1
|
Zhao LL, Liu YJ, Guo QJ, Yan N, Yang J, Han JQ, Xie XH, Luo YS. TPM4 influences the initiation and progression of gastric cancer by modulating ferroptosis via SCD1. Clin Exp Med 2025; 25:115. [PMID: 40214825 PMCID: PMC11991984 DOI: 10.1007/s10238-025-01629-8] [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: 01/31/2025] [Accepted: 03/11/2025] [Indexed: 04/14/2025]
Abstract
Gastric cancer (GC) is a deadly disease with poor prognosis and few treatment options. Tropomyosin 4 (TPM4) is an actin-binding protein that stabilizes the cytoskeleton of cells and has an unclear role in GC. This study aimed to elucidate the role and underlying mechanisms of TPM4 in GC pathogenesis. The expression and diagnostic and prognostic value of TPM4 in GC were analyzed using bioinformatics. A nomogram based on TPM4 expression was created and validated with an external cohort. TPM4-knockdown GC cells and xenograft models in nude mice were used to study the function of TPM4 in vitro and in vivo. Proteomic and rescue experiments confirmed the regulatory effect of TPM4 on stearoyl-CoA desaturase 1 (SCD1) in GC. Immunohistochemistry verified the expression and correlation of the TPM4 and SCD1 proteins in GC tissues. Our study identified TPM4 as an oncogene in GC, suggesting its potential diagnostic and prognostic value. The TPM4-based nomogram showed potential prognostic value for clinical use. TPM4 knockdown inhibited GC cell proliferation, induced ferroptosis, and slowed tumor growth in vivo, which is achieved by inhibiting SCD1 expression. Immunohistochemical analysis of GC tissues revealed elevated expression levels of both TPM4 and SCD1 proteins, with a positive correlation observed between their expression. TPM4 is a promising target for new diagnostic, prognostic, and therapeutic strategies for GC. Downregulation of TPM4 inhibits GC cell growth and induces ferroptosis by suppressing SCD1 expression.
Collapse
Affiliation(s)
- Ling-Lin Zhao
- Research Center for High Altitude Medicine, Key Laboratory of High Altitude Medicine (Ministry of Education), Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Joint Research Key Lab for High Altitude Medicine), Qinghai University, Xining, 810001, China
- Qinghai Provincial People's Hospital, Xining, 810000, China
| | - Yu-Jun Liu
- Department of Oncology, Affiliated Hospital of Qinghai University, Xining, 810001, China
| | - Qi-Jing Guo
- Department of Oncology, Air Force Medical Center, PLA, Beijing, 100142, China
| | - Nan Yan
- Research Center for High Altitude Medicine, Key Laboratory of High Altitude Medicine (Ministry of Education), Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Joint Research Key Lab for High Altitude Medicine), Qinghai University, Xining, 810001, China
| | - Jie Yang
- Research Center for High Altitude Medicine, Key Laboratory of High Altitude Medicine (Ministry of Education), Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Joint Research Key Lab for High Altitude Medicine), Qinghai University, Xining, 810001, China
| | - Jing-Qi Han
- Department of Pathology, Affiliated Hospital of Qinghai University, Xining, 810001, China
| | - Xiao-Hong Xie
- Qinghai Provincial People's Hospital, Xining, 810000, China
| | - Yu-Shuang Luo
- Research Center for High Altitude Medicine, Key Laboratory of High Altitude Medicine (Ministry of Education), Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Joint Research Key Lab for High Altitude Medicine), Qinghai University, Xining, 810001, China.
- Department of Oncology, Affiliated Hospital of Qinghai University, Xining, 810001, China.
| |
Collapse
|
2
|
Li X, Wang Y, Ren M, Liu Q, Li J, Zhang L, Yao S, Tang L, Wen G, An J, Jin H, Tuo B. The role of chloride intracellular channel 4 in tumors. Cancer Cell Int 2025; 25:118. [PMID: 40140845 PMCID: PMC11948840 DOI: 10.1186/s12935-025-03737-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 03/07/2025] [Indexed: 03/28/2025] Open
Abstract
Tumors are among the most predominant health problems in the world, and the annual incidence of cancer is increasing globally; therefore, there is an urgent need to identify effective therapeutic targets. Chloride intracellular channel 4 (CLIC4) belongs to the family of chloride intracellular channels (CLICs), which are widely expressed in various tissues and organs, such as the brain, lung, pancreas, colorectum, and ovary, and play important roles in promoting apoptosis, promoting angiogenesis, maintaining normal proliferation of endothelial cells, and regulating the assembly and reconstruction of the cytoskeleton. The expression and function of CLIC4 in tumors varies. It has been reported that CLIC4 is low expressed in gastric cancer, skin cancer and prostate cancer, suggesting a tumor suppressor role. Interestingly, CLIC4 is overexpressed in pancreatic, ovarian and breast cancers, indicating a cancer-promoting role. CLIC4 expression is dysregulated in some solid tumors, which may be because CLIC4 is involved in the growth, migration or invasion of some cancer cells through various mechanisms. Regulation of CLIC4 expression may be a potential therapeutic strategy for some tumors. CLIC4 may be a promising therapeutic target and a biomarker for some cancers. In this study, we review the role of CLIC4 in several cancers and its value in the diagnosis and treatment of tumors.
Collapse
Affiliation(s)
- Xin Li
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, 149 Dalian Road, Huichuan, Zunyi, 563003, Guizhou, China
| | - Yongfeng Wang
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, 149 Dalian Road, Huichuan, Zunyi, 563003, Guizhou, China
| | - Minmin Ren
- Department of General Surgery, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, 563003, Guizhou Province, China
- Nursing School of Zunyi Medical University, Zunyi, 563003, Guizhou Province, China
| | - Qian Liu
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, 149 Dalian Road, Huichuan, Zunyi, 563003, Guizhou, China
| | - Jiajia Li
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, 149 Dalian Road, Huichuan, Zunyi, 563003, Guizhou, China
| | - Li Zhang
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, 149 Dalian Road, Huichuan, Zunyi, 563003, Guizhou, China
| | - Shun Yao
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, 149 Dalian Road, Huichuan, Zunyi, 563003, Guizhou, China
| | - Lulu Tang
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, 149 Dalian Road, Huichuan, Zunyi, 563003, Guizhou, China
| | - Guorong Wen
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, 149 Dalian Road, Huichuan, Zunyi, 563003, Guizhou, China
| | - Jiaxing An
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, 149 Dalian Road, Huichuan, Zunyi, 563003, Guizhou, China
| | - Hai Jin
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, 149 Dalian Road, Huichuan, Zunyi, 563003, Guizhou, China.
- The Collaborative Innovation Center of Tissue Damage Repair and Regenerative Medicine, Zunyi Medical University, Zunyi, 563003, China.
| | - Biguang Tuo
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, 149 Dalian Road, Huichuan, Zunyi, 563003, Guizhou, China.
- The Collaborative Innovation Center of Tissue Damage Repair and Regenerative Medicine, Zunyi Medical University, Zunyi, 563003, China.
| |
Collapse
|
3
|
Wang D, Zhang Y, Ren D, Meng C, Yang L. Bioinformatics analysis illustrates the functions of miR-377-5p in cervical cancer. Biotechnol Genet Eng Rev 2024; 40:4238-4249. [PMID: 37144663 DOI: 10.1080/02648725.2023.2208453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 04/25/2023] [Indexed: 05/06/2023]
Abstract
Cervical cancer (CC) is a frequent disease in women whose development is related with miRNA disorder. MiR-377-5p plays a negative role in the development of some tumors, while few studies have revealed its role in CC. In this study, the functions of miR-377-5p in CC were investigated by bioinformatics. Briefly, the expression and survival curve of miR-377-5p in CC was analyzed with the Cancer Genome Atlas (TCGA) database, and the abundance of miR-377-5p in clinical samples and CC cell lines were measured by qRT-PCR. Moreover, the MicroRNA Data Integration Portal (miRDIP) database was used to predict targets of miR-377-5p, and the Database for Annotation Visualization and Integrated Discovery (David) was used for enrichment analysis of the functions of the miR-377-5p. The Search Tool for the Retrieval of Interacting Genes (STRING) database was used to screen the hub targets of miR-377-5p. Moreover, the Gene Expression Profiling Interactive Analysis (GEPIA) database was used to analyze the abundance of the genes in CC. Results showed that decreased miR-377-5p was found in the CC tissues and cell lines, and low miR-377-5p was connected with poor prognosis of patients. Besides, the targets of miR-377-5p were enriched in the PI3K/AKT, MAPK and RAS signaling pathways. Moreover, CDC42, FLT1, TPM3 and CAV1 were screened as hub nodes in the targets of miR-377-5p, and increased CDC42, FLT1, TPM3 and CAV1 also indicated the poor survival rates of the patients in the long term. In conclusion, this study suggests that miR-377-5p downregulation is a biomarker event for CC progression.
Collapse
Affiliation(s)
- Dongjie Wang
- Department of Gynaecology, The First People's Hospital of Yunnan Province, Kunming, China
- Department of Gynaecology, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunan, China
| | - Yifeng Zhang
- Department of Gynaecology, The First People's Hospital of Yunnan Province, Kunming, China
- Department of Gynaecology, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunan, China
| | - Dongyan Ren
- Department of Gynaecology, The First People's Hospital of Yunnan Province, Kunming, China
- Department of Gynaecology, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunan, China
| | - Chunmei Meng
- Department of Gynaecology, The First People's Hospital of Yunnan Province, Kunming, China
- Department of Gynaecology, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunan, China
| | - Liufeng Yang
- Department of Gynaecology, The First People's Hospital of Yunnan Province, Kunming, China
- Department of Gynaecology, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunan, China
| |
Collapse
|
4
|
Mollet IG, Viana-Soares R, Cardoso-Pires C, Soares NL, Marto JP, Mendonça M, Queiroga CSF, Carvalho AS, Sequeira CO, Teixeira-Santos L, Fernandes TP, Aloria K, Pereira SA, Matthiesen R, Viana-Baptista M, Vieira HLA. Identification of human circulating factors following remote ischemic conditioning (RIC): Potential impact on stroke. Free Radic Biol Med 2024; 224:23-38. [PMID: 39151835 DOI: 10.1016/j.freeradbiomed.2024.08.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 08/12/2024] [Accepted: 08/13/2024] [Indexed: 08/19/2024]
Abstract
Remote ischemic conditioning (RIC) is a procedure consisting of short cycles of ischemia applied in a limb that activates endogenous protection in distant organs, such as the brain. Despite the promising outcomes of RIC, the biochemical factors governing inter-organ communication remain largely unexplored, particularly in humans. A pilot study on 20 healthy humans was performed to identify potential circulating biochemical factors involved in RIC signalling. Blood was collected before and immediately, 4 and 22 h after the end of RIC. To characterize the responses triggered by RIC, a combination of biochemical and proteomic analysis, along with functional in vitro tests in human cells, were performed. RIC did not alter the levels of nitric oxide, bilirubin and cell-free mitochondrial DNA. In contrast, carboxyhaemoglobin levels increased following RIC at all time points and young subset, suggesting endogenous production of carbon monoxide that is a cytoprotective gasotransmitter. Additionally, the levels of glutathione and cysteinylglycine bound to proteins also increased after RIC, while glutathione catabolism decreased. Plasma proteomic analysis identified overall 828 proteins. Several steps of statistical analysis (Student's t-test, repeated measures ANOVA, with Holm corrected pairwise p-values <0.05 threshold and fold change higher or lower than 100 %) leaded to the identification of 9 proteins with altered circulating levels in response to RIC at 4h and 22h. All 9 proteins are from extracellular space or exosomes, being involved in inflammation, angiogenesis or metabolism control. In addition, RIC-conditioned plasma from young subjects protected microglial cell culture against inflammatory stimuli, indicating an anti-inflammatory effect of RIC. Nevertheless, other functional tests in neurons or endothelial cells had no effect. Overall, we present some evidence for RIC-induced anti-inflammatory and antioxidant responses in healthy human subjects, in particular in young subjects. This study is a first step towards the disclosure of signalling factors involved in RIC-mediated inter-organ communication.
Collapse
Affiliation(s)
- Inês G Mollet
- UCIBIO, Applied Molecular Biosciences Unit, Department of Chemistry, NOVA School of Science and Technology, Universidade Nova de Lisboa, Caparica, Portugal; iNOVA4Health, NOVA Medical School|Faculdade de Ciências Médicas, NMS|FCM, Universidade NOVA de Lisboa, Portugal; Associate Laboratory i4HB, Institute for Health and Bioeconomy, NOVA School of Science and Technology, Universidade NOVA de Lisboa, Caparica, Portugal
| | - Ricardo Viana-Soares
- iNOVA4Health, NOVA Medical School|Faculdade de Ciências Médicas, NMS|FCM, Universidade NOVA de Lisboa, Portugal
| | - Catarina Cardoso-Pires
- UCIBIO, Applied Molecular Biosciences Unit, Department of Chemistry, NOVA School of Science and Technology, Universidade Nova de Lisboa, Caparica, Portugal; Associate Laboratory i4HB, Institute for Health and Bioeconomy, NOVA School of Science and Technology, Universidade NOVA de Lisboa, Caparica, Portugal
| | - Nuno L Soares
- iNOVA4Health, NOVA Medical School|Faculdade de Ciências Médicas, NMS|FCM, Universidade NOVA de Lisboa, Portugal
| | - João Pedro Marto
- iNOVA4Health, NOVA Medical School|Faculdade de Ciências Médicas, NMS|FCM, Universidade NOVA de Lisboa, Portugal; Department of Neurology, Hospital de Egas Moniz, Centro Hospitalar Lisboa Ocidental, Portugal; Centro Clínico Académico de Lisboa CCAL, Lisboa, Portugal
| | - Marcelo Mendonça
- iNOVA4Health, NOVA Medical School|Faculdade de Ciências Médicas, NMS|FCM, Universidade NOVA de Lisboa, Portugal; Champalimaud Research, Champalimaud Center for the Unknown, Lisbon, Portugal
| | - Cláudia S F Queiroga
- iNOVA4Health, NOVA Medical School|Faculdade de Ciências Médicas, NMS|FCM, Universidade NOVA de Lisboa, Portugal
| | - Ana S Carvalho
- iNOVA4Health, NOVA Medical School|Faculdade de Ciências Médicas, NMS|FCM, Universidade NOVA de Lisboa, Portugal
| | - Catarina O Sequeira
- iNOVA4Health, NOVA Medical School|Faculdade de Ciências Médicas, NMS|FCM, Universidade NOVA de Lisboa, Portugal; Centro Clínico Académico de Lisboa CCAL, Lisboa, Portugal
| | - Luísa Teixeira-Santos
- iNOVA4Health, NOVA Medical School|Faculdade de Ciências Médicas, NMS|FCM, Universidade NOVA de Lisboa, Portugal; Centro Clínico Académico de Lisboa CCAL, Lisboa, Portugal
| | - Tatiana P Fernandes
- UCIBIO, Applied Molecular Biosciences Unit, Department of Chemistry, NOVA School of Science and Technology, Universidade Nova de Lisboa, Caparica, Portugal; Associate Laboratory i4HB, Institute for Health and Bioeconomy, NOVA School of Science and Technology, Universidade NOVA de Lisboa, Caparica, Portugal
| | - Kerman Aloria
- Proteomics Core Facility-SGIKER, University of the Basque Country UPV/EHU, Vizcaya, Spain
| | - Sofia A Pereira
- iNOVA4Health, NOVA Medical School|Faculdade de Ciências Médicas, NMS|FCM, Universidade NOVA de Lisboa, Portugal; Centro Clínico Académico de Lisboa CCAL, Lisboa, Portugal
| | - Rune Matthiesen
- iNOVA4Health, NOVA Medical School|Faculdade de Ciências Médicas, NMS|FCM, Universidade NOVA de Lisboa, Portugal
| | - Miguel Viana-Baptista
- iNOVA4Health, NOVA Medical School|Faculdade de Ciências Médicas, NMS|FCM, Universidade NOVA de Lisboa, Portugal; Department of Neurology, Hospital de Egas Moniz, Centro Hospitalar Lisboa Ocidental, Portugal; Centro Clínico Académico de Lisboa CCAL, Lisboa, Portugal
| | - Helena L A Vieira
- UCIBIO, Applied Molecular Biosciences Unit, Department of Chemistry, NOVA School of Science and Technology, Universidade Nova de Lisboa, Caparica, Portugal; iNOVA4Health, NOVA Medical School|Faculdade de Ciências Médicas, NMS|FCM, Universidade NOVA de Lisboa, Portugal; Associate Laboratory i4HB, Institute for Health and Bioeconomy, NOVA School of Science and Technology, Universidade NOVA de Lisboa, Caparica, Portugal.
| |
Collapse
|
5
|
Wojtera B, Ostrowska K, Szewczyk M, Masternak MM, Golusiński W. Chloride intracellular channels in oncology as potential novel biomarkers and personalized therapy targets: a systematic review. Rep Pract Oncol Radiother 2024; 29:258-270. [PMID: 39143969 PMCID: PMC11321771 DOI: 10.5603/rpor.99674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 02/29/2024] [Indexed: 08/16/2024] Open
Abstract
Background The chloride intracellular channels (CLICs) family includes six ion channels (CLIC1-CLIC6) expressed on the cellular level and secreted into interstitial fluid and blood. They are involved in the physiological functioning of multiple systems as well as the pathogenetic processes of cancer. CLICs play essential roles in the tumor microenvironment. The current systematic review aimed at identifying and summarizing the research of CLICs in oncology on clinical material to assess CLICs' potential as novel biomarkers and personalized therapy targets. Materials and methods The authors systematically searched the PubMed database for original articles concerning CLIC research on clinical material of all types of cancer - fluids and tissues. Results Fifty-three articles investigating in summary 3944 clinical samples were qualified for the current review. Studied material included 3438 tumor samples (87%), 437 blood samples (11%), and 69 interstitial fluid samples (2%). Studies investigated 21 cancer types, mostly hepatocellular carcinoma, colorectal, ovarian, and gastric cancer. Importantly, CLIC1, CLIC2, CLIC3, CLIC4, and CLIC5 were differently expressed in cancerous tissues and patients' blood compared to healthy controls. Moreover, CLICs were found to be involved in several cancer-associated signaling pathways, such as PI3K/AKT, MAPK/ERK, and MAPK/p38. Conclusion CLIC family members may be candidates for potential novel cancer biomarkers due to the contrast in their expression between cancerous and healthy tissues and secretion to the interstitial fluid and blood. CLICs are investigated as potential therapeutic targets because of their involvement in cancer pathogenesis and tumor microenvironment.
