1
|
Liu XQ, Shao XR, Liu Y, Dong ZX, Chan SH, Shi YY, Chen SN, Qi L, Zhong L, Yu Y, Lv T, Yang PF, Li LY, Wang XB, Zhang XD, Li X, Zhao W, Sehgal L, Li M, Zhang XD. Tight junction protein 1 promotes vasculature remodeling via regulating USP2/TWIST1 in bladder cancer. Oncogene 2022; 41:502-514. [PMID: 34782718 DOI: 10.1038/s41388-021-02112-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 10/27/2021] [Accepted: 11/01/2021] [Indexed: 12/15/2022]
Abstract
Bladder cancer (BLCA) is the most common malignant tumor of the urinary system and is characterized by high metastatic rates and poor prognosis. The expression of tight junction protein 1 (TJP1) is associated with bladder cancer invasion; however, the mechanism by which TJP1 affects vasculature remodeling remains unknown. In this study, we found that TJP1 expression correlated with tumor angiogenesis and poor overall survival in clinical samples. Furthermore, TJP1 overexpression promoted tumor angiogenesis in BLCA cells and stimulated recruitment of macrophages to tumors by upregulating CCL2 expression. Mechanistically, TJP1 interacted with TWIST1 and enhanced the transcriptional activity of CCL2. The impairment of tumor angiogenesis caused by knockdown of TJP1 was dramatically rescued by overexpression of TWIST1. Furthermore, TJP1 recruited USP2, which deubiquitinated TWIST1, thereby protecting TWIST1 from proteasome-mediated protein degradation. In conclusion, our results suggest that TJP1 controls angiogenesis in BLCA via TWIST1-dependent regulation of CCL2. We demonstrate that TJP1 functions as a scaffold for the interaction between USP2 and TWIST1 and this may provide potential therapeutic targets in bladder cancer.
Collapse
Affiliation(s)
- Xue-Qi Liu
- Molecular Cancer Research Center, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, China
| | - Xin-Rong Shao
- Molecular Cancer Research Center, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, China
| | - Ye Liu
- Department of Pathology, Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Zhao-Xia Dong
- Molecular Cancer Research Center, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, China
| | - Sze-Hoi Chan
- Molecular Cancer Research Center, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, China
| | - Yuan-Yuan Shi
- Molecular Cancer Research Center, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, China
| | - Shu-Na Chen
- Molecular Cancer Research Center, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, China
| | - Lin Qi
- Molecular Cancer Research Center, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, China
| | - Li Zhong
- State Key Laboratory of Oncology in South China, Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yue Yu
- Molecular Cancer Research Center, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, China
| | - Ting Lv
- Department of Pathology, Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Peng-Fei Yang
- Department of Pathology, Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Li-Yan Li
- Molecular Cancer Research Center, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, China
| | - Xiao-Bin Wang
- Andrology section, Center of Reproductive Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xu-Dong Zhang
- Molecular Cancer Research Center, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, China
| | - Xin Li
- Molecular Cancer Research Center, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, China
| | - Wenxue Zhao
- Molecular Cancer Research Center, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, China
| | - Lalit Sehgal
- Division of Hematology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Miao Li
- The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China. .,Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen, China.
| | - Xing-Ding Zhang
- Molecular Cancer Research Center, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, China.
| |
Collapse
|
2
|
Liu G, Zeng T. Sporoderm-Removed Ganoderma lucidum Spore Powder May Suppress the Proliferation, Migration, and Invasion of Esophageal Squamous Cell Carcinoma Cells Through PI3K/AKT/mTOR and Erk Pathway. Integr Cancer Ther 2021; 20:15347354211062157. [PMID: 34841952 PMCID: PMC8649442 DOI: 10.1177/15347354211062157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Tumor metastasis is a key factor of therapeutic failure in tumor patients, but the underlying molecular mechanism remains to be explored and novel effective curative strategies are urgently required. Emerging evidence suggests that sporoderm-removed Ganoderma lucidum spore powder can suppress tumor growth and metastasis. However, the molecular mechanisms of action remain elusive. In the present study, we investigated the effects and mechanisms of sporoderm-removed Ganoderma lucidum spore powder against esophageal squamous cell carcinomas (ESCC). The expression of MCP-1 in esophageal squamous cell carcinoma cells was detected by Western blotting. The MTS assay was used to assess the esophageal squamous cell carcinoma cells viability. The clone formation assay was used to evaluate to the proliferation ability of KYSE140 and KYSE510 cells. Apoptosis and the cell cycle were analyzed by flow cytometry. Wound healing and Transwell assays were used to analyze the migration of KYSE140 and KYSE510 cells. Invasion was also analyzed by the Transwell assay. The expressions of PI3K, AKT/p-AKT, Erk/p-Erk, JNK1, and mTOR were detected by Western blotting. We found that the MCP-1 protein was highly expressed in KYSE140 and KYSE510. In addition, sporoderm-removed Ganoderma lucidum spore powder treatment was found to inhibit esophageal squamous cell carcinoma cell proliferation, to block the cell cycle, to induce cell apoptosis and to inhibit cell migration and invasion. Finally, we found that sporoderm-removed Ganoderma lucidum spore powder decreased the expression of PI3K/AKT/mTOR and Erk signaling pathways. Taken together, these findings demonstrate that sporoderm-removed Ganoderma lucidum spore powder suppresses esophageal squamous cell carcinomas by involving MCP-1, regulated by PI3K/AKT/mTOR and Erk signal pathways.
Collapse
Affiliation(s)
- Guiping Liu
- Department of Medical Laboratory, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, P.R. China.,Department of Laboratory Medicine, Peking University Shenzhen Hospital, Shenzhen, Guangdong, P.R. China
| | - Tao Zeng
- Department of Medical Laboratory, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, P.R. China
| |
Collapse
|
3
|
Ita MI, Wang JH, Fanning N, Kaar G, Lim C, Redmond HP. Plasma circulating cell free messenger RNA as a potential biomarker of melanoma. Acta Oncol 2021; 60:1201-1209. [PMID: 34086522 DOI: 10.1080/0284186x.2021.1928749] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
BACKGROUND Blood borne cell free nucleic acids are increasingly emerging as significant non-invasive adjuncts to current methods of disease status evaluation in cancer patients. In this study, we sought to examine whether significant differences exist in the plasma transcriptomic profile of advanced melanoma patients with a high disease burden compared to patients with a low disease burden or therapeutic response. METHODS Pathway focussed gene expression analysis was performed using cDNA derived from the plasma circulating cell free messenger ribonucleic acid (ccfmRNA) samples of twenty-two patients with advanced melanoma. Patients were assessed with paired blood sample collection and CT scan assessments at baseline and at 3 months follow up. RESULTS We identified several genes which were significantly over-expressed in patients with a low disease burden or therapeutic response; BCL2L1, CXCL9, IDO1, IL13, MIF, MYD88 and TLR4 (p ≤ 0.001, versus high disease burden). There was an increase in the magnitude of fold change (2^ (-dd CT)) of BCL2L1 (p = 0.031), CCL4 (p = 0.001), CCL5 (p = 0.043), CXCL9 (p = 0.012), GZMB (p = 0.023) and TNFSF10 (p = 0.039) genes in patients with therapeutic response at 3 months follow up assessment relative to baseline assessment. Moreover, in stage IV melanoma patients with brain metastases, CCL18, CCR1, CCR4, CD274, CSF2, EGF, and PTGS2 genes were significantly over-expressed (p < 0.001, versus patients without melanoma brain metastasis). CONCLUSION Significant differences were observed in the plasma transcriptomic profile between the various melanoma patient groups, and we postulate that these differences may be exploited to identify novel therapeutic targets or biomarkers relevant to melanoma.
Collapse
Affiliation(s)
- Michael Itak Ita
- Department of Academic Surgery, University College Cork, Cork, Ireland
- Department of Neurosurgery, University College Cork, Cork, Ireland
| | - Jiang Huai Wang
- Department of Academic Surgery, University College Cork, Cork, Ireland
| | - Noel Fanning
- Department of Radiology, University College Cork, Cork, Ireland
| | - George Kaar
- Department of Neurosurgery, University College Cork, Cork, Ireland
| | - Chris Lim
- Department of Neurosurgery, University College Cork, Cork, Ireland
| | | |
Collapse
|
4
|
Myakoshina EB, Kulikova IG, Balatskaya NV, Katargina LA, Saakyan SV. [The role of CXC and CC chemokines in patients with uveal melanoma]. Vestn Oftalmol 2021; 137:31-37. [PMID: 34410054 DOI: 10.17116/oftalma202113704131] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Uveal melanoma is a malignant neoplasm with high metastatic potential; its pathogenesis is currently being studied. Chemokines play a key role not only in the inflammatory response, but also in enhancing angiogenesis, tumor invasiveness, increasing proliferative potential and metastasis. PURPOSE To study the role of chemokines of classes CXC and CC in blood serum and tear fluid of patients with uveal melanoma. MATERIAL AND METHODS The study included 118 people aged 53.7±12.2 years, among them 80 patients with uveal melanoma and 38 healthy donors. Group 1 included 32 patients with small tumors, group 2 (medium-sized tumors) - 26 patients; group 3 (large tumors) was comprised of 22 patients. Chemokines of classes CC (CCL2/MCP-1, CCL3/MIP-1α, CCL4/MIP-1β, CCL5/RANTES, CCL11/Eotaxin) and CXC (CXCL1/GRO-α, CXCL8/IL-8, CXCL10/IP-10, CXCL12/SDF-1α) were determined by multiplex analysis of the blood serum and tear fluid. Statistical processing: Student's t-test, Fisher criteria, and Pierson's chi-squared test (χ2), differences were considered significant at p<0.05. RESULTS Significantly increased level of chemokines with pro-inflammatory (CCL5/RANTES), proliferative (CXCL10/IP-10) and pro-angiogenic (CXCL12/SDF-1α) effects was found in the blood serum of patients with small-sized uveal melanoma in comparison with healthy donors. Concentration of all studied pro-inflammatory, proliferative, and pro-angiogenic chemokines in the lacrimal fluid was found to be significantly elevated in both the affected and the paired "healthy" eyes in all 3 groups of patients, with the maximum content seen in the large tumor group. CONCLUSION The obtained data indicates that early local and systemic immune imbalance can be observed in uveal melanoma, and detection of chemokines can serve as a good reason for developing targeted therapy for small uveal melanoma.
Collapse
Affiliation(s)
- E B Myakoshina
- Helmholtz National Medical Research Center of Eye Diseases, Moscow, Russia
| | - I G Kulikova
- Helmholtz National Medical Research Center of Eye Diseases, Moscow, Russia
| | - N V Balatskaya
- Helmholtz National Medical Research Center of Eye Diseases, Moscow, Russia
| | - L A Katargina
- Helmholtz National Medical Research Center of Eye Diseases, Moscow, Russia
| | - S V Saakyan
- Helmholtz National Medical Research Center of Eye Diseases, Moscow, Russia
| |
Collapse
|
5
|
Oweida A, Paquette B. Reconciling two opposing effects of radiation therapy: stimulation of cancer cell invasion and activation of anti-cancer immunity. Int J Radiat Biol 2021; 99:951-963. [PMID: 34264178 DOI: 10.1080/09553002.2021.1956005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
PURPOSE The damage caused by radiation therapy to cancerous and normal cells inevitably leads to changes in the secretome profile of pro and anti-inflammatory mediators. The inflammatory response depends on the dose of radiation and its fractionation, while the inherent radiosensitivity of each patient dictates the intensity and types of adverse reactions. This review will present an overview of two apparently opposite reactions that may occur after radiation treatment: induction of an antitumor immune response and a protumoral response. Emphasis is placed on the molecular and cellular mechanisms involved. CONCLUSIONS By understanding how radiation changes the balance between anti- and protumoral effects, these forces can be manipulated to optimize radiation oncology treatments.
Collapse
Affiliation(s)
- Ayman Oweida
- Department of Nuclear Medicine and Radiobiology, Faculty of Medicine and Health Sciences, Universite de Sherbrooke, Sherbrooke, Canada
| | - Benoit Paquette
- Department of Nuclear Medicine and Radiobiology, Faculty of Medicine and Health Sciences, Universite de Sherbrooke, Sherbrooke, Canada
| |
Collapse
|
6
|
Wong W, Alouani E, Wei A, Ryu YK, Chabot JA, Manji GA. Future of immunotherapy in pancreas cancer and the trials, tribulations and successes thus far. Semin Oncol 2021; 48:57-68. [PMID: 33965249 DOI: 10.1053/j.seminoncol.2021.02.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 02/02/2021] [Indexed: 11/11/2022]
Abstract
Pancreas ductal adenocarcinoma (PDAC) has a dismal prognosis with a 5-year survival rate of 10%. Currently, chemotherapy remains the standard of care for systemic treatment. Immunotherapy with checkpoint inhibitors unfortunately has not been found to be effective in the treatment of PDAC to date, likely due to the highly desmoplastic and immunosuppressive tumor microenvironment (TME). Treatment targeting pathways against the immunosuppressive mechanisms of PDAC are of mounting interest to improve outcomes in PDAC. In this review, we discuss prior efforts and the current state of immunotherapy in PDAC. We will also review the emerging targets and treatments with significant clinical potential for the treatment of PDAC such as: CD40 pathway, the adenosine pathway, the CXCR4/CXCL12 axis, the CCR2/CCL2 axis, IDO pathway, and others.
