1
|
Feng C, Li Y, Tai Y, Zhang W, Wang H, Lian S, Jin-Si-Han EEMBK, Liu Y, Li X, Chen Q, He M, Lu Z. A neutrophil extracellular traps-related classification predicts prognosis and response to immunotherapy in colon cancer. Sci Rep 2023; 13:19297. [PMID: 37935721 PMCID: PMC10630512 DOI: 10.1038/s41598-023-45558-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 10/20/2023] [Indexed: 11/09/2023] Open
Abstract
Neutrophil extracellular traps (NETs) have been categorized as a form of inflammatory cell death mode of neutrophils (NETosis) involved in natural immunity and the regulation of adaptive immunity. More and more studies revealed the ability of NETs to reshape the tumor immune microenvironment (TIME) by limiting antitumor effector cells, which may impair the efficacy of immunotherapy. To explore whether NETs-related genes make vital impacts on Colon carcinoma (COAD), we have carried out a systematic analysis and showed several findings in the present work. First, we obtained the patient's data from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) dataset, aiming to detect two NETs-associated subtypes by consensus clustering. For the purpose of annotating the roles of NETs-related pathways, gene ontology enrichment analyses were adopted. Next, we constructed a 6 novel NETs-related genes score using the Least Absolute Shrinkage and Selection Operator (LASSO) Cox regression model. We found that the NETs risk score was notably upregulated in COAD patient samples, and its levels were notably correlated with tumor clinicopathological and immune traits. Then, according to NETs-associated molecular subtypes and the risk signature, this study compared immune cell infiltration calculated through the estimate, CIBERSORT, TIMER, ssGSEA algorithms, tumor immune dysfunction, as well as exclusion (TIDE). Furthermore, we confirm that MPO(myeloperoxidase) was significantly upregulated in COAD patient samples, and its levels were significantly linked to tumor malignancy and clinic outcome. Moreover, multiplex immunohistochemistry (mIHC) spatial analysis confirmed that MPO was closely related to Treg and PD-1 + Treg in spatial location which suggested MPO may paly an important role in TIME formation. Altogether, the obtained results indicated that a six NETs-related genes prognostic signature was conducive to estimating the prognosis and response of chemo-/immuno-therapy of COAD patients.
Collapse
Affiliation(s)
- Cheng Feng
- Department of Colorectal Surgery, Sun Yat-sen University Cancer Center, Sun Yat-sen University, Guangzhou, 510515, Guangdong, China
| | - Yuan Li
- Department of Colorectal Surgery, Sun Yat-sen University Cancer Center, Sun Yat-sen University, Guangzhou, 510515, Guangdong, China
| | - Yi Tai
- Department of Musculoskeletal Oncology, Sun Yat-Senen University Cancer Center, Guangzhou, 510515, Guangdong, China
| | - Weili Zhang
- Department of Colorectal Surgery, Sun Yat-sen University Cancer Center, Sun Yat-sen University, Guangzhou, 510515, Guangdong, China
| | - Hao Wang
- Department of Colorectal Surgery, Sun Yat-sen University Cancer Center, Sun Yat-sen University, Guangzhou, 510515, Guangdong, China
| | - Shaopu Lian
- Department of Colorectal Surgery, Sun Yat-sen University Cancer Center, Sun Yat-sen University, Guangzhou, 510515, Guangdong, China
| | - E-Er-Man-Bie-Ke Jin-Si-Han
- Department of Colorectal Surgery, Sun Yat-sen University Cancer Center, Sun Yat-sen University, Guangzhou, 510515, Guangdong, China
| | - Yuanyuan Liu
- Department of Radiation Oncology, The First Hospital of Hunan University of Chinese Medicine, Changsha, 410021, Hunan, China
| | - Xinghui Li
- Department of Cardiology General Hospital of Xinjiang Military Command, No. 359 Youhao North Road, Saybak District, Urumqi, 830001, Xinjiang, China
| | - Qifeng Chen
- Department of Minimally Invasive Interventional Therapy, Liver Cancer Study and Service Group, Sun Yat-Sen University Cancer Center, Guangzhou, 510515, Guangdong, China.
| | - Meng He
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital and Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, 518116, Guangdong, China.
| | - Zhenhai Lu
- Department of Colorectal Surgery, Sun Yat-sen University Cancer Center, Sun Yat-sen University, Guangzhou, 510515, Guangdong, China.
| |
Collapse
|
2
|
Chen J, Xu Q, Liu D, Li X, Guo M, Chen X, Liao J, Lei R, Li W, Huang H, Saw PE, Song E, Yan X, Nie Y. CD146 promotes malignant progression of breast phyllodes tumor through suppressing DCBLD2 degradation and activating the AKT pathway. Cancer Commun (Lond) 2023; 43:1244-1266. [PMID: 37856423 PMCID: PMC10631482 DOI: 10.1002/cac2.12495] [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: 04/19/2023] [Revised: 09/25/2023] [Accepted: 10/08/2023] [Indexed: 10/21/2023] Open
Abstract
BACKGROUND As a rapid-progressing tumor, breast malignant phyllodes tumors (PTs) are challenged by the lack of effective therapeutic strategies and suitable prognostic markers. This study aimed to clarify the role and mechanism of CD146 on promoting PTs malignant progression, and to identify a novel prognosis marker and treatment target of breast malignant PTs. METHODS The expression and prognostic significance of CD146 in PTs was detected through single-cell RNA-sequencing (scRNA-seq), immunostaining, real-time PCR and other methodologies. Functional experiments including proliferation assay, colony formation assay, transwell assay, and collagen contraction assay were conducted to validate the role of CD146 in malignant progression of PTs. The efficacy of anti-CD146 monoclonal antibody AA98 against malignant PTs was corroborated by a malignant PT organoid model and a PT patient-derived xenograft (PDX) model. Transcriptome sequencing, proteomic analysis, co-immunoprecipitation, and pull-down assay was employed to identify the modulating pathway and additional molecular mechanism. RESULTS In this study, the scRNA-seq analysis of PTs disclosed a CD146-positive characteristic in the α-SMA+ fibroblast subset. Furthermore, a progressive elevation in the level of CD146 was observed with the malignant progression of PTs. More importantly, CD146 was found to serve as an independent predictor for recurrence in PT patients. Furthermore, CD146 was found to augment the viability and invasion of PTs. Mechanistically, CD146 acted as a protective "shield" to prevent the degradation of Discoidin, CUB, and LCCL domain-containing protein 2 (DCBLD2), thereby activating the phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT) signaling pathway and enhancing malignant behaviors of PT cells. In the malignant PT organoid and PDX model, a significant suppression of malignant PT growth was observed after the application of AA98. CONCLUSIONS These findings suggested that CD146 served as an efficacious marker for predicting PT malignant progression and showed promise as a prognosis marker and treatment target of breast malignant PTs. The study further unveiled the essential role of the CD146-DCBLD2/PI3K/AKT axis in the malignant progression of PTs.
Collapse
Affiliation(s)
- Jiewen Chen
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationGuangdong‐Hong Kong Joint Laboratory for RNA Medicine, Medical Research CenterSun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdongP. R. China
- Breast Tumor CenterSun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdongP. R. China
- Department of Breast MedicineAffiliated Foshan Maternity and Child Healthcare HospitalSouthern Medical UniversityFoshanGuangdongP. R. China
| | - Qingji Xu
- Key Laboratory of Protein and Peptide PharmaceuticalInstitute of BiophysicsChinese Academy of SciencesBeijingP. R. China
- College of Life SciencesUniversity of Chinese Academy of SciencesBeijingP. R. China
| | - Dan Liu
- Key Laboratory of Protein and Peptide PharmaceuticalInstitute of BiophysicsChinese Academy of SciencesBeijingP. R. China
| | - Xun Li
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationGuangdong‐Hong Kong Joint Laboratory for RNA Medicine, Medical Research CenterSun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdongP. R. China
- Breast Tumor CenterSun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdongP. R. China
| | - Mingyan Guo
- Department of AnesthesiologySun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdongP. R. China
| | - Xuehui Chen
- Key Laboratory of Protein and Peptide PharmaceuticalInstitute of BiophysicsChinese Academy of SciencesBeijingP. R. China
| | - Jianyou Liao
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationGuangdong‐Hong Kong Joint Laboratory for RNA Medicine, Medical Research CenterSun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdongP. R. China
| | - Rong Lei
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationGuangdong‐Hong Kong Joint Laboratory for RNA Medicine, Medical Research CenterSun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdongP. R. China
- Breast Tumor CenterSun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdongP. R. China
| | - Wende Li
- Guangdong Laboratory Animal Monitoring Institute, Guangdong Key Laboratory of Laboratory AnimalGuangzhouGuangdongP. R. China
| | - Hongyan Huang
- Department of Breast SurgeryZhujiang HospitalSouthern Medical UniversityGuangzhouGuangdongP. R. China
| | - Phei Er Saw
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationGuangdong‐Hong Kong Joint Laboratory for RNA Medicine, Medical Research CenterSun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdongP. R. China
| | - Erwei Song
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationGuangdong‐Hong Kong Joint Laboratory for RNA Medicine, Medical Research CenterSun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdongP. R. China
- Breast Tumor CenterSun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdongP. R. China
| | - Xiyun Yan
- Key Laboratory of Protein and Peptide PharmaceuticalInstitute of BiophysicsChinese Academy of SciencesBeijingP. R. China
- College of Life SciencesUniversity of Chinese Academy of SciencesBeijingP. R. China
- Joint Laboratory of Nanozymes in Zhengzhou UniversitySchool of Basic Medical SciencesZhengzhou UniversityZhengzhouHenanP. R. China
| | - Yan Nie
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationGuangdong‐Hong Kong Joint Laboratory for RNA Medicine, Medical Research CenterSun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdongP. R. China
- Breast Tumor CenterSun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdongP. R. China
| |
Collapse
|
3
|
Nojima S. Class IV semaphorins in disease pathogenesis. Pathol Int 2022; 72:471-487. [PMID: 36066011 DOI: 10.1111/pin.13270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 08/16/2022] [Indexed: 12/01/2022]
Abstract
Semaphorins are a large family of secreted and/or transmembrane proteins, originally identified as proteins that function in axon guidance during neuronal development. However, semaphorins play crucial roles in other physiological and pathological processes, including immune responses, angiogenesis, maintenance of tissue homeostasis, and cancer progression. Class IV semaphorins may be present as transmembrane and soluble forms and are implicated in the pathogenesis of various diseases. This review discusses recent progress on the roles of class IV semaphorins determined by clinical and experimental pathology studies.
Collapse
Affiliation(s)
- Satoshi Nojima
- Department of Pathology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan.,Department of Immunopathology, World Premier International Research Center Initiative (WPI), Immunology Frontier Research Center (IFReC), Osaka University, Suita, Osaka, Japan
| |
Collapse
|
4
|
Xie P, Liu JY, Yan H, Wang ZB, Jiang SL, Li X, Liu ZQ. Pan-cancer analyses identify DCBLD2 as an oncogenic, immunological, and prognostic biomarker. Front Pharmacol 2022; 13:950831. [PMID: 36034778 PMCID: PMC9403722 DOI: 10.3389/fphar.2022.950831] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 07/05/2022] [Indexed: 11/13/2022] Open
Abstract
Discoidin, CUB, and LCCL domain-containing protein 2 (DCBLD2) is a two-domain transmembrane protein-coding gene located on chromosome 3, the protein expressed by which acts as the membrane receptor of semaphorin and vascular endothelial growth factor during the development of axons and blood vessels. Although several research evidences at the cellular and clinical levels have associated DCBLD2 with tumorigenesis, nothing is known regarding this gene from a pan-cancer standpoint. In this study, we systematically analyzed the influence of DCBLD2 on prognosis, cancer staging, immune characteristics, and drug sensitivity in a variety of cancers based on a unified and standardized pan-cancer dataset. In addition, we performed GO enrichment analyses and KEGG analyses of DCBLD2-related genes and DCBLD2-binding proteins. Our results showed that DCBLD2 is a potential oncogenic, immunological as well as a prognostic biomarker in terms of pan-cancer, and is expected to contribute to the improvement of tumor prognosis and the development of targeted therapy.