Collapse
Affiliation(s)
- Bartosz Wojtera
- Department of Head and Neck Surgery, Greater Poland Cancer Centre, Poznan University of Medical Sciences, Poznan, Poland
- Radiobiology Laboratory, Department of Medical Physics, Greater Poland Cancer Centre, Poznan University of Medical Sciences, Poznan, Poland
| | - Kamila Ostrowska
- Department of Head and Neck Surgery, Greater Poland Cancer Centre, Poznan University of Medical Sciences, Poznan, Poland
- Radiobiology Laboratory, Department of Medical Physics, Greater Poland Cancer Centre, Poznan University of Medical Sciences, Poznan, Poland
| | - Mateusz Szewczyk
- Department of Head and Neck Surgery, Greater Poland Cancer Centre, Poznan University of Medical Sciences, Poznan, Poland
| | - Michał M. Masternak
- Department of Head and Neck Surgery, Greater Poland Cancer Centre, Poznan University of Medical Sciences, Poznan, Poland
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida, United States
| | - Wojciech Golusiński
- Department of Head and Neck Surgery, Greater Poland Cancer Centre, Poznan University of Medical Sciences, Poznan, Poland
| |
Collapse
|
6
|
Zhou X, Li Z, Chen H, Jiao M, Zhou C, Li H. Relevance Analysis of TPM2 and Clinicopathological Characteristics in Breast Cancer. Int J Gen Med 2024; 17:59-74. [PMID: 38221941 PMCID: PMC10788065 DOI: 10.2147/ijgm.s442004] [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: 10/07/2023] [Accepted: 12/28/2023] [Indexed: 01/16/2024] Open
Abstract
Background The function of tropomyosin 2 (TPM2) in breast cancer is still far understudied. In this study, we aim to explore the roles of TPM2 in breast cancer progression. Methods This research included 155 breast cancer tissues. The expression of TPM2 was analyzed by immunohistochemical staining and grading. The mRNA expression of TPM2 in pan-cancer was analyzed with The Cancer Genome Atlas (TCGA) data plate form. The differential expression of TPM2 protein and the differential promoter methylation level of TPM2 between breast cancer tissues and normal breast tissues were analyzed by the UALCAN online database. The relationship between TPM2 and signaling pathways was interpreted by Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and Gene Set Enrichment Analysis (GSEA) pathway enrichment analyses. The survival curve of TPM2 was analyzed across the Kaplan-Meier plotter online database. Furthermore, the relationship between TPM2 expression and infiltrating macrophages was validated through in vitro co-culture experiments. Results TPM2 expression was significantly down-regulated in breast cancer samples. In addition, TPM2 expression was correlated with lymph node metastasis and high-grade histopathological morphology. The receiver operating characteristic (ROC) curve indicated that TPM2 expression could well distinguish between normal breast tissue and breast cancer tissue. TPM2 may have potential value in breast cancer diagnosis. Bioinformatics analysis illustrated that TPM2 was mainly involved in extracellular matrix organization, collagen fibril organization, cell junction assembly, focal adhesion, cAMP signaling pathway, estrogen signaling pathway, Wnt signaling pathway, and adaptive immune system. TPM2 expression was correlated with immune infiltrating cells and immune checkpoint molecules. Our in vitro co-culture experiments showed that the M2 macrophages could upregulate the expression of TPM2. Conclusion TPM2 may play key roles in breast cancer occurrence and development, especially in cancer metastasis. TPM2 may be a potential biomarker for breast cancer diagnosis.
Collapse
Affiliation(s)
- Xingchen Zhou
- Department of Pathology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, People’s Republic of China
| | - Zhishuang Li
- Department of Pathology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, People’s Republic of China
| | - Huan Chen
- Department of Pathology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, People’s Republic of China
| | - Meng Jiao
- Department of Pathology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, People’s Republic of China
| | - Chengjun Zhou
- Department of Pathology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, People’s Republic of China
| | - Hui Li
- Department of Pathology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, People’s Republic of China
| |
Collapse
|
7
|
Sanchez VC, Craig‐Lucas A, Cataisson C, Carofino BL, Yuspa SH. Crosstalk between tumor and stroma modifies CLIC4 cargo in extracellular vesicles. JOURNAL OF EXTRACELLULAR BIOLOGY 2023; 2:e118. [PMID: 38264628 PMCID: PMC10803055 DOI: 10.1002/jex2.118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 09/08/2023] [Accepted: 09/11/2023] [Indexed: 01/25/2024]
Abstract
Mouse models of breast cancer have revealed that tumor-bearing hosts must express the oxidoreductase CLIC4 to develop lung metastases. In the absence of host CLIC4, primary tumors grow but the lung premetastatic niche is defective for metastatic seeding. Primary breast cancer cells release EVs that incorporate CLIC4 as cargo and circulate in plasma of wildtype tumor-bearing hosts. CLIC4-deficient breast cancer cells also form tumors in wildtype hosts and release EVs in plasma, but these EVs lack CLIC4, suggesting that the tumor is the source of the plasma-derived EVs that carry CLIC4 as cargo. Paradoxically, circulating EVs are also devoid of CLIC4 when CLIC4-expressing primary tumors are grown in CLIC4 knockout hosts. Thus, the incorporation of CLIC4 (and perhaps other factors) as EV cargo released from tumors involves specific signals from the surrounding stroma determined by its genetic composition. Since CLIC4 is also detected in circulating EVs from human breast cancer patients, future studies will address its association with disease.
Collapse
Affiliation(s)
- Vanesa C. Sanchez
- Center for Drug Evaluation and ResearchU.S. Food and Drug AdministrationSilver SpringMarylandUSA
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer InstituteNational Institutes of HealthBethesdaMarylandUSA
| | - Alayna Craig‐Lucas
- Department of SurgeryLehigh Valley Health NetworkAllentownPennsylvaniaUSA
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer InstituteNational Institutes of HealthBethesdaMarylandUSA
| | - Christophe Cataisson
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer InstituteNational Institutes of HealthBethesdaMarylandUSA
| | - Brandi L. Carofino
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer InstituteNational Institutes of HealthBethesdaMarylandUSA
| | - Stuart H. Yuspa
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer InstituteNational Institutes of HealthBethesdaMarylandUSA
| |
Collapse
|
8
|
Beer LA, Yin X, Ding J, Senapati S, Sammel MD, Barnhart KT, Liu Q, Speicher DW, Goldman AR. Identification and verification of plasma protein biomarkers that accurately identify an ectopic pregnancy. Clin Proteomics 2023; 20:37. [PMID: 37715129 PMCID: PMC10503165 DOI: 10.1186/s12014-023-09425-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 08/21/2023] [Indexed: 09/17/2023] Open
Abstract
BACKGROUND Differentiating between a normal intrauterine pregnancy (IUP) and abnormal conditions including early pregnancy loss (EPL) or ectopic pregnancy (EP) is a major clinical challenge in early pregnancy. Currently, serial β-human chorionic gonadotropin (β-hCG) and progesterone are the most commonly used plasma biomarkers for evaluating pregnancy prognosis when ultrasound is inconclusive. However, neither biomarker can predict an EP with sufficient and reproducible accuracy. Hence, identification of new plasma biomarkers that can accurately diagnose EP would have great clinical value. METHODS Plasma was collected from a discovery cohort of 48 consenting women having an IUP, EPL, or EP. Samples were analyzed by liquid chromatography-tandem mass spectrometry (LC-MS/MS) followed by a label-free proteomics analysis to identify significant changes between pregnancy outcomes. A panel of 14 candidate biomarkers were then verified in an independent cohort of 74 women using absolute quantitation by targeted parallel reaction monitoring mass spectrometry (PRM-MS) which provided the capacity to distinguish between closely related protein isoforms. Logistic regression and Lasso feature selection were used to evaluate the performance of individual biomarkers and panels of multiple biomarkers to predict EP. RESULTS A total of 1391 proteins were identified in an unbiased plasma proteome discovery. A number of significant changes (FDR ≤ 5%) were identified when comparing EP vs. non-EP (IUP + EPL). Next, 14 candidate biomarkers (ADAM12, CGA, CGB, ISM2, NOTUM, PAEP, PAPPA, PSG1, PSG2, PSG3, PSG9, PSG11, PSG6/9, and PSG8/1) were verified as being significantly different between EP and non-EP in an independent cohort (FDR ≤ 5%). Using logistic regression models, a risk score for EP was calculated for each subject, and four multiple biomarker logistic models were identified that performed similarly and had higher AUCs than models with single predictors. CONCLUSIONS Overall, four multivariable logistic models were identified that had significantly better prediction of having EP than those logistic models with single biomarkers. Model 4 (NOTUM, PAEP, PAPPA, ADAM12) had the highest AUC (0.987) and accuracy (96%). However, because the models are statistically similar, all markers in the four models and other highly correlated markers should be considered in further validation studies.
Collapse
Affiliation(s)
- Lynn A Beer
- Molecular and Cellular Oncogenesis Program, The Wistar Institute, 3601 Spruce Street, Philadelphia, PA, 19104, USA
| | - Xiangfan Yin
- Molecular and Cellular Oncogenesis Program, The Wistar Institute, 3601 Spruce Street, Philadelphia, PA, 19104, USA
| | - Jianyi Ding
- Molecular and Cellular Oncogenesis Program, The Wistar Institute, 3601 Spruce Street, Philadelphia, PA, 19104, USA
| | - Suneeta Senapati
- Department of Obstetrics and Gynecology, University of Pennsylvania, Philadelphia, PA, USA
| | - Mary D Sammel
- Department of Biostatistics and Informatics, Colorado School of Public Health, Aurora, CO, USA
| | - Kurt T Barnhart
- Department of Obstetrics and Gynecology, University of Pennsylvania, Philadelphia, PA, USA
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, PA, USA
| | - Qin Liu
- Molecular and Cellular Oncogenesis Program, The Wistar Institute, 3601 Spruce Street, Philadelphia, PA, 19104, USA.
| | - David W Speicher
- Molecular and Cellular Oncogenesis Program, The Wistar Institute, 3601 Spruce Street, Philadelphia, PA, 19104, USA.
| | - Aaron R Goldman
- Molecular and Cellular Oncogenesis Program, The Wistar Institute, 3601 Spruce Street, Philadelphia, PA, 19104, USA.
| |
Collapse
|
9
|
Yan J, Xie B, Tian Y, An W, Peng Z, Liu Z, Li J, Li L. MicroRNA-5195-3p mediated malignant biological behaviour of insulin-resistant liver cancer cells via SOX9 and TPM4. BMC Cancer 2023; 23:557. [PMID: 37328795 DOI: 10.1186/s12885-023-11068-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 06/14/2023] [Indexed: 06/18/2023] Open
Abstract
BACKGROUND Primary liver cancer is a malignant tumour of the digestive system, ranking second in cancer mortality in China. In different types of cancer, such as liver cancer, microRNAs (miRNAs) have been shown to be dysregulated. However, little is known about the role of miR-5195-3p in insulin-resistant liver cancer. METHODS AND RESULTS In this study, in vitro and in vivo experiments were conducted to identify the altered biological behaviour of insulin-resistant hepatoma cells (HepG2/IR), and we proved that HepG2/IR cells had stronger malignant biological behaviour. Functional experiments showed that enhanced expression of miR-5195-3p could inhibit the proliferation, migration, invasion, epithelial-mesenchymal transition (EMT) and chemoresistance of HepG2/IR cells, while impaired expression of miR-5195-3p in HepG2 cells resulted in the opposite effects. Bioinformatics prediction and dual luciferase reporter gene assays proved that SOX9 and TPM4 were the target genes of miR-5195-3p in hepatoma cells. CONCLUSIONS In conclusion, our study demonstrated that miR-5195-3p plays a critical role in insulin-resistant hepatoma cells and might be a potential therapeutic target for liver cancer.
Collapse
Affiliation(s)
- Jing Yan
- Department of Clinical Laboratory Center, The Second Hospital of Lanzhou University, Lanzhou, 730000, Gansu, China
- Department of Clinical Laboratory Center, Gansu Provincial Maternity and Child-care Hospital (Gansu Province Central Hospital), Lanzhou, 730000, Gansu, China
| | - Bei Xie
- Department of Medical Laboratory Animal Science, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, Gansu, China.
| | - Ye Tian
- Department of Clinical Laboratory Center, The Second Hospital of Lanzhou University, Lanzhou, 730000, Gansu, China
| | - Wenqin An
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, Gansu, China
| | - Zhiheng Peng
- Department of Clinical Laboratory Center, The Second Hospital of Lanzhou University, Lanzhou, 730000, Gansu, China
| | - Zhuan Liu
- Department of Clinical Laboratory Center, The Second Hospital of Lanzhou University, Lanzhou, 730000, Gansu, China
| | - Jing Li
- Department of Clinical Laboratory Center, The Second Hospital of Lanzhou University, Lanzhou, 730000, Gansu, China
| | - Linjing Li
- Department of Clinical Laboratory Center, The Second Hospital of Lanzhou University, Lanzhou, 730000, Gansu, China.
| |
Collapse
|
10
|
Duan P, Cui J, Li H, Yuan L. Tropomyosin 2 exerts anti-tumor effects in lung adenocarcinoma and is a novel prognostic biomarker. Histol Histopathol 2023; 38:669-680. [PMID: 36102257 DOI: 10.14670/hh-18-514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
BACKGROUND Tropomyosin 2 (TPM2), a member of the actin filament binding protein family, plays distinct roles in the progression of different cancer types. Until now, there has been no study reporting TPM2 expression nor its function in lung adenocarcinoma (LUAD). METHODS In the present study, we examined the expression profile of TPM2 by immunohistochemistry (IHC). The clinical significance of TPM2 was assessed by univariate and multivariate analyses. Function of TPM2 in LUAD was evaluated by knockdown and overexpression strategies in three LUAD cell lines, followed by proliferation and invasion assays. Xenografts were conducted in nude mice to further validate the tumor-related role of TPM2. RESULTS Our results showed that TPM2 was downregulated in LUAD specimens and the low expression of TPM2 was associated with poor outcomes of LUAD patients. Overexpressing TPM2 inhibited cell proliferation and invasion of LUAD cell lines, while silencing TPM2 exerted the opposite effects. The effects of TPM2 in LUAD were further confirmed by xenograft assays. CONCLUSIONS Our results indicated that TPM2 exerted an anti-oncogenic role in LUAD via inhibiting tumor progression, thus providing a novel direction for the prognostic prediction and disease treatment.
