1
|
Haidar Ahmad S, El Baba R, Herbein G. Polyploid giant cancer cells, cytokines and cytomegalovirus in breast cancer progression. Cancer Cell Int 2023; 23:119. [PMID: 37340387 DOI: 10.1186/s12935-023-02971-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Accepted: 06/12/2023] [Indexed: 06/22/2023] Open
Abstract
BACKGROUND Breast cancer is the most common cancer among women. Accumulated evidence over the past decades indicates a very high prevalence of human cytomegalovirus (HCMV) in breast cancer. High-risk HCMV strains possess a direct oncogenic effect displayed by cellular stress, polyploid giant cancer cells (PGCCs) generation, stemness, and epithelial-to-mesenchymal transition (EMT) leading to cancer of aggressive phenotype. Breast cancer development and progression have been regulated by several cytokines where the latter can promote cancer cell survival, help in tumor immune evasion, and initiate the EMT process, thereby resulting in invasion, angiogenesis, and breast cancer metastasis. In the present study, we screened cytokines expression in cytomegalovirus-transformed HMECs (CTH cells) cultures infected with HCMV high-risk strains namely, HCMV-DB and BL, as well as breast cancer biopsies, and analyzed the association between cytokines production, PGCCs count, and HCMV presence in vitro and in vivo. METHODS In CTH cultures and breast cancer biopsies, HCMV load was quantified by real-time qPCR. PGCCs count in CTH cultures and breast cancer biopsies was identified based on cell morphology and hematoxylin and eosin staining, respectively. CTH supernatants were evaluated for the production of TGF-β, IL-6, IL1-β, and IL-10 by ELISA assays. The above-mentioned cytokines expression was assessed in breast cancer biopsies using reverse transcription-qPCR. The correlation analyses were performed using Pearson correlation test. RESULTS The revealed PGCCs/cytokine profile in our in vitro CTH model matched that of the breast cancer biopsies, in vivo. Pronounced cytokine expression and PGCCs count were detected in particularly CTH-DB cultures and basal-like breast cancer biopsies. CONCLUSIONS The analysis of cytokine profiles in PGCCs present mostly in basal-like breast cancer biopsies and derived from CTH cells chronically infected with the high-risk HCMV strains might have the potential to provide novel therapies such as cytokine-based immunotherapy which is a promising field in cancer treatments.
Collapse
Affiliation(s)
- Sandy Haidar Ahmad
- Department Pathogens and Inflammation-EPILAB, EA4266, University of France-Comté, 16 Route de Gray, 25030, Besançon Cedex, France
| | - Ranim El Baba
- Department Pathogens and Inflammation-EPILAB, EA4266, University of France-Comté, 16 Route de Gray, 25030, Besançon Cedex, France
| | - Georges Herbein
- Department Pathogens and Inflammation-EPILAB, EA4266, University of France-Comté, 16 Route de Gray, 25030, Besançon Cedex, France.
- Department of Virology, CHRU Besancon, Besancon, France.
| |
Collapse
|
2
|
Li CY, Anuraga G, Chang CP, Weng TY, Hsu HP, Ta HDK, Su PF, Chiu PH, Yang SJ, Chen FW, Ye PH, Wang CY, Lai MD. Repurposing nitric oxide donating drugs in cancer therapy through immune modulation. J Exp Clin Cancer Res 2023; 42:22. [PMID: 36639681 PMCID: PMC9840268 DOI: 10.1186/s13046-022-02590-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 12/29/2022] [Indexed: 05/30/2023] Open
Abstract
BACKGROUND Nitric oxide-releasing drugs are used for cardiovascular diseases; however, their effects on the tumor immune microenvironment are less clear. Therefore, this study explored the impact of nitric oxide donors on tumor progression in immune-competent mice. METHODS The effects of three different nitric oxide-releasing compounds (SNAP, SNP, and ISMN) on tumor growth were studied in tumor-bearing mouse models. Three mouse tumor models were used: B16F1 melanoma and LL2 lung carcinoma in C57BL/6 mice, CT26 colon cancer in BALB/c mice, and LL2 lung carcinoma in NOD/SCID mice. After nitric oxide treatment, splenic cytokines and lymphocytes were analyzed by cytokine array and flow cytometry, and tumor-infiltrating lymphocytes in the TME were analyzed using flow cytometry and single-cell RNA sequencing. RESULTS Low doses of three exogenous nitric oxide donors inhibited tumor growth in two immunocompetent mouse models but not in NOD/SCID immunodeficient mice. Low-dose nitric oxide donors increase the levels of splenic cytokines IFN-γ and TNF-α but decrease the levels of cytokines IL-6 and IL-10, suggesting an alteration in Th2 cells. Nitric oxide donors increased the number of CD8+ T cells with activation gene signatures, as indicated by single-cell RNA sequencing. Flow cytometry analysis confirmed an increase in infiltrating CD8+ T cells and dendritic cells. The antitumor effect of nitric oxide donors was abolished by depletion of CD8+ T cells, indicating the requirement for CD8+ T cells. Tumor inhibition correlated with a decrease in a subtype of protumor macrophages and an increase in a subset of Arg1-positive macrophages expressing antitumor gene signatures. The increase in this subset of macrophages was confirmed by flow cytometry analysis. Finally, the combination of low-dose nitric oxide donor and cisplatin induced an additive cancer therapeutic effect in two immunocompetent animal models. The enhanced therapeutic effect was accompanied by an increase in the cells expressing the gene signature of NK cell. CONCLUSIONS Low concentrations of exogenous nitric oxide donors inhibit tumor growth in vivo by regulating T cells and macrophages. CD8+ T cells are essential for antitumor effects. In addition, low-dose nitric oxide donors may be combined with chemotherapeutic drugs in cancer therapy in the future.
Collapse
Affiliation(s)
- Chung-Yen Li
- College of Medicine, Institute of basic medical sciences, National Cheng Kung University, Tainan, Taiwan, ROC
| | - Gangga Anuraga
- PhD Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University and Academia Sinica, Taipei, Taiwan, ROC
- Department of Statistics, Faculty of Science and Technology, Universitas PGRI Adi Buana, Surabaya, Indonesia
| | - Chih-Peng Chang
- College of Medicine, Institute of basic medical sciences, National Cheng Kung University, Tainan, Taiwan, ROC
- Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC
| | - Tzu-Yang Weng
- College of Medicine, Institute of basic medical sciences, National Cheng Kung University, Tainan, Taiwan, ROC
| | - Hui-Ping Hsu
- Department of Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC
| | - Hoang Dang Khoa Ta
- PhD Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University and Academia Sinica, Taipei, Taiwan, ROC
| | - Pei-Fang Su
- Department of Statistics, National Cheng Kung University, Tainan, Taiwan, ROC
| | - Pin-Hsuan Chiu
- The Center for Quantitative Sciences, Clinical Medicine Research Center, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC
| | - Shiang-Jie Yang
- College of Medicine, Institute of basic medical sciences, National Cheng Kung University, Tainan, Taiwan, ROC
| | - Feng-Wei Chen
- College of Medicine, Institute of basic medical sciences, National Cheng Kung University, Tainan, Taiwan, ROC
| | - Pei-Hsuan Ye
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC
| | - Chih-Yang Wang
- PhD Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University and Academia Sinica, Taipei, Taiwan, ROC.
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan.
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan.
| | - Ming-Derg Lai
- College of Medicine, Institute of basic medical sciences, National Cheng Kung University, Tainan, Taiwan, ROC.
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC.
| |
Collapse
|
3
|
Cui G, Li G, Pang Z, Florholmen J, Goll R. The presentation and regulation of the IL-8 network in the epithelial cancer stem-like cell niche in patients with colorectal cancer. Biomed Pharmacother 2022; 152:113252. [PMID: 35687912 DOI: 10.1016/j.biopha.2022.113252] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 06/02/2022] [Accepted: 06/02/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Accumulative evidence suggests that the biological behavior of cancer stem-like cells (CSCs) is regulated by their surrounding niche, in which cytokines function as one of the main mediators for the interaction between CSCs and their microenvironment in the colorectal cancer (CRC). METHODS We characterized the presentation of CSCs and the interleukin (IL)- 8 network in the adenoma/CRC epithelium using quantitative real-time PCR (q-PCR), immunohistochemistry (IHC) and double immunofluorescence. In addition, the capacity of IL-1β to stimulate epithelial IL-8 production in colon cancer Caco-2 cells was examined in vitro and the IL-8 product was measured by enzyme-linked immunosorbent assay (ELISA). RESULTS IHC observation showed increased expression of both CSCs and IL-8 in the adenoma and CRC epithelium, and q-PCR results revealed that increased expression of IL-1β transcript was strongly correlated with increased IL-8 transcript levels in both adenoma and CRC tissues. Double immunofluorescence images demonstrated the coexpression of the IL-8 receptors IL-8RA and IL-8RB with LGR5 labeled CSCs in CRC tissue sections. Consistently, in vitro experiments showed that coculture of Caco-2 cells with IL-1β at concentrations of 1, 5, 10 and 20 ng/ml resulted in a dose-dependent release of IL-8, which could be specifically inhibited by cotreatment with the IL-1β receptor antagonist. CONCLUSIONS These results demonstrate activation of the IL-8 network in the niche of CSCs from the precancerous adenoma stage to the CRC stage, which is potentially stimulated by IL-1β in CRC cells.
Collapse
Affiliation(s)
- Guanglin Cui
- Research Group of Gastrointestinal Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China; Faculty of Health Science, Nord University, Campus Levanger, Levanger, Norway.
| | - Gui Li
- Research Group of Gastrointestinal Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhigang Pang
- Research Group of Gastrointestinal Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jon Florholmen
- Department of Gastroenterology, University Hospital of North Norway, University of Tromsø, Tromsø, Norway
| | - Rasmus Goll
- Department of Gastroenterology, University Hospital of North Norway, University of Tromsø, Tromsø, Norway
| |
Collapse
|
4
|
Espinosa Gonzalez M, Volk-Draper L, Bhattarai N, Wilber A, Ran S. Th2 cytokines IL-4, IL-13, and IL-10 promote differentiation of pro-lymphatic progenitors derived from bone marrow myeloid precursors. Stem Cells Dev 2022; 31:322-333. [PMID: 35442077 PMCID: PMC9232236 DOI: 10.1089/scd.2022.0004] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Myeloid-lymphatic endothelial cell progenitors (M-LECP) are a subset of bone marrow (BM)-derived cells characterized by expression of M2-type macrophage markers. We previously showed significant contribution of M-LECP to tumor lymphatic formation and metastasis in human clinical breast tumors and corresponding mouse models. Since M2-type is induced in macrophages by immunosuppressive Th2 cytokines IL-4, IL-13, and IL-10, we hypothesized that these factors might promote pro-lymphatic specification of M-LECP during their differentiation from BM myeloid precursors. To test this hypothesis, we analyzed expression of Th2 cytokines and their receptors in mouse BM cells under conditions leading to M-LECP differentiation, namely, CSF-1 treatment followed by activation of TLR4. We found that under these conditions, all three Th2 receptors were strongly upregulated in >95% of the cells that also secrete endogenous IL-10 but not IL-4 or IL-13 ligands. However, addition of any of the Th2 factors to CSF-1 primed cells significantly increased generation of myeloid-lymphatic progenitors as indicated by co-induction of lymphatic-specific (e.g., Lyve-1, integrin-a9, collectin-12, and stabilin-1) and M2-type markers (e.g., CD163, CD204, CD206, and PD-L1). Antibody-mediated blockade of either IL-10 receptor (IL-10R) or IL-10 ligand significantly reduced both immunosuppressive and lymphatic phenotypes. Moreover, tumor-recruited Lyve-1+ lymphatic progenitors in vivo expressed all Th2 receptors as well as corresponding ligands including IL-4 and IL-13 that were absent in BM cells. This study presents original evidence for the significant role of Th2 cytokines in co-development of immunosuppressive and lymphatic phenotypes in tumor-recruited M2-type myeloid cells. Progenitor-mediated increase in lymphatic vessels can enhance immunosuppression by physical removal of stimulatory immune cells. Thus, targeting Th2 pathways might simultaneously relieve immunosuppression and inhibit differentiation of pro-lymphatic progenitors that ultimately promote tumor spread.
Collapse
Affiliation(s)
- Maria Espinosa Gonzalez
- Southern Illinois University School of Medicine, 12249, Medical Microbiology, Immunology and Cell Biology, Springfield, Illinois, United States;
| | - Lisa Volk-Draper
- Southern Illinois University School of Medicine, 12249, Medical Microbiology, Immunology and Cell Biology, Springfield, Illinois, United States;
| | - Nihit Bhattarai
- Southern Illinois University School of Medicine, 12249, Medical Microbiology, Immunology and Cell Biology, Springfield, Illinois, United States;
| | - Andrew Wilber
- Southern Illinois University School of Medicine, Medical Microbiology, Immunology and Cell Biology, Springfield, Illinois, United States;
| | - Sophia Ran
- Southern Illinois University School of Medicine, 12249, Medical Microbiology, Immunology and Cell Biology, 801 N. Rutledge, P.O. Box 19626, Springfield, Illinois, United States, 62794;
| |
Collapse
|
5
|
Ebersbach C, Beier AMK, Hönscheid P, Sperling C, Jöhrens K, Baretton GB, Thomas C, Sommer U, Borkowetz A, Erb HHH. Influence of Systemic Therapy on the Expression and Activity of Selected STAT Proteins in Prostate Cancer Tissue. Life (Basel) 2022; 12:life12020240. [PMID: 35207527 PMCID: PMC8877682 DOI: 10.3390/life12020240] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 02/01/2022] [Accepted: 02/03/2022] [Indexed: 01/11/2023] Open
Abstract
Signal Transducer and Activator of Transcription (STAT) proteins have been identified as drivers of prostate cancer (PCa) progression and development of aggressive castration-resistant phenotypes. In particular, STAT3, 5, and 6 have been linked to resistance to androgen receptor inhibition and metastasis in in vitro and in vivo models. This descriptive study aimed to validate these preclinical data in tissue obtained from patients with PCa before and while under androgen-deprivation therapy. Therefore, STAT3, 5, and 6 expressions and activity were assessed by immunohistochemistry. The data revealed that STAT3 and 5 changed in PCa. However, there was no relationship between expression and survival. Moreover, due to the heterogeneous nature of PCa, the preclinical results could not be transferred congruently to the patient’s material. A pilot study with a longitudinal patient cohort could also show this heterogeneous influence of systemic therapy on STAT3, 5, and 6 expressions and activity. Even if the main mechanisms were validated, these data demonstrate the urge for better patient-near preclinical models. Therefore, these data reflect the need for investigations of STAT proteins in a longitudinal patient cohort to identify factors responsible for the diverse influence of system therapy on STAT expression.