Collapse
Affiliation(s)
- Pan Xie
- Department of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, and National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacology, Central South University, Changsha, China
| | - Jun-Yan Liu
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, China
| | - Han Yan
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Zhi-Bin Wang
- Department of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, and National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacology, Central South University, Changsha, China
| | - Shi-Long Jiang
- Department of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, and National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacology, Central South University, Changsha, China
| | - Xi Li
- Department of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, and National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacology, Central South University, Changsha, China
- *Correspondence: Zhao-Qian Liu, ; Xi Li,
| | - Zhao-Qian Liu
- Department of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, and National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacology, Central South University, Changsha, China
- *Correspondence: Zhao-Qian Liu, ; Xi Li,
| |
Collapse
|
5
|
Jiang J, Lu Y, Zhang F, Pan T, Zhang Z, Wan Y, Ren X, Zhang R. Semaphorin 4B promotes tumor progression and associates with immune infiltrates in lung adenocarcinoma. BMC Cancer 2022; 22:632. [PMID: 35676688 PMCID: PMC9178879 DOI: 10.1186/s12885-022-09696-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 05/25/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Semaphorins have been found to play important roles in multiple malignancy-related processes. However, the role of Semaphorin 4B (SEMA4B) in lung cancer remains unclear. Here, we aimed to explore the biological functions of SEMA4B in through bioinformatic analysis, in vitro and in vivo assays. In the present study, the possible mechanism by which SEMA4B affected the tumor growth and microenvironment of lung adenocarcinoma (LUAD) were investigated. METHODS The expression of SEMA4B in LUAD was analyzed by bioinformatic analysis and verified by the immunohistochemistry staining. The prognostic value of SEMA4B in LUAD was investigated using the Kaplan-Meier survival and Cox's regression model. After silencing SEMA4B expression, the functions of SEMA4B in LUAD cells were investigated by in vitro experiments, including CCK-8 and plate clone formation. And the effect of SEMA4B on tumor growth and immune infiltration was explored in C57BL/6 mice tumor-bearing models. RESULTS SEMA4B expression was upregulated in LUAD tissues and correlated with later pathological stages and poor prognosis of LUAD patients. Further study found that SEMA4B silencing suppressed the proliferation of lung cancer cells both in vitro and in vivo. Bioinformatic analysis showed that SEMA4B expression was correlated with the increased infiltration of myeloid-derived suppressor cells (MDSCs), T-regs and the decreased infiltration of CD8+ T cell in LUAD. Importantly, in vivo study verified that the infiltration of T-regs and MDSCs in tumor microenvironment (TME) of Xenograft tissues was decreased after SEMA4B silencing. CONCLUSIONS These findings demonstrated SEMA4B might play an oncogenic role in LUAD progression, and be a promising therapeutic target for lung cancer.
Collapse
Affiliation(s)
- Jun Jiang
- Department of Health Service, Base of Health Service, Fourth Military Medical University, Xi'an, China
| | - Yuan Lu
- Department of Respiratory and Critical Care Medicine, Zhongda Hospital, Southeast University, Nanjing, China
| | - Fang Zhang
- Department of Respiratory and Critical Care Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Tao Pan
- Department of Respiratory and Critical Care Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, China
- Translational Medicine Center, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zhipei Zhang
- Department of Thoracic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Yi Wan
- Department of Health Service, Base of Health Service, Fourth Military Medical University, Xi'an, China
| | - Xinling Ren
- Department of Respiratory and Critical Care Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, China.
- Department of Pulmonary Medicine, Shenzhen General Hospital, Shenzhen University, Shenzhen, 518055, Guangdong, China.
| | - Rui Zhang
- State Key Laboratory of Cancer Biology, Department of Immunology, Fourth Military Medical University, Xi'an, 710032, Shaanxi, China.
| |
Collapse
|
6
|
Jiang J, Zhang F, Wan Y, Fang K, Yan ZD, Ren XL, Zhang R. Semaphorins as Potential Immune Therapeutic Targets for Cancer. Front Oncol 2022; 12:793805. [PMID: 35155237 PMCID: PMC8830438 DOI: 10.3389/fonc.2022.793805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 01/04/2022] [Indexed: 11/28/2022] Open
Abstract
Semaphorins are a large class of secreted or membrane-bound molecules. It has been reported that semaphorins play important roles in regulating several hallmarks of cancer, including angiogenesis, metastasis, and immune evasion. Semaphorins and their receptors are widely expressed on tumor cells and immune cells. However, the biological role of semaphorins in tumor immune microenvironment is intricate. The dysregulation of semaphorins influences the recruitment and infiltration of immune cells, leading to abnormal anti-tumor effect. Although the underlying mechanisms of semaphorins on regulating tumor-infiltrating immune cell activation and functions are not fully understood, semaphorins can notably be promising immunotherapy targets for cancer.
Collapse
Affiliation(s)
- Jun Jiang
- Department of Health Service, Fourth Military Medical University, Xi’an, China
- State Key Laboratory of Cancer Biology, Department of Immunology, Fourth Military Medical University, Xi’an, China
| | - Fang Zhang
- Department of Respiratory Medicine, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Yi Wan
- Department of Health Service, Fourth Military Medical University, Xi’an, China
| | - Ke Fang
- Department of Health Service, Fourth Military Medical University, Xi’an, China
| | - Ze-dong Yan
- Department of Biomedical Engineering, Fourth Military Medical University, Xi’an, China
| | - Xin-ling Ren
- Department of Respiratory Medicine, Xijing Hospital, Fourth Military Medical University, Xi’an, China
- Department of Pulmonary Medicine, Shenzhen General Hospital, Shenzhen University, Shenzhen, China
| | - Rui Zhang
- State Key Laboratory of Cancer Biology, Department of Immunology, Fourth Military Medical University, Xi’an, China
| |
Collapse
|
7
|
Suzuki SR, Kuno A, Ozaki H. Cell-to-cell interaction analysis of prognostic ligand-receptor pairs in human pancreatic ductal adenocarcinoma. Biochem Biophys Rep 2021; 28:101126. [PMID: 34522794 PMCID: PMC8426203 DOI: 10.1016/j.bbrep.2021.101126] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 08/25/2021] [Accepted: 08/31/2021] [Indexed: 12/12/2022] Open
Abstract
Cell-to-cell interactions (CCIs) through ligand-receptor (LR) pairs in the tumor microenvironment underlie the poor prognosis of pancreatic ductal adenocarcinoma (PDAC). However, there is scant knowledge of the association of CCIs with PDAC prognosis, which is critical to the identification of potential therapeutic candidates. Here, we sought to identify the LR pairs associated with PDAC patient prognosis by integrating survival analysis and single-cell CCI prediction. Via survival analysis using gene expression from cancer cohorts, we found 199 prognostic LR pairs. CCI prediction based on single-cell RNA-seq data revealed the enriched LR pairs associated with poor prognosis. Notably, the CCIs involved epithelial tumor cells, cancer-associated fibroblasts, and tumor-associated macrophages through integrin-related and ANXA1-FPR pairs. Finally, we determined that CCIs involving 33 poor-prognostic LR pairs were associated with tumor grade. Although the clinical implication of the set of LR pairs must be determined, our results may provide potential therapeutic targets in PDAC.
Collapse
Affiliation(s)
- Sayaka R. Suzuki
- Program in Human Biology, School of Integrative and Global Majors, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575, Japan
- Bioinformatics Laboratory, Faculty of Medicine, University of Tsukuba, Tsukuba 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8577, Japan
| | - Akihiro Kuno
- Program in Human Biology, School of Integrative and Global Majors, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575, Japan
- Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, Tsukuba 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8577, Japan
| | - Haruka Ozaki
- Bioinformatics Laboratory, Faculty of Medicine, University of Tsukuba, Tsukuba 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8577, Japan
- Center for Artificial Intelligence Research, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8577, Japan
| |
Collapse
|
8
|
Han L, Zheng C, Wu S. Long non-coding RNA NEAT1 promotes the malignancy of laryngeal squamous cell carcinoma by regulating the microRNA-204-5p/SEMA4B axis. Oncol Lett 2021; 22:802. [PMID: 34630709 PMCID: PMC8477075 DOI: 10.3892/ol.2021.13063] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Accepted: 06/11/2021] [Indexed: 12/18/2022] Open
Abstract
Laryngeal squamous cell carcinoma (LSCC) is a highly invasive malignant tumor in the head and neck area. As an oncogene, long non-coding RNA (lncRNA) nuclear enriched abundant transcript 1 (NEAT1) promotes cell proliferation, migration and invasion several types of cancer. The present study aimed to reveal the effects of NEAT1 on the progression of LSCC. Reverse transcription-quantitative PCR (RT-qPCR) was used to detect relative mRNA expression levels of NEAT1, microRNA (miR)-204-5p and semaphorin (SEMA) 4B. Kaplan-Meier analysis was used to analyze overall survival times. RNA in-situ hybridization (ISH) exhibited the distribution of NEAT1 and miR-204-5p in tissues. RNA fluorescence ISH was used to analyze the distribution of NEAT1 and miR-204-5p in the cells. Western blot analysis was used to detect the expression level of target proteins. Cell viability was analyzed using a MTT assay, while flow cytometry was used to determine cell apoptosis. Wound healing and Transwell invasion assays were used to value cell migration and invasion, respectively. RNA immunoprecipitation assay, bioinformatics prediction and a dual luciferase reporter assay were used to analyze the target relationship. The RT-qPCR results showed that NEAT1 was highly expressed and miR-204-5p had decreased expression in LSCC tissues and cells compared with that in the normal tissue and the 16HBE-14o cell line, respectively. Knockdown of NEAT1 using small interfering (si) RNA and overexpressed miR-204-5p both effectively inhibited the proliferation, migration and invasion of LSCC cells. Besides, further experiments revealed that miR-204-5p was a target of NEAT1. At the same time, silenced miR-204-5p reversed the anti-tumor effects of si-NEAT1. In addition, SEMA4B was targeted by miR-204-5p in LSCC cells and upregulated SEMA4B weakened the antitumor effects of miR-204-5p in LSCC cells. NEAT1 regulated the expression of SEMA4B by targeting miR-204-5p in LSCC cells. Overall, NEAT1 promoted the proliferation and invasion of LSCC cells by regulating the miR-204-5p/SEMA4B axis.
Collapse
Affiliation(s)
- Ling Han
- Department of Otorhinolaryngology, Shenzhen People's Hospital, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, Guangdong 518020, P.R. China
| | - Chaopan Zheng
- Department of Otorhinolaryngology, Shenzhen People's Hospital, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, Guangdong 518020, P.R. China
| | - Shihai Wu
- Department of Radiation Oncology, Shenzhen People's Hospital, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, Guangdong 518020, P.R. China
| |
Collapse
|
9
|
Alhamoudi KM, Barhoumi T, Al-Eidi H, Asiri A, Nashabat M, Alaamery M, Alharbi M, Alhaidan Y, Tabarki B, Umair M, Alfadhel M. A homozygous nonsense mutation in DCBLD2 is a candidate cause of developmental delay, dysmorphic features and restrictive cardiomyopathy. Sci Rep 2021; 11:12861. [PMID: 34145321 PMCID: PMC8213761 DOI: 10.1038/s41598-021-92026-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 05/13/2021] [Indexed: 12/24/2022] Open
Abstract
DCBLD2 encodes discodin, CUB and LCCL domain-containing protein 2, a type-I transmembrane receptor that is involved in intracellular receptor signalling pathways and the regulation of cell growth. In this report, we describe a 5-year-old female who presented severe clinical features, including restrictive cardiomyopathy, developmental delay, spasticity and dysmorphic features. Trio-whole-exome sequencing and segregation analysis were performed to identify the genetic cause of the disease within the family. A novel homozygous nonsense variant in the DCBLD2 gene (c.80G > A, p.W27*) was identified as the most likely cause of the patient's phenotype. This nonsense variant falls in the extracellular N-terminus of DCBLD2 and thus might affect proper protein function of the transmembrane receptor. A number of in vitro investigations were performed on the proband's skin fibroblasts compared to normal fibroblasts, which allowed a comprehensive assessment resulting in the functional characterization of the identified DCBLD2 nonsense variant in different cellular processes. Our data propose a significant association between the identified variant and the observed reduction in cell proliferation, cell cycle progression, intracellular ROS, and Ca2 + levels, which would likely explain the phenotypic presentation of the patient as associated with lethal restrictive cardiomyopathy.
