1
|
Li S, Yao Q, Li J, Yang H, Qian R, Zheng M, Wu N, Jiang H, Li L, Zeng Z. Inhibition of neutrophil swarming by type I interferon promotes intracellular bacterial evasion. Nat Commun 2024; 15:8663. [PMID: 39375351 PMCID: PMC11458870 DOI: 10.1038/s41467-024-53060-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 09/27/2024] [Indexed: 10/09/2024] Open
Abstract
Listeria monocytogenes (LM) possesses the ability to breach multiple barriers and elicit intricate immune responses. However, there remains a lack of explicit understanding regarding how LM evades innate immune surveillance within the body. Here, we utilized liver intravital imaging to elucidate the dynamic process of LM during infection in the liver. We discovered that LM can rapidly escape from Kupffer cells (KCs) through listeriolysin O (LLO) and proliferate within hepatocytes. Upon LM exposure to the hepatic sinusoids, neutrophils rapidly aggregate at the site of infection. Subsequently, LM can induce type I interferon (IFN-I) production primarily in the spleen, which acts systemically on neutrophils to hamper their swarming by deactivating the ERK pathway, thus evading neutrophil-mediated eradication. Furthermore, our findings suggest that virus-induced IFN-I suppresses neutrophil swarming, and COVID-19 patients exhibit impaired neutrophil aggregation function. In conclusion, our findings provide compelling evidence demonstrating that intracellular bacteria represented by LM can hijack host defense mechanisms against viral infections to evade immune surveillance. Additionally, impaired neutrophil swarming caused by IFN-I is one of the significant factors contributing to the increased susceptibility to bacterial infections following viral infections.
Collapse
Affiliation(s)
- Shimin Li
- Department of Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Qi Yao
- Key Laboratory of Immune Response and Immunotherapy, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Jiajia Li
- Key Laboratory of Immune Response and Immunotherapy, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Haoxiang Yang
- CAS Key Laboratory of Mechanical Behavior and Design of Materials, Department of Modern Mechanics, CAS Center for Excellence in Complex System Mechanics, University of Science and Technology of China, Hefei, China
| | - Rui Qian
- Department of Clinical Laboratory, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Meijuan Zheng
- Department of Clinical Laboratory, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Ning Wu
- The First Affiliated Hospital of Anhui Medical University and Institute of Clinical Immunology Anhui Medical University, Hefei, China
| | - Hongyuan Jiang
- CAS Key Laboratory of Mechanical Behavior and Design of Materials, Department of Modern Mechanics, CAS Center for Excellence in Complex System Mechanics, University of Science and Technology of China, Hefei, China
| | - Lu Li
- Department of Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
| | - Zhutian Zeng
- Department of Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
- Key Laboratory of Immune Response and Immunotherapy, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
| |
Collapse
|
2
|
Park GH, Park E, Lee SJ, Lim K, Kim J, Park JE, Jeong SY. Interferon-Induced Transmembrane Protein 1 (IFITM1) Is Downregulated in Neurofibromatosis Type 1-Associated Malignant Peripheral Nerve Sheath Tumors. Int J Mol Sci 2024; 25:9265. [PMID: 39273214 PMCID: PMC11395022 DOI: 10.3390/ijms25179265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 08/12/2024] [Accepted: 08/14/2024] [Indexed: 09/15/2024] Open
Abstract
Neurofibromatosis type 1 (NF1), an autosomal dominant genetic disorder, is caused by mutations in the NF1 gene, which encodes the GTPase-activating protein neurofibromin. The pathogenesis of the tumor progression of benign plexiform neurofibromas (PNs) and malignant peripheral nerve sheath tumors (MPNSTs) remain unclear. Here, we found that interferon-induced transmembrane protein 1 (IFITM1) was downregulated in MPNST tissues compared to those in PN tissues from patients with NF1. Overexpression of IFITM1 in NF1-associated MPNST cells resulted in a significant decrease in Ras activation (GTP-Ras) and downstream extracellular regulatory kinase 1/2 (ERK1/2) phosphorylation, whereas downregulation of IFITM1 via treatment with small interfering RNA in normal Schwann cells had the opposite result, indicating that expression levels of IFITM1 are closely associated with tumor progression in NF1. Treatment of MPNST cells with interferon-gamma (IFN-γ) significantly augmented the expression of IFITM1, thereby leading to a decrease in Ras and ERK1/2 activation. Despite the small number of patient samples, these findings may potentially provide a new target for chemotherapy in patients with NF1-associated MPNSTs. In xenograft mice injected with MPNST cells, IFN-γ treatment successfully suppressed tumor progression with increased IFITM1 expression and decreased Ras and ERK1/2 activation in tumor tissues. Collectively, these results suggest that IFITM1 is closely involved in MPNST pathogenesis and that IFN-γ is a good candidate for the therapeutic treatment of MPNSTs in NF1.
Collapse
Affiliation(s)
- Gun-Hoo Park
- Department of Medical Genetics, Ajou University School of Medicine, Suwon 16499, Republic of Korea
- Jeju Bio Research Center, Jeju Research Institute, Korea Institute of Ocean Science & Technology (KIOST), Jeju-si 63349, Republic of Korea
| | - Eunkuk Park
- Department of Medical Genetics, Ajou University School of Medicine, Suwon 16499, Republic of Korea
- Jeonbuk Institute for Food-Bioindustry, Jeonju 54810, Republic of Korea
| | - Su-Jin Lee
- Department of Medical Genetics, Ajou University School of Medicine, Suwon 16499, Republic of Korea
- Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon 16499, Republic of Korea
| | - Kyubin Lim
- Department of Medical Genetics, Ajou University School of Medicine, Suwon 16499, Republic of Korea
- Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon 16499, Republic of Korea
| | - Jeonghyun Kim
- Department of Medical Genetics, Ajou University School of Medicine, Suwon 16499, Republic of Korea
- Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon 16499, Republic of Korea
| | - Jun Eun Park
- Department of Pediatrics, Anam Hospital, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Seon-Yong Jeong
- Department of Medical Genetics, Ajou University School of Medicine, Suwon 16499, Republic of Korea
- Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon 16499, Republic of Korea
| |
Collapse
|
3
|
Xiong F, Wang Q, Wu GH, Liu WZ, Wang B, Chen YJ. Direct and indirect effects of IFN-α2b in malignancy treatment: not only an archer but also an arrow. Biomark Res 2022; 10:69. [PMID: 36104718 PMCID: PMC9472737 DOI: 10.1186/s40364-022-00415-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 08/22/2022] [Indexed: 12/02/2022] Open
Abstract
Interferon-α2b (IFN-α2b) is a highly active cytokine that belongs to the interferon-α (IFN-α) family. IFN-α2b has beneficial antiviral, antitumour, antiparasitic and immunomodulatory activities. Direct and indirect antiproliferative effects of IFN-α2b have been found to occur via multiple pathways, mainly the JAK-STAT pathway, in certain cancers. This article reviews mechanistic studies and clinical trials on IFN-α2b. Potential regulators of the function of IFN-α2b were also reviewed, which could be utilized to relieve the poor response to IFN-α2b. IFN-α2b can function not only by enhancing the systematic immune response but also by directly killing tumour cells. Different parts of JAK-STAT pathway activated by IFN-α2b, such as interferon alpha and beta receptors (IFNARs), Janus kinases (JAKs) and IFN‐stimulated gene factor 3 (ISGF3), might serve as potential target for enhancing the pharmacological action of IFN-α2b. Despite some issues that remain to be solved, based on current evidence, IFN-α2b can inhibit disease progression and improve the survival of patients with certain types of malignant tumours. More efforts should be made to address potential adverse effects and complications.
