biomarkers to assess the invasiveness of cancer cells for therapeutic purposes. However, there is still a need to discover new biomarkers for lung cancer. In the future, minimally invasive techniques, such as blood or saliva analyses, may be sufficient for this purpose. Many researchers suggest unconventional biomarkers, like circulating nucleic acids, exosomal proteins, and autoantibodies. This review paper aims to discuss the advantages and limitations of protein biomarkers of invasiveness in lung cancer, to assess their prognostic value, and propose novel biomarker candidates.
Collapse
Affiliation(s)
- Alexandre Mezentsev
- Medical Informatics Laboratory, Yaroslav-the-Wise Novgorod State University, 173003 Veliky Novgorod, Russia
- Center for Theoretical Problems of Physicochemical Pharmacology, 109029 Moscow, Russia
| | - Mikhail Durymanov
- Medical Informatics Laboratory, Yaroslav-the-Wise Novgorod State University, 173003 Veliky Novgorod, Russia
| | - Vladimir A Makarov
- Medical Informatics Laboratory, Yaroslav-the-Wise Novgorod State University, 173003 Veliky Novgorod, Russia
| |
Collapse
|
5
|
Fu Z, Sun G, Li J, Yu H. Identification of hub genes related to metastasis and prognosis of osteosarcoma and establishment of a prognostic model with bioinformatic methods. Medicine (Baltimore) 2024; 103:e38470. [PMID: 38847690 PMCID: PMC11155596 DOI: 10.1097/md.0000000000038470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Accepted: 05/15/2024] [Indexed: 06/10/2024] Open
Abstract
Osteosarcoma (OS) is the most common primary malignant bone tumor occurring in children and adolescents. Improvements in our understanding of the OS pathogenesis and metastatic mechanism on the molecular level might lead to notable advances in the treatment and prognosis of OS. Biomarkers related to OS metastasis and prognosis were analyzed and identified, and a prognostic model was established through the integration of bioinformatics tools and datasets in multiple databases. 2 OS datasets were downloaded from the Gene Expression Omnibus database for data consolidation, standardization, batch effect correction, and identification of differentially expressed genes (DEGs); following that, gene ontology and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses were performed on the DEGs; the STRING database was subsequently used for protein-protein interaction (PPI) network construction and identification of hub genes; hub gene expression was validated, and survival analysis was conducted through the employment of the TARGET database; finally, a prognostic model was established and evaluated subsequent to the screening of survival-related genes. A total of 701 DEGs were identified; by gene ontology and KEGG pathway enrichment analyses, the overlapping DEGs were enriched for 249 biological process terms, 13 cellular component terms, 35 molecular function terms, and 4 KEGG pathways; 13 hub genes were selected from the PPI network; 6 survival-related genes were identified by the survival analysis; the prognostic model suggested that 4 genes were strongly associated with the prognosis of OS. DEGs related to OS metastasis and survival were identified through bioinformatics analysis, and hub genes were further selected to establish an ideal prognostic model for OS patients. On this basis, 4 protective genes including TPM1, TPM2, TPM3, and TPM4 were yielded by the prognostic model.
Collapse
Affiliation(s)
- Zheng Fu
- Department of Orthopedics, Binzhou People’s Hospital, Binzhou,China
- Department of Orthopedics, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, China
| | - Guofeng Sun
- Department of Orthopedics, Binzhou People’s Hospital, Binzhou,China
| | - Jingtian Li
- Department of Orthopedics, Binzhou People’s Hospital, Binzhou,China
| | - Hongjian Yu
- Department of Orthopedics, Binzhou People’s Hospital, Binzhou,China
| |
Collapse
|
6
|
Luo Z, Liu X, Chen Y, Shen L, Qin H, Zha Q, Hu F, Wang Y. Gene features of tumor-specific T cells relevant to immunotherapy, targeted therapy and chemotherapy in lung cancer. Heliyon 2024; 10:e28374. [PMID: 38590880 PMCID: PMC10999884 DOI: 10.1016/j.heliyon.2024.e28374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 03/14/2024] [Accepted: 03/18/2024] [Indexed: 04/10/2024] Open
Abstract
1 Background In lung cancer, the use of small-molecule inhibitors, chemotherapy and immunotherapy has led to unprecedented survival benefits in selected patients. Considering most patients will experience a relapse within a short period of time due to single drug resistance, combination therapy is also particularly important to improve patient prognosis. Therefore, more robust biomarkers to predict responses to immunotherapy, targeted therapy, chemotherapy and rationally drug combination therapies may be helpful in clinical treatment choices. 2 Methods We defined tumor-specific T cells (TSTs) and their features (TSTGs) by single-cell RNA sequencing. We applied LASSO regression to filter out the most survival-relevant TSTGs to form the Tumor-specific T cell score (TSTS). Immunological characteristics, enriched pathways, and mutation were evaluated in high- and low TSTS groups. 3 Results We identified six clusters of T cells as TSTs in lung cancer, and four most robust genes from 9 feature genes expressed only on tumor-specific T cells were screened to construct a tumor-specific T cells score (TSTS). TSTS was positively correlated with immune infiltration and angiogenesis and negatively correlated with malignant cell proliferation. Moreover, potential vascular-immune crosstalk in lung cancer provides the theoretical basis for combined anti-angiogenic and immunotherapy. Noticeable, patients in high TSTS had better response to ICB and targeted therapy and patients in the low TSTS group often benefit from chemotherapy. 4 Conclusion The proposed TSTS is a promising indicator to predict immunotherapy, targeted therapy and chemotherapy responses in lung cancer patients for helping clinical treatment choices.
Collapse
Affiliation(s)
- Ziwei Luo
- Department of Respiratory Medicine, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200120, China
- State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Xuefei Liu
- School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, 518055, China
- Shenzhen Institute of Pediatrics, Shenzhen Children's Hospital, Shenzhen, Guangdong, 518038, China
| | - Ying Chen
- State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Lize Shen
- LC-Bio Technology Co.ltd, Hangzhou, 310018, China
| | - Hui Qin
- Department of Respiratory Medicine, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200120, China
| | - Qiongfang Zha
- Department of Respiratory Medicine, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200120, China
| | - Feng Hu
- Department of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200120, China
| | - Yali Wang
- Department of Respiratory Medicine, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200120, China
| |
Collapse
|
7
|
Cao L, Zhong J, Liu Z, Jiang J, Zhu C, Liu F, Wang B. Increased LOXL2 expression is related to poor prognosis in lung squamous cell carcinoma. J Thorac Dis 2024; 16:581-592. [PMID: 38410543 PMCID: PMC10894394 DOI: 10.21037/jtd-23-1848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 01/22/2024] [Indexed: 02/28/2024]
Abstract
Background The lysyl oxidate-like (LOXL) family was reported to be involved in the process of cancer development. However, the prognostic value of LOXL in lung cancer is unknown. We aimed to study the expression pattern and prognostic value of LOXL family members in lung squamous cell carcinoma (LUSC). Methods The Wilcoxon test and logistic regression analysis were used to study the expression level of LOXLs and its correlation with clinical characteristics. The Kaplan-Meier method and Cox regression analysis were performed to estimate the correlation of LOXsL expression with the survival of LUSC patients. Receiver operator characteristic (ROC) curves were plotted, and areas under the curves (AUCs) were calculated to estimate the diagnostic and prognostic power of LOXL. Cell Counting Kit-8 (CCK-8) assays, wound healing assays and Transwell assays were used to estimate the impact of LOXL2 on LUSC cells. Results LOXL1 and LOXL2 expression was upregulated in LUSC tissues (P<0.001). LOXL1 and LOXL2 showed high diagnostic power in LUSC patients, with AUCs of 0.784 and 0.751, respectively. Patients with high LOXL2 expression levels showed poor overall survival (OS) (P=0.019) and progression-free survival (PFS) (P=0.015). High LOXL2 expression was an independent prognostic factor for poor survival (P=0.026). Inhibition of LOXL2 suppressed proliferation, migration and invasion in LUSC cell lines. Conclusions Increased LOXL2 was related to poor survival in LUSC. LOXL2 may be a potential prognostic biomarker and therapeutic target in LUSC.
Collapse
Affiliation(s)
- Lei Cao
- Department of Thoracic Surgery, Nanjing Chest Hospital, Affiliated Nanjing Brain Hospital Nanjing Medical University, Nanjing, China
| | - Jian Zhong
- Department of Thoracic Surgery, Nanjing Chest Hospital, Affiliated Nanjing Brain Hospital Nanjing Medical University, Nanjing, China
| | - Zicheng Liu
- Department of Thoracic Surgery, Nanjing Chest Hospital, Affiliated Nanjing Brain Hospital Nanjing Medical University, Nanjing, China
| | - Jie Jiang
- Department of Thoracic Surgery, Nanjing Chest Hospital, Affiliated Nanjing Brain Hospital Nanjing Medical University, Nanjing, China
| | - Chenyao Zhu
- Shenzhen Yuce Biotechnology Co., Ltd., Shenzhen, China
| | - Feng Liu
- Department of Thoracic Surgery, Nanjing Chest Hospital, Affiliated Nanjing Brain Hospital Nanjing Medical University, Nanjing, China
| | - Bo Wang
- Department of Thoracic Surgery, Nanjing Chest Hospital, Affiliated Nanjing Brain Hospital Nanjing Medical University, Nanjing, China
| |
Collapse
|
8
|
Li C, Chen S, Fang X, Du Y, Guan XY, Lin R, Xu L, Lan P, Yan Q. LOXL1 promotes tumor cell malignancy and restricts CD8 + T cell infiltration in colorectal cancer. Cell Biol Toxicol 2024; 40:6. [PMID: 38267662 PMCID: PMC10808464 DOI: 10.1007/s10565-024-09840-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 12/05/2023] [Indexed: 01/26/2024]
Abstract
BACKGROUND Colorectal cancer (CRC) is a leading cause of cancer mortality globally. Lymph node metastasis and immunosuppression are main factors of poor prognosis in CRC patients. Lysyl oxidase like 1 (LOXL1), part of the lysyl oxidase (LOX) family, plays a yet unclear role in CRC. This study aimed to identify effective biomarkers predictive of prognosis and efficacy of immunotherapy in CRC patients, and to elucidate the prognostic value, clinical relevance, functional and molecular features, and immunotherapy predictive role of LOXL1 in CRC and pan-cancer. METHODS Weighted gene co-expression network analysis (WGCNA) was employed to explore gene modules related to tumor metastasis and CD8 + T cell infiltration. LOXL1 emerged as a hub gene through differential gene expression and survival analysis. The molecular signatures, functional roles, and immunological characteristics affected by LOXL1 were analyzed in multiple CRC cohorts, cell lines and clinical specimens. Additionally, LOXL1's potential as an immunotherapy response indicator was assessed, along with its role in pan-cancer. RESULTS Turquoise module in WGCNA analysis was identified as the hub module associated with lymph node metastasis and CD8 + T cell infiltration. Aberrant elevated LOXL1 expression was observed in CRC and correlated with poorer differentiation status and prognosis. Molecular and immunological characterization found that LOXL1 might mediate epithelial-mesenchymal transition (EMT) process and immunosuppressive phenotypes of CRC. Functional study found that LOXL1 enhanced tumor cell proliferation, migration and invasion. Moreover, high LOXL1 levels corresponded to reduced CD8 + T cell infiltration and predicted poor clinical outcomes of immunotherapy. Similar trends were also observed at the pan-cancer level. CONCLUSIONS Our findings underscore the critical role of LOXL1 in modulating both malignancy and immunosuppression in CRC. This positions LOXL1 as a promising biomarker for predicting prognosis and the response to immunotherapy in CRC patients.