Collapse
Affiliation(s)
- Kheloud M Alhamoudi
- Medical Genomics Research Department, King Abdullah International Research Center (KAIMRC), King Saud Bin Abdulaziz University for Health Sciences, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, Riyadh, Kingdom of Saudi Arabia
| | - Tlili Barhoumi
- Medical Core Facility and Research Platforms, King Abdullah International Research Center (KAIMRC), King Saud Bin Abdulaziz University for Health Sciences, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, Riyadh, Kingdom of Saudi Arabia
| | - Hamad Al-Eidi
- Medical Genomics Research Department, King Abdullah International Research Center (KAIMRC), King Saud Bin Abdulaziz University for Health Sciences, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, Riyadh, Kingdom of Saudi Arabia
| | - Abdulaziz Asiri
- Faculty of Applied Medical Sciences, University of Bisha, Al Nakhil, 225, Bisha, 67714, Kingdom of Saudi Arabia
| | - Marwan Nashabat
- Division of Genetics, Department of Pediatrics, King Abdullah Specialized Children's Hospital, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, P.O Box 22490, Riyadh, 11426, Kingdom of Saudi Arabia
| | - Manal Alaamery
- Developmental Medicine Department, King Abdullah International Medical Research Center, King Saud Bin Abdulaziz University for Health Sciences, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, Riyadh, Kingdom of Saudi Arabia
| | - Masheal Alharbi
- Medical Genomics Research Department, King Abdullah International Research Center (KAIMRC), King Saud Bin Abdulaziz University for Health Sciences, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, Riyadh, Kingdom of Saudi Arabia
| | - Yazeid Alhaidan
- Medical Genomics Research Department, King Abdullah International Research Center (KAIMRC), King Saud Bin Abdulaziz University for Health Sciences, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, Riyadh, Kingdom of Saudi Arabia
| | - Brahim Tabarki
- Division of Pediatric Neurology, Department of Pediatrics, Prince Sultan Military Medical City, Riyadh, Kingdom of Saudi Arabia
| | - Muhammad Umair
- Medical Genomics Research Department, King Abdullah International Research Center (KAIMRC), King Saud Bin Abdulaziz University for Health Sciences, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, Riyadh, Kingdom of Saudi Arabia
| | - Majid Alfadhel
- Medical Genomics Research Department, King Abdullah International Research Center (KAIMRC), King Saud Bin Abdulaziz University for Health Sciences, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, Riyadh, Kingdom of Saudi Arabia. .,Division of Genetics, Department of Pediatrics, King Abdullah Specialized Children's Hospital, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, P.O Box 22490, Riyadh, 11426, Kingdom of Saudi Arabia.
| |
Collapse
|
10
|
Proctor DM, Bhat V, Suh M, Reichert H, Jiang X, Thompson CM. Inhalation cancer risk assessment for environmental exposure to hexavalent chromium: Comparison of margin-of-exposure and linear extrapolation approaches. Regul Toxicol Pharmacol 2021; 124:104969. [PMID: 34089813 DOI: 10.1016/j.yrtph.2021.104969] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 05/24/2021] [Accepted: 05/26/2021] [Indexed: 10/21/2022]
Abstract
Hexavalent chromium [Cr(VI)] exists in the ambient air at low concentrations (average upperbound ~0.1 ng/m3) yet airborne concentrations typically exceed EPA's Regional Screening Level for residential exposure (0.012 ng/m3) and other similar benchmarks, which assume a mutagenic mode of action (MOA) and use low-dose linear risk assessment models. We reviewed Cr(VI) inhalation unit risk estimates developed by researchers and regulatory agencies for environmental and occupational exposures and the underlying epidemiologic data, updated a previously published MOA analysis, and conducted dose-response modeling of rodent carcinogenicity data to evaluate the need for alternative exposure-response data and risk assessment approaches. Current research supports the role of non-mutagenic key events in the MOA, with growing evidence for epigenetic modifiers. Animal data show a weak carcinogenic response, even at cytotoxic exposures, and highlight the uncertainties associated with the current epidemiological data used in risk assessment. Points of departure from occupational and animal studies were used to determine margins of exposure (MOEs). MOEs range from 1.5 E+3 to 3.3 E+6 with a median of 5 E+5, indicating that current environmental exposures to Cr(VI) in ambient air should be considered of low concern. In this comprehensive review, the divergent results from default linear and MOE assessments support the need for more relevant and robust epidemiologic data, additional mechanistic studies, and refined risk assessment strategies.
Collapse
Affiliation(s)
- Deborah M Proctor
- ToxStrategies, Inc, 27001 La Paz Rd, Suite 260, Mission Viejo, CA, 92691, USA.
| | | | - Mina Suh
- ToxStrategies, Inc, 27001 La Paz Rd, Suite 260, Mission Viejo, CA, 92691, USA
| | | | | | | |
Collapse
|
11
|
Xie P, Yuan FQ, Huang MS, Zhang W, Zhou HH, Li X, Liu ZQ. DCBLD2 Affects the Development of Colorectal Cancer via EMT and Angiogenesis and Modulates 5-FU Drug Resistance. Front Cell Dev Biol 2021; 9:669285. [PMID: 34095137 PMCID: PMC8170045 DOI: 10.3389/fcell.2021.669285] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 04/27/2021] [Indexed: 01/05/2023] Open
Abstract
Background: DCBLD2 is highly expressed in various cancers, including colorectal cancer. DCBLD2 overexpression promotes tumor occurrence, development, and metastasis. However, DCBLD2 sensitivity to chemotherapy drugs and its mechanism on tumor development are unknown. Methods: DCBLD2 expression differences in cancer and normal tissues were obtained from GEO and TCGA databases. DCBLD2 influence on prognosis was also compared, and the database analysis results were verified via the analysis of clinical samples. GDSC database was used to analyze the effect of DCBLD2 expression difference on 5-FU drug sensitivity on tumor cells. CCK-8, clone formation, scratch, Transwell invasion and migration assays were used to assess DCBLD2 effects on the proliferation, metastasis, and 5-FU drug sensitivity on HCT116 and Caco-2 colorectal cancer cells. Angiogenesis and Matrigel plug assays were used to study the effect of DCBLD2 on angiogenesis. Q-RCR and Western Blot were used to analyze DCBLD2 impact on the EMT signaling pathway, and TAP-MS assay with Co-IP verification was used to identify the downstream target proteins binding to DCBLD2. Results: Both database and clinical sample validation results showed that the expression of DCBLD2 in colorectal cancer tissues was significantly higher than that in normal tissues, leading to poor prognosis of patients. GDSC database analysis showed that DCBLD2 overexpression caused tumor cell resistance to 5-FU. The results of in vitro and in vivo experiments showed that the inhibition of DCBLD2 reduced the proliferation, migration and invasion of colorectal cancer cells, inhibited the angiogenesis of endothelial cells, and enhanced the drug sensitivity to 5-FU. The results of q-RCR and Western Blot experiments showed that the inhibition of DCBLD2 can suppress the EMT signal. The results of TAP-MS assay showed that the proteins bound to DCBLD2 were enriched to the Focal adhesion pathway. The results of Co-IP assay show that DCBLD2 can combine with ITGB1, the key factor of Focal adhesion pathway. Conclusion: DCBLD2 may affect the development of colorectal cancer by regulating cell proliferation and motility, and modulate 5-FU resistance. Down-regulation of DCBLD2 can inhibit EMT signal and angiogenesis. DCBLD2 can combine with ITGB1, the key signal factor of the Focal adhesion pathway.
Collapse
Affiliation(s)
- Pan Xie
- Hunan Key Laboratory of Pharmacogenetics, Department of Clinical Pharmacology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.,Institute of Clinical Pharmacology, Central South University, Changsha, China
| | - Fu-Qiang Yuan
- Hunan Key Laboratory of Pharmacogenetics, Department of Clinical Pharmacology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.,Institute of Clinical Pharmacology, Central South University, Changsha, China
| | - Ma-Sha Huang
- Hunan Key Laboratory of Pharmacogenetics, Department of Clinical Pharmacology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.,Institute of Clinical Pharmacology, Central South University, Changsha, China
| | - Wei Zhang
- Hunan Key Laboratory of Pharmacogenetics, Department of Clinical Pharmacology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.,Institute of Clinical Pharmacology, Central South University, Changsha, China
| | - Hong-Hao Zhou
- Hunan Key Laboratory of Pharmacogenetics, Department of Clinical Pharmacology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.,Institute of Clinical Pharmacology, Central South University, Changsha, China
| | - Xi Li
- Hunan Key Laboratory of Pharmacogenetics, Department of Clinical Pharmacology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.,Institute of Clinical Pharmacology, Central South University, Changsha, China
| | - Zhao-Qian Liu
- Hunan Key Laboratory of Pharmacogenetics, Department of Clinical Pharmacology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.,Institute of Clinical Pharmacology, Central South University, Changsha, China
| |
Collapse
|
12
|
Coppo R, Orso F, Virga F, Dalmasso A, Baruffaldi D, Nie L, Clapero F, Dettori D, Quirico L, Grassi E, Defilippi P, Provero P, Valdembri D, Serini G, Sadeghi MM, Mazzone M, Taverna D. ESDN inhibits melanoma progression by blocking E-selectin expression in endothelial cells via STAT3. Cancer Lett 2021; 510:13-23. [PMID: 33862151 DOI: 10.1016/j.canlet.2021.04.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 03/10/2021] [Accepted: 04/07/2021] [Indexed: 02/07/2023]
Abstract
An interactive crosstalk between tumor and stroma cells is essential for metastatic melanoma progression. We evidenced that ESDN/DCBLD2/CLCP1 plays a crucial role in endothelial cells during the spread of melanoma. Precisely, increased extravasation and metastasis formation were revealed in ESDN-null mice injected with melanoma cells, even if the primary tumor growth, vessel permeability, and angiogenesis were not enhanced. Interestingly, improved adhesion of melanoma cells to ESDN-depleted endothelial cells was observed, due to the presence of higher levels of E-selectin transcripts/proteins in ESDN-defective cells. In accordance with these results, anticorrelation was observed between ESDN and E-selectin in human endothelial cells. Most importantly, our data revealed that cimetidine, an E-selectin inhibitor, was able to block cell adhesion, extravasation, and metastasis formation in ESDN-null mice, underlying a major role of ESDN in E-selectin transcription upregulation, which according to our data, may presumably be linked to STAT3. Based on our results, we propose a protective role for ESDN during the spread of melanoma and reveal its therapeutic potential.
Collapse
Affiliation(s)
- Roberto Coppo
- Molecular Biotechnology Center (MBC), University of Torino, Torino, Italy; Dept. Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Francesca Orso
- Molecular Biotechnology Center (MBC), University of Torino, Torino, Italy; Dept. Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Federico Virga
- Molecular Biotechnology Center (MBC), University of Torino, Torino, Italy; Dept. Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy; VIB Center for Cancer Biology, Department of Oncology, University of Leuven, Leuven, Belgium
| | - Alberto Dalmasso
- Molecular Biotechnology Center (MBC), University of Torino, Torino, Italy; Dept. Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Desirée Baruffaldi
- Molecular Biotechnology Center (MBC), University of Torino, Torino, Italy; Dept. Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Lei Nie
- Section of Cardiovascular Medicine and Cardiovascular Research Center, Yale School of Medicine, New Haven, CT, USA; Veterans Affairs Connecticut Healthcare System, West Haven, CT, USA
| | - Fabiana Clapero
- Candiolo Cancer Institute, Fondazione del Piemonte per l'Oncologia (FPO) Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), 10060, Candiolo, Torino, Italy
| | - Daniela Dettori
- Molecular Biotechnology Center (MBC), University of Torino, Torino, Italy; Dept. Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Lorena Quirico
- Molecular Biotechnology Center (MBC), University of Torino, Torino, Italy; Dept. Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Elena Grassi
- Molecular Biotechnology Center (MBC), University of Torino, Torino, Italy; Dept. Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Paola Defilippi
- Molecular Biotechnology Center (MBC), University of Torino, Torino, Italy; Dept. Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Paolo Provero
- Molecular Biotechnology Center (MBC), University of Torino, Torino, Italy; Dept. Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy; Center for Translational Genomics and Bioinformatics, San Raffaele Scientific Institute, Milano, Italy
| | - Donatella Valdembri
- Candiolo Cancer Institute, Fondazione del Piemonte per l'Oncologia (FPO) Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), 10060, Candiolo, Torino, Italy; Department of Oncology, University of Torino School of Medicine, 10060, Candiolo, Torino, Italy
| | - Guido Serini
- Candiolo Cancer Institute, Fondazione del Piemonte per l'Oncologia (FPO) Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), 10060, Candiolo, Torino, Italy; Department of Oncology, University of Torino School of Medicine, 10060, Candiolo, Torino, Italy
| | - Mehran M Sadeghi
- Section of Cardiovascular Medicine and Cardiovascular Research Center, Yale School of Medicine, New Haven, CT, USA; Veterans Affairs Connecticut Healthcare System, West Haven, CT, USA
| | - Massimiliano Mazzone
- Molecular Biotechnology Center (MBC), University of Torino, Torino, Italy; Dept. Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy; VIB Center for Cancer Biology, Department of Oncology, University of Leuven, Leuven, Belgium
| | - Daniela Taverna
- Molecular Biotechnology Center (MBC), University of Torino, Torino, Italy; Dept. Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy.
| |
Collapse
|
13
|
Transmembrane protein DCBLD2 is correlated with poor prognosis and affects phenotype by regulating epithelial-mesenchymal transition in human glioblastoma cells. Neuroreport 2021; 32:507-517. [PMID: 33788813 DOI: 10.1097/wnr.0000000000001611] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVE We attempt to investigate the biological function of the discoidin, complement C1r/C1s,Uegf, and Bmp1 and Limulus factor C, Coch, and Lgl domain-containing 2 (DCBLD2) in glioblastoma, as well as its effect on the epithelial-mesenchymal transition (EMT) process. METHODS The public expression data of glioblastoma samples and normal brain samples from The Cancer Genome Atlas database, Genotype-Tissue Expression database and Chinese Glioma Genome Atlas database were used to analyze the expression of DCBLD2 and its relationship with the survival of patients with glioblastoma. Quantitative real-time PCR and western blot were used to evaluate mRNA and protein levels of DCBLD2. Cell viabilities were tested using Cell Counting Kit-8 and clone formation assays. Cell invasive and migratory abilities were measured by transwell assays. RESULTS DCBLD2 expression was upregulated in glioblastoma and has a significantly positive correlation with the WHO classification. In addition, high expression of DCBLD2 was closely correlated with poor prognosis in primary and recurrent patients with glioblastoma. What is more, we found that knockdown of DCBLD2 notably reduced the cell proliferative, invasive and migratory capacities by elevating the expression of E-cadherin and inhibiting the expression of vimentin, snail, slug and twist. However, overexpression of DCBLD2 presented the opposite results. CONCLUSION The current study reveals that high expression of DCBLD2 is closely related to poor prognosis in glioblastoma and can significantly enhance the tumor cell viability and metastasis by activating the EMT process, suggesting that DCBLD2 may be a possible biomarker for glioblastoma treatment.