Collapse
Affiliation(s)
- Peng Duan
- Department of Oncology, The Third People's Hospital of Qingdao, Qingdao, China
| | - Jing Cui
- Department of Emergency, The Third People's Hospital of Qingdao, Qingdao, China
| | - Hongyan Li
- Department of Oncology, The Third People's Hospital of Qingdao, Qingdao, China
| | - Lei Yuan
- Department of Respiratory, The Third People's Hospital of Qingdao, Qingdao, China.
| |
Collapse
|
11
|
Ramírez A, Ogonaga-Borja I, Acosta B, Chiliquinga AJ, de la Garza J, Gariglio P, Ocádiz-Delgado R, Bañuelos C, Camacho J. Ion Channels and Personalized Medicine in Gynecological Cancers. Pharmaceuticals (Basel) 2023; 16:800. [PMID: 37375748 DOI: 10.3390/ph16060800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Revised: 05/22/2023] [Accepted: 05/24/2023] [Indexed: 06/29/2023] Open
Abstract
Targeted therapy against cancer plays a key role in delivering safer and more efficient treatments. In the last decades, ion channels have been studied for their participation in oncogenic processes because their aberrant expression and/or function have been associated with different types of malignancies, including ovarian, cervical, and endometrial cancer. The altered expression or function of several ion channels have been associated with tumor aggressiveness, increased proliferation, migration, invasion, and metastasis of cancer cells and with poor prognosis in gynecological cancer patients. Most ion channels are integral membrane proteins easily accessible by drugs. Interestingly, a plethora of ion channel blockers have demonstrated anticancer activity. Consequently, some ion channels have been proposed as oncogenes, cancer, and prognostic biomarkers, as well as therapeutic targets in gynecological cancers. Here, we review the association of ion channels with the properties of cancer cells in these tumors, which makes them very promising candidates to be exploited in personalized medicine. The detailed analysis of the expression pattern and function of ion channels could help to improve the clinical outcomes in gynecological cancer patients.
Collapse
Affiliation(s)
- Ana Ramírez
- Facultad de Ciencias Químicas e Ingeniería, Universidad Autónoma de Baja California, Calzada Universidad 14418, Parque Industrial Internacional, Tijuana 22390, Mexico
| | - Ingrid Ogonaga-Borja
- Grupo de Investigación de Ciencias en Red, Universidad Técnica del Norte, Av. 17 de Julio 5-21, Ibarra 100105, Ecuador
- Departamento de Farmacología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Av. Instituto Politécnico Nacional 2508, Ciudad de Mexico 07360, Mexico
| | - Brenda Acosta
- Grupo de Investigación de Ciencias en Red, Universidad Técnica del Norte, Av. 17 de Julio 5-21, Ibarra 100105, Ecuador
- Departamento de Farmacología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Av. Instituto Politécnico Nacional 2508, Ciudad de Mexico 07360, Mexico
| | - Andrea Jazmín Chiliquinga
- Grupo de Investigación de Ciencias en Red, Universidad Técnica del Norte, Av. 17 de Julio 5-21, Ibarra 100105, Ecuador
| | - Jaime de la Garza
- Unidad de Oncología Torácica y Laboratorio de Medicina Personalizada, Instituto Nacional de Cancerología (INCan), San Fernando No. 22, Tlalpan, Ciudad de Mexico14080, Mexico
| | - Patricio Gariglio
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Av. Instituto Politécnico Nacional 2508, Ciudad de Mexico 07360, Mexico
| | - Rodolfo Ocádiz-Delgado
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Av. Instituto Politécnico Nacional 2508, Ciudad de Mexico 07360, Mexico
| | - Cecilia Bañuelos
- Programa Transdisciplinario en Desarrollo Científico y Tecnológico para la Sociedad, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Av. Instituto Politécnico Nacional 2508, Ciudad de Mexico 07360, Mexico
| | - Javier Camacho
- Departamento de Farmacología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Av. Instituto Politécnico Nacional 2508, Ciudad de Mexico 07360, Mexico
| |
Collapse
|
12
|
Guo Q, Zhao L, Yan N, Li Y, Guo C, Dang S, Shen X, Han J, Luo Y. Integrated pan-cancer analysis and experimental verification of the roles of tropomyosin 4 in gastric cancer. Front Immunol 2023; 14:1148056. [PMID: 36993958 PMCID: PMC10041708 DOI: 10.3389/fimmu.2023.1148056] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 02/27/2023] [Indexed: 03/14/2023] Open
Abstract
ObjectiveTo investigate the function of tropomyosin 4 (TPM4) using pan-cancer data, especially in gastric cancer (GC), using comprehensive bioinformatics analysis and molecular experiments.MethodsWe used UCSC Xena, The Cancer Genome Atlas (TCGA), Genotype-Tissue Expression Project (GTEx), TIMER2.0, GEPIA, cBioPortal, Xiantao tool, and UALCAN websites and databases for the extraction of pan-cancer data on TPM4. TPM4 expression was investigated with respect to prognosis, genetic alterations, epigenetic alterations, and immune infiltration. RNA22, miRWalk, miRDB, Starbase 2.0, and Cytoscape were used for identifying and constructing the regulatory networks of lncRNAs, miRNAs, and TPM4 in GC. Data from GSCALite, drug bank databases, and Connectivity Map (CMap) were used to analyze the sensitivity of drugs dependent on TPM4 expression. Gene Ontology (GO), enrichment analyses of the Kyoto Encyclopedia of Genes and Genomes (KEGG), wound healing assays, and (Matrigel) transwell experiments were used to investigate the biological functions of TPM4 in GC.ResultThe findings of the comprehensive pan-cancer analysis revealed that TPM4 has a certain diagnostic and prognosis value in most cancers. Alterations in the expression of TPM4, including duplications and deep mutations, and epigenetic alterations revealed that TPM4 expression is related to the expression of DNA methylation inhibitors and RNA methylation regulators at high concentrations. Besides, TPM4 expression was found to correlate with immune cell infiltration, immune checkpoint (ICP) gene expression, the tumor mutational burden (TMB), and microsatellite instability (MSI). Neoantigens (NEO) were also found to influence its response to immunotherapy. A lncRNA-miRNA -TPM4 network was found to regulate GC development and progression. TPM4 expression was related to docetaxel,5-fluorouracil, and eight small molecular targeted drugs sensitivity. Gene function enrichment analyses revealed that genes that were co-expressed with TPM4 were enriched within the extracellular matrix (ECM)-related pathways. Wound-healing and (Matrigel) transwell assays revealed that TPM4 promotes cell migration and invasion. TPM4, as an oncogene, plays a biological role, perhaps via ECM remodeling in GC.ConclusionsTPM4 is a prospective marker for the diagnosis, treatment outcome, immunology, chemotherapy, and small molecular drugs targeted for pan-cancer treatment, including GC treatment. The lncRNA-miRNA-TPM4network regulates the mechanism underlying GC progression. TPM4 may facilitate the invasion and migration of GC cells, possibly through ECM remodeling.
Collapse
Affiliation(s)
- Qijing Guo
- Research Center for High Altitude Medicine, Key Laboratory of High Altitude Medicine (Ministry of Education), Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinhai Province (Qinghai-Utah Joint Research Key Lab for High Altitude Medicine), Laboratory for High Altitude Medicine of Qinghai Province, Qinghai University, Xining, China
- Department of Oncology, Affiliated Hospital of Qinghai University, Xining, China
| | - Linglin Zhao
- Research Center for High Altitude Medicine, Key Laboratory of High Altitude Medicine (Ministry of Education), Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinhai Province (Qinghai-Utah Joint Research Key Lab for High Altitude Medicine), Laboratory for High Altitude Medicine of Qinghai Province, Qinghai University, Xining, China
| | - Nan Yan
- Research Center for High Altitude Medicine, Key Laboratory of High Altitude Medicine (Ministry of Education), Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinhai Province (Qinghai-Utah Joint Research Key Lab for High Altitude Medicine), Laboratory for High Altitude Medicine of Qinghai Province, Qinghai University, Xining, China
| | - Yan Li
- Department of Oncology, Affiliated Hospital of Qinghai University, Xining, China
| | - Cuiping Guo
- Department of Oncology, Affiliated Hospital of Qinghai University, Xining, China
| | - Shengyan Dang
- Department of Oncology, Affiliated Hospital of Qinghai University, Xining, China
| | - Xianliang Shen
- Department of Oncology, Affiliated Hospital of Qinghai University, Xining, China
| | - Jianfang Han
- Department of Oncology, Affiliated Hospital of Qinghai University, Xining, China
| | - Yushuang Luo
- Research Center for High Altitude Medicine, Key Laboratory of High Altitude Medicine (Ministry of Education), Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinhai Province (Qinghai-Utah Joint Research Key Lab for High Altitude Medicine), Laboratory for High Altitude Medicine of Qinghai Province, Qinghai University, Xining, China
- Department of Oncology, Affiliated Hospital of Qinghai University, Xining, China
- *Correspondence: Yushuang Luo,
| |
Collapse
|
13
|
Al Khamici H, Sanchez VC, Yan H, Cataisson C, Michalowski AM, Yang HH, Li L, Lee MP, Huang J, Yuspa SH. The oxidoreductase CLIC4 is required to maintain mitochondrial function and resistance to exogenous oxidants in breast cancer cells. J Biol Chem 2022; 298:102275. [PMID: 35863434 PMCID: PMC9418444 DOI: 10.1016/j.jbc.2022.102275] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 06/01/2022] [Accepted: 06/02/2022] [Indexed: 02/07/2023] Open
Abstract
The chloride intracellular channel-4 (CLIC4) is one of the six highly conserved proteins in the CLIC family that share high structural homology with GST-omega in the GST superfamily. While CLIC4 is a multifunctional protein that resides in multiple cellular compartments, the discovery of its enzymatic glutaredoxin-like activity in vitro suggested that it could function as an antioxidant. Here, we found that deleting CLIC4 from murine 6DT1 breast tumor cells using CRISPR enhanced the accumulation of reactive oxygen species (ROS) and sensitized cells to apoptosis in response to H2O2 as a ROS-inducing agent. In intact cells, H2O2 increased the expression of both CLIC4 mRNA and protein. In addition, increased superoxide production in 6DT1 cells lacking CLIC4 was associated with mitochondrial hyperactivity including increased mitochondrial membrane potential and mitochondrial organelle enlargement. In the absence of CLIC4, however, H2O2-induced apoptosis was associated with low expression and degradation of the antiapoptotic mitochondrial protein Bcl2 and the negative regulator of mitochondrial ROS, UCP2. Furthermore, transcriptomic profiling of H2O2-treated control and CLIC4-null cells revealed upregulation of genes associated with ROS-induced apoptosis and downregulation of genes that sustain mitochondrial functions. Accordingly, tumors that formed from transplantation of CLIC4-deficient 6DT1 cells were highly necrotic. These results highlight a critical role for CLIC4 in maintaining redox-homeostasis and mitochondrial functions in 6DT1 cells. Our findings also raise the possibility of targeting CLIC4 to increase cancer cell sensitivity to chemotherapeutic drugs that are based on elevating ROS in cancer cells.
Collapse
Affiliation(s)
- Heba Al Khamici
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Center, National Institutes of Health. Bethesda, Maryland, USA
| | - Vanesa C Sanchez
- Office of Science, Division of Nonclinical Science, Center for Tobacco Products, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - Hualong Yan
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Center, National Institutes of Health. Bethesda, Maryland, USA
| | - Christophe Cataisson
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Center, National Institutes of Health. Bethesda, Maryland, USA
| | - Aleksandra M Michalowski
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Center, National Institutes of Health. Bethesda, Maryland, USA
| | - Howard H Yang
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Center, National Institutes of Health. Bethesda, Maryland, USA
| | - Luowei Li
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Center, National Institutes of Health. Bethesda, Maryland, USA
| | - Maxwell P Lee
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Center, National Institutes of Health. Bethesda, Maryland, USA
| | - Jing Huang
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Center, National Institutes of Health. Bethesda, Maryland, USA
| | - Stuart H Yuspa
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Center, National Institutes of Health. Bethesda, Maryland, USA.
| |
Collapse
|
14
|
Wang J, Yang Y, Du B. Clinical Characterization and Prognostic Value of TPM4 and Its Correlation with Epithelial–Mesenchymal Transition in Glioma. Brain Sci 2022; 12:brainsci12091120. [PMID: 36138856 PMCID: PMC9497136 DOI: 10.3390/brainsci12091120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 08/19/2022] [Accepted: 08/21/2022] [Indexed: 11/26/2022] Open
Abstract
Tropomyosin 4 (TPM4) has been reported as an oncogenic gene across different malignancies. However, the role of TPM4 in glioma remains unclear. This study aimed to determine the clinical characterization and prognostic value of TPM4 in gliomas. Transcriptome expression and clinical information were collected from the CGGA and TCGA datasets, which included 998 glioma patients. ScRNA-seq data were obtained from CGGA. R software was utilized for statistical analyses. There was a positive correlation between TPM4 and WHO grades. IDH-wildtype and mesenchymal subtype gliomas were accompanied by TPM4 upregulation. GO and GSEA analysis suggested that TPM4 was profoundly associated with epithelial-to-mesenchymal transition (EMT). Subsequent GSVA revealed a robust correlation between TPM4 and three signaling pathways of EMT (hypoxia, TGF-β, PI3K/AKT). Furthermore, TPM4 showed a synergistic effect with mesenchymal biomarkers, particularly with N-cadherin, Slug, Snail, TWIST1, and vimentin. ScRNA-seq analysis suggested that higher TPM4 was mainly attributed to tumor cells and macrophages and associated with tumor cell progression and macrophage polarization. Finally, high TPM4 was significantly associated with unfavorable outcomes. In conclusion, our findings indicate that TPM4 is significantly correlated with more malignant characteristics of gliomas, potentially through involvement in EMT. TPM4 could predict worse survival for patients with glioma.
Collapse
Affiliation(s)
- Jin Wang
- Department of Emergency, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University), Shenzhen 518020, China
| | - Ying Yang
- Department of Pediatrics, Futian Women and Children Health Institute, Shenzhen 518045, China
| | - Bo Du
- Department of Emergency, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University), Shenzhen 518020, China
- Correspondence: ; Tel.: +86-159-1414-1979
| |
Collapse
|
15
|
Single-Cell RNA Sequencing Reveals the Role of Epithelial Cell Marker Genes in Predicting the Prognosis of Colorectal Cancer Patients. DISEASE MARKERS 2022; 2022:8347125. [PMID: 35968507 PMCID: PMC9372514 DOI: 10.1155/2022/8347125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 07/09/2022] [Accepted: 07/21/2022] [Indexed: 11/17/2022]
Abstract
Single-cell RNA sequencing (scRNA-seq) is increasingly used in studies on gastrointestinal cancers. This study investigated the prognostic value of epithelial cell-associated biomarkers in colorectal cancer (CRC) using scRNA-seq data. We downloaded and analysed scRNA-seq data from four CRC samples from the Gene Expression Omnibus (GEO), and we identified marker genes of malignant epithelial cells (MECs) using CRC transcriptome and clinical data downloaded from The Cancer Genome Atlas (TCGA) and GEO as training and validation cohorts, respectively. In the TCGA training cohort, weighted gene correlation network analysis, univariate Cox proportional hazard model (Cox) analysis, and least absolute shrinkage and selection operator regression analysis were performed on the marker genes of MEC subsets to identify a signature of nine prognostic MEC-related genes (MECRGs) and calculate a risk score based on the signature. CRC patients were divided into high- and low-risk groups according to the median risk score. We found that the MECRG risk score was significantly correlated with the clinical features and overall survival of CRC patients, and that CRC patients in the high-risk group showed a significantly shorter survival time. The univariate and multivariate Cox regression analyses showed that the MECRG risk score can serve as an independent prognostic factor for CRC patients. Gene set enrichment analysis revealed that the MECRG signature genes are involved in fatty acid metabolism, p53 signalling, and other pathways. To increase the clinical application value, we constructed a MECRG nomogram by combining the MECRG risk score with other independent prognostic factors. The validity of the nomogram is based on receiver operating characteristics and calibration curves. The MECRG signature and nomogram models were well validated in the GEO dataset. In conclusion, we established an epithelial cell marker gene-based risk assessment model based on scRNA-seq analysis of CRC samples for predicting the prognosis of CRC patients.