Collapse
Affiliation(s)
- Chenxi Li
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-Sen University, Room 703, Building No. 3, 26 Yuancun ERheng Road, Guangzhou, 510655, China
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-Sen University, Room 703, Building No. 3, 26 Yuancun ERheng Road, Guangzhou, 510655, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Room 703, Building No. 3, 26 Yuancun ERheng Road, Guangzhou, 510655, China
| | - Siqi Chen
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-Sen University, Room 703, Building No. 3, 26 Yuancun ERheng Road, Guangzhou, 510655, China
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-Sen University, Room 703, Building No. 3, 26 Yuancun ERheng Road, Guangzhou, 510655, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Room 703, Building No. 3, 26 Yuancun ERheng Road, Guangzhou, 510655, China
| | - Xiaona Fang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou, China
- Department of Pediatric Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yaqing Du
- Institute of Basic Medical Sciences, School of Life Sciences and Biopharmaceuticals, Guangdong Pharmaceutical University, Guangzhou, China
| | - Xin-Yuan Guan
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong, China
- State Key Laboratory for Liver Research, The University of Hong Kong, Hong Kong, China
| | - Runhua Lin
- Department of Pathology, Shantou University Medical College, Shantou, China
| | - Liang Xu
- Department of Pathology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
| | - Ping Lan
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-Sen University, Room 703, Building No. 3, 26 Yuancun ERheng Road, Guangzhou, 510655, China.
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-Sen University, Room 703, Building No. 3, 26 Yuancun ERheng Road, Guangzhou, 510655, China.
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Room 703, Building No. 3, 26 Yuancun ERheng Road, Guangzhou, 510655, China.
- State Key Laboratory of Oncology in South China, Sun Yat-sen University, Guangzhou, China.
| | - Qian Yan
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-Sen University, Room 703, Building No. 3, 26 Yuancun ERheng Road, Guangzhou, 510655, China.
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-Sen University, Room 703, Building No. 3, 26 Yuancun ERheng Road, Guangzhou, 510655, China.
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Room 703, Building No. 3, 26 Yuancun ERheng Road, Guangzhou, 510655, China.
| |
Collapse
|
9
|
Baghy K, Ladányi A, Reszegi A, Kovalszky I. Insights into the Tumor Microenvironment-Components, Functions and Therapeutics. Int J Mol Sci 2023; 24:17536. [PMID: 38139365 PMCID: PMC10743805 DOI: 10.3390/ijms242417536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 11/25/2023] [Accepted: 12/13/2023] [Indexed: 12/24/2023] Open
Abstract
Similarly to our healthy organs, the tumor tissue also constitutes an ecosystem. This implies that stromal cells acquire an altered phenotype in tandem with tumor cells, thereby promoting tumor survival. Cancer cells are fueled by abnormal blood vessels, allowing them to develop and proliferate. Tumor-associated fibroblasts adapt their cytokine and chemokine production to the needs of tumor cells and alter the peritumoral stroma by generating more collagen, thereby stiffening the matrix; these processes promote epithelial-mesenchymal transition and tumor cell invasion. Chronic inflammation and the mobilization of pro-tumorigenic inflammatory cells further facilitate tumor expansion. All of these events can impede the effective administration of tumor treatment; so, the successful inhibition of tumorous matrix remodeling could further enhance the success of antitumor therapy. Over the last decade, significant progress has been made with the introduction of novel immunotherapy that targets the inhibitory mechanisms of T cell activation. However, extensive research is also being conducted on the stromal components and other cell types of the tumor microenvironment (TME) that may serve as potential therapeutic targets.
Collapse
Affiliation(s)
- Kornélia Baghy
- Department of Pathology and Experimental Cancer Research, Semmelweis University, 1085 Budapest, Hungary;
| | - Andrea Ladányi
- Department of Surgical and Molecular Pathology and the National Tumor Biology Laboratory, National Institute of Oncology, 1122 Budapest, Hungary;
| | - Andrea Reszegi
- Department of Pediatrics, College of Medicine, University of Florida, Gainesville, FL 32610, USA;
- Department of Pathology, Forensic and Insurance Medicine, Semmelweis University, 1091 Budapest, Hungary
| | - Ilona Kovalszky
- Department of Pathology and Experimental Cancer Research, Semmelweis University, 1085 Budapest, Hungary;
| |
Collapse
|
10
|
Xie W, Peng Z, Zhou X, Xia Q, Chen M, Zheng X, Sun H, Zou H, Xu L, Du Z, Li E, Wu B. The Expression Pattern and Clinical Significance of Lysyl Oxidase Family in Gliomas. DOKL BIOCHEM BIOPHYS 2023; 510:132-143. [PMID: 37582875 DOI: 10.1134/s1607672922600269] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 03/03/2023] [Accepted: 03/09/2023] [Indexed: 08/17/2023]
Abstract
LOX (Lysyl oxidase) family participates in the catalysis of collagen and elastin to maintain ECM homeostasis. Glioma is the most common primary brain tumor and LOX family has not been systemic studied in glioma. In this study, we found LOX family members are upregulated expressed in gliomas samples. A protein-protein interaction network (PPIN) was construct to visualize and understand the differential expression pattern, as well as functional annotation, for LOX family and their interacting proteins, which involved in collagen fibril organization and MAPK signaling pathway. Through subcellular localization distribution, the LOX family members distribute both intracellular and extracellular. All five LOX members are consistently significantly correlate with dendritic cell both in immune infiltrate of GBM and LGG. Survival analysis showed that high expression of LOX family is associated with a poor prognosis of gliomas patients. These analyses provide important clues to identify the potential biological roles for LOX family in gliomas, which might serve as diagnosis markers.
Collapse
Affiliation(s)
- Weijie Xie
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, 515041, Shantou, China
| | - Zhongte Peng
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, 515041, Shantou, China
| | - Xiao Zhou
- Department of Central Laboratory, Shantou Central Hospital, 515041, Shantou, China
| | - Qiaoxi Xia
- Department of Central Laboratory, Shantou Central Hospital, 515041, Shantou, China
| | - Mantong Chen
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, 515041, Shantou, China
| | - Xiaoqi Zheng
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, 515041, Shantou, China
| | - Hong Sun
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, 515041, Shantou, China
| | - Haiying Zou
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, 515041, Shantou, China
| | - Liyan Xu
- Institute of Oncologic Pathology, Shantou University Medical College, 515041, Shantou, China
| | - Zepeng Du
- Department of Central Laboratory, Shantou Central Hospital, 515041, Shantou, China
| | - Enmin Li
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, 515041, Shantou, China.
| | - Bingli Wu
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, 515041, Shantou, China.
| |
Collapse
|
11
|
Liu F, Wu Q, Dong Z, Liu K. Integrins in cancer: Emerging mechanisms and therapeutic opportunities. Pharmacol Ther 2023:108458. [PMID: 37245545 DOI: 10.1016/j.pharmthera.2023.108458] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 05/10/2023] [Accepted: 05/22/2023] [Indexed: 05/30/2023]
Abstract
Integrins are vital surface adhesion receptors that mediate the interactions between the extracellular matrix (ECM) and cells and are essential for cell migration and the maintenance of tissue homeostasis. Aberrant integrin activation promotes initial tumor formation, growth, and metastasis. Recently, many lines of evidence have indicated that integrins are highly expressed in numerous cancer types and have documented many functions of integrins in tumorigenesis. Thus, integrins have emerged as attractive targets for the development of cancer therapeutics. In this review, we discuss the underlying molecular mechanisms by which integrins contribute to most of the hallmarks of cancer. We focus on recent progress on integrin regulators, binding proteins, and downstream effectors. We highlight the role of integrins in the regulation of tumor metastasis, immune evasion, metabolic reprogramming, and other hallmarks of cancer. In addition, integrin-targeted immunotherapy and other integrin inhibitors that have been used in preclinical and clinical studies are summarized.
Collapse
Affiliation(s)
- Fangfang Liu
- Research Center of Basic Medicine, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China; China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan 450008, China
| | - Qiong Wu
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan 450008, China; Department of Pathophysiology, School of Basic Medical Sciences, College of Medicine, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Zigang Dong
- Research Center of Basic Medicine, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China; China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan 450008, China; Department of Pathophysiology, School of Basic Medical Sciences, College of Medicine, Zhengzhou University, Zhengzhou, Henan 450001, China; State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou, Henan 450000, China; Tianjian Advanced Biomedical Laboratory, Zhengzhou University, Zhengzhou, Henan 450001, China.
| | - Kangdong Liu
- Research Center of Basic Medicine, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China; China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan 450008, China; Department of Pathophysiology, School of Basic Medical Sciences, College of Medicine, Zhengzhou University, Zhengzhou, Henan 450001, China; State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou, Henan 450000, China; Tianjian Advanced Biomedical Laboratory, Zhengzhou University, Zhengzhou, Henan 450001, China; Cancer Chemoprevention International Collaboration Laboratory, Zhengzhou, Henan 450000, China.
| |
Collapse
|
12
|
Zeltz C, Navab R, Heljasvaara R, Kusche-Gullberg M, Lu N, Tsao MS, Gullberg D. Integrin α11β1 in tumor fibrosis: more than just another cancer-associated fibroblast biomarker? J Cell Commun Signal 2022; 16:649-660. [PMID: 35378690 PMCID: PMC8978763 DOI: 10.1007/s12079-022-00673-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Accepted: 03/04/2022] [Indexed: 12/13/2022] Open
Abstract
There is currently an increased interest in understanding the role of the tumor microenvironment (TME) in tumor growth and progression. In this context the role of integrins in cancer-associated fibroblasts (CAFs) will need to be carefully re-evaluated. Fibroblast-derived cells are not only in the focus in tumors, but also in tissue fibrosis as well as in inflammatory conditions. The recent transcriptional profiling of what has been called "the pan-fibroblast cell lineage" in mouse and human tissues has identified novel transcriptional biomarker mRNAs encoding the secreted ECM proteins dermatopontin and collagen XV as well as the phosphatidylinositol-anchored membrane protein Pi16. Some of the genes identified in these fibroblasts scRNA-seq datasets will be useful for rigorous comparative characterizations of fibroblast-derived cell subpopulations. At the same time, it will be a challenge in the coming years to validate these transcriptional mRNA datasets at the protein-(expression) and at tissue-(distribution) levels and to find useful protein biomarker reagents that will facilitate fibroblast profiling at the cell level. In the current review we will focus on the role of the collagen-binding integrin α11β1 in CAFs, summarizing our own work as well as published datasets with information on α11 mRNA expression in selected tumors. Our experimental data suggest that α11β1 is more than just another biomarker and that it as a functional collagen receptor in the TME is playing a central role in regulating collagen assembly and matrix remodeling, which in turn impact tumor growth and metastasis.
