1
|
Ke B, Jin P, Wang XJ, Liu N, Liang H, Zhang RP. Oncogenic and immunological role of EDIL3 in human tumours: From pan-cancer analysis to validation in gastric cancer. Heliyon 2024; 10:e32291. [PMID: 38882287 PMCID: PMC11180325 DOI: 10.1016/j.heliyon.2024.e32291] [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: 01/02/2024] [Revised: 05/30/2024] [Accepted: 05/31/2024] [Indexed: 06/18/2024] Open
Abstract
Background Epidermal growth factor-like repeats and discoidin I-like domains 3 (EDIL3) is a secreted extracellular matrix protein implicated in diverse physiological and pathological processes including embryonic development, angiogenesis, and anti-inflammatory responses. Recent reports have indicated that EDIL3 play critical roles in carcinogenesis and progression of many cancers. Herein, we performed a pan-cancer investigation to study the potential functions of EDIL3 in various cancers and experimentally validate its function in gastric cancer (GC). Methods We analysed EDIL3 expression profiles in different tumours using The Cancer Genome Atlas database. The Kaplan-Meier Plotter was used to investigate the prognostic value of EDIL3, while receiver operating characteristic curve was performed to analyze its diagnostic efficacy. Several bioinformatics tools were used to study the association between EDIL3 and promoter methylation, gene enrichment analysis, immune infiltration, immune-related genes, and drug sensitivity. Molecular biology experiments were conducted to validate the tumorigenic effects of EDIL3. Results EDIL3 is variably expressed in different cancers and is closely associated with clinical outcomes. An inverse correlation between EDIL3 and DNA methylation has been observed in 13 cancers. Enrichment analysis indicated that EDIL3 is correlated with many cellular pathways such as extracellular matrix receptor interactions and focal adhesion. EDIL3 was tightly associated with immune infiltration and immune checkpoints. EDIL3 knockdown can promote GC calls apoptosis while preventing proliferation, migration, and invasion in vitro. Conclusion EDIL3 is a promising prognostic, diagnostic, and immunological biomarker in various cancers, which could be applied as a new target for cancer therapy.
Collapse
Affiliation(s)
- Bin Ke
- Department of Gastric Surgery, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Key Laboratory of Digestive Cancer, 300060, Tianjin, China
| | - Peng Jin
- Department of Gastric Surgery, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Key Laboratory of Digestive Cancer, 300060, Tianjin, China
| | - Xue-Jun Wang
- Department of Gastric Surgery, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Key Laboratory of Digestive Cancer, 300060, Tianjin, China
| | - Ning Liu
- Department of Gastric Surgery, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Key Laboratory of Digestive Cancer, 300060, Tianjin, China
| | - Han Liang
- Department of Gastric Surgery, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Key Laboratory of Digestive Cancer, 300060, Tianjin, China
| | - Ru-Peng Zhang
- Department of Gastric Surgery, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Key Laboratory of Digestive Cancer, 300060, Tianjin, China
| |
Collapse
|
2
|
Li M, Zhong D, Li G. Regulatory role of local tissue signal Del-1 in cancer and inflammation: a review. Cell Mol Biol Lett 2021; 26:31. [PMID: 34217213 PMCID: PMC8254313 DOI: 10.1186/s11658-021-00274-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 06/28/2021] [Indexed: 12/29/2022] Open
Abstract
Developmental endothelial locus-1 (Del-1) is a secretory, multifunctional domain protein. It can bind to integrins and phosphatidylserine. As a local tissue signal, it plays a regulatory role in the cancer microenvironment and inflammation. Del-1 has destructive effects in most cancers and is associated with the progression and invasion of some cancers. In contrast, Del-1 also plays a protective role in inflammation. Del-1 regulates inflammation by regulating the generation of neutrophils in bone marrow, inhibiting the recruitment and migration of neutrophils and accelerating the clearance of neutrophils by macrophages. Del-1 and IL-17 are reciprocally regulated, and their balance maintains immune system homeostasis. Del-1 is expected to become a new therapeutic target for inflammatory disorders such as multiple sclerosis.
Collapse
Affiliation(s)
- Meng Li
- Department of Neurology, First Affiliated Hospital of Harbin Medical University, No. 23 Youzheng Road, Harbin, 150001, Heilongjiang, China
| | - Di Zhong
- Department of Neurology, First Affiliated Hospital of Harbin Medical University, No. 23 Youzheng Road, Harbin, 150001, Heilongjiang, China.
| | - Guozhong Li
- Department of Neurology, First Affiliated Hospital of Harbin Medical University, No. 23 Youzheng Road, Harbin, 150001, Heilongjiang, China
| |
Collapse
|
3
|
Chung JW, Kim HT, Ha YS, Lee EH, Chun SY, Lee CH, Byeon KH, Choi SH, Lee JN, Kim BS, Kim TH, Yoo ES, Yoon GS, Baek MC, Kwon TG. Identification of a novel non-invasive biological marker to overcome the shortcomings of PSA in diagnosis and risk stratification for prostate cancer: Initial prospective study of developmental endothelial locus-1 protein. PLoS One 2021; 16:e0250254. [PMID: 33901217 PMCID: PMC8075267 DOI: 10.1371/journal.pone.0250254] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 04/01/2021] [Indexed: 12/27/2022] Open
Abstract
Objective This prospective study sought to clarify the developmental endothelial locus-1 (Del-1) protein as values of diagnosis and risk stratification of prostate cancer (PCa). Design From February 2017 to December 2019, a total 458 patients who underwent transrectal ultrasound guided prostate biopsy or surgery of benign prostatic hyperplasia agreed to research of Del-1 protein. We prospectively compared and analyzed the Del-1 protein and prostate specific antigen (PSA) in relation to the patients’ demographic and clinicopathological characteristics. Results Mean age was 68.86±8.55 years. Mean PSA and Del-1 protein was 21.72±89.37, 0.099±0.145, respectively. Two hundred seventy-six (60.3%) patients were diagnosed as PCa. Among them, 181 patients underwent radical prostatectomy (RP). There were significant differences in Del-1 protein between benign and PCa group (0.066±0.131 vs 0.121±0.149, respectively, p<0.001). When we set the cut-off value of del-1 protein as 0.120, in patients with 3≤PSA≤8, positive predictive value and specificity of Del-1 protein (≥0.120) for predicting PCa were 88.9% (56/63) and 93.5% (101/108), respectively. Among 181 patients who underwent RP, there were significant differences in Del-1 protein according to stage (pT2 vs pT3a vs ≥pT3b) (0.113±0.078, 0.171±0.121, 0.227±0.161, respectively, p<0.001) and to Gleason score (6 (3+3) or 7 (3+4) vs 7 (4+3) or 8 (4+4) vs 9 or 10) (0.134±0.103, 0.150±0.109, 0.212±0.178, respectively, P = 0.044). Multivariate analysis showed that PSA, Del-1 protein and high Gleason score (≥9) were the independent prognostic factors for predicting higher pT stage (≥3b). Furthermore, age, PSA and Del-1 protein were independent prognostic factors for predicting significant PCa. Conclusion Patients with PCa showed higher expression of Del-1 protein than benign patients. Del-1 protein increased with the stage and Gleason score of PCa. Collaboration with PSA, Del-1 protein can be a non-invasive useful marker for diagnosis and risk stratification of PCa.