Collapse
Affiliation(s)
- Celina Ebersbach
- Department of Urology, Technische Universität Dresden, 01307 Dresden, Germany; (C.E.); (A.-M.K.B.); (C.T.); (A.B.)
- Mildred Scheel Early Career Center, Department of Urology, Medical Faculty and University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Alicia-Marie K. Beier
- Department of Urology, Technische Universität Dresden, 01307 Dresden, Germany; (C.E.); (A.-M.K.B.); (C.T.); (A.B.)
- Mildred Scheel Early Career Center, Department of Urology, Medical Faculty and University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Pia Hönscheid
- Institute of Pathology, Universitätsklinikum Carl Gustav Carus Dresden, 01307 Dresden, Germany; (P.H.); (C.S.); (K.J.); (G.B.B.); (U.S.)
- National Center for Tumor Diseases Partner Site Dresden and German Cancer Center Heidelberg, 69120 Heidelberg, Germany
| | - Christian Sperling
- Institute of Pathology, Universitätsklinikum Carl Gustav Carus Dresden, 01307 Dresden, Germany; (P.H.); (C.S.); (K.J.); (G.B.B.); (U.S.)
- National Center for Tumor Diseases Partner Site Dresden and German Cancer Center Heidelberg, 69120 Heidelberg, Germany
| | - Korinna Jöhrens
- Institute of Pathology, Universitätsklinikum Carl Gustav Carus Dresden, 01307 Dresden, Germany; (P.H.); (C.S.); (K.J.); (G.B.B.); (U.S.)
- Tumor and Normal Tissue Bank of the University Cancer Center (UCC), University Hospital and Faculty of Medicine, Technische Universität Dresden, 01307 Dresden, Germany
| | - Gustavo B. Baretton
- Institute of Pathology, Universitätsklinikum Carl Gustav Carus Dresden, 01307 Dresden, Germany; (P.H.); (C.S.); (K.J.); (G.B.B.); (U.S.)
- National Center for Tumor Diseases Partner Site Dresden and German Cancer Center Heidelberg, 69120 Heidelberg, Germany
- Tumor and Normal Tissue Bank of the University Cancer Center (UCC), University Hospital and Faculty of Medicine, Technische Universität Dresden, 01307 Dresden, Germany
| | - Christian Thomas
- Department of Urology, Technische Universität Dresden, 01307 Dresden, Germany; (C.E.); (A.-M.K.B.); (C.T.); (A.B.)
- National Center for Tumor Diseases Partner Site Dresden and German Cancer Center Heidelberg, 69120 Heidelberg, Germany
| | - Ulrich Sommer
- Institute of Pathology, Universitätsklinikum Carl Gustav Carus Dresden, 01307 Dresden, Germany; (P.H.); (C.S.); (K.J.); (G.B.B.); (U.S.)
- National Center for Tumor Diseases Partner Site Dresden and German Cancer Center Heidelberg, 69120 Heidelberg, Germany
- Tumor and Normal Tissue Bank of the University Cancer Center (UCC), University Hospital and Faculty of Medicine, Technische Universität Dresden, 01307 Dresden, Germany
| | - Angelika Borkowetz
- Department of Urology, Technische Universität Dresden, 01307 Dresden, Germany; (C.E.); (A.-M.K.B.); (C.T.); (A.B.)
| | - Holger H. H. Erb
- Department of Urology, Technische Universität Dresden, 01307 Dresden, Germany; (C.E.); (A.-M.K.B.); (C.T.); (A.B.)
- Correspondence:
| |
Collapse
|
6
|
D’Andrea MR, Cereda V, Coppola L, Giordano G, Remo A, De Santis E. Propensity for Early Metastatic Spread in Breast Cancer: Role of Tumor Vascularization Features and Tumor Immune Infiltrate. Cancers (Basel) 2021; 13:cancers13235917. [PMID: 34885027 PMCID: PMC8657227 DOI: 10.3390/cancers13235917] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Revised: 11/17/2021] [Accepted: 11/22/2021] [Indexed: 02/05/2023] Open
Abstract
Breast cancer is a complex and highly heterogeneous disease consisting of various subtypes. It is classified into human epidermal growth receptor 2 (HER-2)-enriched, luminal A, luminal B and basal-like/triple negative (TNBC) breast cancer, based on histological and molecular features. At present, clinical decision-making in breast cancer is focused only on the assessment of tumor cells; nevertheless, it has been recognized that the tumor microenvironment (TME) plays a critical biologic role in breast cancer. This is constituted by a large group of immune and non-immune cells, but also by non-cellular components, such as several cytokines. TME is deeply involved in angiogenesis, immune-evasion strategies, and propensity for early metastatic spread, impacting on prognosis and prediction of response to specific treatments. In this review, we focused our attention on the early morphological changes of tumor microenvironment (tumor vasculature features, presence of immune and non-immune cells infiltrating the stroma, levels of cytokines) during breast cancer development. At the same time, we correlate these characteristics with early metastatic propensity (defined as synchronous metastasis or early recurrence) with particular attention to breast cancer subtypes.
Collapse
Affiliation(s)
- Mario Rosario D’Andrea
- Clinical Oncology Unit, San Paolo Hospital, Largo Donatori del Sangue 1, Civitavecchia, 00053 Rome, Italy;
| | - Vittore Cereda
- Clinical Oncology Unit, San Paolo Hospital, Largo Donatori del Sangue 1, Civitavecchia, 00053 Rome, Italy;
- Correspondence: ; Tel.: +39-07-6659-1230
| | - Luigi Coppola
- Unit of Anatomy, Pathological Histology and Diagnostic Cytology, Department of Diagnostic and Pharma-Ceutical Services, Sandro Pertini Hospital, 00157 Rome, Italy;
| | - Guido Giordano
- Unit of Medical Oncology and Biomolecular Therapy, Department of Medical and Surgical Sciences, University of Foggia, Policlinico Riuniti, 71122 Foggia, Italy;
| | - Andrea Remo
- Pathology Unit, Mater Salutis Hospital, ULSS9, Legnago, 37045 Verona, Italy;
| | - Elena De Santis
- Department of Anatomical, Histological, Forensic Medicine and Orthopedic Sciences, Sapienza University of Rome, 00185 Rome, Italy;
| |
Collapse
|
7
|
Ibrahim YF, Refaie MM, Kamel MY, Ahmed SM, Moussa RA, Bayoumi AM, Ibrahim MA. Molecular mechanisms underlying the effect of diacerein on trichloroacetic acid-induced hepatic pre-neoplastic lesions in rats. Hum Exp Toxicol 2021; 40:S788-S803. [PMID: 34794354 DOI: 10.1177/09603271211056331] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
CONCLUSION IL-1β mediates angiogenesis indirectly, as it has been shown to induce hypoxia-inducible factor-1α (HIF-1α) which upregulates VEGF.
Collapse
Affiliation(s)
- Yasmine F Ibrahim
- Department of Pharmacology, Faculty of Medicine, 68877Minia University, Minia, Egypt
| | - Marwa Mm Refaie
- Department of Pharmacology, Faculty of Medicine, 68877Minia University, Minia, Egypt
| | - Maha Y Kamel
- Department of Pharmacology, Faculty of Medicine, 68877Minia University, Minia, Egypt
| | - Sara M Ahmed
- Department of Pharmacology, Faculty of Medicine, 68877Minia University, Minia, Egypt
| | - Rabab A Moussa
- Department of Pathology, Faculty of Medicine, 68877Minia University, Minia, Egypt
| | - Asmaa Ma Bayoumi
- Department of Biochemistry, Faculty of Pharmacy, 68877Minia University, Minia, Egypt.,Department of Biochemistry, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Mohamed A Ibrahim
- Department of Pharmacology, Faculty of Medicine, 68877Minia University, Minia, Egypt
| |
Collapse
|
8
|
Erb HHH, Culig Z, Stope MB. IL-4 Counteracts the Cytotoxic Effects of Peripheral Blood Mononuclear Cells on Hormone-sensitive Prostate Cancer Cells. In Vivo 2021; 35:1973-1977. [PMID: 34182471 DOI: 10.21873/invivo.12465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 04/29/2021] [Accepted: 05/18/2021] [Indexed: 11/10/2022]
Abstract
BACKGROUND/AIM Proinflammatory cytokines play an essential role in the development and progression of prostate cancer (PCa). Especially interleukine (IL-)6 is involved in the development of aggressive PCa. Peripheral blood mononuclear cells (PBMC) have been reported to interact with cancer cells and subsequently lead to increased production of pro-inflammatory cytokines. However, the role of anti-nflammatory cytokines, such as IL-4 is still largely unexplored in prostate cancer. In the present study, we investigated the effects of IL-4 on PBMC co-cultured with PCa cells. MATERIALS AND METHODS PBMC were co-culured with the PCa cell lines LNCaP and LNCaP-IL6+. To avoid cell-cell contact, cancer and immune cells were separated using cell culture inserts with a 0.4 μm pore size membrane. Cell growth was assessed using the [3-(4, 5-dimethylthiazol-2-yl)-2, 5 diphenyl tetrazolium bromide] (MTT) assay. Cytokine levels were measured using a BD™Cytometric Bead Array. RESULTS Cell viability of LNCaP cells decreased massively when cells were co-cultured with PBMC. Pre-incubation with IL-4 could partly rescue the observed effect of cell viability of LNCaP cells co-cultured with PBMC. In contrast, cell viability of the LNCaP-IL6+ cell line was not affected when co-cultured with PBMC. CONCLUSION IL-4 counteracts the cytotoxic effects of PBMC on hormone-sensitive LNCaP cells and is involved in the immune escape and development of aggressive phenotypes of PCa.
Collapse
Affiliation(s)
- Holger H H Erb
- Department of Urology, Technische Universität Dresden, Dresden, Germany.,UroFors Consortium (Natural Scientists in Urological Research) of the German Society of Urology, Düsseldorf, Germany
| | - Zoran Culig
- Experimental Urology, Department of Urology, University of Innsbruck, Innsbruck, Austria
| | - Matthias B Stope
- UroFors Consortium (Natural Scientists in Urological Research) of the German Society of Urology, Düsseldorf, Germany; .,Department of Gynecology and Gynecological Oncology, University Hospital Bonn, Bonn, Germany
| |
Collapse
|
9
|
Rey F, Messa L, Pandini C, Launi R, Barzaghini B, Micheletto G, Raimondi MT, Bertoli S, Cereda C, Zuccotti GV, Cancello R, Carelli S. Transcriptome Analysis of Subcutaneous Adipose Tissue from Severely Obese Patients Highlights Deregulation Profiles in Coding and Non-Coding Oncogenes. Int J Mol Sci 2021; 22:1989. [PMID: 33671464 PMCID: PMC7922682 DOI: 10.3390/ijms22041989] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 02/12/2021] [Accepted: 02/16/2021] [Indexed: 02/07/2023] Open
Abstract
Obesity is a major risk factor for a large number of secondary diseases, including cancer. Specific insights into the role of gender differences and secondary comorbidities, such as type 2 diabetes (T2D) and cancer risk, are yet to be fully identified. The aim of this study is thus to find a correlation between the transcriptional deregulation present in the subcutaneous adipose tissue of obese patients and the oncogenic signature present in multiple cancers, in the presence of T2D, and considering gender differences. The subcutaneous adipose tissue (SAT) of five healthy, normal-weight women, five obese women, five obese women with T2D and five obese men were subjected to RNA-sequencing, leading to the identification of deregulated coding and non-coding RNAs, classified for their oncogenic score. A panel of DE RNAs was validated via Real-Time PCR and oncogene expression levels correlated the oncogenes with anthropometrical parameters, highlighting significant trends. For each analyzed condition, we identified the deregulated pathways associated with cancer, the prediction of possible prognosis for different cancer types and the lncRNAs involved in oncogenic networks and tissues. Our results provided a comprehensive characterization of oncogenesis correlation in SAT, providing specific insights into the possible molecular targets implicated in this process. Indeed, the identification of deregulated oncogenes also in SAT highlights hypothetical targets implicated in the increased oncogenic risk in highly obese subjects. These results could shed light on new molecular targets to be specifically modulated in obesity and highlight which cancers should receive the most attention in terms of better prevention in obesity-affected patients.
Collapse
Affiliation(s)
- Federica Rey
- Department of Biomedical and Clinical Sciences “L. Sacco”, School of Medicine, University of Milano, Via Grassi 74, 20157 Milano, Italy; (F.R.); (R.L.); (G.V.Z.)
- Pediatric Clinical Research Centre Fondazione “Romeo ed Enrica Invernizzi”, University of Milano, Via G.B. Grassi 74, 20157 Milano, Italy
| | - Letizia Messa
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta”, Politecnico di Milano, Piazza Leonardo da Vinci 32, 20133 Milano, Italy; (L.M.); (B.B.); (M.T.R.)
| | - Cecilia Pandini
- Genomic and Post-Genomic Centre, IRCCS Mondino Foundation, 27100 Pavia, Italy; (C.P.); (C.C.)
| | - Rossella Launi
- Department of Biomedical and Clinical Sciences “L. Sacco”, School of Medicine, University of Milano, Via Grassi 74, 20157 Milano, Italy; (F.R.); (R.L.); (G.V.Z.)
- Pediatric Clinical Research Centre Fondazione “Romeo ed Enrica Invernizzi”, University of Milano, Via G.B. Grassi 74, 20157 Milano, Italy
| | - Bianca Barzaghini
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta”, Politecnico di Milano, Piazza Leonardo da Vinci 32, 20133 Milano, Italy; (L.M.); (B.B.); (M.T.R.)
| | - Giancarlo Micheletto
- Department of Pathophysiology and Transplantation, INCO and Department of General Surgery, Istituto Clinico Sant’Ambrogio, University of Milan, Via Francesco Sforza 35, 20122 Milano, Italy;
| | - Manuela Teresa Raimondi
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta”, Politecnico di Milano, Piazza Leonardo da Vinci 32, 20133 Milano, Italy; (L.M.); (B.B.); (M.T.R.)
| | - Simona Bertoli
- Obesity Unit—Laboratory of Nutrition and Obesity Research, Department of Endocrine and Metabolic Diseases, IRCCS Istituto Auxologico Italiano, Via Ariosto 9, 20145 Milano, Italy; (S.B.); (R.C.)
- International Center for the Assessment of Nutritional Status (ICANS), Department of Food, Environmental and Nutritional Sciences (DeFENS), University of Milan, Via Celoria 2, 20133 Milano, Italy
| | - Cristina Cereda
- Genomic and Post-Genomic Centre, IRCCS Mondino Foundation, 27100 Pavia, Italy; (C.P.); (C.C.)
| | - Gian Vincenzo Zuccotti
- Department of Biomedical and Clinical Sciences “L. Sacco”, School of Medicine, University of Milano, Via Grassi 74, 20157 Milano, Italy; (F.R.); (R.L.); (G.V.Z.)