Collapse
|
14
|
DCBLD2 Mediates Epithelial-Mesenchymal Transition-Induced Metastasis by Cisplatin in Lung Adenocarcinoma. Cancers (Basel) 2021; 13:cancers13061403. [PMID: 33808696 PMCID: PMC8003509 DOI: 10.3390/cancers13061403] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 03/10/2021] [Accepted: 03/13/2021] [Indexed: 12/29/2022] Open
Abstract
Growing evidence suggests that cisplatin and other chemotherapeutic agents promote tumor metastasis while inhibiting tumor growth, which is a critical issue for certain patients in clinical practices. However, the role of chemotherapeutics in promoting tumor metastasis and the molecular mechanism involved are unclear. Here, we investigated the roles of cisplatin in promoting tumor metastasis in lung adenocarcinoma (LUAD). We demonstrated that cisplatin promoted epithelial-mesenchymal transition (EMT), cell motility, and metastasis in vitro and in vivo. The bioinformatic analysis and molecular biology approaches also indicated that DCBLD2 (Discoidin, CUB and LCCL domain containing 2) is a key gene that mediates cisplatin-induced metastasis. DCBLD2 stabilizes β-catenin by phosphorylating GSK3β and transporting accumulated β-catenin to the nucleus to promote the expression of EMT-related transcriptional factors (TFs), ultimately resulting in tumor metastasis. We also identified that cisplatin enhanced DCBLD2 expression by phosphorylating ERK and hence the AP-1-driven transcription of DCBLD2. Furthermore, DCBLD2-specific siRNAs encapsulated by nanocarriers prominently inhibit cisplatin-induced metastasis in vivo. Therefore, DCBLD2 plays a key role in cisplatin-induced metastasis in LUAD and is a potential target for preventing chemotherapy-induced metastasis in vivo.
Collapse
|
15
|
Baietti MF, Zhao P, Crowther J, Sewduth RN, De Troyer L, Debiec-Rychter M, Sablina AA. Loss of 9p21 Regulatory Hub Promotes Kidney Cancer Progression by Upregulating HOXB13. Mol Cancer Res 2021; 19:979-990. [PMID: 33619226 DOI: 10.1158/1541-7786.mcr-20-0705] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 12/24/2020] [Accepted: 02/16/2021] [Indexed: 11/16/2022]
Abstract
Loss of chromosome 9p21 is observed in one-thirds of clear-cell renal cell carcinoma (ccRCC) and is associated with poorer patient survival. Unexpectedly, 9p21 LOH does not lead to decreased expression of the 9p21 tumor suppressor genes, CDKN2A and CDKN2B, suggesting alternative mechanisms of 9p-mediated tumorigenesis. Concordantly, CRISPR-mediated 9p21 deletion promotes growth of immortalized human embryonic kidney epithelial cells independently of the CDKN2A/B pathway inactivation. The 9p21 locus has a highly accessible chromatin structure, suggesting that 9p21 loss might contribute to kidney cancer progression by dysregulating genes distal to the 9p21 locus. We identified several 9p21 regulatory hubs by assessing which of the 9p21-interacting genes are dysregulated in 9p21-deleted kidney cells and ccRCCs. By focusing on the analysis of the homeobox gene 13 (HOXB13) locus, we found that 9p21 loss relieves the HOXB13 locus, decreasing HOXB13 methylation and promoting its expression. Upregulation of HOXB13 facilitates cell growth and is associated with poorer survival of patients with ccRCC. IMPLICATIONS: The results of our study propose a novel tumor suppressive mechanism on the basis of coordinated expression of physically associated genes, providing a better understanding of the role of chromosomal deletions in cancer.
Collapse
Affiliation(s)
- Maria Francesca Baietti
- VIB-KU Leuven Center for Cancer Biology, Leuven, Belgium. .,Department of Oncology, KU Leuven, Leuven, Belgium
| | - Peihua Zhao
- VIB-KU Leuven Center for Cancer Biology, Leuven, Belgium.,Department of Oncology, KU Leuven, Leuven, Belgium
| | - Jonathan Crowther
- VIB-KU Leuven Center for Cancer Biology, Leuven, Belgium.,Department of Oncology, KU Leuven, Leuven, Belgium
| | - Raj Nayan Sewduth
- VIB-KU Leuven Center for Cancer Biology, Leuven, Belgium.,Department of Oncology, KU Leuven, Leuven, Belgium
| | - Linde De Troyer
- VIB-KU Leuven Center for Cancer Biology, Leuven, Belgium.,Department of Oncology, KU Leuven, Leuven, Belgium
| | - Maria Debiec-Rychter
- Department of Human Genetics, KU Leuven, Leuven, Belgium.,Department of Pathology, University Hospitals KU Leuven, Leuven, Belgium
| | - Anna A Sablina
- VIB-KU Leuven Center for Cancer Biology, Leuven, Belgium. .,Department of Oncology, KU Leuven, Leuven, Belgium
| |
Collapse
|
16
|
Schmoker AM, Weinert JL, Markwood JM, Albretsen KS, Lunde ML, Weir ME, Ebert AM, Hinkle KL, Ballif BA. FYN and ABL Regulate the Interaction Networks of the DCBLD Receptor Family. Mol Cell Proteomics 2020; 19:1586-1601. [PMID: 32606017 PMCID: PMC8015000 DOI: 10.1074/mcp.ra120.002163] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Indexed: 12/23/2022] Open
Abstract
The Discoidin, CUB, and LCCL domain-containing protein (DCBLD) family consists of two type-I transmembrane scaffolding receptors, DCBLD1 and DCBLD2, which play important roles in development and cancer. The nonreceptor tyrosine kinases FYN and ABL are known to drive phosphorylation of tyrosine residues in YXXP motifs within the intracellular domains of DCBLD family members, which leads to the recruitment of the Src homology 2 (SH2) domain of the adaptors CT10 regulator of kinase (CRK) and CRK-like (CRKL). We previously characterized the FYN- and ABL-driven phosphorylation of DCBLD family YXXP motifs. However, we have identified additional FYN- and ABL-dependent phosphorylation sites on DCBLD1 and DCBLD2. This suggests that beyond CRK and CRKL, additional DCBLD interactors may be regulated by FYN and ABL activity. Here, we report a quantitative proteomics approach in which we map the FYN- and ABL-regulated interactomes of DCBLD family members. We found FYN and ABL regulated the binding of several signaling molecules to DCBLD1 and DCBLD2, including members of the 14-3-3 family of adaptors. Biochemical investigation of the DCBLD2/14-3-3 interaction revealed ABL-induced binding of 14-3-3 family members directly to DCBLD2.
Collapse
Affiliation(s)
- Anna M Schmoker
- Department of Biology, University of Vermont, Marsh Life Sciences, Burlington, Vermont, USA.
| | - Jaye L Weinert
- Department of Biology, University of Vermont, Marsh Life Sciences, Burlington, Vermont, USA
| | - Jacob M Markwood
- Department of Biology, Norwich University, Northfield, Vermont, USA
| | | | - Michelle L Lunde
- Department of Biology, Norwich University, Northfield, Vermont, USA
| | - Marion E Weir
- Department of Biology, University of Vermont, Marsh Life Sciences, Burlington, Vermont, USA
| | - Alicia M Ebert
- Department of Biology, University of Vermont, Marsh Life Sciences, Burlington, Vermont, USA
| | - Karen L Hinkle
- Department of Biology, Norwich University, Northfield, Vermont, USA
| | - Bryan A Ballif
- Department of Biology, University of Vermont, Marsh Life Sciences, Burlington, Vermont, USA.
| |
Collapse
|
17
|
Chen J, Cai Y, Xu R, Pan J, Zhou J, Mei J. Identification of four hub genes as promising biomarkers to evaluate the prognosis of ovarian cancer in silico. Cancer Cell Int 2020; 20:270. [PMID: 32595417 PMCID: PMC7315561 DOI: 10.1186/s12935-020-01361-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Accepted: 06/17/2020] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Ovarian cancer (OvCa) is one of the most fatal cancers among females in the world. With growing numbers of individuals diagnosed with OvCa ending in deaths, it is urgent to further explore the potential mechanisms of OvCa oncogenesis and development and related biomarkers. METHODS The gene expression profiles of GSE49997 were downloaded from the Gene Expression Omnibus (GEO) database. Weighted gene co-expression network analysis (WGCNA) was applied to explore the most potent gene modules associated with the overall survival (OS) and progression-free survival (PFS) events of OvCa patients, and the prognostic values of these genes were exhibited and validated based on data from training and validation sets. Next, protein-protein interaction (PPI) networks were built by GeneMANIA. Besides, enrichment analysis was conducted using DAVID website. RESULTS According to the WGCNA analysis, a total of eight modules were identified and four hub genes (MM > 0.90) in the blue module were reserved for next analysis. Kaplan-Meier analysis exhibited that these four hub genes were significantly associated with worse OS and PFS in the patient cohort from GSE49997. Moreover, we validated the short-term (4-years) and long-term prognostic values based on the GSE9891 data, respectively. Last, PPI networks analysis, Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis revealed several potential mechanisms of four hub genes and their co-operators participating in OvCa progression. CONCLUSION Four hub genes (COL6A3, CRISPLD2, FBN1 and SERPINF1) were identified to be associated with the prognosis in OvCa, which might be used as monitoring biomarkers to evaluate survival time of OvCa patients.
Collapse
Affiliation(s)
- Jingxuan Chen
- School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166 China
- Cytoskeleton Research Group & First Clinical Medicine College, Nanjing Medical University, No. 101 Longmian Road, Nanjing, 211166 China
| | - Yun Cai
- Department of Bioinformatics, Nanjing Medical University, Nanjing, 211166 China
| | - Rui Xu
- School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166 China
- Cytoskeleton Research Group & First Clinical Medicine College, Nanjing Medical University, No. 101 Longmian Road, Nanjing, 211166 China
| | - Jiadong Pan
- First Clinical Medicine College, Nanjing Medical University, Nanjing, 211166 China
| | - Jie Zhou
- Department of Gynecology and Obstetrics, Affiliated Wuxi Maternal and Child Health Hospital of Nanjing Medical University, No.48, Huaishu Road, Wuxi, 214023 China
| | - Jie Mei
- Cytoskeleton Research Group & First Clinical Medicine College, Nanjing Medical University, No. 101 Longmian Road, Nanjing, 211166 China
- First Clinical Medicine College, Nanjing Medical University, Nanjing, 211166 China
| |
Collapse
|
18
|
Feng L, Guo X, Li T, Yao C, Xia H, Jiang Z, Jia J, Fang Y, Shi L, Lu CA, Lou J. Novel DNA methylation biomarkers for hexavalent chromium exposure: an epigenome-wide analysis. Epigenomics 2020; 12:221-233. [DOI: 10.2217/epi-2019-0216] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Aim: We aimed to identify differential methylation of genes that could illuminate the biological mechanisms of chromium (VI) toxicity in this exposure-control study. Materials & methods: DNA methylation was measured in blood samples collected from electroplating workers and controls using a combination of Infinium Methylation450K Chip and targeted-bisulfite sequencing. QuantiGene assay was used to detect the mRNA expression of differentially methylated genes. Inductively coupled plasma–mass spectrometry was used to quantify metals in blood and urine samples. The cytosine–phosphate–guanine sites methylation and gene expression were confirmed in a human lymphoblastoid cell line. Results & conclusion: A total of 131 differentially methylated cytosine–phosphate–guanine sites were found between exposures and controls. DNA methylation of SEMA4B may serve as a potential biomarker for chromium (VI) exposure.