Collapse
Affiliation(s)
- Jae-Wook Chung
- Department of Urology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Hyun Tae Kim
- Department of Urology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Yun-Sok Ha
- Department of Urology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Eun Hye Lee
- Biomedical Research Institute, Kyungpook National University, Daegu, Republic of Korea
| | - So Young Chun
- Biomedical Research Institute, Kyungpook National University, Daegu, Republic of Korea
| | - Chan-Hyeong Lee
- Department of Molecular Medicine, Cell and Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu, Korea
| | - Kyeong Hyeon Byeon
- Department of Urology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Seock Hwan Choi
- Department of Urology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Jun Nyung Lee
- Department of Urology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Bum Soo Kim
- Department of Urology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Tae-Hwan Kim
- Department of Urology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Eun Sang Yoo
- Department of Urology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Ghil Suk Yoon
- Department of Pathology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Moon-Chang Baek
- Department of Molecular Medicine, Cell and Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu, Korea
- * E-mail: (MCB); (TGK)
| | - Tae Gyun Kwon
- Department of Urology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
- Joint Institute for Regenerative Medicine, Kyungpook National University, Daegu, Republic of Korea
- * E-mail: (MCB); (TGK)
| |
Collapse
|
4
|
An Integrated Approach of the Potential Underlying Molecular Mechanistic Paradigms of SARS-CoV-2-Mediated Coagulopathy. Indian J Clin Biochem 2021; 36:387-403. [PMID: 33875909 PMCID: PMC8047580 DOI: 10.1007/s12291-021-00972-3] [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: 12/13/2020] [Accepted: 04/03/2021] [Indexed: 02/06/2023]
Abstract
Coronavirus disease 2019 (Covid-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a pandemic disease which has affected more than 6.2 million people globally, with numbers mounting considerably daily. However, till date, no specific treatment modalities are available for Covid-19 and also not much information is known about this disease. Recent studies have revealed that SARS-CoV-2 infection is associated with the generation of thrombosis and coagulopathy. Fundamentally, it has been believed that a diverse array of signalling pathways might be responsible for the activation of coagulation cascade during SARS-CoV-2 infection. Henceforth, a detailed understanding of these probable underlying molecular mechanistic pathways causing thrombosis in Covid-19 disease deserves an urgent exploration. Therefore, in this review, the hypothetical crosstalk between distinct signalling pathways including apoptosis, inflammation, hypoxia and angiogenesis attributable for the commencement of thrombotic events during SARS-CoV-2 infection has been addressed which might further unravel promising therapeutic targets in Covid-19 disease.
Collapse
|
5
|
Oplawski M, Dziobek K, Zmarzły N, Grabarek B, Tomala B, Leśniak E, Adwent I, Januszyk P, Dąbruś D, Boroń D. Evaluation of Changes in the Expression Pattern of EDIL3 in Different Grades of Endometrial Cancer. Curr Pharm Biotechnol 2019; 20:483-488. [PMID: 30961491 PMCID: PMC6806535 DOI: 10.2174/1389201020666190408112822] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 03/01/2019] [Accepted: 03/26/2019] [Indexed: 01/26/2023]
Abstract
BACKGROUND EDIL3 is an extracellular matrix protein that plays a key role in angiogenesis. Changes in the pattern of its expression also affect cellular processes and the tumor microenvironment. Elevated level of EDIL3 is considered an unfavorable prognostic marker of survival. OBJECTIVE The aim of this study was to evaluate the changes in EDIL3 expression in endometrial cancer at various degrees of its differentiation (G1-G3) and to discuss its potential role as a molecular diagnostic marker and therapeutic target. METHODS The study group consisted of 45 patients with endometrial cancer: G1, 17; G2, 15; G3, 13. The control group (C) included 15 patients without neoplastic changes. The expression of EDIL3 was assessed using immunohistochemistry. Statistical analysis was performed using the Statistica 12 PL software (p<0.05). RESULTS Analysis of EDIL3 expression showed that the average optical density of the reaction product in G1 reached 130% of the control, while the values in G2 and G3 were 153% and 158%, respectively. Regardless of the endometrial cancer grade, an increase in EDIL3 level was observed compared to the control. CONCLUSION In our study, we demonstrated overexpression of EDIL3 protein in endometrial cancer. Differences in expression between degrees of tumor differentiation suggest the potential of using changes in EDIL3 level as a new complementary diagnostic marker and target for anti-angiogenic therapy.