Collapse
Affiliation(s)
- Cédric Zeltz
- Department of Biomedicine, Matrix Biology Group, Centre for Cancer Biomarkers, University of Bergen, Jonas Lies vei 91, 5009, Bergen, Norway
| | - Roya Navab
- Princess Margaret Cancer Center, University Health Network, Toronto, ON, M5G 1L7, Canada
| | - Ritva Heljasvaara
- Oulu Center for Cell-Matrix Research, Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Marion Kusche-Gullberg
- Department of Biomedicine, Matrix Biology Group, Centre for Cancer Biomarkers, University of Bergen, Jonas Lies vei 91, 5009, Bergen, Norway
| | - Ning Lu
- Department of Biomedicine, Matrix Biology Group, Centre for Cancer Biomarkers, University of Bergen, Jonas Lies vei 91, 5009, Bergen, Norway
| | - Ming-Sound Tsao
- Princess Margaret Cancer Center, University Health Network, Toronto, ON, M5G 1L7, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, M5G 1X8, Canada
| | - Donald Gullberg
- Department of Biomedicine, Matrix Biology Group, Centre for Cancer Biomarkers, University of Bergen, Jonas Lies vei 91, 5009, Bergen, Norway.
| |
Collapse
|
13
|
Weerakoon AT, Condon N, Cox TR, Sexton C, Cooper C, Meyers IA, Thomson D, Ford PJ, Roy S, Symons AL. Dynamic dentin: A quantitative microscopic assessment of age and spatial changes to matrix architecture, peritubular dentin, and collagens types I and III. J Struct Biol 2022; 214:107899. [PMID: 36208858 DOI: 10.1016/j.jsb.2022.107899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 08/16/2022] [Accepted: 09/19/2022] [Indexed: 12/07/2022]
Abstract
To investigate age and site-related changes to human dentin collagen, sound human teeth collected from donors aged 13-29 (young) and 50-74 (aged) years (n = 9/group) were cut to shallow and deep sites. Dentin collagen orientation and fibril bundling was investigated using the Picrosirius Red (PSR) stain observed under cross-polarized light microscopy (Pol), and collagen distribution was investigated using Confocal Laser Scanning Microscopy (CLSM). Collagen types III to I distribution in peritubular dentin (PTD) was revealed using Herovici stain and brightfield microscopy. Image analysis software and linear mixed modelling quantified outcomes. In situ dentin collagen was observed using Xenon Plasma Focussed Ion Beam Scanning Electron Microscopy (Xe PFIB-SEM). The PSR-Pol analysis revealed less coherently aligned and more bundled collagen fibrils in aged dentin (P = 0.005). Deep inner dentin collagen in both groups were less coherently aligned with reduced bundling. Regardless of age, CLSM showed collagen distribution remained stable; and more collagen type III was detectable in PTD located in inner dentin (Young: P = 0.006; Aged: P = 0.008). Observations following Xe PFIB-SEM cross-sectioning showed apatite-like deposits surrounding large intratubular collagen fibers, and evidence of smaller intertubular dentin collagen fibrils in situ. In conclusion, aging changes collagen network architecture, but not distribution or content.
Collapse
Affiliation(s)
- Arosha T Weerakoon
- School of Dentistry, The University of Queensland, Brisbane, Queensland, Australia.
| | - Nicholas Condon
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | - Thomas R Cox
- Garvan Institute of Medical Research & School of Clinical Medicine, UNSW, Sydney, Australia
| | - Christopher Sexton
- School of Dentistry, The University of Queensland, Brisbane, Queensland, Australia
| | - Crystal Cooper
- Central Analytical Research Facility, Queensland University of Technology, Brisbane, Queensland, Australia; Centre for Microscopy, Characterisation and Analysis, University of Western Australia, Perth, Western Australia, Australia
| | - Ian A Meyers
- School of Dentistry, The University of Queensland, Brisbane, Queensland, Australia
| | - David Thomson
- School of Dentistry, The University of Queensland, Brisbane, Queensland, Australia
| | - Pauline J Ford
- School of Dentistry, The University of Queensland, Brisbane, Queensland, Australia
| | - Sandrine Roy
- Translational Research Institute, Brisbane, Queensland, Australia; Olympus Life Science, Australia
| | - Anne L Symons
- School of Dentistry, The University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|
14
|
Li C, Qiu Y, Zhang Y. Research Progress on Therapeutic Targeting of Cancer-Associated Fibroblasts to Tackle Treatment-Resistant NSCLC. Pharmaceuticals (Basel) 2022; 15:1411. [PMID: 36422541 PMCID: PMC9696940 DOI: 10.3390/ph15111411] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 11/09/2022] [Accepted: 11/11/2022] [Indexed: 08/04/2023] Open
Abstract
Non-small cell lung cancer (NSCLC) accounts for most lung cancer cases and is the leading cause of cancer-related deaths worldwide. Treatment options for lung cancer are no longer limited to surgery, radiotherapy, and chemotherapy, as targeted therapy and immunotherapy offer a new hope for patients. However, drug resistance in chemotherapy and targeted therapy, and the low response rates to immunotherapy remain important challenges. Similar to tumor development, drug resistance occurs because of significant effects exerted by the tumor microenvironment (TME) along with cancer cell mutations. Cancer-associated fibroblasts (CAFs) are a key component of the TME and possess multiple functions, including cross-talking with cancer cells, remodeling of the extracellular matrix (ECM), secretion of various cytokines, and promotion of epithelial-mesenchymal transition, which in turn provide support for the growth, invasion, metastasis, and drug resistance of cancer cells. Therefore, CAFs represent valuable therapeutic targets for lung cancer. Herein, we review the latest progress in the use of CAFs as potential targets and mediators of drug resistance for NSCLC treatment. We explored the role of CAFs on the regulation of the TME and surrounding ECM, with particular emphasis on treatment strategies involving combined CAF targeting within the current framework of cancer treatment.
Collapse
|
15
|
Wong KY, Cheung AH, Chen B, Chan WN, Yu J, Lo KW, Kang W, To KF. Cancer-associated fibroblasts in nonsmall cell lung cancer: From molecular mechanisms to clinical implications. Int J Cancer 2022; 151:1195-1215. [PMID: 35603909 PMCID: PMC9545594 DOI: 10.1002/ijc.34127] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 05/08/2022] [Accepted: 05/12/2022] [Indexed: 11/14/2022]
Abstract
Lung cancer is the common and leading cause of cancer death worldwide. The tumor microenvironment has been recognized to be instrumental in tumorigenesis. To have a deep understanding of the molecular mechanism of nonsmall cell lung carcinoma (NSCLC), cancer-associated fibroblasts (CAFs) have gained increasing research interests. CAFs belong to the crucial and dominant cell population in the tumor microenvironment to support the cancer cells. The interplay and partnership between cancer cells and CAFs contribute to each stage of tumorigenesis. CAFs exhibit prominent heterogeneity and secrete different kinds of cytokines and chemokines, growth factors and extracellular matrix proteins involved in cancer cell proliferation, invasion, metastasis and chemoresistance. Many studies focused on the protumorigenic functions of CAFs, yet many challenges about the heterogeneity of CAFS remain unresolved. This review comprehensively summarized the tumor-promoting role and molecular mechanisms of CAFs in NSCLC, including their origin, phenotypic changes and heterogeneity and their functional roles in carcinogenesis. Meanwhile, we also highlighted the updated molecular classifications based on the molecular features and functional roles of CAFs. With the development of cutting-edge platforms and further investigations of CAFs, novel therapeutic strategies for accurately targeting CAFs in NSCLC may be developed based on the increased understanding of the relevant molecular mechanisms.
Collapse
Affiliation(s)
- Kit Yee Wong
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational OncologyPrince of Wales Hospital, The Chinese University of Hong KongHong KongSARChina
- Institute of Digestive Disease, State Key Laboratory of Digestive Disease, The Chinese University of Hong KongHong KongSARChina
- Li Ka Shing Institute of Health Science, Sir Y.K. Pao Cancer Center, The Chinese University of Hong KongHong KongSARChina
| | - Alvin Ho‐Kwan Cheung
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational OncologyPrince of Wales Hospital, The Chinese University of Hong KongHong KongSARChina
- Institute of Digestive Disease, State Key Laboratory of Digestive Disease, The Chinese University of Hong KongHong KongSARChina
- Li Ka Shing Institute of Health Science, Sir Y.K. Pao Cancer Center, The Chinese University of Hong KongHong KongSARChina
| | - Bonan Chen
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational OncologyPrince of Wales Hospital, The Chinese University of Hong KongHong KongSARChina
- Institute of Digestive Disease, State Key Laboratory of Digestive Disease, The Chinese University of Hong KongHong KongSARChina
- Li Ka Shing Institute of Health Science, Sir Y.K. Pao Cancer Center, The Chinese University of Hong KongHong KongSARChina
| | - Wai Nok Chan
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational OncologyPrince of Wales Hospital, The Chinese University of Hong KongHong KongSARChina
- Institute of Digestive Disease, State Key Laboratory of Digestive Disease, The Chinese University of Hong KongHong KongSARChina
- Li Ka Shing Institute of Health Science, Sir Y.K. Pao Cancer Center, The Chinese University of Hong KongHong KongSARChina
| | - Jun Yu
- Institute of Digestive Disease, State Key Laboratory of Digestive Disease, The Chinese University of Hong KongHong KongSARChina
- Department of Medicine and TherapeuticsThe Chinese University of Hong KongHong KongSARChina
| | - Kwok Wai Lo
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational OncologyPrince of Wales Hospital, The Chinese University of Hong KongHong KongSARChina
- Institute of Digestive Disease, State Key Laboratory of Digestive Disease, The Chinese University of Hong KongHong KongSARChina
- Li Ka Shing Institute of Health Science, Sir Y.K. Pao Cancer Center, The Chinese University of Hong KongHong KongSARChina
| | - Wei Kang
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational OncologyPrince of Wales Hospital, The Chinese University of Hong KongHong KongSARChina
- Institute of Digestive Disease, State Key Laboratory of Digestive Disease, The Chinese University of Hong KongHong KongSARChina
- Li Ka Shing Institute of Health Science, Sir Y.K. Pao Cancer Center, The Chinese University of Hong KongHong KongSARChina
| | - Ka Fai To
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational OncologyPrince of Wales Hospital, The Chinese University of Hong KongHong KongSARChina
- Institute of Digestive Disease, State Key Laboratory of Digestive Disease, The Chinese University of Hong KongHong KongSARChina
- Li Ka Shing Institute of Health Science, Sir Y.K. Pao Cancer Center, The Chinese University of Hong KongHong KongSARChina
| |
Collapse
|
16
|
Lysyl Oxidase Family Proteins: Prospective Therapeutic Targets in Cancer. Int J Mol Sci 2022; 23:ijms232012270. [PMID: 36293126 PMCID: PMC9602794 DOI: 10.3390/ijms232012270] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Revised: 10/03/2022] [Accepted: 10/08/2022] [Indexed: 11/06/2022] Open
Abstract
The lysyl oxidase (LOX) family, consisting of LOX and LOX-like proteins 1–4 (LOXL1–4), is responsible for the covalent crosslinking of collagen and elastin, thus maintaining the stability of the extracellular matrix (ECM) and functioning in maintaining connective tissue function, embryonic development, and wound healing. Recent studies have found the aberrant expression or activity of the LOX family occurs in various types of cancer. It has been proved that the LOX family mainly performs tumor microenvironment (TME) remodeling function and is extensively involved in tumor invasion and metastasis, immunomodulation, proliferation, apoptosis, etc. With relevant translational research in progress, the LOX family is expected to be an effective target for tumor therapy. Here, we review the research progress of the LOX family in tumor progression and therapy to provide novel insights for future exploration of relevant tumor mechanism and new therapeutic targets.