- Pediatric Clinical Research Centre Fondazione “Romeo ed Enrica Invernizzi”, University of Milano, Via G.B. Grassi 74, 20157 Milano, Italy
- Department of Pediatrics, Children’s Hospital “V. Buzzi”, Via Lodovico Castelvetro 32, 20154 Milano, Italy
| | - Raffaella Cancello
- Obesity Unit—Laboratory of Nutrition and Obesity Research, Department of Endocrine and Metabolic Diseases, IRCCS Istituto Auxologico Italiano, Via Ariosto 9, 20145 Milano, Italy; (S.B.); (R.C.)
| | - Stephana Carelli
- Department of Biomedical and Clinical Sciences “L. Sacco”, School of Medicine, University of Milano, Via Grassi 74, 20157 Milano, Italy; (F.R.); (R.L.); (G.V.Z.)
- Pediatric Clinical Research Centre Fondazione “Romeo ed Enrica Invernizzi”, University of Milano, Via G.B. Grassi 74, 20157 Milano, Italy
| |
Collapse
|
10
|
Kim K, Sohn YJ, Lee R, Yoo HJ, Kang JY, Choi N, Na D, Yeon JH. Cancer-Associated Fibroblasts Differentiated by Exosomes Isolated from Cancer Cells Promote Cancer Cell Invasion. Int J Mol Sci 2020; 21:ijms21218153. [PMID: 33142759 PMCID: PMC7662577 DOI: 10.3390/ijms21218153] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 10/20/2020] [Accepted: 10/28/2020] [Indexed: 12/12/2022] Open
Abstract
Cancer-associated fibroblasts (CAFs) in the cancer microenvironment play an essential role in metastasis. Differentiation of endothelial cells into CAFs is induced by cancer cell-derived exosomes secreted from cancer cells that transfer molecular signals to surrounding cells. Differentiated CAFs facilitate migration of cancer cells to different regions through promoting extracellular matrix (ECM) modifications. However, in vitro models in which endothelial cells exposed to cancer cell-derived exosomes secreted from various cancer cell types differentiate into CAFs or a microenvironmentally controlled model for investigating cancer cell invasion by CAFs have not yet been studied. In this study, we propose a three-dimensional in vitro cancer cell invasion model for real-time monitoring of the process of forming a cancer invasion site through CAFs induced by exosomes isolated from three types of cancer cell lines. The invasiveness of cancer cells with CAFs induced by cancer cell-derived exosomes (eCAFs) was significantly higher than that of CAFs induced by cancer cells (cCAFs) through physiological and genetic manner. In addition, different genetic tendencies of the invasion process were observed in the process of invading cancer cells according to CAFs. Our 3D microfluidic platform helps to identify specific interactions among multiple factors within the cancer microenvironment and provides a model for cancer drug development.
Collapse
Affiliation(s)
- Kimin Kim
- Department of Integrative Biosciences, University of Brain Education (UBE), Cheonan 31228, Korea; (K.K.); (Y.J.S.); (R.L.); (H.J.Y.)
| | - Yeh Joo Sohn
- Department of Integrative Biosciences, University of Brain Education (UBE), Cheonan 31228, Korea; (K.K.); (Y.J.S.); (R.L.); (H.J.Y.)
| | - Ruri Lee
- Department of Integrative Biosciences, University of Brain Education (UBE), Cheonan 31228, Korea; (K.K.); (Y.J.S.); (R.L.); (H.J.Y.)
| | - Hye Ju Yoo
- Department of Integrative Biosciences, University of Brain Education (UBE), Cheonan 31228, Korea; (K.K.); (Y.J.S.); (R.L.); (H.J.Y.)
| | - Ji Yoon Kang
- Center for BioMicrosystems, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea; (J.Y.K.); (N.C.)
- Division of Bio-Medical Science & Technology (Biomedical Engineering), KIST School, Korea University of Science and Technology (UST), Seoul 02792, Korea
| | - Nakwon Choi
- Center for BioMicrosystems, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea; (J.Y.K.); (N.C.)
- Division of Bio-Medical Science & Technology (Biomedical Engineering), KIST School, Korea University of Science and Technology (UST), Seoul 02792, Korea
| | - Dokyun Na
- Department of Biomedical Engineering, Chung-Ang University, Seoul 06974, Korea
- Correspondence: (D.N.); (J.H.Y.); Tel.: +82-2-820-5690 (D.N.); +82-41-529-2621 (J.H.Y.); Fax: +82-2-814-2651 (D.N.); +82-41-529-2674 (J.H.Y.)
| | - Ju Hun Yeon
- Department of Integrative Biosciences, University of Brain Education (UBE), Cheonan 31228, Korea; (K.K.); (Y.J.S.); (R.L.); (H.J.Y.)
- Correspondence: (D.N.); (J.H.Y.); Tel.: +82-2-820-5690 (D.N.); +82-41-529-2621 (J.H.Y.); Fax: +82-2-814-2651 (D.N.); +82-41-529-2674 (J.H.Y.)
| |
Collapse
|
11
|
Wang J, Liu T, Chen J. Oncolytic Measles Virus Encoding Interleukin-12 Mediated Antitumor Activity and Immunologic Control of Colon Cancer In Vivo and Ex Vivo. Cancer Biother Radiopharm 2020; 36:774-782. [PMID: 32783751 DOI: 10.1089/cbr.2019.3084] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Background: In this study, we used an oncolytic measles virus encoding interleukin 12 (IL-12) to treat colon cancer in vivo and ex vivo to investigate its effect on the viability and apoptosis of colon cancer cells. Method: A rat model was established to evaluate the immunostimulatory capabilities and therapeutic efficacy of vectors encoding an IL-12 fusion protein (MeVac FmIL-12 vectors). TUNEL staining, western blot, and enzyme-linked immunosorbent assay were performed to examine the impacts of MeVac FmIL-12 on the expression of inflammatory cytokines. Cell transfection was carried out to validate the anti-tumor role of MeVac FmIL-12 in vitro. Flow cytometry and MTT assay were performed to assess the effects of MeVac FmIL-12 on cell apoptosis and viability. Result: High concentrations (10-1000 ng/mL) of murine IL-12 fusion protein (FmIL-12) decreased the production of interferon γ (IFN-γ) in a concentration-dependent manner and reflected FmIL-12-induced overstimulation. Rats treated with MeVac vectors encoding FmIL-12 showed a significantly increased level of FmIL-12 overtime and a concentration-dependent (0.01-10 ng/mL) increase in IFN-γ production. MeVac FmIL-12 also increased the expression of inflammatory cytokines (IFN-γ, tumor necrosis factor α, and IL-6) both in vivo and in vitro. MeVac FmIL-12 promoted cell apoptosis and reduced cell viability, which helped to trigger a systemic anti-tumor immune response, both in vivo and in vitro. Conclusion: In this study, we suggested that MeVac FmIL-12 enhanced the therapeutic efficacy of tumor treatment by improving anti-tumor immunity.
Collapse
Affiliation(s)
- Jian Wang
- Department of Clinical Laboratory, The Affiliated Hospital of Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| | - Tao Liu
- Department of Medical Oncology, and The Affiliated Hospital of Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| | - Jie Chen
- Department of Pathology, The Affiliated Hospital of Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| |
Collapse
|
12
|
Cytokines and Chemokines as Mediators of Prostate Cancer Metastasis. Int J Mol Sci 2020; 21:ijms21124449. [PMID: 32585812 PMCID: PMC7352203 DOI: 10.3390/ijms21124449] [Citation(s) in RCA: 104] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 06/19/2020] [Accepted: 06/21/2020] [Indexed: 12/16/2022] Open
Abstract
The consequences of prostate cancer metastasis remain severe, with huge impact on the mortality and overall quality of life of affected patients. Despite the convoluted interplay and cross talk between various cell types and secreted factors in the metastatic process, cytokine and chemokines, along with their receptors and signaling axis, constitute important factors that help drive the sequence of events that lead to metastasis of prostate cancer. These proteins are involved in extracellular matrix remodeling, epithelial-mesenchymal-transition, angiogenesis, tumor invasion, premetastatic niche creation, extravasation, re-establishment of tumor cells in secondary organs as well as the remodeling of the metastatic tumor microenvironment. This review presents an overview of the main cytokines/chemokines, including IL-6, CXCL12, TGFβ, CXCL8, VEGF, RANKL, CCL2, CX3CL1, IL-1, IL-7, CXCL1, and CXCL16, that exert modulatory roles in prostate cancer metastasis. We also provide extensive description of their aberrant expression patterns in both advanced disease states and metastatic sites, as well as their functional involvement in the various stages of the prostate cancer metastatic process.
Collapse
|
13
|
Zhang Y, Fan K, Xu X, Wang A. The TGF-β1 Induces the Endothelial-to-Mesenchymal Transition via the UCA1/miR-455/ZEB1 Regulatory Axis in Human Umbilical Vein Endothelial Cells. DNA Cell Biol 2020; 39:1264-1273. [PMID: 32584608 DOI: 10.1089/dna.2019.5194] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Transforming growth factor-beta 1 (TGF-β1) plays important roles in the endothelial-to-mesenchymal transition (EndMT). Recently, long noncoding RNAs (lncRNAs) have been identified to be involved in the physiological and pathological processes of human diseases. However, the role of endothelial lncRNAs in the TGF-β1-mediated control of angiogenesis and its underlying mechanism remains largely unclear. In this study, we first demonstrated that TGF-β1 induced EndMT; promoted cell viability, proliferation, migration, and tube formation of human umbilical vein endothelial cells (HUVECs). Second, our study displayed that TGF-β1 upregulated the lncRNA UCA1 expression in HUVECs, knocked down UCA1 with small interfering RNAs, and inhibited the function of TGF-β1 in HUVECs. Third, our study showed that UCA1 was located in the cytoplasm and absorbed miR-455 in TGF-β1-treated HUVECs. Further, the miR-455 inhibitor restored the role of the inhibited UCA1 in HUVECs treated with TGF-β1. Fourth, our study revealed that miR-455 inhibited ZEB1 expression, and overexpression of ZEB1 restored the role of miR-455 in HUVECs treated with TGF-β1. Finally, our study revealed that UCA1 exerted its role via regulating the UCA1/miR-455/ZEB1 regulatory axis in HUVECs treated with TGF-β1. Collectively, our study identified the role of the UCA1/miR-455/ZEB1 pathway in HUVECs treated with TGF-β1 and indicated the potential therapeutic role of this regulatory axis in angiogenesis.
Collapse
Affiliation(s)
- Ying Zhang
- Department of Anesthesiology, Shanghai Sixth People's Hospital East Affiliated to Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Kun Fan
- Department of Anesthesiology, Shanghai Sixth People's Hospital East Affiliated to Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Xiaotao Xu
- Department of Anesthesiology, Shanghai Sixth People's Hospital East Affiliated to Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Aizhong Wang
- Department of Anesthesiology, Shanghai Sixth People's Hospital East Affiliated to Shanghai University of Medicine & Health Sciences, Shanghai, China
| |
Collapse
|
14
|
Chen X, Xu C, Hong S, Xia X, Cao Y, McDermott J, Mu Y, Han JDJ. Immune Cell Types and Secreted Factors Contributing to Inflammation-to-Cancer Transition and Immune Therapy Response. Cell Rep 2020; 26:1965-1977.e4. [PMID: 30759403 DOI: 10.1016/j.celrep.2019.01.080] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 09/17/2018] [Accepted: 01/22/2019] [Indexed: 12/23/2022] Open
Abstract
Although chronic inflammation increases many cancers' risk, how inflammation facilitates cancer development is still not well studied. Recognizing whether and when inflamed tissues transition to cancerous tissues is of utmost importance. To unbiasedly infer molecular events, immune cell types, and secreted factors contributing to the inflammation-to-cancer (I2C) transition, we develop a computational package called "SwitchDetector" based on liver, gastric, and colon cancer I2C data. Using it, we identify angiogenesis associated with a common critical transition stage for multiple I2C events. Furthermore, we infer infiltrated immune cell type composition and their secreted or suppressed extracellular proteins to predict expression of important transition stage genes. This identifies extracellular proteins that may serve as early-detection biomarkers for pre-cancer and early-cancer stages. They alone or together with I2C hallmark angiogenesis genes are significantly related to cancer prognosis and can predict immune therapy response. The SwitchDetector and I2C database are publicly available at www.inflammation2cancer.org.
Collapse
Affiliation(s)
- Xingwei Chen
- Key Laboratory of Computational Biology, CAS Center for Excellence in Molecular Cell Science, Collaborative Innovation Center for Genetics and Developmental Biology, Chinese Academy of Sciences-Max Planck Partner Institute for Computational Biology, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Chi Xu
- Key Laboratory of Computational Biology, CAS Center for Excellence in Molecular Cell Science, Collaborative Innovation Center for Genetics and Developmental Biology, Chinese Academy of Sciences-Max Planck Partner Institute for Computational Biology, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shengjun Hong
- Key Laboratory of Computational Biology, CAS Center for Excellence in Molecular Cell Science, Collaborative Innovation Center for Genetics and Developmental Biology, Chinese Academy of Sciences-Max Planck Partner Institute for Computational Biology, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xian Xia
- Key Laboratory of Computational Biology, CAS Center for Excellence in Molecular Cell Science, Collaborative Innovation Center for Genetics and Developmental Biology, Chinese Academy of Sciences-Max Planck Partner Institute for Computational Biology, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yaqiang Cao
- Key Laboratory of Computational Biology, CAS Center for Excellence in Molecular Cell Science, Collaborative Innovation Center for Genetics and Developmental Biology, Chinese Academy of Sciences-Max Planck Partner Institute for Computational Biology, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Joseph McDermott
- Key Laboratory of Computational Biology, CAS Center for Excellence in Molecular Cell Science, Collaborative Innovation Center for Genetics and Developmental Biology, Chinese Academy of Sciences-Max Planck Partner Institute for Computational Biology, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai 200031, China
| | - Yonglin Mu
- Key Laboratory of Computational Biology, CAS Center for Excellence in Molecular Cell Science, Collaborative Innovation Center for Genetics and Developmental Biology, Chinese Academy of Sciences-Max Planck Partner Institute for Computational Biology, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jing-Dong J Han
- Key Laboratory of Computational Biology, CAS Center for Excellence in Molecular Cell Science, Collaborative Innovation Center for Genetics and Developmental Biology, Chinese Academy of Sciences-Max Planck Partner Institute for Computational Biology, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
15
|
Shahid A, Bharadwaj M. The connection between the Th17 cell related cytokines and cancer stem cells in cancer: Novel therapeutic targets. Immunol Lett 2019; 213:9-20. [PMID: 31278971 DOI: 10.1016/j.imlet.2019.07.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 06/25/2019] [Accepted: 07/02/2019] [Indexed: 02/08/2023]
Abstract
Cancer Stem Cells (CSCs) are the subpopulation of cells present in the different types of cancers with capabilities of self-renewal, differentiation, and tumorigenicity when transplanted into an animal host. The research work on the CSC has been providing a promising approach for the improvement of cancer therapies in the future. The CSCs have a close connection with the cytokines related with the T helper 17 (Th17) cell and other factors present in the tumor microenvironment, and these play a pivotal role in tumor progression and metastasis. The properties of CSCs are well defined in various type of tumor which is mainly developed by chemically and spontaneously in murine cancer model but in human defined primarily on acute myeloid leukemia, glioma, and breast cancer. The role of Th1, Th2, Natural Killer cells are well described in the cancer biology, but the Th17 cells are the subset which is recently exploited, and lots of research are going on. In this Review, we summarize current findings of the characteristics and functions of the Th17 cell and its signature cytokines in different cancers and their interconnections with cancer stem cells and with their markers. We have also discussed the functional properties of CSCs and how the CSCs markers can be distinguished from normal stem cells markers. We have also talked about the strategies that are efficiently targeting of CSCs and Th17 cells in different cancers.