Collapse
Affiliation(s)
- Marcin Oplawski
- Department of Gynecology and Obstetrics with Gynecologic Oncology, Ludwik Rydygier Memorial Specialized Hospital, Krakow, Poland
| | - Konrad Dziobek
- Center of Oncology, M. Sklodowska-Curie Memorial Institute, Cracow Branch, Poland
| | - Nikola Zmarzły
- Department of Molecular Biology, School of Pharmacy with the Division of Laboratory Medicine in Sosnowiec, Medical University of Silesia in Katowice, Poland
| | - Beniamin Grabarek
- Department of Molecular Biology, School of Pharmacy with the Division of Laboratory Medicine in Sosnowiec, Medical University of Silesia in Katowice, Poland
| | - Barbara Tomala
- Faculty of Health Science, Public Higher Medical Professional School in Opole, Poland
| | - Ewa Leśniak
- Faculty of Health Science, Public Higher Medical Professional School in Opole, Poland
| | - Iwona Adwent
- Department of Molecular Biology, School of Pharmacy with the Division of Laboratory Medicine in Sosnowiec, Medical University of Silesia in Katowice, Poland
| | - Piotr Januszyk
- Department of Molecular Biology, School of Pharmacy with the Division of Laboratory Medicine in Sosnowiec, Medical University of Silesia in Katowice, Poland
| | - Dariusz Dąbruś
- Faculty of Health Science, Public Higher Medical Professional School in Opole, Poland
| | - Dariusz Boroń
- Department of Gynecology and Obstetrics with Gynecologic Oncology, Ludwik Rydygier Memorial Specialized Hospital, Krakow, Poland.,Faculty of Health Science, Public Higher Medical Professional School in Opole, Poland.,Department of Histology and Cell Pathology, School of Medicine with the Division of Dentistry in Zabrze, Medical University of Silesia in Katowice, Poland
| |
Collapse
|
6
|
Dzuricky M, Xiong S, Weber P, Chilkoti A. Avidity and Cell Uptake of Integrin-Targeting Polypeptide Micelles is Strongly Shape-Dependent. NANO LETTERS 2019; 19:6124-6132. [PMID: 31389705 DOI: 10.1021/acs.nanolett.9b02095] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
We describe a genetically encoded micelle for targeted delivery consisting of a diblock polypeptide with segments derived from repetitive protein motifs inspired by Drosophila melanogaster Rec-1 resilin and human tropoelastin with a C-terminal fusion of an integrin-targeting fibronectin type III domain. By systematically varying the weight fraction of the hydrophilic elastin-like polypeptide (ELP) block and molecular weight of the diblock polypeptide, we designed micelles of different morphologies that modulate the binding avidity of the human wild-type 10th fibronectin domain (Fn3) as a function of shape. We show that wormlike micelles that present the Fn3 domain have a 1000-fold greater avidity for the αvβ3 receptor compared to the monomer ligand and an avidity that is greater than a clinically relevant antibody that is driven by their multivalency. The amplified avidity of these micelles leads to significantly increased cellular internalization, a feature that may have utility for the intracellular delivery of drugs that are loaded into the core of these micelles.
Collapse
Affiliation(s)
- Michael Dzuricky
- Department of Biomedical Engineering , Duke University , Durham , North Carolina 27708 , United States
| | - Sinan Xiong
- Department of Biomedical Engineering , Duke University , Durham , North Carolina 27708 , United States
| | - Patrick Weber
- Department of Biomedical Engineering , Duke University , Durham , North Carolina 27708 , United States
- Swiss Nanoscience Institute , University of Basel , Basel 4056 , Switzerland
| | - Ashutosh Chilkoti
- Department of Biomedical Engineering , Duke University , Durham , North Carolina 27708 , United States
| |
Collapse
|
7
|
Evans CE. Hypoxia and HIF activation as a possible link between sepsis and thrombosis. Thromb J 2019; 17:16. [PMID: 31423111 PMCID: PMC6693167 DOI: 10.1186/s12959-019-0205-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 07/23/2019] [Indexed: 01/01/2023] Open
Abstract
Risk factors for thrombosis include hypoxia and sepsis, but the mechanisms that control sepsis-induced thrombus formation are incompletely understood. A recent article published in Thrombosis Journal: (i) reviews the role of endothelial cells in the pathogenesis of sepsis-associated microthrombosis; (ii) describes a novel ‘two-path unifying theory’ of hemostatic discorders; and (iii) refers to hypoxia as a consequence of microthrombus formation in sepsis patients. The current article adds to this review by describing how sepsis and thrombus formation could be linked through hypoxia and activation of hypoxia-inducible transcription factors (HIFs). In other words, hypoxia and HIF activation may be a cause as well as a consequence of thrombosis in sepsis patients. While microthrombosis reduces microvascular blood flow causing local hypoxia and tissue ischemia, sepsis-induced increases in HIF1 activation could conversely increase the expression of coagulant factors and integrins that promote thrombus formation, and stimulate the formation of pro-thrombotic neutrophil extracellular traps. A better understanding of the role of cell-specific HIFs in thrombus formation could lead to the development of novel prophylactic therapies for individuals at risk of thrombosis.
Collapse
Affiliation(s)
- Colin E Evans
- 1Program for Lung and Vascular Biology, Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL USA.,2Department of Pediatrics, Division of Critical Care, Northwestern University Feinberg School of Medicine, Chicago, IL USA
| |
Collapse
|
8
|
Latteyer S, Christoph S, Theurer S, Hönes GS, Schmid KW, Führer D, Moeller LC. Thyroxine promotes lung cancer growth in an orthotopic mouse model. Endocr Relat Cancer 2019; 26:565-574. [PMID: 30893642 DOI: 10.1530/erc-18-0353] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 03/20/2019] [Indexed: 12/30/2022]
Abstract
Thyroid hormones are important for physiology and homeostasis. In addition to nuclear thyroid hormone receptors, the plasma membrane protein integrin αvβ3 has been recognized as a receptor for both thyroxine (T4) and triiodothyronine (T3). Here, we studied whether thyroid hormone promotes growth of murine lung cancer via αvβ3 in vivo. Murine Lewis lung carcinoma cells (3LL), stably transfected with luciferase, were injected into mouse lungs. Tumor growth in untreated mice was compared to hypothyroid mice and hypothyroid mice treated with T3 or T4 with or without the αvβ3 inhibitor 3,5,3',5'-tetraiodothyroacetic acid (Tetrac). Tumor progression was determined by serial in vivo imaging of bioluminescence emitted from the tumor. Tumor weight was recorded at the end of the experiment. Neoangiogenesis was determined by immunohistochemistry for CD31. Tumor growth was reduced in hypothyroidism and increased by T4 treatment. Strikingly, only T4 but not T3 treatment promoted tumor growth. This T4 effect was abrogated by the αvβ3 inhibitor Tetrac. Tumor weight and neoangiogenesis were also significantly increased only in T4-treated mice. The T4 effect on tumor weight and neoangiogenesis was abolished by Tetrac. In vitro, T4 did not stimulate 3LL cell proliferation or signaling pathway activation. We conclude that T4 promotes lung cancer growth in this orthotopic mouse model. The tumor-promoting effect is mediated via the plasma membrane integrin αvβ3 and increased neoangiogenesis rather than direct stimulation of 3LL cells. These data suggest that such effects of levothyroxine may need to be considered in cancer patients on T4 substitution.