Collapse
Affiliation(s)
- Lingfang Feng
- Institute of Occupational Diseases, Zhejiang Academy of Medical Sciences, Hangzhou 310013, PR China
| | - Xinnian Guo
- Institute of Occupational Diseases, Zhejiang Academy of Medical Sciences, Hangzhou 310013, PR China
| | - Tao Li
- Institute of Occupational Diseases, Zhejiang Academy of Medical Sciences, Hangzhou 310013, PR China
| | - Chunji Yao
- Institute of Hygiene, Zhejiang Academy of Medical Sciences, Hangzhou 310013, PR China
| | - Hailing Xia
- Institute of Occupational Diseases, Zhejiang Academy of Medical Sciences, Hangzhou 310013, PR China
| | - Zhaoqiang Jiang
- Institute of Occupational Diseases, Zhejiang Academy of Medical Sciences, Hangzhou 310013, PR China
| | - Junlin Jia
- Center for Biostatistics, Bioinformatics & Big Data, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, PR China
| | - Yuan Fang
- Institute of Occupational Diseases, Zhejiang Academy of Medical Sciences, Hangzhou 310013, PR China
| | - Li Shi
- Institute of Occupational Diseases, Zhejiang Academy of Medical Sciences, Hangzhou 310013, PR China
| | - Chensheng Alex Lu
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Jianlin Lou
- Institute of Occupational Diseases, Zhejiang Academy of Medical Sciences, Hangzhou 310013, PR China
| |
Collapse
|
19
|
Ko PH, Lenka G, Chen YA, Chuang EY, Tsai MH, Sher YP, Lai LC. Semaphorin 5A suppresses the proliferation and migration of lung adenocarcinoma cells. Int J Oncol 2019; 56:165-177. [PMID: 31789397 PMCID: PMC6910195 DOI: 10.3892/ijo.2019.4932] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 11/13/2019] [Indexed: 12/26/2022] Open
Abstract
Semaphorin 5A (SEMA5A), a member of the semaphorin family, plays an important role in axonal guidance. Previously, the authors identified another possible role of SEMA5A as a prognostic biomarker for non-smoking women with lung adenocarcinoma in Taiwan, and this phenomenon has been validated in other ethnic groups. However, the functional significance of SEMA5A in lung adenocarcinoma remains unclear. Therefore, we assessed the function of SEMA5A in three lung adenocarcinoma cell lines in this study. Kaplan-Meier Plotter for lung cancer was conducted for survival analyses. Reverse transcription-quantitative PCR (RT-qPCR) and western blot analysis were performed to investigate the expression and post-translational regulation of SEMA5A in lung adenocar-cinoma cell lines. A pre-designed PyroMark CpG assay and 5-aza-2′-deoxycytidine treatment were used to measure the methylation levels of SEMA5A. The biological functions of lung adenocarcinoma cells overexpressing SEMA5A were investigated by microarrays, and validated both in vitro (proliferation, colony formation and migration assays) and in vivo (tumor xenografts) experiments. The results revealed that the hypermethylation of SEMA5A and the cleavage of the extracellular domain of SEMA5A were responsible for the downregulation of the SEMA5A levels in lung adenocarcinoma cells (A549 and H1299) as compared to the normal controls. Functional analysis of SEMA5A-regulated genes revealed that they were involved in cellular growth and proliferation. The overexpression of SEMA5A in A549 and H1299 cells significantly decreased the proliferation (P<0.01), colony formation (P<0.001) and migratory ability (P<0.01) of the cells. The suppressive effects of SEMA5A on the proliferative and migratory ability of the cells were also observed in both in vitro and in vivo experiments using brain metastatic Bm7 lung adenocarcinoma cells. On the whole, the findings of this study suggest a suppressive role for SEMA5A in lung adenocarcinoma involving the inhibition of the proliferation and migration of lung transformed cells.
Collapse
Affiliation(s)
- Pin-Hao Ko
- Graduate Institute of Physiology, College of Medicine, National Taiwan University, Taipei 10051, Taiwan, R.O.C
| | - Govinda Lenka
- Graduate Institute of Physiology, College of Medicine, National Taiwan University, Taipei 10051, Taiwan, R.O.C
| | - Yu-An Chen
- Bioinformatics and Biostatistics Core, Center of Genomic and Precision Medicine, National Taiwan University, Taipei 10055, Taiwan, R.O.C
| | - Eric Y Chuang
- Bioinformatics and Biostatistics Core, Center of Genomic and Precision Medicine, National Taiwan University, Taipei 10055, Taiwan, R.O.C
| | - Mong-Hsun Tsai
- Bioinformatics and Biostatistics Core, Center of Genomic and Precision Medicine, National Taiwan University, Taipei 10055, Taiwan, R.O.C
| | - Yuh-Pyng Sher
- Graduate Institute of Clinical Medical Science, China Medical University, Taichung 40402, Taiwan, R.O.C
| | - Liang-Chuan Lai
- Graduate Institute of Physiology, College of Medicine, National Taiwan University, Taipei 10051, Taiwan, R.O.C
| |
Collapse
|
20
|
The DCBLD receptor family: emerging signaling roles in development, homeostasis and disease. Biochem J 2019; 476:931-950. [PMID: 30902898 DOI: 10.1042/bcj20190022] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 02/20/2019] [Accepted: 03/04/2019] [Indexed: 02/08/2023]
Abstract
The discoidin, CUB, and LCCL domain-containing (DCBLD) receptor family are composed of the type-I transmembrane proteins DCBLD1 and DCBLD2 (also ESDN and CLCP1). These proteins are highly conserved across vertebrates and possess similar domain structure to that of neuropilins, which act as critical co-receptors in developmental processes. Although DCBLD1 remains largely uncharacterized, the functional and mechanistic roles of DCBLD2 are emerging. This review provides a comprehensive discussion of this presumed receptor family, ranging from structural and signaling aspects to their associations with cancer, physiology, and development.
Collapse
|
21
|
Movassagh H, Koussih L, Shan L, Gounni AS. The regulatory role of semaphorin 3E in allergic asthma. Int J Biochem Cell Biol 2018; 106:68-73. [PMID: 30447428 DOI: 10.1016/j.biocel.2018.11.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 11/06/2018] [Accepted: 11/12/2018] [Indexed: 10/27/2022]
Abstract
Semaphorins were originally discovered as essential mediators involved in regulation of axonal growth during development of the nervous system. Ubiquitously expressed on various organs, they control several cellular functions by regulating essential signaling pathways. Among them, semaphorin3E binds plexinD1 as the primary receptor and mediates regulatory effects on cell migration, proliferation, and angiogenesis considered major physiological and pathological features in health and disease. Recent in vitro and in vivo experimental evidence demonstrate a key regulator role of semaphorin3E on airway inflammation, hyperresponsivenss and remodeling in allergic asthma. Herein, we aim to provide a broad overview of the biology of semaphorin family and review the recently discovered regulatory role of semaphorin3E in modulating immune cells and structural cells function in the airways. These findings support the concept of semaphorin3E/plexinD1 axis as a therapeutic target in allergic asthma.
Collapse
Affiliation(s)
- Hesam Movassagh
- Department of Immunology, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Latifa Koussih
- Department of Immunology, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Lianyu Shan
- Department of Immunology, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Abdelilah S Gounni
- Department of Immunology, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada.
| |
Collapse
|
22
|
Bossé Y, Amos CI. A Decade of GWAS Results in Lung Cancer. Cancer Epidemiol Biomarkers Prev 2018; 27:363-379. [PMID: 28615365 PMCID: PMC6464125 DOI: 10.1158/1055-9965.epi-16-0794] [Citation(s) in RCA: 152] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Revised: 12/06/2016] [Accepted: 04/20/2017] [Indexed: 01/03/2023] Open
Abstract
Genome-wide association studies (GWAS) were successful to identify genetic factors robustly associated with lung cancer. This review aims to synthesize the literature in this field and accelerate the translation of GWAS discoveries into results that are closer to clinical applications. A chronologic presentation of published GWAS on lung cancer susceptibility, survival, and response to treatment is presented. The most important results are tabulated to provide a concise overview in one read. GWAS have reported 45 lung cancer susceptibility loci with varying strength of evidence and highlighted suspected causal genes at each locus. Some genetic risk loci have been refined to more homogeneous subgroups of lung cancer patients in terms of histologic subtypes, smoking status, gender, and ethnicity. Overall, these discoveries are an important step for future development of new therapeutic targets and biomarkers to personalize and improve the quality of care for patients. GWAS results are on the edge of offering new tools for targeted screening in high-risk individuals, but more research is needed if GWAS are to pay off the investment. Complementary genomic datasets and functional studies are needed to refine the underlying molecular mechanisms of lung cancer preliminarily revealed by GWAS and reach results that are medically actionable. Cancer Epidemiol Biomarkers Prev; 27(4); 363-79. ©2018 AACRSee all articles in this CEBP Focus section, "Genome-Wide Association Studies in Cancer."
Collapse
Affiliation(s)
- Yohan Bossé
- Institut Universitaire de Cardiologie et de Pneumologie de Québec, Quebec, Canada.
- Department of Molecular Medicine, Laval University, Quebec, Canada
| | - Christopher I Amos
- Department of Biomedical Data Science, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire
| |
Collapse
|
23
|
Abstract
Semaphorins are extracellular signaling proteins that are essential for the development and maintenance of many organs and tissues. The more than 20-member semaphorin protein family includes secreted, transmembrane and cell surface-attached proteins with diverse structures, each characterized by a single cysteine-rich extracellular sema domain, the defining feature of the family. Early studies revealed that semaphorins function as axon guidance molecules, but it is now understood that semaphorins are key regulators of morphology and motility in many different cell types including those that make up the nervous, cardiovascular, immune, endocrine, hepatic, renal, reproductive, respiratory and musculoskeletal systems, as well as in cancer cells. Semaphorin signaling occurs predominantly through Plexin receptors and results in changes to the cytoskeletal and adhesive machinery that regulate cellular morphology. While much remains to be learned about the mechanisms underlying the effects of semaphorins, exciting work has begun to reveal how semaphorin signaling is fine-tuned through different receptor complexes and other mechanisms to achieve specific outcomes in various cellular contexts and physiological systems. These and future studies will lead to a more complete understanding of semaphorin-mediated development and to a greater understanding of how these proteins function in human disease.
Collapse
Affiliation(s)
- Laura Taylor Alto
- Departments of Neuroscience and Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Jonathan R Terman
- Departments of Neuroscience and Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.
| |
Collapse
|
24
|
Ge X, Shi QM, Ding Z, Ju Q, Wang H, Wang Q, Li MX, Chen G, Wang HX, Xu LC. Association Between CRISPLD2 Polymorphisms and the Risk of Nonsyndromic Clefts of the Lip and/or Palate: A Meta-analysis. Cleft Palate Craniofac J 2018; 55:328-334. [PMID: 29437515 DOI: 10.1177/1055665617738995] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE Nonsyndromic clefts of the lip and/or palate (NSCL/P) are one of the most common polygenic diseases. Recently, many studies focused on the association between CRISPLD2 polymorphisms and NSCL/P risk. However, some studies have shown opposite results. In this study, meta-analysis was used to confirm whether CRISPLD2 polymorphism was associated with NSCL/P, and the possible mechanism between CRISPLD2 and NSCL/P was explored. METHODS Relevant studies were conducted on PubMed, Ovid, EBSCO, CINAHL, FMRS, Web of Science, CNKI, and Wanfang databases from their inception up to June 31, 2016. Review Manager 5.0.24 was used to analyze whether CRISPLD2 polymorphism was involved in NSCL/P by pooling odds ratios (ORs) and 95% confidence intervals (CIs). Potential publication bias was evaluated by visual inspection of the funnel plot. RESULTS CRISPLD2 rs4783099 was associated with cleft lip and/or palate (CL/P) statistically (OR = 3.18, P < .01). Compared to genotype TT, genotypes CC and CT were correlated significantly (OR = 2.04, P = .04) with CL/P. No evidence showed an association between genetic variation at the CRISPLD2 locus and cleft palate only (CP). CONCLUSION The polymorphism of CRISPLD2 rs4783099 is correlated with an increased risk of CL/P.
Collapse
Affiliation(s)
- Xing Ge
- 1 School of Public Health, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Qiao-Mei Shi
- 1 School of Public Health, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Zhen Ding
- 1 School of Public Health, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Qiang Ju
- 1 School of Public Health, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Hui Wang
- 1 School of Public Health, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Qi Wang
- 1 School of Public Health, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Meng-Xue Li
- 1 School of Public Health, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Gang Chen
- 1 School of Public Health, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Heng-Xue Wang
- 1 School of Public Health, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Li-Chun Xu
- 1 School of Public Health, Xuzhou Medical University, Xuzhou, Jiangsu, China
| |
Collapse
|
25
|
A fast algorithm for Bayesian multi-locus model in genome-wide association studies. Mol Genet Genomics 2017; 292:923-934. [DOI: 10.1007/s00438-017-1322-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Accepted: 04/18/2017] [Indexed: 12/27/2022]
|
26
|
Sun T, Yang L, Kaur H, Pestel J, Looso M, Nolte H, Krasel C, Heil D, Krishnan RK, Santoni MJ, Borg JP, Bünemann M, Offermanns S, Swiercz JM, Worzfeld T. A reverse signaling pathway downstream of Sema4A controls cell migration via Scrib. J Cell Biol 2016; 216:199-215. [PMID: 28007914 PMCID: PMC5223600 DOI: 10.1083/jcb.201602002] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 09/30/2016] [Accepted: 11/13/2016] [Indexed: 11/22/2022] Open
Abstract
Semaphorins comprise a large family of ligands that regulate key cellular functions through their receptors, plexins. In this study, we show that the transmembrane semaphorin 4A (Sema4A) can also function as a receptor, rather than a ligand, and transduce signals triggered by the binding of Plexin-B1 through reverse signaling. Functionally, reverse Sema4A signaling regulates the migration of various cancer cells as well as dendritic cells. By combining mass spectrometry analysis with small interfering RNA screening, we identify the polarity protein Scrib as a downstream effector of Sema4A. We further show that binding of Plexin-B1 to Sema4A promotes the interaction of Sema4A with Scrib, thereby removing Scrib from its complex with the Rac/Cdc42 exchange factor βPIX and decreasing the activity of the small guanosine triphosphatase Rac1 and Cdc42. Our data unravel a role for Plexin-B1 as a ligand and Sema4A as a receptor and characterize a reverse signaling pathway downstream of Sema4A, which controls cell migration.