Collapse
Affiliation(s)
- Ayaz Shahid
- Molecular Biology Group, National Institute of Cancer Prevention and Research, Indian Council of Medical Research (ICMR), Department of Health Research, Noida, 201301, India
| | - Mausumi Bharadwaj
- Molecular Biology Group, National Institute of Cancer Prevention and Research, Indian Council of Medical Research (ICMR), Department of Health Research, Noida, 201301, India.
| |
Collapse
|
16
|
Cohen EN, Fouad TM, Lee BN, Arun BK, Liu D, Tin S, Gutierrez Barrera AM, Miura T, Kiyokawa I, Yamashita J, Alvarez RH, Valero V, Woodward WA, Shen Y, Ueno NT, Cristofanilli M, Reuben JM. Elevated serum levels of sialyl Lewis X (sLe X) and inflammatory mediators in patients with breast cancer. Breast Cancer Res Treat 2019; 176:545-556. [PMID: 31054033 DOI: 10.1007/s10549-019-05258-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 04/26/2019] [Indexed: 12/17/2022]
Abstract
PURPOSE The carbohydrate sialyl LewisX (sLeX) mediates cell adhesion, is critical in the normal function of immune cells, and is frequently over-expressed on cancer cells. We assessed the association, differential levels, and prognostic value of sLeX and inflammatory cytokines/chemokines in breast cancer sera. METHODS We retrospectively measured sLeX and a panel of cytokines/chemokines in the sera of 26 non-invasive ductal carcinoma in situ (DCIS), 154 invasive non-metastatic breast cancer (non-MBC), 63 metastatic breast cancer (MBC) patients, and 43 healthy controls. Differences in sLeX and inflammatory cytokines among and between patient groups and healthy controls were assessed with nonparametric tests and we performed survival analysis for the prognostic potential of sLeX using a cut-off of 8 U/mL as previously defined. RESULTS Median serum sLeX was significantly higher than controls for invasive breast cancer patients (MBC and non-MBC) but not DCIS. In univariate analysis, we confirmed patients with serum sLeX > 8 U/mL have a significantly shorter progression-free survival (PFS) (P = 0.0074) and overall survival (OS (P = 0.0003). Similarly, patients with high serum MCP-1 and IP-10 had shorter OS (P = 0.001 and P < 0.001, respectively) and PFS (P = 0.010 and P < 0.001, respectively). sLeX, MCP-1 and IP-10 remained significant in multivariate survival analysis. CONCLUSION Elevated serum sLeX was associated with invasive cancer but not DCIS. High serum sLeX levels were associated with inflammatory mediators and may play a role in facilitating local invasion of breast tumor. Furthermore, serum MCP-1, IP-10 and sLeX may have prognostic value in breast cancer.
Collapse
Affiliation(s)
- Evan N Cohen
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, 1220 Holcombe Blvd, Houston, TX, 77030, USA.,Department of Hematopathology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Tamer M Fouad
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, 1220 Holcombe Blvd, Houston, TX, 77030, USA.,Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe, Unit 3552, Houston, TX, 77030, USA.,Department of Medical Oncology, The National Cancer Institute, Cairo University, Kasr El-Aini Road, Cairo, 11796, Egypt
| | - Bang-Ning Lee
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, 1220 Holcombe Blvd, Houston, TX, 77030, USA.,Department of Hematopathology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA.,Michael E. DeBakey Veterans Affairs Medical Center, Conroe, TX, USA
| | - Banu K Arun
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe, Unit 3552, Houston, TX, 77030, USA
| | - Diane Liu
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, 1400 Pressler Street, Houston, TX, 77030, USA
| | - Sanda Tin
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, 1220 Holcombe Blvd, Houston, TX, 77030, USA.,Department of Hematopathology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Angelica M Gutierrez Barrera
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe, Unit 3552, Houston, TX, 77030, USA
| | - Toshihide Miura
- Nittobo Medical Co., Ltd., 1, Shiojima Fukuhara, Fukuyama, Koriyama, Fukushima, Japan
| | - Iwao Kiyokawa
- Nittobo Medical Co., Ltd., 1, Shiojima Fukuhara, Fukuyama, Koriyama, Fukushima, Japan
| | - Jun Yamashita
- Nittobo Medical Co., Ltd., 1, Shiojima Fukuhara, Fukuyama, Koriyama, Fukushima, Japan
| | - Ricardo H Alvarez
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, 1220 Holcombe Blvd, Houston, TX, 77030, USA.,Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe, Unit 3552, Houston, TX, 77030, USA.,Cancer Treatment Centers of America, Newnan, GA, USA
| | - Vicente Valero
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe, Unit 3552, Houston, TX, 77030, USA
| | - Wendy A Woodward
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, 1220 Holcombe Blvd, Houston, TX, 77030, USA.,Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe, Unit 76468, Houston, TX, 77030, USA
| | - Yu Shen
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, 1400 Pressler Street, Houston, TX, 77030, USA
| | - Naoto T Ueno
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, 1220 Holcombe Blvd, Houston, TX, 77030, USA.,Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe, Unit 3552, Houston, TX, 77030, USA
| | - Massimo Cristofanilli
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, 1220 Holcombe Blvd, Houston, TX, 77030, USA.,Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe, Unit 3552, Houston, TX, 77030, USA.,Feinberg School of Medicine, Northwestern Univeristy, Chicago, IL, USA
| | - James M Reuben
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, 1220 Holcombe Blvd, Houston, TX, 77030, USA. .,Department of Hematopathology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA.
| |
Collapse
|
17
|
Adamo H, Hammarsten P, Hägglöf C, Dahl Scherdin T, Egevad L, Stattin P, Halin Bergström S, Bergh A. Prostate cancer induces C/EBPβ expression in surrounding epithelial cells which relates to tumor aggressiveness and patient outcome. Prostate 2019; 79:435-445. [PMID: 30536410 DOI: 10.1002/pros.23749] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 11/08/2018] [Indexed: 02/04/2023]
Abstract
BACKGROUND Implantation of rat prostate cancer cells into the normal rat prostate results in tumor-stimulating adaptations in the tumor-bearing organ. Similar changes are seen in prostate cancer patients and they are related to outcome. One gene previously found to be upregulated in the non-malignant part of tumor-bearing prostate lobe in rats was the transcription factor CCAAT/enhancer-binding protein-β (C/EBPβ). METHODS To explore this further, we examined C/EBPβ expression by quantitative RT-PCR, immunohistochemistry, and Western blot in normal rat prostate tissue surrounding slow-growing non-metastatic Dunning G, rapidly growing poorly metastatic (AT-1), and rapidly growing highly metastatic (MatLyLu) rat prostate tumors-and also by immunohistochemistry in a tissue microarray (TMA) from prostate cancer patients managed by watchful waiting. RESULTS In rats, C/EBPβ mRNA expression was upregulated in the surrounding tumor-bearing prostate lobe. In tumors and in the surrounding non-malignant prostate tissue, C/EBPβ was detected by immunohistochemistry in some epithelial cells and in infiltrating macrophages. The magnitude of glandular epithelial C/EBPβ expression in the tumor-bearing prostates was associated with tumor size, distance to the tumor, and metastatic capacity. In prostate cancer patients, high expression of C/EBPβ in glandular epithelial cells in the surrounding tumor-bearing tissue was associated with accumulation of M1 macrophages (iNOS+) and favorable outcome. High expression of C/EBPβ in tumor epithelial cells was associated with high Gleason score, high tumor cell proliferation, metastases, and poor outcome. CONCLUSIONS This study suggest that the expression of C/EBP-beta, a transcription factor mediating multiple biological effects, is differentially expressed both in the benign parts of the tumor-bearing prostate and in prostate tumors, and that alterations in this may be related to patient outcome.
Collapse
Affiliation(s)
- Hanibal Adamo
- Department of Medical Biosciences, Pathology, Umeå University, Umeå, Sweden
| | - Peter Hammarsten
- Department of Medical Biosciences, Pathology, Umeå University, Umeå, Sweden
| | - Christina Hägglöf
- Department of Medical Biosciences, Pathology, Umeå University, Umeå, Sweden
| | - Tove Dahl Scherdin
- Department of Medical Biosciences, Pathology, Umeå University, Umeå, Sweden
| | - Lars Egevad
- Department of Oncology-Pathology, Karolinska University Hospital, Stockholm, Sweden
| | - Pär Stattin
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | | | - Anders Bergh
- Department of Medical Biosciences, Pathology, Umeå University, Umeå, Sweden
| |
Collapse
|
18
|
Bloom MJ, Saksena SD, Swain GP, Behar MS, Yankeelov TE, Sorace AG. The effects of IKK-beta inhibition on early NF-kappa-B activation and transcription of downstream genes. Cell Signal 2018; 55:17-25. [PMID: 30543861 DOI: 10.1016/j.cellsig.2018.12.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 12/05/2018] [Accepted: 12/10/2018] [Indexed: 02/07/2023]
Abstract
Small molecule approaches targeting the nuclear factor kappa B (NF-kB) pathway, a regulator of inflammation, have thus far proven unsuccessful in the clinic in part due to the complex pleiotropic nature of this network. Downstream effects depend on multiple factors including stimulus-specific temporal patterns of NF-kB activity. Despite considerable advances, genome-level impact of changes in temporal NF-kB activity caused by inhibitors and their stimulus dependency remains unexplored. This study evaluates the effects of pathway inhibitors on early NF-κB activity and downstream gene transcription. 3T3 fibroblasts were treated with SC-514, an inhibitor targeted to the NF-kB pathway, prior to stimulation with interleukin 1 beta (IL-1β) or tumor necrosis factor alpha (TNF-α). Stimulus induced NF-κB activation was quantified using immunofluorescence imaging over 90-minutes and gene expression tracked over 6-hours using mRNA TagSeq. When stimulated with IL-1β or TNF-α, significant differences (P < 0.05, two-way ANOVA), were observed in the temporal profiles of NF-κB activation between treated and untreated cells. Increasing numbers of differentially expressed genes (P < 0.01) were observed at higher inhibitor concentrations. Individual gene expression profiles varied in an inhibitor concentration and stimulus-dependent manner. The results in this study demonstrate small molecule inhibitors acting on pleiotropic pathway components can alter signal dynamics in a stimulus-dependent manner and affect gene response in complex ways.
Collapse
Affiliation(s)
- Meghan J Bloom
- Biomedical Engineering, The University of Texas, Austin, TX, USA.
| | - Sachit D Saksena
- Computational and Systems Biology, Massachusetts Institute of Technology, Cambridge, MA, USA.
| | - George P Swain
- Biomedical Engineering, The University of Texas, Austin, TX, USA.
| | - Marcelo S Behar
- Biomedical Engineering, The University of Texas, Austin, TX, USA
| | - Thomas E Yankeelov
- Biomedical Engineering, The University of Texas, Austin, TX, USA; Diagnostic Medicine, The University of Texas, Austin, TX, USA; Livestrong Cancer Institutes, The University of Texas, Austin, TX, USA; Oncology, The University of Texas, Austin, TX, USA; Institute for Computational and Engineering Sciences, The University of Texas, Austin, TX, USA.
| | - Anna G Sorace
- Biomedical Engineering, The University of Texas, Austin, TX, USA; Diagnostic Medicine, The University of Texas, Austin, TX, USA; Livestrong Cancer Institutes, The University of Texas, Austin, TX, USA; Oncology, The University of Texas, Austin, TX, USA.
| |
Collapse
|
19
|
Role of tumor microenvironment in cancer stem cell chemoresistance and recurrence. Int J Biochem Cell Biol 2018; 103:115-124. [DOI: 10.1016/j.biocel.2018.08.011] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 08/16/2018] [Accepted: 08/18/2018] [Indexed: 12/13/2022]
|
20
|
Banzola I, Mengus C, Wyler S, Hudolin T, Manzella G, Chiarugi A, Boldorini R, Sais G, Schmidli TS, Chiffi G, Bachmann A, Sulser T, Spagnoli GC, Provenzano M. Expression of Indoleamine 2,3-Dioxygenase Induced by IFN-γ and TNF-α as Potential Biomarker of Prostate Cancer Progression. Front Immunol 2018; 9:1051. [PMID: 29896191 PMCID: PMC5986916 DOI: 10.3389/fimmu.2018.01051] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2018] [Accepted: 04/27/2018] [Indexed: 12/22/2022] Open
Abstract
Inflammation has been suggested to play an important role in onset and progression of prostate cancer (PCa). Histological analysis of prostatectomy specimens has revealed focal inflammation in early stage lesions of this malignancy. We addressed the role of inflammatory stimuli in the release of PCa-specific, tumor-derived soluble factors (PCa-TDSFs) already reported to be mediators of PCa morbidity, such as indoleamine 2,3-dioxygenase (IDO) and interleukin (IL)-6. Inflammation-driven production and functions of PCa-TDFSs were tested "in vitro" by stimulating established cell lines (CA-HPV-10 and PC3) with IFN-γ or TNF-α. Expression of genes encoding IDO, IL-6, IFN-γ, TNF-α, and their receptors was investigated in tumor tissues of PCa patients undergoing radical prostatectomy, in comparison with benign prostatic hyperplasia (BPH) specimens. IFN-γ and TNF-α-treatment resulted in the induction of IDO and IL-6 gene expression and release in established cell lines, suggesting that the elicitation of PCa-TDSFs by these cytokines might contribute to progression of cancer into an untreatable phenotype. An analysis based on timing of biochemical recurrence revealed the prognostic value of IDO but not IL-6 gene expression in predicting recurrence-free survival in patients (RFS) with PCa. In addition, a urine-based mRNA biomarker study revealed the diagnostic potential of IDO gene expression in urines of men at risk of PCa development.
