1
|
Gazzeri S, Zubchuk N, Montaudon E, Nemati F, Huot-Marchand S, Berardi G, Pucciarelli A, Dib Y, Nerini D, Oddou C, Pezet M, David-Boudet L, Ardin C, de Fraipont F, Maraver A, Girard N, Decaudin D, Toffart AC, Eymin B. PPP3CB overexpression mediates EGFR TKI resistance in lung tumors via calcineurin/MEK/ERK signaling. Life Sci Alliance 2024; 7:e202402873. [PMID: 39353739 PMCID: PMC11447527 DOI: 10.26508/lsa.202402873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 09/23/2024] [Accepted: 09/24/2024] [Indexed: 10/04/2024] Open
Abstract
Despite initial high response rates to first-line EGFR TKI, all non-small-cell lung cancer (NSCLC) with EGFR-activating mutation will ultimately develop resistance to treatment. Identification of resistance mechanisms is critical to adapt treatment and improve patient outcomes. Here, we show that a PPP3CB transcript that encodes full-length catalytic subunit 2B of calcineurin accumulates in EGFR-mutant NSCLC cells with acquired resistance against different EGFR TKIs and in post-progression biopsies of NSCLC patients treated with EGFR TKIs. Neutralization of PPP3CB by siRNA or inactivation of calcineurin by cyclosporin A induces apoptosis in resistant cells treated with EGFR TKIs. Mechanistically, EGFR TKIs increase the cytosolic level of calcium and trigger activation of a calcineurin/MEK/ERK pathway that prevents apoptosis. Combining EGFR, calcineurin, and MEK inhibitors overcomes resistance to EGFR TKI in both in vitro and in vivo models. Our results identify PPP3CB overexpression as a new mechanism of acquired resistance to EGFR TKIs, and provide a promising therapeutic approach for NSCLC patients that progress under TKI treatment.
Collapse
Affiliation(s)
- Sylvie Gazzeri
- University Grenoble Alpes, Inserm U1209, CNRS UMR 5309, Team RNA Splicing, Cell Signaling and Response to Therapy, Institute for Advanced Biosciences, Grenoble, France
| | - Nadiia Zubchuk
- University Grenoble Alpes, Inserm U1209, CNRS UMR 5309, Team RNA Splicing, Cell Signaling and Response to Therapy, Institute for Advanced Biosciences, Grenoble, France
| | - Elodie Montaudon
- Laboratory of Preclinical Investigation, Translational Research Department, Institut Curie, PSL Research University, Paris, France
| | - Fariba Nemati
- Laboratory of Preclinical Investigation, Translational Research Department, Institut Curie, PSL Research University, Paris, France
| | - Sarah Huot-Marchand
- University Grenoble Alpes, Inserm U1209, CNRS UMR 5309, Team RNA Splicing, Cell Signaling and Response to Therapy, Institute for Advanced Biosciences, Grenoble, France
| | - Giulia Berardi
- Department of Pneumology and Physiology, Grenoble-Alpes University Hospital, Grenoble, France
| | - Amelie Pucciarelli
- University Grenoble Alpes, Inserm U1209, CNRS UMR 5309, Team RNA Splicing, Cell Signaling and Response to Therapy, Institute for Advanced Biosciences, Grenoble, France
| | - Yassir Dib
- University Grenoble Alpes, Inserm U1209, CNRS UMR 5309, Team RNA Splicing, Cell Signaling and Response to Therapy, Institute for Advanced Biosciences, Grenoble, France
| | - Dylan Nerini
- University Grenoble Alpes, Inserm U1209, CNRS UMR 5309, Team RNA Splicing, Cell Signaling and Response to Therapy, Institute for Advanced Biosciences, Grenoble, France
| | - Christiane Oddou
- University Grenoble Alpes, Inserm U1209, CNRS UMR 5309, Team Epigenetics, Immunity, Metabolism, Cell Signaling and Cancer, Institute for Advanced Biosciences, Grenoble, France
| | - Mylène Pezet
- University Grenoble Alpes, Inserm U1209, CNRS UMR 5309, Platform MicroCell, Institute for Advanced Biosciences, Grenoble, France