Collapse
|
16
|
Hossain KR, Escobar Bermeo JD, Warton K, Valenzuela SM. New Approaches and Biomarker Candidates for the Early Detection of Ovarian Cancer. Front Bioeng Biotechnol 2022; 10:819183. [PMID: 35223789 PMCID: PMC8867026 DOI: 10.3389/fbioe.2022.819183] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Accepted: 01/24/2022] [Indexed: 11/13/2022] Open
Affiliation(s)
- K R Hossain
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia
| | - J D Escobar Bermeo
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia.,ARC Research Hub for Integrated Device for End-user Analysis at Low-levels (IDEAL), Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia
| | - K Warton
- School of Women's and Children's Health, Faculty of Medicine and Health, University of New South Wales, South Wales, NSW, Australia
| | - S M Valenzuela
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia.,ARC Research Hub for Integrated Device for End-user Analysis at Low-levels (IDEAL), Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia
| |
Collapse
|
17
|
Revealing therapeutic targets and mechanism of baicalin for anti-chronic gastritis using proteomic analysis of the gastric tissue. J Chromatogr B Analyt Technol Biomed Life Sci 2022; 1196:123214. [DOI: 10.1016/j.jchromb.2022.123214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 02/19/2022] [Accepted: 03/04/2022] [Indexed: 11/24/2022]
|
18
|
Tian Z, Zhao J, Wang Y. The prognostic value of TPM1-4 in hepatocellular carcinoma. Cancer Med 2021; 11:433-446. [PMID: 34850589 PMCID: PMC8729055 DOI: 10.1002/cam4.4453] [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: 08/31/2021] [Revised: 10/30/2021] [Accepted: 11/03/2021] [Indexed: 12/11/2022] Open
Abstract
Background Despite advances in multiple disciplinary diagnoses and treatments, the prognosis of hepatocellular carcinoma (HCC) remains poor. Some evidence has identified that the aberrant expression of tropomyosins (TPMs) is involved with some cancers development. However, prognostic values of TPMs in HCC have not been thoroughly investigated. Methods Original TPM1–4 mRNA expression of TCGA HCC data and GTEx was downloaded from UCSC XENA. Oncomine database and GSE46408 were used for verification. Clinical stages and survival analysis of TPM1–4 in HCC were performed by GEPIA2. cBioPortal was utilized to assess TPM1–4 gene alteration in HCC. TIMER2.0 was used for investigating the relevance of TPM1–4 to tumor‐infiltrating immune cells in HCC. Additionally, we constructed a TPM1–4 prognostic model to explore the value of TPM1–4 for prognostic evaluation in HCC. LinkedOmics was applied to elucidate TPM3 co‐expression networks in HCC. Results This present study showed that TPM1–4 was upregulated in all HCC tissues, and TPM3 overexpression was correlated with poor survival outcomes in patients with HCC. Besides, TPM3 amplification was the main altered type in TPM1–4 genetic alteration, which affected the prognosis of HCC patients. The risk model revealed that TPM1, TPM2, and TPM3 were applied to risk assessment of HCC prognosis, among which TPM3 expression was significantly higher in the high‐risk group than that in the low‐risk group. Univariate and multivariate cox regression analyses indicated that TPM3 may be an independent prognostic factor of HCC prognosis. In addition, TPM3 co‐expression genes mainly participated in the cell cycle by maintaining microtubule cytoskeleton in HCC progression. TPM1–4 was associated with some tumor‐infiltrating immune cells in HCC. Conclusion Our study detected that the expression level of TPM1–4 was all remarkably elevated in HCC, suggesting that TPM1–4 may serve an important role in HCC development. High TPM3 expression was found to be correlated with poor overall survival, and TPM3 may be an independent prognostic factor for HCC.
Collapse
Affiliation(s)
- Zhihui Tian
- Gastroenterology Ward One, Shanxi Province Cancer Hospital, Taiyuan, Shanxi, China
| | - Jian Zhao
- Gastroenterology Ward One, Shanxi Province Cancer Hospital, Taiyuan, Shanxi, China
| | - Yusheng Wang
- Gastroenterology Ward One, Shanxi Province Cancer Hospital, Taiyuan, Shanxi, China
| |
Collapse
|
19
|
Proteomic Signatures of Diffuse and Intestinal Subtypes of Gastric Cancer. Cancers (Basel) 2021; 13:cancers13235930. [PMID: 34885041 PMCID: PMC8656738 DOI: 10.3390/cancers13235930] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 11/20/2021] [Accepted: 11/23/2021] [Indexed: 12/14/2022] Open
Abstract
Gastric cancer is a leading cause of death from cancer globally. Gastric cancer is classified into intestinal, diffuse and indeterminate subtypes based on histology according to the Laurén classification. The intestinal and diffuse subtypes, although different in histology, demographics and outcomes, are still treated in the same fashion. This study was designed to discover proteomic signatures of diffuse and intestinal subtypes. Mass spectrometry-based proteomics using tandem mass tags (TMT)-based multiplexed analysis was used to identify proteins in tumor tissues from patients with diffuse or intestinal gastric cancer with adjacent normal tissue control. A total of 7448 or 4846 proteins were identified from intestinal or diffuse subtype, respectively. This quantitative mass spectrometric analysis defined a proteomic signature of differential expression across the two subtypes, which included gremlin1 (GREM1), bcl-2-associated athanogene 2 (BAG2), olfactomedin 4 (OLFM4), thyroid hormone receptor interacting protein 6 (TRIP6) and melanoma-associated antigen 9 (MAGE-A9) proteins. Although GREM1, BAG2, OLFM4, TRIP6 and MAGE-A9 have all been previously implicated in tumor progression and metastasis, they have not been linked to intestinal or diffuse subtypes of gastric cancer. Using immunohistochemical labelling of a tissue microarray comprising of 124 cases of gastric cancer, we validated the proteomic signature obtained by mass spectrometry in the discovery cohort. Our findings should help investigate the pathogenesis of these gastric cancer subtypes and potentially lead to strategies for early diagnosis and treatment.
Collapse
|
20
|
Ciregia F, Cetani F, Pardi E, Soggiu A, Piras C, Zallocco L, Borsari S, Ronci M, Caruso V, Marcocci C, Mazzoni MR, Lucacchini A, Giusti L. Parathyroid Carcinoma and Adenoma Co-existing in One Patient: Case Report and Comparative Proteomic Analysis. Cancer Genomics Proteomics 2021; 18:781-796. [PMID: 34697069 DOI: 10.21873/cgp.20297] [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: 04/24/2021] [Revised: 09/02/2021] [Accepted: 09/17/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND/AIM The lack of specific parathyroid carcinoma (PC) biomarkers in clinical practice points out the importance of analyzing the proteomic signature of this cancer. We performed a comparative proteomic analysis of PC and parathyroid adenoma (PA) co-existing in the same patient. PATIENTS AND METHODS PC and PA were taken from a 63-year-old patient. Using two-dimensional differential gel electrophoresis (2D-DIGE) coupled to mass spectrometry we examined the differences between PC and PA proteins. For validation, additional PC and PA samples were obtained from 10 patients. Western blot analysis was used to validate the difference of expression observed with 2D-DIGE analysis. Bioinfomatic analysis was performed using QIAGEN's Ingenuity Pathways Analysis (IPA) to determine the predominant canonical pathways and interaction networks involved. RESULTS Thirty-three differentially expressed proteins were identified in PC compared to PA. Among these, ubiquitin C-terminal hydrolase-L1 (UCH-L1) was highly overexpressed in PC. The result was confirmed by Western Blot analysis in additional PC samples. CONCLUSION Our comparative proteomic analysis of co-existing neoplasms allowed detecting specific and peculiar differences between PC and PA overcoming population biological variability.
Collapse
Affiliation(s)
- Federica Ciregia
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy.,Department of Rheumatology, GIGA Research, Centre Hospitalier Universitaire (CHU) de Liège, University of Liège, Liège, Belgium
| | - Filomena Cetani
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Elena Pardi
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Alessio Soggiu
- Surgical and Dental Sciences-One Health Unit, Department of Biomedical, University of Milano, Milan, Italy
| | - Cristian Piras
- Department of Health Sciences, Campus Universitario "S. Venuta", University "Magna Græcia" of Catanzaro, Catanzaro, Italy
| | | | - Simona Borsari
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Maurizio Ronci
- Department of Pharmacy, University G. d'Annunzio of Chieti-Pescara, Chieti, Italy
| | - Vanni Caruso
- School of Pharmacy & Pharmacology - College of Health and Medicine, University of Tasmania, Hobart, TAS, Australia
| | - Claudio Marcocci
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | | | - Antonio Lucacchini
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Laura Giusti
- School of Pharmacy, University of Camerino, Camerino, Italy
| |
Collapse
|
21
|
Zhou X, Zhu X, Yao J, Wang X, Wang N. Comprehensive analysis of clinical prognosis and molecular immune characterization of tropomyosin 4 in pancreatic cancer. Invest New Drugs 2021; 39:1469-1483. [PMID: 33983530 DOI: 10.1007/s10637-021-01128-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 05/05/2021] [Indexed: 11/26/2022]
Abstract
Pancreatic cancer (PC) is one of the most lethal human solid malignancies with devastating prognosis, making biomarker detection considerably important. Immune infiltrates in microenvironment is associated with patients' survival in PC. The role of Tropomyosin 4 (TPM4) gene in PC has not been reported. Our study first identifies TPM4 expression and its potential biological functions in PC. The potential oncogenic roles of TPM4 was examined using the datasets of TCGA (The cancer genome atlas) and GEO (Gene expression omnibus). We investigated the clinical significance and prognostic value of TPM4 gene based on The Gene Expression Profiling Interactive Analysis (GEPIA) and survival analysis. TIMER and TISIDB databases were used to analyze the correlations between TPM4 gene and tumor-infiltrating immune cells. We found that the expression level of TPM4 was upregulated in PC malignant tissues with the corresponding normal tissues as controls. High TPM4 expression was correlated with the worse clinicopathological features and poor prognosis in PC cohorts. The positive association between TPM4 expression and tumor-infiltrating immune cells was identified in tumor microenvironment (TME). Moreover, functional enrichment analysis suggested that TPM4 might participate in cell adhesion and promote tumor cell migration. This is the first comprehensive study to disclose that TPM4 may serve as a novel prognostic biomarker associating with immune infiltrates and provide a potential therapeutic target for the treatment of PC.
Collapse
Affiliation(s)
- Xue Zhou
- Department of Nephrology, Tianjin Haihe Hospital, Tianjin, 300350, China
| | - Xiaowei Zhu
- Tianjin Third Central Hospital, 83 Jintang Road, Hedong District, Tianjin, 300170, China
| | - Junchao Yao
- Department of Hepatobiliary Surgery, Tianjin Third Central Hospital, Tianjin, 300170, China
| | - Xue Wang
- Department of Respiratory Medicine, Tianjin Third Central Hospital, Tianjin, 300170, China
| | - Ning Wang
- Tianjin Third Central Hospital, 83 Jintang Road, Hedong District, Tianjin, 300170, China.
| |
Collapse
|
22
|
Ryu J, Thomas SN. Quantitative Mass Spectrometry-Based Proteomics for Biomarker Development in Ovarian Cancer. Molecules 2021; 26:molecules26092674. [PMID: 34063568 PMCID: PMC8125593 DOI: 10.3390/molecules26092674] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 04/29/2021] [Accepted: 05/01/2021] [Indexed: 12/11/2022] Open
Abstract
Ovarian cancer is the most lethal gynecologic malignancy among women. Approximately 70–80% of patients with advanced ovarian cancer experience relapse within five years and develop platinum-resistance. The short life expectancy of patients with platinum-resistant or platinum-refractory disease underscores the need to develop new and more effective treatment strategies. Early detection is a critical step in mitigating the risk of disease progression from early to an advanced stage disease, and protein biomarkers have an integral role in this process. The best biological diagnostic tool for ovarian cancer will likely be a combination of biomarkers. Targeted proteomics methods, including mass spectrometry-based approaches, have emerged as robust methods that can address the chasm between initial biomarker discovery and the successful verification and validation of these biomarkers enabling their clinical translation due to the robust sensitivity, specificity, and reproducibility of these versatile methods. In this review, we provide background information on the fundamental principles of biomarkers and the need for improved treatment strategies in ovarian cancer. We also provide insight into the ways in which mass spectrometry-based targeted proteomics approaches can provide greatly needed solutions to many of the challenges related to ovarian cancer biomarker development.
Collapse
|
23
|
Jing J, Du Z, Qin W. Proteome Analysis of Urinary Biomarkers in Acute Hypercoagulable State Rat Model. Front Mol Biosci 2021; 8:634606. [PMID: 33996895 PMCID: PMC8119894 DOI: 10.3389/fmolb.2021.634606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Accepted: 02/18/2021] [Indexed: 11/13/2022] Open
Abstract
Thrombotic diseases are usually preceded by a hypercoagulable state in the body. This study aimed to screen potential urinary biomarkers for hypercoagulable state based on proteome analysis. Wistar rats were administered with the hemostatic agent etamsylate to establish hypercoagulable state. Urine samples were collected for proteome analysis. We found 20 proteins with levels more than 1.5-fold in difference between control rats and model rats. We searched human homologs of 20 rat proteins and identified 13 human proteins. Of the 13 human homologous proteins, nine were members of human core urinary proteome. Human homologous proteins of differential proteins were highly expressed in 31 human tissues, especially in the kidneys followed by digestive system and reproductive system. Surprisingly, we did not identify known coagulation factors as differential proteins in the urine of model rats. Hypercoagulable state of the body may not involve direct changes in coagulation factors but causes the changes upstream of the coagulation cascade system. Common differential urinary proteins between different hypercoagulable states suggest some common pathways in the formation of hypercoagulable states and may serve as potential biomarkers for the prevention and treatment of thrombotic diseases.
Collapse
Affiliation(s)
- Jian Jing
- Beijing Key Lab of Genetic Engineering and Biotechnology, College of Life Sciences, Beijing Normal University, Beijing, China
| | - Zhenhuan Du
- Beijing Key Lab of Genetic Engineering and Biotechnology, College of Life Sciences, Beijing Normal University, Beijing, China
| | - Weiwei Qin
- Beijing Key Lab of Genetic Engineering and Biotechnology, College of Life Sciences, Beijing Normal University, Beijing, China
| |
Collapse
|
24
|
Lee SY, Kwon J, Lee KA. Bcl2l10 induces metabolic alterations in ovarian cancer cells by regulating the TCA cycle enzymes SDHD and IDH1. Oncol Rep 2021; 45:47. [PMID: 33649794 PMCID: PMC7934226 DOI: 10.3892/or.2021.7998] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 02/03/2021] [Indexed: 01/07/2023] Open
Abstract
Bcl2‑like‑10 (Bcl2l10) has both oncogenic and tumor suppressor functions depending on the type of cancer. It has been previously demonstrated that the suppression of Bcl2l10 in ovarian cancer SKOV3 and A2780 cells causes cell cycle arrest and enhances cell proliferation, indicating that Bcl2l10 is a tumor suppressor gene in ovarian cancer cells. The aim of the present study was to identify possible downstream target genes and investigate the underlying mechanisms of action of Bcl2l10 in ovarian cancer cells. RNA sequencing (RNA‑Seq) was performed to obtain a list of differentially expressed genes (DEGs) in Bcl2l10‑suppressed SKOV3 and A2780 cells. The RNA‑Seq data were validated by reverse transcription‑quantitative PCR (RT‑qPCR) and western blot analysis, and the levels of metabolites after Bcl2l10‑knockdown were measured using colorimetric assay kits. Pathway enrichment analysis revealed that the commonly downregulated genes in SKOV3 and A2780 cells after Bcl2l10‑knockdown were significantly enriched in metabolic pathways. The analysis of the DEGs identified from RNA‑Seq and validated by RT‑qPCR revealed that succinate dehydrogenase complex subunit D (SDHD) and isocitrate dehydrogenase 1 (IDH1), which are key enzymes of the TCA cycle that regulate oncometabolite production, may be potential downstream targets of Bcl2l10. Furthermore, Bcl2l10‑knockdown induced the accumulation of succinate and isocitrate through the downregulation of SDHD and IDH1. The present study was the first to elucidate the metabolic regulatory functions of Bcl2l10 in ovarian cancer cells, and the results indicated that Bcl2l10 may serve as a potential therapeutic target in ovarian cancer.