Collapse
Affiliation(s)
- S Latteyer
- Department of Endocrinology, Diabetes and Metabolism, University of Duisburg-Essen, Essen, Germany
| | - S Christoph
- Clinic for Bone Marrow Transplants, University of Duisburg-Essen, Essen, Germany
| | - S Theurer
- Institute of Pathology, University of Duisburg-Essen, Essen, Germany
| | - G S Hönes
- Department of Endocrinology, Diabetes and Metabolism, University of Duisburg-Essen, Essen, Germany
| | - K W Schmid
- Institute of Pathology, University of Duisburg-Essen, Essen, Germany
| | - D Führer
- Department of Endocrinology, Diabetes and Metabolism, University of Duisburg-Essen, Essen, Germany
| | - L C Moeller
- Department of Endocrinology, Diabetes and Metabolism, University of Duisburg-Essen, Essen, Germany
| |
Collapse
|
9
|
Arun AS, Tepper CG, Lam KS. Identification of integrin drug targets for 17 solid tumor types. Oncotarget 2018; 9:30146-30162. [PMID: 30046394 PMCID: PMC6059022 DOI: 10.18632/oncotarget.25731] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Accepted: 06/12/2018] [Indexed: 12/12/2022] Open
Abstract
Integrins are contributors to remodeling of the extracellular matrix and cell migration. Integrins participate in the assembly of the actin cytoskeleton, regulate growth factor signaling pathways, cell proliferation, and control cell motility. In solid tumors, integrins are involved in promoting metastasis to distant sites, and angiogenesis. Integrins are a key target in cancer therapy and imaging. Integrin antagonists have proven successful in halting invasion and migration of tumors. Overexpressed integrins are prime anti-cancer drug targets. To streamline the development of specific integrin cancer therapeutics, we curated data to predict which integrin heterodimers are pausible therapeutic targets against 17 different solid tumors. Computational analysis of The Cancer Genome Atlas (TCGA) gene expression data revealed a set of integrin targets that are differentially expressed in tumors. Filtered by FPKM (Fragments Per Kilobase of transcript per Million mapped reads) expression level, overexpressed subunits were paired into heterodimeric protein targets. By comparing the RNA-seq differential expression results with immunohistochemistry (IHC) data, overexpressed integrin subunits were validated. Biologics and small molecule drug compounds against these identified overexpressed subunits and heterodimeric receptors are potential therapeutics against these cancers. In addition, high-affinity and high-specificity ligands against these integrins can serve as efficient vehicles for delivery of cancer drugs, nanotherapeutics, or imaging probes against cancer.
Collapse
Affiliation(s)
- Adith S Arun
- Department of Biochemistry and Molecular Medicine, University of California Davis School of Medicine, UC Davis NCI-Designated Comprehensive Cancer Center, Sacramento, CA 95817, USA
| | - Clifford G Tepper
- Department of Biochemistry and Molecular Medicine, University of California Davis School of Medicine, UC Davis NCI-Designated Comprehensive Cancer Center, Sacramento, CA 95817, USA
| | - Kit S Lam
- Department of Biochemistry and Molecular Medicine, University of California Davis School of Medicine, UC Davis NCI-Designated Comprehensive Cancer Center, Sacramento, CA 95817, USA
| |
Collapse
|
10
|
Jeong D, Ban S, Oh S, Jin Lee S, Yong Park S, Koh YW. Prognostic Significance of EDIL3 Expression and Correlation with Mesenchymal Phenotype and Microvessel Density in Lung Adenocarcinoma. Sci Rep 2017; 7:8649. [PMID: 28819306 PMCID: PMC5561239 DOI: 10.1038/s41598-017-08851-9] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Accepted: 07/14/2017] [Indexed: 12/31/2022] Open
Abstract
We examined the prognostic significance of Epidermal Growth Factor-like repeats and Discoidin I-Like Domains 3 (EDIL3) expression and its correlations with mesenchymal phenotype and microvessel density in non-small cell lung carcinoma (NSCLC). A total of 268 NSCLC specimens were evaluated retrospectively by immunohistochemical staining for EDIL3, EMT markers (e-cadherin, β-catenin, and vimentin), and CD31 to measure microvessel density. EDIL3, e-cadherin, β-catenin, and vimentin were expressed in 16%, 22.8%, 3.7%, and 10.1% of the specimens, respectively. The mRNA level of EDIL3 in tumor was correlated with the level of EDIL3 protein expression using immunohistochemistry. In lung adenocarcinoma patients, EDIL3 expression was significantly correlated with low e-cadherin expression, high vimentin expression, and increased microvessel density (P < 0.001, P = 0.001, and P = 0.023, respectively). In lung squamous cell carcinoma patients, EDIL3 expression was significantly correlated with low e-cadherin expression and high vimentin expression (P = 0.021 and P = 0.002, respectively). In lung adenocarcinoma patients, EDIL3 was an independent prognostic factor for overall survival in a multivariate analysis (hazard ratio: 2.552, P = 0.004). EDIL3 is significantly correlated with mesenchymal phenotype, angiogenesis, and tumor progression in lung adenocarcinoma.
