1
|
Thapa R, Gupta S, Gupta G, Bhat AA, Smriti, Singla M, Ali H, Singh SK, Dua K, Kashyap MK. Epithelial-mesenchymal transition to mitigate age-related progression in lung cancer. Ageing Res Rev 2024; 102:102576. [PMID: 39515620 DOI: 10.1016/j.arr.2024.102576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 10/27/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024]
Abstract
Epithelial-Mesenchymal Transition (EMT) is a fundamental biological process involved in embryonic development, wound healing, and cancer progression. In lung cancer, EMT is a key regulator of invasion and metastasis, significantly contributing to the fatal progression of the disease. Age-related factors such as cellular senescence, chronic inflammation, and epigenetic alterations exacerbate EMT, accelerating lung cancer development in the elderly. This review describes the complex mechanism among EMT and age-related pathways, highlighting key regulators such as TGF-β, WNT/β-catenin, NOTCH, and Hedgehog signalling. We also discuss the mechanisms by which oxidative stress, mediated through pathways involving NRF2 and ROS, telomere attrition, regulated by telomerase activity and shelterin complex, and immune system dysregulation, driven by alterations in cytokine profiles and immune cell senescence, upregulate or downregulate EMT induction. Additionally, we highlighted pathways of transcription such as SNAIL, TWIST, ZEB, SIRT1, TP53, NF-κB, and miRNAs regulating these processes. Understanding these mechanisms, we highlight potential therapeutic interventions targeting these critical molecules and pathways.
Collapse
Affiliation(s)
- Riya Thapa
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Saurabh Gupta
- Chameli Devi Institute of Pharmacy, Department of Pharmacology, Indore, Madhya Pradesh, India
| | - Gaurav Gupta
- Centre for Research Impact & Outcome-Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India.
| | - Asif Ahmad Bhat
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Smriti
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Madhav Singla
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Haider Ali
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144411, India; Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, Australia
| | - Kamal Dua
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, Australia; Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, NSW 2007, Australia
| | - Manoj Kumar Kashyap
- Molecular Oncology Laboratory, Amity Stem Cell Institute, Amity Medical School, Amity University Haryana, Panchgaon (Manesar), Gurugram, Haryana, India.
| |
Collapse
|
2
|
Wang Y, Wang W, Wang H, Qin L, Zhang M, Zhang Y, Wang Y, Hao C, Qu M, Wang G. Clinical prognostic significance of xeroderma pigmentosum group C and IFN‑γ in non‑small cell lung cancer. Oncol Lett 2024; 27:259. [PMID: 38646492 PMCID: PMC11027110 DOI: 10.3892/ol.2024.14392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 02/28/2024] [Indexed: 04/23/2024] Open
Abstract
Lung cancer is the most common cancer in the world due to its high incidence and recurrence. Genetic instability is one of the main factors leading to its occurrence, development and poor prognosis. Decreased xeroderma pigmentosum group C (XPC) expression notably enhances the stem cell properties of lung cancer cells and increases their proliferation and migration. Additionally, patients with lung cancer and low XPC expression had a poor prognosis. The purpose of the present study was to analyze the effect of XPC and IFN-γ on the clinical prognosis of patients with non-small cell lung cancer (NSCLC). Lung adenocarcinoma specimens were collected from a total of 140 patients with NSCLC. Additionally, from these 140 patients, 48 paracarcinoma tissue specimens were also collected, which were later used to construct tissue microarrays. The expression of XPC and IFN-γ in cancer tissues and in paraneoplastic tissues was detected using immunohistochemistry. The prognosis and overall survival of patients were determined through telephone follow-up. The results showed a positive correlation between expression of XPC and IFN-γ in NSCLC. Additionally, high expression of both markers was associated with a favorable prognosis in patients with NSCLC. The aforementioned findings suggest that the expression of XPC and IFN-γ has prognostic value in clinical practice and is expected to become a marker for clinical application.