Collapse
|
4
|
EMT and Inflammation: Crossroads in HCC. J Gastrointest Cancer 2022; 54:204-212. [PMID: 35020133 DOI: 10.1007/s12029-021-00801-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/29/2021] [Indexed: 10/19/2022]
Abstract
Hepatocellular carcinoma is one of the major causes of cancer-related deaths worldwide and is associated with several inflammatory mediators, since 90% of HCCs occur based on chronic hepatitis B or C, alcoholism or increasingly metabolic syndrome-associated inflammation. EMT is a physiological process, with coordinated changes in epithelial gene signatures and is regulated by multiple factors, including cytokines and growth factors such as TGFβ, EGF, and FGF. Recent reports propose a strong association between EMT and inflammation, which is also correlated with tumor aggressiveness and poor outcomes. Cellular heterogeneity results collectively as an outcome of EMT, inflammation, and the tumor microenvironment, and it plays a fundamental role in the progression, complexity of cancer, and chemoresistance. In this review, we highlight recent developments concerning the association of EMT and inflammation in the context of HCC progression. Identifying potential EMT-related biomarkers and understanding EMT regulatory molecules will likely contribute to promising developments in clinical practice and will be a valuable tool for predicting metastasis in general and specifically in HCC.
Collapse
|
5
|
Meng X, Chen Y, Macip S, Leppard K. PML-II regulates ERK and AKT signal activation and IFNα-induced cell death. Cell Commun Signal 2021; 19:70. [PMID: 34215258 PMCID: PMC8252201 DOI: 10.1186/s12964-021-00756-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 06/03/2021] [Indexed: 11/23/2022] Open
Abstract
Background The requirement of promyelocytic leukaemia protein (PML) in interferon (IFN)-induced cell apoptosis is well-established. However, the exact mechanisms by which the multiple isoforms of PML protein participate in this process remain not well-understood. We previously demonstrated that PML isoform II (PML-II) positively regulates induced gene expression during a type I IFN response and evaluate here how PML-II contributes to IFNα-induced cell death. Methods HeLa cells were transiently depleted of PML-II by siRNA treatment and the response of these cells to treatment with IFNα assessed by molecular assays of mRNA and proteins associated with IFN and apoptosis responses. Results In HeLa cells, death during IFNα stimulation was reduced by prior PML-II depletion. PML-II removal also considerably decreased the induced expression of pro-apoptotic ISGs such as ISG54 (IFIT2), and substantially impaired or prevented expression of PUMA and TRAIL, proteins that are associated with the intrinsic and extrinsic apoptotic pathways respectively. Thirdly, PML-II depletion enhanced ERK and AKT pro-survival signaling activation suggesting that PML-II normally suppresses signaling via these pathways, and that lack of PML-II hence led to greater than normal activation of AKT signaling upon IFNα stimulation and consequently increased resistance to IFNα-induced apoptosis. Conclusions The positive contribution of PML-II to the expression of various IFNα-induced pro-apoptotic proteins and its inhibition of pro-survival signaling together provide a mechanistic explanation for reduced apoptosis under conditions of PML deficiency and may account for at least part of the role of PML as a tumor suppressor gene. Video Abstract
Supplementary Information The online version contains supplementary material available at 10.1186/s12964-021-00756-5.
Collapse
Affiliation(s)
- Xueqiong Meng
- School of Basic Medicine, Henan University of Science and Technology, Luoyang, China.,School of Life Sciences, University of Warwick, Coventry, UK
| | - Yixiang Chen
- School of Basic Medicine, Henan University of Science and Technology, Luoyang, China.,Henan International Joint Laboratory of Thrombosis and Hemostasis, Luoyang, China.,Mechanisms of Cancer and Aging Laboratory, Department of Molecular and Cell Biology, University of Leicester, Leicester, UK
| | - Salvador Macip
- Mechanisms of Cancer and Aging Laboratory, Department of Molecular and Cell Biology, University of Leicester, Leicester, UK.,FoodLab, Faculty of Health Sciences, Universitat Oberta de Catalunya, Barcelona, Spain
| | - Keith Leppard
- School of Life Sciences, University of Warwick, Coventry, UK.
| |
Collapse
|
6
|
Yu S, Xu T, Dai J, Ma M, Tang H, Chi Z, Si L, Cui C, Sheng X, Kong Y, Guo J. TERT copy gain predicts the outcome of high-dose interferon α-2b therapy in acral melanoma. Onco Targets Ther 2018; 11:4097-4104. [PMID: 30046245 PMCID: PMC6054280 DOI: 10.2147/ott.s158239] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Background Asian populations are more likely to develop acral melanoma (AM) than Caucasians. The high-dose interferon (HD-IFN) α-2b regimen is the main adjuvant treatment for AM. TERT encodes the catalytic subunit of telomerase reverse transcriptase, which plays an important role in melanoma. Frequent TERT mutation and increased TERT gene expression have been described in AM. Our study aimed to investigate the status and the clinical significance of TERT copy number in a large cohort of patients with AM and to analyze the relationship between TERT copy number gain and the efficiency of HD-IFN. Patients and methods A total of 573 melanoma samples were retrospectively collected and analyzed for TERT copy number via Sanger sequencing. Clinical data of patients were also collected. Results TERT copy gain (copy number >2) was detected in 257 of the 573 patients with AM (44.9%). Of the 573 patients, 81 (14.1%) had a high copy gain (copy number >4). Patients with ulceration showed a significantly higher copy gain rate of TERT compared to the patients without ulceration (P=0.028). Patients with a tumor thicker than 4 mm also had a higher copy number rate of TERT than those with <4 mm (P=0.048). Our results showed that the overall survival (OS) was not significantly different between patients with and without TERT copy gain (P=0.890). However, among the 278 patients who received an HD-IFN regimen, Kaplan–Meier survival analysis demonstrated a significant correlation between TERT copy gain and relapse-free survival (RFS) (P=0.008). In addition, multivariate Cox regression assays validated TERT copy gain to be an independent prognostic factor of RFS for patients with AM undergoing HD-IFN therapy (hazard ratio =1.50; P=0.019). Conclusion The copy number status of TERT might be a predictor for HD-IFN efficacy, but it is not a prognostic factor of OS in patients with AM.