Collapse
Affiliation(s)
- Tianliang Sun
- Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Lida Yang
- Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Harmandeep Kaur
- Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Jenny Pestel
- Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Mario Looso
- Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Hendrik Nolte
- Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Cornelius Krasel
- Institute of Pharmacology and Clinical Pharmacy, Biochemical-Pharmacological Center, University of Marburg, 35043 Marburg, Germany
| | - Daniel Heil
- Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Ramesh K Krishnan
- Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Marie-Josée Santoni
- Cell Polarity, Cell Signaling and Cancer, Equipe labellisée Ligue Contre le Cancer, Institut National de la Santé et de la Recherche Médicale, U1068, 13009 Marseille, France.,Institut Paoli-Calmettes, 13009 Marseille, France.,Aix-Marseille Université, 13284 Marseille, France.,Centre National de la Recherche Scientifique, UMR7258, 13273 Marseille, France
| | - Jean-Paul Borg
- Cell Polarity, Cell Signaling and Cancer, Equipe labellisée Ligue Contre le Cancer, Institut National de la Santé et de la Recherche Médicale, U1068, 13009 Marseille, France.,Institut Paoli-Calmettes, 13009 Marseille, France.,Aix-Marseille Université, 13284 Marseille, France.,Centre National de la Recherche Scientifique, UMR7258, 13273 Marseille, France
| | - Moritz Bünemann
- Institute of Pharmacology and Clinical Pharmacy, Biochemical-Pharmacological Center, University of Marburg, 35043 Marburg, Germany
| | - Stefan Offermanns
- Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany.,Medical Faculty, University of Frankfurt, 60590 Frankfurt am Main, Germany
| | - Jakub M Swiercz
- Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Thomas Worzfeld
- Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany .,Institute of Pharmacology, Biochemical-Pharmacological Center, University of Marburg, 35043 Marburg, Germany
| |
Collapse
|
27
|
Neufeld G, Mumblat Y, Smolkin T, Toledano S, Nir-Zvi I, Ziv K, Kessler O. The semaphorins and their receptors as modulators of tumor progression. Drug Resist Updat 2016; 29:1-12. [DOI: 10.1016/j.drup.2016.08.001] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2016] [Revised: 07/31/2016] [Accepted: 08/23/2016] [Indexed: 12/16/2022]
|
28
|
Gurrapu S, Tamagnone L. Transmembrane semaphorins: Multimodal signaling cues in development and cancer. Cell Adh Migr 2016; 10:675-691. [PMID: 27295627 DOI: 10.1080/19336918.2016.1197479] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Semaphorins constitute a large family of membrane-bound and secreted proteins that provide guidance cues for axon pathfinding and cell migration. Although initially discovered as repelling cues for axons in nervous system, they have been found to regulate cell adhesion and motility, angiogenesis, immune function and tumor progression. Notably, semaphorins are bifunctional cues and for instance can mediate both repulsive and attractive functions in different contexts. While many studies focused so far on the function of secreted family members, class 1 semaphorins in invertebrates and class 4, 5 and 6 in vertebrate species comprise around 14 transmembrane semaphorin molecules with emerging functional relevance. These can signal in juxtacrine, paracrine and autocrine fashion, hence mediating long and short range repulsive and attractive guidance cues which have a profound impact on cellular morphology and functions. Importantly, transmembrane semaphorins are capable of bidirectional signaling, acting both in "forward" mode via plexins (sometimes in association with receptor tyrosine kinases), and in "reverse" manner through their cytoplasmic domains. In this review, we will survey known molecular mechanisms underlying the functions of transmembrane semaphorins in development and cancer.
Collapse
Affiliation(s)
- Sreeharsha Gurrapu
- a Department of Oncology , University of Torino c/o IRCCS , Candiolo ( TO ), Italy.,b Candiolo Cancer Institute, IRCCS-FPO , Candiolo ( TO ), Italy
| | - Luca Tamagnone
- a Department of Oncology , University of Torino c/o IRCCS , Candiolo ( TO ), Italy.,b Candiolo Cancer Institute, IRCCS-FPO , Candiolo ( TO ), Italy
| |
Collapse
|
29
|
Jian H, Zhao Y, Liu B, Lu S. SEMA4B inhibits growth of non-small cell lung cancer in vitro and in vivo. Cell Signal 2015; 27:1208-13. [PMID: 25746385 DOI: 10.1016/j.cellsig.2015.02.027] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Revised: 02/17/2015] [Accepted: 02/26/2015] [Indexed: 01/03/2023]
Abstract
We have recently shown that Semaphorin 4B (SEMA4B) inhibits the invasion of non-small cell lung cancer (NSCLC) through PI3K-dependent suppression of MMP9 activation. In the current study, we evaluated whether SEMA4B may also affect the growth of NSCLC. We thus used two human NSCLC lines, A549 and Calu-3, to examine our hypothesis. We found that overexpression of SEMA4B significantly decreased NSCLC cell growth, while SEMA4B inhibition significantly increased NSCLC cell growth, both in vitro and in vivo in an implanted NSCLC model. Adaptation of SEMA4B in NSCLC cells did not alter cell apoptosis, but changed the cell proliferation. Further analyses show that SEMA4B may induce FoxO1 nuclear retention through suppressing PI3K/Akt signaling pathway, which subsequently inhibited cell growth through the direct nuclear target of FoxO1, p21. Our study thus demonstrate a role of SEMA4B in suppressing NSCLC growth, besides its role in inhibiting cell metastasis, and highlights SEMA4B as a promising therapeutic target for NSCLC.
Collapse
Affiliation(s)
- Hong Jian
- Shanghai Lung Cancer Center, Shanghai Chest Hospital Affiliated to Shanghai Jiaotong University, Shanghai, China
| | - Yi Zhao
- Shanghai Lung Cancer Center, Shanghai Chest Hospital Affiliated to Shanghai Jiaotong University, Shanghai, China
| | - Bin Liu
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Shun Lu
- Shanghai Lung Cancer Center, Shanghai Chest Hospital Affiliated to Shanghai Jiaotong University, Shanghai, China.
| |
Collapse
|
30
|
SEMA4b inhibits MMP9 to prevent metastasis of non-small cell lung cancer. Tumour Biol 2014; 35:11051-6. [PMID: 25095981 DOI: 10.1007/s13277-014-2409-8] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Accepted: 07/27/2014] [Indexed: 10/24/2022] Open
Abstract
Semaphorin 4B (SEMA4b) has been shown to play a substantial role in the invasion of non-small cell lung cancer (NSCLC). However, the regulation mechanism of SEMA4b is largely elusive. Here, we reported significant decrease in SEMA4b level and significant increase in MMP9 level in NSCLC compared with the adjacent normal tissue from the same patient. We thus used two human NSCLC lines, A549 and Calu-3, to examine whether SEMA4b may affect the expression of MMP9 in NSCLC. We found that overexpression of SEMA4b significantly decreased MMP9 level, while SEMA4b inhibition significantly increased MMP9 level. The adapted MMP9 levels affected the potential of NSCLC invasiveness correspondingly. To define the signal transduction cascades downstream of SEMA4b for regulation of MMP9 expression, we inhibited PI3K, ERK/MAPK, or JNK signaling pathway in SEMA4b knockout NSCLC and found that only inhibition of PI3K signaling pathway significantly decreased MMP9 activation. Our data thus suggest that SEMA4b may activate PI3K signaling pathway to promote MMP9 expression, which subsequently increases metastasis of NSCLC. Our study thus highlights SEMA4b as a novel therapeutic target for NSCLC.
Collapse
|
31
|
Hypoxia and hypoxia-inducible factor 1 repress SEMA4B expression to promote non-small cell lung cancer invasion. Tumour Biol 2014; 35:4949-55. [DOI: 10.1007/s13277-014-1651-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2013] [Accepted: 01/12/2014] [Indexed: 12/23/2022] Open
|
32
|
Martinelli E, Troiani T, D'Aiuto E, Morgillo F, Vitagliano D, Capasso A, Costantino S, Ciuffreda LP, Merolla F, Vecchione L, De Vriendt V, Tejpar S, Nappi A, Sforza V, Martini G, Berrino L, De Palma R, Ciardiello F. Antitumor activity of pimasertib, a selective MEK 1/2 inhibitor, in combination with PI3K/mTOR inhibitors or with multi-targeted kinase inhibitors in pimasertib-resistant human lung and colorectal cancer cells. Int J Cancer 2013; 133:2089-101. [PMID: 23629727 DOI: 10.1002/ijc.28236] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2012] [Accepted: 04/05/2013] [Indexed: 01/09/2023]
Abstract
The RAS/RAF/MEK/MAPK and the PTEN/PI3K/AKT/mTOR pathways are key regulators of proliferation and survival in human cancer cells. Selective inhibitors of different transducer molecules in these pathways have been developed as molecular targeted anti-cancer therapies. The in vitro and in vivo anti-tumor activity of pimasertib, a selective MEK 1/2 inhibitor, alone or in combination with a PI3K inhibitor (PI3Ki), a mTOR inhibitor (everolimus), or with multi-targeted kinase inhibitors (sorafenib and regorafenib), that block also BRAF and CRAF, were tested in a panel of eight human lung and colon cancer cell lines. Following pimasertib treatment, cancer cell lines were classified as pimasertib-sensitive (IC50 for cell growth inhibition of 0.001 µM) or pimasertib-resistant. Evaluation of basal gene expression profiles by microarrays identified several genes that were up-regulated in pimasertib-resistant cancer cells and that were involved in both RAS/RAF/MEK/MAPK and PTEN/PI3K/AKT/mTOR pathways. Therefore, a series of combination experiments with pimasertib and either PI3Ki, everolimus, sorafenib or regorafenib were conducted, demonstrating a synergistic effect in cell growth inhibition and induction of apoptosis with sustained blockade in MAPK- and AKT-dependent signaling pathways in pimasertib-resistant human colon carcinoma (HCT15) and lung adenocarcinoma (H1975) cells. Finally, in nude mice bearing established HCT15 and H1975 subcutaneous tumor xenografts, the combined treatment with pimasertib and BEZ235 (a dual PI3K/mTOR inhibitor) or with sorafenib caused significant tumor growth delays and increase in mice survival as compared to single agent treatment. These results suggest that dual blockade of MAPK and PI3K pathways could overcome intrinsic resistance to MEK inhibition.
Collapse
Affiliation(s)
- Erika Martinelli
- Oncologia Medica, Dipartimento Medico- Chirurgico di Internistica Clinica e Sperimentale, F. Magrassi e A. Lanzara, Seconda Universitá degli Studi di Napoli, Via S. Pansini 5, 80131, Napoli, Italia
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
[RNA interference library research progress and its application in cancer research]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2013; 16:102-6. [PMID: 23425903 PMCID: PMC6000388 DOI: 10.3779/j.issn.1009-3419.2013.02.08] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
RNA interference is a homologous mRNA special degradation phenomenon which is caused by the double-stranded RNA. RNAi library is a pooled library that is artificially constructed using RNAi technology. As RNAi library has made a major breakthrough in the field of genetic research, it has been widely used in the field of medical research, especially in the field of cancer research. This review discussed the research progress of RNAi library and its applications in cancer research.