Collapse
Affiliation(s)
- Dongjun Jeong
- Department of Pathology, College of Medicine, Soonchunhyang University, Chonan, Republic of Korea
| | - Seona Ban
- Soonchunhyang medical science research institute, College of medicine, Soonchunhyang University, Chonan, Republic of Korea
| | - Seunghyun Oh
- Soonchunhyang medical science research institute, College of medicine, Soonchunhyang University, Chonan, Republic of Korea
| | - Su Jin Lee
- Department of Nuclear Medicine and Molecular Imaging, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Seong Yong Park
- Department of Thoracic and Cardiovascular Surgery, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Young Wha Koh
- Department of Pathology, Ajou University School of Medicine, Suwon, Republic of Korea.
| |
Collapse
|
11
|
Liu J, Hao Y, Wang Y, Hu S, Xu K, Lu C. Candidate methylated genes in osteoarthritis explored by bioinformatics analysis. Knee 2016; 23:1035-1043. [PMID: 27810435 DOI: 10.1016/j.knee.2016.09.020] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Revised: 08/01/2016] [Accepted: 09/20/2016] [Indexed: 02/02/2023]
Abstract
BACKGROUND This study aimed to explore potential novel genes correlated with osteoarthritis (OA). METHODS The gene expression profile of GSE48422 was downloaded from the Gene Expression Omnibus (GEO) database. This dataset included five arthritic cartilage samples and five non-arthritic cartilage samples from five female OA patients. Differentially methylated genes (DMGs) between the two kinds of samples were identified, followed by their functional analysis and protein-protein interaction (PPI) analysis. Furthermore, the Comparative Toxicogenomics Database (CTD) was used to further identify OA-related genes among these DMGs. RESULTS In total, 965 hypermethylated genes and 112 hypomethylated genes were identified in the arthritic cartilage samples. The hypermethylated genes (e.g., ADCY4 and ADCY6) were significantly related to the calcium signaling pathway and gonadotropin-releasing hormone signaling pathway, while the hypomethylated genes were implicated in the mammalian target of rapamycin signaling pathway. In the PPI network, several genes had a higher degree, such as ADCY4, ADCY6 and GPR17, and they interacted with each other. Additionally, 565 DMGs were predicted to be associated with OA, and five of them (e.g., COMP and EDIL3) were previously identified as OA markers. CONCLUSIONS The methylation of genes ADCY4, ADCY6 and GPR17, as well as the gonadotropin-releasing hormone signaling pathway, was newly found to be potentially associated with OA. They may be novel OA markers.
Collapse
Affiliation(s)
- Jie Liu
- Shaanxi University of Chinese Medicine, Shiji Avenue, Xi'an-Xianyang New Economic Zone, Shaanxi 712046, PR China
| | - Yangquan Hao
- Department of Osteonecrosis and Joint Reconstruction, Honghui Hospital, Xi'an Jiao Tong University Health Science Center, 555 Youyi East Road, Xi'an, Shaanxi 710068, PR China.
| | - Yugui Wang
- Shaanxi University of Chinese Medicine, Shiji Avenue, Xi'an-Xianyang New Economic Zone, Shaanxi 712046, PR China
| | - Shouye Hu
- Department of Osteonecrosis and Joint Reconstruction, Honghui Hospital, Xi'an Jiao Tong University Health Science Center, 555 Youyi East Road, Xi'an, Shaanxi 710068, PR China
| | - Ke Xu
- Department of Osteonecrosis and Joint Reconstruction, Honghui Hospital, Xi'an Jiao Tong University Health Science Center, 555 Youyi East Road, Xi'an, Shaanxi 710068, PR China
| | - Chao Lu
- Department of Osteonecrosis and Joint Reconstruction, Honghui Hospital, Xi'an Jiao Tong University Health Science Center, 555 Youyi East Road, Xi'an, Shaanxi 710068, PR China
| |
Collapse
|
12
|
Abstract
L1 cell adhesion molecule (L1CAM) is the prototype member of the L1-family of closely related neural adhesion molecules. L1CAM is differentially expressed in the normal nervous system as well as pathological tissues and displays a wide range of biological activities. In human malignancies, L1CAM plays a vital role in tumor growth, invasion and metastasis. Recently, increasing evidence has suggested that L1CAM exerts a variety of functions at different steps of tumor progression through a series of signaling pathways. In addition, L1CAM has been identified as a promising target for cancer therapy by using synthetic and natural inhibitors. In this review, we provide an up-to-date overview of the role of L1CAM involved in cancers and the rationale for L1CAM as a novel molecular target for cancer therapy.
Collapse
Affiliation(s)
- Xinzhe Yu
- a Department of Pancreatic Surgery, Huashan Hospital , Fudan University , Shanghai , China
| | - Feng Yang
- a Department of Pancreatic Surgery, Huashan Hospital , Fudan University , Shanghai , China
| | - De-Liang Fu
- a Department of Pancreatic Surgery, Huashan Hospital , Fudan University , Shanghai , China
| | - Chen Jin
- a Department of Pancreatic Surgery, Huashan Hospital , Fudan University , Shanghai , China
| |
Collapse
|
13
|
Lee SH, Kim DY, Jing F, Kim H, Yun CO, Han DJ, Choi EY. Del-1 overexpression potentiates lung cancer cell proliferation and invasion. Biochem Biophys Res Commun 2015; 468:92-8. [DOI: 10.1016/j.bbrc.2015.10.159] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Accepted: 10/30/2015] [Indexed: 12/26/2022]
|
14
|
Beckham CJ, Olsen J, Yin PN, Wu CH, Ting HJ, Hagen FK, Scosyrev E, Messing EM, Lee YF. Bladder cancer exosomes contain EDIL-3/Del1 and facilitate cancer progression. J Urol 2014; 192:583-92. [PMID: 24530986 DOI: 10.1016/j.juro.2014.02.035] [Citation(s) in RCA: 166] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/11/2014] [Indexed: 02/08/2023]
Abstract
PURPOSE High grade bladder cancer is an extremely aggressive malignancy associated with high rates of morbidity and mortality. Understanding how exosomes may affect bladder cancer progression could reveal novel therapeutic targets. MATERIALS AND METHODS Exosomes derived from human bladder cancer cell lines and the urine of patients with high grade bladder cancer were assessed for the ability to promote cancer progression in standard assays. Exosomes purified from the high grade bladder cancer cell line TCC-SUP and the nonmalignant urothelial cell line SV-HUC were submitted for mass spectrometry analysis. EDIL-3 was identified and selected for further analysis. Western blot was done to determine EDIL-3 levels in urinary exosomes from patients with high grade bladder cancer. shRNA gene knockdown and recombinant EDIL-3 were applied to study EDIL-3 function. RESULTS Exosomes isolated from high grade bladder cancer cells and the urine of patients with high grade bladder cancer promoted angiogenesis and migration of bladder cancer cells and endothelial cells. We silenced EDIL-3 expression and found that shEDIL-3 exosomes did not facilitate angiogenesis, and urothelial and endothelial cell migration. Moreover, exosomes purified from the urine of patients with high grade bladder cancer contained significantly higher EDIL-3 levels than exosomes from the urine of healthy controls. EDIL-3 activated epidermal growth factor receptor signaling while blockade of epidermal growth factor receptor signaling abrogated this EDIL-3 induced bladder cell migration. CONCLUSIONS Exosomes derived from the urine of patients with bladder cancer contains bioactive molecules such as EDIL-3. Identifying these components and their associated oncogenic pathways could lead to novel therapeutic targets and treatment strategies.