Collapse
Affiliation(s)
- Sifan Yu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, Beijing 100142, China, ;
| | - Tianxiao Xu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, Beijing 100142, China, ;
| | - Jie Dai
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, Beijing 100142, China, ;
| | - Meng Ma
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, Beijing 100142, China, ;
| | - Huan Tang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, Beijing 100142, China, ;
| | - Zhihong Chi
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, Beijing 100142, China, ;
| | - Lu Si
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, Beijing 100142, China, ;
| | - Chuanliang Cui
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, Beijing 100142, China, ;
| | - Xinan Sheng
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, Beijing 100142, China, ;
| | - Yan Kong
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, Beijing 100142, China, ;
| | - Jun Guo
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, Beijing 100142, China, ;
| |
Collapse
|
7
|
Wang Y, Nan J, Willard B, Wang X, Yang J, Stark GR. Negative regulation of type I IFN signaling by phosphorylation of STAT2 on T387. EMBO J 2017; 36:202-212. [PMID: 27852626 PMCID: PMC5239994 DOI: 10.15252/embj.201694834] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Revised: 10/06/2016] [Accepted: 10/11/2016] [Indexed: 01/17/2023] Open
Abstract
The transcription factor ISGF3, comprised of IRF9 and tyrosine-phosphorylated STATs 1 and 2, transmits the signal from the type I interferon receptor to the genome. We have discovered a novel phosphorylation of STAT2 on T387 that negatively regulates this response. In most untreated cell types, the majority of STAT2 is phosphorylated on T387 constitutively. In response to interferon-β, the T387A mutant of STAT2 is much more effective than wild-type STAT2 in mediating the expression of many interferon-stimulated genes, in protecting cells against virus infection, and in inhibiting cell growth. Interferon-β-treated cells expressing wild-type STAT2 contain much less ISGF3 capable of binding to an interferon-stimulated response element than do cells expressing T387A STAT2. T387 lies in a cyclin-dependent kinase (CDK) consensus sequence, and CDK inhibitors decrease T387 phosphorylation. Using CDK inhibitors to reverse the constitutive inhibitory phosphorylation of T387 of U-STAT2 might enhance the efficacy of type I interferons in many different clinical settings.
Collapse
Affiliation(s)
- Yuxin Wang
- Key Laboratory of Marine Drugs, Ministry of Education Ocean University of China, Qingdao, Shandong, China
- Department of Cancer Biology, Lerner Research Institute The Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Jing Nan
- Department of Cancer Biology, Lerner Research Institute The Cleveland Clinic Foundation, Cleveland, OH, USA
- Institute of Cancer Biology & Drug Screening, School of Life Sciences Lanzhou University, Lanzhou, Gansu, China
| | - Belinda Willard
- Proteomics and Metabolomics Laboratory, Lerner Research Institute The Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Xin Wang
- Department of Immunology, Lerner Research Institute The Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Jinbo Yang
- Key Laboratory of Marine Drugs, Ministry of Education Ocean University of China, Qingdao, Shandong, China
- Institute of Cancer Biology & Drug Screening, School of Life Sciences Lanzhou University, Lanzhou, Gansu, China
| | - George R Stark
- Department of Cancer Biology, Lerner Research Institute The Cleveland Clinic Foundation, Cleveland, OH, USA
| |
Collapse
|
8
|
Lin YM, Yu BC, Chiu WT, Sun HY, Chien YC, Su HC, Yen SY, Lai HW, Bai CH, Young KC, Tsao CW. Fluoxetine regulates cell growth inhibition of interferon-α. Int J Oncol 2016; 49:1746-54. [DOI: 10.3892/ijo.2016.3650] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2016] [Accepted: 07/25/2016] [Indexed: 11/05/2022] Open
|
9
|
Gaudio E, Tarantelli C, Kwee I, Barassi C, Bernasconi E, Rinaldi A, Ponzoni M, Cascione L, Targa A, Stathis A, Goodstal S, Zucca E, Bertoni F. Combination of the MEK inhibitor pimasertib with BTK or PI3K-delta inhibitors is active in preclinical models of aggressive lymphomas. Ann Oncol 2016; 27:1123-1128. [PMID: 26961147 DOI: 10.1093/annonc/mdw131] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Accepted: 03/02/2016] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Lymphomas are among the most common human cancers and represent the cause of death for still too many patients. The B-cell receptor with its downstream signaling pathways represents an important therapeutic target for B-cell lymphomas. Here, we evaluated the activity of the MEK1/2 inhibitor pimasertib as single agent and in combination with other targeted drugs in lymphoma preclinical models. MATERIALS AND METHODS Cell lines derived mature B-cell lymphomas were exposed to increasing doses of pimasertib alone. Immunoblotting and gene expression profiling were performed. Combination of pimasertib with idelalisib or ibrutinib was assessed. RESULTS Pimasertib as single agent exerted a dose-dependent antitumor activity across a panel of 23 lymphoma cell lines, although at concentrations higher than reported for solid tumors. Strong synergism was observed with pimasertib combined with the PI3K inhibitor idelalisib and the BTK inhibitor ibrutinib in cell lines derived from diffuse large B-cell lymphoma (DLBCL) and mantle cell lymphoma. The data were confirmed in an in vivo experiment treating DLBCL xenografts with pimasertib and ibrutinib. CONCLUSION The data presented here provide the basis for further investigation of regimens including pimasertib in relapsed and refractory lymphomas.
Collapse
Affiliation(s)
- E Gaudio
- Lymphoma and Genomics Research Program, IOR Institute of Oncology Research, Bellinzona
| | - C Tarantelli
- Lymphoma and Genomics Research Program, IOR Institute of Oncology Research, Bellinzona
| | - I Kwee
- Lymphoma and Genomics Research Program, IOR Institute of Oncology Research, Bellinzona; Dalle Molle Institute for Artificial Intelligence (IDSIA), Manno; Swiss Institute of Bioinformatics (SIB), Lausanne, Switzerland
| | - C Barassi
- Lymphoma and Genomics Research Program, IOR Institute of Oncology Research, Bellinzona
| | - E Bernasconi
- Lymphoma and Genomics Research Program, IOR Institute of Oncology Research, Bellinzona
| | - A Rinaldi
- Lymphoma and Genomics Research Program, IOR Institute of Oncology Research, Bellinzona
| | - M Ponzoni
- Unit of Lymphoid Malignancies, Department of Onco-Haematology, San Raffaele Scientific Institute, Milan, Italy
| | - L Cascione
- Lymphoma and Genomics Research Program, IOR Institute of Oncology Research, Bellinzona; IOSI Oncology Institute of Southern Switzerland, Bellinzona, Switzerland
| | - A Targa
- Lymphoma and Genomics Research Program, IOR Institute of Oncology Research, Bellinzona
| | - A Stathis
- IOSI Oncology Institute of Southern Switzerland, Bellinzona, Switzerland
| | - S Goodstal
- Translational and Biomarker Research, Translational Innovation Platform Oncology, EMD Serono Research and Development Institute, Billerica, USA
| | - E Zucca
- IOSI Oncology Institute of Southern Switzerland, Bellinzona, Switzerland
| | - F Bertoni
- Lymphoma and Genomics Research Program, IOR Institute of Oncology Research, Bellinzona; IOSI Oncology Institute of Southern Switzerland, Bellinzona, Switzerland.
| |
Collapse
|
10
|
Li J, Lehmann C, Chen X, Romerio F, Lu W. Total chemical synthesis of human interferon alpha-2b via native chemical ligation. J Pept Sci 2015; 21:554-60. [PMID: 25810135 DOI: 10.1002/psc.2760] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2014] [Revised: 01/17/2015] [Accepted: 01/19/2015] [Indexed: 02/04/2023]
Abstract
Interferon-alpha (IFNα) is a cytokine that orchestrates innate and adaptive immune responses and potently inhibits proliferation of normal and tumor cells. These properties have warranted the use of IFNα in clinical practice for the treatment of several viral infections and malignancies. However, overexpression of IFNα leads to immunopathology observed in the context of chronic viral infections and autoimmune conditions. Thus, it is desirable to develop therapeutic approaches that aim at suppressing excessive IFNα production. To that end, artificial evolution of peptides from phage display libraries represents a strategy that seeks to disrupt the interaction between IFNα and its cell surface receptor and thus inhibit the ensuing biological effects. Mirror-image phage display that screens peptide libraries against the D-enantiomer is particularly attractive because it allows for identification of proteolysis-resistant D-peptide inhibitors. This approach, however, relies on the availability of chemically synthesized D-IFNα composed entirely of D-amino acids. Here, we describe the synthesis and biological properties of IFNα2b of 165 amino acid residues produced by native chemical ligation, which represents an important first step toward the discovery of D-peptide antagonists with potential therapeutic applications.