Collapse
|
34
|
Lan Q, Hsiung CA, Matsuo K, Hong YC, Seow A, Wang Z, Hosgood HD, Chen K, Wang JC, Chatterjee N, Hu W, Wong MP, Zheng W, Caporaso N, Park JY, Chen CJ, Kim YH, Kim YT, Landi MT, Shen H, Lawrence C, Burdett L, Yeager M, Yuenger J, Jacobs KB, Chang IS, Mitsudomi T, Kim HN, Chang GC, Bassig BA, Tucker M, Wei F, Yin Z, Wu C, An SJ, Qian B, Lee VHF, Lu D, Liu J, Jeon HS, Hsiao CF, Sung JS, Kim JH, Gao YT, Tsai YH, Jung YJ, Guo H, Hu Z, Hutchinson A, Wang WC, Klein R, Chung CC, Oh IJ, Chen KY, Berndt SI, He X, Wu W, Chang J, Zhang XC, Huang MS, Zheng H, Wang J, Zhao X, Li Y, Choi JE, Su WC, Park KH, Sung SW, Shu XO, Chen YM, Liu L, Kang CH, Hu L, Chen CH, Pao W, Kim YC, Yang TY, Xu J, Guan P, Tan W, Su J, Wang CL, Li H, Sihoe ADL, Zhao Z, Chen Y, Choi YY, Hung JY, Kim JS, Yoon HI, Cai Q, Lin CC, Park IK, Xu P, Dong J, Kim C, He Q, Perng RP, Kohno T, Kweon SS, et alLan Q, Hsiung CA, Matsuo K, Hong YC, Seow A, Wang Z, Hosgood HD, Chen K, Wang JC, Chatterjee N, Hu W, Wong MP, Zheng W, Caporaso N, Park JY, Chen CJ, Kim YH, Kim YT, Landi MT, Shen H, Lawrence C, Burdett L, Yeager M, Yuenger J, Jacobs KB, Chang IS, Mitsudomi T, Kim HN, Chang GC, Bassig BA, Tucker M, Wei F, Yin Z, Wu C, An SJ, Qian B, Lee VHF, Lu D, Liu J, Jeon HS, Hsiao CF, Sung JS, Kim JH, Gao YT, Tsai YH, Jung YJ, Guo H, Hu Z, Hutchinson A, Wang WC, Klein R, Chung CC, Oh IJ, Chen KY, Berndt SI, He X, Wu W, Chang J, Zhang XC, Huang MS, Zheng H, Wang J, Zhao X, Li Y, Choi JE, Su WC, Park KH, Sung SW, Shu XO, Chen YM, Liu L, Kang CH, Hu L, Chen CH, Pao W, Kim YC, Yang TY, Xu J, Guan P, Tan W, Su J, Wang CL, Li H, Sihoe ADL, Zhao Z, Chen Y, Choi YY, Hung JY, Kim JS, Yoon HI, Cai Q, Lin CC, Park IK, Xu P, Dong J, Kim C, He Q, Perng RP, Kohno T, Kweon SS, Chen CY, Vermeulen R, Wu J, Lim WY, Chen KC, Chow WH, Ji BT, Chan JKC, Chu M, Li YJ, Yokota J, Li J, Chen H, Xiang YB, Yu CJ, Kunitoh H, Wu G, Jin L, Lo YL, Shiraishi K, Chen YH, Lin HC, Wu T, Wu YL, Yang PC, Zhou B, Shin MH, Fraumeni JF, Lin D, Chanock SJ, Rothman N. Genome-wide association analysis identifies new lung cancer susceptibility loci in never-smoking women in Asia. Nat Genet 2012; 44:1330-5. [PMID: 23143601 DOI: 10.1038/ng.2456] [Show More Authors] [Citation(s) in RCA: 254] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2012] [Accepted: 10/05/2012] [Indexed: 02/06/2023]
Abstract
To identify common genetic variants that contribute to lung cancer susceptibility, we conducted a multistage genome-wide association study of lung cancer in Asian women who never smoked. We scanned 5,510 never-smoking female lung cancer cases and 4,544 controls drawn from 14 studies from mainland China, South Korea, Japan, Singapore, Taiwan and Hong Kong. We genotyped the most promising variants (associated at P < 5 × 10(-6)) in an additional 1,099 cases and 2,913 controls. We identified three new susceptibility loci at 10q25.2 (rs7086803, P = 3.54 × 10(-18)), 6q22.2 (rs9387478, P = 4.14 × 10(-10)) and 6p21.32 (rs2395185, P = 9.51 × 10(-9)). We also confirmed associations reported for loci at 5p15.33 and 3q28 and a recently reported finding at 17q24.3. We observed no evidence of association for lung cancer at 15q25 in never-smoking women in Asia, providing strong evidence that this locus is not associated with lung cancer independent of smoking.
Collapse
Affiliation(s)
- Qing Lan
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Osella-Abate S, Novelli M, Quaglino P, Orso F, Ubezio B, Tomasini C, Berardengo E, Bernengo MG, Taverna D. Expression of AP-2α, AP-2γ and ESDN in primary melanomas: correlation with histopathological features and potential prognostic value. J Dermatol Sci 2012; 68:202-4. [PMID: 23036739 DOI: 10.1016/j.jdermsci.2012.09.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2012] [Revised: 09/04/2012] [Accepted: 09/05/2012] [Indexed: 10/27/2022]
|
36
|
Pandiri AR, Sills RC, Ziglioli V, Ton TVT, Hong HHL, Lahousse SA, Gerrish KE, Auerbach SS, Shockley KR, Bushel PR, Peddada SD, Hoenerhoff MJ. Differential transcriptomic analysis of spontaneous lung tumors in B6C3F1 mice: comparison to human non-small cell lung cancer. Toxicol Pathol 2012; 40:1141-59. [PMID: 22688403 DOI: 10.1177/0192623312447543] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Lung cancer is the leading cause of cancer-related death in people and is mainly due to environmental factors such as smoking and radon. The National Toxicology Program (NTP) tests various chemicals and mixtures for their carcinogenic hazard potential. In the NTP chronic bioassay using B6C3F1 mice, the incidence of lung tumors in treated and control animals is second only to the liver tumors. In order to study the molecular mechanisms of chemically induced lung tumors, an understanding of the genetic changes that occur in spontaneous lung (SL) tumors from untreated control animals is needed. The authors have evaluated the differential transcriptomic changes within SL tumors compared to normal lungs from untreated age-matched animals. Within SL tumors, several canonical pathways associated with cancer (eukaryotic initiation factor 2 signaling, RhoA signaling, PTEN signaling, and mammalian target of rapamycin signaling), metabolism (Inositol phosphate metabolism, mitochondrial dysfunction, and purine and pyramidine metabolism), and immune responses (FcγR-mediated phagocytosis, clathrin-mediated endocytosis, interleukin 8 signaling, and CXCR4 signaling) were altered. Meta-analysis of murine SL tumors and human non-small cell lung cancer transcriptomic data sets revealed a high concordance. These data provide important information on the differential transcriptomic changes in murine SL tumors that will be critical to our understanding of chemically induced lung tumors and will aid in hazard analysis in the NTP 2-year carcinogenicity bioassays.
Collapse
Affiliation(s)
- Arun R Pandiri
- Cellular and Molecular Pathology Branch, National Toxicology Program-NTP, National Institute of Environmental Health Sciences-NIEHS, Research Triangle Park, North Carolina, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Pasaje CFA, Bae JS, Park BL, Cheong HS, Kim JH, Jang AS, Uh ST, Park CS, Shin HD. DCBLD2 gene variations correlate with nasal polyposis in Korean asthma patients. Lung 2012; 190:199-207. [PMID: 22261696 DOI: 10.1007/s00408-011-9354-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2011] [Accepted: 11/28/2011] [Indexed: 11/24/2022]
Abstract
BACKGROUND Nasal polyps are abnormal lesions that cause airway obstruction and can occur along with other respiratory diseases. On account of its association with aspirin exacerbated respiratory disease (AERD), the human discoidin, CUB and LCCL domain containing 2 (DCBLD2) is hypothesized to be a candidate gene for the development of nasal polyps in asthma patients. METHODS A total of 12 single-nucleotide polymorphisms (SNPs) were genotyped in 467 Korean asthma patients who were stratified further into 108 AERD and 353 aspirin-tolerant asthma (ATA) subgroups. Five major haplotypes were inferred from pairwise comparison of the polymorphisms. The patients were matched to control for confounds, and differences in the frequency distribution of DCBLD2 SNPs and haplotypes were analyzed using logistic models via various modes of genetic inheritance. RESULTS Results reveal significant association of rs828618 and DCBLD2_ht1 with nasal polyposis in the overall asthma patients group (P = 0.006, P(corr) = 0.05). Interestingly, the strength of association were maintained in the ATA subgroup (P = 0.007, P(corr) = 0.06), and moderate correlation was detected in the AERD subgroup (P = 0.04-0.05, P(corr) > 0.05). CONCLUSIONS Although further replication and validation are needed, these findings suggest that DCBLD2 could be a potential marker and drug target for treatment of nasal polyposis in Korean asthma patients.
Collapse
|
38
|
Resch E, Hiss JA, Schreiner A, Schneider G, Starzinski-Powitz A. Long signal peptides of RGMa and DCBLD2 are dissectible into subdomains according to the NtraC model. ACTA ACUST UNITED AC 2011; 7:942-51. [DOI: 10.1039/c0mb00254b] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
39
|
Shi J, Jiao X, Song T, Zhang B, Qin C, Cao F. CRISPLD2 polymorphisms are associated with non-syndromic cleft lip with or without cleft palate in a northern Chinese population. Eur J Oral Sci 2010; 118:430-3. [PMID: 20662919 DOI: 10.1111/j.1600-0722.2010.00743.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Non-syndromic cleft lip with or without cleft palate (NSCLP) is the most common craniofacial birth defect. This complex genetic disorder results from interactions between genes and environmental factors. Numerous genes have been reported in studies demonstrating association between the cleft lip and palate phenotypes and the alleles at single-nucleotide polymorphisms (SNPs) within specific genes. Recently, the cysteine-rich secretory protein LCCL domain containing 2 (CRISPLD2) has been revealed to be a novel candidate gene for NSCLP. The SNPs rs1546124, rs4783099 and rs16974880 in CRISPLD2 were highly significant in Caucasian and Hispanic multiplex families but showed no association in Colombian and Irish populations. In the current study, we examined these three SNPs in a northern Chinese population and found an association between these polymorphisms and NSCLP in both single-marker and haplotype analyses. Our data further strengthen the conclusion that altered CRISPLD2 is associated with NSCLP susceptibility.
Collapse
Affiliation(s)
- Jinna Shi
- Department of Periodontology, School of Stomatology, the First Affiliated Hospital, Harbin Medical University, Harbin, China
| | | | | | | | | | | |
Collapse
|
40
|
Lu TP, Tsai MH, Lee JM, Hsu CP, Chen PC, Lin CW, Shih JY, Yang PC, Hsiao CK, Lai LC, Chuang EY. Identification of a novel biomarker, SEMA5A, for non-small cell lung carcinoma in nonsmoking women. Cancer Epidemiol Biomarkers Prev 2010; 19:2590-7. [PMID: 20802022 DOI: 10.1158/1055-9965.epi-10-0332] [Citation(s) in RCA: 221] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Although cigarette smoking is the major risk factor for lung cancer, only 7% of female lung cancer patients in Taiwan have a history of smoking. The genetic mechanisms of carcinogenesis in nonsmokers are unclear, but semaphorins have been suggested to play a role as lung tumor suppressors. This report is a comprehensive analysis of the molecular signature of nonsmoking female lung cancer patients in Taiwan, with a particular focus on the semaphorin gene family. METHODS Sixty pairs of tumor and adjacent normal lung tissue specimens were analyzed by using Affymetrix U133plus2.0 expression arrays. Differentially expressed genes in tumor tissues were identified by a paired t test and validated by reverse transcriptase-PCR and immunohistochemistry. Functional analysis was conducted by using Ingenuity Pathway Analysis as well as gene set enrichment analysis and sigPathway algorithms. Kaplan-Meier survival analyses were used to evaluate the association of SEMA5A expression and clinical outcome. RESULTS We identified 687 differentially expressed genes in non-small cell lung carcinoma (NSCLC). Many of these genes, most notably the semaphorin family, were participants in the axon guidance signaling pathway. The downregulation of SEMA5A in tumor tissue, both at the transcriptional and translational levels, was associated with poor survival among nonsmoking women with NSCLC. CONCLUSIONS In summary, several semaphorin gene family members were identified as potential therapeutic targets, and SEMA5A may be useful as a prognostic biomarker for NSCLC, which may also be gender specific in Taiwanese patients. IMPACT A novel biomarker for NSCLC is identified.
Collapse
Affiliation(s)
- Tzu-Pin Lu
- Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taipei, Taiwan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Letra A, Menezes R, Cooper ME, Fonseca RF, Tropp S, Govil M, Granjeiro JM, Imoehl SR, Mansilla MA, Murray JC, Castilla EE, Orioli IM, Czeizel AE, Ma L, Chiquet BT, Hecht JT, Vieira AR, Marazita ML. CRISPLD2 variants including a C471T silent mutation may contribute to nonsyndromic cleft lip with or without cleft palate. Cleft Palate Craniofac J 2010; 48:363-70. [PMID: 20815724 DOI: 10.1597/09-227] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
OBJECTIVE To assess the association between nonsyndromic (NS) cleft lip with or without cleft palate (CL(P)) and single-nucleotide polymorphisms (SNPs) within the CRISPLD2 gene (cysteine-rich secretory protein LCCL domain containing 2). DESIGN Four SNPs within the CRISPLD2 gene domain (rs1546124, rs8061351, rs2326398, rs4783099) were genotyped to test for association via family-based association methods. PARTICIPANTS A total of 5826 individuals from 1331 families in which one or more family member is affected with CL(P). RESULTS Evidence of association was seen for SNP rs1546124 in U.S. (p = .02) and Brazilian (p = .04) Caucasian cohorts. We also found association of SNP rs1546124 with cleft palate alone (CP) in South Americans (Guatemala and ECLAMC) and combined Hispanics (Guatemala, ECLAMC, and Texas Hispanics; p = .03 for both comparisons) and with both cleft lip with cleft palate (CLP; p = .04) and CL(P) (p = .02) in North Americans. Strong evidence of association was found for SNP rs2326398 with CP in Asian populations (p = .003) and with CL(P) in Hispanics (p = .03) and also with bilateral CL(P) in Brazilians (p = .004). In Brazilians, SNP rs8061351 showed association with cleft subgroups incomplete CL(P) (p = .004) and unilateral incomplete CL(P) (p = .003). Prediction of SNP functionality revealed that the C allele in the C471T silent mutation (overrepresented in cases with CL(P) presents two putative exonic splicing enhancer motifs and creates a binding site AP-2 alpha, a transcription factor involved in craniofacial development. CONCLUSIONS Our results support the hypothesis that variants in the CRISPLD2 gene may be involved in the etiology of NS CL(P).