Collapse
Affiliation(s)
- Carla J Beckham
- Department of Urology, University of Rochester, Rochester, New York.
| | - Jayme Olsen
- Department of Genetics, University of Rochester, Rochester, New York
| | - Peng-Nien Yin
- Department of Urology, University of Rochester, Rochester, New York
| | - Chia-Hao Wu
- Department of Urology, University of Rochester, Rochester, New York
| | - Huei-Ju Ting
- Department of Biological Science and Technology, National University of Tainan, Taiwan, Republic of China
| | - Fred K Hagen
- Department of Biochemistry and Biophysics Proteomics Center, University of Rochester, Rochester, New York
| | - Emelian Scosyrev
- Department of Urology, University of Rochester, Rochester, New York
| | - Edward M Messing
- Department of Urology, University of Rochester, Rochester, New York
| | - Yi-Fen Lee
- Department of Urology, University of Rochester, Rochester, New York.
| |
Collapse
|
15
|
Hashimoto Y, Tazawa H, Teraishi F, Kojima T, Watanabe Y, Uno F, Yano S, Urata Y, Kagawa S, Fujiwara T. The hTERT promoter enhances the antitumor activity of an oncolytic adenovirus under a hypoxic microenvironment. PLoS One 2012; 7:e39292. [PMID: 22720091 PMCID: PMC3376103 DOI: 10.1371/journal.pone.0039292] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2011] [Accepted: 05/18/2012] [Indexed: 12/11/2022] Open
Abstract
Hypoxia is a microenvironmental factor that contributes to the invasion, progression and metastasis of tumor cells. Hypoxic tumor cells often show more resistance to conventional chemoradiotherapy than normoxic tumor cells, suggesting the requirement of novel antitumor therapies to efficiently eliminate the hypoxic tumor cells. We previously generated a tumor-specific replication-competent oncolytic adenovirus (OBP-301: Telomelysin), in which the human telomerase reverse transcriptase (hTERT) promoter drives viral E1 expression. Since the promoter activity of the hTERT gene has been shown to be upregulated by hypoxia, we hypothesized that, under hypoxic conditions, the antitumor effect of OBP-301 with the hTERT promoter would be more efficient than that of the wild-type adenovirus 5 (Ad5). In this study, we investigated the antitumor effects of OBP-301 and Ad5 against human cancer cells under a normoxic (20% oxygen) or a hypoxic (1% oxygen) condition. Hypoxic condition induced nuclear accumulation of the hypoxia-inducible factor-1α and upregulation of hTERT promoter activity in human cancer cells. The cytopathic activity of OBP-301 was significantly higher than that of Ad5 under hypoxic condition. Consistent with their cytopathic activity, the replication of OBP-301 was significantly higher than that of Ad5 under the hypoxic condition. OBP-301-mediated E1A was expressed within hypoxic areas of human xenograft tumors in mice. These results suggest that the cytopathic activity of OBP-301 against hypoxic tumor cells is mediated through hypoxia-mediated activation of the hTERT promoter. Regulation of oncolytic adenoviruses by the hTERT promoter is a promising antitumor strategy, not only for induction of tumor-specific oncolysis, but also for efficient elimination of hypoxic tumor cells.
Collapse
Affiliation(s)
- Yuuri Hashimoto
- Department of Gastroenterological Surgery, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Hiroshi Tazawa
- Department of Gastroenterological Surgery, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
- Center for Gene and Cell Therapy, Okayama University Hospital, Okayama, Japan
| | - Fuminori Teraishi
- Department of Gastroenterological Surgery, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Toru Kojima
- Department of Gastroenterological Surgery, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Yuichi Watanabe
- Department of Gastroenterological Surgery, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Futoshi Uno
- Department of Gastroenterological Surgery, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Shuya Yano
- Department of Gastroenterological Surgery, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | | | - Shunsuke Kagawa
- Department of Gastroenterological Surgery, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Toshiyoshi Fujiwara
- Department of Gastroenterological Surgery, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
- * E-mail:
| |
Collapse
|
16
|
Nixon B, Mitchell LA, Anderson AL, Mclaughlin EA, O'bryan MK, Aitken RJ. Proteomic and functional analysis of human sperm detergent resistant membranes. J Cell Physiol 2011; 226:2651-65. [DOI: 10.1002/jcp.22615] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
17
|
Tang NH, Chen YL, Wang XQ, Li XJ, Wu Y, Zou QL, Chen YZ. N-terminal and C-terminal heparin-binding domain polypeptides derived from fibronectin reduce adhesion and invasion of liver cancer cells. BMC Cancer 2010; 10:552. [PMID: 20939933 PMCID: PMC2965728 DOI: 10.1186/1471-2407-10-552] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2009] [Accepted: 10/13/2010] [Indexed: 12/01/2022] Open
Abstract
Background Fibronectin (FN) is known to be a large multifunction glycoprotein with binding sites for many substances, including N-terminal and C-terminal heparin-binding domains. We investigated the effects of highly purified rhFNHN29 and rhFNHC36 polypeptides originally cloned from the two heparin-binding domains on the adhesion and invasion of highly metastatic human hepatocellular carcinoma cells (MHCC97H) and analyzed the underlying mechanism involved. Methods The MHCC97H cells that adhered to FN in the presence of various concentrations of rhFNHN29 and rhFNHC36 polypeptides were stained with crystal violet and measured, and the effects of rhFNHN29 and rhFNHC36 on the invasion of the MHCC97H cells were then detected using the Matrigel invasion assay as well as a lung-metastasis mouse model. The expression level of integrins and focal adhesion kinase (FAK) phosphotyrosyl protein was examined by Western blot, and the activity of matrix metalloproteinases (MMPs) and activator protein 1 (AP-1) was analyzed by gelatin zymography and the electrophoretic mobility band-shift assay (EMSA), respectively. Results Both of the polypeptides rhFNHN29 and rhFNHC36 inhibited adhesion and invasion of MHCC97H cells; however, rhFNHC36 exhibited inhibition at a lower dose than rhFNHN29. These inhibitory effects were mediated by integrin αvβ3 and reversed by a protein tyrosine phosphatase inhibitor. Polypeptides rhFNHN29 and rhFNHC36 abrogated the tyrosine phosphorylation of focal adhesion kinase (p-FAK) and activation of activator protein 1 (AP-1), resulting in the decrease of integrin αv, β3 and β1 expression as well as the reduction of MMP-9 activity. Conclusions Polypeptides rhFNHN29 and rhFNHC36 could potentially be applicable to human liver cancer as anti-adhesive and anti-invasive agents.