Collapse
Affiliation(s)
- Jing Li
- Institute of Human Virology, University of Maryland School of Medicine, 725 West Lombard Street, Baltimore, MD, 21201, USA.,Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, China
| | - Clara Lehmann
- Institute of Human Virology, University of Maryland School of Medicine, 725 West Lombard Street, Baltimore, MD, 21201, USA.,First Department of Internal Medicine, University of Cologne, Cologne, Germany
| | - Xishan Chen
- Institute of Human Virology, University of Maryland School of Medicine, 725 West Lombard Street, Baltimore, MD, 21201, USA
| | - Fabio Romerio
- Institute of Human Virology, University of Maryland School of Medicine, 725 West Lombard Street, Baltimore, MD, 21201, USA
| | - Wuyuan Lu
- Institute of Human Virology, University of Maryland School of Medicine, 725 West Lombard Street, Baltimore, MD, 21201, USA
| |
Collapse
|
11
|
Involvement of ERK pathway in interferon alpha-mediated antiviral activity against hepatitis C virus. Cytokine 2015; 72:17-24. [DOI: 10.1016/j.cyto.2014.11.031] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Revised: 11/23/2014] [Accepted: 11/26/2014] [Indexed: 01/19/2023]
|
12
|
Parody JP, Ceballos MP, Quiroga AD, Frances DE, Carnovale CE, Pisani GB, Alvarez ML, Carrillo MC. FoxO3a modulation and promotion of apoptosis by interferon-α2b in rat preneoplastic liver. Liver Int 2014; 34:1566-77. [PMID: 24289330 DOI: 10.1111/liv.12421] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2013] [Accepted: 11/21/2013] [Indexed: 12/27/2022]
Abstract
BACKGROUND FoxO3a, a member of the FOXO family of transcription factors, is expressed in adult liver and modulates the expression of genes involved in apoptosis. FoxO3a is post-translationally regulated, negatively by PI3K/Akt and MAPK/Erk and positively by oxidative stress/JNK pathways. In previous works, we have demonstrated that interferon-α2b (IFN-α2b) induces apoptosis of hepatic preneoplastic foci through the production of reactive oxygen species (ROS). AIMS To investigate the post-translational signal events triggered by the oxidative stress induced by IFN-α2b and the modulation of FoxO3a transcriptional activity during these events in rat preneoplastic liver. METHODS Adult male Wistar rats were subjected to a two-phase model of hepatocarcinogenesis. A group of animals received IFN-α2b and another group received IFN-α2b and ascorbic acid (ASC), by intraperitoneal injection. Lipid peroxidation, immunohistochemistry, immunoblotting, co-immunoprecipitation and sqRT-PCR assays were performed to explore the role of ROS, JNK, Akt, Erk, FoxO3a, β-catenin and PUMA in the IFN-α2b-mediated apoptotic mechanism. RESULTS In vivo IFN-α2b treatment induced endogenous production of ROS which activated JNK. IFN-α2b blocked the activation of Akt and Erk, avoiding FoxO3a activity repression. Activated JNK was responsible for the nuclear translocation and transcriptional activity of FoxO3a which positively modulated the expression of PUMA, a proapoptotic player. In addition, nuclear FoxO3a competed for the nuclear β-catenin associated to TCF, inhibiting the canonical Wnt signalling pathway. CONCLUSIONS The data presented here propose a model in which in vivo IFN-α2b treatment induces nuclear translocation and transcriptional activity of FoxO3a, triggering the mitochondrial apoptotic pathway in hepatic preneoplastic foci.
Collapse
Affiliation(s)
- Juan P Parody
- Instituto de Fisiología Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Rosario, Argentina
| | | | | | | | | | | | | | | |
Collapse
|
13
|
Asmana Ningrum R. Human interferon alpha-2b: a therapeutic protein for cancer treatment. SCIENTIFICA 2014; 2014:970315. [PMID: 24741445 PMCID: PMC3967813 DOI: 10.1155/2014/970315] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/03/2014] [Accepted: 01/22/2014] [Indexed: 06/03/2023]
Abstract
Human interferon alpha (hIFNα) is a wide biological activity cytokine that is used in hepatitis and cancer treatments. It regulates many genes that are involved in antiviral and antiproliferative activities. This mini review focuses on human interferon alpha-2b (hIFNα-2b) as therapeutic protein for cancer treatment. The review covers hIFNα-2b molecular characteristic and its molecular mechanism by Janus activated kinase/signal transducer activation of transcription (JAK-STAT) pathway. The JAK-STAT pathway regulates not only proteins involved in inhibition of proliferation but also apoptosis. As additional discussion of clinical applications, the use of recombinant hIFNα-2b (rhIFNα-2b) as therapeutic protein in several types of cancer is also explained.
Collapse
Affiliation(s)
- Ratih Asmana Ningrum
- Research Center for Biotechnology, Cibinong Science Center, Jalan Raya Bogor km 46, Cibinong, Bogor, West Java 16911, Indonesia
| |
Collapse
|
14
|
Nishimura Y, Natsume A, Ito M, Hara M, Motomura K, Fukuyama R, Sumiyoshi N, Aoki I, Saga T, Lee HJ, Wakabayashi T, Kim SU. Interferon-β Delivery via Human Neural Stem Cell Abates Glial Scar Formation in Spinal Cord Injury. Cell Transplant 2013; 22:2187-201. [DOI: 10.3727/096368912x657882] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Glial scar formation is the major impedance to axonal regrowth after spinal cord injury (SCI), and scar-modulating treatments have become a leading therapeutic goal for SCI treatment. In this study, human neural stem cells (NSCs) encoding interferon-β (INF-β) gene were administered intravenously to mice 1 week after SCI. Animals receiving NSCs encoding IFN-β exhibited significant neurobehavioral improvement, electrophysiological recovery, suppressed glial scar formation, and preservation of nerve fibers in lesioned spinal cord. Systemic evaluation of SCI gliosis lesion site with lesion-specific microdissection, genome-wide microarray, and MetaCore pathway analysis identified upregulation of toll-like receptor 4 (TLR4) in SCI gliosis lesion site, and this led us to focus on TLR4 signaling in reactive astrocytes. Examination of primary astrocytes from TLR4 knockout mice, and in vivo inhibition of TLR4, revealed that the effect of IFN-β on the suppression of glial scar formation in SCI requires TLR4 stimulation. These results suggest that IFN-β delivery via intravenous injection of NSCs following SCI inhibits glial scar formation in spinal cord through stimulation of TLR4 signaling.