Collapse
Affiliation(s)
- Yongming Wang
- Department of Thoracic Surgery, Shandong Provincial Hospital, Shandong University, Jinan, Shandong 250021, P.R. China
- Department of Thoracic Surgery, Translational Medical Center, Weifang Second People's Hospital (Weifang Respiratory Disease Hospital), Weifang, Shandong 261041, P.R. China
| | - Weiyu Wang
- Department of Thoracic Surgery, Translational Medical Center, Weifang Second People's Hospital (Weifang Respiratory Disease Hospital), Weifang, Shandong 261041, P.R. China
- Department of Otolaryngology, and Head and Neck Surgery, Shandong Provincial ENT Hospital, Jinan, Shandong 250022, P.R. China
| | - Huaijie Wang
- Department of Thoracic Surgery, Translational Medical Center, Weifang Second People's Hospital (Weifang Respiratory Disease Hospital), Weifang, Shandong 261041, P.R. China
| | - Liya Qin
- Department of Thoracic Surgery, Translational Medical Center, Weifang Second People's Hospital (Weifang Respiratory Disease Hospital), Weifang, Shandong 261041, P.R. China
- Biopharmaceutical Laboratory, Key Laboratory of Shandong Province Colleges and Universities, School of Life Sciences and Technology, Weifang Medical University, Weifang, Shandong 261053, P.R. China
| | - Meijia Zhang
- Department of Thoracic Surgery, Translational Medical Center, Weifang Second People's Hospital (Weifang Respiratory Disease Hospital), Weifang, Shandong 261041, P.R. China
| | - Yong Zhang
- Department of Thoracic Surgery, Translational Medical Center, Weifang Second People's Hospital (Weifang Respiratory Disease Hospital), Weifang, Shandong 261041, P.R. China
| | - Yubing Wang
- Biopharmaceutical Laboratory, Key Laboratory of Shandong Province Colleges and Universities, School of Life Sciences and Technology, Weifang Medical University, Weifang, Shandong 261053, P.R. China
| | - Changcheng Hao
- Department of Thoracic Surgery, Translational Medical Center, Weifang Second People's Hospital (Weifang Respiratory Disease Hospital), Weifang, Shandong 261041, P.R. China
| | - Meihua Qu
- Department of Thoracic Surgery, Translational Medical Center, Weifang Second People's Hospital (Weifang Respiratory Disease Hospital), Weifang, Shandong 261041, P.R. China
- Biopharmaceutical Laboratory, Key Laboratory of Shandong Province Colleges and Universities, School of Life Sciences and Technology, Weifang Medical University, Weifang, Shandong 261053, P.R. China
| | - Gongchao Wang
- Department of Thoracic Surgery, Shandong Provincial Hospital, Shandong University, Jinan, Shandong 250021, P.R. China
| |
Collapse
|
3
|
Thome CD, Tausche P, Hohenberger K, Yang Z, Krammer S, Trufa DI, Sirbu H, Schmidt J, Finotto S. Short-chain fatty acids induced lung tumor cell death and increased peripheral blood CD4+ T cells in NSCLC and control patients ex vivo. Front Immunol 2024; 15:1328263. [PMID: 38650948 PMCID: PMC11033355 DOI: 10.3389/fimmu.2024.1328263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 03/14/2024] [Indexed: 04/25/2024] Open
Abstract
Background Despite therapy advances, one of the leading causes of cancer deaths still remains lung cancer. To improve current treatments or prevent non-small cell lung cancer (NSCLC), the role of the nutrition in cancer onset and progression needs to be understood in more detail. While in colorectal cancer, the influence of local microbiota derived SCFAs have been well investigated, the influence of SCFA on lung cancer cells via peripheral blood immune system should be investigated more deeply. In this respect, nutrients absorbed via the gut might affect the tumor microenvironment (TME) and thus play an important role in tumor cell growth. Objective This study focuses on the impact of the short-chain fatty acid (SCFA) Sodium Butyrate (SB), on lung cancer cell survival. We previously described a pro-tumoral role of glucose on A549 lung adenocarcinoma cell line. In this study, we wanted to know if SB would counteract the effect of glucose and thus cultured A549 and H520 in vitro with and without SB in the presence or absence of glucose and investigated how the treatment with SB affects the survival of lung cancer cells and its influence on immune cells fighting against lung cancer. Methods In this study, we performed cell culture experiments with A549, H520 and NSCLC-patient-derived epithelial cells under different SB levels. To investigate the influence on the immune system, we performed in vitro culture of peripheral mononuclear blood cells (PBMC) from control, smoker and lung cancer patients with increasing SB concentrations. Results To investigate the effect of SB on lung tumor cells, we first analyzed the effect of 6 different concentrations of SB on A549 cells at 48 and 72 hours cell culture. Here we found that, SB treatment reduced lung cancer cell survival in a concentration dependent manner. We next focused our deeper analysis on the two concentrations, which caused the maximal reduction in cell survival. Here, we observed that SB led to cell cycle arrest and induced early apoptosis in A549 lung cancer cells. The expression of cell cycle regulatory proteins and A549 lung cancer stem cell markers (CD90) was induced. Additionally, this study explored the role of interferon-gamma (IFN-γ) and its receptor (IFN-γ-R1) in combination with SB treatment, revealing that, although IFN-γ-R1 expression was increased, IFN-γ did not affect the efficacy of SB in reducing tumor cell viability. Furthermore, we examined the effects of SB on immune cells, specifically CD8+ T cells and natural killer (NK) cells from healthy individuals, smokers, and NSCLC patients. SB treatment resulted in a decreased production of IFN-γ and granzyme B in CD8+ T cells and NK cells. Moreover, SB induced IFN-γ-R1 in NK cells and CD4+ T cells in the absence of glucose both in PBMCs from controls and NSCLC subjects. Conclusion Overall, this study highlights the potential of SB in inhibiting lung cancer cell growth, triggering apoptosis, inducing cell cycle arrest, and modulating immune responses by activating peripheral blood CD4+ T cells while selectively inducing IFN-γ-R1 in NK cells in peripheral blood and inhibiting peripheral blood CD8+ T cells and NK cells. Thus, understanding the mechanisms of action of SB in the TME and its influence on the immune system provide valuable insights of potentially considering SB as a candidate for adjunctive therapies in NSCLC.