Collapse
|
17
|
Zeltz C, Khalil M, Navab R, Tsao MS. Collagen Type XI Inhibits Lung Cancer-Associated Fibroblast Functions and Restrains the Integrin Binding Site Availability on Collagen Type I Matrix. Int J Mol Sci 2022; 23:ijms231911722. [PMID: 36233024 PMCID: PMC9569509 DOI: 10.3390/ijms231911722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 09/20/2022] [Accepted: 09/26/2022] [Indexed: 12/02/2022] Open
Abstract
The tumor microenvironment, including cancer-associated fibroblast (CAF), plays an active role in non-small cell lung cancer (NSCLC) development and progression. We previously reported that collagen type XI and integrin α11, a collagen receptor, were upregulated in NSCLC; the latter promotes tumor growth and metastasis. We here explored the role of collagen type XI in NSCLC stroma. We showed that the presence of collagen type XI in collagen type I matrices inhibits CAF-mediated collagen remodeling and cell migration. This resulted in the inhibition of CAF-dependent lung-tumor cell invasion. Among the collagen receptors expressed on CAF, we determined that DDR2 and integrin α2β1, but not integrin α11β1, mediated the high-affinity binding to collagen type XI. We further demonstrated that collagen type XI restrained the integrin binding site availability on collagen type I matrices, thus limiting cell interaction with collagen type I. As a consequence, CAFs failed to activate FAK, p38 and Akt one hour after they interacted with collagen type I/XI. We concluded that collagen type XI may have a competitive negative feedback role on the binding of collagen type I to its receptors.
Collapse
Affiliation(s)
- Cédric Zeltz
- Princess Margaret Cancer Center, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Maryam Khalil
- Princess Margaret Cancer Center, University Health Network, Toronto, ON M5G 1L7, Canada
- Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Roya Navab
- Princess Margaret Cancer Center, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Ming-Sound Tsao
- Princess Margaret Cancer Center, University Health Network, Toronto, ON M5G 1L7, Canada
- Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada
- Departments of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada
- Correspondence:
| |
Collapse
|
18
|
Luo C, Li B, Liu C, Dong R, Hu C, Liu J, Hu L, Liao X, Zhou J, Xu L, Liu S, Yuan D, Jiang W, Yan J, Li Y. Lysyl oxidase family gene polymorphisms and risk of aneurysmal subarachnoid hemorrhage: a case-control study. ANNALS OF TRANSLATIONAL MEDICINE 2022; 10:925. [PMID: 36172092 PMCID: PMC9511205 DOI: 10.21037/atm-22-3484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 08/08/2022] [Indexed: 11/15/2022]
Abstract
Background Aneurysmal subarachnoid hemorrhage (aSAH) is a devastating disease caused by intracranial aneurysm (IA) rupture. Lysyl oxidase (LOX) family genes (LOX-like [LOXL] 1-4) have roles in collagen cross-linking in the extracellular matrix (ECM) and may be associated with IA rupture. We aimed to explore the association between LOX polymorphisms and the risk of aSAH. Methods This case-control study included 2 cohorts: 133 single ruptured and 115 unruptured IA patients, and 65 multiple ruptured and 71 unruptured IA patients. Genotyping of 27 single nucleotide polymorphisms (SNPs) in LOX was performed. Logistic regression analysis was performed to calculate the odds ratios (ORs) and 95% confidence intervals (CIs) of the SNPs of LOX and the risk of aSAH. Results LOX rs1800449 and LOXL4 rs3793692 were positively associated with the risk of single IA rupture in the recessive model (OR =5.66, 2.06; 95% CI =1.22–26.24, 1.11–3.82, respectively) and LOX rs10519694 demonstrated a protective effect on single IA rupture (dominant model: OR =0.42, 95% CI =0.21–0.83; recessive model: OR =0.16, 95% CI =0.04–0.65; additive model: OR =0.46, 95% CI =0.28–0.78). LOXL1 rs2165241, LOXL2 rs1063582, and LOXL3 rs17010021 showed risk effects on multiple IAs rupture. LOXL3 rs17010022 showed a protective effect on multiple IAs ruptures (dominant model: OR =0.41, 95% CI =0.21–0.82; additive model: OR =0.51, 95% CI =0.30–0.85). Conclusions LOX and LOXL4 may be susceptibility genes for single IA rupture, whereas LOXL1-3 may have a role in susceptibility to multiple IAs ruptures in the Chinese population, suggesting that LOX family genes may be associated with aSAH.
Collapse
Affiliation(s)
- Chun Luo
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, China
| | - Bingyang Li
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, China.,Department of Information Statistics, Changsha Hospital of Traditional Chinese Medicine (Changsha Eighth Hospital), Changsha, China
| | - Chao Liu
- Department of Neurosurgery, Central Hospital of Zhuzhou, Zhuzhou, China
| | - Rui Dong
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, China
| | - Chongyu Hu
- Department of Neurology, Hunan People's Hospital, Changsha, China
| | - Junyu Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Liming Hu
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, China
| | - Xin Liao
- Department of Biomedical Sciences, Faculty of Health Sciences, University of Macau, Macau, China
| | - Jilin Zhou
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Lu Xu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Songlin Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Dun Yuan
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Weixi Jiang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Junxia Yan
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, China.,The Hunan Provincial Key Laboratory of Clinical Epidemiology, Xiangya School of Public Health, Central South University, Changsha, China
| | - Yifeng Li
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
19
|
Laurentino TDS, Soares RDS, Marie SKN, Oba-Shinjo SM. Correlation of Matrisome-Associatted Gene Expressions with LOX Family Members in Astrocytomas Stratified by IDH Mutation Status. Int J Mol Sci 2022; 23:ijms23179507. [PMID: 36076905 PMCID: PMC9455728 DOI: 10.3390/ijms23179507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 08/02/2022] [Accepted: 08/16/2022] [Indexed: 12/05/2022] Open
Abstract
Tumor cell infiltrative ability into surrounding brain tissue is a characteristic of diffusely infiltrative astrocytoma and is strongly associated with extracellular matrix (ECM) stiffness. Collagens are the most abundant ECM scaffolding proteins and contribute to matrix organization and stiffness. LOX family members, copper-dependent amine oxidases, participate in the collagen and elastin crosslinking that determine ECM tensile strength. Common IDH mutations in lower-grade gliomas (LGG) impact prognosis and have been associated with ECM stiffness. We analyzed the expression levels of LOX family members and matrisome-associated genes in astrocytoma stratified by malignancy grade and IDH mutation status. A progressive increase in expression of all five LOX family members according to malignancy grade was found. LOX, LOXL1, and LOXL3 expression correlated with matrisome gene expressions. LOXL1 correlations were detected in LGG with IDH mutation (IDHmut), LOXL3 correlations in LGG with IDH wild type (IDHwt) and strong LOX correlations in glioblastoma (GBM) were found. These increasing correlations may explain the increment of ECM stiffness and tumor aggressiveness from LGG-IDHmut and LGG-IDHwt through to GBM. The expression of the mechanosensitive transcription factor, β-catenin, also increased with malignancy grade and was correlated with LOXL1 and LOXL3 expression, suggesting involvement of this factor in the outside–in signaling pathway.
Collapse
|
20
|
Martínez-Nieto GA, Teppo HR, Petrelius N, Izzi V, Devarajan R, Petäistö T, Liu H, Kim KS, Karppinen SM, Ruotsalainen H, Koivunen J, Mäki JM, Walker GC, Pihlajaniemi T, Gullberg D, Heljasvaara R. Upregulated integrin α11 in the stroma of cutaneous squamous cell carcinoma promotes skin carcinogenesis. Front Oncol 2022; 12:981009. [PMID: 36003785 PMCID: PMC9393502 DOI: 10.3389/fonc.2022.981009] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 07/12/2022] [Indexed: 11/17/2022] Open
Abstract
Integrin α11β1 is a collagen-binding integrin that is needed to induce and maintain the myofibroblast phenotype in fibrotic tissues and during wound healing. The expression of the α11 is upregulated in cancer-associated fibroblasts (CAFs) in various human neoplasms. We investigated α11 expression in human cutaneous squamous cell carcinoma (cSCC) and in benign and premalignant human skin lesions and monitored its effects on cSCC development by subjecting α11-knockout (Itga11−/−) mice to the DMBA/TPA skin carcinogenesis protocol. α11-deficient mice showed significantly decreased tumor cell proliferation, leading to delayed tumor development and reduced tumor burden. Integrin α11 expression was significantly upregulated in the desmoplastic tumor stroma of human and mouse cSCCs, and the highest α11 expression was detected in high-grade tumors. Our results point to a reduced ability of α11-deficient stromal cells to differentiate into matrix-producing and tumor-promoting CAFs and suggest that this is one causative mechanism underlying the observed decreased tumor growth. An unexpected finding in our study was that, despite reduced CAF activation, the α11-deficient skin tumors were characterized by the presence of thick and regularly aligned collagen bundles. This finding was attributed to a higher expression of TGFβ1 and collagen crosslinking lysyl oxidases in the Itga11-/- tumor stroma. In summary, our data suggest that α11β1 operates in a complex interactive tumor environment to regulate ECM synthesis and collagen organization and thus foster cSCC growth. Further studies with advanced experimental models are still needed to define the exact roles and molecular mechanisms of stromal α11β1 in skin tumorigenesis.
Collapse
Affiliation(s)
- Guillermo A. Martínez-Nieto
- ECM-Hypoxia Research Unit, Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Hanna-Riikka Teppo
- Cancer Research and Translational Medicine Research Unit, University of Oulu, Oulu, Finland
- Medical Research Center, Oulu University Hospital and University of Oulu, Oulu, Finland
- Department of Pathology, Oulu University Hospital, Oulu, Finland
| | - Noora Petrelius
- ECM-Hypoxia Research Unit, Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Valerio Izzi
- ECM-Hypoxia Research Unit, Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
- Research Unit of Biomedicine, University of Oulu, Oulu, Finland
- Finnish Cancer Institute, Helsinki, Finland
| | - Raman Devarajan
- ECM-Hypoxia Research Unit, Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Tiina Petäistö
- ECM-Hypoxia Research Unit, Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Hengshuo Liu
- Matrix Biology Group, Department of Biomedicine, Centre for Cancer Biomarkers, University of Bergen, Bergen, Norway
| | - Kris S. Kim
- Department of Chemistry, University of Toronto, Toronto, ON, Canada
| | - Sanna-Maria Karppinen
- ECM-Hypoxia Research Unit, Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Heli Ruotsalainen
- ECM-Hypoxia Research Unit, Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Jarkko Koivunen
- ECM-Hypoxia Research Unit, Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Joni M. Mäki
- ECM-Hypoxia Research Unit, Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | | | - Taina Pihlajaniemi
- ECM-Hypoxia Research Unit, Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Donald Gullberg
- Matrix Biology Group, Department of Biomedicine, Centre for Cancer Biomarkers, University of Bergen, Bergen, Norway
| | - Ritva Heljasvaara
- ECM-Hypoxia Research Unit, Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
- Matrix Biology Group, Department of Biomedicine, Centre for Cancer Biomarkers, University of Bergen, Bergen, Norway
- *Correspondence: Ritva Heljasvaara,
| |
Collapse
|
21
|
Wang C, Yang J. Mechanical forces: The missing link between idiopathic pulmonary fibrosis and lung cancer. Eur J Cell Biol 2022; 101:151234. [DOI: 10.1016/j.ejcb.2022.151234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 05/05/2022] [Accepted: 05/05/2022] [Indexed: 11/25/2022] Open
|
22
|
Su Y, Zhang X, Li S, Xie W, Guo J. Emerging roles of the copper-CTR1 axis in tumorigenesis. Mol Cancer Res 2022; 20:1339-1353. [PMID: 35604085 DOI: 10.1158/1541-7786.mcr-22-0056] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 03/09/2022] [Accepted: 05/17/2022] [Indexed: 11/16/2022]
Abstract
Physiological roles of copper in metabolic homeostasis have been well established, however, whether and how copper is dysregulated in tumors and contributes to tumorigenesis are not recapitulated. Here, we comprehensively summarize the potential origins of copper accumulation in diseases especially in cancers by dysregulating copper transporter 1 (CTR1) or ATPase copper transporting alpha/beta (ATP7A/B) and further demonstrate the underlying mechanism of copper contributing to tumorigenesis. Specifically, in addition to modulating reactive oxygen species (ROS), angiogenesis, immune response, and metabolic homeostasis, copper recently has drawn more attention by directly binding to oncoproteins such as MEK, ULK, Memo, and PDK1 to activate distinct oncogenic signals and account for tumorigenesis. In the end, we disclose the emerging applications of copper in cancer diagnosis and highlight the promising strategies to target the copper-CTR1 axis for cancer therapies.