Collapse
Affiliation(s)
| | - Atsushi Natsume
- Department of Neurosurgery, Nagoya University, Nagoya, Japan
| | - Motokazu Ito
- Department of Neurosurgery, Nagoya University, Nagoya, Japan
| | - Masahito Hara
- Department of Neurosurgery, Nagoya University, Nagoya, Japan
| | - Kazuya Motomura
- Department of Neurosurgery, Nagoya University, Nagoya, Japan
| | | | | | - Ichio Aoki
- MR Molecular Imaging Team, Molecular Imaging Center, National Institute of Radiological Sciences, Chiba, Japan
| | - Tsuneo Saga
- MR Molecular Imaging Team, Molecular Imaging Center, National Institute of Radiological Sciences, Chiba, Japan
| | - Hong J. Lee
- Medical Research Institute, Chung-Ang University College of Medicine, Seoul, Korea
| | | | - Seung U. Kim
- Medical Research Institute, Chung-Ang University College of Medicine, Seoul, Korea
- Division of Neurology, Department of Medicine, UBC Hospital, University of British Columbia, Vancouver, Canada
| |
Collapse
|
15
|
Yin H, Xie F, Zhang J, Yang Y, Deng B, Sun J, Wang Q, Qu X, Mao H. Combination of interferon-α and 5-fluorouracil induces apoptosis through mitochondrial pathway in hepatocellular carcinoma in vitro. Cancer Lett 2011; 306:34-42. [PMID: 21474236 DOI: 10.1016/j.canlet.2011.02.017] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2011] [Revised: 02/14/2011] [Accepted: 02/15/2011] [Indexed: 11/30/2022]
Abstract
Many clinical reports have proven that the combination therapy of interferon-alpha plus 5-fluorouracil is remarkably effective for advanced hepatocellular carcinoma (HCC). However, the mechanism of this therapy is not well understood. Here, we demonstrated that the combination therapy synergistically inhibited the growth of Fas-negative HCC cells, arrested cell-cycle progression and induced apoptosis. Moreover, the combination therapy significantly increased the protein expression of caspase-8, activated Bid and cytochrome c. Meanwhile, the expression of anti-apoptotic gene Bcl-xL was reduced and intracellular calcium elevated obviously during the early stage of treatment. Therefore, mitochondrial pathway was involved in the apoptosis of Fas-negative HCC cells induced by IFN-α/5-FU and Ca(2+) partially promoted the beneficial effect against HCC.
Collapse
Affiliation(s)
- Haipeng Yin
- Institute of Basic Medical Sciences, Qi Lu Hospital, Shandong University, Jinan, PR China
| | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Huang W, Yang AH, Matsumoto D, Collette W, Marroquin L, Ko M, Aguirre S, Younis HS. PD0325901, a Mitogen-Activated Protein Kinase Kinase Inhibitor, Produces Ocular Toxicity in a Rabbit Animal Model of Retinal Vein Occlusion. J Ocul Pharmacol Ther 2009; 25:519-30. [DOI: 10.1089/jop.2009.0060] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Affiliation(s)
- Wenhu Huang
- Drug Safety, Pfizer Global Research and Development, San Diego, California
| | - Amy H. Yang
- Drug Safety, Pfizer Global Research and Development, San Diego, California
| | - Diane Matsumoto
- Drug Safety, Pfizer Global Research and Development, San Diego, California
| | - Walter Collette
- Drug Safety, Pfizer Global Research and Development, San Diego, California
| | - Lisa Marroquin
- Drug Safety, Pfizer Global Research and Development, San Diego, California
| | - Mira Ko
- Drug Safety, Pfizer Global Research and Development, San Diego, California
| | - Shirley Aguirre
- Drug Safety, Pfizer Global Research and Development, San Diego, California
| | - Husam S. Younis
- Drug Safety, Pfizer Global Research and Development, San Diego, California
| |
Collapse
|
17
|
Ito M, Natsume A, Takeuchi H, Shimato S, Ohno M, Wakabayashi T, Yoshida J. Type I interferon inhibits astrocytic gliosis and promotes functional recovery after spinal cord injury by deactivation of the MEK/ERK pathway. J Neurotrauma 2009; 26:41-53. [PMID: 19196180 DOI: 10.1089/neu.2008.0646] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
Formation of a glial scar is one of the major obstacles to axonal growth after injury to the adult CNS. In this study, we have addressed this issue by focusing on reactive astrocytes in a mouse model of spinal cord injury (SCI). First, we attempted to identify profile changes in the expression of astrocytic gliosis 10 days after injury by using gliosis-specific microdissection, genome-wide microarray, and MetaCore(trade mark) pathway analysis. This systematic data processing revealed many intriguing activated pathways. However, considering that proliferation/mitosis is one of the most prominent features of reactive astrocytes, we focused on the functional role of the Ras-MEK-ERK signaling cascades in reactive astrocytes. SCI-induced proliferation of reactive astrocytes in the lesion is in accordance with the increase in the expression and phosphorylation of MEK-ERK. Second, to reduce reactive gliosis after SCI, liposomes containing the interferon-beta (IFN-beta) gene were administered locally 30 min after injury. At 14 days after this treatment, GFAP-positive intensity and MEK-ERK phosphorylation at the lesion were reduced. In the animals receiving the IFN-beta gene, significant recovery of neurobehavior and parameters of electrophysiology following SCI was revealed by assessments of rotarod performance and improvements in the Basso Mouse Scale for locomotion and cortical motor-evoked potentials. SCI resulted in the degeneration of biotinylated dextran amine-labeled descending corticospinal tract axons, but the IFN-beta gene delivery induced regrowth of a large number of corticospinal tract axons. These results suggest that liposome-mediated IFN-beta gene delivery inhibits glial scar formation after SCI and promotes functional recovery.
Collapse
Affiliation(s)
- Motokazu Ito
- Department of Neurosurgery, Nagoya University School of Medicine , Nagoya, Japan
| | | | | | | | | | | | | |
Collapse
|
18
|
Tochizawa S, Masumori N, Yanai Y, Ohmoto Y, Yabuuchi Y, Tsukamoto T. Antitumor effects of a combination of interferon-alpha and sorafenib on human renal carcinoma cell lines. ACTA ACUST UNITED AC 2009; 29:271-8. [PMID: 19129670 DOI: 10.2220/biomedres.29.271] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
To support the role of interferon (IFN)-alpha and sorafenib combination therapy against renal cell carcinoma (RCC), the effects of IFN-alpha and sorafenib on tumor growth, vascular endothelial growth factor (VEGF) production, and phosphorylation levels of extracellular signal-regulated kinase (ERK) and mitogen-activated protein/ERK kinase (MEK) were examined using several cultured RCC cell lines (ACHN, Caki-1, Caki-2, SMKT-R1, SMKT-R2, SMKT-R3 and SMKT-R4). IFN-alpha or sorafenib alone inhibited the proliferation of all the cell lines except Caki-2, while combined treatment with the two agents showed enhanced inhibitory effects compared to treatment with each agent alone. VEGF production was inhibited by IFN-alpha alone in ACHN and SMKT-R2 cells and by sorafenib alone in ACHN, Caki-1, SMKT-R1 and SMKT-R2 cells. However, sorafenib increased VEGF production by Caki-2 cells. Interestingly, combined treatment with the two agents suppressed VEGF production by SMKT-R1 and SMKT-R2 cells more strongly than IFN-alpha or sorafenib alone. Although phosphorylated ERK (p-ERK) was increased after 30 min of treatment with IFN-alpha alone, no difference was observed between control and IFN-alpha-treated cells after 2 h. Sorafenib decreased p-ERK in ACHN, Caki-1, SMKT-R1 and SMKT-R2 cells, but increased p-ERK in Caki-2, SMKT-R3 and SMKT-R4 cells, after 2 h. Combined treatment with IFN-alpha and sorafenib decreased p-ERK compared to treatment with each agent alone in all cell lines except Caki-2. However, IFN-alpha did not inhibit the p-ERK increase induced by sorafenib in Caki-2 cells. Phosphorylated MEK showed similar patterns to p-ERK after the various treatments. In conclusion, combined treatment with IFN-alpha and sorafenib suppressed cell proliferation and VEGF production more strongly than treatment with each agent alone in several RCC cell lines.