Collapse
Affiliation(s)
- Su-Yeon Lee
- Department of Biomedical Science, College of Life Science, CHA University, Seongnam, Gyeonggi 13488, Republic of Korea
| | - Jinie Kwon
- Department of Biomedical Science, College of Life Science, CHA University, Seongnam, Gyeonggi 13488, Republic of Korea
| | - Kyung-Ah Lee
- Department of Biomedical Science, College of Life Science, CHA University, Seongnam, Gyeonggi 13488, Republic of Korea,Correspondence to: Professor Kyung-Ah Lee, Department of Biomedical Science, College of Life Science, CHA University, 335 Pangyo-ro, Bundang, Seongnam, Gyeonggi 13488, Republic of Korea, E-mail:
| |
Collapse
|
25
|
Altamura C, Greco MR, Carratù MR, Cardone RA, Desaphy JF. Emerging Roles for Ion Channels in Ovarian Cancer: Pathomechanisms and Pharmacological Treatment. Cancers (Basel) 2021; 13:668. [PMID: 33562306 PMCID: PMC7914442 DOI: 10.3390/cancers13040668] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 01/21/2021] [Accepted: 02/04/2021] [Indexed: 12/20/2022] Open
Abstract
Ovarian cancer (OC) is the deadliest gynecologic cancer, due to late diagnosis, development of platinum resistance, and inadequate alternative therapy. It has been demonstrated that membrane ion channels play important roles in cancer processes, including cell proliferation, apoptosis, motility, and invasion. Here, we review the contribution of ion channels in the development and progression of OC, evaluating their potential in clinical management. Increased expression of voltage-gated and epithelial sodium channels has been detected in OC cells and tissues and shown to be involved in cancer proliferation and invasion. Potassium and calcium channels have been found to play a critical role in the control of cell cycle and in the resistance to apoptosis, promoting tumor growth and recurrence. Overexpression of chloride and transient receptor potential channels was found both in vitro and in vivo, supporting their contribution to OC. Furthermore, ion channels have been shown to influence the sensitivity of OC cells to neoplastic drugs, suggesting a critical role in chemotherapy resistance. The study of ion channels expression and function in OC can improve our understanding of pathophysiology and pave the way for identifying ion channels as potential targets for tumor diagnosis and treatment.
Collapse
Affiliation(s)
- Concetta Altamura
- Department of Biomedical Sciences and Human Oncology, School of Medicine, University of Bari Aldo Moro, 70124 Bari, Italy; (M.R.G.); (M.R.C.); (J.-F.D.)
| | - Maria Raffaella Greco
- Department of Biomedical Sciences and Human Oncology, School of Medicine, University of Bari Aldo Moro, 70124 Bari, Italy; (M.R.G.); (M.R.C.); (J.-F.D.)
- Department of Biosciences, Biotechnologies, and Biopharmaceutics, University of Bari Aldo Moro, 70125 Bari, Italy;
| | - Maria Rosaria Carratù
- Department of Biomedical Sciences and Human Oncology, School of Medicine, University of Bari Aldo Moro, 70124 Bari, Italy; (M.R.G.); (M.R.C.); (J.-F.D.)
| | - Rosa Angela Cardone
- Department of Biosciences, Biotechnologies, and Biopharmaceutics, University of Bari Aldo Moro, 70125 Bari, Italy;
| | - Jean-François Desaphy
- Department of Biomedical Sciences and Human Oncology, School of Medicine, University of Bari Aldo Moro, 70124 Bari, Italy; (M.R.G.); (M.R.C.); (J.-F.D.)
| |
Collapse
|
26
|
Luo Y, Yu P, Zhao J, Guo Q, Fan B, Diao Y, Jin Y, Wu J, Zhang C. Inhibitory Effect of Crocin Against Gastric Carcinoma via Regulating TPM4 Gene. Onco Targets Ther 2021; 14:111-122. [PMID: 33442270 PMCID: PMC7800707 DOI: 10.2147/ott.s254167] [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: 03/16/2020] [Accepted: 11/21/2020] [Indexed: 11/23/2022] Open
Abstract
Background Gastric cancer (GC) is one of the most common malignant tumors and the second most frequent cause of cancer death worldwide. Crocin is a kind of bioactive constituent found in the stigmas of saffron, which has shown various pharmacological activities. Methods In this study, we investigated the inhibitory effect of crocin on gastric cancer AGS cells proliferation and explored the underlying mechanism. A series of methods were used including cell counting kit assay, gene microarray analysis, qRT-PCR, Celigo image cytometry, cell clone formation assay, Western blot, and cell xenograft growth in vivo. Results The results indicated that crocin inhibited AGS cells proliferation and promoted cell apoptosis. Further studies suggested that crocin decreased a series of genes expression, among which TPM4 gene downregulation inhibited the tumor cells proliferation and tumor growth in mice, and overexpression of TPM4 gene abolishes the inhibitory effect of crocin. Further study using microarray analysis suggested that knocking down of TPM4 altered genes related to the proliferation and apoptosis of cells. Discussion Crocin could inhibit the gastric cancer cells AGS cells proliferation by regulating TPM4 gene expression, and TPM4 may be a promising therapeutic target for GC treatment.
Collapse
Affiliation(s)
- Yushuang Luo
- Department of Oncology, Affiliated Hospital of Qinghai University, Xining 810001, People's Republic of China
| | - Pengjie Yu
- Department of Oncology, Affiliated Hospital of Qinghai University, Xining 810001, People's Republic of China
| | - Junhui Zhao
- Department of Oncology, Affiliated Hospital of Qinghai University, Xining 810001, People's Republic of China
| | - Qijing Guo
- Department of Oncology, Affiliated Hospital of Qinghai University, Xining 810001, People's Republic of China
| | - Baohua Fan
- Department of Oncology, Affiliated Hospital of Qinghai University, Xining 810001, People's Republic of China
| | - Yinzhuo Diao
- Department of Oncology, Affiliated Hospital of Qinghai University, Xining 810001, People's Republic of China
| | - Yulong Jin
- Department of Oncology, Affiliated Hospital of Qinghai University, Xining 810001, People's Republic of China
| | - Jing Wu
- Department of Oncology, Affiliated Hospital of Qinghai University, Xining 810001, People's Republic of China
| | - Chengwu Zhang
- Department of Oncology, Affiliated Hospital of Qinghai University, Xining 810001, People's Republic of China
| |
Collapse
|
27
|
Li L, Ye T, Zhang Q, Li X, Ma L, Yan J. The expression and clinical significance of TPM4 in hepatocellular carcinoma. Int J Med Sci 2021; 18:169-175. [PMID: 33390785 PMCID: PMC7738955 DOI: 10.7150/ijms.49906] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 10/07/2020] [Indexed: 12/24/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is known as the fifth most common cancer in the world for its poor prognosis. New diagnostic markers and treatments are urgent to discover. To evaluate the protein expression of Tropomyosin4 (TPM4) and investigate its prognostic value in HCC, we collected 110 patients with different degrees of HCC and 10 patients with normal hepatic tissues and performed immunohistochemistry. Western bot was used to evaluate the expression of TPM4 in three HCC cell lines (HepG2, Huh7, SMMC-7721) and normal liver cell line LO2, as well as 7 HCC tissues and 7 normal hepatic tissues. The results of TPM4 staining revealed that TPM4 expression in HCC was higher than that in normal hepatic tissues, which was positive in 51.8% (n=57) and negative in 48.2% (n=53) while in normal hepatic tissues positive staining was in 10% (n=1) and negative staining was in 90% (n=9) (P=0.011). And the expression of TPM4 was related to pT status, grade and stage (P<0.001, P=0.015 and P<0.001, respectively). Western blot results indicated that TPM4 was high expressed in HCC cell line and HCC tissues. In conclusion, we believe that TPM4 can be applied as a diagnostic and prognostic marker to assist the management of HCC.
Collapse
Affiliation(s)
- Linjing Li
- Department of Clinical Laboratory Center, The Second Hospital of Lanzhou University, Lanzhou 730000, P.R. China
| | - Tao Ye
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China, 510515
| | - Qingyan Zhang
- Center of Reproductive Medicine, Department of Obstetrics and Gynecology, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China, 510080
| | - Xin Li
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China, 510515
| | - Li Ma
- Department of Clinical Laboratory Center, The Second Hospital of Lanzhou University, Lanzhou 730000, P.R. China
| | - Jing Yan
- Department of Clinical Laboratory Center, The Second Hospital of Lanzhou University, Lanzhou 730000, P.R. China
| |
Collapse
|
28
|
Challenges and Opportunities in Clinical Applications of Blood-Based Proteomics in Cancer. Cancers (Basel) 2020; 12:cancers12092428. [PMID: 32867043 PMCID: PMC7564506 DOI: 10.3390/cancers12092428] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 08/23/2020] [Accepted: 08/25/2020] [Indexed: 12/12/2022] Open
Abstract
Simple Summary The traditional approach in identifying cancer related protein biomarkers has focused on evaluation of a single peptide/protein in tissue or circulation. At best, this approach has had limited success for clinical applications, since multiple pathological tumor pathways may be involved during initiation or progression of cancer which diminishes the significance of a single candidate protein/peptide. Emerging sensitive proteomic based technologies like liquid chromatography mass spectrometry (LC-MS)-based quantitative proteomics can provide a platform for evaluating serial serum or plasma samples to interrogate secreted products of tumor–host interactions, thereby revealing a more “complete” repertoire of biological variables encompassing heterogeneous tumor biology. However, several challenges need to be met for successful application of serum/plasma based proteomics. These include uniform pre-analyte processing of specimens, sensitive and specific proteomic analytical platforms and adequate attention to study design during discovery phase followed by validation of discovery-level signatures for prognostic, predictive, and diagnostic cancer biomarker applications. Abstract Blood is a readily accessible biofluid containing a plethora of important proteins, nucleic acids, and metabolites that can be used as clinical diagnostic tools in diseases, including cancer. Like the on-going efforts for cancer biomarker discovery using the liquid biopsy detection of circulating cell-free and cell-based tumor nucleic acids, the circulatory proteome has been underexplored for clinical cancer biomarker applications. A comprehensive proteome analysis of human serum/plasma with high-quality data and compelling interpretation can potentially provide opportunities for understanding disease mechanisms, although several challenges will have to be met. Serum/plasma proteome biomarkers are present in very low abundance, and there is high complexity involved due to the heterogeneity of cancers, for which there is a compelling need to develop sensitive and specific proteomic technologies and analytical platforms. To date, liquid chromatography mass spectrometry (LC-MS)-based quantitative proteomics has been a dominant analytical workflow to discover new potential cancer biomarkers in serum/plasma. This review will summarize the opportunities of serum proteomics for clinical applications; the challenges in the discovery of novel biomarkers in serum/plasma; and current proteomic strategies in cancer research for the application of serum/plasma proteomics for clinical prognostic, predictive, and diagnostic applications, as well as for monitoring minimal residual disease after treatments. We will highlight some of the recent advances in MS-based proteomics technologies with appropriate sample collection, processing uniformity, study design, and data analysis, focusing on how these integrated workflows can identify novel potential cancer biomarkers for clinical applications.
Collapse
|
29
|
Carofino BL, Dinshaw KM, Ho PY, Cataisson C, Michalowski AM, Ryscavage A, Alkhas A, Wong NW, Koparde V, Yuspa SH. Head and neck squamous cancer progression is marked by CLIC4 attenuation in tumor epithelium and reciprocal stromal upregulation of miR-142-3p, a novel post-transcriptional regulator of CLIC4. Oncotarget 2019; 10:7251-7275. [PMID: 31921386 PMCID: PMC6944452 DOI: 10.18632/oncotarget.27387] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 12/02/2019] [Indexed: 02/06/2023] Open
Abstract
Chloride intracellular channel 4 (CLIC4) is a tumor suppressor implicated in processes including growth arrest, differentiation, and apoptosis. CLIC4 protein expression is diminished in the tumor parenchyma during progression in squamous cell carcinoma (SCC) and other neoplasms, but the underlying mechanisms have not been identified. Data from The Cancer Genome Atlas suggest this is not driven by genomic alterations. However, screening and functional assays identified miR-142-3p as a regulator of CLIC4. CLIC4 and miR-142-3p expression are inversely correlated in head and neck (HN) SCC and cervical SCC, particularly in advanced stage cancers. In situ localization revealed that stromal immune cells, not tumor cells, are the predominant source of miR-142-3p in HNSCC. Furthermore, HNSCC single-cell expression data demonstrated that CLIC4 is lower in tumor epithelial cells than in stromal fibroblasts and endothelial cells. Tumor-specific downregulation of CLIC4 was confirmed in an SCC xenograft model concurrent with immune cell infiltration and miR-142-3p upregulation. These findings provide the first evidence of CLIC4 regulation by miRNA. Furthermore, the distinct localization of CLIC4 and miR-142-3p within the HNSCC tumor milieu highlight the limitations of bulk tumor analysis and provide critical considerations for both future mechanistic studies and use of miR-142-3p as a HNSCC biomarker.
Collapse
Affiliation(s)
- Brandi L. Carofino
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Kayla M. Dinshaw
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
- Department of Molecular and Cellular Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Pui Yan Ho
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
- Department of Pediatrics, Division of Stem Cell Transplantation and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Christophe Cataisson
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Aleksandra M. Michalowski
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Andrew Ryscavage
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | | | - Nathan W. Wong
- CCR Collaborative Bioinformatics Resource (CCBR), Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
- Advanced Biomedical Computational Science, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Vishal Koparde
- CCR Collaborative Bioinformatics Resource (CCBR), Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
- Advanced Biomedical Computational Science, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Stuart H. Yuspa
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| |
Collapse
|
30
|
Thorsen SF, Gromova I, Christensen IJ, Fredriksson S, Andersen CL, Nielsen HJ, Stenvang J, Moreira JM. Gel-Based Proteomics of Clinical Samples Identifies Potential Serological Biomarkers for Early Detection of Colorectal Cancer. Int J Mol Sci 2019; 20:ijms20236082. [PMID: 31810358 PMCID: PMC6929140 DOI: 10.3390/ijms20236082] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 11/18/2019] [Accepted: 11/18/2019] [Indexed: 12/15/2022] Open
Abstract
The burden of colorectal cancer (CRC) is considerable-approximately 1.8 million people are diagnosed each year with CRC and of these about half will succumb to the disease. In the case of CRC, there is strong evidence that an early diagnosis leads to a better prognosis, with metastatic CRC having a 5-year survival that is only slightly greater than 10% compared with up to 90% for stage I CRC. Clearly, biomarkers for the early detection of CRC would have a major clinical impact. We implemented a coherent gel-based proteomics biomarker discovery platform for the identification of clinically useful biomarkers for the early detection of CRC. Potential protein biomarkers were identified by a 2D gel-based analysis of a cohort composed of 128 CRC and site-matched normal tissue biopsies. Potential biomarkers were prioritized and assays to quantitatively measure plasma expression of the candidate biomarkers were developed. Those biomarkers that fulfilled the preset criteria for technical validity were validated in a case-control set of plasma samples, including 70 patients with CRC, adenomas, or non-cancer diseases and healthy individuals in each group. We identified 63 consistently upregulated polypeptides (factor of four-fold or more) in our proteomics analysis. We selected 10 out of these 63 upregulated polypeptides, and established assays to measure the concentration of each one of the ten biomarkers in plasma samples. Biomarker levels were analyzed in plasma samples from healthy individuals, individuals with adenomas, CRC patients, and patients with non-cancer diseases and we identified one protein, tropomyosin 3 (Tpm3) that could discriminate CRC at a significant level (p = 0.0146). Our results suggest that at least one of the identified proteins, Tpm3, could be used as a biomarker in the early detection of CRC, and further studies should provide unequivocal evidence for the real-life clinical validity and usefulness of Tpm3.