Collapse
Affiliation(s)
- Yaqing Su
- First Affiliated Hospital of Sun Yat-sen University, guangzhou, guangdong, China
| | - Xiaomei Zhang
- First Affiliated Hospital of Sun Yat-sen University, China
| | - Shaoqiang Li
- The First Affiliatd Hospital of Sun Yat-sen University, Guangzhou, China
| | - Wei Xie
- First Affiliated Hospital of Sun Yat-sen University, China
| | - Jianping Guo
- First Affiliated Hospital of Sun Yat-sen University, guangzhou, guangdong, China
| |
Collapse
|
23
|
Feng B, Wu J, Shen B, Jiang F, Feng J. Cancer-associated fibroblasts and resistance to anticancer therapies: status, mechanisms, and countermeasures. Cancer Cell Int 2022; 22:166. [PMID: 35488263 PMCID: PMC9052457 DOI: 10.1186/s12935-022-02599-7] [Citation(s) in RCA: 57] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 04/19/2022] [Indexed: 12/12/2022] Open
Abstract
Cancer-associated fibroblasts (CAFs) are critical components of the tumor microenvironment (TME) with diverse functions such as extracellular matrix (ECM) remodeling, modulation of metabolism and angiogenesis, and crosstalk with both cancer cells and infiltrating immune cells by production of growth factors, cytokines, and chemokines. Within the TME milieu, CAFs exhibit morphological and functional transitions with relatively specific markers and hold tremendous potential to facilitate tumorigenesis, development, and resistance towards multiple therapeutic strategies including chemotherapy, radiotherapy, targeted therapy, anti-angiogenesis therapy, immunotherapy, and endocrine therapy. Accordingly, CAFs themselves and the downstream effectors and/or signaling pathways are potential targets for optimizing the sensitivity of anti-cancer therapies. This review aims to provide a detailed landscape of the role that CAFs play in conferring therapeutic resistance in different cancers and the underlying mechanisms. The translational and therapeutic perspectives of CAFs in the individualized treatment of malignant tumors are also discussed.
Collapse
Affiliation(s)
- Bing Feng
- Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, 42 Baiziting, Nanjing, 210009, China
| | - Jianzhong Wu
- Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, 42 Baiziting, Nanjing, 210009, China
| | - Bo Shen
- Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, 42 Baiziting, Nanjing, 210009, China
| | - Feng Jiang
- Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, 42 Baiziting, Nanjing, 210009, China.
| | - Jifeng Feng
- Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, 42 Baiziting, Nanjing, 210009, China.
| |
Collapse
|
24
|
Perryman L, Gray SG. Fibrosis in Mesothelioma: Potential Role of Lysyl Oxidases. Cancers (Basel) 2022; 14:981. [PMID: 35205728 PMCID: PMC8870010 DOI: 10.3390/cancers14040981] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 02/07/2022] [Accepted: 02/08/2022] [Indexed: 12/13/2022] Open
Abstract
Immunotherapies (such as checkpoint inhibitors) and standard chemotherapies (such as cisplatin) have limitations in the successful treatment of malignant pleural mesothelioma (MPM). Fibrosis is the accumulation of collagen in the extracellular matrix (ECM) of tissues, making them denser than that of healthy tissues and thereby affecting drug delivery and immune cell infiltration. Moreover, fibrosis severely affects the patient's breathing and quality of life. The production of collagen and its assembly is highly regulated by various enzymes such as lysyl oxidases. Many solid tumors aberrantly express the family of lysyl oxidases (LOX/LOXL). This review examines how LOX/LOXLs were found to be dysregulated in noncancerous and cancerous settings, discusses their roles in solid tumor fibrosis and pathogenesis and explores the role of fibrosis in the development and poor clinical outcomes of patients with MPM. We examine the current preclinical status of drugs targeting LOX/LOXLs and how the incorporation of such drugs may have therapeutic benefits in the treatment and management of patients with MPM.
Collapse
Affiliation(s)
- Lara Perryman
- Drug Discovery Department, Pharmaxis Ltd., Sydney, NSW 2086, Australia;
| | - Steven G. Gray
- Thoracic Oncology, Labmed Directorate, St James’s Hospital, D08 RX0X Dublin, Ireland
| |
Collapse
|
25
|
Meng K, Hu X, Zheng G, Qian C, Xin Y, Guo H, He R, Ge M, Xu J. Identification of prognostic biomarkers for papillary thyroid carcinoma by a weighted gene co‐expression network analysis. Cancer Med 2022; 11:2006-2019. [PMID: 35152572 PMCID: PMC9089218 DOI: 10.1002/cam4.4602] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 12/07/2021] [Accepted: 12/08/2021] [Indexed: 12/13/2022] Open
Abstract
Aim Methods Results Conclusions
Collapse
Affiliation(s)
- Kexin Meng
- Otolaryngology& Head and Neck Center, Cancer Center, Department of Head and Neck Surgery Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College) Hangzhou Zhejiang China
- Key Laboratory of Endocrine Gland Diseases of Zhejiang Province Hangzhou Zhejiang China
| | - Xiaotian Hu
- Otolaryngology& Head and Neck Center, Cancer Center, Department of Head and Neck Surgery Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College) Hangzhou Zhejiang China
- Qingdao University Qingdao Shandong China
| | - Guowan Zheng
- Otolaryngology& Head and Neck Center, Cancer Center, Department of Head and Neck Surgery Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College) Hangzhou Zhejiang China
- Key Laboratory of Endocrine Gland Diseases of Zhejiang Province Hangzhou Zhejiang China
| | - Chenhong Qian
- Otolaryngology& Head and Neck Center, Cancer Center, Department of Head and Neck Surgery Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College) Hangzhou Zhejiang China
- Bengbu Medical College Bengbu China
| | - Ying Xin
- Otolaryngology& Head and Neck Center, Cancer Center, Department of Head and Neck Surgery Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College) Hangzhou Zhejiang China
- Key Laboratory of Endocrine Gland Diseases of Zhejiang Province Hangzhou Zhejiang China
| | - Haiwei Guo
- Otolaryngology& Head and Neck Center, Cancer Center, Department of Head and Neck Surgery Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College) Hangzhou Zhejiang China
- Key Laboratory of Endocrine Gland Diseases of Zhejiang Province Hangzhou Zhejiang China
| | - Ru He
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College Hangzhou China
| | - Minghua Ge
- Otolaryngology& Head and Neck Center, Cancer Center, Department of Head and Neck Surgery Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College) Hangzhou Zhejiang China
- Key Laboratory of Endocrine Gland Diseases of Zhejiang Province Hangzhou Zhejiang China
| | - Jiajie Xu
- Otolaryngology& Head and Neck Center, Cancer Center, Department of Head and Neck Surgery Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College) Hangzhou Zhejiang China
- Key Laboratory of Endocrine Gland Diseases of Zhejiang Province Hangzhou Zhejiang China
| |
Collapse
|
26
|
Wang L, Cao S, Zhai R, Zhao Y, Song G. Systematic Analysis of Expression and Prognostic Values of Lysyl Oxidase Family in Gastric Cancer. Front Genet 2022; 12:760534. [PMID: 35126449 PMCID: PMC8812723 DOI: 10.3389/fgene.2021.760534] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 12/16/2021] [Indexed: 01/14/2023] Open
Abstract
Background: Gastric cancer (GC) remains the fifth most commonly diagnosed malignancy worldwide, with a poor prognosis. The lysyl oxidase (LOX) family, a type of secreted copper-dependent amine oxidases, is comprised of LOX and four LOX-like (LOXL) 1–4 isoforms and has been reported to be dysregulated in a number of different type cancers. However, the diverse expression patterns and prognostic values of LOX family in GC have yet to be systematically analyzed. Methods: ONCOMINE, GEPIA, UALCAN, Kaplan–Meier Plotter, LOGpc, cBioPortal, GeneMANIA and Metascape databases were utilized in this study to analyze the expression, prognostic values, mutations and functional networks of LOX family in GC. Results: The mRNA expression levels of LOX, LOXL1 and LOXL2 were significantly higher in GC, the expression level of LOXL3 was contrary in different databases, while the expression level of LOXL4 made no difference; the expression levels of LOX, LOXL1 and LOXL3 were higher in stages 2–4 than that of normal tissues and stage 1, while the mRNA level of LOXL2 in stage 1–4 was higher than normal tissues; patients with high expression of LOX and LOXL 2-4 had poor OS; the genes correlated with LOX and LOXL2 were enriched in extracellular matrix organization, vasculature development and skeletal system development. Conclusion: Our results indicated that the LOX family, especially LOX and LOXL2, might play an important role in GC oncogenesis, and they may become biomarkers for predicting tumor prognosis and potential targets for tumor therapy.
Collapse
Affiliation(s)
- Li Wang
- Department of Gastrointestinal Surgery, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Shan Cao
- Department of Respiratory, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Rujun Zhai
- Department of Gastrointestinal Surgery, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Yang Zhao
- Radiology Department, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Guodong Song
- Department of Gastrointestinal Surgery, The Second Hospital of Tianjin Medical University, Tianjin, China
| |
Collapse
|
27
|
Jones JO, Moody WM, Shields JD. Microenvironmental modulation of the developing tumour: an immune-stromal dialogue. Mol Oncol 2021; 15:2600-2633. [PMID: 32741067 PMCID: PMC8486574 DOI: 10.1002/1878-0261.12773] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 07/03/2020] [Accepted: 07/27/2020] [Indexed: 12/17/2022] Open
Abstract
Successful establishment of a tumour relies on a cascade of interactions between cancer cells and stromal cells within an evolving microenvironment. Both immune and nonimmune cellular components are key factors in this process, and the individual players may change their role from tumour elimination to tumour promotion as the microenvironment develops. While the tumour-stroma crosstalk present in an established tumour is well-studied, aspects in the early tumour or premalignant microenvironment have received less attention. This is in part due to the challenges in studying this process in the clinic or in mouse models. Here, we review the key anti- and pro-tumour factors in the early microenvironment and discuss how understanding this process may be exploited in the clinic.