Collapse
Affiliation(s)
- Shiro Tochizawa
- Free Radical Research Institute, Otsuka Pharmaceutical Co. Ltd, Tokushima, Japan.
| | | | | | | | | | | |
Collapse
|
19
|
Kaluz S, Kaluzová M, Liao SY, Lerman M, Stanbridge EJ. Transcriptional control of the tumor- and hypoxia-marker carbonic anhydrase 9: A one transcription factor (HIF-1) show? Biochim Biophys Acta Rev Cancer 2009; 1795:162-72. [PMID: 19344680 DOI: 10.1016/j.bbcan.2009.01.001] [Citation(s) in RCA: 124] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2008] [Revised: 01/08/2009] [Accepted: 01/12/2009] [Indexed: 12/22/2022]
Abstract
Transcriptional activation by hypoxia is mediated by the hypoxia-inducible factor (HIF) via binding to the hypoxia-responsive element (HRE). Hypoxia in solid tumors associates with poorer outcome of the disease and reliable cellular markers of tumor hypoxia would represent a valuable diagnostic marker and a potential therapeutic target. In this category, carbonic anhydrase IX (CAIX) is one of the most promising candidates. Here, we summarize the knowledge about transcriptional regulation of CA9. The HRE is the central regulatory element in the CA9 promoter, whereas other elements are limited to lesser roles of amplification of signals received at the HRE. The analysis of known mechanisms of activation of CA9 reveals the prominent role of the HIF-1 pathway. Experimental paradigms with uncoupled HIF-1alpha stability and transcriptional activity (pericellular hypoxia, proteasomal inhibitor) provide evidence that CA9 expression monitors transcriptional activity of HIF-1, rather than the abundance of HIF-1alpha. Furthermore, these paradigms could provide a corollary to some of the apparently discordant cases (CAIX+, HIF-1alpha-) or (CAIX-, HIF-1alpha+) observed in vivo. In conclusion, the existing data support the notion that CA9, due to the unique structure of its promoter, is one of the most sensitive endogenous sensors of HIF-1 activity.
Collapse
Affiliation(s)
- Stefan Kaluz
- Department of Microbiology and Molecular Genetics, University of California, Irvine, CA 92697-4025, USA.
| | | | | | | | | |
Collapse
|
20
|
Zitzmann K, Brand S, Baehs S, Göke B, Meinecke J, Spöttl G, Meyer H, Auernhammer CJ. Novel interferon-lambdas induce antiproliferative effects in neuroendocrine tumor cells. Biochem Biophys Res Commun 2006; 344:1334-41. [PMID: 16650825 DOI: 10.1016/j.bbrc.2006.04.043] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2006] [Accepted: 04/09/2006] [Indexed: 11/23/2022]
Abstract
Interferon-alpha (IFN-alpha) is used for biotherapy of neuroendocrine carcinomas. The interferon-lambdas (IL-28A/B and IL-29) are a novel group of interferons. In this study, we investigated the effects of the IFN-lambdas IL-28A and IL-29 on human neuroendocrine BON1 tumor cells. Similar to IFN-alpha, incubation of BON1 cells with IL-28A (10 ng/ml) and IL-29 (10 ng/ml) induced phosphorylation of STAT1, STAT2, and STAT3, significantly decreased cell numbers in a proliferation assay, and induced apoptosis as demonstrated by poly(ADP-ribose) polymerase (PARP)-cleavage, caspase-3-cleavage, and DNA-fragmentation. Stable overexpression of suppressor of cytokine signaling proteins (SOCS1 and SOCS3) completely abolished the aforementioned effects indicating that SOCS proteins act as negative regulators of IFN-lambda signaling in BON1 cells. In conclusion, the novel IFN-lambdas IL-28A and IL-29 potently induce STAT signaling and antiproliferative effects in neuroendocrine BON1 tumor cells. Thus, IFN-lambdas may hint a promising new approach in the antiproliferative therapy of neuroendocrine tumors.
Collapse
Affiliation(s)
- Kathrin Zitzmann
- Department of Internal Medicine II, University-Hospital Munich-Grosshadern, University of Munich, Munich, Germany
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Inamura K, Matsuzaki Y, Uematsu N, Honda A, Tanaka N, Uchida K. Rapid inhibition of MAPK signaling and anti-proliferation effect via JAK/STAT signaling by interferon-α in hepatocellular carcinoma cell lines. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2005; 1745:401-10. [PMID: 16054712 DOI: 10.1016/j.bbamcr.2005.06.003] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2005] [Revised: 06/09/2005] [Accepted: 06/10/2005] [Indexed: 01/04/2023]
Abstract
The potential anti-proliferation effect of interferon-alpha (IFN-alpha) against hepatocellular carcinoma (HCC) and its growth inhibitory mechanisms remain unclear. We examined four human HCC cell lines and every cell line had the anti-proliferative effect of IFN-alpha. The PLC/PRF/5 cell line, which expressed the IFN receptor most abundantly, responded most effectively to IFN-alpha stimulation. Here, we delineate the anti-proliferative effect of IFN-alpha via the MAPK pathway in human HCC cell lines. IFN-alpha retarded G1/S transition with no evidence of apoptosis and inhibited cell proliferation. IFN-alpha diminished the phosphorylation of both extracellular signal-regulated kinase (ERK) and mitogen-activated ERK-regulating kinase (MEK), but not Raf, within 5 min. Knockdown of signal transducers of activation and transcription1 (STAT1) or Janus kinase1 (JAK1) suppressed the reduction of phosphorylation both of ERK and MEK and diminished the growth inhibition by IFN-alpha. These results suggest that IFN-alpha induces anti-proliferative signaling via the JAK/STAT pathway downstream of IFN-alpha receptors and may reduce the growth stimulation signaling by cross-talk with the MEK/ERK pathway without IFN-alpha-induced transcription.