| | - Laurence David-Boudet
- Department of Cytology and Pathology, Grenoble-Alpes University Hospital, Grenoble, France
| | - Camille Ardin
- University Grenoble Alpes, Inserm U1209, CNRS UMR 5309, Team RNA Splicing, Cell Signaling and Response to Therapy, Institute for Advanced Biosciences, Grenoble, France
- Department of Pneumology and Physiology, Grenoble-Alpes University Hospital, Grenoble, France
| | - Florence de Fraipont
- University Grenoble Alpes, Inserm U1209, CNRS UMR 5309, Team RNA Splicing, Cell Signaling and Response to Therapy, Institute for Advanced Biosciences, Grenoble, France
- Medical Unit of Molecular Genetic (Hereditary Diseases and Oncology), Grenoble-Alpes University Hospital, Grenoble, France
| | - Antonio Maraver
- Institut de Recherche en Cancérologie de Montpellier, INSERM U1194-ICM-Université de Montpellier, Montpellier, France
| | - Nicolas Girard
- Institut du Thorax Curie-Montsouris, Institut Curie, Paris, France
| | - Didier Decaudin
- Laboratory of Preclinical Investigation, Translational Research Department, Institut Curie, PSL Research University, Paris, France
- Department of Medical Oncology, Institut Curie, Paris, France
| | - Anne-Claire Toffart
- University Grenoble Alpes, Inserm U1209, CNRS UMR 5309, Team RNA Splicing, Cell Signaling and Response to Therapy, Institute for Advanced Biosciences, Grenoble, France
- Department of Pneumology and Physiology, Grenoble-Alpes University Hospital, Grenoble, France
| | - Beatrice Eymin
- University Grenoble Alpes, Inserm U1209, CNRS UMR 5309, Team RNA Splicing, Cell Signaling and Response to Therapy, Institute for Advanced Biosciences, Grenoble, France
| |
Collapse
|
2
|
Jiang H, Liu M, Deng Y, Zhang C, Dai L, Zhu B, Ou Y, Zhu Y, Hu C, Yang L, Li J, Bai Y, Yang D. Identification of prostate cancer bone metastasis related genes and potential therapy targets by bioinformatics and in vitro experiments. J Cell Mol Med 2024; 28:e18511. [PMID: 39098992 PMCID: PMC11298316 DOI: 10.1111/jcmm.18511] [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: 04/23/2024] [Revised: 06/16/2024] [Accepted: 06/18/2024] [Indexed: 08/06/2024] Open
Abstract
The aetiology of bone metastasis in prostate cancer (PCa) remains unclear. This study aims to identify hub genes involved in this process. We utilized machine learning, GO, KEGG, GSEA, Single-cell analysis, ROC methods to identify hub genes for bone metastasis in PCa using the TCGA and GEO databases. Potential drugs targeting these genes were identified. We validated these results using 16 specimens from patients with PCa and analysed the relationship between the hub genes and clinical features. The impact of APOC1 on PCa was assessed through in vitro experiments. Seven hub genes with AUC values of 0.727-0.926 were identified. APOC1, CFH, NUSAP1 and LGALS1 were highly expressed in bone metastasis tissues, while NR4A2, ADRB2 and ZNF331 exhibited an opposite trend. Immunohistochemistry further confirmed these results. The oxidative phosphorylation pathway was significantly enriched by the identified genes. Aflatoxin B1, benzo(a)pyrene, cyclosporine were identified as potential drugs. APOC1 expression was correlated with clinical features of PCa metastasis. Silencing APOC1 significantly inhibited PCa cell proliferation, clonality, and migration in vitro. This study identified 7 hub genes that potentially facilitate bone metastasis in PCa through mitochondrial metabolic reprogramming. APOC1 emerged as a promising therapeutic target and prognostic marker for PCa with bone metastasis.