Collapse
Affiliation(s)
- Irina Banzola
- Oncology Research Unit, Department of Urology, University Hospital of Zurich, Zurich, Switzerland
- Department of Oncology and Children’s Research Center (CRC), University Children’s Hospital, Zurich, Switzerland
| | - Chantal Mengus
- Department of Biomedicine, University Hospital of Basel, Basel, Switzerland
| | - Stephen Wyler
- Department of Urology, University Hospital of Basel, Basel, Switzerland
| | - Tvrko Hudolin
- Department of Urology, University Hospital of Basel, Basel, Switzerland
| | - Gabriele Manzella
- Department of Oncology and Children’s Research Center (CRC), University Children’s Hospital, Zurich, Switzerland
| | - Alberto Chiarugi
- Department of Preclinical and Clinical Pharmacology, University of Florence, Florence, Italy
| | - Renzo Boldorini
- Department of Health Science, School of Medicine, Università degli Studi del Piemonte Orientale, Novara, Italy
| | - Giovanni Sais
- Oncology Research Unit, Department of Urology, University Hospital of Zurich, Zurich, Switzerland
| | - Tobias S. Schmidli
- Oncology Research Unit, Department of Urology, University Hospital of Zurich, Zurich, Switzerland
| | - Gabriele Chiffi
- Oncology Research Unit, Department of Urology, University Hospital of Zurich, Zurich, Switzerland
| | | | - Tullio Sulser
- Oncology Research Unit, Department of Urology, University Hospital of Zurich, Zurich, Switzerland
| | - Giulio C. Spagnoli
- Department of Biomedicine, University Hospital of Basel, Basel, Switzerland
- CNR Institute of Translational Pharmacology, Rome, Italy
| | - Maurizio Provenzano
- Oncology Research Unit, Department of Urology, University Hospital of Zurich, Zurich, Switzerland
| |
Collapse
|
21
|
Hernández-Esquivel MA, Pérez-Torres A, Romero-Romero L, Reyes-Matute A, Loaiza B, Mellado-Sánchez G, Pavón L, Medina-Rivero E, Pestell RG, Pérez-Tapia SM, Velasco-Velázquez MA. The dialyzable leukocyte extract TransferonTM inhibits tumor growth and brain metastasis in a murine model of prostate cancer. Biomed Pharmacother 2018; 101:938-944. [DOI: 10.1016/j.biopha.2018.03.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 03/02/2018] [Accepted: 03/05/2018] [Indexed: 12/25/2022] Open
|
22
|
Pindiprolu SKSS, Krishnamurthy PT, Chintamaneni PK. Pharmacological targets of breast cancer stem cells: a review. Naunyn Schmiedebergs Arch Pharmacol 2018; 391:463-479. [PMID: 29476201 DOI: 10.1007/s00210-018-1479-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Accepted: 02/13/2018] [Indexed: 02/07/2023]
Abstract
Breast cancers contain small population of tumor-initiating cells called breast cancer stem cells (BCSCs), which are spared even after chemotherapy. Recently, BCSCs are implicated to be a cause of metastasis, tumor relapse, and therapy resistance in breast cancer. BCSCs have unique molecular mechanisms, which can be targeted to eliminate them. These include surface biomarkers, proteins involved in self-renewal pathways, drug efflux transporters, apoptotic/antiapoptotic proteins, autophagy, metabolism, and microenvironment regulation. The complex molecular mechanisms behind the survival of BCSCs and pharmacological targets for elimination of BCSCs are described in this review.
Collapse
Affiliation(s)
- Sai Kiran S S Pindiprolu
- Department of Pharmacology, JSS College of Pharmacy (Jagadguru Sri Shivarathreeshwara University), Rocklands, Udhagamandalam, Tamil Nadu, 643001, India
| | - Praveen T Krishnamurthy
- Department of Pharmacology, JSS College of Pharmacy (Jagadguru Sri Shivarathreeshwara University), Rocklands, Udhagamandalam, Tamil Nadu, 643001, India.
| | - Pavan Kumar Chintamaneni
- Department of Pharmacology, JSS College of Pharmacy (Jagadguru Sri Shivarathreeshwara University), Rocklands, Udhagamandalam, Tamil Nadu, 643001, India
| |
Collapse
|
23
|
Erb HHH, Guggenberger F, Santer FR, Culig Z. Interleukin-4 induces a CD44 high /CD49b high PC3 subpopulation with tumor-initiating characteristics. J Cell Biochem 2018; 119:4103-4112. [PMID: 29236307 PMCID: PMC5900863 DOI: 10.1002/jcb.26607] [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: 08/04/2017] [Accepted: 12/04/2017] [Indexed: 12/22/2022]
Abstract
Pro‐ and anti‐inflammatory cytokines may influence proliferation, migration, invasion, and other cellular events of prostate cancer (PCa) cells. The hyaluronan receptor CD44, which is regulated by Interleukin (IL)‐4, is a prostate basal cell marker. CD44high/CD49bhigh expressing cells have been demonstrated to have tumor‐initiating characteristics. Here, we aimed to analyze the effects of long‐term IL‐4 treatment on CD44/CD49b expression, migration, proliferation, and clonogenic potential of basal‐like PCa cells. To this end PC3 cells were treated over 30 passages with 5 ng/mL IL‐4 (PC3‐IL4) resulting in an increased population of CD44high expressing cells. This was concurrent with a clonal outgrowth of cuboid‐shaped cells, with increased size and light absorbance properties. Flow cytometry revealed that the PC3‐IL4 CD44high expressing subpopulation corresponds to the CD49bhigh population. Isolation of the PC3‐IL4 CD44high/CD49bhigh subpopulation via fluorescence‐associated cell sorting showed increased migrative, proliferative, and clonogenic potential compared to the CD44low/CD49blow subpopulation. In conclusion, IL‐4 increases a PC3 subpopulation with tumor‐initiating characteristics. Thus, IL‐4, similar to other cytokines may be a regulator of tumor‐initiation and hence, may present a suitable therapy target in combination with current treatment options.
Collapse
Affiliation(s)
- Holger H H Erb
- Department of Urology and Pediatric Urology, University Medical Center Mainz, Mainz, Germany
| | - Fabian Guggenberger
- Division of Experimental Urology, Department of Urology, Medical University of Innsbruck, Innsbruck, Austria
| | - Frédéric R Santer
- Division of Experimental Urology, Department of Urology, Medical University of Innsbruck, Innsbruck, Austria
| | - Zoran Culig
- Division of Experimental Urology, Department of Urology, Medical University of Innsbruck, Innsbruck, Austria.,Center of Biomolecular and Cellular Engineering, International Clinical Research Center, St. Annés University Hospital Brno, Brno, Czech Republic
| |
Collapse
|
24
|
Pindiprolu SKSS, Krishnamurthy PT, Chintamaneni PK, Karri VVSR. Nanocarrier based approaches for targeting breast cancer stem cells. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2017; 46:885-898. [PMID: 28826237 DOI: 10.1080/21691401.2017.1366337] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Breast cancer stem cells (BCSCs) are heterogeneous subpopulation of tumour initiating cells within breast tumours. They are spared even after chemotherapy and responsible for tumour relapse. Targeting BCSCs is, therefore, necessary to achieve radical cure in breast cancer. Despite the availability of agents targeting BCSCs, their clinical application is limited due to their off-target effects and bioavailability issues. Nanotechnology based drug carriers (nanocarriers) offer various advantages to deliver anti-BCSCs agents specifically to their target sites by overcoming their bioavailability issues. In this review, we describe various strategies for targeting BCSCs using nanocarriers.
Collapse
Affiliation(s)
- Sai Kiran S S Pindiprolu
- a Department of Pharmacology , JSS College of Pharmacy (A Constituent College of Jagadguru Sri Shivarathreeshwara University) , Ootacamund , Tamil Nadu , India
| | - Praveen T Krishnamurthy
- a Department of Pharmacology , JSS College of Pharmacy (A Constituent College of Jagadguru Sri Shivarathreeshwara University) , Ootacamund , Tamil Nadu , India
| | - Pavan Kumar Chintamaneni
- a Department of Pharmacology , JSS College of Pharmacy (A Constituent College of Jagadguru Sri Shivarathreeshwara University) , Ootacamund , Tamil Nadu , India
| | - Veera Venkata Satyanarayana Reddy Karri
- b Department of Pharmaceutics , JSS College of Pharmacy (A Constituent College of Jagadguru Sri Shivarathreeshwara University) , Ootacamund , Tamil Nadu , India
| |
Collapse
|
25
|
The immunosuppressive cytokine interleukin-4 increases the clonogenic potential of prostate stem-like cells by activation of STAT6 signalling. Oncogenesis 2017; 6:e342. [PMID: 28553931 PMCID: PMC5523058 DOI: 10.1038/oncsis.2017.23] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Revised: 02/24/2017] [Accepted: 03/03/2017] [Indexed: 12/11/2022] Open
Abstract
Interleukin-4 plays a critical role in the regulation of immune responses and has been detected at high levels in the tumour microenvironment of cancer patients, where concentrations correlate with the grade of malignancy. In prostate cancer, interleukin-4 has been associated with activation of the androgen receptor, increased proliferation and activation of survival pathways such as Akt and NF-κB. However, its role in therapy resistance has not yet been determined. Here we investigate the influence of interleukin-4 on primary epithelial cells from prostate cancer patients. Our data demonstrate an increase in the clonogenic potential of these cells when cultured in the presence of interleukin-4. In addition, a Phospho-Kinase Array revealed that in contrast to previously published work, signal transducer and activator of transcription6 (STAT6) is the only signalling molecule activated after interleukin-4 treatment. Using the STAT6-specific inhibitor AS1517499 we could confirm the role of STAT6 in increasing colony-forming frequency. However, clonogenic recovery assays revealed that interleukin-4 does not rescue the effects of either irradiation or docetaxel treatment. We therefore propose that although the interleukin-4/STAT6 axis does not appear to be involved in therapy resistance, it does play a crucial role in the colony-forming abilities of the basal cell population in prostate cancer. IL-4 may therefore contribute to disease relapse by providing a niche that is favourable for the clonogenic growth of prostate cancer stem cells.
Collapse
|
26
|
Nandy SB, Lakshmanaswamy R. Cancer Stem Cells and Metastasis. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2017; 151:137-176. [DOI: 10.1016/bs.pmbts.2017.07.007] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
27
|
Bryson BL, Junk DJ, Cipriano R, Jackson MW. STAT3-mediated SMAD3 activation underlies Oncostatin M-induced Senescence. Cell Cycle 2016; 16:319-334. [PMID: 27892764 DOI: 10.1080/15384101.2016.1259037] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Cytokines in the developing tumor microenvironment (TME) can drive transformation and subsequent progression toward metastasis. Elevated levels of the Interleukin-6 (IL-6) family cytokine Oncostatin M (OSM) in the breast TME correlate with aggressive, metastatic cancers, increased tumor recurrence, and poor patient prognosis. Paradoxically, OSM engages a tumor-suppressive, Signal Transducer and Activator of Transcription 3 (STAT3)-dependent senescence response in normal and non-transformed human mammary epithelial cells (HMEC). Here, we identify a novel link between OSM-activated STAT3 signaling and the Transforming Growth Factor-β (TGF-β) signaling pathway that engages senescence in HMEC. Inhibition of functional TGF-β/SMAD signaling by expressing a dominant-negative TGF-β receptor, treating with a TGF-β receptor inhibitor, or suppressing SMAD3 expression using a SMAD3-shRNA prevented OSM-induced senescence. OSM promoted a protein complex involving activated-STAT3 and SMAD3, induced the nuclear localization of SMAD3, and enhanced SMAD3-mediated transcription responsible for senescence. In contrast, expression of MYC (c-MYC) from a constitutive promoter abrogated senescence and strikingly, cooperated with OSM to promote a transformed phenotype, epithelial-mesenchymal transition (EMT), and invasiveness. Our findings suggest that a novel STAT3/SMAD3-signaling axis is required for OSM-mediated senescence that is coopted during the transformation process to confer aggressive cancer cell properties. Understanding how developing cancer cells bypass OSM/STAT3/SMAD3-mediated senescence may help identify novel targets for future "pro-senescence" therapies aiming to reengage this hidden tumor-suppressive response.
Collapse
Affiliation(s)
- Benjamin L Bryson
- a Department of Pathology , School of Medicine, Case Western Reserve University , Cleveland , OH , USA
| | - Damian J Junk
- a Department of Pathology , School of Medicine, Case Western Reserve University , Cleveland , OH , USA
| | - Rocky Cipriano
- a Department of Pathology , School of Medicine, Case Western Reserve University , Cleveland , OH , USA
| | - Mark W Jackson
- a Department of Pathology , School of Medicine, Case Western Reserve University , Cleveland , OH , USA.,b Case Comprehensive Cancer Center , Case Western Reserve University , Cleveland , OH , USA
| |
Collapse
|
28
|
Christopher AF, Gupta M, Bansal P. Micronome revealed miR-19a/b as key regulator of SOCS3 during cancer related inflammation of oral squamous cell carcinoma. Gene 2016; 594:30-40. [PMID: 27581787 DOI: 10.1016/j.gene.2016.08.044] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Revised: 08/20/2016] [Accepted: 08/26/2016] [Indexed: 12/21/2022]
Abstract
Although significant advances have been established in molecular biology of Oral squamous cell carcinoma (OSCC), innovative strategies are still required to further understand detailed molecular mechanisms. Using bioinformatic approach, we aim to explore the potential miRNA-mRNA pairs in cancer related inflammatory response and investigate their potential roles as signature miRNA and proteins in the signaling pathway. Firstly, the differentially expressed genes of OSCC were selected which then underwent gene ontology to identify genes engaged in inflammatory response and its regulation. Validated miRNAs were retrieved and miRNAs with complete complementarily with their targets were visualized for miRNA-mRNA regulatory network. Protein-protein interactions of inflammatory and its regulatory genes were analyzed for interacting genes involved in signaling pathway. Eight universal miRNAs were obtained for inflammation and its regulation. miRNA-19a/b showed significant influence in controlling inflammatory response in OSCC. Therefore, micronome on deregulated genes in inflammation identifies miRNA-mRNA pairs which have high potential to be targeted for diagnostic and treatment applications in OSCC.