Collapse
Affiliation(s)
- Carolin D. Thome
- Department of Molecular Pneumology, University Medical School Hospital Erlangen (UKER) Friedrich-Alexander-University (FAU), Erlangen-Nürnberg, Germany
| | - Patrick Tausche
- Department of Molecular Pneumology, University Medical School Hospital Erlangen (UKER) Friedrich-Alexander-University (FAU), Erlangen-Nürnberg, Germany
| | - Katja Hohenberger
- Department of Molecular Pneumology, University Medical School Hospital Erlangen (UKER) Friedrich-Alexander-University (FAU), Erlangen-Nürnberg, Germany
| | - Zuqin Yang
- Department of Molecular Pneumology, University Medical School Hospital Erlangen (UKER) Friedrich-Alexander-University (FAU), Erlangen-Nürnberg, Germany
| | - Susanne Krammer
- Department of Molecular Pneumology, University Medical School Hospital Erlangen (UKER) Friedrich-Alexander-University (FAU), Erlangen-Nürnberg, Germany
| | - Denis I. Trufa
- Department of Thoracic Surgery, University Medical School Hospital Erlangen (UKER), Friedrich-Alexander University of Erlangen-Nürnberg (FAU), Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen, Germany
| | - Horia Sirbu
- Department of Thoracic Surgery, University Medical School Hospital Erlangen (UKER), Friedrich-Alexander University of Erlangen-Nürnberg (FAU), Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen, Germany
| | - Joachim Schmidt
- Department of Anesthesiology, University Medical School Hospital Erlangen (UKER), Friedrich-Alexander University of Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Susetta Finotto
- Department of Molecular Pneumology, University Medical School Hospital Erlangen (UKER) Friedrich-Alexander-University (FAU), Erlangen-Nürnberg, Germany
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen, Germany
| |
Collapse
|
4
|
Grard M, Chatelain C, Delaunay T, Pons-Tostivint E, Bennouna J, Fonteneau JF. Homozygous Co-Deletion of Type I Interferons and CDKN2A Genes in Thoracic Cancers: Potential Consequences for Therapy. Front Oncol 2021; 11:695770. [PMID: 34249754 PMCID: PMC8266377 DOI: 10.3389/fonc.2021.695770] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 06/07/2021] [Indexed: 12/24/2022] Open
Abstract
Homozygous deletion (HD) of the tumor suppressor gene CDKN2A is the most frequent genetic alteration in malignant pleural mesothelioma and is also frequent in non-small cell lung cancers. This HD is often accompanied by the HD of the type I interferons (IFN I) genes that are located closed to the CDKN2A gene on the p21.3 region of chromosome 9. IFN I genes encode sixteen cytokines (IFN-α, IFN-β…) that are implicated in cellular antiviral and antitumor defense and in the induction of the immune response. In this review, we discuss the potential influence of IFN I genes HD on thoracic cancers therapy and speak in favor of better taking these HD into account in patients monitoring.
Collapse
Affiliation(s)
- Marion Grard
- Université de Nantes, Inserm, CRCINA, Nantes, France.,Labex IGO, Immunology Graft Oncology, Nantes, France
| | - Camille Chatelain
- Université de Nantes, Inserm, CRCINA, Nantes, France.,Labex IGO, Immunology Graft Oncology, Nantes, France
| | - Tiphaine Delaunay
- Université de Nantes, Inserm, CRCINA, Nantes, France.,Labex IGO, Immunology Graft Oncology, Nantes, France
| | - Elvire Pons-Tostivint
- Université de Nantes, Inserm, CRCINA, Nantes, France.,Labex IGO, Immunology Graft Oncology, Nantes, France.,CHU de Nantes, oncologie thoracique et digestive, Université de Nantes, Nantes, France
| | - Jaafar Bennouna
- Université de Nantes, Inserm, CRCINA, Nantes, France.,Labex IGO, Immunology Graft Oncology, Nantes, France.,CHU de Nantes, oncologie thoracique et digestive, Université de Nantes, Nantes, France
| | - Jean-François Fonteneau
- Université de Nantes, Inserm, CRCINA, Nantes, France.,Labex IGO, Immunology Graft Oncology, Nantes, France
| |
Collapse
|
5
|
Zhao J, Cheng M, Gai J, Zhang R, Du T, Li Q. SPOCK2 Serves as a Potential Prognostic Marker and Correlates With Immune Infiltration in Lung Adenocarcinoma. Front Genet 2020; 11:588499. [PMID: 33244319 PMCID: PMC7683796 DOI: 10.3389/fgene.2020.588499] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 10/21/2020] [Indexed: 12/31/2022] Open
Abstract
Lung adenocarcinoma (LUAD) is one of the major types of lung cancer. Tumor-infiltrating immune cells (TIICs) are positively associated with overall survival (OS) in LUAD. The SPARC/osteonectin, cwcv and kazal-like domains proteoglycan 2 (SPOCK2) is a complex type of secreted proteoglycan involved in forming a protective barrier against viral infection. The purpose of this study was to investigate the relationship between SPOCK2 and TIICs and the prognostic role of SPOCK2 in LUAD. The GEPIA2, GEO, CPTAC, and HPA databases were analyzed to examine both the mRNA and protein expression of SPOCK2 in LUAD. GEPIA2 and the Kaplan-Meier Plotter (KM Plotter) were used to evaluate the prognostic value of SPOCK2 in LUAD patients. TCGA data were examined for a correlation between SPOCK2 expression and clinical characteristics. Gene enrichment analyses were performed to explore the underlying mechanism of SPOCK2 based on LinkedOmics. RegNetwork was used to predict the regulators of SPOCK2. The correlation between SPOCK2 and TIICs, including immune infiltration level and relative proportion was investigated via TIMER. KM Plotter was also used to evaluate the prognostic role of SPOCK2 expression in LUAD with enriched and decreased TIIC subgroups. We found SPOCK2 was significantly downregulated in LUAD compared with that in non-tumor controls and was correlated with clinical parameters. Moreover, a high SPOCK2 expression level predicted better survival. The SPOCK2-associated regulatory network was constructed. SPOCK2 influenced the TIIC infiltration level and relative proportion in LUAD. Furthermore, a high SPOCK2 expression level was associated with a favorable prognosis in enriched CD4 + T cells and macrophage subgroups in LUAD. In conclusion, a high level of SPOCK2 expression predicted favorable prognosis and was significantly correlated with TIICs in LUAD. Therefore, the expression of SPOCK2 may affect the prognosis of LUAD partly due to TIICs.