Collapse
Affiliation(s)
- James O. Jones
- MRC Cancer UnitHutchison/MRC Research CentreUniversity of CambridgeCambridgeUK
- Department of OncologyCambridge University Hospitals NHS Foundation TrustCambridgeUK
| | - William M. Moody
- MRC Cancer UnitHutchison/MRC Research CentreUniversity of CambridgeCambridgeUK
| | | |
Collapse
|
28
|
Cancer-associated fibroblasts: overview, progress, challenges, and directions. Cancer Gene Ther 2021; 28:984-999. [PMID: 33712707 DOI: 10.1038/s41417-021-00318-4] [Citation(s) in RCA: 159] [Impact Index Per Article: 53.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Revised: 02/12/2021] [Accepted: 02/25/2021] [Indexed: 01/30/2023]
Abstract
Tumors are one of the main causes of death in humans. The development of safe and effective methods for early diagnosis and treatment of tumors is a difficult problem that needs to be solved urgently. It is well established that the occurrence of tumors involves complex biological mechanisms, and the tumor microenvironment (TME) plays an important role in regulating the biological behavior of tumors. Cancer-associated fibroblasts (CAFs) are a group of activated fibroblasts with significant heterogeneity and plasticity in the tumor microenvironment. They secrete a variety of active factors to regulate tumor occurrence, development, metastasis, and therapeutic resistance. Although most studies suggest that CAFs have significant tumor-promoting functions, some evidence indicates that they may have certain tumor-suppressive functions in the early stage of tumors. Current research on CAFs continues to face many challenges, and the heterogeneity of their origin, phenotype, and function is a major difficulty and hot spot. To provide new perspectives for the research on CAFs and tumor diagnosis and treatment, this review summarizes the definition, origin, biomarkers, generation mechanism, functions, heterogeneity, plasticity, subpopulations, pre-metastasis niches (PMN), immune microenvironment, and targeted therapy of CAFs, describes the research progress and challenges, and proposes possible future research directions based on existing reports.
Collapse
|
29
|
Chen C, Hou J, Yu S, Li W, Wang X, Sun H, Qin T, Claret FX, Guo H, Liu Z. Role of cancer-associated fibroblasts in the resistance to antitumor therapy, and their potential therapeutic mechanisms in non-small cell lung cancer. Oncol Lett 2021; 21:413. [PMID: 33841574 PMCID: PMC8020389 DOI: 10.3892/ol.2021.12674] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Accepted: 02/26/2021] [Indexed: 12/13/2022] Open
Abstract
Non-small cell lung cancer (NSCLC) is a malignant tumor with high morbidity and mortality rates, which seriously endangers human health. Although treatment methods continue to evolve, the emergence of drug resistance is inevitable and seriously hinders the treatment of NSCLC. The tumor microenvironment (TME) protects tumor cells from the effects of chemotherapeutic drugs, which can lead to drug resistance. Cancer-associated fibroblasts (CAFs) are an important component of the TME, and various studies have demonstrated that CAFs play a crucial role in drug resistance in NSCLC. However, the drug resistance mechanism of CAFs and whether CAFs can be used as a target to reverse the resistance of tumor cells remain unclear. The present review discusses this issue and describes the heterogeneity of CAF markers, as well as their origins and resident organs, and the role and mechanism of this heterogeneity in NSCLC progression. Furthermore, the mechanism of CAF-mediated NSCLC resistance to chemotherapy, targeted therapy and immunotherapy is introduced, and strategies to reverse this resistance are described.
Collapse
Affiliation(s)
- Congcong Chen
- School of Life Science, Northwest University, Xi'an, Shaanxi 710069, P.R. China
| | - Jia Hou
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Sizhe Yu
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Wenyuan Li
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Xiao Wang
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Hong Sun
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Tianjie Qin
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Francois X. Claret
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston TX77030, USA
| | - Hui Guo
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
- Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education of China, Xi'an, Shaanxi 710061, P.R. China
| | - Zhiyan Liu
- School of Life Science, Northwest University, Xi'an, Shaanxi 710069, P.R. China
- Department of Respiratory and Critical Care Medicine, Xi'an No. 3 Hospital, The Affiliated Hospital of Northwest University, Xi'an, Shaanxi 710018, P.R. China
| |
Collapse
|
30
|
Mahboubi-Rabbani M, Zarghi A. Lipoxygenase Inhibitors as Cancer Chemopreventives: Discovery, Recent Developments and Future Perspectives. Curr Med Chem 2021; 28:1143-1175. [PMID: 31820690 DOI: 10.2174/0929867326666191210104820] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 10/31/2019] [Accepted: 11/10/2019] [Indexed: 11/22/2022]
Abstract
BACKGROUND Leukotrienes (LTs) constitute a bioactive group of Polyunsaturated Fatty Acid (PUFA) metabolites molded by the enzymatic activity of lipoxygenase (LO) and have a pivotal role in inflammation and allergy. Evidence is accumulating both by in vitro cell culture experiments and animal tumor model studies in support of the direct involvement of aberrant metabolism of arachidonic acid (ACD) in the development of several types of human cancers such as lung, prostate, pancreatic and colorectal malignancies. Several independent experimental data suggest a correlation between tumoral cells viability and LO gene expression, especially, 5-lipoxygenase (5-LO). Overexpressed 5-LO cells live longer, proliferate faster, invade more effectively through extracellular matrix destruction and activate the anti-apoptotic signaling mechanisms more intensively compared to the normal counterparts. Thus, some groups of lipoxygenase inhibitors may be effective as promising chemopreventive agents. METHODS A structured search of bibliographic databases for peer-reviewed research literature regarding the role of LO in the pathogenesis of cancer was performed. The characteristics of screened papers were summarized and the latest advances focused on the discovery of new LO inhibitors as anticancer agents were discussed. RESULTS More than 180 papers were included and summarized in this review; the majority was about the newly designed and synthesized 5-LO inhibitors as anti-inflammatory and anticancer agents. The enzyme's structure, 5-LO pathway, 5-LO inhibitors structure-activity relationships as well as the correlation between these drugs and a number of most prevalent human cancers were described. In most cases, it has been emphasized that dual cyclooxygenase-2/5-lipoxygenase (COX-2/5-LO) or dual 5-lipoxygenase/microsomal prostaglandin E synthase-1 (5-LO/mPGES-1) inhibitors possess considerable inhibitory activities against their target enzymes as well as potent antiproliferative effects. Several papers disclosing 5-lipoxygenase activating protein (FLAP) antagonists as a new group of 5-LO activity regulators are also subject to this review. Also, the potential of 12-lipoxygenase (12- LO) and 15-lipoxygenase (15-LO) inhibitors as chemopreventive agents was outlined to expand the scope of new anticancer agents discovery. Some peptides and peptidomimetics with anti-LT activities were described as well. In addition, the cytotoxic effects of lipoxygenase inhibitors and their adverse effects were discussed and some novel series of natural-product-derived inhibitors of LO was also discussed in this review. CONCLUSION This review gives insights into the novel lipoxygenase inhibitors with anticancer activity as well as the different molecular pharmacological strategies to inhibit the enzyme effectively. The findings confirm that certain groups of LO inhibitors could act as promising chemopreventive agents.
Collapse
Affiliation(s)
- Mohammad Mahboubi-Rabbani
- Department of Medicinal Chemistry, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Afshin Zarghi
- Department of Medicinal Chemistry, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
31
|
Targeting Lysyl Oxidase Family Meditated Matrix Cross-Linking as an Anti-Stromal Therapy in Solid Tumours. Cancers (Basel) 2021; 13:cancers13030491. [PMID: 33513979 PMCID: PMC7865543 DOI: 10.3390/cancers13030491] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 01/20/2021] [Accepted: 01/22/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary To improve efficacy of solid cancer treatment, efforts have shifted towards targeting both the cancer cells and the surrounding tumour tissue they grow in. The lysyl oxidase (LOX) family of enzymes underpin the fibrotic remodeling of the tumour microenvironment to promote both cancer growth, spread throughout the body and modulate response to therapies. This review examines how the lysyl oxidase family is involved in tumour development, how they can be targeted, and their potential as diagnostic and prognostic biomarkers in solid tumours. Abstract The lysyl oxidase (LOX) family of enzymes are a major driver in the biogenesis of desmoplastic matrix at the primary tumour and secondary metastatic sites. With the increasing interest in and development of anti-stromal therapies aimed at improving clinical outcomes of cancer patients, the Lox family has emerged as a potentially powerful clinical target. This review examines how lysyl oxidase family dysregulation in solid cancers contributes to disease progression and poor patient outcomes, as well as an evaluation of the preclinical landscape of LOX family targeting therapeutics. We also discuss the suitability of the LOX family as a diagnostic and/or prognostic marker in solid tumours.
Collapse
|
32
|
James DS, Campagnola PJ. Recent Advancements in Optical Harmonic Generation Microscopy: Applications and Perspectives. BME FRONTIERS 2021; 2021:3973857. [PMID: 37849910 PMCID: PMC10521653 DOI: 10.34133/2021/3973857] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 12/14/2020] [Indexed: 10/19/2023] Open
Abstract
Second harmonic generation (SHG) and third harmonic generation (THG) microscopies have emerged as powerful imaging modalities to examine structural properties of a wide range of biological tissues. Although SHG and THG arise from very different contrast mechanisms, the two are complimentary and can often be collected simultaneously using a modified multiphoton microscope. In this review, we discuss the needed instrumentation for these modalities as well as the underlying theoretical principles of SHG and THG in tissue and describe how these can be leveraged to extract unique structural information. We provide an overview of recent advances showing how SHG microscopy has been used to evaluate collagen alterations in the extracellular matrix and how this has been used to advance our knowledge of cancers, fibroses, and the cornea, as well as in tissue engineering applications. Specific examples using polarization-resolved approaches and machine learning algorithms are highlighted. Similarly, we review how THG has enabled developmental biology and skin cancer studies due to its sensitivity to changes in refractive index, which are ubiquitous in all cell and tissue assemblies. Lastly, we offer perspectives and outlooks on future directions of SHG and THG microscopies and present unresolved questions, especially in terms of overall miniaturization and the development of microendoscopy instrumentation.
Collapse
Affiliation(s)
- Darian S. James
- Department of Biomedical Engineering, University of Wisconsin-Madison, 1550 Engineering Dr, Madison, WI 53706, USA
| | - Paul J. Campagnola
- Department of Biomedical Engineering, University of Wisconsin-Madison, 1550 Engineering Dr, Madison, WI 53706, USA
| |
Collapse
|
33
|
Yuan R, Li Y, Yang B, Jin Z, Xu J, Shao Z, Miao H, Ren T, Yang Y, Li G, Song X, Hu Y, Wang X, Huang Y, Liu Y. LOXL1 exerts oncogenesis and stimulates angiogenesis through the LOXL1-FBLN5/αvβ3 integrin/FAK-MAPK axis in ICC. MOLECULAR THERAPY. NUCLEIC ACIDS 2021; 23:797-810. [PMID: 33614230 PMCID: PMC7868718 DOI: 10.1016/j.omtn.2021.01.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 01/05/2021] [Indexed: 02/07/2023]
Abstract
Aberrant expression of lysyl oxidase-like 1 (LOXL1) reportedly leads to fibrous diseases. Recent studies have revealed its role in cancers. In this study, we observed an elevated level of LOXL1 in the tissues and sera of patients with intrahepatic cholangiocarcinoma (ICC) compared with levels in nontumor tissues and sera of unaffected individuals. Overexpression of LOXL1 in RBE and 9810 cell lines promoted cell proliferation, colony formation, and metastasis in vivo and in vitro and induced angiogenesis. In contrast, depletion of LOXL1 showed the opposite effects. We further showed that LOXL1 interacted with fibulin 5 (FBLN5), which regulates angiogenesis, through binding to the αvβ3 integrin in an arginine-glycine-aspartic (Arg-Gly-Asp) domain-dependent mechanism and enhanced the focal adhesion kinase (FAK)-mitogen-activated protein kinase (MAPK) signaling pathway inside vascular endothelial cells. Our findings shed light on the molecular mechanism underlying LOXL1 regulation of angiogenesis in ICC development and indicate that the LOXL1-FBLN5/αvβ3 integrin/FAK-MAPK axis might be the critical pathological link leading to angiogenesis in ICC.