Collapse
Affiliation(s)
- Ajay Francis Christopher
- Division of Clinical Research, University Centre of Excellence in Research, Baba Farid University of Health Science, Faridkot 151203, Punjab, India
| | - Mridula Gupta
- Division of Clinical Research, University Centre of Excellence in Research, Baba Farid University of Health Science, Faridkot 151203, Punjab, India
| | - Parveen Bansal
- Division of Clinical Research, University Centre of Excellence in Research, Baba Farid University of Health Science, Faridkot 151203, Punjab, India.
| |
Collapse
|
29
|
IGF1R as a Key Target in High Risk, Metastatic Medulloblastoma. Sci Rep 2016; 6:27012. [PMID: 27255663 PMCID: PMC4891740 DOI: 10.1038/srep27012] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Accepted: 05/12/2016] [Indexed: 12/21/2022] Open
Abstract
Risk or presence of metastasis in medulloblastoma causes substantial treatment-related morbidity and overall mortality. Through the comparison of cytokines and growth factors in the cerebrospinal fluid (CSF) of metastatic medulloblastoma patients with factors also in conditioned media of metastatic MYC amplified medulloblastoma or leptomeningeal cells, we were led to explore the bioactivity of IGF1 in medulloblastoma by elevated CSF levels of IGF1, IGF-sequestering IGFBP3, IGFBP3-cleaving proteases (MMP and tPA), and protease modulators (TIMP1 and PAI-1). IGF1 led not only to receptor phosphorylation but also accelerated migration/adhesion in MYC amplified medulloblastoma cells in the context of appropriate matrix or meningothelial cells. Clinical correlation suggests a peri-/sub-meningothelial source of IGF-liberating proteases that could facilitate leptomeningeal metastasis. In parallel, studies of key factors responsible for cell autonomous growth in MYC amplified medulloblastoma prioritized IGF1R inhibitors. Together, our studies identify IGF1R as a high value target for clinical trials in high risk medulloblastoma.
Collapse
|
30
|
Affiliation(s)
- Miyoung Yang
- Department of Anatomy, School of Medicine, Wonkwang University, Iksan, Jeonbuk, South Korea
| | - Changjong Moon
- Department of Veterinary Anatomy and Animal Behavior, College of Veterinary Medicine, Chonnam National University, Gwangju, South Korea
| |
Collapse
|
31
|
Bharti R, Dey G, Ojha PK, Rajput S, Jaganathan SK, Sen R, Mandal M. Diacerein-mediated inhibition of IL-6/IL-6R signaling induces apoptotic effects on breast cancer. Oncogene 2015; 35:3965-75. [PMID: 26616855 DOI: 10.1038/onc.2015.466] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Revised: 09/29/2015] [Accepted: 10/05/2015] [Indexed: 02/08/2023]
Abstract
Interleukin-6 (IL-6) signaling network has been implicated in oncogenic transformations making it attractive target for the discovery of novel cancer therapeutics. In this study, potent antiproliferative and apoptotic effect of diacerein were observed against breast cancer. In vitro apoptosis was induced by this drug in breast cancer cells as verified by increased sub-G1 population, LIVE/DEAD assay, cell cytotoxicity and presence of terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL)-positive cells, as well as downregulation of antiapoptotic proteins Bcl-2 and Bcl-xL and upregulation of apoptotic protein Bax. In addition, apoptosis induction was found to be caspase dependent. Further molecular investigations indicated that diacerein instigated apoptosis was associated with inhibition of IL-6/IL-6R autocrine signaling axis. Suppression of STAT3, MAPK and Akt pathways were also observed as a consequence of diacerein-mediated upstream inhibition of IL-6/IL-6R. Fluorescence study and western blot analysis revealed cytosolic accumulation of STAT3 in diacerein-treated cells. The docking study showed diacerein/IL-6R interaction that was further validated by competitive binding assay and isothermal titration calorimetry. Most interestingly, it was found that diacerein considerably suppressed tumor growth in MDA-MB-231 xenograft model. The in vivo antitumor effect was correlated with decreased proliferation (Ki-67), increased apoptosis (TUNEL) and inhibition of IL-6/IL-6R-mediated STAT3, MAPK and Akt pathway in tumor remnants. Taken together, diacerein offered a novel blueprint for cancer therapy by hampering IL-6/IL-6R/STAT3/MAPK/Akt network.
Collapse
Affiliation(s)
- R Bharti
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, India
| | - G Dey
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, India
| | - P K Ojha
- Drug Theoretics and Cheminformatics Laboratory, Division of Medicinal and Pharmaceutical Chemistry, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, India
| | - S Rajput
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, India
| | - S K Jaganathan
- IJN-UTM Cardiovascular Engineering Centre, Faculty of Biosciences and Medical Engineering, Universiti Teknologi, Malaysia
| | - R Sen
- Department of Biotechnology, Indian Institute of Technology Kharagpur, Kharagpur, India
| | - M Mandal
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, India
| |
Collapse
|
32
|
Almatroodi SA, McDonald CF, Darby IA, Pouniotis DS. Characterization of M1/M2 Tumour-Associated Macrophages (TAMs) and Th1/Th2 Cytokine Profiles in Patients with NSCLC. CANCER MICROENVIRONMENT 2015; 9:1-11. [PMID: 26319408 DOI: 10.1007/s12307-015-0174-x] [Citation(s) in RCA: 98] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Accepted: 08/17/2015] [Indexed: 12/11/2022]
Abstract
Lung cancer is one of the most commonly reported cancers, and is known to be associated with a poor prognosis. The function of tumour-associated macrophages (TAMs) in lung cancer patients is multifaceted and the literature shows conflicting roles. (I) To analyze the Th1 and Th2 cytokine levels that contribute to the differentiation of M1 and M2 macrophage populations in the serum of patients with NSCLC versus non-cancer controls; and (II) To characterize the M1 and M2 macrophage populations within TAMs in different subtypes of NSCLC compared to non-tumour tissue. The Th1 and Th2 cytokine levels were analyzed in serum using the Bio-Plex assay. In addition, TAMs subsets from non-tumour and tumour tissues were analyzed using immunohistochemistry (IHC). The level of IL-1β, IL-4, IL-6 and IL-8 was found to be increased in the serum of patients with large cell carcinoma but not in other NSCLC subtypes compared to non-cancer controls. In addition, the expression of CD68 and M2 marker CD163 was found to be increased (P ≤ 0.0001) in all NSCLC subtypes compared to non-tumour tissues. In contrast, the expression of iNOS (M1 marker) was decreased in the tumour tissue of patients with adenocarcinoma (P ≤ 0.01) and squamous carcinoma (P ≤ 0.05) but not in large cell carcinoma compared to non-tumour tissue. The results of this study indicate that NSCLC might have the ability to alter phenotype within the lung tumour areas in the local environment (TAMs) but not in the bloodstream in the systemic environment (serum) except for large cell carcinoma.
Collapse
Affiliation(s)
- S A Almatroodi
- Cancer & Tissue Repair Laboratory, School of Medical Sciences, RMIT University, P.O. Box 71, Bundoora, Victoria, Australia, 3083. .,Applied Medical Sciences College, Qassim University, Buraidah, Saudi Arabia.
| | - C F McDonald
- Institute for Breathing & Sleep, Austin Health, Heidelberg, Victoria, 3084, Australia
| | - I A Darby
- Cancer & Tissue Repair Laboratory, School of Medical Sciences, RMIT University, P.O. Box 71, Bundoora, Victoria, Australia, 3083
| | - D S Pouniotis
- Cancer & Tissue Repair Laboratory, School of Medical Sciences, RMIT University, P.O. Box 71, Bundoora, Victoria, Australia, 3083
| |
Collapse
|
33
|
Immature myeloid cells and tolerogenic cytokine profile in lung adenocarcinoma metastatic lymph nodes assessed by endobronchial ultrasound. Tumour Biol 2015; 37:953-61. [DOI: 10.1007/s13277-015-3885-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2015] [Accepted: 08/03/2015] [Indexed: 01/15/2023] Open
|
34
|
Malaponte G, Signorelli SS, Bevelacqua V, Polesel J, Taborelli M, Guarneri C, Fenga C, Umezawa K, Libra M. Increased Levels of NF-kB-Dependent Markers in Cancer-Associated Deep Venous Thrombosis. PLoS One 2015; 10:e0132496. [PMID: 26192925 PMCID: PMC4507873 DOI: 10.1371/journal.pone.0132496] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2015] [Accepted: 06/15/2015] [Indexed: 12/17/2022] Open
Abstract
Several studies highlight the role of inflammatory markers in thrombosis as well as in cancer. However, their combined role in cancer-associated deep vein thrombosis (DVT) and the molecular mechanisms, involved in its pathophysiology, needs further investigations. In the present study, C-reactive protein, interleukin-6 (IL-6), tumor necrosis factor-α (TNF-α), interleukin-1 (IL-1β), matrix metalloproteases-9 (MMP-9), vascular endothelial growth factor (VEGF), tissue factor (TF), fibrinogen and soluble P-selectin, were analyzed in plasma and in monocyte samples from 385 cancer patients, of whom 64 were concomitantly affected by DVT (+). All these markers were higher in cancer patients DVT+ than in those DVT-. Accordingly, significantly higher NF-kB activity was observed in cancer patients DVT+ than DVT-. Significant correlation between data obtained in plasma and monocyte samples was observed. NF-kB inhibition was associated with decreased levels of all molecules in both cancer DVT+ and DVT-. To further demonstrate the involvement of NF-kB activation by the above mentioned molecules, we treated monocyte derived from healthy donors with a pool of sera from cancer patients with and without DVT. These set of experiments further suggest the significant role played by some molecules, regulated by NF-kB, and detected in cancer patients with DVT. Our data support the notion that NF-kB may be considered as a therapeutic target for cancer patients, especially those complicated by DVT. Treatment with NF-kB inhibitors may represent a possible strategy to prevent or reduce the risk of DVT in cancer patients.
Collapse
Affiliation(s)
- Grazia Malaponte
- Department of Biomedical and Biotechnological Sciences, Section of General & Clinical Pathology and Oncology, University of Catania, Catania, Italy
| | - Salvatore S. Signorelli
- Department of Clinical and Experimental Medicine, University of Catania, Medical Angiology Unit, Garibaldi Hospital, Catania, Italy
- * E-mail:
| | - Valentina Bevelacqua
- Department of Biomedical and Biotechnological Sciences, Section of General & Clinical Pathology and Oncology, University of Catania, Catania, Italy
| | - Jerry Polesel
- Epidemiology and Biostatistics Unit, Centro di Riferimento Oncologico-National Cancer Institute, Aviano, Italy
| | - Martina Taborelli
- Epidemiology and Biostatistics Unit, Centro di Riferimento Oncologico-National Cancer Institute, Aviano, Italy
| | - Claudio Guarneri
- Department of Clinical and Experimental Medicine, Section of Dermatology, University of Messina, Messina, Italy
| | - Concettina Fenga
- Department of Environmental and Health Sciences (S.A.S.T.A.S.), University of Messina, Messina, Italy
| | - Kazou Umezawa
- Department of Molecular Target Medicine Screening, Aichi Medical University, Nagakute, Japan
| | - Massimo Libra
- Department of Biomedical and Biotechnological Sciences, Section of General & Clinical Pathology and Oncology, University of Catania, Catania, Italy
| |
Collapse
|
35
|
Chen R, Chen B. Siltuximab (CNTO 328): a promising option for human malignancies. DRUG DESIGN DEVELOPMENT AND THERAPY 2015; 9:3455-8. [PMID: 26170629 PMCID: PMC4494175 DOI: 10.2147/dddt.s86438] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Siltuximab (CNTO 328) is a promising antibody-drug conjugate targeting cytokine interleukin-6 (IL-6). It is highly binding to IL-6 and thus neutralizing IL-6 bioactivity and promoting death of tumor cell. In this review, we mainly focus on the mechanisms, clinical studies, and adverse effect of siltuximab in the treatment of human malignancies. We also provide our recommendations for siltuximab treatment in the future.
Collapse
Affiliation(s)
- Runzhe Chen
- Department of Hematology and Oncology, Zhongda Hospital, Medical School, Southeast University, Nanjing, Jiangsu Province, People's Republic of China
| | - Baoan Chen
- Department of Hematology and Oncology, Zhongda Hospital, Medical School, Southeast University, Nanjing, Jiangsu Province, People's Republic of China
| |
Collapse
|
36
|
Fibulin-3 is a novel TGF-β pathway inhibitor in the breast cancer microenvironment. Oncogene 2015; 34:5635-47. [PMID: 25823021 PMCID: PMC4589427 DOI: 10.1038/onc.2015.13] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Revised: 12/17/2014] [Accepted: 01/05/2015] [Indexed: 12/11/2022]
Abstract
TGF-β is an important regulator of breast cancer progression. However, how the breast cancer microenvironment regulates TGF-β signaling during breast cancer progression remains largely unknown. Here, we identified fibulin-3 as a secreted protein in the breast cancer microenvironment, which efficiently inhibits TGF-β signaling in both breast cancer cells and endothelial cells. Mechanistically, fibulin-3 interacts with the type I TGF-β receptor (TβRI) to block TGF-β induced complex formation of TβRI with the type II TGF-β receptor (TβRII) and subsequent downstream TGF-β signaling. Fibulin-3 expression decreases during breast cancer progression, with low fibulin-3 levels correlating with a poorer prognosis. Functionally, high fibulin-3 levels inhibited TGF-β-induced EMT, migration, invasion and endothelial permeability, while loss of fibulin-3 expression/function promoted these TGF-β-mediated effects. Further, restoring fibulin-3 expression in breast cancer cells inhibited TGF-β signaling, breast cancer cell EMT, invasion and metastasis in vivo. These studies provide a novel mechanism for how TGF-β signaling is regulated by the tumor microenvironment, and provide insight into targeting the TGF-β signaling pathway in human breast cancer patients.
Collapse
|
37
|
Esquivel-Velázquez M, Ostoa-Saloma P, Palacios-Arreola MI, Nava-Castro KE, Castro JI, Morales-Montor J. The role of cytokines in breast cancer development and progression. J Interferon Cytokine Res 2015; 35:1-16. [PMID: 25068787 PMCID: PMC4291218 DOI: 10.1089/jir.2014.0026] [Citation(s) in RCA: 329] [Impact Index Per Article: 36.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Accepted: 05/23/2014] [Indexed: 12/12/2022] Open
Abstract
Cytokines are highly inducible, secretory proteins that mediate intercellular communication in the immune system. They are grouped into several protein families that are referred to as tumor necrosis factors, interleukins, interferons, and colony-stimulating factors. In recent years, it has become clear that some of these proteins as well as their receptors are produced in the organisms under physiological and pathological conditions. The exact initiation process of breast cancer is unknown, although several hypotheses have emerged. Inflammation has been proposed as an important player in tumor initiation, promotion, angiogenesis, and metastasis, all phenomena in which cytokines are prominent players. The data here suggest that cytokines play an important role in the regulation of both induction and protection in breast cancer. This knowledge could be fundamental for the proposal of new therapeutic approaches to particularly breast cancer and other cancer-related disorders.