Collapse
|
42
|
Orso F, Corà D, Ubezio B, Provero P, Caselle M, Taverna D. Identification of functional TFAP2A and SP1 binding sites in new TFAP2A-modulated genes. BMC Genomics 2010; 11:355. [PMID: 20525283 PMCID: PMC2890567 DOI: 10.1186/1471-2164-11-355] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2010] [Accepted: 06/03/2010] [Indexed: 12/20/2022] Open
Abstract
Background Different approaches have been developed to dissect the interplay between transcription factors (TFs) and their cis-acting sequences on DNA in order to identify TF target genes. Here we used a combination of computational and experimental approaches to identify novel direct targets of TFAP2A, a key TF for a variety of physiological and pathological cellular processes. Gene expression profiles of HeLa cells either silenced for TFAP2A by RNA interference or not were previously compared and a set of differentially expressed genes was revealed. Results The regulatory regions of 494 TFAP2A-modulated genes were analyzed for the presence of TFAP2A binding sites, employing the canonical TFAP2A Positional Weight Matrix (PWM) reported in Jaspar http://jaspar.genereg.net/. 264 genes containing at least 2 high score TFAP2A binding sites were identified, showing a central role in "Cellular Movement" and "Cellular Development". In an attempt to identify TFs that could cooperate with TFAP2A, a statistically significant enrichment for SP1 binding sites was found for TFAP2A-activated but not repressed genes. The direct binding of TFAP2A or SP1 to a random subset of TFAP2A-modulated genes was demonstrated by Chromatin ImmunoPrecipitation (ChIP) assay and the TFAP2A-driven regulation of DCBLD2/ESDN/CLCP1 gene studied in details. Conclusions We proved that our computational approaches applied to microarray selected genes are valid tools to identify functional TF binding sites in gene regulatory regions as confirmed by experimental validations. In addition, we demonstrated a fine-tuned regulation of DCBLD2/ESDN transcription by TFAP2A.
Collapse
Affiliation(s)
- Francesca Orso
- Molecular Biotechnology Center (MBC), Department of Oncological Sciences, University of Torino, Torino, Italy
| | | | | | | | | | | |
Collapse
|
43
|
Abstract
Targeting cell infection using herpes simplex virus type 1 (HSV-1) vectors is a complicated issue as the process involves multiple interactions of viral envelope glycoproteins and cellular host surface proteins. In this study, we have inserted a human glioma-specific peptide sequence (denoted as MG11) into a peptide display HSV-1 amplicon vector replacing the heparan sulfate-binding domain of glycoprotein C (gC). The modified MG11:gC envelope recombinant vectors were subsequently packaged into virions in the presence of helper virus deleted for gC. Our results showed that the tropism of these HSV-1 recombinant virions was increased for human glioma cells in culture as compared with wild-type virions. The binding of these recombinant virions could also be blocked effectively by pre-incubating the cells with the glioma-specific peptide, indicating that MG11 peptide and the recombinant virions competed for the same or similar receptor-binding sites on the cell surface of human glioma cells. Furthermore, preferential homing of these virions was shown in xenograft glioma mouse model following intravascular delivery. Taken together, these results validated the hypothesis that HSV-1 binding to cells can be redirected to human gliomas through the incorporation of MG11 peptide sequence to the virions.
Collapse
|
44
|
Abstract
Neuroendocrine tumours (NETs) comprise a heterogenous group of malignancies with an often unpredictable course, and with limited treatment options. Thus, new diagnostic, prognostic, and therapeutic markers are needed. To shed new lights into the biology of NETs, we have by cDNA transcript profiling, sought to identify genes that are either up- or downregulated in NE as compared with non-NE tumour cells. A panel of six NET and four non-NET cell lines were examined, and out of 12 743 genes examined, we studied in detail the 200 most significantly differentially expressed genes in the comparison. In addition to potential new diagnostic markers (NEFM, CLDN4, PEROX2), the results point to genes that may be involved in the tumorigenesis (BEX1, TMEPAI, FOSL1, RAB32), and in the processes of invasion, progression and metastasis (MME, STAT3, DCBLD2) of NETs. Verification by real time qRT–PCR showed a high degree of consistency to the microarray results. Furthermore, the protein expression of some of the genes were examined. The results of our study has opened a window to new areas of research, by uncovering new candidate genes and proteins to be further investigated in the search for new prognostic, predictive, and therapeutic markers in NETs.
Collapse
|
45
|
Neufeld G, Kessler O. The semaphorins: versatile regulators of tumour progression and tumour angiogenesis. Nat Rev Cancer 2008; 8:632-45. [PMID: 18580951 DOI: 10.1038/nrc2404] [Citation(s) in RCA: 305] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The semaphorins and their receptors, the neuropilins and the plexins, were originally characterized as constituents of the complex regulatory system responsible for the guidance of axons during the development of the central nervous system. However, a growing body of evidence indicates that various semaphorins can either promote or inhibit tumour progression through the promotion or inhibition of processes such as tumour angiogenesis, tumour metastasis and tumour cell survival. This Review focuses on the emerging role of the semaphorins in cancer.
Collapse
Affiliation(s)
- Gera Neufeld
- Cancer Research and Vascular Biology Center, The Bruce Rappaport Faculty of Medicine, Technion, Israel Institute of Technology, P.O. Box 9679, 1 Efron Street, Haifa, 31096, Israel.
| | | |
Collapse
|
46
|
Potiron VA, Roche J, Drabkin HA. Semaphorins and their receptors in lung cancer. Cancer Lett 2008; 273:1-14. [PMID: 18625544 DOI: 10.1016/j.canlet.2008.05.032] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2008] [Revised: 02/28/2008] [Accepted: 05/23/2008] [Indexed: 12/30/2022]
Abstract
Semaphorins are a large family of secreted, transmembrane and GPI-linked proteins initially characterized in the development of the nervous system and axonal guidance. Semaphorins are expressed in many tissues where they regulate normal development, organ morphogenesis, immunity and angiogenesis. They affect the cytoskeleton, actin filament organization, microtubules and cell adhesion. Semaphorin signaling is transduced by plexins, which in the case of most class-3 semaphorins requires high-affinity neuropilin receptors. The neuropilins also function as receptors for VEGF and other growth factors, and their expression is often abnormal in tumors. In cancer, semaphorins have both tumor suppressor and tumor promoting functions. We review here the current status of semaphorins and their receptors in tumor development with a focus on lung cancer.
Collapse
Affiliation(s)
- Vincent A Potiron
- Division of Hematology/Oncology, Medical University of South Carolina, P.O. Box 250623, 96 Jonathan Lucas Street, Charleston, SC 29425, USA
| | | | | |
Collapse
|
47
|
Orso F, Penna E, Cimino D, Astanina E, Maione F, Valdembri D, Giraudo E, Serini G, Sismondi P, De Bortoli M, Taverna D. AP-2alpha and AP-2gamma regulate tumor progression via specific genetic programs. FASEB J 2008; 22:2702-14. [PMID: 18443366 DOI: 10.1096/fj.08-106492] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The events occurring during tumor formation and progression display similarities to some of the steps in embryonic morphogenesis. The family of AP-2 proteins consists of five different transcription factors (alpha, beta, gamma, delta, and epsilon) that play relevant roles in embryonic development, as demonstrated by the phenotypes of the corresponding knockout mice. Here, we show that AP-2alpha and AP-2gamma proteins play an essential role in tumorigenesis. Down-modulation of AP-2 expression in tumor cells by RNA interference (RNAi) led to enhanced tumor growth and reduced chemotherapy-induced cell death, as well as migration and invasion. Most of these biological modulations were rescued by AP-2 overexpression. We observed that increased xenotransplant growth was mostly due to highly enhanced proliferation of the tumor cells together with reduced innate immune cell recruitment. Moreover, we showed that migration impairment was mediated, at least in part, by secreted factors. To identify the genetic programs involved in tumorigenesis, we performed whole genome microarray analysis of AP-2alpha knockdown cells and observed that AP-2alpha regulates specific genes involved in cell cycle, cell death, adhesion, and migration. In particular, we showed that ESDN, EREG, and CXCL2 play a major role in AP-2 controlled migration, as ablation of any of these genes severely altered migration.
Collapse
Affiliation(s)
- Francesca Orso
- Institute for Cancer Research and Treatment, University of Torino, Via Nizza, 52, 10126 Torino, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Chiquet BT, Lidral AC, Stal S, Mulliken JB, Moreno LM, Arco-Burgos M, Valencia-Ramirez C, Blanton SH, Hecht JT. CRISPLD2: a novel NSCLP candidate gene. Hum Mol Genet 2007; 16:2241-8. [PMID: 17616516 PMCID: PMC3755375 DOI: 10.1093/hmg/ddm176] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Non-syndromic cleft lip with or without cleft palate (NSCLP) results from the complex interaction between genes and environmental factors. Candidate gene analysis and genome scans have been employed to identify the genes contributing to NSCLP. In this study, we evaluated the 16q24.1 chromosomal region, which has been identified by multiple genome scans as an NSCLP region of interest. Two candidate genes were found in the region: interferon regulatory factor 8 (IRF8) and cysteine-rich secretory protein LCCL domain containing 2 (CRISPLD2). Initially, Caucasian and Hispanic NSCLP multiplex families and simplex parent-child trios were genotyped for single nucleotide polymorphisms (SNPs) in both IRF8 and CRISPLD2. CRISPLD2 was subsequently genotyped in a data set comprised of NSCLP families from Colombia, South America. Linkage disequilibrium analysis identified a significant association between CRISPLD2 and NSCLP in both our Caucasian and Hispanic NSCLP cohorts. SNP rs1546124 and haplotypes between rs1546124 and either rs4783099 or rs16974880 were significant in the Caucasian multiplex population (P=0.01, P=0.002 and P=0.001, respectively). An altered transmission of CRISPLD2 SNPs rs8061351 (P=0.02) and rs2326398 (P=0.06) was detected in the Hispanic population. No association was found between CRISPLD2 and our Colombian population or IRF8 and NSCLP. In situ hybridization showed that CRISPLD2 is expressed in the mandible, palate and nasopharynx regions during craniofacial development at E13.5-E17.5, respectively. Altogether, these data suggest that genetic variation in CRISPLD2 has a role in the etiology of NSCLP.
Collapse
Affiliation(s)
- Brett T. Chiquet
- Department of Pediatrics, University of Texas Medical School at Houston
- University of Texas Dental Branch at Houston, Houston, TX 77030, USA
| | - Andrew C. Lidral
- Dows Institute for Dental Research, University of Iowa, Iowa City, IA, USA
- Department of Orthodontics, University of Iowa, Iowa City, IA, USA
| | - Samuel Stal
- Texas Children's Hospital, Houston, TX 77030, USA
| | | | - Lina M. Moreno
- Dows Institute for Dental Research, University of Iowa, Iowa City, IA, USA
| | - Mauricio Arco-Burgos
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Consuelo Valencia-Ramirez
- College of Dentistry, University of Antioquia, Medellín, Colombia, South America, Miami, FL 33101, USA
| | - Susan H. Blanton
- University of Miami Miller School of Medicine, Miami, FL 33101, USA
| | | |
Collapse
|
49
|
Rizzolio S, Tamagnone L. Semaphorin signals on the road to cancer invasion and metastasis. Cell Adh Migr 2007; 1:62-8. [PMID: 19329883 DOI: 10.4161/cam.1.2.4570] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Semaphorins are a large family of secreted and membrane-bound molecules initially implicated in the development of the nervous system and in axon guidance. More recently, they have been found to regulate cell adhesion and cell motility, angiogenesis, immune function and tumour progression. Notably, Semaphorins have been implicated with opposite functions in cancer: either as putative tumor suppressors and anti-angiogenic factors, or as mediating tumour angiogenesis, invasion and metastasis. Interestingly, Semaphorins may display divergent activities in different cell types. These multifaceted functions may be explained by the involvement of different kinds of semaphorin receptor complexes, and by the consequent activation of multiple signaling pathways, in different cells or different functional stages. Semaphorin signaling is largely mediated by the Plexins. However, semaphorin receptor complexes may also include Neuropilins and tyrosine kinases implicated in cancer. In this review, we will focus on major open questions concerning the potential role of Semaphorin signals in cancer.
Collapse
Affiliation(s)
- Sabrina Rizzolio
- Institute for Cancer Research and Treatment (IRCC), Division of Molecular Oncology, University of Torino Medical School, Candiolo, Italy
| | | |
Collapse
|