Collapse
Affiliation(s)
- Jinming Zhao
- Department of Pathology, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Ming Cheng
- Department of Pathology, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Junda Gai
- Department of Pathology, College of Basic Medical Sciences, China Medical University, Shenyang, China.,Department of Pathology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Ruochen Zhang
- Department of Chronic Disease Epidemiology, Yale School of Public Health, Yale University, New Haven, CT, United States
| | - Tengjiao Du
- Department of Pathology, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Qingchang Li
- Department of Pathology, College of Basic Medical Sciences, China Medical University, Shenyang, China.,Department of Pathology, The First Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
6
|
Liang P, Wang G, Liu X, Wang Z, Wang J, Gao W. Spatiotemporal combination of thermosensitive polypeptide fused interferon and temozolomide for post-surgical glioblastoma immunochemotherapy. Biomaterials 2020; 264:120447. [PMID: 33069137 DOI: 10.1016/j.biomaterials.2020.120447] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Revised: 10/06/2020] [Accepted: 10/09/2020] [Indexed: 02/08/2023]
Abstract
Cancer recurrence post surgical resection is of considerable challenge especially in glioblastoma (GBM) therapy. Herein, we demonstrate that interferon-alpha (IFN) fused to a body temperature-sensitive elastin-like polypeptide (IFN-ELP(V)) formed a depot in situ when injected into GBM resection cavity in a mouse brain orthotopic model of GBM. Notably, IFN-ELP(V) in the depot showed a zero-order release kinetics, resulting in dramatically improved pharmacokinetics and biodistribution, and thus inhibited GBM recurrence by stimulating antitumor immunoresponse as compared to IFN. More importantly, when combined with subsequent intraperitoneal injection of temozolomide (TMZ), IFN-ELP(V) could much more effectively suppress post-surgical GBM recurrence than IFN, leading to a remarkably enhanced GBM-free survival rate (60%) over IFN (12.5%). Our findings implicate that the spatiotemporally-programmed combination of IFN-ELP(V) and TMZ leads to the synergy of post-surgical GBM immunochemotherapy, thereby providing a new and effective strategy for cancer therapy.
Collapse
Affiliation(s)
- Ping Liang
- Department of Neurosurgery, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, 102218, PR China
| | - Guihuai Wang
- Department of Neurosurgery, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, 102218, PR China
| | - Xinyu Liu
- Department of Geriatric Dentistry, Beijing Laboratory of Biomedical Materials, Peking University School and Hospital of Stomatology, Beijing, 100081, China; Biomedical Engineering Department, Peking University, Beijing, 100191, PR China
| | - Zhuoran Wang
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Protein and Peptide Pharmaceutical, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, PR China
| | - Jing Wang
- Department of Neurosurgery, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, 102218, PR China
| | - Weiping Gao
- Department of Geriatric Dentistry, Beijing Laboratory of Biomedical Materials, Peking University School and Hospital of Stomatology, Beijing, 100081, China; Biomedical Engineering Department, Peking University, Beijing, 100191, PR China.
| |
Collapse
|
7
|
IL-10 suppresses IFN-γ-mediated signaling in lung adenocarcinoma. Clin Exp Med 2020; 20:449-459. [PMID: 32306136 DOI: 10.1007/s10238-020-00626-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 04/02/2020] [Indexed: 01/04/2023]
Abstract
Interleukin-10 (IL-10) is a pleiotropic cytokine produced by a wide variety of cells. It has been implicated in cancer progression, and at times, it has seemingly contradictory effects. The impact of IL-10 on immune components in the context of cancer has been intensively investigated, but its effect on cancer cells remains poorly understood. In this study, we examined the expression of IL-10 and IL-10 receptor 1 (IL-10R1) in resected locally advanced lung adenocarcinoma by immunohistochemistry. IL-10 immunoreactivity was stronger in intraepithelial regions than in stroma. The amount of IL-10 found either in intraepithelial or in stromal regions had no prognostic value, but the relative distribution of IL-10 in these two locations was related to cancer-immune phenotypes. High expression of IL-10R1 by tumor cells was significantly correlated with poor prognosis, suggesting that IL-10-mediated signaling may induce cancer cell intrinsic effects that promote cancer progression. Functional analysis using human lung adenocarcinoma cell lines revealed that IL-10 did not directly affect cell proliferation and migration. Incubation of cancer cells with IL-10 suppressed interferon-γ (IFN-γ)-induced STAT1 phosphorylation and inhibited the transcription of IFN-γ-targeted genes, such as CXCL9, CXCL10, and PD-L1. IL-10 enhanced IFN-γ-induced SOCS1 and SOCS3 expression, an effect that might be responsible for the downregulation of STAT1 activity in cancer cells. Our findings provide a rationale for targeting IL-10 on cancer cells as a potential strategy for treating cancer.