Collapse
Affiliation(s)
- Winston Wong
- Division of Hematology and Oncology, Columbia University Irving Medical Center, and New York Presbyterian Hospital, New York, NY
| | - Emily Alouani
- Division of Hematology and Oncology, Columbia University Irving Medical Center, and New York Presbyterian Hospital, New York, NY
| | - Alexander Wei
- Division of Hematology and Oncology, Columbia University Irving Medical Center, and New York Presbyterian Hospital, New York, NY
| | - Yun Kyoung Ryu
- Division of Hematology and Oncology, Columbia University Irving Medical Center, and New York Presbyterian Hospital, New York, NY
| | - John A Chabot
- Division of Hematology and Oncology, Columbia University Irving Medical Center, and New York Presbyterian Hospital, New York, NY
| | - Gulam A Manji
- Division of Hematology and Oncology, Columbia University Irving Medical Center, and New York Presbyterian Hospital, New York, NY.
| |
Collapse
|
7
|
Noel M, O'Reilly EM, Wolpin BM, Ryan DP, Bullock AJ, Britten CD, Linehan DC, Belt BA, Gamelin EC, Ganguly B, Yin D, Joh T, Jacobs IA, Taylor CT, Lowery MA. Phase 1b study of a small molecule antagonist of human chemokine (C-C motif) receptor 2 (PF-04136309) in combination with nab-paclitaxel/gemcitabine in first-line treatment of metastatic pancreatic ductal adenocarcinoma. Invest New Drugs 2020; 38:800-811. [PMID: 31297636 PMCID: PMC7211198 DOI: 10.1007/s10637-019-00830-3] [Citation(s) in RCA: 116] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 07/01/2019] [Indexed: 12/20/2022]
Abstract
Background In pancreatic ductal adenocarcinoma (PDAC), the chemokine (C-C motif) ligand 2 (CCL2)/chemokine (C-C motif) receptor 2 (CCR2) axis plays a key role in immunosuppressive properties of the tumor microenvironment, patient prognosis, and chemoresistance. This phase Ib study assessed the effects of the orally administered CCR2 inhibitor PF-04136309 in combination with nab-paclitaxel and gemcitabine in patients with previously untreated metastatic PDAC. Methods Patients received PF-04136309 twice daily (BID) continuously plus nab-paclitaxel (125 mg/m2) and gemcitabine (1000 mg/m2) administered on days 1, 8, and 15 of each 28-day cycle. The primary objectives were to evaluate safety and tolerability, characterize dose-limiting toxicities (DLTs), and determine the recommended phase II dose (RP2D) of PF-04136309. Results In all, 21 patients received PF-04136309 at a starting dose of 500 mg or 750 mg BID. The RP2D was identified to be 500 mg BID. Of 17 patients treated at the 500 mg BID starting dose, three (17.6%) experienced a total of four DLTs, including grade 3 dysesthesia, diarrhea, and hypokalemia and one event of grade 4 hypoxia. Relative to the small number of patients (n = 21), a high incidence (24%) of pulmonary toxicity was observed in this study. The objective response rate for 21 patients was 23.8% (95% confidence interval: 8.2-47.2%). Levels of CD14 + CCR2+ inflammatory monocytes (IM) decreased in the peripheral blood, but did not accumulate in the bone marrow. Conclusions PF-04136309 in combination with nab-paclitaxel plus gemcitabine had a safety profile that raises concern for synergistic pulmonary toxicity and did not show an efficacy signal above nab-paclitaxel and gemcitabine. ClinicalTrials.gov identifier: NCT02732938.
Collapse
Affiliation(s)
- Marcus Noel
- Department of Medicine, Division of Hematology/Oncology, University of Rochester Medical Center School of Medicine & Dentistry, Rochester, NY, USA
| | - Eileen M O'Reilly
- Department of Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell Medical College, New York, NY, USA
| | - Brian M Wolpin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - David P Ryan
- MGH Cancer Center, Division of Hematogy-Oncology, Massachusetts General Hospital, Boston, MA, USA
| | - Andrea J Bullock
- Division of Hematology/Oncology, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Carolyn D Britten
- Division of Hematology/Oncology, Medical University of South Carolina, Charleston, SC, USA
| | - David C Linehan
- Department of Surgery, University of Rochester Medical Center School of Medicine & Dentistry, Rochester, NY, USA
| | - Brian A Belt
- Department of Surgery, University of Rochester, Rochester, NY, USA
| | - Eric C Gamelin
- Early Oncology Development and Clinical Research, Pfizer Inc, 219 East 42nd Street, New York, NY, 10017, USA
| | - Bishu Ganguly
- Early Oncology Development and Clinical Research, Pfizer Inc, 219 East 42nd Street, New York, NY, 10017, USA
- Lyell Immunopharma Inc, Palo Alto, CA, USA
| | - Donghua Yin
- Early Oncology Development and Clinical Research, Pfizer Inc, 219 East 42nd Street, New York, NY, 10017, USA
| | - Tenshang Joh
- Early Oncology Development and Clinical Research, Pfizer Inc, 219 East 42nd Street, New York, NY, 10017, USA
| | - Ira A Jacobs
- Early Oncology Development and Clinical Research, Pfizer Inc, 219 East 42nd Street, New York, NY, 10017, USA.
| | - Carrie T Taylor
- Early Oncology Development and Clinical Research, Pfizer Inc, 219 East 42nd Street, New York, NY, 10017, USA
| | - Maeve A Lowery
- Trinity St James's Cancer Institute, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
8
|
Pereira FV, Melo ACL, Silva MB, de Melo FM, Terra FF, Castro IA, Perandini LA, Miyagi MT, Sato FT, Origassa CST, Hiyane MI, Donato J, Wasinski F, Araujo RC, Festuccia WTL, da Silva JS, Camara NOS. Interleukin-6 and the Gut Microbiota Influence Melanoma Progression in Obese Mice. Nutr Cancer 2020; 73:642-651. [DOI: 10.1080/01635581.2020.1764982] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Affiliation(s)
- Felipe V. Pereira
- Laboratory of Transplantation Immunobiology, Department of Immunology, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, São Paulo, Brazil
| | - Amanda C. L. Melo
- Laboratory of Transplantation Immunobiology, Department of Immunology, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, São Paulo, Brazil
| | - Marina B. Silva
- Laboratory of Transplantation Immunobiology, Department of Immunology, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, São Paulo, Brazil
| | - Filipe M. de Melo
- Department of Microbiology, Immunology and Parasitology, Escola Paulista de Medicina- Universidade Federal de São Paulo (EPM-UNIFESP), São Paulo, São Paulo, Brazil
| | - Fernanda F. Terra
- Laboratory of Transplantation Immunobiology, Department of Immunology, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, São Paulo, Brazil
| | - Iris A. Castro
- Laboratory of Transplantation Immunobiology, Department of Immunology, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, São Paulo, Brazil
| | - Luiz A. Perandini
- Laboratory of Transplantation Immunobiology, Department of Immunology, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, São Paulo, Brazil
| | - Marcelli T. Miyagi
- Laboratory of Transplantation Immunobiology, Department of Immunology, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, São Paulo, Brazil
| | - Fabio T. Sato
- Institute of Biology, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Clarice S. T. Origassa
- Laboratory of Transplantation Immunobiology, Department of Immunology, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, São Paulo, Brazil
| | - Meire I. Hiyane
- Laboratory of Transplantation Immunobiology, Department of Immunology, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, São Paulo, Brazil
| | - Jose Donato
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, São Paulo, Brazil
| | - Frederick Wasinski
- Department of Biophysics, Federal University of São Paulo, São Paulo, São Paulo, Brazil
| | - Ronaldo C. Araujo
- Department of Biophysics, Federal University of São Paulo, São Paulo, São Paulo, Brazil
| | - William T. L. Festuccia
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, São Paulo, Brazil
| | - Joao Santana da Silva
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Niels Olsen S. Camara
- Laboratory of Transplantation Immunobiology, Department of Immunology, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, São Paulo, Brazil
| |
Collapse
|
9
|
López González M, van de Ven R, de Haan H, van Eck van der Sluijs J, Dong W, van Beusechem VW, de Gruijl TD. Oncolytic adenovirus ORCA-010 increases the type 1 T cell stimulatory capacity of melanoma-conditioned dendritic cells. Clin Exp Immunol 2020; 201:145-160. [PMID: 32301504 PMCID: PMC7366753 DOI: 10.1111/cei.13442] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Revised: 03/25/2020] [Accepted: 03/26/2020] [Indexed: 12/16/2022] Open
Abstract
Immune checkpoint blockade has resulted in durable responses in patients with metastatic melanoma, but only in a fraction of treated patients. For immune checkpoint inhibitors (ICI) to be effective, sufficient infiltration with tumor‐reactive T cells is essential. Oncolytic viruses (OV) selectively replicate in and lyse tumor cells and so induce an immunogenic form of cell death, providing at once a source of tumor‐associated (neo)antigens and of danger signals that together induce effective T cell immunity and tumor infiltration. Melanoma‐associated suppression of dendritic cell (DC) differentiation effectively hampers OV‐ or immune checkpoint inhibitor (ICI)‐induced anti‐tumor immunity, due to a consequent inability to prime and attract anti‐tumor effector T cells. Here, we set out to study the effect of ORCA‐010, a clinical stage oncolytic adenovirus, on DC differentiation and functionality in the context of human melanoma. In melanoma and monocyte co‐cultures, employing a panel of five melanoma cell lines with varying origins and oncogenic mutation status, we observed clear suppression of DC development with apparent skewing of monocyte differentiation to a more M2‐macrophage‐like state. We established the ability of ORCA‐010 to productively infect and lyse the melanoma cells. Moreover, although ORCA‐010 was unable to restore DC differentiation, it induced activation and an increased co‐stimulatory capacity of monocyte‐derived antigen‐presenting cells. Their subsequent ability to prime effector T cells with a type I cytokine profile was significantly increased in an allogeneic mixed leukocyte reaction. Our findings suggest that ORCA‐010 is a valuable immunotherapeutic agent for melanoma.
Collapse
Affiliation(s)
- M López González
- Department of Medical Oncology, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam, the Netherlands
| | - R van de Ven
- Department of Medical Oncology, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam, the Netherlands.,Otolaryngology/Head-Neck Surgery, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam, the Netherlands
| | - H de Haan
- Department of Medical Oncology, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam, the Netherlands
| | - J van Eck van der Sluijs
- Department of Medical Oncology, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam, the Netherlands
| | - W Dong
- ORCA Therapeutics, 's-Hertogenbosch, the Netherlands
| | - V W van Beusechem
- Department of Medical Oncology, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam, the Netherlands.,ORCA Therapeutics, 's-Hertogenbosch, the Netherlands
| | - T D de Gruijl
- Department of Medical Oncology, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam, the Netherlands
| |
Collapse
|
10
|
Mesenchymal stem/stromal cells stably transduced with an inhibitor of CC chemokine ligand 2 ameliorate bronchopulmonary dysplasia and pulmonary hypertension. Cytotherapy 2020; 22:180-192. [PMID: 32139242 DOI: 10.1016/j.jcyt.2020.01.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 01/21/2020] [Accepted: 01/22/2020] [Indexed: 12/20/2022]
Abstract
Perinatal bronchopulmonary dysplasia (BPD) is defined as lung injury in preterm infants caused by various factors, resulting in serious respiratory dysfunction and high mortality. The administration of mesenchymal stem/stromal cells (MSCs) to treat/prevent BPD has proven to have certain therapeutic effects. However, MSCs can only weakly regulate macrophage function, which is strongly involved in the development of BPD. 7ND-MSCs are MSCs transfected with 7ND, a truncated version of CC chemokine ligand 2 (CCL2) that promotes macrophage activation, using a lentiviral vector. In the present study, we show in a BPD rat model that 7ND-MSC administration, but not MSCs alone, ameliorated the impaired alveolarization evaluated by volume density and surface area in the lung tissue, as well as pulmonary artery remodeling and pulmonary hypertension induced by BPD. In addition, 7ND-MSCs, but not MSCs alone, reduced M1 macrophages and the messenger RNA expressions of interleukin-6 and CCL2 in the lung tissue. Thus, the present study showed the treatment effect of 7ND-MSCs in a BPD rat model, which was more effective than that of MSCs alone.
Collapse
|
11
|
Zhang J, Endres S, Kobold S. Enhancing tumor T cell infiltration to enable cancer immunotherapy. Immunotherapy 2020; 11:201-213. [PMID: 30730277 DOI: 10.2217/imt-2018-0111] [Citation(s) in RCA: 88] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Cancer immunotherapy has changed the treatment landscape for cancer patients, especially for those with metastatic spread. While the immunotherapeutic armamentarium is constantly growing, as exemplified by approved compounds, clinical outcome remains variable both within and across entities. A sufficient infiltration into the tumor microenvironment and successful activation of effector T lymphocytes against tumor cells have been identified as predictors for responses to T cell-based immunotherapies. However, tumor cells have developed a variety of mechanisms to reduce T cell homing and access to the tumor tissue to prevent activity of anticancer immunity. As a consequence, investigations have interrogated strategies to improve the efficacy of cancer immunotherapies by enhancing T cell infiltration into tumor tissues. In this review, we summarize mechanisms of how tumor tissue shapes immune suppressive microenvironment to prevent T cell access to the tumor site. We focus on current strategies to improve cancer immunotherapies through enhancing T cell infiltration.