Collapse
Affiliation(s)
- Nan-Hong Tang
- Fujian Institute of Hepatobiliary Surgery, Union Hospital, Fujian Medical University, Fuzhou, China
| | | | | | | | | | | | | |
Collapse
|
18
|
Sun JC, Liang XT, Pan K, Wang H, Zhao JJ, Li JJ, Ma HQ, Chen YB, Xia JC. High expression level of EDIL3 in HCC predicts poor prognosis of HCC patients. World J Gastroenterol 2010; 16:4611-5. [PMID: 20857535 PMCID: PMC2945496 DOI: 10.3748/wjg.v16.i36.4611] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To determine the role of epidermal growth factor-like repeats and discoidin I-like domains 3 (EDIL3) in pathogenesis of hepatocellular carcinoma (HCC) by investigating the EDIL3 expression in HCC and its prognostic value for HCC.
METHODS: EDIL3 expression was detected in 101 HCC surgical tissue samples with immunohistochemistry method, and its relation with clinicopathologic features and prognosis of HCC patients was analyzed.
RESULTS: EDIL3 was highly expressed in 48.5% of the HCC patients. Although the EDIL3 expression level did not correlate with any clinicopathological parameters, Kaplan-Meier survival analysis showed that high expression level of EDIL3 resulted in a significantly poor prognosis of HCC patients (log-rank test, P = 0.010). Multivariate Cox’s analysis showed that the EDIL3 expression level was a significant and independent prognostic parameter for the overall survival rate of HCC patients (hazard ratio = 1.978, 95% confidence interval = 1.139-3.435, P = 0.015).
CONCLUSION: High expression level of EDIL3 predicts poor prognosis of HCC patients. EDIL3 may be a potential target of antiangiogenic therapy for HCC.
Collapse
|
19
|
|
20
|
Spano D, Russo R, Di Maso V, Rosso N, Terracciano LM, Roncalli M, Tornillo L, Capasso M, Tiribelli C, Iolascon A. Galectin-1 and its involvement in hepatocellular carcinoma aggressiveness. Mol Med 2009; 16:102-15. [PMID: 20200618 DOI: 10.2119/molmed.2009.00119] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2009] [Accepted: 12/13/2009] [Indexed: 12/13/2022] Open
Abstract
Hepatocellular carcinoma is one of the most common cancers worldwide. Despite several efforts to elucidate hepatocellular carcinoma molecular pathogenesis, it is still not fully understood. To acquire further insights into the molecular mechanisms of hepatocellular carcinoma, we performed a systematic functional genomic approach on human HuH-7 and JHH-6 cells. The subsequent analysis of the differentially expressed genes in human specimens revealed a molecular signature of 11 genes from which we selected the LGALS1 gene, which was overexpressed in hepatocellular carcinoma. The expression analysis in humans of Galectin-1 (Gal-1), the protein encoded by LGALS1, showed a Gal-1 preferential accumulation in the stromal tissue around hepatocellular carcinoma tumors. Moreover, a significant association between increased expression of Gal-1 in hepatocellular carcinoma and the presence of metastasis was observed. Interestingly, Gal-1 overexpression resulted in an increase of cell migration and invasion. In conclusion, this study provides a portfolio of targets useful for future investigations into molecular marker-discovery studies on a large number of patients and functional assays. In addition, our data provide evidence that Gal-1 plays a role in hepatocellular carcinoma cell migration and invasion, and we suggest that further studies should be conducted to fully establish the role of Gal-1 in hepatocellular carcinoma pathogenesis and evaluate Gal-1 as a potential molecular therapeutic target.
Collapse
|
21
|
Zhao ZS, Wang YY, Ye ZY, Tao HQ. Prognostic value of tumor-related molecular expression in gastric carcinoma. Pathol Oncol Res 2009; 15:589-96. [PMID: 19294533 DOI: 10.1007/s12253-009-9158-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2008] [Accepted: 02/17/2009] [Indexed: 01/22/2023]
Abstract
In order to identify reliable molecular markers for prognostic prediction in gastric carcinoma, we evaluated the expression of six molecular markers, namely bFGF, IGF-2, HGF, MMP-9, integrin beta3 and uPA in gastric cancer. There was a significant correlation between the expression of these markers and the depth of tumor invasion, vessel invasion, lymph node and distant metastasis, TNM stage and microvessel density. The average survival time and 5-year survival rate of patients with positive expression of molecular markers was higher than those with negative expression. Multivariate analysis showed that abnormal expression of bFGF, MMP-9 and uPA, as well as depth of invasion, lymph node and distant metastasis and TNM stage were independently related to poor prognosis of gastric cancer. MMP-9, bFGF and uPA are potential candidates for development as clinically applicable molecular prognostic markers for gastric carcinoma, and may be effective therapeutic targets for the disease in the future.