Collapse
Affiliation(s)
- Ruiyan Yuan
- State Key Laboratory of Oncogenes and Related Genes, Department of General Surgery, Shanghai Key Laboratory of Biliary Tract Disease Research, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Yang Li
- State Key Laboratory of Oncogenes and Related Genes, Department of General Surgery, Shanghai Key Laboratory of Biliary Tract Disease Research, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Bo Yang
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, Department of Surgery, First Affiliated Hospital of Wenzhou Medical University, Baixiang Road, Wenzhou 325000, China
| | - Zhaohui Jin
- State Key Laboratory of Oncogenes and Related Genes, Department of General Surgery, Shanghai Key Laboratory of Biliary Tract Disease Research, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Jiacheng Xu
- Endoscopy Center and Endoscopy Research Institute, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Shanghai 200032, China
| | - Ziyu Shao
- State Key Laboratory of Oncogenes and Related Genes, Department of General Surgery, Shanghai Key Laboratory of Biliary Tract Disease Research, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Huijie Miao
- State Key Laboratory of Oncogenes and Related Genes, Department of General Surgery, Shanghai Key Laboratory of Biliary Tract Disease Research, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Tai Ren
- State Key Laboratory of Oncogenes and Related Genes, Department of General Surgery, Shanghai Key Laboratory of Biliary Tract Disease Research, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Yang Yang
- State Key Laboratory of Oncogenes and Related Genes, Department of General Surgery, Shanghai Key Laboratory of Biliary Tract Disease Research, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Guoqiang Li
- State Key Laboratory of Oncogenes and Related Genes, Department of General Surgery, Shanghai Key Laboratory of Biliary Tract Disease Research, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Xiaoling Song
- State Key Laboratory of Oncogenes and Related Genes, Department of General Surgery, Shanghai Key Laboratory of Biliary Tract Disease Research, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Yunping Hu
- State Key Laboratory of Oncogenes and Related Genes, Department of General Surgery, Shanghai Key Laboratory of Biliary Tract Disease Research, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Xu'an Wang
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200120, China
| | - Ying Huang
- State Key Laboratory of Oncogenes and Related Genes, Department of General Surgery, Shanghai Key Laboratory of Biliary Tract Disease Research, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Yingbin Liu
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200120, China
| |
Collapse
|
34
|
Hu Q, Masuda T, Kuramitsu S, Tobo T, Sato K, Kidogami S, Nambara S, Ueda M, Tsuruda Y, Kuroda Y, Ito S, Oki E, Mori M, Mimori K. Potential association of LOXL1 with peritoneal dissemination in gastric cancer possibly via promotion of EMT. PLoS One 2020; 15:e0241140. [PMID: 33095806 PMCID: PMC7584171 DOI: 10.1371/journal.pone.0241140] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 10/08/2020] [Indexed: 12/27/2022] Open
Abstract
Background Peritoneal dissemination (PD) frequently occurs in gastric cancer (GC) and is incurable. In this study, we aimed to identify novel PD-associated genes and clarify their clinical and biological significance in GC. Materials and methods We identified LOXL1 as a PD-associated candidate gene by in silico analysis of GC datasets (highly disseminated peritoneal GC cell line and two freely available GC datasets, GSE15459 and TCGA). Next, we evaluated the clinical significance of LOXL1 expression using RT-qPCR and immunohistochemistry staining (IHC) in a validation cohort (Kyushu cohort). Moreover, we performed gene expression analysis, including gene set enrichment analysis (GSEA) with GSE15459 and TCGA datasets. Finally, we performed a series of in vitro experiments using GC cells. Results In silico analysis showed that LOXL1 was overexpressed in tumor tissues of GC patients with PD and in highly disseminated peritoneal GC cells, relative to that in the control GC patients and cells, respectively. High expression of LOXL1 was a poor prognostic factor in the TCGA dataset. Next, IHC showed that LOXL1 was highly expressed in GC cells. High LOXL1 mRNA expression was associated with poorly differentiated histological type, lymph node metastasis, and was an independent poor prognostic factor in the Kyushu validation cohort. Moreover, LOXL1 expression was positively correlated with the EMT (epithelial-mesenchymal transition) gene set in GSEA. Finally, LOXL1-overexpressing GC cells changed their morphology to a spindle-like form. LOXL1 overexpression reduced CDH1 expression; increased the expression of VIM, CDH2, SNAI2, and PLS3; and promoted the migration capacity of GC cells. Conclusions LOXL1 is associated with PD in GC, possibly through the induction of EMT.
Collapse
Affiliation(s)
- Qingjiang Hu
- Department of Surgery, Kyushu University Beppu Hospital, Beppu, Japan
- Department of Surgery and Science, Kyushu University Hospital, Fukuoka, Japan
| | - Takaaki Masuda
- Department of Surgery, Kyushu University Beppu Hospital, Beppu, Japan
| | - Shotaro Kuramitsu
- Department of Surgery, Kyushu University Beppu Hospital, Beppu, Japan
| | - Taro Tobo
- Department of Clinical Laboratory Medicine, Kyushu University Beppu Hospital, Beppu, Japan
| | - Kuniaki Sato
- Department of Surgery, Kyushu University Beppu Hospital, Beppu, Japan
| | - Shinya Kidogami
- Department of Surgery, Kyushu University Beppu Hospital, Beppu, Japan
| | - Sho Nambara
- Department of Surgery, Kyushu University Beppu Hospital, Beppu, Japan
| | - Masami Ueda
- Department of Surgery, Kyushu University Beppu Hospital, Beppu, Japan
| | - Yusuke Tsuruda
- Department of Surgery, Kyushu University Beppu Hospital, Beppu, Japan
| | - Yosuke Kuroda
- Department of Surgery, Kyushu University Beppu Hospital, Beppu, Japan
| | - Shuhei Ito
- Department of Surgery, Kyushu University Beppu Hospital, Beppu, Japan
| | - Eiji Oki
- Department of Surgery and Science, Kyushu University Hospital, Fukuoka, Japan
| | - Masaki Mori
- Department of Surgery and Science, Kyushu University Hospital, Fukuoka, Japan
| | - Koshi Mimori
- Department of Surgery, Kyushu University Beppu Hospital, Beppu, Japan
- * E-mail:
| |
Collapse
|
35
|
Parker AL, Cox TR. The Role of the ECM in Lung Cancer Dormancy and Outgrowth. Front Oncol 2020; 10:1766. [PMID: 33014869 PMCID: PMC7516130 DOI: 10.3389/fonc.2020.01766] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 08/06/2020] [Indexed: 12/19/2022] Open
Abstract
The dissemination of tumor cells to local and distant sites presents a significant challenge in the clinical management of many solid tumors. These cells may remain dormant for months or years before overt metastases are re-awakened. The components of the extracellular matrix, their posttranslational modifications and their associated factors provide mechanical, physical and chemical cues to these disseminated tumor cells. These cues regulate the proliferative and survival capacity of these cells and lay the foundation for their engraftment and colonization. Crosstalk between tumor cells, stromal and immune cells within primary and secondary sites is fundamental to extracellular matrix remodeling that feeds back to regulate tumor cell dormancy and outgrowth. This review will examine the role of the extracellular matrix and its associated factors in establishing a fertile soil from which individual tumor cells and micrometastases establish primary and secondary tumors. We will focus on the role of the lung extracellular matrix in providing the architectural support for local metastases in lung cancer, and distant metastases in many solid tumors. This review will define how the matrix and matrix associated components are collectively regulated by lung epithelial cells, fibroblasts and resident immune cells to orchestrate tumor dormancy and outgrowth in the lung. Recent advances in targeting these lung-resident tumor cell subpopulations to prevent metastatic disease will be discussed. The development of novel matrix-targeted strategies have the potential to significantly reduce the burden of metastatic disease in lung and other solid tumors and significantly improve patient outcome in these diseases.
Collapse
Affiliation(s)
- Amelia L Parker
- Garvan Institute of Medical Research, Darlinghurst, NSW, Australia.,St Vincent's Clinical School, UNSW Sydney, Darlinghurst, NSW, Australia
| | - Thomas R Cox
- Garvan Institute of Medical Research, Darlinghurst, NSW, Australia.,St Vincent's Clinical School, UNSW Sydney, Darlinghurst, NSW, Australia
| |
Collapse
|
36
|
Hu L, Wang J, Wang Y, Wu L, Wu C, Mao B, Maruthi Prasad E, Wang Y, Chin YE. LOXL1 modulates the malignant progression of colorectal cancer by inhibiting the transcriptional activity of YAP. Cell Commun Signal 2020; 18:148. [PMID: 32912229 PMCID: PMC7488294 DOI: 10.1186/s12964-020-00639-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 08/07/2020] [Indexed: 12/24/2022] Open
Abstract
Background LOX-like 1 (LOXL1) is a lysyl oxidase, and emerging evidence has revealed its effect on malignant cancer progression. However, its role in colorectal cancer (CRC) and the underlying molecular mechanisms have not yet been elucidated. Methods LOXL1 expression in colorectal cancer was detected by immunohistochemistry, western blotting and real-time PCR. In vitro, colony formation, wound healing, migration and invasion assays were performed to investigate the effects of LOXL1 on cell proliferation, migration and invasion. In vivo, metastasis models and mouse xenografts were used to assess tumorigenicity and metastasis ability. Molecular biology experiments were utilized to reveal the underlying mechanisms by which LOXL1 modulates the Hippo pathway. Results LOXL1 was highly expressed in normal colon tissues compared with cancer tissues. In vitro, silencing LOXL1 in CRC cell lines dramatically enhanced migration, invasion, and colony formation, while overexpression of LOXL1 exerted the opposite effects. The results of the in vivo experiments demonstrated that the overexpression of LOXL1 in CRC cell lines drastically inhibited metastatic progression and tumour growth. Mechanistically, LOXL1 inhibited the transcriptional activity of Yes-associated protein (YAP) by interacting with MST1/2 and increasing the phosphorylation of MST1/2. Conclusions LOXL1 may function as an important tumour suppressor in regulating tumour growth, invasion and metastasis via negative regulation of YAP activity. Video abstract
Graphical abstract ![]()
Collapse
Affiliation(s)
- Lin Hu
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Jing Wang
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Yunliang Wang
- Department of General surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Linpeng Wu
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Chao Wu
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Bo Mao
- School of Biology and Basic Medical Science, Soochow University, Suzhou, China
| | - E Maruthi Prasad
- Department of Cell Biology and Genetics, Shenzhen key of Laboratory of Translational medicine of Tumor, Shenzhen University Health science center, Shenzhen, China
| | - Yuhong Wang
- Department of Pathology, The First Affiliated Hospital of Soochow University, Suzhou, China.
| | - Y Eugene Chin
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China.