Collapse
Affiliation(s)
- Jin Zhang
- Center of Integrated Protein Science Munich (CIPS-M) & Division of Clinical Pharmacology, Klinikum der Universität München, Lindwurmstrasse 2a, 80337 Munich, Germany, Member of the German Center of Lung Research
| | - Stefan Endres
- Center of Integrated Protein Science Munich (CIPS-M) & Division of Clinical Pharmacology, Klinikum der Universität München, Lindwurmstrasse 2a, 80337 Munich, Germany, Member of the German Center of Lung Research
| | - Sebastian Kobold
- Center of Integrated Protein Science Munich (CIPS-M) & Division of Clinical Pharmacology, Klinikum der Universität München, Lindwurmstrasse 2a, 80337 Munich, Germany, Member of the German Center of Lung Research
| |
Collapse
|
12
|
Long-Term Local Injection of RAGE-Aptamer Suppresses the Growth of Malignant Melanoma in Nude Mice. JOURNAL OF ONCOLOGY 2019; 2019:7387601. [PMID: 31565056 PMCID: PMC6746150 DOI: 10.1155/2019/7387601] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 05/12/2019] [Accepted: 08/08/2019] [Indexed: 02/06/2023]
Abstract
Accumulating evidence has suggested the pathological role of advanced glycation end products (AGEs) and their receptor RAGE axis in aging-associated disorders, including cancers. In this study, we examined the effects of local injection of RAGE-aptamer adjacent to the tumor on G361 melanoma growth in nude mice. We further investigated the effects of RAGE-aptamer on oxidative stress generation, RAGE, vascular endothelial growth factor (VEGF), and monocyte chemoattractant protein-1 (MCP-1) gene expression in Nε-(carboxymethyl)lysine (CML)-exposed G361 melanoma cells in vitro. Local injection of RAGE-aptamer adjacent to the tumor dramatically decreased the growth of G361 melanoma in nude mice, which was associated with reduced expression of CML, RAGE, nitrotyrosine, VEGF, CD31, and von Willebrand factor, markers of endothelial cells in G361 tumors. Furthermore, RAGE-aptamer inhibited the binding of CML to V-domain of RAGE and blocked the CML-induced increases in oxidative stress generation, RAGE, VEGF, and MCP-1 mRNA levels in G361 melanoma cells. Our present findings suggest that long-term local injection of RAGE-aptamer adjacent to the tumor could inhibit melanoma growth in nude mice partly by suppressing tumor angiogenesis via blockade of the CML-RAGE interaction. Local injection of RAGE-aptamer may be a feasible therapeutic tool for the treatment of malignant melanoma.
Collapse
|
13
|
Thymoquinone Enhances the Effect of Gamma Knife in B16-F10 Melanoma Through Inhibition of Phosphorylated STAT3. World Neurosurg 2019; 128:e570-e581. [PMID: 31054338 DOI: 10.1016/j.wneu.2019.04.205] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Revised: 04/20/2019] [Accepted: 04/22/2019] [Indexed: 01/26/2023]
Abstract
BACKGROUND Patients with brain metastasis from melanoma have a dismal prognosis with poor survival time. Gamma Knife (GK) is an effective treatment to control brain metastasis from melanoma. Thymoquinone (TQ) has emerged as a potential therapeutic option due to its antiproliferative effects on various cancers. The purpose of the study was to assess the effect of GK on B16-F10 melanoma cells in vitro and intracerebral melanoma in vivo, and its synergistic effect in combination with TQ. METHODS The effects of GK and combination treatment of GK and TQ were studied on B16-F10 melanoma cells by evaluating cytotoxicity with an adenosine triphosphate assay, apoptosis by acridine orange staining, and genotoxicity by comet assay. Western blot analysis was performed to investigate the expression of STAT3, p-STAT3 (Tyr705), JAK2, p-JAK2, caspase-3, Bax, Bcl-2, survivin, and β-actin. Expression of inflammatory cytokines was assessed by enzyme-linked immunosorbent assay. GK alone and in combination with TQ was assessed in an established intracerebral melanoma tumor in mice. RESULTS The effects of GK on cytotoxicity, genotoxicity, and apoptosis were enhanced by TQ in B16-F10 melanoma cells. GK induced apoptosis through inhibition of p-STAT3 expression, which in turn regulated pro- and antiapoptotic proteins such as caspase-3, Bax, Bcl-2, and survivin. Adding TQ to GK irradiation further enhanced this apoptotic effect of GK irradiation. GK was shown to reduce the levels of tumor-related inflammatory cytokines in B16-F10 melanoma cells. This effect was more pronounced when TQ was added to GK irradiation. GK with 15 Gy increased the survival of mice with intracerebral melanoma compared with untreated mice. However, despite the additive effect of TQ in addition to GK irradiation on B16-F10 melanoma cells in vitro, TQ did not add any significant survival benefit to GK treatment in mice with intracerebral melanoma. CONCLUSIONS Our findings suggest that TQ would be a potential therapeutic agent in addition to GK to enhance the antitumor effect of irradiation. Further studies are required to support our findings.
Collapse
|
14
|
Abstract
Uveal melanoma (UM) is the major intraocular malignancy in adults, of which the molecular biology is still unknown. Therefore, this study was designed to determine the aqueous concentrations of angiogenic, inflammatory, and chemotactic cytokines in eyes with UM.Aqueous humor samples were collected from 38 patients with UM and 22 patients undergoing cataract surgery. Interleukin 6, 8 (IL-6, IL-8, respectively), interferon-inducible protein-10 (IP-10), placental growth factor1 (PIGF1), regulated on activation, normal T Cell expressed and secreted (RANTES), monocyte chemoattractant protein-1 (MCP-1), nerve growth factor-beta (NGF-β), epidermal growth factor (EGF), basic fibroblast growth factor (bFGF), and vascular endothelia growth factor A (VEGF-A) were assessed by multiplex bead assay.In the study group, significantly higher concentrations of IL-6 (P = .006), IL-8 (P = .018), IP-10 (P = .004), RANTES (P = .008), MCP-1 (P = .02), NGF-β (P = .013), EGF (P < .001), PIGF1 (P = .01), bFGF (P = .016), and VEGF (P = .017) were measured, when compared with the control group.Several angiogenic, inflammatory, and chemotactic cytokines are highly expressed in the aqueous humor of the UM eyes, which provides new insights into the pathophysiology of UM and could be potential targets for treatment.
Collapse
Affiliation(s)
- Yong Cheng
- Department of Ophthalmology, Peking University People's Hospital, Eye Diseases and Optometry Institute, Beijing Key Laboratory of Diagnosis and Therapy of Retinal and Choroid Diseases, College of Optometry, Peking University Health Science Center
| | - Jing Feng
- Department of Ophthalmology, Peking University People's Hospital, Eye Diseases and Optometry Institute, Beijing Key Laboratory of Diagnosis and Therapy of Retinal and Choroid Diseases, College of Optometry, Peking University Health Science Center
- Department of Ophthalmology, Beijing ChaoYang Hospital, Beijing, China
| | - Xuemei Zhu
- Department of Ophthalmology, Peking University People's Hospital, Eye Diseases and Optometry Institute, Beijing Key Laboratory of Diagnosis and Therapy of Retinal and Choroid Diseases, College of Optometry, Peking University Health Science Center
| | - Jianhong Liang
- Department of Ophthalmology, Peking University People's Hospital, Eye Diseases and Optometry Institute, Beijing Key Laboratory of Diagnosis and Therapy of Retinal and Choroid Diseases, College of Optometry, Peking University Health Science Center
| |
Collapse
|
15
|
Preisner F, Leimer U, Sandmann S, Zoernig I, Germann G, Koellensperger E. Impact of Human Adipose Tissue-Derived Stem Cells on Malignant Melanoma Cells in An In Vitro Co-culture Model. Stem Cell Rev Rep 2018; 14:125-140. [PMID: 29064018 DOI: 10.1007/s12015-017-9772-y] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
This study focuses on the interactions of human adipose tissue-derived stem cells (ADSCs) and malignant melanoma cells (MMCs) with regard to future cell-based skin therapies. The aim was to identify potential oncological risks as ADSCs could unintentionally be sited within the proximity of the tumor microenvironment of MMCs. An indirect co-culture model was used to analyze interactions between ADSCs and four different established melanoma cell lines (G-361, SK-Mel-5, MeWo and A2058) as well as two low-passage primary melanoma cell cultures (M1 and M2). Doubling time, migration and invasion, angiogenesis, quantitative real-time PCR of 229 tumor-associated genes and multiplex protein assays of 20 chemokines and growth factors and eight matrix metalloproteinases (MMPs) were evaluated. Co-culture with ADSCs significantly increased migration capacity of G-361, SK-Mel-5, A2058, MeWo and M1 and invasion capacity of G-361, SK-Mel-5 and A2058 melanoma cells. Furthermore, conditioned media from all ADSC-MMC-co-cultures induced tube formation in an angiogenesis assay in vitro. Gene expression analysis of ADSCs and MMCs, especially of low-passage melanoma cell cultures, revealed an increased expression of various genes with tumor-promoting activities, such as CXCL12, PTGS2, IL-6, and HGF upon ADSC-MMC-co-culture. In this context, a significant increase (up to 5,145-fold) in the expression of numerous tumor-associated proteins could be observed, e.g. several pro-angiogenic factors, such as VEGF, IL-8, and CCL2, as well as different matrix metalloproteinases, especially MMP-2. In conclusion, the current report clearly demonstrates that a bi-directional crosstalk between ADSCs and melanoma cells can enhance different malignant properties of melanoma cells in vitro.
Collapse
Affiliation(s)
- Fabian Preisner
- ETHIANUM - Clinic for Plastic, Aesthetic and Reconstructive Surgery, Spine, Orthopedic and Hand Surgery, Preventive Medicine, Voßstraße 6, 69115, Heidelberg, Germany
| | - Uwe Leimer
- ETHIANUM - Clinic for Plastic, Aesthetic and Reconstructive Surgery, Spine, Orthopedic and Hand Surgery, Preventive Medicine, Voßstraße 6, 69115, Heidelberg, Germany
| | - Stefanie Sandmann
- ETHIANUM - Clinic for Plastic, Aesthetic and Reconstructive Surgery, Spine, Orthopedic and Hand Surgery, Preventive Medicine, Voßstraße 6, 69115, Heidelberg, Germany
| | - Inka Zoernig
- Department of Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg, Heidelberg University Hospital, Im Neuenheimer Feld 460, 60120, Heidelberg, Germany
| | - Guenter Germann
- ETHIANUM - Clinic for Plastic, Aesthetic and Reconstructive Surgery, Spine, Orthopedic and Hand Surgery, Preventive Medicine, Voßstraße 6, 69115, Heidelberg, Germany
| | - Eva Koellensperger
- ETHIANUM - Clinic for Plastic, Aesthetic and Reconstructive Surgery, Spine, Orthopedic and Hand Surgery, Preventive Medicine, Voßstraße 6, 69115, Heidelberg, Germany.
| |
Collapse
|
16
|
Jacquelot N, Duong CPM, Belz GT, Zitvogel L. Targeting Chemokines and Chemokine Receptors in Melanoma and Other Cancers. Front Immunol 2018; 9:2480. [PMID: 30420855 PMCID: PMC6215820 DOI: 10.3389/fimmu.2018.02480] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 10/08/2018] [Indexed: 12/19/2022] Open
Abstract
The tumor microenvironment is highly heterogeneous. It is composed of a diverse array of immune cells that are recruited continuously into lesions. They are guided into the tumor through interactions between chemokines and their receptors. A variety of chemokine receptors are expressed on the surface of both tumor and immune cells rendering them sensitive to multiple stimuli that can subsequently influence their migration and function. These features significantly impact tumor fate and are critical in melanoma control and progression. Indeed, particular chemokine receptors expressed on tumor and immune cells are strongly associated with patient prognosis. Thus, potential targeting of chemokine receptors is highly attractive as a means to quench or eliminate unconstrained tumor cell growth.
Collapse
Affiliation(s)
- Nicolas Jacquelot
- Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia.,Department of Medical Biology, University of Melbourne, Melbourne, VIC, Australia
| | - Connie P M Duong
- Gustave Roussy Comprehensive Cancer Institute, Villejuif, France.,INSERM U1015, Villejuif, France
| | - Gabrielle T Belz
- Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia.,Department of Medical Biology, University of Melbourne, Melbourne, VIC, Australia
| | - Laurence Zitvogel
- Gustave Roussy Comprehensive Cancer Institute, Villejuif, France.,INSERM U1015, Villejuif, France.,Faculty of Medicine, Paris Sud/Paris XI University, LeKremlin-Bicêtre, France.,Center of Clinical Investigations in Biotherapies of Cancer (CICBT) 1428, Villejuif, France
| |
Collapse
|
17
|
Phinney BB, Ray AL, Peretti AS, Jerman SJ, Grim C, Pinchuk IV, Beswick EJ. MK2 Regulates Macrophage Chemokine Activity and Recruitment to Promote Colon Tumor Growth. Front Immunol 2018; 9:1857. [PMID: 30298062 PMCID: PMC6160543 DOI: 10.3389/fimmu.2018.01857] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 07/27/2018] [Indexed: 12/21/2022] Open
Abstract
A major risk factor for colon cancer growth and progression is chronic inflammation. We have shown that the MAPK-activated protein kinase 2 (MK2) pathway is critical for colon tumor growth in colitis-associated and spontaneous colon cancer models. This pathway is known to regulate expression of the tumor-promoting cytokines, IL-1, IL-6, and TNF-α. However, little is known about the ability of MK2 to regulate chemokine production. This is the first study to demonstrate this pathway also regulates the chemokines, MCP-1, Mip-1α, and Mip-2α (MMM). We show that these chemokines induce tumor cell growth and invasion in vitro and that MK2 inhibition suppresses tumor cell production of chemokines and reverses the resulting pro-tumorigenic effects. Addition of MMM to colon tumors in vivo significantly enhances tumor growth in control tumors and restores tumor growth in the presence of MK2 inhibition. We also demonstrate that MK2 signaling is critical for chemokine expression and macrophage influx to the colon tumor microenvironment. MK2 signaling in macrophages was essential for inflammatory cytokine/chemokine production, whereas MK2−/− macrophages or MK2 inhibition suppressed cytokine expression. We show that addition of bone marrow-derived macrophages to the tumor microenvironment enhances tumor growth in control tumors and restores tumor growth in tumors treated with MK2 inhibitors, while addition of MK2−/− macrophages had no effect. This is the first study to demonstrate the critical role of the MK2 pathway in chemokine production, macrophage influx, macrophage function, and tumor growth.