Collapse
Affiliation(s)
- Zhong-Sheng Zhao
- Department of Pathology, Zhejiang Provincal People's Hospital, 158 shangtang road, 310014, Hangzhou, China.
| | | | | | | |
Collapse
|
22
|
Shah GV, Muralidharan A, Thomas S, Gokulgandhi M, Mudit M, Khanfar M, El Sayed K. Identification of a small molecule class to enhance cell-cell adhesion and attenuate prostate tumor growth and metastasis. Mol Cancer Ther 2009; 8:509-20. [PMID: 19276166 DOI: 10.1158/1535-7163.mct-08-0693] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Expression of calcitonin (CT) and its receptor (CTR) is elevated in advanced prostate cancer, and activated CT-CTR autocrine axis plays a pivotal role in tumorigenicity and metastatic potential of multiple prostate cancer cell lines. Recent studies suggest that CT promotes prostate cancer metastasis by reducing cell-cell adhesion through the disassembly of tight and adherens junctions and activation of beta-catenin signaling. We attempted to identify a class of molecules that enhances cell-cell adhesion of prostate cells and reverses the disruptive actions of CT on tight and adherens junctions. Screening several compounds led to the emergence of phenyl-methylene hydantoin (PMH) as a lead candidate that can augment cell-cell adhesion and abolish disruptive actions of CT on junctional complexes. PMH reduced invasiveness of PC-3M cells and abolished proinvasive actions of CT. Importantly, PMH did not display significant cytotoxicity on PC-3M cells at the tested doses. I.p. administered PMH and its S-ethyl derivative remarkably decreased orthotopic tumor growth and inhibited the formation of tumor micrometastases in distant organs of nude mice. PMH treatment also reduced the growth of spontaneous tumors in LPB-Tag mice to a significant extent without any obvious cytotoxic effects. By virtue of its ability to stabilize cell junctions, PMH could reverse the effect of CT on junctional disruption and metastasis, which strengthens the possibility of using PMH as a potential drug candidate for CT-positive androgen-independent prostate cancers.
Collapse
Affiliation(s)
- Girish V Shah
- Department of Pharmaceutical Sciences, University of Louisiana College of Pharmacy, 1800 Bienville Drive, Monroe, LA 71209, USA.
| | | | | | | | | | | | | |
Collapse
|
23
|
Zou X, Qiao H, Jiang X, Dong X, Jiang H, Sun X. Downregulation of developmentally regulated endothelial cell locus-1 inhibits the growth of colon cancer. J Biomed Sci 2008; 16:33. [PMID: 19292890 PMCID: PMC2666667 DOI: 10.1186/1423-0127-16-33] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2008] [Accepted: 10/16/2008] [Indexed: 11/13/2022] Open
Abstract
Developmentally regulated endothelial cell locus-1 (Del1) is an embryonic angiogenic factor expressed in early embryonic endothelial cells, but recently has been found to be expressed in some forms of cancers including colon and breast cancers, and melanoma, and human cancer cell lines. Overexpression of Del1 accelerates tumor growth by enhancing vascular formation, implying Del1 may be a potential target for anti-angiogenic cancer therapy. The study aims to investigate whether downregulation of Del1 could inhibit the growth of tumors established in nude Balb/c mice by subcutaneous implantation of human LS-174T colon cancer cells. The shRNA expression vectors targeting human Del1, and vascular endothelial growth factor (VEGF) were constructed. Gene transfection of Del1-shRNA downregulated expression of Del1 in LS-174T cells in vivo and in vitro, but did not alter the proliferative or survival properties of cells in vitro. Gene transfection of VEGF-shRNA downregulated expression of both VEGF and Del1 in LS-174T cells in vivo and in vitro. Both Del1-shRNA and VEGF-shRNA gene therapies exhibited anti-tumor activities and they also showed a synergistic effect in suppressing growth of colon tumors by anti-angiogenesis and anti-proliferation. Although further investigation to clarify the mechanisms explaining the role of Del1 in tumor growth, and the interaction between VEGF and Del1, is required, the results indicate that downregulation of Del1 presents a potent therapeutic strategy to combat colon cancer.
Collapse
Affiliation(s)
- Xiaolong Zou
- The Hepatosplenic Surgery Center, Department of General Surgery, The First Clinical College of Harbin Medical University, Harbin 150001, PR China
| | - Haiquan Qiao
- The Hepatosplenic Surgery Center, Department of General Surgery, The First Clinical College of Harbin Medical University, Harbin 150001, PR China
| | - Xian Jiang
- The Hepatosplenic Surgery Center, Department of General Surgery, The First Clinical College of Harbin Medical University, Harbin 150001, PR China
| | - Xuesong Dong
- The Hepatosplenic Surgery Center, Department of General Surgery, The First Clinical College of Harbin Medical University, Harbin 150001, PR China
| | - Hongchi Jiang
- The Hepatosplenic Surgery Center, Department of General Surgery, The First Clinical College of Harbin Medical University, Harbin 150001, PR China
| | - Xueying Sun
- The Hepatosplenic Surgery Center, Department of General Surgery, The First Clinical College of Harbin Medical University, Harbin 150001, PR China
- Department of Molecular Medicine & Pathology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| |
Collapse
|
24
|
Abstract
Integrins are a class of cell adhesion molecules that regulate interactions between cells and their extracellular matrix (ECM). Several specific integrin receptors have been identified in intervertebral discs, including the fibronectin-binding integrin receptors alpha(5) beta(1), alpha(v) beta(3) and the collagen-binding integrin receptors alpha(1) beta(1), alpha(2) beta(1), and, alpha(v) beta(1). But the integrins expressions in degenerated intervertebral discs are still unknown. In our study, the expressions of alpha(1), alpha(2), alpha(5), alpha(v), beta(1), beta(3) integrin subunits, collagens, and fibronectin in normal and herniated intervertebral discs of human were determined. Specimens of human lumbar intervertebral discs were divided into 3 groups: normal discs (n = 10), protrusion (n = 15), and extrusion (n = 15). Real-time quantitative reverse transcription polymerase chain reaction (RT-PCR) and immunoprecipitation were used to evaluate the alpha(1), alpha(2), alpha(5),alpha(v), beta(1), and beta(3) integrin subunits messenger ribonucleic acid (mRNA) and protein expressions. RT-PCR was also performed to measure the mRNA level of collagen I, collagen II, and fibronectin. The expressions of alpha(5) and beta(1) subunits were increased in herniated discs, especially in the discs of extrusion. But as to alpha(1), alpha(2), alpha(v) and beta(3), their expressions had no difference among the discs. Fibronectin, whose binding integrin receptor was alpha(5) beta(1) was also increased. And in herniated discs, the collagen I was increased, but the collagen II was decreased. The expressions of some integrin subunits were increased in herniated discs. These results may be attributed to the interaction between cells and the ECM in the process of degeneration.
Collapse
Affiliation(s)
- Maosheng Xia
- Department of Orthopaedics, The First Hospital of China Medical University, Shenyang, PR China
| | | |
Collapse
|