Collapse
Affiliation(s)
- Marcela Esquivel-Velázquez
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, México City, México
| | - Pedro Ostoa-Saloma
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, México City, México
| | | | - Karen E. Nava-Castro
- Centro de Investigación Sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, SSA, Cuernavaca, Morelos, México
| | - Julieta Ivonne Castro
- Centro de Investigación Sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, SSA, Cuernavaca, Morelos, México
| | - Jorge Morales-Montor
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, México City, México
| |
Collapse
|
38
|
|
39
|
Hahn AM, Myers JD, McFarland EK, Lee S, Jerde TJ. Interleukin-driven insulin-like growth factor promotes prostatic inflammatory hyperplasia. J Pharmacol Exp Ther 2014; 351:605-15. [PMID: 25292180 DOI: 10.1124/jpet.114.218693] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Prostatic inflammation is of considerable importance to urologic research because of its association with benign prostatic hyperplasia and prostate cancer. However, the mechanisms by which inflammation leads to proliferation and growth remain obscure. Here, we show that insulin-like growth factors (IGFs), previously known as critical developmental growth factors during prostate organogenesis, are induced by inflammation as part of the proliferative recovery to inflammation. Using genetic models and in vivo IGF receptor blockade, we demonstrate that the hyperplastic response to inflammation depends on interleukin-1-driven IGF signaling. We show that human prostatic hyperplasia is associated with IGF pathway activation specifically localized to foci of inflammation. This demonstrates that mechanisms of inflammation-induced epithelial proliferation and hyperplasia involve the induction of developmental growth factors, further establishing a link between inflammatory and developmental signals and providing a mechanistic basis for the management of proliferative diseases by IGF pathway modulation.
Collapse
Affiliation(s)
- Alana M Hahn
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, Indiana (A.M.H., J.D.M., T.J.J.); Cellular and Molecular Biology, University of Wisconsin-Madison, Madison, Wisconsin (E.K.M., S.L.); and Melvin and Bren Simon Cancer Center, Indiana Basic Urological Research Working Group, Indiana University, Indianapolis, Indiana (T.J.J.)
| | - Jason D Myers
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, Indiana (A.M.H., J.D.M., T.J.J.); Cellular and Molecular Biology, University of Wisconsin-Madison, Madison, Wisconsin (E.K.M., S.L.); and Melvin and Bren Simon Cancer Center, Indiana Basic Urological Research Working Group, Indiana University, Indianapolis, Indiana (T.J.J.)
| | - Eliza K McFarland
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, Indiana (A.M.H., J.D.M., T.J.J.); Cellular and Molecular Biology, University of Wisconsin-Madison, Madison, Wisconsin (E.K.M., S.L.); and Melvin and Bren Simon Cancer Center, Indiana Basic Urological Research Working Group, Indiana University, Indianapolis, Indiana (T.J.J.)
| | - Sanghee Lee
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, Indiana (A.M.H., J.D.M., T.J.J.); Cellular and Molecular Biology, University of Wisconsin-Madison, Madison, Wisconsin (E.K.M., S.L.); and Melvin and Bren Simon Cancer Center, Indiana Basic Urological Research Working Group, Indiana University, Indianapolis, Indiana (T.J.J.)
| | - Travis J Jerde
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, Indiana (A.M.H., J.D.M., T.J.J.); Cellular and Molecular Biology, University of Wisconsin-Madison, Madison, Wisconsin (E.K.M., S.L.); and Melvin and Bren Simon Cancer Center, Indiana Basic Urological Research Working Group, Indiana University, Indianapolis, Indiana (T.J.J.)
| |
Collapse
|
40
|
Davare MA, Lal S, Peckham JL, Prajapati SI, Gultekin SH, Rubin BP, Keller C. Secreted meningeal chemokines, but not VEGFA, modulate the migratory properties of medulloblastoma cells. Biochem Biophys Res Commun 2014; 450:555-60. [PMID: 24928387 DOI: 10.1016/j.bbrc.2014.06.018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Accepted: 06/03/2014] [Indexed: 02/01/2023]
Abstract
Leptomeningeal metastasis is a cause of morbidity and mortality in medulloblastoma, but the understanding of molecular mechanisms driving this process is nascent. In this study, we examined the secretory chemokine profile of medulloblastoma cells (DAOY) and a meningothelial cell line (BMEN1). Conditioned media (CM) of meningothelial cells increased adhesion, spreading and migration of medulloblastoma. VEGFA was identified at elevated levels in the CM from BMEN1 cells (as compared to DAOY CM); however, recombinant VEGFA alone was insufficient to enhance medulloblastoma cell migration. In addition, bevacizumab, the VEGFA scavenging monoclonal antibody, did not block the migratory phenotype induced by the CM. These results reveal that paracrine factors secreted by meningothelial cells can influence migration and adherence of medulloblastoma tumor cells, but VEGFA may not be a specific target for therapeutic intervention in this context.
Collapse
Affiliation(s)
- Monika A Davare
- Pediatric Cancer Biology Program, Papé Family Pediatric Research Institute, Department of Pediatrics, Oregon Health & Science University, Portland, OR 97239, USA.
| | - Sangeet Lal
- Pediatric Cancer Biology Program, Papé Family Pediatric Research Institute, Department of Pediatrics, Oregon Health & Science University, Portland, OR 97239, USA
| | - Jennifer L Peckham
- Pediatric Cancer Biology Program, Papé Family Pediatric Research Institute, Department of Pediatrics, Oregon Health & Science University, Portland, OR 97239, USA
| | - Suresh I Prajapati
- Greehey Children's Cancer Research Institute, University of Texas Health Science Center, San Antonio, TX 78229, USA
| | - Sakir H Gultekin
- Department of Pathology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Brian P Rubin
- Department of Anatomic Pathology, Taussig Cancer Center and Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195, USA; Department Molecular Genetics, Taussig Cancer Center and Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, 44195 USA
| | - Charles Keller
- Pediatric Cancer Biology Program, Papé Family Pediatric Research Institute, Department of Pediatrics, Oregon Health & Science University, Portland, OR 97239, USA.
| |
Collapse
|
41
|
Hoefer J, Kern J, Ofer P, Eder IE, Schäfer G, Dietrich D, Kristiansen G, Geley S, Rainer J, Gunsilius E, Klocker H, Culig Z, Puhr M. SOCS2 correlates with malignancy and exerts growth-promoting effects in prostate cancer. Endocr Relat Cancer 2014; 21:175-87. [PMID: 24280133 PMCID: PMC3907181 DOI: 10.1530/erc-13-0446] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Deregulation of cytokine and growth factor signaling due to an altered expression of endogenous regulators is well recognized in prostate cancer (PCa) and other cancers. Suppressor of cytokine signaling 2 (SOCS2) is a key regulator of the GH, IGF, and prolactin signaling pathways that have been implicated in carcinogenesis. In this study, we evaluated the expression patterns and functional significance of SOCS2 in PCa. Protein expression analysis employing tissue microarrays from two independent patient cohorts revealed a significantly enhanced expression in tumor tissue compared with benign tissue as well as association with Gleason score and disease progression. In vitro and in vivo assays uncovered the involvement of SOCS2 in the regulation of cell growth and apoptosis. Functionally, SOCS2 knockdown inhibited PCa cell proliferation and xenograft growth in a CAM assay. Decreased cell growth after SOCS2 downregulation was associated with cell-cycle arrest and apoptosis. In addition, we proved that SOCS2 expression is significantly elevated upon androgenic stimulation in androgen receptor (AR)-positive cell lines, providing a possible mechanistic explanation for high SOCS2 levels in PCa tissue. Consequently, SOCS2 expression correlated with AR expression in the malignant tissue of patients. On the whole, our study linked increased SOCS2 expression in PCa with a pro-proliferative role in vitro and in vivo.
Collapse
Affiliation(s)
- Julia Hoefer
- Experimental Urology, Department of UrologyInnsbruck Medical UniversityAnichstrasse 35A-6020, InnsbruckAustria
| | - Johann Kern
- Oncotyrol Laboratory for Tumor Biology and AngiogenesisInnsbruckAustria
| | - Philipp Ofer
- Experimental Urology, Department of UrologyInnsbruck Medical UniversityAnichstrasse 35A-6020, InnsbruckAustria
| | - Iris E Eder
- Experimental Urology, Department of UrologyInnsbruck Medical UniversityAnichstrasse 35A-6020, InnsbruckAustria
| | - Georg Schäfer
- Experimental Urology, Department of UrologyInnsbruck Medical UniversityAnichstrasse 35A-6020, InnsbruckAustria
| | - Dimo Dietrich
- Institute of PathologyUniversity Hospital BonnBonnGermany
| | | | - Stephan Geley
- Division of Molecular PathophysiologyInnsbruck Biocenter Medical University InnsbruckInnsbruckAustria
| | - Johannes Rainer
- Division of Molecular PathophysiologyInnsbruck Biocenter Medical University InnsbruckInnsbruckAustria
| | | | - Helmut Klocker
- Experimental Urology, Department of UrologyInnsbruck Medical UniversityAnichstrasse 35A-6020, InnsbruckAustria
| | - Zoran Culig
- Experimental Urology, Department of UrologyInnsbruck Medical UniversityAnichstrasse 35A-6020, InnsbruckAustria
- Correspondence should be addressed to Z Culig or M Puhr Emails: or
| | - Martin Puhr
- Experimental Urology, Department of UrologyInnsbruck Medical UniversityAnichstrasse 35A-6020, InnsbruckAustria
- Correspondence should be addressed to Z Culig or M Puhr Emails: or
| |
Collapse
|
42
|
Yang M, Kim J, Kim JS, Kim SH, Kim JC, Kang MJ, Jung U, Shin T, Wang H, Moon C. Hippocampal dysfunctions in tumor-bearing mice. Brain Behav Immun 2014; 36:147-55. [PMID: 24513875 DOI: 10.1016/j.bbi.2013.10.022] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2013] [Revised: 10/07/2013] [Accepted: 10/22/2013] [Indexed: 01/29/2023] Open
Abstract
Individuals with cancer are particularly susceptible to depression and cognitive impairment. However, the precise mechanisms underlying cancer-induced hippocampal dysfunction are poorly understood. We investigated the effects of a peripheral tumor on emotional behavior, hippocampus-dependent memory and associated molecular and cellular features using an experimental animal model. Behavioral alterations were examined; stress-related parameters measured; hippocampal neurogenesis evaluated; and the levels of pro-inflammatory cytokines, brain-derived neurotrophic factor (BDNF) and cyclooxygenase-2 (COX-2) assayed, 2 weeks after inoculation of adult BALB/c mice with cells of a colon carcinoma cell line (CT26). As the tumors developed, CT26-inoculated mice showed significant increases in the depression-like behavior (measured using the tail suspension test) and memory impairment (in terms of object recognition) compared with vehicle-inoculated controls. The presence of a peripheral tumor significantly elevated the hippocampal levels of mRNAs encoding interleukin-6 (IL-6) and tumor necrosis factor-α, as well as plasma IL-6 and corticosterone levels. Additionally, the adrenal glands became enlarged, and the numbers of Ki-67-positive proliferating hippocampal cells and doublecortin-positive immature progenitor neurons, as well as the constitutive levels of mRNAs encoding BDNF and COX-2 were significantly reduced. Therefore, a peripheral tumor alone may be sufficient to induce hippocampal dysfunction, possibly by reducing the rate of neurogenesis and the levels of BDNF and COX-2 in that tissue and also by increasing stress-related parameters and the circulating levels of pro-inflammatory cytokines.
Collapse
|
43
|
Wang SW, Sun YM. The IL-6/JAK/STAT3 pathway: potential therapeutic strategies in treating colorectal cancer (Review). Int J Oncol 2014; 44:1032-40. [PMID: 24430672 DOI: 10.3892/ijo.2014.2259] [Citation(s) in RCA: 227] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Accepted: 01/07/2014] [Indexed: 11/06/2022] Open
Abstract
Among the cytokines linked to inflammation-associated cancer, interleukin (IL)-6 drives many of the cancer 'hallmarks' through downstream activation of the Janus kinase/signal transducer and activator of transcription 3 (JAK/STAT3) signaling pathway. Additionally, dysregulation of the interleukin (IL)-6-mediated JAK/STAT3 signaling pathway is closely related to the development of diverse human solid tumors including colorectal cancer (CRC). On this basis, modulation of the IL-6/JAK/STAT3 signaling pathway is currently being widely explored to develop novel therapies for CRC. The present review details the mechanisms and roles of the IL-6/JAK/STAT3 pathway in CRC, describes current therapeutic strategies, and the search for potential therapeutic approaches to treat CRC.
Collapse
Affiliation(s)
- Shu-Wei Wang
- Department of Minimally Invasive Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, P.R. China
| | - Yue-Ming Sun
- Department of Minimally Invasive Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, P.R. China
| |
Collapse
|
44
|
Vidal SJ, Rodriguez-Bravo V, Galsky M, Cordon-Cardo C, Domingo-Domenech J. Targeting cancer stem cells to suppress acquired chemotherapy resistance. Oncogene 2013; 33:4451-63. [PMID: 24096485 DOI: 10.1038/onc.2013.411] [Citation(s) in RCA: 191] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2013] [Revised: 08/12/2013] [Accepted: 08/12/2013] [Indexed: 12/18/2022]
Abstract
Acquired resistance has curtailed cancer survival since the dawn of the chemotherapy age more than half a century ago. Although the application of stem cell (SC) concepts to cancer captured the imagination of scientists for many years, only the last decade has yielded substantial evidence that cancer SCs (CSCs) contribute to chemotherapy resistance. Recent studies suggest that the functional and molecular properties of CSCs constitute therapeutic opportunities to improve the efficacy of chemotherapy. Here we review how these properties have stimulated combination strategies that suppress acquired resistance across a spectrum of malignancies. The clinical implementation of these strategies promises to rejuvenate the effort against an enduring challenge.