Collapse
Affiliation(s)
- Brandon B Phinney
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
| | - Anita L Ray
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
| | - Amanda S Peretti
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
| | - Stephanie J Jerman
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
| | - Carl Grim
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of Texas Medical Branch, Galveston, TX, United States
| | - Irina V Pinchuk
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of Texas Medical Branch, Galveston, TX, United States
| | - Ellen J Beswick
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
| |
Collapse
|
18
|
Hatiboglu MA, Kocyigit A, Guler EM, Akdur K, Nalli A, Karatas E, Tuzgen S. Thymoquinone Induces Apoptosis in B16-F10 Melanoma Cell Through Inhibition of p-STAT3 and Inhibits Tumor Growth in a Murine Intracerebral Melanoma Model. World Neurosurg 2018; 114:e182-e190. [DOI: 10.1016/j.wneu.2018.02.136] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2017] [Accepted: 02/23/2018] [Indexed: 11/16/2022]
|
19
|
Luo S, Zhou C, Zhang J, Chen M, Li H, Zheng S, Quan J. Mutant monocyte chemoattractant protein-1 protein (7ND) inhibits osteoclast differentiation and reduces oral squamous carcinoma cell bone invasion. Oncol Lett 2018; 15:7760-7768. [PMID: 29725470 PMCID: PMC5920317 DOI: 10.3892/ol.2018.8308] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Accepted: 01/23/2018] [Indexed: 12/31/2022] Open
Abstract
The seven-amino acid truncated (7ND) protein is an N-terminal deletion mutant of monocyte chemoattractant protein-1 (MCP-1) and it functions as a dominant-negative inhibitor. 7ND and wild-type MCP-1 form a heterodimer, which binds to MCP-1 receptors and inhibits monocyte chemotaxis. In the present study, the 7ND protein was cloned, expressed and purified. An MTT assay revealed that the proliferation of oral squamous cell carcinoma (OSCC) SCC25 cells was not affected following 3 days of treatment with synthetic 7ND protein. Serial dilutions of the 7ND protein were tested for monocyte migration and osteoclast differentiation, and tartrate-resistant acid phosphatase staining demonstrated that significantly fewer osteoclasts were differentiated from cluster of differentiation 14+ (CD14+) monocytes using magnetic activated cell sorting. Immunofluorescence confirmed these results and significantly less F-actin staining was observed in 7ND-treated osteoclasts. Furthermore, bone invasion was examined by subcutaneously injecting SCC25 cells into the area overlaying the calvariae of nude mice. The results demonstrated that the average tumor volume of SCC25 cells with 7ND protein was similar to the average volume of tumors formed by untreated SCC25 cells. Flow cytometric analysis suggested that the CD14+ subpopulation in the bone marrow of 7ND-treated mice was reduced compared with that of untreated mice. Micro-computed tomography imaging revealed significantly less bone resorption in the calvariae injected with SCC25 cells plus the 7ND protein. Taken together, the results of the present study demonstrated the potential therapeutic value of the 7ND protein. The 7ND MCP-1 variant not only functions in vitro to inhibit osteoclast differentiation, but also reduces the progression of bone invasion by OSCC cells in vivo.
Collapse
Affiliation(s)
- Shuyu Luo
- Department of Oral Pathology, School and Hospital of Stomatology, Tianjin Medical University, Heping, Tianjin 300070, P.R. China
| | - Chuanxiang Zhou
- Department of Oral Pathology, Peking University School and Hospital of Stomatology, Haidian, Beijing 100081, P.R. China
| | - Jianming Zhang
- Department of Stomatology, General Hospital of Tianjin Medical University, Heping, Tianjin 300052, P.R. China
| | - Mengshan Chen
- School of Public Health, Tianjin Medical University, Heping, Tianjin 300070, P.R. China
| | - Hongjie Li
- Department of Oral Pathology, School and Hospital of Stomatology, Tianjin Medical University, Heping, Tianjin 300070, P.R. China
| | - Shanchuan Zheng
- Department of Stomatology, WuQing People's Hospital, Wuqing, Tianjin 301700, P.R. China
| | - Jingjing Quan
- Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-Sen University and Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, Guangdong 510080, P.R. China
| |
Collapse
|
20
|
Surcel M, Constantin C, Caruntu C, Zurac S, Neagu M. Inflammatory Cytokine Pattern Is Sex-Dependent in Mouse Cutaneous Melanoma Experimental Model. J Immunol Res 2017; 2017:9212134. [PMID: 29318162 PMCID: PMC5727748 DOI: 10.1155/2017/9212134] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 10/09/2017] [Accepted: 10/22/2017] [Indexed: 12/16/2022] Open
Abstract
We present the evaluation of inflammatory cytokines in mouse cutaneous melanoma experimental model, as markers of disease evolution. Moreover, to test our experimental model, we have used low doses of dacarbazine (DTIC). C57 BL/6J mouse of both sexes were subjected to experimental cutaneous melanoma and treated with low doses of DTIC. Clinical parameters and serum cytokines were followed during tumor evolution and during DTIC therapy. Cytokine/chemokine pattern was assessed using xMAP technology and the following molecules were quantified: interleukins (IL)-1-beta, IL-6, IL-10, IL-12 (p70), interferon (IFN)-gamma, granulocyte macrophage colony-stimulating factor (GM-CSF), tumor necrosis factor (TNF)-alpha, macrophage inflammatory protein (MIP)-1alpha, monocyte chemoattractant protein (MCP-1), and keratinocyte-derived chemokine (KC). Significant differences were found between normal females and males mice, female mice having a statistically higher serum concentration of IL-1-beta compared to male mice, while males have a significantly higher concentration of MIP-1-alpha. During melanoma evolution in the female group, IL-1-beta, MIP-1-alpha, and KC circulatory levels were found 10-fold increased, while other cytokines doubled their values. In the male mice group, only circulatory KC increased 4 times, while IL-1-beta and TNF-alpha doubled their circulatory values. Various serum cytokines correlated with the disease evolution in cutaneous melanoma mouse model.
Collapse
Affiliation(s)
- Mihaela Surcel
- Immunology Department, “Victor Babes” National Institute of Pathology, 99-101 Spl. Independentei, 050096 Bucharest, Romania
- Faculty of Biology, University of Bucharest, 91-95 Spl. Independentei, 76201 Bucharest, Romania
| | - Carolina Constantin
- Immunology Department, “Victor Babes” National Institute of Pathology, 99-101 Spl. Independentei, 050096 Bucharest, Romania
- Colentina University Hospital, 19-21 Stefan cel Mare Blv., 020125 Bucharest, Romania
| | - Constantin Caruntu
- “Carol Davila” University of Pharmacy and Medicine, 37 Dionisie Lupu Street, 020021 Bucharest, Romania
| | - Sabina Zurac
- Colentina University Hospital, 19-21 Stefan cel Mare Blv., 020125 Bucharest, Romania
- “Carol Davila” University of Pharmacy and Medicine, 37 Dionisie Lupu Street, 020021 Bucharest, Romania
| | - Monica Neagu
- Immunology Department, “Victor Babes” National Institute of Pathology, 99-101 Spl. Independentei, 050096 Bucharest, Romania
- Faculty of Biology, University of Bucharest, 91-95 Spl. Independentei, 76201 Bucharest, Romania
- Colentina University Hospital, 19-21 Stefan cel Mare Blv., 020125 Bucharest, Romania
| |
Collapse
|
21
|
Nakamura N, Matsui T, Ishibashi Y, Sotokawauchi A, Fukami K, Higashimoto Y, Yamagishi SI. RAGE-aptamer Attenuates the Growth and Liver Metastasis of Malignant Melanoma in Nude Mice. Mol Med 2017; 23:295-306. [PMID: 29387865 DOI: 10.2119/molmed.2017.00099] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Accepted: 09/13/2017] [Indexed: 12/12/2022] Open
Abstract
Epidemiological studies have suggested the link between cumulative diabetic exposure and cancer. Interaction of advanced glycation end products (AGEs) with their receptor (RAGE) may contribute to the phenomenon. We examined here the effects of DNA aptamer raised against RAGE (RAGE-aptamer) on growth and liver metastasis of G361 melanoma in nude mice. Malignant melanoma cells were intradermally injected into the upper flank region of nude mice, which received continuous administration of RAGE-aptamer (38.4 pmol/day/g body weight) or vehicle intraperitoneally by an osmotic pump up to 42 days. RAGE-aptamer significantly reduced levels of 8-hydroxy-2'-deoxy-guanosine, AGEs, RAGE, proliferating nuclear antigen, cyclin D1, vascular endothelial growth factor (VEGF), monocyte chemoattractant protein-1 (MCP-1), and CD31 and Mac-3, respective markers of endothelial cells and macrophages in tumors of nude mice and suppressed the proliferation and liver metastasis of malignant melanoma. Furthermore, RAGE-aptamer attenuated the AGE-induced oxidative stress generation, proliferation, and VEGF and MCP-1 gene expression in both G361 melanoma cells and endothelial cells. The present findings suggest that RAGE-aptamer could attenuate melanoma growth and liver metastasis in nude mice by suppressing the tumor angiogenesis and macrophage infiltration via inhibition of the AGE-RAGE system. RAGE-aptamer may be a novel therapeutic tool for the treatment of malignant melanoma.
Collapse
Affiliation(s)
- Nobutaka Nakamura
- Department of Pathophysiology and Therapeutics of Diabetic Vascular Complications, Kurume University School of Medicine, Kurume, Japan
| | - Takanori Matsui
- Department of Pathophysiology and Therapeutics of Diabetic Vascular Complications, Kurume University School of Medicine, Kurume, Japan
| | - Yuji Ishibashi
- Department of Pathophysiology and Therapeutics of Diabetic Vascular Complications, Kurume University School of Medicine, Kurume, Japan
| | - Ami Sotokawauchi
- Department of Pathophysiology and Therapeutics of Diabetic Vascular Complications, Kurume University School of Medicine, Kurume, Japan
| | - Kei Fukami
- Department of Medicine, Kurume University School of Medicine, Kurume, Japan
| | | | - Sho-Ichi Yamagishi
- Department of Pathophysiology and Therapeutics of Diabetic Vascular Complications, Kurume University School of Medicine, Kurume, Japan
| |
Collapse
|
22
|
Poon C, Chowdhuri S, Kuo CH, Fang Y, Alenghat FJ, Hyatt D, Kani K, Gross ME, Chung EJ. Protein Mimetic and Anticancer Properties of Monocyte-Targeting Peptide Amphiphile Micelles. ACS Biomater Sci Eng 2017; 3:3273-3282. [PMID: 29302619 DOI: 10.1021/acsbiomaterials.7b00600] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Monocyte chemoattractant protein-1 (MCP-1) stimulates the migration of monocytes to inflammatory sites, leading to the progression of many diseases. Recently, we described a monocyte-targeting peptide amphiphile micelle (MCP-1 PAM) incorporated with the chemokine receptor CCR2 binding motif of MCP-1, which has a high affinity for monocytes in atherosclerotic plaques. We further report here the biomimetic components of MCP-1 PAMs and the influence of the nanoparticle upon binding to monocytes. We report that MCP-1 PAMs have enhanced secondary structure compared to the MCP-1 peptide. As a result, MCP-1 PAMs displayed improved binding and chemoattractant properties to monocytes, which upregulated the inflammatory signaling pathways responsible for monocyte migration. Interestingly, when MCP-1 PAMs were incubated in the presence of prostate cancer cells in vitro, the particle displayed anticancer efficacy by reducing CCR2 expression. Given that monocytes play an important role in tumor cell migration and invasion, our results demonstrate that PAMs can improve the native biofunctional properties of the peptide and may be used as an effective inhibitor to prevent chemokine-receptor interactions that promote disease progression.