Collapse
|
8
|
Mitselou A, Grammeniatis V, Varouktsi A, Papadatos SS, Katsanos K, Galani V. Proinflammatory cytokines in irritable bowel syndrome: a comparison with inflammatory bowel disease. Intest Res 2020; 18:115-120. [PMID: 32013318 PMCID: PMC7000645 DOI: 10.5217/ir.2019.00125] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 12/03/2019] [Indexed: 12/13/2022] Open
Abstract
Background/Aims Irritable bowel syndrome (IBS) is a common disease often considered as a functional intestinal disorder. Inflammation in IBS is a quite intriguing theory. The aim of this study was to investigate tumor necrosis factor (TNF)-α, interleukin (IL)-1β and IL-6 expression in inflammatory bowel disease (IBD) patients, IBS patients and normal controls. Methods IBS and IBD patients along with normal controls were recruited in the study. In all groups, 2 pinch biopsies were taken at each of 3 anatomical sites (terminal ileum, cecum, and rectum). IBS patients were also subcategorized according to the syndrome clinical manifestations. Two monoclonal antibodies (mAb), TNF-α mAb and IL-6 mAb, and one polyclonal antibody IL-1β mAb were applied for immunohistochemical analysis. Results In IBD patients intensity of TNF-α and IL-1β were lower than in IBS patients or controls, while IL-6 was significantly increased comparing to the aforementioned groups. In IBS patients TNF-α was increased comparing to IBD patients or controls, while IL-6 and IL-1β were similar to controls. In IBS subgroups, TNF-α was lower in diarrhea predominant IBS patients and higher constipation predominant IBS patients. Differences among IBS subgroups regarding IL-6 and IL-1β were nonsignificant. Conclusions IL-6 seems to be the most important proinflammatory cytokine in IBD patients, while TNF-α could play a more significant role in IBS pathogenesis.
Collapse
Affiliation(s)
- Antigony Mitselou
- Department of Forensic Pathology, Medical School University of Ioannina, Ioannina, Greece
| | | | - Anna Varouktsi
- Department of Internal Medicine, Ippokratio Hospital, Thessaloniki, Greece
| | - Stamatis S Papadatos
- 3rd Department of Internal Medicine, Sotiria General Hospital, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | | | - Vasiliki Galani
- Department of Anatomy, Histology, and Embryology, Medical School University of Ioannina, Ioannina, Greece
| |
Collapse
|
9
|
Galani V, Varouktsi A, Papadatos SS, Mitselou A, Sainis I, Constantopoulos S, Dalavanga Y. The role of apoptosis defects in malignant mesothelioma pathogenesis with an impact on prognosis and treatment. Cancer Chemother Pharmacol 2019; 84:241-253. [DOI: 10.1007/s00280-019-03878-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 05/18/2019] [Indexed: 01/09/2023]
|
10
|
Zhao H, Dong N, Liu T, Zhang P, Zheng Y, Yang L, Ren X. Clinical Significance of Serum Type III Interferons in Patients with Gastric Cancer. J Interferon Cytokine Res 2019; 39:155-163. [PMID: 30672717 DOI: 10.1089/jir.2018.0119] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Type III interferon (IFN) is a newly established IFN that shows roles different from those of type I and II IFNs. However, the effect of type III IFN on the prognosis of patients with gastric cancer (GC) is controversial. This study aimed to investigate the effects of serum IFN-α, IFN-β, IFN-γ, IFN-λ1, and IFN-λ2/3 levels on the survival of patients with GC. LEGENDplex bead-based immunoassays were used to analyze the serum IFN-α, IFN-β, IFN-γ, IFN-λ1, and IFN-λ2/3 levels in patients with GC and healthy volunteers. Flow cytometry was used to test the IFN concentrations. Compared with the healthy controls, the patients with GC had significantly decreased serum IFN-α, IFN-γ, and IFN-λ1 levels, but significantly increased serum IFN-λ2/3 level. Analysis of the serum IFN concentrations and clinical parameters of the patients with GC showed significant correlations of serum IFN-α, IFN-β, IFN-γ, and IFN-λ2/3 levels with clinical stages. Serum IFN-λ1 levels significantly correlated with tumor location, histopathology, and lymph node involvement. Serum IFN-λ2/3 levels significantly correlated with lymph node involvement and distant metastasis. In addition, serum IFN-α, IFN-β, and IFN-γ levels significantly correlated with clinical stage and lymph node metastasis. Serum IFN-β and IFN-γ levels also significantly correlated with Lauren classification. Furthermore, the patients with stage IV GC exhibited significantly lower IFN-λ2/3 levels and higher IFN-α, IFN-β, and IFN-γ levels than the patients with stage I-III GC. The univariate analysis revealed that tumor sizes of >5 cm, higher clinical stage, and high IFN-λ2/3 level were significantly associated with poor prognosis. This study shows the relationship between serum IFN level and GC progression. High serum IFN-λ2/3 levels were associated with poor prognosis and could be a novel biomarker for evaluating GC progression and predicting the outcomes of nonmetastatic GC.