Collapse
Affiliation(s)
- Stine F. Thorsen
- Institute of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, 2100 Copenhagen, Denmark;
| | - Irina Gromova
- Danish Cancer Society Research Center, 2100 Copenhagen, Denmark;
| | - Ib J. Christensen
- Department of Surgical Gastroenterology, Hvidovre Hospital, University of Copenhagen, 2650 Hvidovre, Denmark; (I.J.C.); (H.J.N.)
| | | | - Claus L. Andersen
- Department of Molecular Medicine, Aarhus University Hospital, 8200 Aarhus, Denmark;
| | - Hans J. Nielsen
- Department of Surgical Gastroenterology, Hvidovre Hospital, University of Copenhagen, 2650 Hvidovre, Denmark; (I.J.C.); (H.J.N.)
| | - Jan Stenvang
- Institute of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, 2100 Copenhagen, Denmark;
- Correspondence: (J.S.); (J.M.A.M.)
| | - José M.A. Moreira
- Institute of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, 2100 Copenhagen, Denmark;
- Correspondence: (J.S.); (J.M.A.M.)
| |
Collapse
|
31
|
Wang CCN, Li CY, Cai JH, Sheu PCY, Tsai JJP, Wu MY, Li CJ, Hou MF. Identification of Prognostic Candidate Genes in Breast Cancer by Integrated Bioinformatic Analysis. J Clin Med 2019; 8:1160. [PMID: 31382519 PMCID: PMC6723760 DOI: 10.3390/jcm8081160] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 07/31/2019] [Accepted: 07/31/2019] [Indexed: 12/24/2022] Open
Abstract
Breast cancer is one of the most common malignancies. However, the molecular mechanisms underlying its pathogenesis remain to be elucidated. The present study aimed to identify the potential prognostic marker genes associated with the progression of breast cancer. Weighted gene coexpression network analysis was used to construct free-scale gene coexpression networks, evaluate the associations between the gene sets and clinical features, and identify candidate biomarkers. The gene expression profiles of GSE48213 were selected from the Gene Expression Omnibus database. RNA-seq data and clinical information on breast cancer from The Cancer Genome Atlas were used for validation. Four modules were identified from the gene coexpression network, one of which was found to be significantly associated with patient survival time. The expression status of 28 genes formed the black module (basal); 18 genes, dark red module (claudin-low); nine genes, brown module (luminal), and seven genes, midnight blue module (nonmalignant). These modules were clustered into two groups according to significant difference in survival time between the groups. Therefore, based on betweenness centrality, we identified TXN and ANXA2 in the nonmalignant module, TPM4 and LOXL2 in the luminal module, TPRN and ADCY6 in the claudin-low module, and TUBA1C and CMIP in the basal module as the genes with the highest betweenness, suggesting that they play a central role in information transfer in the network. In the present study, eight candidate biomarkers were identified for further basic and advanced understanding of the molecular pathogenesis of breast cancer by using co-expression network analysis.
Collapse
Affiliation(s)
- Charles C N Wang
- Department of Bioinformatics and Medical Engineering, Asia University, Taichung 413, Taiwan
| | - Chia Ying Li
- Department of Surgery, Show Chwan Memorial Hospital, Changhua 500, Taiwan
- Graduate Institute of Biomedical Engineering, National Chung Hsing University, Taichung 402, Taiwan
| | - Jia-Hua Cai
- Department of Bioinformatics and Medical Engineering, Asia University, Taichung 413, Taiwan
| | - Phillip C-Y Sheu
- Department of EECS and BME, University of California, Irvine, CA 92697, USA
| | - Jeffrey J P Tsai
- Department of Bioinformatics and Medical Engineering, Asia University, Taichung 413, Taiwan
| | - Meng-Yu Wu
- Department of Emergency Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei 231, Taiwan
- Department of Emergency Medicine, School of Medicine, Tzu Chi University, Hualien 970, Taiwan
| | - Chia-Jung Li
- Department of Obstetrics and Gynecology, Kaohsiung Veterans General Hospital, Kaohsiung 813, Taiwan.
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung 804, Taiwan.
| | - Ming-Feng Hou
- Division of Breast Surgery, Department of Surgery, Center for Cancer Research,Kaohsiung Medical University Chung-Ho Memorial Hospital, Kaohsiung 807, Taiwan.
- Graduate Institute of Clinical Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- National Sun Yat-Sen University-Kaohsiung Medical University Joint Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- National Chiao Tung University-Kaohsiung Medical University Joint Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| |
Collapse
|
32
|
Zhou S, Ma X, Wang ZJ, Zhang WY, Jiang H, Li SD, Zhang TZ, Du J, Lu Z. Research on the establishment of a TPM3 monoclonal stable transfected PANC-1 cell line and the experiment of the EMT occurrence in human pancreatic cancer. Onco Targets Ther 2019; 12:5577-5587. [PMID: 31371995 PMCID: PMC6628969 DOI: 10.2147/ott.s212689] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 06/21/2019] [Indexed: 01/16/2023] Open
Abstract
Background: Pancreatic cancer is one of the most aggressive human malignancies that is associated with early metastasis and chemoresistance. Tropomyosin (TPM) is an indispensable regulatory protein for muscle contraction, Abnormal expressions of TPM gene are closely related to the carcinogenesis and metastasis of malignant tumors. Purpose: In this experiment, a monoclonal stable transfected cell line was established by the knock-down of TMP3 expression in PANC-1 cells with the lentivirus method, and the impacts of the downregulated TPM3 gene expression on the EMT-related molecules and biological behaviors of PANC-1 cells were explored. Methods: Based on the TPM3 gene sequence, we designed the RNA interference sequence, constructed and screened out the recombinant plasmid segment TPM3-shRNA with the optimal silencing effect, and carried out lentivirus titer determination and packaging. The recombinant lentiviral interference vector LV-TPM3-shRNA was transfected into PANC-1 cells; the transfection efficiency was then evaluated to screen out the monoclonal stable transfected PANC-1 cell line with downregulated TPM3 expression. The qRT-PCR and Western blot were used to detect the changes in the gene- and protein-levels expressions of EMT-related transcription factors in the target cell line and to respectively test the variations of the invasion and proliferation capacities. Results: It is shown that the monoclonal stable transfected PANC-1 cell line with downregulated TPM3 expression was successfully established with the recombinant lentiviral vector. After knocking down the expression of TPM3 gene in PANC-1 cells, EMT occurred in the cells; the cell phenotype showed malignant transformation, and the in vitro biological behaviors of the cells (such as proliferation and invasion) were enhanced to different degrees. Conclusion: It is indicated that the TPM3 gene can be a potential target spot for the treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Shuo Zhou
- Department of Emergency Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu 233000, Anhui, People's Republic of China
| | - Xiang Ma
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu 233000, Anhui, People's Republic of China
| | - Zhen-Jie Wang
- Department of Emergency Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu 233000, Anhui, People's Republic of China
| | - Wei-Yue Zhang
- Department of Emergency Medicine, The Second People's Hospital of Bengbu City, Bengbu 233000, Anhui, People's Republic of China
| | - Hai Jiang
- Department of Emergency Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu 233000, Anhui, People's Republic of China
| | - San-Dang Li
- Department of Emergency Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu 233000, Anhui, People's Republic of China
| | - Tai-Zhe Zhang
- Department of Emergency Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu 233000, Anhui, People's Republic of China
| | - Jie Du
- Department of Emergency Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu 233000, Anhui, People's Republic of China
| | - Zheng Lu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu 233000, Anhui, People's Republic of China
| |
Collapse
|
33
|
Zhao X, Jiang M, Wang Z. TPM4 promotes cell migration by modulating F-actin formation in lung cancer. Onco Targets Ther 2019; 12:4055-4063. [PMID: 31239699 PMCID: PMC6554522 DOI: 10.2147/ott.s198542] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Accepted: 04/08/2019] [Indexed: 12/29/2022] Open
Abstract
Background: Tropomyosin 4 (TPM4) is a member of the tropomyosin family of actin-binding proteins. Abnormal level of TPM4 is found in several cancers, and TPM4 is considered as a potential detecting marker for ovarian cancer, breast cancer, colon cancer, keratoacanthoma and esophageal squamous cell carcinoma. In this paper, the function of TPM4 in lung cancer cell lines was determined. Materials and methods: TPM4 knockout cells were constructed by CRISPR/CAS9 technique. TPM4 overexpression cells were also constructed based on TPM4 knockout cells. Cell growth ability was detected by MTS assay. The potency of cell motility was investigated using transwell assay and wound scratch assay. The protein levels in lung cancer cells were determined by western-blot. Immunofluorescence technique was used to image the structure of F-actin. Results: As a result, TPM4 downregulation and TPM4 upregulation cell models were obtained successfully. Cell motility was inhibited by the suppression of TPM4 while cell migration was enhanced in TPM4 upregulated cells. But TPM4 was not involved in cell proliferation and EMT progression. Microfilaments were depolymerized result from the suppression of TPM4 expression. And F-actin assembly was increased when TPM4 was upregulated. Conclusion: In summary, TPM4 was able to promote cell motility by altering the actin cytoskeleton directly.
Collapse
Affiliation(s)
- Xiaoting Zhao
- Department of Cellular and Molecular Biology, Beijing Chest Hospital, Capital Medical University /Beijing Tuberculosis and Thoracic Tumor Research Institute, Tongzhou, Beijing, People's Republic of China
| | - Mei Jiang
- Department of Cellular and Molecular Biology, Beijing Chest Hospital, Capital Medical University /Beijing Tuberculosis and Thoracic Tumor Research Institute, Tongzhou, Beijing, People's Republic of China
| | - Ziyu Wang
- Department of Cellular and Molecular Biology, Beijing Chest Hospital, Capital Medical University /Beijing Tuberculosis and Thoracic Tumor Research Institute, Tongzhou, Beijing, People's Republic of China
| |
Collapse
|
34
|
Bjerre MT, Strand SH, Nørgaard M, Kristensen H, Rasmussen AK, Mortensen MM, Fredsøe J, Mouritzen P, Ulhøi B, Ørntoft T, Borre M, Sørensen KD. Aberrant DOCK2, GRASP, HIF3A and PKFP Hypermethylation has Potential as a Prognostic Biomarker for Prostate Cancer. Int J Mol Sci 2019; 20:ijms20051173. [PMID: 30866497 PMCID: PMC6429171 DOI: 10.3390/ijms20051173] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 02/12/2019] [Accepted: 02/28/2019] [Indexed: 12/18/2022] Open
Abstract
Prostate cancer (PCa) is a clinically heterogeneous disease and currently, accurate diagnostic and prognostic molecular biomarkers are lacking. This study aimed to identify novel DNA hypermethylation markers for PCa with future potential for blood-based testing. Accordingly, to search for genes specifically hypermethylated in PCa tissue samples and not in blood cells or other cancer tissue types, we performed a systematic analysis of genome-wide DNA methylation data (Infinium 450K array) available in the Marmal-aid database for 4072 malignant/normal tissue samples of various types. We identified eight top candidate markers (cg12799885, DOCK2, FBXO30, GRASP, HIF3A, MOB3B, PFKP, and TPM4) that were specifically hypermethylated in PCa tissue samples and hypomethylated in other benign and malignant tissue types, including in peripheral blood cells. Potential as diagnostic and prognostic biomarkers was further assessed by the quantitative methylation specific PCR (qMSP) analysis of 37 nonmalignant and 197 PCa tissue samples from an independent population. Here, all eight hypermethylated candidates showed high sensitivity (75–94%) and specificity (84–100%) for PCa. Furthermore, DOCK2, GRASP, HIF3A and PKFP hypermethylation was significantly associated with biochemical recurrence (BCR) after radical prostatectomy (RP; 197 patients), independent of the routine clinicopathological variables. DOCK2 is the most promising single candidate marker (hazard ratio (HR) (95% confidence interval (CI)): 1.96 (1.24–3.10), adjusted p = 0.016; multivariate cox regression). Further validation studies are warranted and should investigate the potential value of these hypermethylation candidate markers for blood-based testing also.
Collapse
Affiliation(s)
- Marianne T Bjerre
- Department of Molecular Medicine (MOMA), Aarhus University Hospital (AUH), Palle Juul-Jensens Boulevard 99, 8200 Aarhus N, Denmark.
- Department of Urology, Aarhus University Hospital (AUH), Palle Juul-Jensens Boulevard 99, 8200 Aarhus N, Denmark.
| | - Siri H Strand
- Department of Molecular Medicine (MOMA), Aarhus University Hospital (AUH), Palle Juul-Jensens Boulevard 99, 8200 Aarhus N, Denmark.
| | - Maibritt Nørgaard
- Department of Molecular Medicine (MOMA), Aarhus University Hospital (AUH), Palle Juul-Jensens Boulevard 99, 8200 Aarhus N, Denmark.
| | | | | | - Martin Mørck Mortensen
- Department of Urology, Aarhus University Hospital (AUH), Palle Juul-Jensens Boulevard 99, 8200 Aarhus N, Denmark.
| | - Jacob Fredsøe
- Department of Molecular Medicine (MOMA), Aarhus University Hospital (AUH), Palle Juul-Jensens Boulevard 99, 8200 Aarhus N, Denmark.
| | - Peter Mouritzen
- Exiqon ⁻ a Qiagen company, Skelstedet 16, 2950 Vedbæk, Denmark.
| | - Benedicte Ulhøi
- Department of Pathology, Aarhus University Hospital (AUH), Palle Juul-Jensens Boulevard 99, 8200 Aarhus N, Denmark.
| | - Torben Ørntoft
- Department of Molecular Medicine (MOMA), Aarhus University Hospital (AUH), Palle Juul-Jensens Boulevard 99, 8200 Aarhus N, Denmark.
| | - Michael Borre
- Department of Urology, Aarhus University Hospital (AUH), Palle Juul-Jensens Boulevard 99, 8200 Aarhus N, Denmark.
| | - Karina D Sørensen
- Department of Molecular Medicine (MOMA), Aarhus University Hospital (AUH), Palle Juul-Jensens Boulevard 99, 8200 Aarhus N, Denmark.
| |
Collapse
|
35
|
Proteomic profile of histotroph during early embryo development in mares. Theriogenology 2019; 125:224-235. [DOI: 10.1016/j.theriogenology.2018.11.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 10/31/2018] [Accepted: 11/04/2018] [Indexed: 01/16/2023]
|
36
|
Xu Y, Xu J, Feng J, Li J, Jiang C, Li X, Zou S, Wang Q, Li Y. Expression of CLIC1 as a potential biomarker for oral squamous cell carcinoma: a preliminary study. Onco Targets Ther 2018; 11:8073-8081. [PMID: 30519049 PMCID: PMC6239106 DOI: 10.2147/ott.s181936] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Purpose CLIC1, a member of the highly conserved class ion-channel protein family, is frequently upregulated in multiple human malignancies and has been demonstrated to play a critical role in cell proliferation, apoptosis, and invasion. However, limited is known about its expression, biological functions, and action mechanism in oral malignancies. We aimed to evaluate whether CLIC1 could be a biomarker for oral squamous cell carcinoma (OSCC). Methods Immunohistochemistry was used to analyze the expression of CLIC1 in tissue. CLIC1 protein and mRNA were measured through Western immunoblotting and quantitative real-time PCR. CLIC1 protein expression in plasma was detected via ELISA. A total of 72 OSCC specimens were recruited in this study for evaluation of correlations of CLIC1 with clinicopathological features and survival. Results CLIC1 was significantly overexpressed in tissue and plasma of OSCC patients. It was found that upregulated CLIC1 was distinctly correlated with histological grade, TNM stage, and tumor size. Meanwhile, Kaplan–Meier survival analysis showed that OSCC patients with high CLIC1 expression had remarkably poorer overall survival rate than those with low CLIC1 expression. Multivariate Cox regression analysis revealed that CLIC1 was the independent prognostic factor for overall survival rate of OSCC patients. In addition, Pearson correlation analysis showed that CLIC1 was associated with multiple tumor-associated genes. Conclusion These results indicated that CLIC1 acts as a molecular target in OSCC and may present a novel diagnostic marker and therapeutic target for OSCC.