Collapse
Affiliation(s)
- Christopher Poon
- Department of Biomedical Engineering, University of Southern California, 1042 Downey Way, Los Angeles, California 90089, United States
| | - Sampreeti Chowdhuri
- Department of Biomedical Engineering, University of Southern California, 1042 Downey Way, Los Angeles, California 90089, United States
| | - Cheng-Hsiang Kuo
- Section of Pulmonary and Critical Care, Department of Medicine, University of Chicago, 5841 South Maryland Avenue, Chicago, Illinois 60637, United States
| | - Yun Fang
- Section of Pulmonary and Critical Care, Department of Medicine, University of Chicago, 5841 South Maryland Avenue, Chicago, Illinois 60637, United States
| | - Francis J Alenghat
- Section of Cardiology, Department of Medicine, University of Chicago, 5841 South Maryland Avenue, Chicago, Illinois 60637, United States
| | - Danielle Hyatt
- Section of Cardiology, Department of Medicine, University of Chicago, 5841 South Maryland Avenue, Chicago, Illinois 60637, United States
| | - Kian Kani
- Lawrence J. Ellison Institute for Transformative Medicine of USC, Keck School of Medicine, University of Southern California, 2250 Alcazar Street, Los Angeles, California 90089, United States
| | - Mitchell E Gross
- Lawrence J. Ellison Institute for Transformative Medicine of USC, Keck School of Medicine, University of Southern California, 2250 Alcazar Street, Los Angeles, California 90089, United States
| | - Eun Ji Chung
- Department of Biomedical Engineering, University of Southern California, 1042 Downey Way, Los Angeles, California 90089, United States
| |
Collapse
|
23
|
Wang H, Yang L, Wang D, Zhang Q, Zhang L. Pro-tumor activities of macrophages in the progression of melanoma. Hum Vaccin Immunother 2017; 13:1556-1562. [PMID: 28441072 PMCID: PMC5512774 DOI: 10.1080/21645515.2017.1312043] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Macrophages are located in essentially all tissues due to their “janitor” function. Macrophages can exert either anti- or pro-tumor activities depending upon the specific tumor microenvironment they inhabit. Substantial evidence indicates that macrophages, owing to their plasticity, can be reeducated to adopt a protumoral phenotype within a tumor microenvironment through the help of growth factors in the microenvironment and intercellular interactions. As the lethality of malignant melanoma is due to its aggressive capacity for metastasis and resistance to therapy, considerable effort has gone toward treatment of metastatic melanoma. In the present review, we focus on the pro-tumor activities of macrophages in melanoma. Based upon the information presented in this review it is anticipated that new therapies will soon be developed that target pro-tumor activities of macrophages for use in the treatment of melanoma.
Collapse
Affiliation(s)
- Huafeng Wang
- a Modern College of Arts and Science, or School of Life Science, Shanxi Normal University , Linfen , China
| | - Luhong Yang
- a Modern College of Arts and Science, or School of Life Science, Shanxi Normal University , Linfen , China
| | - Dong Wang
- b Central Blood Station of Tianjin , Tianjin , China
| | - Qi Zhang
- c Nankai Hospital , Tianjin , China
| | - Lijuan Zhang
- d Research Center of Basic Medical Sciences , Tianjin Medical University , Tianjin , China
| |
Collapse
|
24
|
Wang H, Zhang L, Yang L, Liu C, Zhang Q, Zhang L. Targeting macrophage anti-tumor activity to suppress melanoma progression. Oncotarget 2017; 8:18486-18496. [PMID: 28060744 PMCID: PMC5392344 DOI: 10.18632/oncotarget.14474] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 12/27/2016] [Indexed: 01/09/2023] Open
Abstract
By phagocytosing cancer cells and their cellular debris, macrophages play a critical role in nonspecific defense (innate immunity) and, as antigen presenters, they help initiate specific defense mechanisms (adaptive immunity). Malignant melanoma is a lethal disease due to its aggressive capacity for metastasis and resistance to therapy. For decades, considerable effort has gone into development of an effective immunotherapy for treatment of metastatic melanoma. In this review, we focus on the anti-tumor activities of macrophages in melanoma and their potential as therapeutic targets in melanoma. Although macrophages can be re-educated through intercellular signaling to promote tumor survival owing to their plasticity, we expect that targeting the anti-tumor activity of macrophages remains a promising strategy for melanoma inhibition. The combination of tumoricidal macrophage activation and other treatments such as surgery, chemotherapy, and radiotherapy, may provide an effective and comprehensive anti-melanoma strategy.
Collapse
Affiliation(s)
- Huafeng Wang
- Modern College of Arts and Science, or School of Life Science, Shanxi Normal University, Linfen, China
| | - Lijuan Zhang
- Modern College of Arts and Science, or School of Life Science, Shanxi Normal University, Linfen, China
- Research Center of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Luhong Yang
- Modern College of Arts and Science, or School of Life Science, Shanxi Normal University, Linfen, China
| | - Chengfang Liu
- Department of Human Anatomy, Shanxi Medical University, Shanxi Sheng, China
| | | | - Linjing Zhang
- Modern College of Arts and Science, or School of Life Science, Shanxi Normal University, Linfen, China
| |
Collapse
|
25
|
Abstract
The human body combats infection and promotes wound healing through the remarkable process of inflammation. Inflammation is characterized by the recruitment of stromal cell activity including recruitment of immune cells and induction of angiogenesis. These cellular processes are regulated by a class of soluble molecules called cytokines. Based on function, cell target, and structure, cytokines are subdivided into several classes including: interleukins, chemokines, and lymphokines. While cytokines regulate normal physiological processes, chronic deregulation of cytokine expression and activity contributes to cancer in many ways. Gene polymorphisms of all types of cytokines are associated with risk of disease development. Deregulation RNA and protein expression of interleukins, chemokines, and lymphokines have been detected in many solid tumors and hematopoetic malignancies, correlating with poor patient prognosis. The current body of literature suggests that in some tumor types, interleukins and chemokines work against the human body by signaling to cancer cells and remodeling the local microenvironment to support the growth, survival, and invasion of primary tumors and enhance metastatic colonization. Some lymphokines are downregulated to suppress tumor progression by enhancing cytotoxic T cell activity and inhibiting tumor cell survival. In this review, we will describe the structure/function of several cytokine families and review our current understanding on the roles and mechanisms of cytokines in tumor progression. In addition, we will also discuss strategies for exploiting the expression and activity of cytokines in therapeutic intervention.
Collapse
Affiliation(s)
- M Yao
- University of Kansas Medical Center, Kansas City, KS, United States
| | - G Brummer
- University of Kansas Medical Center, Kansas City, KS, United States
| | - D Acevedo
- University of Kansas Medical Center, Kansas City, KS, United States
| | - N Cheng
- University of Kansas Medical Center, Kansas City, KS, United States.
| |
Collapse
|
26
|
The Effect of MCP-1/CCR2 on the Proliferation and Senescence of Epidermal Constituent Cells in Solar Lentigo. Int J Mol Sci 2016; 17:ijms17060948. [PMID: 27314341 PMCID: PMC4926481 DOI: 10.3390/ijms17060948] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Revised: 06/07/2016] [Accepted: 06/08/2016] [Indexed: 12/15/2022] Open
Abstract
Solar lentigo (SL) is a representative photoaging skin disorder. Alteration of the main epidermal constituent cells-keratinocytes and melanocytes-in relation to the photoaged dermal environment or chemokine/cytokine network is suggested as its pathogenesis. Among these, we focused on monocyte chemoattractant protein-1 (MCP-1), as it is known to be associated with tissue aging. For the first time, we report that the MCP-1 receptor, CCR2, is expressed in normal human melanocytes. In SL tissue, there was an increase of CCR2+Melan A+ melanocytes with positivity to Rb protein compared to peri-lesional normal skin. MCP-1 induced the proliferation of normal human melanocytes without a significant change in the melanin content. MCP-1 treatment in normal human keratinocytes showed an increase in senescence-associated β-galactosidase staining and p53 and p21 protein expressions. In summary, MCP-1 may participate in the development of SL by affecting epidermal constituent cells, for example, by inducing melanocyte proliferation and keratinocyte senescence.
Collapse
|
27
|
Kretzer IF, Maria DA, Guido MC, Contente TC, Maranhão RC. Simvastatin increases the antineoplastic actions of paclitaxel carried in lipid nanoemulsions in melanoma-bearing mice. Int J Nanomedicine 2016; 11:885-904. [PMID: 27022257 PMCID: PMC4788363 DOI: 10.2147/ijn.s88546] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
PURPOSE Lipid nanoemulsions (LDEs) that bind to low-density lipoprotein (LDL) receptors used as carriers of paclitaxel (PTX) can decrease toxicity and increase PTX antitumoral action. The administration of simvastatin (Simva), which lowers LDL-cholesterol, was tested as an adjuvant to commercial PTX and to PTX associated with LDE (LDE-PTX). MATERIALS AND METHODS B16F10 melanoma-bearing mice were treated with saline solution or LDE (controls), Simva, PTX, PTX and Simva, LDE-PTX, and LDE-PTX and Simva: PTX dose 17.5 μmol/kg (three intraperitoneal injections, 3 alternate days): Simva 50 mg/kg/day by gavage, 9 consecutive days. RESULTS Compared with saline controls, 95% tumor-growth inhibition was achieved by LDE-PTX and Simva, 61% by LDE-PTX, 44% by PTX and Simva, and 43% by PTX. Simva alone had no effect. Metastasis developed in only 37% of the LDE-PTX and Simva, 60% in LDE-PTX, and 90% in PTX and Simva groups. Survival rates were higher in LDE-PTX and Simva and in LDE-PTX groups. The LDE-PTX and Simva group presented tumors with reduced cellular density and increased collagen fibers I and III. Tumors from all groups showed reduction in immunohistochemical expression of ICAM, MCP-1, and MMP-9; LDE-PTX and Simva presented the lowest MMP-9 expression. Expression of p21 was increased in the Simva, LDE-PTX, and LDE-PTX and Simva groups. In the Simva and LDE-PTX and Simva groups, expression of cyclin D1, a proliferation and survival promoter of tumor cells, was decreased. Therapy with LDE-PTX and Simva showed negligible toxicity compared with PTX and Simva, which resulted in weight loss and myelosuppression. CONCLUSION Simva increased the antitumor activity of PTX carried in LDE but not of PTX commercial presentation, possibly because statins increase the expression of LDL receptors that internalize LDE-PTX.
Collapse
Affiliation(s)
- Iara F Kretzer
- Laboratory of Metabolism and Lipids, Heart Institute of the Medical School Hospital, University of São Paulo, São Paulo, Brazil; Department of Clinical Chemistry, Faculty of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Durvanei A Maria
- Biochemistry and Biophysics Laboratories, Butantan Institute, São Paulo, Brazil
| | - Maria C Guido
- Laboratory of Metabolism and Lipids, Heart Institute of the Medical School Hospital, University of São Paulo, São Paulo, Brazil
| | - Thaís C Contente
- Laboratory of Metabolism and Lipids, Heart Institute of the Medical School Hospital, University of São Paulo, São Paulo, Brazil
| | - Raul C Maranhão
- Laboratory of Metabolism and Lipids, Heart Institute of the Medical School Hospital, University of São Paulo, São Paulo, Brazil; Department of Clinical Chemistry, Faculty of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
28
|
Tang J, Chen Z, Sun B, Dong J, Liu J, Zhou H, Wang L, Bai R, Miao Q, Zhao Y, Chen C, Liu Y. Polyhydroxylated fullerenols regulate macrophage for cancer adoptive immunotherapy and greatly inhibit the tumor metastasis. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2015; 12:945-954. [PMID: 26733256 DOI: 10.1016/j.nano.2015.11.021] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Revised: 11/19/2015] [Accepted: 11/25/2015] [Indexed: 12/26/2022]
Abstract
UNLABELLED Adoptive immunotherapy is a highly effective approach for cancer treatment. Several potential adoptive immunotherapies have high (though reversible) toxicities with disappointing results. Polyhydroxylated fullerenols have been demonstrated as promising antitumor drugs with low toxicities. In this study, we investigate whether polyhydroxylated fullerenols (C60(OH)22 and Gd@C82(OH)22) contribute to cancer immunotherapy by regulating macrophages. Our results show that fullerenols treatment enhances mitochondrial metabolism, phagocytosis and cytokine secretion. Moreover, activated macrophages inhibit the growth of several cancer cell types. It is likely that this inhibition is dependent on an NF-κB-mediated release of multiple cytokines. Using a lung metastasis model, we also show that autologous macrophages greatly suppress cancer cell metastasis to lung when they are activated by C60(OH)22 and Gd@C82(OH)22. More importantly, Gd@C82(OH)22 are shown to have stronger ability than C60(OH)22 to improve the macrophage function, which shed light on the rational design for nanomedicine and clinical application. FROM THE CLINICAL EDITOR The interest in the use of immunotherapy in cancer has rekindled recently. However, many approaches have shown disappointing results. In this study, the authors investigated the effects of polyhydroxylated fullerenol nanoparticles on regulating macrophages for immunotherapy. These positive findings may point a novel way to cancer treatment.
Collapse
Affiliation(s)
- Jinglong Tang
- Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology of China and Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing, China; Center for Nanochemistry, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Zhiyun Chen
- Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology of China and Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing, China
| | - Baoyun Sun
- Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology of China and Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing, China
| | - Jinquan Dong
- Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology of China and Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing, China
| | - Jing Liu
- Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology of China and Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing, China
| | - Huige Zhou
- Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology of China and Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing, China
| | - Liming Wang
- Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology of China and Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing, China
| | - Ru Bai
- Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology of China and Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing, China
| | - Qing Miao
- Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology of China and Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing, China; Department of Surgery, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Yuliang Zhao
- Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology of China and Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing, China
| | - Chunying Chen
- Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology of China and Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing, China
| | - Ying Liu
- Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology of China and Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing, China.
| |
Collapse
|
29
|
Proudfoot AEI, Bonvin P, Power CA. Targeting chemokines: Pathogens can, why can't we? Cytokine 2015; 74:259-67. [PMID: 25753743 DOI: 10.1016/j.cyto.2015.02.011] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Accepted: 02/11/2015] [Indexed: 12/19/2022]
Abstract
Chemoattractant cytokines, or chemokines, are the largest sub-family of cytokines. About 50 distinct chemokines have been identified in humans. Their principal role is to stimulate the directional migration of leukocytes, which they achieve through activation of their receptors, following immobilization on cell surface glycosaminoglycans (GAGs). Chemokine receptors belong to the G protein-coupled 7-transmembrane receptor family, and hence their identification brought great promise to the pharmaceutical industry, since this receptor class is the target for a large percentage of marketed drugs. Unfortunately, the development of potent and efficacious inhibitors of chemokine receptors has not lived up to the early expectations. Several approaches to targeting this system will be described here, which have been instrumental in establishing paradigms in chemokine biology. Whilst drug discovery programs have not yet elucidated how to make successful drugs targeting the chemokine system, it is now known that certain parasites have evolved anti-chemokine strategies in order to remain undetected by their hosts. What can we learn from them?