Collapse
Affiliation(s)
- Hua Zhao
- 1 Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,2 Department of Immunology, National Clinical Research Center for Cancer, Tianjin, China.,3 Key Laboratories of Cancer Prevention and Therapy, Tianjin, China.,4 Department of Immunology, Tianjin's Clinical Research Center for Cancer, Tianjin, China.,5 Key Laboratories of Cancer Immunology and Biotherapy, Tianjin, China
| | - Nan Dong
- 1 Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,2 Department of Immunology, National Clinical Research Center for Cancer, Tianjin, China.,3 Key Laboratories of Cancer Prevention and Therapy, Tianjin, China.,4 Department of Immunology, Tianjin's Clinical Research Center for Cancer, Tianjin, China.,5 Key Laboratories of Cancer Immunology and Biotherapy, Tianjin, China
| | - Ting Liu
- 1 Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,2 Department of Immunology, National Clinical Research Center for Cancer, Tianjin, China.,3 Key Laboratories of Cancer Prevention and Therapy, Tianjin, China.,4 Department of Immunology, Tianjin's Clinical Research Center for Cancer, Tianjin, China.,5 Key Laboratories of Cancer Immunology and Biotherapy, Tianjin, China
| | - Peng Zhang
- 1 Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,2 Department of Immunology, National Clinical Research Center for Cancer, Tianjin, China.,3 Key Laboratories of Cancer Prevention and Therapy, Tianjin, China.,4 Department of Immunology, Tianjin's Clinical Research Center for Cancer, Tianjin, China.,5 Key Laboratories of Cancer Immunology and Biotherapy, Tianjin, China
| | - Yawen Zheng
- 1 Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,2 Department of Immunology, National Clinical Research Center for Cancer, Tianjin, China.,3 Key Laboratories of Cancer Prevention and Therapy, Tianjin, China.,4 Department of Immunology, Tianjin's Clinical Research Center for Cancer, Tianjin, China.,5 Key Laboratories of Cancer Immunology and Biotherapy, Tianjin, China
| | - Lili Yang
- 1 Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,2 Department of Immunology, National Clinical Research Center for Cancer, Tianjin, China.,3 Key Laboratories of Cancer Prevention and Therapy, Tianjin, China.,4 Department of Immunology, Tianjin's Clinical Research Center for Cancer, Tianjin, China.,5 Key Laboratories of Cancer Immunology and Biotherapy, Tianjin, China
| | - Xiubao Ren
- 1 Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,2 Department of Immunology, National Clinical Research Center for Cancer, Tianjin, China.,3 Key Laboratories of Cancer Prevention and Therapy, Tianjin, China.,4 Department of Immunology, Tianjin's Clinical Research Center for Cancer, Tianjin, China.,5 Key Laboratories of Cancer Immunology and Biotherapy, Tianjin, China
| |
Collapse
|
11
|
Kazaana A, Sano E, Yoshimura S, Makita K, Hara H, Yoshino A, Ueda T. Promotion of TRAIL/Apo2L-induced apoptosis by low-dose interferon-β in human malignant melanoma cells. J Cell Physiol 2019; 234:13510-13524. [PMID: 30613977 DOI: 10.1002/jcp.28029] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 11/30/2018] [Indexed: 12/29/2022]
Abstract
Interferon β (IFN-β) is considered a signaling molecule with important therapeutic potential in cancer since IFN-β-induced gene transcription mediates antiproliferation and cell death induction. Whereas, TNF-related apoptosis inducing ligand/Apo2 ligand (TRAIL/Apo2L) has emerged as a promising anticancer agent because it induces apoptosis specifically in cancer cells. In this study, we elucidated that IFN-β augments TRAIL-induced apoptosis synergistically using five human malignant melanoma cells. All of these cells were induced apoptosis by TRAIL. Whereas, the response against IFN-β was different in amelanotic cells (A375 and CRL1579) and melanotic cells (G361, SK-MEL-28, and MeWo). The responsibility of amelanotic cells against IFN-β was higher than those of melanotic cells. The synergism of IFN-β and TRAIL were correlated with the responsibilities of the cells against IFN-β. The synergistic interaction was confirmed by a combination index based on the Chou-Talalay method. The upregulation of apoptosis in amelanotic cells was caused by very low doses of IFN-β (over 0.1 IU/ml). Both of p53-mediated intrinsic pathway and Fas-related extrinsic pathway were activated by IFN-β alone and combination with TRAIL. Further, TRAIL death receptors (DR4 and DR5) were upregulated by a low-dose IFN-β (over 0.1 IU/ml) and the expression was more promoted by the combination with TRAIL. It was clarified that the upregulation of DR5 is associated with the declination of viability.