Collapse
Affiliation(s)
- S J Vidal
- 1] Department of Pathology, Mount Sinai Icahn School of Medicine, New York, NY, USA [2] Columbia University College of Physicians and Surgeons, New York, NY, USA
| | - V Rodriguez-Bravo
- Molecular Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - M Galsky
- Oncology Department, Tisch Cancer Institute, Mount Sinai Icahn School of Medicine, New York, NY, USA
| | - C Cordon-Cardo
- Department of Pathology, Mount Sinai Icahn School of Medicine, New York, NY, USA
| | - J Domingo-Domenech
- Department of Pathology, Mount Sinai Icahn School of Medicine, New York, NY, USA
| |
Collapse
|
45
|
Erb HHH, Langlechner RV, Moser PL, Handle F, Casneuf T, Verstraeten K, Schlick B, Schäfer G, Hall B, Sasser K, Culig Z, Santer FR. IL6 sensitizes prostate cancer to the antiproliferative effect of IFNα2 through IRF9. Endocr Relat Cancer 2013; 20:677-89. [PMID: 23913484 PMCID: PMC3753051 DOI: 10.1530/erc-13-0222] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Development and progression of prostate cancer (PCa) are associated with chronic inflammation. The cytokine interleukin 6 (IL6) can influence progression, differentiation, survival, and angiogenesis of PCa. To identify novel pathways that are triggered by IL6, we performed a gene expression profiling of two PCa cell lines, LNCaP and MDA PCa 2b, treated with 5 ng/ml IL6. Interferon (IFN) regulatory factor 9 (IRF9) was identified as one of the most prevalent IL6-regulated genes in both cell lines. IRF9 is a mediator of type I IFN signaling and acts together with STAT1 and 2 to activate transcription of IFN-responsive genes. The IL6 regulation of IRF9 was confirmed at mRNA and protein levels by quantitative real-time PCR and western blot respectively in both cell lines and could be blocked by the anti-IL6 antibody Siltuximab. Three PCa cell lines, PC3, Du-145, and LNCaP-IL6+, with an autocrine IL6 loop displayed high expression of IRF9. A tissue microarray with 36 PCa tissues showed that IRF9 protein expression is moderately elevated in malignant areas and positively correlates with the tissue expression of IL6. Downregulation and overexpression of IRF9 provided evidence for an IFN-independent role of IRF9 in cellular proliferation of different PCa cell lines. Furthermore, expression of IRF9 was essential to mediate the antiproliferative effects of IFNα2. We concluded that IL6 is an inducer of IRF9 expression in PCa and a sensitizer for the antiproliferative effects of IFNα2.
Collapse
Affiliation(s)
- Holger H H Erb
- Division of Experimental Urology, Department of UrologyInnsbruck Medical University6020, InnsbruckAustria
| | - Regina V Langlechner
- Division of Experimental Urology, Department of UrologyInnsbruck Medical University6020, InnsbruckAustria
| | - Patrizia L Moser
- Department of PathologyInnsbruck Medical University6020, InnsbruckAustria
| | - Florian Handle
- Division of Experimental Urology, Department of UrologyInnsbruck Medical University6020, InnsbruckAustria
| | - Tineke Casneuf
- Oncology Biomarkers, Janssen Research and DevelopmentBeerseBelgium
| | | | - Bettina Schlick
- Oncotyrol Center for Personalized Medicine6020, InnsbruckAustria
| | - Georg Schäfer
- Division of Experimental Urology, Department of UrologyInnsbruck Medical University6020, InnsbruckAustria
| | - Brett Hall
- Oncology Biomarkers Janssen Research and DevelopmentSpring House, PennsylvaniaUSA
| | - Kate Sasser
- Oncology Biomarkers Janssen Research and DevelopmentSpring House, PennsylvaniaUSA
| | - Zoran Culig
- Division of Experimental Urology, Department of UrologyInnsbruck Medical University6020, InnsbruckAustria
- Correspondence should be addressed to Z Culig or F R Santer Emails: or
| | - Frédéric R Santer
- Division of Experimental Urology, Department of UrologyInnsbruck Medical University6020, InnsbruckAustria
| |
Collapse
|
46
|
Agalliu I, Xue X, Cushman M, Cornell E, Hsing AW, Kaplan RC, Anastos K, Rajpathak S, Ho GYF. Detectability and reproducibility of plasma levels of chemokines and soluble receptors. RESULTS IN IMMUNOLOGY 2013; 3:79-84. [PMID: 24600562 DOI: 10.1016/j.rinim.2013.07.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Revised: 07/26/2013] [Accepted: 07/29/2013] [Indexed: 12/26/2022]
Abstract
BACKGROUND Multiplex assays are available to measure an array of circulating chemokines, soluble cytokine receptors and growth factors. However, there is limited information regarding whether these analytes are suitable for large-scale epidemiological studies to assess their relationships with chronic diseases, including cancer. METHODS We examined detectability, assay repeatability, and 3-year within-subject reproducibility of plasma levels of 25 chemokines and 11 soluble receptors of cytokines and growth factors selected from the Human Millipore Panels. Plasma samples were obtained from 36 men (average age 62 years) and 17 women (average age 32 years) who participated in two epidemiological studies. Inter-assay and within-subject reproducibility were assessed by intraclass correlation coefficients (ICC). RESULTS All analytes, except lymphotactin (47% detectability), were detectable in >90% of plasma samples. Inter-assay reproducibility for all analytes in 36 men tested three times on separate days were good to excellent (ICCs: 0.71-1.00). Within-subject reproducibility in 17 women sampled three times in three years were excellent (ICC ≥ 0.75) for five chemokines (eotaxin, fractalkine, 6Ckine, eotaxin 3, and SDF-1α+β) and three soluble receptors (sIL-1R2, sIL-4R and sVEGFR2); ICCs were fair to good (0.4 ≤ ICC < 0.75) for 15 chemokines and eight soluble receptors. However, five chemokines (GRO, IP-10, MIP-1β, BCA-1, and MIP-3α) had ICC < 0.4, suggesting biological variability. CONCLUSION Multiplex assays for plasma levels of selected chemokines and soluble receptors showed good to excellent assay detectability and repeatability. Most analytes also had good 3-year within-subject reproducibility, indicating that a single measurement of these analytes may be used to assess biomarker-disease associations.
Collapse
Affiliation(s)
- Ilir Agalliu
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Xiaonan Xue
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Mary Cushman
- Department of Medicine, University of Vermont, Colchester, VT, USA
| | - Elaine Cornell
- Department of Pathology, University of Vermont, Colchester, VT, USA
| | - Ann W Hsing
- Cancer Prevention Institute of California, Freemont, CA, USA ; Stanford Cancer Institute, Palo Alto, CA, USA
| | - Robert C Kaplan
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Kathryn Anastos
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA ; Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Swapnil Rajpathak
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA ; Current address: Merck & Co., Inc., Whitehouse Station, NJ, USA
| | - Gloria Y F Ho
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
47
|
Mechanisms by which interleukin-6 attenuates cell invasion and tumorigenesis in human bladder carcinoma cells. BIOMED RESEARCH INTERNATIONAL 2013; 2013:791212. [PMID: 23762858 PMCID: PMC3671296 DOI: 10.1155/2013/791212] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/25/2013] [Accepted: 04/23/2013] [Indexed: 12/27/2022]
Abstract
Interleukin-6, a multifunctional cytokine, contributes to tumor cell proliferation and differentiation. However, the biological mechanisms that are affected by the expression of interleukin-6 in bladder cancer cells remain unclear. We evaluated the effects of interleukin-6 expression in human bladder carcinoma cells in vitro and in vivo. The results of interleukin-6-knockdown experiments in T24 cells and interleukin-6-overexpression experiments in HT1376 cells revealed that interleukin-6 reduced cell proliferation, migration, and invasion in vitro. Xenograft animal studies indicated that the overexpression of interleukin-6 downregulated tumorigenesis of bladder cells and that interleukin-6 knockdown reversed this effect. The results of RT-PCR, immunoblotting, and reporter assays indicated that the overexpression of interleukin-6 upregulated the expression of the mammary serine protease inhibitor (MASPIN), N-myc downstream gene 1 (NDRG1), and KAI1 proteins in HT1376 cells and that interleukin-6 knockdown reduced the expression of these proteins in T24 cells. In addition, results of immunoblotting assays revealed that interleukin-6 modulated epithelial-mesenchymal transitions by upregulating the expression of the E-cadherin, while downregulation N-cadherin and vimentin proteins. Our results suggest that the effects of interleukin-6 on the regulation of epithelial-mesenchymal transitions and the expressions of the MASPIN, NDRG1, and KAI1 genes attribute to the modulation of tumorigenesis in human bladder carcinoma cells.
Collapse
|
48
|
Sofra M, Fei PC, Fabrizi L, Marcelli ME, Claroni C, Gallucci M, Ensoli F, Forastiere E. Immunomodulatory effects of total intravenous and balanced inhalation anesthesia in patients with bladder cancer undergoing elective radical cystectomy: preliminary results. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2013; 32:6. [PMID: 23374147 PMCID: PMC3577511 DOI: 10.1186/1756-9966-32-6] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/10/2012] [Accepted: 01/27/2013] [Indexed: 01/05/2023]
Abstract
Background Although surgery and anesthesia induce immunesuppression, remains largely unknown whether various anesthetic techniques have different immunosuppressive effects on cancer patients. Therefore, the aim of this study was to investigate the influence of total intravenous anesthesia with target-controlled infusion (TIVA-TCI) and balanced inhalation anesthesia (BAL) on the peri-operative levels of inflammatory cytokines and regulatory T cells (Tregs) in patients with bladder cancer undergoing surgery. Methods Twenty eight consecutive patients with bladder cancer who underwent radical cystectomy were prospectively randomized into two groups to receive TIVA-TCI (n = 14) or BAL (n = 14). Before the induction of anesthesia (T0), 6–8 hours (T1) post-surgery, and 5 days post-surgery (T2), Tregs and serum levels of interleukin -1beta (IL-1β), interferon-gamma (IFN-γ), tumor necrosis factor-alpha (TNF-α), interleukin −2 (IL-2), interleukin −6 (IL-6), and interleukin −10 (IL-10) were measured. Results In the peri-operative period all cancer patients showed a marked and significant increase in IL-6. Moreover, TIVA-TCI patients also showed a higher increase in IFN-γ, whereas in BAL patients Tregs were reduced by approximately 30% during surgery. The incidence of infections, metastases, and death was similar in both groups. Conclusions The increase in the Th1 response in the TIVA-TCI group and the reduction in Tregs in the BAL group seem to balance the immunosuppressive effect induced by IL-6. Therefore TIVA-TCI and BAL can be both used in major surgery in patients with bladder cancer without worsening the outcome.
Collapse
Affiliation(s)
- Maria Sofra
- Department of Anesthesiology, Regina Elena, Rome National Cancer Institute, Via Elio Chianesi 53, 00144, Rome, Italy
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Shen Z, Ye Y, Kauttu T, Seppänen H, Vainionpää S, Wang S, Mustonen H, Puolakkainen P. The novel focal adhesion gene kindlin-2 promotes the invasion of gastric cancer cells mediated by tumor-associated macrophages. Oncol Rep 2012; 29:791-7. [PMID: 23151599 DOI: 10.3892/or.2012.2137] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2012] [Accepted: 09/20/2012] [Indexed: 12/13/2022] Open
Abstract
Kindlin-2 is a novel focal adhesion gene mediating the cell-extracellular matrix (ECM) adhesion. Tumor-associated macrophages (TAMs) play an important role in linking chronic inflammation to cancer progression. Both kindlin-2 and TAMs have been found to promote the invasion of gastric cancer cells in our previous studies. However, the correlation between kindlin-2 and TAMs remains unclear. Real-time RT-PCR was used to investigate kindlin-2 expression in the AGS, NCI and Hs-746T gastric cancer cell lines co-cultured with TAMs under normal or hypoxic conditions. IL8, IL10, IL11, IL17b, IL18, IL22 and IL24 expressions were measured by real-time RT-PCR in the gastric cancer lines with varying levels of kindlin-2 expression, as well as after downregulation of kindlin-2 mRNA expression by the siRNA method. We found that kindlin-2 was upregulated in all three gastric cancer cell lines when co-cultured with TAMs under normal conditions. Under hypoxic conditions, the induction of kindlin-2 expression induced by macrophages was significantly downregulated in the Hs-746T cell line. IL8, IL11, IL17b, IL22 and IL24 expression was significantly higher in gastric cell lines with high kindlin-2 expression. Downregulation of kindlin-2 mRNA decreased IL10, IL11, IL17b, IL22 and IL24 expression but IL8 and IL18 expression was upregulated. Therefore, the novel focal adhesion gene kindlin-2 may play an important role in promoting the invasion of gastric cancer cells mediated by TAMs through regulating interleukin expression.
Collapse
Affiliation(s)
- Zhanlong Shen
- Department of Gastroenterological Surgery, Peking University People's Hospital, Beijing 100044, PR China
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Tsui KH, Chang YL, Feng TH, Chung LC, Lee TY, Chang PL, Juang HH. Growth differentiation factor-15 upregulates interleukin-6 to promote tumorigenesis of prostate carcinoma PC-3 cells. J Mol Endocrinol 2012; 49:153-63. [PMID: 22872134 DOI: 10.1530/jme-11-0149] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Growth differentiation factor-15 (GDF15), a member of the transforming growth factor-β superfamily, is associated with human cancer progress. We evaluated the role GDF15 plays in tumorigenesis of prostate carcinoma PC-3 cells. Results from real-time RT-PCR and ELISA revealed that expression of GDF15 was approximately threefold higher in LNCaP cells than in PC-3 cells. Other prostate cell lines (PZ-HPV-7, CA-HPV-10, and DU145 cells) expressed extremely low levels of GDF15. Stable overexpression of GDF15 in PC-3 cells enhanced the degree of cell proliferation and invasion as shown in the (3)H-thymidine incorporation assay and in the Matrigel invasion assay respectively. Soft agar assays and xenograft animal studies indicated that overexpression of GDF15 in PC-3 cells increased tumorigenesis in vitro and in vivo. Results from RT-PCR, immunoblot, and reporter assays revealed that overexpression of GDF15 resulted in decreased expression of maspin and upregulation of interleukin-6 (IL6), matriptase, and N-myc downstream-regulated gene 1 (NDRG1) expression. Further studies revealed that overexpression of IL6 enhanced GDF15 expression in LNCaP cells while knockdown of IL6 blocked the expression of GDF15 in PC-3 cells, suggesting that expression of GDF15 is upregulated by IL6. This study demonstrated that expression of GDF15 induces cell proliferation, invasion, and tumorigenesis of prostate carcinoma PC-3 cells. The enhancement of tumorigenesis and invasiveness of prostate carcinoma cells that stably overexpress GDF15 may be caused by the dysregulation of maspin, matriptase, and IL6 gene expression. The expression of GDF15 and IL6 is controlled via a positive feedback loop in PC-3 cells.
Collapse
Affiliation(s)
- Ke-Hung Tsui
- Department of Urology Bioinformation Center, Chang Gung Memorial Hospital, Kwei-Shan, Tao-Yuan, Taiwan
| | | | | | | | | | | | | |
Collapse
|