| |
Collapse
|
37
|
Wang Z, Gao L, Guo X, Lian W, Deng K, Xing B. Development and Validation of a Novel DNA Methylation-Driven Gene Based Molecular Classification and Predictive Model for Overall Survival and Immunotherapy Response in Patients With Glioblastoma: A Multiomic Analysis. Front Cell Dev Biol 2020; 8:576996. [PMID: 33015072 PMCID: PMC7494802 DOI: 10.3389/fcell.2020.576996] [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: 06/28/2020] [Accepted: 08/18/2020] [Indexed: 12/29/2022] Open
Abstract
Purpose Glioblastoma (GBM) is the most common primary malignant tumor of the central nervous system, with a 5-year overall survival (OS) rate of only 5.6%. This study aimed to develop a novel DNA methylation-driven gene (MDG)-based molecular classification and risk model for individualized prognosis prediction for GBM patients. Methods The DNA methylation profiles (458 samples) and gene expression profiles (376 samples) of patients were enrolled to identify MDGs using the MethylMix algorithm. Unsupervised consensus clustering was performed to develop the MDG-based molecular classification. By performing the univariate, least absolute shrinkage and selection operator (LASSO), and multivariate Cox regression analysis, a MDG-based prognostic model was developed and validated. Then, Bisulfite Amplicon Sequencing (BSAS) and quantitative real-time polymerase chain reaction (qPCR) were performed to verify the methylation and expressions of MDGs in GBM cell lines. Results A total of 199 MDGs were identified, the expression patterns of which enabled TCGA and CGGA GBM patients to be divided into 2 clusters by unsupervised consensus clustering. Cluster 1 patients commonly exhibited a poor prognosis, were older in age, and were more sensitive to immunotherapies. Then, six MDGs (ANKRD10, BMP2, LOXL1, RPL39L, TMEM52, and VILL) were further selected to construct the prognostic risk score model, which was validated in the CGGA cohort. Kaplan-Meier survival analysis demonstrated that high-risk patients had significantly poorer OS than low-risk patients (logrank P = 3.338 × 10-6). Then, a prognostic nomogram was constructed and validated. Calibration plots, receiver operating characteristic curves, and decision curve analysis indicated excellent predictive performance for the nomogram in both the TCGA training and CGGA validation cohorts. Finally, in vitro BSAS and qPCR analysis validated that the expressions of the MDGs were negatively regulated by methylations of target genes, especially promoter region methylation. Conclusion The MDG-based prognostic model could serve as a promising prognostic indicator and potential therapeutic target to facilitate individualized survival prediction and better treatment options for GBM patients.
Collapse
Affiliation(s)
- Zihao Wang
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lu Gao
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaopeng Guo
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wei Lian
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Kan Deng
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Bing Xing
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
38
|
Munir H, Mazzaglia C, Shields JD. Stromal regulation of tumor-associated lymphatics. Adv Drug Deliv Rev 2020; 161-162:75-89. [PMID: 32783989 DOI: 10.1016/j.addr.2020.08.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Revised: 07/27/2020] [Accepted: 08/06/2020] [Indexed: 02/08/2023]
Abstract
Recent advances have identified a growing array of roles played by lymphatics in the tumor microenvironment, from providing a route of metastasis to immune modulation. The tumor microenvironment represents an exceptionally complex, dynamic niche comprised of a diverse mixture of cancer cells and normal host cells termed the stroma. This review discusses our current understanding of stromal elements and how they regulate lymphatic growth and functional properties in the tumor context.
Collapse
Affiliation(s)
- Hafsa Munir
- MRC Cancer Unit, Hutchison/MRC Research Centre, University of Cambridge, Box 197 Cambridge Biomedical Campus, Cambridge, CB2 0XZ
| | - Corrado Mazzaglia
- MRC Cancer Unit, Hutchison/MRC Research Centre, University of Cambridge, Box 197 Cambridge Biomedical Campus, Cambridge, CB2 0XZ
| | - Jacqueline D Shields
- MRC Cancer Unit, Hutchison/MRC Research Centre, University of Cambridge, Box 197 Cambridge Biomedical Campus, Cambridge, CB2 0XZ.
| |
Collapse
|
39
|
Greene AG, Eivers SB, Dervan EWJ, O'Brien CJ, Wallace DM. Lysyl Oxidase Like 1: Biological roles and regulation. Exp Eye Res 2020; 193:107975. [PMID: 32070696 DOI: 10.1016/j.exer.2020.107975] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 01/12/2020] [Accepted: 02/13/2020] [Indexed: 12/11/2022]
Abstract
Lysyl Oxidase Like 1 (LOXL1) is a gene that encodes for the LOXL1 enzyme. This enzyme is required for elastin biogenesis and collagen cross-linking, polymerising tropoelastin monomers into elastin polymers. Its main role is in elastin homeostasis and matrix remodelling during injury, fibrosis and cancer development. Because of its vast range of biological functions, abnormalities in LOXL1 underlie many disease processes. Decreased LOXL1 expression is observed in disorders of elastin such as Cutis Laxa and increased expression is reported in fibrotic disease such as Idiopathic Pulmonary Fibrosis. LOXL1 is also downregulated in the lamina cribrosa in pseudoexfoliation glaucoma and genetic variants in the LOXL1 gene have been linked with an increased risk of developing pseudoexfoliation glaucoma and pseudoexfoliation syndrome. However the two major risk alleles are reversed in certain ethnic groups and are present in a large proportion of the normal population, implying complex genetic and environmental regulation is involved in disease pathogenesis. It also appears that the non-coding variants in intron 1 of LOXL1 may be involved in the regulation of LOXL1 expression. Gene alteration may occur via a number of epigenetic and post translational mechanisms such as DNA methylation, long non-coding RNAs and microRNAs. These may represent future therapeutic targets for disease. Environmental factors such as hypoxia, oxidative stress and ultraviolet radiation exposure alter LOXL1 expression, and it is likely a combination of these genetic and environmental factors that influence disease development and progression. In this review, we discuss LOXL1 properties, biological roles and regulation in detail with a focus on pseudoexfoliation syndrome and glaucoma.
Collapse
Affiliation(s)
- Alison G Greene
- UCD Clinical Research Centre, School of Medicine, University College Dublin, Ireland.
| | - Sarah B Eivers
- UCD Clinical Research Centre, School of Medicine, University College Dublin, Ireland
| | - Edward W J Dervan
- Dept. of Ophthalmology, Mater Misericordiae University Hospital, Eccles Street, Dublin 7, Ireland
| | - Colm J O'Brien
- UCD Clinical Research Centre, School of Medicine, University College Dublin, Ireland; Dept. of Ophthalmology, Mater Misericordiae University Hospital, Eccles Street, Dublin 7, Ireland
| | - Deborah M Wallace
- UCD Clinical Research Centre, School of Medicine, University College Dublin, Ireland
| |
Collapse
|
40
|
Schlötzer-Schrehardt U, Zenkel M. The role of lysyl oxidase-like 1 (LOXL1) in exfoliation syndrome and glaucoma. Exp Eye Res 2019; 189:107818. [PMID: 31563608 DOI: 10.1016/j.exer.2019.107818] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 09/23/2019] [Accepted: 09/25/2019] [Indexed: 12/27/2022]
Abstract
Exfoliation syndrome (XFS) is an age-related systemic disease that affects the extracellular matrix. It increases the risk of glaucoma (exfoliation glaucoma, XFG) and susceptibility to diseases of elastin-rich connective tissues. LOXL1 (lysyl oxidase-like 1) is still recognized as the major genetic effect locus in XFS and XFG in all populations worldwide, although its genetic architecture is incompletely understood. LOXL1 is a key cross-linking enzyme in elastic fiber formation and remodeling, which is compatible with the pathogenetic concept of XFS as a specific type of elastosis. This review provides an overview on the current knowledge about the role of LOXL1 in the etiology and pathophysiology of XFS and XFG. It covers the known genetic associations at the LOXL1 locus, potential mechanisms of gene regulation, implications of LOXL1 in XFS-associated fibrosis and connective tissue homeostasis, its role in the development of glaucoma and associated systemic diseases, and the currently available LOXL1-based in vivo and in vitro models. Finally, it also identifies gaps in knowledge and suggests potential areas for future research.
Collapse
Affiliation(s)
| | - Matthias Zenkel
- Department of Ophthalmology, University of Erlangen-Nürnberg, Schwabachanlage 6, 91054, Erlangen, Germany
| |
Collapse
|
41
|
What Is the Fuss about Integrins and the Tumor Microenvironment? Cancers (Basel) 2019; 11:cancers11091296. [PMID: 31484335 PMCID: PMC6770914 DOI: 10.3390/cancers11091296] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 08/28/2019] [Indexed: 01/17/2023] Open
|
42
|
Zeltz C, Primac I, Erusappan P, Alam J, Noel A, Gullberg D. Cancer-associated fibroblasts in desmoplastic tumors: emerging role of integrins. Semin Cancer Biol 2019; 62:166-181. [PMID: 31415910 DOI: 10.1016/j.semcancer.2019.08.004] [Citation(s) in RCA: 176] [Impact Index Per Article: 35.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 08/01/2019] [Accepted: 08/05/2019] [Indexed: 02/06/2023]
Abstract
The tumor microenvironment (TME) is a complex meshwork of extracellular matrix (ECM) macromolecules filled with a collection of cells including cancer-associated fibroblasts (CAFs), blood vessel associated smooth muscle cells, pericytes, endothelial cells, mesenchymal stem cells and a variety of immune cells. In tumors the homeostasis governing ECM synthesis and turnover is disturbed resulting in abnormal blood vessel formation and excessive fibrillar collagen accumulations of varying stiffness and organization. The disturbed ECM homeostasis opens up for new types of paracrine, cell-cell and cell-ECM interactions with large consequences for tumor growth, angiogenesis, metastasis, immune suppression and resistance to treatments. As a main producer of ECM and paracrine signals the CAF is a central cell type in these events. Whereas the paracrine signaling has been extensively studied in the context of tumor-stroma interactions, the nature of the numerous integrin-mediated cell-ECM interactions occurring in the TME remains understudied. In this review we will discuss and dissect the role of known and potential CAF interactions in the TME, during both tumorigenesis and chemoresistance-induced events, with a special focus on the "interaction landscape" in desmoplastic breast, lung and pancreatic cancers. As an example of the multifaceted mode of action of the stromal collagen receptor integrin α11β1, we will summarize our current understanding on the role of this CAF-expressed integrin in these three tumor types.
Collapse
Affiliation(s)
- Cédric Zeltz
- Department of Biomedicine and Centre for Cancer Biomarkers, University of Bergen, Bergen, Norway; Princess Margaret Cancer Center, University Health Network, Toronto, Canada
| | - Irina Primac
- Laboratory of Tumor and Development Biology, GIGA-Cancer, University of Liege (ULiège), Liege, Belgium
| | - Pugazendhi Erusappan
- Department of Biomedicine and Centre for Cancer Biomarkers, University of Bergen, Bergen, Norway; Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Jahedul Alam
- Department of Biomedicine and Centre for Cancer Biomarkers, University of Bergen, Bergen, Norway
| | - Agnes Noel
- Laboratory of Tumor and Development Biology, GIGA-Cancer, University of Liege (ULiège), Liege, Belgium
| | - Donald Gullberg
- Department of Biomedicine and Centre for Cancer Biomarkers, University of Bergen, Bergen, Norway.
| |
Collapse
|