Collapse
Affiliation(s)
- Ying Xu
- College of Stomatology, Chongqing Medical University, Chongqing, China, .,Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing, China, .,Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China,
| | - Jie Xu
- College of Stomatology, Chongqing Medical University, Chongqing, China, .,Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing, China, .,Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China,
| | - Jiali Feng
- College of Stomatology, Chongqing Medical University, Chongqing, China, .,Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing, China, .,Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China,
| | - Jie Li
- College of Stomatology, Chongqing Medical University, Chongqing, China, .,Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing, China, .,Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China,
| | - Chao Jiang
- College of Stomatology, Chongqing Medical University, Chongqing, China, .,Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing, China, .,Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China,
| | - Xian Li
- College of Stomatology, Chongqing Medical University, Chongqing, China, .,Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing, China, .,Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China,
| | - Sihai Zou
- College of Stomatology, Chongqing Medical University, Chongqing, China, .,Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing, China, .,Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China,
| | - Qian Wang
- College of Stomatology, Chongqing Medical University, Chongqing, China, .,Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing, China, .,Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China,
| | - Yong Li
- College of Stomatology, Chongqing Medical University, Chongqing, China, .,Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing, China, .,Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China,
| |
Collapse
|
37
|
CLIC1 and CLIC4 complement CA125 as a diagnostic biomarker panel for all subtypes of epithelial ovarian cancer. Sci Rep 2018; 8:14725. [PMID: 30282979 PMCID: PMC6170428 DOI: 10.1038/s41598-018-32885-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 09/18/2018] [Indexed: 01/20/2023] Open
Abstract
New plasma and tissue biomarkers of epithelial ovarian cancer (EOC) could improve early diagnosis and post-diagnosis clinical management. Here we investigated tissue staining and tissue secretion of CLIC1 and CLIC4 across EOC subtypes. CLIC1 and CLIC4 are two promising biomarkers we previously showed were elevated in EOC patient sera. Individually, CLIC1 or CLIC4 stained larger percentages of malignant tumors across all EOC subtypes compared with CA125, particularly early stage and mucinous tumors. CLIC4 also stained benign tumors but staining was limited to nuclei; whereas malignant tumors showed diffuse cellular staining of stromal and tumor cells. Both proteins were shed by all EOC subtypes tumors in short term organ culture at more consistent levels than CA125, supporting their potential as pan-subtype serum and tissue biomarkers. Elevated CLIC4 expression, but not CLIC1 expression, was a negative indicator of patient survival, and CLIC4 knockdown in cultured cells decreased cell proliferation and migration indicating a potential role in tumor progression. These results suggest CLIC1 and CLIC4 are promising serum and tissue biomarkers as well as potential therapeutic targets for all EOC subtypes. This justifies development of high throughput serum/plasma biomarker assays to evaluate utility of a biomarker panel consisting of CLIC1, CLIC4 and CA125.
Collapse
|
38
|
Zare M, Hadi F, Alivand MR. Considering the downregulation of Tpm1.6 and Tpm1.7 in squamous cell carcinoma of esophagus as a potent biomarker. Per Med 2018; 15:361-370. [PMID: 30259780 DOI: 10.2217/pme-2018-0015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
AIM Squamous cell carcinoma of esophagus (SCCE) is an aggressive disease with a poor prognosis. Tropomyosins attach to actin microfilaments, providing its stability. Nonmuscle cells express Tpm isoforms such as Tpm1.6 and Tpm1.7 which are involved in cytoskeleton functional properties regulation. MATERIALS & METHODS The expression of Tpm1.6 and Tpm1.7 was analyzed in SCCE tissues and its association with clinicopathological parameters and survival of patients was assessed. RESULTS Tpm1.6 and Tpm1.7, besides TPM1 mRNA decreased considerably in SCCE tissues relative to normal esophageal tissues (p < 0.001). TPM1 downregulation level was significantly associated with the degree of tumor differentiation (p = 0.017). CONCLUSION Tpm1.6 and Tpm1.7 suppression play a crucial role in esophagus tumorigenesis and could be associated with SCCE poor prognosis.
Collapse
Affiliation(s)
- Maryam Zare
- Department of Biology, Faculty of Sciences, Payame Noor University, Tehran, Iran
| | - Faranak Hadi
- Department of Biology, Faculty of Sciences, Lorestan University, Khorramabad, Iran
| | - Mohammad Reza Alivand
- Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
39
|
The clinical significance and biological function of tropomyosin 4 in colon cancer. Biomed Pharmacother 2018; 101:1-7. [PMID: 29455030 DOI: 10.1016/j.biopha.2018.01.166] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2018] [Revised: 01/31/2018] [Accepted: 01/31/2018] [Indexed: 01/06/2023] Open
Abstract
Tropomyosin 4 (TPM4) has been found to be dys-regulated, and function as oncogene or anti-oncogene in human cancers. However, there was no report on the clinical significance and biological function of TPM4 in colon cancer. This study was designed to investigate the clinical value and biological function of TPM4 in colon cancer. Thus, we detected the TPM4 expression in colon cancer clinical samples, and conducted the gain-of-function in colon cancer cell lines. In our results, TPM4 mRNA and protein expressions were reduced in colon cancer tissues and cell lines compared with normal colon tissues and colon epithelial cell line, respectively. TPM4 protein low-expression was obviously associated with clinical stage, T classification (invasion depth), N classification (lymph node metastasis), distant metastasis and differentiation. Survival analysis showed low-expression of TPM4 was an unfavorable independent prognostic factor for colon cancer patients. Moreover, the experiments in vitro suggested up-regulated TPM4 expression suppressed colon cancer cell migration, invasion and metastasis-associated gene expression including MMP-2, MMP-9 and MT1-MMP, but had no effect on cell proliferation. In conclusion, TPM4 is associated with clinical progression in colon cancer patient and acts as a tumor suppressor in colon cancer cell.
Collapse
|
40
|
Jiang W, Liu L, Chen Y. Simultaneous Detection of Human C-Terminal p53 Isoforms by Single Template Molecularly Imprinted Polymers (MIPs) Coupled with Liquid Chromatography-Tandem Mass Spectrometry (LC-MS/MS)-Based Targeted Proteomics. Anal Chem 2018; 90:3058-3066. [DOI: 10.1021/acs.analchem.7b02890] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Wenting Jiang
- School of Pharmacy, Nanjing Medical University, 818 Tian Yuan East Road, Nanjing, Jiangsu, China, 211166
| | - Liang Liu
- School of Pharmacy, Nanjing Medical University, 818 Tian Yuan East Road, Nanjing, Jiangsu, China, 211166
| | - Yun Chen
- School of Pharmacy, Nanjing Medical University, 818 Tian Yuan East Road, Nanjing, Jiangsu, China, 211166
- China State Key Laboratory of Reproductive Medicine, Nanjing, China 210029
| |
Collapse
|
41
|
Alečković M, Wei Y, LeRoy G, Sidoli S, Liu DD, Garcia BA, Kang Y. Identification of Nidogen 1 as a lung metastasis protein through secretome analysis. Genes Dev 2017; 31:1439-1455. [PMID: 28827399 PMCID: PMC5588926 DOI: 10.1101/gad.301937.117] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 07/31/2017] [Indexed: 11/24/2022]
Abstract
Secreted proteins play crucial roles in mediating tumor-stroma interactions during metastasis of cancer to different target organs. To comprehensively profile secreted proteins involved in lung metastasis, we applied quantitative mass spectrometry-based proteomics and identified 392 breast cancer-derived and 302 melanoma-derived proteins secreted from highly lung metastatic cells. The cancer-specific lung metastasis secretome signatures (LMSSs) displayed significant prognostic value in multiple cancer clinical data sets. Moreover, we observed a significant overlap of enriched pathways between the LMSSs of breast cancer and melanoma despite an overall small overlap of specific proteins, suggesting that common biological processes are executed by different proteins to enable the two cancer types to metastasize to the lung. Among the novel candidate lung metastasis proteins, Nidogen 1 (NID1) was confirmed to promote lung metastasis of breast cancer and melanoma, and its expression is correlated with poor clinical outcomes. In vitro functional analysis further revealed multiple prometastatic functions of NID1, including enhancing cancer cell migration and invasion, promoting adhesion to the endothelium and disrupting its integrity, and improving vascular tube formation capacity. As a secreted prometastatic protein, NID1 may be developed as a new biomarker for disease progression and therapeutic target in breast cancer and melanoma.
Collapse
Affiliation(s)
- Maša Alečković
- Department of Molecular Biology, Princeton University, Princeton, New Jersey 08544, USA
| | - Yong Wei
- Department of Molecular Biology, Princeton University, Princeton, New Jersey 08544, USA
| | - Gary LeRoy
- Department of Molecular Biology, Princeton University, Princeton, New Jersey 08544, USA
| | - Simone Sidoli
- Epigenetics Program, Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Daniel D Liu
- Department of Molecular Biology, Princeton University, Princeton, New Jersey 08544, USA
| | - Benjamin A Garcia
- Epigenetics Program, Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Yibin Kang
- Department of Molecular Biology, Princeton University, Princeton, New Jersey 08544, USA
| |
Collapse
|
42
|
Liu XX, Ye H, Wang P, Li LX, Zhang Y, Zhang JY. Proteomic-based identification of HSP70 as a tumor-associated antigen in ovarian cancer. Oncol Rep 2017; 37:2771-2778. [PMID: 28339059 DOI: 10.3892/or.2017.5525] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Accepted: 11/21/2016] [Indexed: 11/06/2022] Open
Abstract
Ovarian cancer commonly presents without prominent symptoms and is consequently diagnosed at advanced stages with unfavorable prognosis. Novel serological biomarkers for the early detection and clinical management of ovarian cancer are imminently needed. Proteomic-based methods for biomarker discovery are promising strategies implemented in cancer research. The aim of the present study was to identify new tumor antigens from the ovarian cancer cell line SKOV3 and their associated autoantibodies in sera of patients with ovarian cancer employing proteomic-based approaches. Proteins from the ovarian cancer cell line SKOV3 were extracted by two‑dimensional polyacrylamide gel electrophoresis (2-DE) followed by western blotting and antibody reaction with sera from patients with ovarian cancer and normal controls. Positive spots were excised from Coomassie blue‑stained gels and identified by liquid chromatography‑tandem mass spectrometry (LC-MS/MS). The 2-DE analysis results revealed a total of 14 protein spots on the gel, and 7 proteins were finally identified by LC-MS/MS. In the subsequent experiment, using immunoassay on ovarian cancer sera and tissue-array slides, the well-known protein HSP70 was selected in order to validate this proteomic-based approach. In conclusion, the proteomic method used in the present study is a powerful instrument for identifying novel serum markers that may exhibit clinical usefulness in cancer.
Collapse
Affiliation(s)
- Xin-Xin Liu
- Center for Tumor Biotherapy, The First Affiliated Hospital and College of Public Health, Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Hua Ye
- Center for Tumor Biotherapy, The First Affiliated Hospital and College of Public Health, Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Peng Wang
- Center for Tumor Biotherapy, The First Affiliated Hospital and College of Public Health, Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Liu-Xia Li
- Center for Tumor Biotherapy, The First Affiliated Hospital and College of Public Health, Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Yi Zhang
- Center for Tumor Biotherapy, The First Affiliated Hospital and College of Public Health, Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Jian-Ying Zhang
- Center for Tumor Biotherapy, The First Affiliated Hospital and College of Public Health, Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| |
Collapse
|
43
|
Ulmasov B, Bruno J, Oshima K, Cheng YW, Holly SP, Parise LV, Egan TM, Edwards JC. CLIC1 null mice demonstrate a role for CLIC1 in macrophage superoxide production and tissue injury. Physiol Rep 2017; 5:e13169. [PMID: 28275112 PMCID: PMC5350177 DOI: 10.14814/phy2.13169] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Revised: 01/16/2017] [Accepted: 01/17/2017] [Indexed: 12/23/2022] Open
Abstract
We generated and studied CLIC1 null (C1KO) mice to investigate the physiological role of this protein. C1KO and matched wild-type (WT) mice were studied in two models of acute toxic tissue injury. CLIC1 expression is upregulated following acute injury of WT kidney and pancreas and is absent in C1KOs. Acute tissue injury is attenuated in the C1KOs and this correlates with an absence of the rise in tissue reactive oxygen species (ROS) that is seen in WT mice. Infiltration of injured tissue by inflammatory cells was comparable between WT and C1KOs. Absence of CLIC1 increased PMA-induced superoxide production by isolated peritoneal neutrophils but dramatically decreased PMA-induced superoxide production by peritoneal macrophages. CLIC1 is expressed in both neutrophils and macrophages in a peripheral pattern consistent with either plasma membrane or the cortical cytoskeleton in resting cells and redistributes away from the periphery following PMA stimulation in both cell types. Absence of CLIC1 had no effect on redistribution or dephosphorylation of Ezrin/ERM cytoskeleton in macrophages. Plasma membrane chloride conductance is altered in the absence of CLIC1, but not in a way that would be expected to block superoxide production. NADPH oxidase redistributes from an intracellular compartment to the plasma membrane when WT macrophages are stimulated to produce superoxide and this redistribution fails to occur in C1KO macrophages. We conclude that the role of CLIC1 in macrophage superoxide production is to support redistribution of NADPH oxidase to the plasma membrane, and not through major effects on ERM cytoskeleton or by acting as a plasma membrane chloride channel.
Collapse
Affiliation(s)
- Barbara Ulmasov
- Department of Internal Medicine, Saint Louis University, St. Louis, Missouri
| | - Jonathan Bruno
- Department of Internal Medicine, Saint Louis University, St. Louis, Missouri
- UNC Kidney Center, University of North Carolina, Chapel Hill, North Carolina
| | - Kiyoko Oshima
- Department of Pathology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Yao-Wen Cheng
- UNC Kidney Center, University of North Carolina, Chapel Hill, North Carolina
| | - Stephen P Holly
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, North Carolina
| | - Leslie V Parise
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, North Carolina
| | - Terrance M Egan
- Department of Pharmacology and Physiology, Saint Louis University, St. Louis, Missouri
| | - John C Edwards
- Department of Internal Medicine, Saint Louis University, St. Louis, Missouri
- UNC Kidney Center, University of North Carolina, Chapel Hill, North Carolina
- Department of Pharmacology and Physiology, Saint Louis University, St. Louis, Missouri
| |
Collapse
|
44
|
Faria SS, Morris CFM, Silva AR, Fonseca MP, Forget P, Castro MS, Fontes W. A Timely Shift from Shotgun to Targeted Proteomics and How It Can Be Groundbreaking for Cancer Research. Front Oncol 2017; 7:13. [PMID: 28265552 PMCID: PMC5316539 DOI: 10.3389/fonc.2017.00013] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 01/17/2017] [Indexed: 01/10/2023] Open
Abstract
The fact that cancer is a leading cause of death all around the world has naturally sparked major efforts in the pursuit of novel and more efficient biomarkers that could better serve as diagnostic tools, prognostic predictors, or therapeutical targets in the battle against this type of disease. Mass spectrometry-based proteomics has proven itself as a robust and logical alternative to the immuno-based methods that once dominated the field. Nevertheless, intrinsic limitations of classic proteomic approaches such as the natural gap between shotgun discovery-based methods and clinically applicable results have called for the implementation of more direct, hypothesis-based studies such as those made available through targeted approaches, that might be able to streamline biomarker discovery and validation as a means to increase survivability of affected patients. In fact, the paradigm shifting potential of modern targeted proteomics applied to cancer research can be demonstrated by the large number of advancements and increasing examples of new and more useful biomarkers found during the course of this review in different aspects of cancer research. Out of the many studies dedicated to cancer biomarker discovery, we were able to devise some clear trends, such as the fact that breast cancer is the most common type of tumor studied and that most of the research for any given type of cancer is focused on the discovery diagnostic biomarkers, with the exception of those that rely on samples other than plasma and serum, which are generally aimed toward prognostic markers. Interestingly, the most common type of targeted approach is based on stable isotope dilution-selected reaction monitoring protocols for quantification of the target molecules. Overall, this reinforces that notion that targeted proteomics has already started to fulfill its role as a groundbreaking strategy that may enable researchers to catapult the number of viable, effective, and validated biomarkers in cancer clinical practice.