Collapse
Affiliation(s)
- Amanda E I Proudfoot
- Geneva Research Centre, Merck Serono S.A., 9 chemin des Mines, 1202 Genève and NovImmune S.A., 14 chemin des Aulx, 1228 Plan-les-Ouates, Geneva, Switzerland.
| | - Pauline Bonvin
- Geneva Research Centre, Merck Serono S.A., 9 chemin des Mines, 1202 Genève and NovImmune S.A., 14 chemin des Aulx, 1228 Plan-les-Ouates, Geneva, Switzerland.
| | - Christine A Power
- Geneva Research Centre, Merck Serono S.A., 9 chemin des Mines, 1202 Genève, Switzerland.
| |
Collapse
|
30
|
Shen Y, Zhang FQ, Wei X. Truncated monocyte chemoattractant protein-1 can alleviate cardiac injury in mice with viral myocarditis via infiltration of mononuclear cells. Microbiol Immunol 2014; 58:195-201. [PMID: 24401088 DOI: 10.1111/1348-0421.12130] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2013] [Revised: 12/21/2013] [Accepted: 01/04/2014] [Indexed: 11/29/2022]
Abstract
BALB/c mice inoculated intraperitoneally with coxsackievirus group B type 3 (CVB3) were allocated to five groups; namely, a viral myocarditis group infected with CVB3 alone (control group), an antibody intervention group that received intracardiac anti-MCP-1, an antibody intervention control group that received goat IgG, a tMCP-1 intervention group that received plasmid pVMt expressing tMCP-1, and a tMCP-1 intervention control group that received plasmid pVAX1. There was also a normal control group. The ratio of murine heart weight to body weight, pathological score of myocardial tissue, serum creatine kinase-MB titers and CVB3 loading of myocardial tissue were assessed. The cardiac lesions in mice that received 20, 40 or 60 µg pVMt (P < 0.05) were less severe than those in control mice with untreated viral myocarditis. In addition, fewer mononuclear cells had infiltrated the myocardium of mice who received 40 or 60 µg pVMt intramyocardially (P < 0.01), whereas there was no difference in mononuclear cell infiltration between mice with viral myocarditis and those that received 20 µg pVMt (P > 0.05). There was also no difference between mice that received anti-MCP-1 antibody and those that received 40 µg pVMt in ratio of HW/BW, serum CK-MB titers and pathological score (P > 0.05). This study showed that tMCP-1 can alleviate cardiac lesions and cardiac injury in mice with viral myocarditis via infiltration of mononuclear cells. Thus, tMCP-1 may be an alternative to anti-MCP-1 antibody treatment of viral myocarditis. Further research is required.
Collapse
Affiliation(s)
- Yan Shen
- First Affiliated Hospital, Zhengzhou University, 1 Jian She Road, Zhengzhou, 450052, China
| | | | | |
Collapse
|
31
|
Quan J, Morrison NA, Johnson NW, Gao J. MCP-1 as a Potential Target to Inhibit the Bone Invasion by Oral Squamous Cell Carcinoma. J Cell Biochem 2014; 115:1787-98. [DOI: 10.1002/jcb.24849] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2013] [Accepted: 05/12/2014] [Indexed: 12/18/2022]
Affiliation(s)
- Jingjing Quan
- Guanghua School of Stomatology; Hospital of Stomatology; Sun Yat-sen University & Guangdong Provincial Key Laboratory of Stomatology; Guangzhou Guangdong 510055 China
- School of Medical Science; Griffith University; QLD 4222 Australia
| | | | | | - Jin Gao
- School of Dentistry and Oral Health; Griffith University; QLD 4222 Australia
| |
Collapse
|
32
|
Zhou J, Xiang Y, Yoshimura T, Chen K, Gong W, Huang J, Zhou Y, Yao X, Bian X, Wang JM. The role of chemoattractant receptors in shaping the tumor microenvironment. BIOMED RESEARCH INTERNATIONAL 2014; 2014:751392. [PMID: 25110692 PMCID: PMC4119707 DOI: 10.1155/2014/751392] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Accepted: 06/17/2014] [Indexed: 12/13/2022]
Abstract
Chemoattractant receptors are a family of seven transmembrane G protein coupled receptors (GPCRs) initially found to mediate the chemotaxis and activation of immune cells. During the past decades, the functions of these GPCRs have been discovered to not only regulate leukocyte trafficking and promote immune responses, but also play important roles in homeostasis, development, angiogenesis, and tumor progression. Accumulating evidence indicates that chemoattractant GPCRs and their ligands promote the progression of malignant tumors based on their capacity to orchestrate the infiltration of the tumor microenvironment by immune cells, endothelial cells, fibroblasts, and mesenchymal cells. This facilitates the interaction of tumor cells with host cells, tumor cells with tumor cells, and host cells with host cells to provide a basis for the expansion of established tumors and development of distant metastasis. In addition, many malignant tumors of the nonhematopoietic origin express multiple chemoattractant GPCRs that increase the invasiveness and metastasis of tumor cells. Therefore, GPCRs and their ligands constitute targets for the development of novel antitumor therapeutics.
Collapse
Affiliation(s)
- Jiamin Zhou
- Laboratory of Molecular Immunoregulation, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA
- Endoscopic Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yi Xiang
- Laboratory of Molecular Immunoregulation, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA
- Department of Pulmonary Medicine, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, China
| | - Teizo Yoshimura
- Laboratory of Molecular Immunoregulation, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA
| | - Keqiang Chen
- Laboratory of Molecular Immunoregulation, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA
| | - Wanghua Gong
- Basic Research Program, Leidos Biomedical Research, Inc., Frederick, MD 21702, USA
| | - Jian Huang
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Ye Zhou
- Department of Gastric Cancer and Soft Tissue Surgery, Fudan University Cancer Center, Shanghai 200032, China
| | - Xiaohong Yao
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Xiuwu Bian
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Ji Ming Wang
- Laboratory of Molecular Immunoregulation, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA
| |
Collapse
|
33
|
Ji WT, Chen HR, Lin CH, Lee JW, Lee CC. Monocyte chemotactic protein 1 (MCP-1) modulates pro-survival signaling to promote progression of head and neck squamous cell carcinoma. PLoS One 2014; 9:e88952. [PMID: 24586454 PMCID: PMC3929549 DOI: 10.1371/journal.pone.0088952] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2013] [Accepted: 01/16/2014] [Indexed: 12/04/2022] Open
Abstract
Background Monocyte chemotactic protein-1 (MCP-1) recruits monocytes and macrophages to inflammation sites, and inflammatory infiltration correlates with the progression of head and neck squamous cell carcinoma (HNSCC). This study aims to determine whether MCP-1 expression is related to HNSCC malignancy and patient survival. We also investigated the relationship between MCP-1 expression and the phosphorylation state of the pro-survival pathway factors Akt, ERK, and STAT3. Methods Expression of MCP-1 and related proteins in HNSCC cell lines was investigated using western blotting. HNSCC patients (34) without distant metastasis at diagnosis were recruited for tissue specimen evaluation of MCP-1 expression and clinical outcomes. The relationship between MCP-1 expression and survival was evaluated using the Cox proportional hazard model with stepwise selection. Results High-grade HNSCC cell lines were found to have higher levels of active Akt, ERK, and/or STAT3 than did lower grade cell lines under serum-free condition. OCSL, the most malignant cell line, had the highest level of endogenous MCP-1. Administration of exogenous recombinant MCP-1 increased phosphorylation of Akt, ERK, and STAT3 in a dose- and time-dependent manner and increased cellular resistance to serum starvation. Inhibition of Akt, ERK, or STAT3 reduced cell growth and caused cell death. Long-term survival of HNSCC patients was negatively associated with the histological intensity of MCP-1, implicating MCP-1 as a potential prognostic marker for HNSCC. Conclusions These results suggest that overexpressed MCP-1 in cancer cells may promote HNSCC progression through upregulating pro-survival signaling pathways. High cellular MCP-1 expression is related to poor overall survival rate in HNSCC patients.
Collapse
Affiliation(s)
- Wen-Tsai Ji
- Department of Life Science, Institute of Molecular Biology and Institute of Biomedical Science, College of Science, National Chung Cheng University, Min-Hsiung, Chia-Yi, Taiwan
| | - Hau-Ren Chen
- Department of Life Science, Institute of Molecular Biology and Institute of Biomedical Science, College of Science, National Chung Cheng University, Min-Hsiung, Chia-Yi, Taiwan
| | - Chun-Hsuan Lin
- Department of Medical Research, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Chia-Yi, Taiwan
| | - Jeng-Woei Lee
- Department of Life Sciences, Tzu-Chi University, Hualien, Taiwan
- Institute of Medical Sciences, Tzu-Chi University, Hualien, Taiwan
- * E-mail: (C-CL); (J-WL)
| | - Ching-Chih Lee
- Department of Otolaryngology, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Chia-Yi, Taiwan
- School of Medicine, Tzu Chi University, Hualian, Taiwan
- * E-mail: (C-CL); (J-WL)
| |
Collapse
|
34
|
Neagu M, Constantin C, Martin D, Albulescu L, Iacob N, Ighigeanu D. Whole body microwave irradiation for improved dacarbazine therapeutical action in cutaneous melanoma mouse model. Radiol Res Pract 2013; 2013:414816. [PMID: 24377047 PMCID: PMC3860147 DOI: 10.1155/2013/414816] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Revised: 09/23/2013] [Accepted: 09/30/2013] [Indexed: 12/13/2022] Open
Abstract
A cutaneous melanoma mouse model was used to test the efficacy of a new therapeutical approach that uses low doses of cytostatics in conjunction with mild whole body microwave exposure of 2.45 GHz in order to enhance cytostatics antitumoral effect. Materials and Methods. A microwave exposure system for C57BL/6 mouse whole body microwave irradiation was designed; groups of 40 mice (males and females) bearing experimental tumours were subjected to a combined therapy comprising low doses of dacarbazine in combination with mild whole body irradiation. Clinical parameters and serum cytokine testing using xMAP technology were performed. Results. The group that was subjected to combined therapy, microwave and cytostatic, had the best clinical evolution in terms of overall survival, tumour volume, and metastatic potential. At day 14 the untreated group had 100% mortality, while in the combined therapy group 40% of mice were surviving. Quantifying serum IL-1 β , IL-6, IL-10, IL-12 (p70), IFN- γ , GM-CSF, TNF- α , MIP-1 α , MCP-1, and KC during tumorigenesis and therapy found that the combined experimental therapy decreases all the inflammatory cytokines, except chemokine MCP-1 that was found increased, suggesting an increase of the anti-tumoral immune response triggered by the combined therapy. The overall metastatic process is decreased in the combined therapy group.
Collapse
Affiliation(s)
- Monica Neagu
- Immunology Department, Immunobiology Laboratory, “Victor Babes” National Institute of Pathology, 99-101 Splaiul Independentei, sector 5, Bucharest 050096, Romania
| | - Carolina Constantin
- Immunology Department, Immunobiology Laboratory, “Victor Babes” National Institute of Pathology, 99-101 Splaiul Independentei, sector 5, Bucharest 050096, Romania
| | - Diana Martin
- National Institute for Laser, Plasma and Radiation Physics, 409 Atomistilor Street, Magurele 077125, Romania
| | - Lucian Albulescu
- Department of Infectious Diseases and Immunology, Virology Division, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 1, 3584 CL Utrecht, The Netherlands
| | - Nicusor Iacob
- National Institute for Laser, Plasma and Radiation Physics, 409 Atomistilor Street, Magurele 077125, Romania
| | - Daniel Ighigeanu
- National Institute for Laser, Plasma and Radiation Physics, 409 Atomistilor Street, Magurele 077125, Romania
| |
Collapse
|
35
|
Heterogeneity of functional properties of Clone 66 murine breast cancer cells expressing various stem cell phenotypes. PLoS One 2013; 8:e78725. [PMID: 24265713 PMCID: PMC3827106 DOI: 10.1371/journal.pone.0078725] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2013] [Accepted: 09/19/2013] [Indexed: 01/06/2023] Open
Abstract
INTRODUCTION Breast cancer grows, metastasizes and relapses from rare, therapy resistant cells with a stem cell phenotype (cancer stem cells/CSCs). However, there is a lack of studies comparing the functions of CSCs isolated using different phenotypes in order to determine if CSCs are homogeneous or heterogeneous. METHODS Cells with various stem cell phenotypes were isolated by sorting from Clone 66 murine breast cancer cells that grow orthotopically in immune intact syngeneic mice. These populations were compared by in vitro functional assays for proliferation, growth, sphere and colony formation; and in vivo limiting dilution analysis of tumorigenesis. RESULTS The proportion of cells expressing CD44(high)CD24(low/neg), side population (SP) cells, ALDH1(+), CD49f(high), CD133(high), and CD34(high) differed, suggesting heterogeneity. Differences in frequency and size of tumor spheres from these populations were observed. Higher rates of proliferation of non-SP, ALDH1(+), CD34(low), and CD49f(high) suggested properties of transit amplifying cells. Colony formation was higher from ALDH1(-) and non-SP cells than ALDH1(+) and SP cells suggesting a progenitor phenotype. The frequency of clonal colonies that grew in agar varied and was differentially altered by the presence of Matrigel™. In vivo, fewer cells with a stem cell phenotype were needed for tumor formation than "non-stem" cells. Fewer SP cells were needed to form tumors than ALDH1(+) cells suggesting further heterogeneities of cells with stem phenotypes. Different levels of cytokines/chemokines were produced by Clone 66 with RANTES being the highest. Whether the heterogeneity reflects soluble factor production remains to be determined. CONCLUSIONS These data demonstrate that Clone 66 murine breast cancer cells that express stem cell phenotypes are heterogeneous and exhibit different functional properties, and this may also be the case for human breast cancer stem cells.