Collapse
Affiliation(s)
- Akira Kazaana
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa-shi, Japan
| | - Emiko Sano
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa-shi, Japan
| | - Sodai Yoshimura
- Department of Neurological Surgery, Nihon University School of Medicine, Tokyo, Japan
| | - Kotaro Makita
- Department of Neurological Surgery, Nihon University School of Medicine, Tokyo, Japan
| | - Hiroyuki Hara
- Department of Functional Morphology, Nihon University School of Medicine, Tokyo, Japan
| | - Atsuo Yoshino
- Department of Neurological Surgery, Nihon University School of Medicine, Tokyo, Japan
| | - Takuya Ueda
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa-shi, Japan
| |
Collapse
|
12
|
Interferon-γ and Smac mimetics synergize to induce apoptosis of lung cancer cells in a TNFα-independent manner. Cancer Cell Int 2018; 18:84. [PMID: 29946223 PMCID: PMC6001173 DOI: 10.1186/s12935-018-0579-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 06/06/2018] [Indexed: 12/28/2022] Open
Abstract
Background The prognosis of lung cancer is very poor and hence new therapeutic strategies are urgently desired. In this study, we searched for efficacious Smac mimetic-based combination therapies with biomarkers to predict responses for non-small cell lung cancer (NSCLC). Methods NSCLC cell lines and normal human alveolar epithelial cells were treated with Smac mimetics plus IFNγ or other agonists and cell viabilities were assessed by MTS assay, cell counting, flow cytometry and cell colony assay. Western blot analysis was performed to assess the cleavage (activation) of caspases and expression of signaling molecules. Caspase activity was determined to verify caspase activation. The pathways involved in NSCLC cell death were investigated using specific inhibitors. Results We found that IFNγ could cooperate with various Smac mimetics to trigger a profound apoptosis in a number of NSCLC cell lines that are competent for IFNγ signaling (i.e. expressing IFNγ receptor-1 and STAT1) but have low expression levels of inhibitor of apoptosis proteins survivin and livin without harming normal human lung epithelial cells. IFNγ co-treatment with a novel class dimeric Smac mimetic AZD5582 eradicated NSCLC cell colony formation. Unlike IFNγ, IFNα, IFNλ, TNFα, or TRAIL alone or plus AZD5582 had minor effects on NSCLC cell viability. IFNγ/AZD5582-induced cell death in NSCLC cells was independent of TNFα autocrine but relied on apoptosis mediated by JAK kinase, caspase 8 and RIPK1 pathways. Conclusion Our results indicate that IFNγ and Smac mimetics can synergize to induce apoptosis of NSCLC cells and suggest that IFNγ and Smac mimetic regimen may be a novel and efficacious apoptosis targeted therapy with biomarkers to predict responses for NSCLC cells.
Collapse
|
13
|
Bello-Rivero I, Garcia-Vega Y, Duncan-Roberts Y, Vazquez-Blomquistc D, Santana-Milian H, Besada-Perez V, Rios-Cabrera M. HeberFERON, a new formulation of IFNs with improved pharmacodynamics: Perspective for cancer treatment. Semin Oncol 2018; 45:27-33. [PMID: 30318081 DOI: 10.1053/j.seminoncol.2018.04.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 04/20/2018] [Indexed: 12/22/2022]
Abstract
The rational combination of recombinant IFN-α2b and IFN-γ resulted in a new formulation of interferons (HeberFERON) with improved pharmacodynamics. In basal cell carcinomas HeberFERON produces a more rapid antitumor effect and results in a larger number of complete responses. In patients with glioblastoma multiforme, the administration of HeberFERON after surgery and radiotherapy results in an estimated overall survival of 19 months. Patients with stage III or IV renal cell carcinoma also appear to benefit from the intravenous administration of HeberFERON, with prolongation of survival and good quality of live. HeberFERON offers a promising alternative formulation of interferons for the treatment of cancer with a very favorable safety profile.
Collapse
Affiliation(s)
- Iraldo Bello-Rivero
- Clinical Research Department, Center for Genetic Engineering and Biotechnology, Havana, Cuba.
| | | | | | | | - Hector Santana-Milian
- Formulation Department, Center for Genetic Engineering and Biotechnology, Havana, Cuba
| | - Vladimir Besada-Perez
- Proteomic Department, Center for Genetic Engineering and Biotechnology, Havana, Cuba
| | | |
Collapse
|
14
|
Liu CP, Zhang X, Tan QL, Xu WX, Zhou CY, Luo M, Li X, Huang RY, Zeng X. NF-κB pathways are involved in M1 polarization of RAW 264.7 macrophage by polyporus polysaccharide in the tumor microenvironment. PLoS One 2017; 12:e0188317. [PMID: 29155869 PMCID: PMC5695768 DOI: 10.1371/journal.pone.0188317] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 11/03/2017] [Indexed: 12/30/2022] Open
Abstract
Bladder cancer is one of the most malignant tumors closely associated with macrophages. Polyporus polysaccharide (PPS) has shown excellent efficacy in treating bladder cancer with minimal side effects. However, the molecular mechanisms underlying the effects of PPS in inhibiting bladder cancer remain unclear. In this study, we used macrophages cultured alone or with T24 human bladder cancer cell culture supernatant as study models. We found that PPS enhanced the activities of IFN-γ-stimulated RAW 264.7 macrophages, as shown by the release of inducible nitric oxide synthase (INOS), secretion of tumor necrosis factor (TNF)-α and interleukin (IL)-6, phagocytosis activity, as well as expression of M1 phenotype indicators, such as CD40, CD284 and CD86. PPS acted upstream in activation cascade of nuclear factor (NF)-κB signaling pathways by interfering with IκB phosphorylation. In addition, PPS regulated NF-κB (P65) signaling by interfering with Toll-like receptor (TLR)-4, INOS and cyclooxygenase (COX)-2. Our results indicate that PPS activates macrophages through TLR4/NF-κB signaling pathways.