Collapse
Affiliation(s)
- Kentaro Inamura
- Department of Molecular Biology and Molecular Oncology, Graduate School of Comprehensive Human Science, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba, Ibaraki 305-8575, Japan
| | | | | | | | | | | |
Collapse
|
22
|
Caraglia M, Marra M, Pelaia G, Maselli R, Caputi M, Marsico SA, Abbruzzese A. Alpha-interferon and its effects on signal transduction pathways. J Cell Physiol 2005; 202:323-35. [PMID: 15389589 DOI: 10.1002/jcp.20137] [Citation(s) in RCA: 95] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Interferon-alpha (IFNalpha) is a recombinant protein widely used in the therapy of several neoplasms such as myeloma, renal cell carcinoma, epidermoid cervical and head and neck tumors, and melanoma. IFNalpha, the first cytokine to be produced by recombinant DNA technology, has emerged as an important regulator of cancer cell growth and differentiation, affecting cellular communication and signal transduction pathways. However, the way by which tumor cell growth is directly suppressed by IFNalpha is not well known. Wide evidence exists on the possibility that cancer cells undergo apoptosis after the exposure to the cytokine. Here we will review the consolidate signal transducer and activator of transcription (STAT)-dependent mechanism of action of IFNalpha. We will discuss data obtained by us and others on the triggering of the stress-dependent kinase pathway induced by IFNalpha and its correlations with the apoptotic process. The regulation of the expression of proteins involved in apoptosis occurrence will be also described. In this regard, IFNalpha is emerging as a post-translational controller of the intracellular levels of the apoptosis-related protein tissue transglutaminase (tTG). This new way of regulation of tTG occurs through the modulation of their proteasome-dependent degradation induced by the cytokine. Until today, inconsistent data have been obtained regarding the clinical effectiveness of IFNalpha in the therapy of solid tumors. In fact, the benefit of IFNalpha treatment is limited to some neoplasms while others are completely or partially resistant. The mechanisms of tumor resistance to IFNalpha have been studied in vitro. The alteration of JAK-STAT components of the IFNalpha-induced signaling, can be indeed a mechanism of resistance to IFN. However, we have recently described a reactive mechanism of protection of tumor cells from the apoptosis induced by IFNalpha dependent on the epidermal growth factor (EGF)-mediated Ras/extracellular signal regulated kinase (Erk) signaling. The involvement of the Ras-->Erk pathway in the protection of tumor cells from the apoptosis induced by IFNalpha is further demonstrated by both Ras inactivation by RASN17 transfection and mitogen extracellular signal regulated kinase 1 (Mek-1) inhibition by exposure to PD098059. These data strongly suggest that the specific disruption of the latter could be a useful approach to potentiate the antitumour activity of IFNalpha against human tumors based on the new mechanistic insights achieved in the last years.
Collapse
Affiliation(s)
- Michele Caraglia
- Department of Biochemistry and Biophysics, Second University of Naples, Via Costantinopoli, Naples, Italy.
| | | | | | | | | | | | | |
Collapse
|
23
|
Peleg-Shulman T, Tsubery H, Mironchik M, Fridkin M, Schreiber G, Shechter Y. Reversible PEGylation: A Novel Technology To Release Native Interferon α2 over a Prolonged Time Period. J Med Chem 2004; 47:4897-904. [PMID: 15369394 DOI: 10.1021/jm0497693] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Many peptide and protein drugs have a short circulatory half-life in vivo. The covalent attachment of polyethylene glycol (PEG) chains (PEGylation) can overcome this deficiency, but pegylated peptides and proteins are often inactive. In this study, we present a novel PEG-IFNalpha2 conjugate, PEG(40)-FMS-IFNalpha2, capable of regenerating native interferon alpha2 (IFNalpha2) at a slow rate under physiological conditions. A 2-sulfo-9-fluorenylmethoxycarbonyl (FMS) containing bifunctional reagent, MAL-FMS-NHS, has been synthesized, enabling the linkage of a 40 kDa PEG-SH to IFNalpha2 through a slowly hydrolyzable bond. By use of a BIAcore binding assay, the in vitro rate of regeneration of native interferon was estimated to have a half-life of 65 h. Following subcutaneous administration to rats and monitoring circulating antiviral activity, active IFNalpha2 levels peaked at 50 h, with substantial levels still being detected 200 h after administration. This value contrasts with a half-life of about 1 h measured for unmodified interferon. The concentration of active IFNalpha2 scaled linearly with the quantity injected. Comparing subcutaneous to intravenous administration of PEG(40)-FMS-IFNalpha2, we found that the long circulatory lifetime of IFNalpha2 was affected both by the slow rate of absorption of the PEGylated protein from the subcutaneous volume and by the slow rate of discharge from the PEG in circulation. A numerical simulation of the results was in good agreement with the results observed in vivo. The pharmacokinetic profile of this novel IFNalpha2 conjugate combines a prolonged maintenance in vivo with the regeneration of active-native IFNalpha2, ensuring ready access to peripheral tissues and thus an overall advantage over currently used formulations.
Collapse
Affiliation(s)
- Tal Peleg-Shulman
- Department of Biological Chemistry and Organic Chemistry, The Weizmann Institute of Science, Rehovot 76100, Israel
| | | | | | | | | | | |
Collapse
|
24
|
Stamatos NM, Curreli S, Zella D, Cross AS. Desialylation of glycoconjugates on the surface of monocytes activates the extracellular signal-related kinases ERK 1/2 and results in enhanced production of specific cytokines. J Leukoc Biol 2003; 75:307-13. [PMID: 14634064 DOI: 10.1189/jlb.0503241] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Modulation of the sialic acid content of cell-surface glycoproteins and glycolipids influences the functional capacity of cells of the immune system. The role of sialidase(s) and the consequent desialylation of cell surface glycoconjugates in the activation of monocytes have not been established. In this study, we show that desialylation of glycoconjugates on the surface of purified monocytes using exogenous neuraminidase (NANase) activated extracellular signal-regulated kinase 1/2 (ERK 1/2), an intermediate in intracellular signaling pathways. Elevated levels of phosphorylated ERK 1/2 were detected in desialylated monocytes after 2 h of NANase treatment, and increased amounts persisted for at least 2 additional hours. Desialylation of cell surface glycoconjugates also led to increased production of interleukin (IL)-6, macrophage inflammatory protein (MIP)-1alpha, and MIP-1beta by NANase-treated monocytes that were maintained in culture. Neither increased levels of phosphorylated ERK 1/2 nor enhanced production of cytokines were detected when NANase was heat-inactivated before use, demonstrating the specificity of NANase action. Treatment of monocytes with gram-negative bacterial lipopolysaccharide (LPS) also led to enhanced production of IL-6, MIP-1alpha, and MIP-1beta. The amount of each of these cytokines that was produced was markedly increased when monocytes were desialylated with NANase before exposure to LPS. These results suggest that changes in the sialic acid content of surface glycoconjugates influence the activation of monocytes.
Collapse
Affiliation(s)
- Nicholas M Stamatos
- Institute of Human Virology, Department of Medicine, University of Maryland Medical Center, University of Maryland, Baltimore, MD 21201, USA.
| | | | | | | |
Collapse
|
25
|
Abstract
The second part of this review examines the use of recombinant interferon-alpha (rIFNalpha) in the following solid tumours: superficial bladder cancer, Kaposi's sarcoma, head and neck cancer, gastrointestinal cancers, lung cancer, mesothelioma and ovarian, breast and cervical malignancies. In superficial bladder cancer, intravesical rIFNalpha has a promising role as second-line therapy in patients resistant or intolerant to intravesical bacille Calmette-Guérin (BCG). In HIV-associated Kaposi's sarcoma, rIFNalpha is active as monotherapy and in combination with antiretroviral agents, especially in patients with CD4 counts >200/mm(3), no prior opportunistic infections and nonvisceral disease. rIFNalpha has shown encouraging results when used in combination with retinoids in the chemoprevention of head and neck squamous cell cancers. It is effective in the chemoprevention of hepatocellular cancer in hepatitis C-seropositive patients. In neuroendocrine tumours, including carcinoid tumour, low-dosage (</=3 MU) or intermediate-dosage (5 to 10 MU) rIFNalpha is indicated as second-line treatment, either with octreotide or alone in patients resistant to somatostatin analogues. Intracavitary IFNalpha may be useful in malignant pleural effusions from mesothelioma. Similarly, intraperitoneal IFNalpha may have a role in the treatment of minimal residual disease in ovarian cancer. In breast cancer, the only possible role for IFNalpha appears to be intralesional administration for resistant disease. IFNalpha may have a role as a radiosensitising agent for the treatment of cervical cancer; however, this requires confirmation in randomised trials. On the basis of current evidence, the routine use of rIFNalpha is not recommended in the therapy of head and neck squamous cell cancers, upper gastrointestinal tract, colorectal and lung cancers, or mesothelioma. Pegylated IFNalpha (peginterferon-alpha) is an exciting development that offers theoretical advantages of increased efficacy, reduced toxicity and improved compliance. Further data from randomised studies in solid tumours are needed where rIFNalpha has activity, such as neuroendocrine tumours, minimal residual disease in ovarian cancer, and cervical cancer. A better understanding of the biological mechanisms that determine response to rIFNalpha is needed. Studies of IFNalpha-stimulated gene expression, which are now feasible, should help to identify molecular predictors of response and allow us to target therapy more selectively to patients with solid tumours responsive to IFNalpha.