Collapse
|
36
|
Kehlen A, Haegele M, Menge K, Gans K, Immel UD, Hoang-Vu C, Klonisch T, Demuth HU. Role of glutaminyl cyclases in thyroid carcinomas. Endocr Relat Cancer 2013. [PMID: 23183267 DOI: 10.1530/erc-12-0053] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
CCL2 is a chemokine known to recruit monocytes/macrophages to sites of inflammation. CCL2 is also associated with tumor progression in several cancer types. Recently, we showed that the N-terminus of CCL2 is modified to a pyroglutamate (pE)-residue by both glutaminyl cyclases (QC (QPCT)) and its isoenzyme (isoQC (QPCTL)). The pE-residue increases stability against N-terminal degradation by aminopeptidases. Here, we report an upregulation of QPCT expression in tissues of patients with thyroid carcinomas compared with goiter tissues, whereas QPCTL was not regulated. In thyroid carcinoma cell lines, QPCT gene expression correlates with the mRNA levels of its substrate CCL2. Both QPCT and CCL2 are regulated in a NF-κB-dependent pathway shown by stimulation with TNFa and IL1b as well as by inhibition with the IKK2 inhibitor and RNAi of p50. In the culture supernatant of thyroid carcinoma cells, equal amounts of pECCL2 and total CCL2 were detected by two ELISAs discriminating between total CCL2 and pECCL2, concluding that all CCL2 is secreted as pECCL2. Activation of the CCL2/CCR2 pathway by recombinant CCL2 increased tumor cell migration of FTC238 cells in scratch assays as well as thyroid carcinoma cell-derived CCL2-induced migration of monocytic THP1 cells. Suppression of CCL2 signaling by CCR2 antagonist, IKK2 inhibitor, and QPCT RNAi reduced FTC238 cell growth measured by WST8 proliferation assays. Our results reveal new evidence for a novel role of QC in thyroid carcinomas and provide an intriguing rationale for the use of QC inhibitors as a means of blocking pECCL2 formation and preventing thyroid cancer metastasis.
Collapse
Affiliation(s)
- Astrid Kehlen
- Probiodrug AG, Weinbergweg 22, Halle, Saale, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Takahashi R, Hirata Y, Sakitani K, Nakata W, Kinoshita H, Hayakawa Y, Nakagawa H, Sakamoto K, Hikiba Y, Ijichi H, Moses HL, Maeda S, Koike K. Therapeutic effect of c-Jun N-terminal kinase inhibition on pancreatic cancer. Cancer Sci 2013; 104:337-44. [PMID: 23237571 DOI: 10.1111/cas.12080] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2012] [Revised: 12/04/2012] [Accepted: 12/07/2012] [Indexed: 12/25/2022] Open
Abstract
c-Jun N-terminal kinase (JNK) is a member of the mitogen-activated protein kinase (MAPK) family, and it is reportedly involved in the development of several cancers. However, the role of JNK in pancreatic cancer has not been elucidated. We assessed t he involvement of JNK in the development of pancreatic cancer and investigated the therapeutic effect of JNK inhibitors on this deadly cancer. Small interfering RNAs against JNK or the JNK inhibitor SP600125 were used to examine the role of JNK in cellular proliferation and the cell cycles of pancreatic cancer cell lines. Ptf1a(cre/+) ;LSL-Kras(G12D/+) ;Tgfbr2(flox/flox) mice were treated with the JNK inhibitor to examine pancreatic histology and survival. The effect of JNK inhibition on tumor angiogenesis was also assessed using cell lines and murine pancreatic cancer specimens. JNK was frequently activated in human and murine pancreatic cancer in vitro and in vivo. Growth of human pancreatic cancer cell lines was suppressed by JNK inhibition through G1 arrest in the cell cycle with decreased cyclin D1 expression. In addition, oncogenic K-ras expression led to activation of JNK in pancreatic cancer cell lines. Treatment of Ptf1a(cre/+) ;LSL-Kras(G12D/+) ;Tgfbr2(flox/flox) mice with the JNK inhibitor decreased growth of murine pancreatic cancer and prolonged survival of the mice significantly. Angiogenesis was also decreased by JNK inhibition in vitro and in vivo. In conclusion, activation of JNK promotes development of pancreatic cancer, and JNK may be a potential therapeutic target for pancreatic cancer.
Collapse
Affiliation(s)
- Ryota Takahashi
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Lee CS, Jun IH, Kim TI, Byeon SH, Koh HJ, Lee SC. Expression of 12 cytokines in aqueous humour of uveal melanoma before and after combined Ruthenium-106 brachytherapy and transpupillary thermotherapy. Acta Ophthalmol 2012; 90:e314-20. [PMID: 22429778 DOI: 10.1111/j.1755-3768.2012.02392.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
PURPOSE To determine the aqueous humour levels of 12 cytokines in eyes with uveal melanoma and whether their expression changes after combined Ru-106 brachytherapy and transpupillary thermotherapy (TTT). METHODS Aqueous humour samples were collected from 20 patients with previously untreated uveal melanoma undergoing combined Ru-106 brachytherapy and TTT, both at the time of plaque placement and removal. Using multiplex biochip array technology, 12 different cytokines were measured, including interleukin (IL)-1α, IL-1β, IL-2, IL-4, IL-6, IL-8, IL-10, vascular endothelial growth factor (VEGF), tumour necrosis factor (TNF)-α, interferon (IFN)-γ, epidermal growth factor (EGF) and monocyte chemoattractant protein (MCP)-1. Aqueous humour from 20 patients undergoing cataract surgery was used as control. RESULTS IL-6, IL-8, IFN-γ and MCP-1 were highly expressed in uveal melanoma, whereas IL-2, IL-10 and TNF- α were low in expression. There was a positive correlation between tumour height and IL-8 level (p = 0.020). Vascular endothelial growth factor tends to be highly expressed in melanoma-containing eyes (p = 0.056). Levels of IL-6, IL-8 and IL-1β increased after the mean 117 ± 38 hrs of brachytherapy and adjunctive TTT with a tumour apex dose of 61 ± 28 Gy and a scleral contact dose of 786 ± 226 Gy. Increase in levels of IL-6 (p = 0.003) and IL-8 (p = 0.046) positively correlated with scleral contact dose. CONCLUSIONS Cytokines such as IL-6, IL-8, IFN-γ and MCP-1 may be implicated in the progression of uveal melanoma. Ocular irradiation from a Ru-106 plaque promoted an increase in the levels of IL-6, IL-8 and IL-1β, modulation of which could be useful in managing radiation-related side effects.
Collapse
Affiliation(s)
- Christopher Seungkyu Lee
- Department of Ophthalmology, The Institute of Vision Research, Yonsei University College of Medicine, Seoul, Korea
| | | | | | | | | | | |
Collapse
|
39
|
Role of bone marrow-derived cells in angiogenesis: focus on macrophages and pericytes. CANCER MICROENVIRONMENT 2012; 5:225-36. [PMID: 22528877 DOI: 10.1007/s12307-012-0106-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2012] [Accepted: 04/03/2012] [Indexed: 02/06/2023]
Abstract
Tumor growth relies on the formation of new blood vessels to receive an adequate supply of oxygen and nutrient. This process is facilitated by both the remodeling of the pre-existing vasculatures and the recruitment of the progenitor/stem cells originated from bone marrow-derived cells (BMDCs). Evidences from both animal studies and human trials have reported that these tumor-associated BMDCs differentiate into a series of stromal cells including macrophages and pericytes, and regulate tumor angiogenesis in various aspects. Macrophages constitute a large portion of the BMDCs infiltrated in the tumor microenvironment, and have been shown to disrupt the balance of pro- and anti-angiogenic signalings by the secretion of various cytokines. Pericytes, mainly derived from the subpopulation of PDGFRβ(+) BMDCs, can provide both pro-survival signaling and mechanical support to maintain the newly formed endothelium via the direct interactions with endothelial cells. In the current review, we summarize the recruitment mechanisms of BMDC-derived macrophages and pericytes within tumor microenvironment, and also review the contribution of these cells to the different aspects of angiogenesis, with particular emphasis on their therapeutic implications as potential targets for anti-tumor strategies.
Collapse
|
40
|
Severin IC, Souza ALS, Davis JH, Musolino N, Mack M, Power CA, Proudfoot AEI. Properties of 7ND-CCL2 are modulated upon fusion to Fc. Protein Eng Des Sel 2012; 25:213-22. [PMID: 22388887 DOI: 10.1093/protein/gzs008] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
7ND, a truncated version of the chemokine MCP-1/CCL2 lacking amino acids 2-8, is a potent antagonist of CCR2. In contrast to CCL2, 7ND is an obligate monomer. Similar to other chemokines, the in vivo half-life of 7ND is very short and its use as an antagonist in disease models is thus limited. We therefore constructed a 7ND-Fc fusion protein to extend the half-life of 7ND and overcome its limitations as a potential therapeutic antagonist. When we tested the properties of the fusion molecule in vitro, we found to our surprise that 7ND-Fc, in contrast to 7ND, produced a distinct, albeit small, chemotactic response in THP-1 cells, and a robust chemotactic response in L1.2 cells stably transfected with CCR2. To test whether this unexpected observation might be due to the bivalency of 7ND-Fc stemming from the dimeric nature of Fc fusions, we produced a heterodimeric Fc fusion which displays only one 7ND moiety, using a technology called strand exchange of engineered CH3 domains (SEED). The monovalent construct had properties equivalent to the parent 7ND. Furthermore, partial agonist activity appears to depend on receptor density as well as the signaling pathway examined. However, we were able to show that 7ND-Fc, but not 7ND alone, has antagonistic activity in experimental autoimmune encephalomyelitis, a murine model of multiple sclerosis.
Collapse
Affiliation(s)
- India C Severin
- Merck Serono Geneva Research Centre, 9, Chemin des Mines, 1202 Geneva, Switzerland
| | | | | | | | | | | | | |
Collapse
|
41
|
Li Z, Yao L, Li J, Zhang W, Wu X, Liu Y, Lin M, Su W, Li Y, Liang D. Celastrol nanoparticles inhibit corneal neovascularization induced by suturing in rats. Int J Nanomedicine 2012; 7:1163-73. [PMID: 22419865 PMCID: PMC3298384 DOI: 10.2147/ijn.s27860] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
PURPOSE Celastrol, a traditional Chinese medicine, is widely used in anti-inflammation and anti-angiogenesis research. However, the poor water solubility of celastrol restricts its further application. This paper aims to study the effect of celastrol nanoparticles (CNPs) on corneal neovascularization (CNV) and determine the possible mechanism. METHODS To improve the hydrophilicity of celastrol, celastrol-loaded poly(ethylene glycol)-block-poly(ɛ-caprolactone) nanopolymeric micelles were developed. The characterization of CNPs was measured by dynamic light scattering and transmission electron microscopy analysis. Celastrol loading content and release were assessed by ultraviolet-visible analysis and high performance liquid chromatography, respectively. In vitro, human umbilical vein endothelial cell proliferation and capillary-like tube formation were assayed. In vivo, suture-induced CNV was chosen to evaluate the effect of CNPs on CNV in rats. Immunohistochemistry for CD68 assessed the macrophage infiltration of the cornea on day 6 after surgery. Real-time quantitative reverse transcription-polymerase chain reaction and enzyme-linked immunosorbent assay were used to evaluate the messenger ribonucleic acid and protein levels, respectively, of vascular endothelial growth factor, matrix metalloproteinase 9, and monocyte chemoattractant protein 1 in the cornea. RESULTS The mean diameter of CNPs with spherical shape was 48 nm. The celastrol loading content was 7.36%. The release behavior of CNPs in buffered solution (pH 7.4) showed a typical two-phase release profile. CNPs inhibited the proliferation of human umbilical vein endothelial cells in a dose-independent manner and suppressed the capillary structure formation. After treatment with CNPs, the length and area of CNV reduced from 1.16 ± 0.18 mm to 0.49 ± 0.12 mm and from 7.71 ± 0.94 mm(2) to 2.29 ± 0.61 mm(2), respectively. Macrophage infiltration decreased significantly in the CNP-treated corneas. CNPs reduced the expression of vascular endothelial growth factor, matrix metalloproteinase 9, and monocyte chemoattractant protein 1 in the cornea on day 6 after suturing. CONCLUSION CNPs significantly inhibited suture-induced CNV by suppressing macrophage infiltration and the expression of vascular endothelial growth factor and matrix metalloproteinase 9 in the rat cornea.