Collapse
Affiliation(s)
- Chun-Ping Liu
- Department of Integrated Chinese medicine immunization and Section Rheumatology Research, The Second Affiliated Hospital, Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China
- Discipline of Integrated Chinese and Western Medicine in Guangzhou University of Chinese medicine, Guangzhou, China
| | - Xian Zhang
- Department of Integrated Chinese medicine immunization and Section Rheumatology Research, The Second Affiliated Hospital, Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China
| | - Qing-Long Tan
- Department of Integrated Chinese medicine immunization and Section Rheumatology Research, The Second Affiliated Hospital, Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China
| | - Wen-Xing Xu
- Department of Integrated Chinese medicine immunization and Section Rheumatology Research, The Second Affiliated Hospital, Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China
| | - Chang-Yuan Zhou
- Department of Integrated Chinese medicine immunization and Section Rheumatology Research, The Second Affiliated Hospital, Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China
| | - Min Luo
- Department of Integrated Chinese medicine immunization and Section Rheumatology Research, The Second Affiliated Hospital, Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China
| | - Xiong Li
- Department of Integrated Chinese medicine immunization and Section Rheumatology Research, The Second Affiliated Hospital, Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China
| | - Run-Yue Huang
- Department of Integrated Chinese medicine immunization and Section Rheumatology Research, The Second Affiliated Hospital, Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China
- Discipline of Integrated Chinese and Western Medicine in Guangzhou University of Chinese medicine, Guangzhou, China
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangzhou, China
- * E-mail: (XZ); (RYH)
| | - Xing Zeng
- Department of Integrated Chinese medicine immunization and Section Rheumatology Research, The Second Affiliated Hospital, Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China
- Discipline of Integrated Chinese and Western Medicine in Guangzhou University of Chinese medicine, Guangzhou, China
- * E-mail: (XZ); (RYH)
| |
Collapse
|
15
|
Mechanism of splenic cell death and host mortality in a Plasmodium yoelii malaria model. Sci Rep 2017; 7:10438. [PMID: 28874800 PMCID: PMC5585408 DOI: 10.1038/s41598-017-10776-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Accepted: 08/14/2017] [Indexed: 12/14/2022] Open
Abstract
Malaria is a fatal disease that displays a spectrum of symptoms and severity, which are determined by complex host-parasite interactions. It has been difficult to study the effects of parasite strains on disease severity in human infections, but the mechanisms leading to specific disease phenotypes can be investigated using strains of rodent malaria parasites that cause different disease symptoms in inbred mice. Using a unique mouse malaria model, here we investigated the mechanisms of splenic cell death and their relationship to control of parasitemia and host mortality. C57BL/6 mice infected with Plasmodium yoelii nigeriensis N67C display high levels of pro-inflammatory cytokines and chemokines (IL-6, IFN-γ, TNF-α, CXCL1, and CCL2) and extensive splenic damage with dramatic reduction of splenic cell populations. These disease phenotypes were rescued in RAG2−/−, IFN-γ−/−, or T cell depleted mice, suggesting IFN-γ and T cell mediated disease mechanisms. Additionally, apoptosis was one of the major pathways involved in splenic cell death, which coincides with the peaks of pro-inflammatory cytokines. Our results demonstrate the critical roles of T cells and IFN-γ in mediating splenic cell apoptosis, parasitemia control, and host lethality and thus may provide important insights for preventing/reducing morbidity associated with severe malaria in humans.
Collapse
|
16
|
Galani V, Papadatos SS, Alexiou G, Galani A, Kyritsis AP. In Vitro and In Vivo Preclinical Effects of Type I IFNs on Gliomas. J Interferon Cytokine Res 2017; 37:139-146. [PMID: 28387596 DOI: 10.1089/jir.2016.0094] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
The interferons (IFNs) are a family of cytokines with diverse cellular actions such as control of cell proliferation and regulation of immune responses; therefore, they have been extensively studied as antitumor agents for a variety of malignancies, including gliomas. Type I IFNs exert their antitumor effects either directly, by targeting the tumor cells or the tumor stem cells, or indirectly, by regulating the anticancer activities of the immune system. More specifically, IFN-beta and IFN-alpha exhibit antiproliferative effects by p53 induction, CD8+ T-lymphocyte and macrophage activation, chemokine secretion, and miR-21 downregulation. In vitro and in vivo studies provide evidence that immunotherapy could have a role in glioma treatment, especially when first-line therapeutic interventions fail to produce durable responses. These effects are more obvious when combining IFN-beta with classical antitumor therapies such as temozolamide, an oral chemotherapeutic, for both newly diagnosed and recurrent gliomas. However, further clinical studies are needed to determine whether IFNs will have a definite place in the management of gliomas.
Collapse
Affiliation(s)
- Vasiliki Galani
- 1 Department of Anatomy-Histology-Embryology, Faculty of Medicine, University of Ioannina , Ioannina, Greece
| | - Stamatis S Papadatos
- 2 3rd Department of Internal Medicine, Athens School of Medicine, National and Kapodistrian University of Athens , Sotiria General Hospital, Athens, Greece
| | - George Alexiou
- 3 Neurosurgical Institute, University of Ioannina , Ioannina, Greece
| | - Angeliki Galani
- 4 Department of Environmental and Natural Resources Management, University of Patras , Patra, Greece
| | - Athanasios P Kyritsis
- 3 Neurosurgical Institute, University of Ioannina , Ioannina, Greece .,5 Department of Neurology, Faculty of Medicine, University of Ioannina , Ioannina, Greece
| |
Collapse
|