Collapse
Affiliation(s)
- Sundar Santhanam
- Department of Oncology, Leicester Royal Infirmary, Leicester, UK.
| | | | | |
Collapse
|
26
|
Lee M, Koh WS. Raf-independent and MEKK1-dependent activation of NF-kappaB by hydrogen peroxide in 70Z/3 pre-B lymphocyte tumor cells. J Cell Biochem 2003; 88:545-56. [PMID: 12532330 DOI: 10.1002/jcb.10376] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
We have previously demonstrated that hydrogen peroxide (H(2)O(2)) treatment of murine 70Z/3 pre-B lymphocytes inhibits the immune response to lipopolysaccharide by attenuating signaling through c-Jun N-terminal kinase (JNK) activation. In the present study, we further examined the signaling intermediates responsible for immunosuppression by H(2)O(2), focusing on NF-kappaB, a dimeric transcription factor whose activation is implicated in a number of immune response. Treatment of 70Z/3 pre-B cells with H(2)O(2) caused activation of NF-kappaB in the nuclei by detection of NF-kappaB specific DNA binding, concomitant with phosphorylation of IkappaBalpha. H(2)O(2) stimulation of NF-kappaB occurred within 20 min of treatment, reached maximum level at 60 min, and sustained for 2 h or more. Especially, MEK1 may contribute to H(2)O(2)-induced NF-kappaB activation as shown in the inhibition of NF-kappaB binding activity by the MEK1 inhibitor, PD 98059, and H(2)O(2)-induced MEK1 activation. However, H(2)O(2) exhibited no effect on the activity of Raf-1 kinase, which was an upstream activator of MEK1. Furthermore, B-58l and alpha-hydroxyfarnesylphosphonic acid, two inhibitors of Ras, did not block NF-kappaB activation. In addition, the transient transfection of a dominant negative Ras (RasN17) construct showed a negligible inhibitory effect on the activation of NF-kappaB by H(2)O(2). Instead, treatment of 70Z/3 cells with H(2)O(2) resulted in the activation of MAPK kinase kinase 1 (MEKK1) as well as JNK. Therefore, our data suggest that H(2)O(2) regulates the activity of NF-kappaB by MEK1 activation through MEKK1-dependent but Ras/Raf-independent mechanism.
Collapse
Affiliation(s)
- Michael Lee
- Korea Institute of Toxicology, Korea Research Institute of Chemical Technology, P.O. Box 107, Yusong, Daejeon 305-600, Republic of Korea.
| | | |
Collapse
|
27
|
Romerio F, Zella D. MEK and ERK inhibitors enhance the anti-proliferative effect of interferon-alpha2b. FASEB J 2002; 16:1680-2. [PMID: 12206994 DOI: 10.1096/fj.02-0120fje] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Interferon (IFN)-alpha, initially characterized as an antiviral cytokine, affects several cellular functions. It is used in clinical practice for the treatment of several tumors, including hematopoietic malignancies, due to its antiproliferative effects. To better characterize the molecular mechanism(s) underlying this property, we conducted our studies in purified primary CD4+ T cells stimulated with anti-CD3 and interleukin (IL)-2. Upon treatment with IFN-alpha, the cells were blocked in the G0/G1 phase of the cell cycle and exhibited impaired entry into S phase and reduced proliferation. Moreover, we detected short- and long-term inhibition of extracellular signal-regulated kinase (ERK) and mitogen-activated ERK-regulating kinase (MEK) function, known to control cellular proliferation. The activity of the upstream regulators, Ras and Raf-1, was not affected. Analysis of downstream events controlled by the MEK/ERK pathway showed reduced activity of cyclin-dependent kinase (Cdk)-2 and -4, high levels of the mitotic inhibitors, p21Waf1 and p27Kipl, and decreased cyclin D and E expression. When IFN-alpha was used in combination with MEK and ERK inhibitors, we observed a dose-dependent additive effect in reducing cellular proliferation. Our data demonstrate that IFN-alpha may be associated with other molecules to inhibit cellular growth by targeting the MEK/ERK pathway. This may eventually lead to new clinical strategies to strengthen its anticancer effect.
Collapse
Affiliation(s)
- Fabio Romerio
- Institute of Human Virology, University of Maryland Biotechnology Institute, Baltimore, Maryland 2120, USA
| | | |
Collapse
|
28
|
Reddy SM, Hsiao KHK, Abernethy VE, Fan H, Longacre A, Lieberthal W, Rauch J, Koh JS, Levine JS. Phagocytosis of apoptotic cells by macrophages induces novel signaling events leading to cytokine-independent survival and inhibition of proliferation: activation of Akt and inhibition of extracellular signal-regulated kinases 1 and 2. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2002; 169:702-13. [PMID: 12097372 DOI: 10.4049/jimmunol.169.2.702] [Citation(s) in RCA: 89] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Recent evidence indicates that phagocytic clearance of apoptotic cells, initially thought to be a silent event, can modulate macrophage (M phi) function. We show in this work that phagocytic uptake of apoptotic cells or bodies, in the absence of serum or soluble survival factors, inhibits apoptosis and maintains viability of primary cultures of murine peritoneal and bone marrow M phi with a potency approaching that of serum-supplemented medium. Apoptotic uptake also profoundly inhibits the proliferation of bone marrow M phi stimulated to proliferate by M-CSF. While inhibition of proliferation is an unusual property for survival factors, the combination of increased survival and decreased proliferation may aid the M phi in its role as a scavenger during resolution of inflammation. The ability of apoptotic cells to promote survival and inhibit proliferation appears to be the result of simultaneous activation of Akt and inhibition of the mitogen-activated protein kinases extracellular signal-regulated kinase (ERK)1 and ERK2 (ERK1/2). While several activators of the innate immune system, or danger signals, also inhibit apoptosis and proliferation, danger signals and necrotic cells differ from apoptotic cells in that they activate, rather than inhibit, ERK1/2. These signaling differences may underlie the opposing tendencies of apoptotic cells and danger signals in promoting tolerance vs immunity.
Collapse
Affiliation(s)
- Suman M Reddy
- Renal Section, Evans Memorial Department of Clinical Research, Department of Medicine, Boston University Medical Center, Boston, MA 02118, USA
| | | | | | | | | | | | | | | | | |
Collapse
|