Collapse
Affiliation(s)
- Sara S Faria
- Mastology Program, Federal University of Uberlandia (UFU) , Uberlandia , Brazil
| | - Carlos F M Morris
- Laboratory of Biochemistry and Protein Chemistry, Department of Cell Biology, Institute of Biology, University of Brasilia , Brasília , Brazil
| | - Adriano R Silva
- Laboratory of Biochemistry and Protein Chemistry, Department of Cell Biology, Institute of Biology, University of Brasilia , Brasília , Brazil
| | - Micaella P Fonseca
- Laboratory of Biochemistry and Protein Chemistry, Department of Cell Biology, Institute of Biology, University of Brasilia, Brasília, Brazil; Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Patrice Forget
- Department of Anesthesiology and Perioperative Medicine, Universitair Ziekenhuis Brussel, Vrije Universiteit of Brussel , Brussels , Belgium
| | - Mariana S Castro
- Laboratory of Biochemistry and Protein Chemistry, Department of Cell Biology, Institute of Biology, University of Brasilia , Brasília , Brazil
| | - Wagner Fontes
- Laboratory of Biochemistry and Protein Chemistry, Department of Cell Biology, Institute of Biology, University of Brasilia , Brasília , Brazil
| |
Collapse
|
45
|
Zou Q, Yang Z, Li D, Liu Z, Yuan Y. Association of chloride intracellular channel 4 and Indian hedgehog proteins with survival of patients with pancreatic ductal adenocarcinoma. Int J Exp Pathol 2017; 97:422-429. [PMID: 28205343 DOI: 10.1111/iep.12213] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2016] [Accepted: 10/18/2016] [Indexed: 12/30/2022] Open
Abstract
Pancreatic cancer is the fourth most common cause of cancer-related mortality. Novel molecular biomarkers need to be identified for personalized medicine and to improve survival. The aim of this study was to examine chloride intracellular channel 4 (CLIC4) and Indian Hedgehog (Ihh) expression in benign and malignant lesions of the pancreas and to examine the eventual association between CLIC4 and Ihh expression, with clinicopathological features and prognosis of pancreatic cancer. A retrospective study of specimens collected from January 2000 to December 2011 at the Department of Pathology of the Second and Third Xiangya Hospitals, Central South University was undertaken to explore this question. Immunohistochemistry of CLIC4 and Ihh was performed with EnVision™ in 106 pancreatic ductal adenocarcinoma specimens, 35 paracancer samples (2 cm away from the tumour, when possible or available), 55 benign lesions and 13 normal tissue samples. CLIC4 and Ihh expression in pancreatic ductal adenocarcinoma were significantly higher than in paracancer tissue and benign lesions (CLIC4: P = 0.009 and Ihh: P < 0.0001; CLIC4: P = 0.0004 and Ihh: P = 0.0001 respectively). CLIC4 and Ihh expression was negative in normal pancreatic tissues. The expression of CLIC4 and Ihh was associated significantly with tumour grade, lymph node metastasis, tumour invasion and poor overall survival. Thus CLIC4 and Ihh could serve as biological markers for the progression, metastasis and/or invasiveness of pancreatic ductal adenocarcinoma.
Collapse
Affiliation(s)
- Qiong Zou
- Department of Pathology, Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhulin Yang
- Research Laboratory of Hepatobiliary Diseases, Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Daiqiang Li
- Department of Pathology, Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ziru Liu
- Research Laboratory of Hepatobiliary Diseases, Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yuan Yuan
- Department of Pathology, Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
46
|
Pan H, Gu L, Liu B, Li Y, Wang Y, Bai X, Li L, Wang B, Peng Q, Yao Z, Tang Z. Tropomyosin-1 acts as a potential tumor suppressor in human oral squamous cell carcinoma. PLoS One 2017; 12:e0168900. [PMID: 28182650 PMCID: PMC5300227 DOI: 10.1371/journal.pone.0168900] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Accepted: 12/05/2016] [Indexed: 01/16/2023] Open
Abstract
It is widely accepted that oral squamous cell carcinoma (OSCC) is a major contributor to the incidence and mortality of neck and head cancer. Tropomyosin-1 (TPM1), which is expressed at a low level, has been considered a prominent tumor-suppressing gene in a variety of solid tumors, although the precise mechanism of the TPM1 gene in OSCC progression remains unknown. We found that TPM1 expression levels decreased in OSCC patients and OSCC cell lines. The overall and cancer-specific survival of patients who exhibited low TPM1 levels were inferior to those of patients who had high TPM1 levels. It was also found that OSCC patients who suffered from disease stageⅠ-Ⅱ were more likely to have an up-regulated TPM1 expression level, and OSCC patients with lymph node metastasis had a higher probability of exhibiting reduced TPM1 expression. We show that overexpression of TPM1 can promote cell apoptosis and inhibit migration. Our results suggest that TPM1 can suppress tumors in OSCC, and the TPM1 expression level is related to OSCC patient prognosis.
Collapse
Affiliation(s)
- Hao Pan
- Department of Oral & Maxillofacial Surgery, Xiangya Stomatological Hospital & School of Stomatology, Central South University, Changsha, Hunan, China
| | - Liqun Gu
- Department of Oral & Maxillofacial Surgery, Xiangya Stomatological Hospital & School of Stomatology, Central South University, Changsha, Hunan, China
| | - Binjie Liu
- Department of Periodontics, Xiangya Stomatological Hospital & School of Stomatology, Central South University, Changsha, Hunan, China
| | - Yiping Li
- Department of Prosthodontics, Xiangya Stomatological Hospital & School of Stomatology, Central South University, Changsha, Hunan, China
| | - Yuehong Wang
- Department of Prosthodontics, Xiangya Stomatological Hospital & School of Stomatology, Central South University, Changsha, Hunan, China
| | - Xinna Bai
- Department of Conservative Dentistry & Endodontics, Xiangya Stomatological Hospital & School of Stomatology, Central South University, Changsha, Hunan, China
| | - Long Li
- Department of Oral Pathology, Xiangya Stomatological Hospital & School of Stomatology, Central South University, Changsha, Hunan, China
| | - Baisheng Wang
- Department of Oral & Maxillofacial Surgery, Xiangya Stomatological Hospital & School of Stomatology, Central South University, Changsha, Hunan, China
| | - Qian Peng
- Department of Oral & Maxillofacial Surgery, Xiangya Stomatological Hospital & School of Stomatology, Central South University, Changsha, Hunan, China
| | - Zhigang Yao
- Department of Oral Pathology, Xiangya Stomatological Hospital & School of Stomatology, Central South University, Changsha, Hunan, China
| | - Zhangui Tang
- Department of Oral & Maxillofacial Surgery, Xiangya Stomatological Hospital & School of Stomatology, Central South University, Changsha, Hunan, China
| |
Collapse
|
47
|
Peruzzo R, Biasutto L, Szabò I, Leanza L. Impact of intracellular ion channels on cancer development and progression. EUROPEAN BIOPHYSICS JOURNAL : EBJ 2016; 45:685-707. [PMID: 27289382 PMCID: PMC5045486 DOI: 10.1007/s00249-016-1143-0] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Revised: 05/13/2016] [Accepted: 05/17/2016] [Indexed: 12/13/2022]
Abstract
Cancer research is nowadays focused on the identification of possible new targets in order to try to develop new drugs for curing untreatable tumors. Ion channels have emerged as "oncogenic" proteins, since they have an aberrant expression in cancers compared to normal tissues and contribute to several hallmarks of cancer, such as metabolic re-programming, limitless proliferative potential, apoptosis-resistance, stimulation of neo-angiogenesis as well as cell migration and invasiveness. In recent years, not only the plasma membrane but also intracellular channels and transporters have arisen as oncological targets and were proposed to be associated with tumorigenesis. Therefore, the research is currently focusing on understanding the possible role of intracellular ion channels in cancer development and progression on one hand and, on the other, on developing new possible drugs able to modulate the expression and/or activity of these channels. In a few cases, the efficacy of channel-targeting drugs in reducing tumors has already been demonstrated in vivo in preclinical mouse models.
Collapse
Affiliation(s)
| | - Lucia Biasutto
- CNR Institute of Neuroscience, Padua, Italy
- Department of Biomedical Sciences, University of Padua, Padua, Italy
| | - Ildikò Szabò
- Department of Biology, University of Padua, Padua, Italy
- CNR Institute of Neuroscience, Padua, Italy
| | - Luigi Leanza
- Department of Biology, University of Padua, Padua, Italy.
| |
Collapse
|
48
|
Savill SA, Leitch HF, Harvey JN, Thomas TH. Inflammatory Adipokines Decrease Expression of Two High Molecular Weight Isoforms of Tropomyosin Similar to the Change in Type 2 Diabetic Patients. PLoS One 2016; 11:e0162908. [PMID: 27649540 PMCID: PMC5029944 DOI: 10.1371/journal.pone.0162908] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 08/30/2016] [Indexed: 01/04/2023] Open
Abstract
Cardiovascular disease and cancer are increased in Type 2 diabetes. TPM1 and TPM4 genes encode proteins associated with cardiovascular and neoplastic disease. High (HMW) and low (LMW) molecular weight isoforms from TPM1 and TPM4 are altered in several cancer cells and the 3'UTR of TPM1 mRNA is tumour suppressive. Leukocytes influence cardiovascular and neoplastic disease by immunosurveillance for cancer and by chronic inflammation in Type 2 diabetes and cardiovascular disease. The aim was to determine changes in expression of isoforms from TPM1 and TPM4 genes in leukocytes from Type 2 diabetic patients and to use the leukocyte cell line THP1 to identify possible mediators of changes in the patients. Gene expression was determined by RT-qPCR. In diabetes, expression of HMW isoforms from TPM1 were markedly decreased (0.55 v 1.00; p = 0.019) but HMW isoforms from TPM4 were not significantly different (0.76 v 1.00; p = 0.205). Within individual variance in expression of HMW isoforms was very high. The change in expression in HMW isoforms from TPM1 and TPM4 was replicated in THP1 cells treated with 1 ng/ml TNFα (0.10 and 0.12 v 1.00 respectively) or 10 ng/ml IL-1α (0.17 and 0.14 v 1.00 respectively). Increased insulin or glucose concentrations had no substantial effects on TPM1 or TPM4 expression. Decreased TPM1 mRNA resulted in decreases in HMW protein levels. Expression of HMW isoforms from TPM1 is decreased in Type 2 diabetes. This is probably due to increased levels of inflammatory cytokines TNFα and IL-1α in Type 2 diabetes. Lower levels of TPM1 mRNA reduce tumour suppression and could contribute to increased cancer risk in Type 2 diabetes. Decreased HMW tropomyosin isoforms are associated with cancer. Decreased HMW isoforms give rise to cells that are more plastic, motile, invasive and prone to dedifferentiation resulting in leukocytes that are more invasive but less functionally effective.
Collapse
Affiliation(s)
- Stuart A. Savill
- Betsi Cadwaladr University Health Board, Croesnewydd Road, Wrexham, United Kingdom
- * E-mail:
| | - Helen F. Leitch
- Betsi Cadwaladr University Health Board, Croesnewydd Road, Wrexham, United Kingdom
| | - John N. Harvey
- Betsi Cadwaladr University Health Board, Croesnewydd Road, Wrexham, United Kingdom
- School of Medical Sciences, Wrexham Academic Unit, Bangor University, Bangor, United Kingdom
| | - Trevor H. Thomas
- Betsi Cadwaladr University Health Board, Croesnewydd Road, Wrexham, United Kingdom
| |
Collapse
|
49
|
Setti M, Osti D, Richichi C, Ortensi B, Del Bene M, Fornasari L, Beznoussenko G, Mironov A, Rappa G, Cuomo A, Faretta M, Bonaldi T, Lorico A, Pelicci G. Extracellular vesicle-mediated transfer of CLIC1 protein is a novel mechanism for the regulation of glioblastoma growth. Oncotarget 2016; 6:31413-27. [PMID: 26429879 PMCID: PMC4741615 DOI: 10.18632/oncotarget.5105] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Accepted: 09/18/2015] [Indexed: 01/09/2023] Open
Abstract
Little progresses have been made in the treatment of glioblastoma (GBM), the most aggressive and lethal among brain tumors. Recently we have demonstrated that Chloride Intracellular Channel-1 (CLIC1) is overexpressed in GBM compared to normal tissues, with highest expression in patients with poor prognosis. Moreover, CLIC1-silencing in cancer stem cells (CSCs) isolated from human GBM patients negatively influences proliferative capacity and self-renewal properties in vitro and impairs the in vivo tumorigenic potential. Here we show that CLIC1 exists also as a circulating protein, secreted via extracellular vesicles (EVs) released by either cell lines or GBM-derived CSCs. Extracellular vesicles (EVs), comprising exosomes and microvesicles based on their composition and biophysical properties, have been shown to sustain tumor growth in a variety of model systems, including GBM. Interestingly, treatment of GBM cells with CLIC1-containing EVs stimulates cell growth both in vitro and in vivo in a CLIC1-dose dependent manner. EVs derived from CLIC1-overexpressing GBM cells are strong inducers of proliferation in vitro and tumor engraftment in vivo. These stimulations are significantly attenuated by treatment of GBM cells with EVs derived from CLIC1-silenced cells. However, CLIC1 modulation appears to have no direct role in EV structure, biogenesis and secretion. These findings reveal that, apart from the function of CLIC1 cellular reservoir, CLIC1 contained in EVs is a novel regulator of GBM growth.
Collapse
Affiliation(s)
- Matteo Setti
- Department of Experimental Oncology, European Institute of Oncology, Milan, Italy
| | - Daniela Osti
- Department of Experimental Oncology, European Institute of Oncology, Milan, Italy
| | - Cristina Richichi
- Department of Experimental Oncology, European Institute of Oncology, Milan, Italy
| | - Barbara Ortensi
- Department of Experimental Oncology, European Institute of Oncology, Milan, Italy
| | - Massimiliano Del Bene
- Department of Neurosurgery, Fondazione IRCCS Istituto Neurologico C. Besta, Milan, Italy
| | - Lorenzo Fornasari
- Department of Experimental Oncology, European Institute of Oncology, Milan, Italy
| | - Galina Beznoussenko
- Institute of Molecular Oncology (IFOM) of The Italian Foundation for Cancer Research (FIRC), Milan, Italy
| | - Alexandre Mironov
- Institute of Molecular Oncology (IFOM) of The Italian Foundation for Cancer Research (FIRC), Milan, Italy
| | - Germana Rappa
- Department of Experimental Oncology, European Institute of Oncology, Milan, Italy
| | - Alessandro Cuomo
- Department of Experimental Oncology, European Institute of Oncology, Milan, Italy
| | - Mario Faretta
- Department of Experimental Oncology, European Institute of Oncology, Milan, Italy
| | - Tiziana Bonaldi
- Department of Experimental Oncology, European Institute of Oncology, Milan, Italy
| | - Aurelio Lorico
- Cancer Research Center, Roseman University of Health Sciences with Roseman University College of Medicine, Las Vegas, NV, USA
| | - Giuliana Pelicci
- Department of Experimental Oncology, European Institute of Oncology, Milan, Italy.,Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy
| |
Collapse
|
50
|
Meo AD, Pasic MD, Yousef GM. Proteomics and peptidomics: moving toward precision medicine in urological malignancies. Oncotarget 2016; 7:52460-52474. [PMID: 27119500 PMCID: PMC5239567 DOI: 10.18632/oncotarget.8931] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Accepted: 04/16/2016] [Indexed: 12/31/2022] Open
Abstract
Urological malignancies are a major cause of morbidity and mortality worldwide. Advances in early detection, diagnosis, prognosis and prediction of treatment response can significantly improve patient care. Proteomic and peptidomic profiling studies are at the center of kidney, prostate and bladder cancer biomarker discovery and have shown great promise for improved clinical assessment. Mass spectrometry (MS) is the most widely employed method for proteomic and peptidomic analyses. A number of MS platforms have been developed to facilitate accurate identification of clinically relevant markers in various complex biological samples including tissue, urine and blood. Furthermore, protein profiling studies have been instrumental in the successful introduction of several diagnostic multimarker tests into the clinic. In this review, we will provide a brief overview of high-throughput technologies for protein and peptide based biomarker discovery. We will also examine the current state of kidney, prostate and bladder cancer biomarker research as well as review the journey toward successful clinical implementation.
Collapse
Affiliation(s)
- Ashley Di Meo
- Department of Laboratory Medicine, and The Keenan Research Centre for Biomedical Science at The Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Maria D. Pasic
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Department of Laboratory Medicine, St. Joseph's Health Centre, Toronto, Ontario, Canada
| | - George M. Yousef
- Department of Laboratory Medicine, and The Keenan Research Centre for Biomedical Science at The Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|