Collapse
Affiliation(s)
- Zhanrong Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Celec P, Hodosy J, Gardlík R, Behuliak M, Pálffy R, Pribula M, Jáni P, Turňa J, Sebeková K. The effects of anti-inflammatory and anti-angiogenic DNA vaccination on diabetic nephropathy in rats. Hum Gene Ther 2012; 23:158-66. [PMID: 21939398 DOI: 10.1089/hum.2011.030] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Inflammation and angiogenesis play a crucial role in the pathomechanism of diabetic nephropathy. Monocyte chemoattractant protein 1 (MCP) is a key regulator of the immune system in kidneys, and its inhibition with a dominant-negative mutant lacking the N-terminal amino acids 2-8 (7ND) reduces renal fibrosis. Angiomotin (Amot) is a novel angiogenesis modulator. We studied the effects of inhibition of Amot and MCP using DNA vaccination on incipient diabetic nephropathy in rats. Plasmid DNA (with either 7ND or human Amot) was electroporated twice into hind-limb muscles of rats with streptozotocin-induced diabetes mellitus. Sham-electroporated diabetic rats and healthy animals served as controls. After 4 months, renal histology and biochemical analyses were performed. In sham-electroporated diabetic rats, glomerular histology revealed pathological changes. 7ND and Amot treatments reduced glomerular hypertrophy and periodic acid-Schiff positivity. In both treated groups, the expression of profibrotic (transforming growth factor-β, collagen 1), proinflammatory (interleukin-6, tumor necrosis factor-α), and proangiogenic (vascular endothelial growth factor) genes in the renal cortex was lower than in the diabetic group without treatment. The mentioned renoprotective effects could be mediated via higher total antioxidant capacity and improved glycemic control. Anti-angiogenic and anti-inflammatory DNA vaccination ameliorates the progression of glomerular pathology in an animal model of diabetic nephropathy.
Collapse
Affiliation(s)
- Peter Celec
- Institute of Molecular Biomedicine, Comenius University , 811 08 Bratislava, Slovakia.
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Tumor cells and tumor-associated macrophages: secreted proteins as potential targets for therapy. Clin Dev Immunol 2011; 2011:565187. [PMID: 22162712 PMCID: PMC3227419 DOI: 10.1155/2011/565187] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2011] [Revised: 09/09/2011] [Accepted: 09/20/2011] [Indexed: 01/02/2023]
Abstract
Inflammatory pathways, meant to defend the organism against infection and injury, as a byproduct, can promote an environment which favors tumor growth and metastasis. Tumor-associated macrophages (TAMs), which constitute a significant part of the tumor-infiltrating immune cells, have been linked to the growth, angiogenesis, and metastasis of a variety of cancers, most likely through polarization of TAMs to the M2 (alternative) phenotype. The interaction between tumor cells and macrophages provides opportunities for therapy. This paper will discuss secreted proteins as targets for intervention.
Collapse
|
44
|
Chevigné A, Fievez V, Schmit JC, Deroo S. Engineering and screening the N-terminus of chemokines for drug discovery. Biochem Pharmacol 2011; 82:1438-56. [DOI: 10.1016/j.bcp.2011.07.091] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2011] [Revised: 07/21/2011] [Accepted: 07/22/2011] [Indexed: 01/21/2023]
|
45
|
Das ND, Jung KH, Park JH, Mondol MAM, Shin HJ, Lee HS, Park KS, Choi MR, Kim KS, Kim MS, Lee SR, Chai YG. Terminalia chebula extract acts as a potential NF-κB inhibitor in human lymphoblastic T cells. Phytother Res 2011; 25:927-34. [PMID: 21509843 DOI: 10.1002/ptr.3398] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Terminalia chebula (TC) is native to southern Asia to southwestern China and is used in traditional medicine for the treatment of human ailments including malignant tumors and diabetes. This plant also has antibacterial and immunomodulatory properties. Nuclear factor kappa-light chain-enhancer of activated B cells (NF-κB) is responsible for the expression of numerous genes involved in cell survival, proliferation, angiogenesis, inflammation, invasion and metastasis, among other processes. This study aims to assess the NF-κB inhibitory effect of TC extract in human lymphoblastic T (Jurkat) cells. The effects of TC extract were investigated using the FRET-based Gene Blazer technique in transfected Jurkat-NF-κB-RE-bla cells. The concentration of TC extract required for NF-κB inhibition was determined by a cell proliferation assay. Treatment with TC extract (50 μg/mL) inhibited NF-κB activity and protected against IκBα degradation and strongly suppressed IκBα phosphorylation in Jurkat-NF-κB-RE-bla cells. This treatment might be crucial for inhibiting NF-κB translocation and activation. In addition, the TC extract downregulated certain NF-κB regulated genes, including IL-8 and MCP-1, in Jurkat-NF-κB-RE-bla cells. Moreover, gallic acid was identified from the TC extract demonstrating its ability to inhibit NF-κB activity in Jurkat-NF-κB-RE-bla cells. Further studies to identify the role of gallic acid in NF-κB inhibition may uncover the crucial antiinflammatory and antitumor properties of the TC extract.
Collapse
Affiliation(s)
- Nando Dulal Das
- Division of Molecular and Life Science, Hanyang University, Ansan, 426-791, Korea
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Rafei M, Deng J, Boivin MN, Williams P, Matulis SM, Yuan S, Birman E, Forner K, Yuan L, Castellino C, Boise LH, MacDonald TJ, Galipeau J. A MCP1 fusokine with CCR2-specific tumoricidal activity. Mol Cancer 2011; 10:121. [PMID: 21943176 PMCID: PMC3189909 DOI: 10.1186/1476-4598-10-121] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2011] [Accepted: 09/24/2011] [Indexed: 11/16/2022] Open
Abstract
Background The CCL2 chemokine is involved in promoting cancer angiogenesis, proliferation and metastasis by malignancies that express CCR2 receptor. Thus the CCL2/CCR2 axis is an attractive molecular target for anticancer drug development. Methods We have generated a novel fusion protein using GMCSF and an N-terminal truncated version of MCP1/CCL2 (6-76) [hereafter GMME1] and investigated its utility as a CCR2-specific tumoricidal agent. Results We found that distinct to full length CCL2 or its N-truncated derivative (CCL2 5-76), GMME1 bound to CCR2 on mouse lymphoma EG7, human multiple myeloma cell line U266, or murine and human medulloblastoma cell lines, and led to their death by apoptosis. We demonstrated that GMME1 specifically blocked CCR2-associated STAT3 phosphorylation and up-regulated pro-apoptotic BAX. Furthermore, GMME1 significantly inhibited EG7 tumor growth in C57BL/6 mice, and induced apoptosis of primary myeloma cells from patients. Conclusion Our data demonstrate that GMME1 is a fusokine with a potent, CCR2 receptor-mediated pro-apoptotic effect on tumor cells and could be exploited as a novel biological therapy for CCR2+ malignancies including lymphoid and central nervous system malignancies.
Collapse
Affiliation(s)
- Moutih Rafei
- The Montreal Center for Experimental Therapeutics in Cancer, McGill University, Montreal, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Abstract
In this review, we introduce the changing public perception of vaccines and immunotherapy in cancer treatments. We discuss the roles that different immunosuppressive cells play in the tumor microenvironment. Tumor associated macrophages (TAMs) and M1 and M2 macrophage phenotypes are discussed in depth. Additionally, the role that myeloid derived suppressor cells (MDSC) and T regulatory cells (Tregs) play in the tumor microenvironment is addressed. Highlighted are examples of therapies used against each suppressive cell type, which vary from the hypothetical to the ineffective; the inefficient to the successful. A variety of treatments have been tried to combat this fundamental problem, indeed the cause that allows cancerous mutated cells to survive, multiply and overtake the body. Efficient methods to disable each particular suppressive type of cell have been introduced; this review summarizes the discussion with a table to guide future development. We see gene therapy as the most innovative and flexible method to lead the charge to specifically modifying the tumor microenvironment.
Collapse
Affiliation(s)
- Elizabeth A. Vasievich
- Division of Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Leaf Huang
- Division of Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| |
Collapse
|
48
|
Kakinoki K, Nakamoto Y, Kagaya T, Tsuchiyama T, Sakai Y, Nakahama T, Mukaida N, Kaneko S. Prevention of intrahepatic metastasis of liver cancer by suicide gene therapy and chemokine ligand 2/monocyte chemoattractant protein-1 delivery in mice. J Gene Med 2011; 12:1002-13. [PMID: 21157824 DOI: 10.1002/jgm.1528] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND The prognosis of patients with hepatocellular carcinoma (HCC) remains poor, largely as a result of intrahepatic metastasis. Using a mouse model of intrahepatic metastasis, we investigated whether chemokine ligand 2/monocyte chemoattractant protein-1 (CCL2/MCP-1) could potentiate the antitumor effects of the herpes simplex virus thymidine kinase/ganciclovir (HSV-tk/GCV) system. METHODS Mouse hepatoma cells infected with recombinant adenovirus vectors expressing HSV-tk, CCL2/MCP-1 and LacZ at multiplicities of infection of Ad-tk/Ad-MCP1 = 3/0.03 (T/M(Low)), 3/3 (T/M(High)) and Ad-tk/Ad-LacZ = 3/3 (T/L) were injected into BALB/c mice. RESULTS Intrahepatic tumor growth was significantly lower in T/M(Low) mice. By contrast, no tumor suppression was observed in T/M(High) mice. The tumor-specific cytolytic activities of splenocytes from T/M(Low) and T/M(High) mice were comparable. Immunohistochemical analysis of liver tissues showed similar infiltration by Mac-1(+) and T cells in these animals, whereas the proportions of classical activated (M1) monocytes/macrophages were significantly higher in T/M(Low) mice. In addition, interleukin-12 production was elevated in these tissues. Vascular endothelial growth factor-A expression and CD31(+) microvessels were increased in T/M(High) mice. CONCLUSIONS Collectively, these results demonstrate that an adequate amount of CCL2/MCP-1, together with the HSV-tk/GCV system, may induce T helper 1-polarized antitumor effects without inducing tumor angiogenesis in the microenvironment of intrahepatic HCC progression.
Collapse
Affiliation(s)
- Kaheita Kakinoki
- Disease Control and Homeostasis, Graduate School of Medical Science, Kanazawa University, Kanazawa, Japan
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Díaz-Valdés N, Basagoiti M, Dotor J, Aranda F, Monreal I, Riezu-Boj JI, Borrás-Cuesta F, Sarobe P, Feijoó E. Induction of monocyte chemoattractant protein-1 and interleukin-10 by TGFbeta1 in melanoma enhances tumor infiltration and immunosuppression. Cancer Res 2010; 71:812-21. [PMID: 21159663 DOI: 10.1158/0008-5472.can-10-2698] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Melanoma progression is associated with the expression of different growth factors, cytokines, and chemokines. Because TGFβ1 is a pleiotropic cytokine involved not only in physiologic processes but also in cancer development, we analyzed in A375 human melanoma cells, the effect of TGFβ1 on monocyte chemoattractant protein-1 (MCP-1) and interleukin-10 (IL-10) expression, two known factors responsible for melanoma progression. TGFβ1 increased the expression of MCP-1 and IL-10 in A375 cells, an effect mediated by the cross-talk between Smad, PI3K (phosphoinositide 3-kinase)/AKT, and BRAF-MAPK (mitogen activated protein kinase) signaling pathways. Supernatants from TGFβ1-treated A375 cells enhanced MCP-1-dependent migration of monocytes, which, in turn, expressed high levels of TGF,β1, bFGF, and VEGF mRNA. Moreover, these supernatants also inhibited functional properties of dendritic cells through IL-10-dependent mechanisms. When using in vitro, the TGFβ1-blocking peptide P144, TGFβ1-dependent Smad3 phosphorylation, and expression of MCP-1 and IL-10 were inhibited. In vivo, treatment of A375 tumor-bearing athymic mice with P144 significantly reduced tumor growth, associated with a lower macrophage infiltrate and decreased intratumor MCP-1 and VEGF levels, as well as angiogenesis. Finally, in C57BL/6 mice with B16-OVA melanoma tumors, when administered with immunotherapy, P144 decreased tumor growth and intratumor IL-10 levels, linked to enhanced activation of dendritic cells and natural killer cells, as well as anti-OVA T-cell responses. These results show new effects of TGFβ1 on melanoma cells, which promote tumor progression and immunosuppression, strongly reinforcing the relevance of this cytokine as a molecular target in melanoma.
Collapse
Affiliation(s)
- Nancy Díaz-Valdés
- Universidad de Navarra, Centro de Investigación Médica Aplicada, Área de Hepatología y Terapia Génica, Pamplona, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Foster WK, Ford NL. Investigating the effect of longitudinal micro-CT imaging on tumour growth in mice. Phys Med Biol 2010; 56:315-26. [PMID: 21160110 DOI: 10.1088/0031-9155/56/2/002] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
The aim of this study is to determine the impact of longitudinal micro-CT imaging on the growth of B16F1 tumours in C57BL/6 mice. Sixty mice received 2 × 10(5) B16F1 cells subcutaneously in the hind flank and were divided into control (no scan), 'low-dose' (80 kVp, 70 mA, 8 s, 0.07 Gy), 'medium-dose' (80 kVp, 50 mA, 30 s, 0.18 Gy) and 'high-dose' (80 kVp, 50 mA, 50 s, 0.30 Gy) groups. All imaging was performed on a fast volumetric micro-CT scanner (GE Locus Ultra, London, Canada). Each mouse was imaged on days 4, 8, 12 and 16. After the final imaging session, each tumour was excised, weighed on an electronic balance, imaged to obtain the final tumour volume and processed for histology. Final tumour volume was used to evaluate the impact of longitudinal micro-CT imaging on the tumour growth. An ANOVA indicated no statistically significant difference in tumour volume (p = 0.331, α = β = 0.1) when discriminating against a treatment-sized effect. Histological samples revealed no observable differences in apoptosis or cell proliferation. We conclude that four imaging sessions, using standard protocols, over the course of 16 days did not cause significant changes in final tumour volume for B16F1 tumours in female C57BL/6 mice (ANOVA, α = β = 0.1, p = 0.331).
Collapse
Affiliation(s)
- W Kyle Foster
- Department of Physics, Ryerson University, Toronto, Ontario M5B 2K3, Canada
| | | |
Collapse
|