Collapse
Affiliation(s)
- Haiyang Jiang
- Department of Urology IThe Third Affiliated Hospital of Kunming Medical University (Peking University Cancer Hospital Yunnan, Yunnan Cancer Hospital, Cancer Center of Yunnan Province)KunmingYunnanChina
- Department of Urology IIThe second Affiliated Hospital of Kunming Medical UniversityKunmingYunnanChina
| | - Mingcheng Liu
- Department of Human Cell Biology and Genetics, School of MedicineSouthern University of Science and TechnologyShenzhenChina
| | - Yingfei Deng
- Pathology‐DepartmentThe Third Affiliated Hospital of Kunming Medical University (Peking University Cancer Hospital Yunnan, Yunnan Cancer Hospital, Cancer Center of Yunnan Province)KunmingYunnanChina
| | - Chongjian Zhang
- Department of Urology IThe Third Affiliated Hospital of Kunming Medical University (Peking University Cancer Hospital Yunnan, Yunnan Cancer Hospital, Cancer Center of Yunnan Province)KunmingYunnanChina
| | - Longguo Dai
- Department of Urology IThe Third Affiliated Hospital of Kunming Medical University (Peking University Cancer Hospital Yunnan, Yunnan Cancer Hospital, Cancer Center of Yunnan Province)KunmingYunnanChina
| | - Bingyu Zhu
- Department of Urology IThe Third Affiliated Hospital of Kunming Medical University (Peking University Cancer Hospital Yunnan, Yunnan Cancer Hospital, Cancer Center of Yunnan Province)KunmingYunnanChina
| | - Yitian Ou
- Department of Urology IIThe second Affiliated Hospital of Kunming Medical UniversityKunmingYunnanChina
| | - Yong Zhu
- Department of Urology IThe Third Affiliated Hospital of Kunming Medical University (Peking University Cancer Hospital Yunnan, Yunnan Cancer Hospital, Cancer Center of Yunnan Province)KunmingYunnanChina
| | - Chen Hu
- Department of Urology IThe Third Affiliated Hospital of Kunming Medical University (Peking University Cancer Hospital Yunnan, Yunnan Cancer Hospital, Cancer Center of Yunnan Province)KunmingYunnanChina
| | - Libo Yang
- Department of Urology IThe Third Affiliated Hospital of Kunming Medical University (Peking University Cancer Hospital Yunnan, Yunnan Cancer Hospital, Cancer Center of Yunnan Province)KunmingYunnanChina
| | - Jun Li
- Department of Urology IThe Third Affiliated Hospital of Kunming Medical University (Peking University Cancer Hospital Yunnan, Yunnan Cancer Hospital, Cancer Center of Yunnan Province)KunmingYunnanChina
| | - Yu Bai
- Department of Urology IThe Third Affiliated Hospital of Kunming Medical University (Peking University Cancer Hospital Yunnan, Yunnan Cancer Hospital, Cancer Center of Yunnan Province)KunmingYunnanChina
| | - Delin Yang
- Department of Urology IIThe second Affiliated Hospital of Kunming Medical UniversityKunmingYunnanChina
| |
Collapse
|
3
|
Chen H, Fang S, Zhu X, Liu H. Cancer-associated fibroblasts and prostate cancer stem cells: crosstalk mechanisms and implications for disease progression. Front Cell Dev Biol 2024; 12:1412337. [PMID: 39092186 PMCID: PMC11291335 DOI: 10.3389/fcell.2024.1412337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Accepted: 07/05/2024] [Indexed: 08/04/2024] Open
Abstract
The functional heterogeneity and ecological niche of prostate cancer stem cells (PCSCs), which are major drivers of prostate cancer development and treatment resistance, have attracted considerable research attention. Cancer-associated fibroblasts (CAFs), which are crucial components of the tumor microenvironment (TME), substantially affect PCSC stemness. Additionally, CAFs promote PCSC growth and survival by releasing signaling molecules and modifying the surrounding environment. Conversely, PCSCs may affect the characteristics and behavior of CAFs by producing various molecules. This crosstalk mechanism is potentially crucial for prostate cancer progression and the development of treatment resistance. Using organoids to model the TME enables an in-depth study of CAF-PCSC interactions, providing a valuable preclinical tool to accurately evaluate potential target genes and design novel treatment strategies for prostate cancer. The objective of this review is to discuss the current research on the multilevel and multitarget regulatory mechanisms underlying CAF-PCSC interactions and crosstalk, aiming to inform therapeutic approaches that address challenges in prostate cancer treatment.
Collapse
Affiliation(s)
| | | | | | - Hao Liu
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
4
|
Sato Y, Habara M, Hanaki S, Sharif J, Tomiyasu H, Miki Y, Shimada M. Calcineurin/NFATc1 pathway represses cellular cytotoxicity by modulating histone H3 expression. Sci Rep 2024; 14:14732. [PMID: 38926604 PMCID: PMC11208570 DOI: 10.1038/s41598-024-65769-9] [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: 12/31/2023] [Accepted: 06/24/2024] [Indexed: 06/28/2024] Open
Abstract
Excess amounts of histones in the cell induce mitotic chromosome loss and genomic instability, and are therefore detrimental to cell survival. In yeast, excess histones are degraded by the proteasome mediated via the DNA damage response factor Rad53. Histone expression, therefore, is tightly regulated at the protein level. Our understanding of the transcriptional regulation of histone genes is far from complete. In this study, we found that calcineurin inhibitor treatment increased histone protein levels, and that the transcription factor NFATc1 (nuclear factor of activated T cells 1) repressed histone transcription and acts downstream of the calcineurin. We further revealed that NFATc1 binds to the promoter regions of many histone genes and that histone transcription is downregulated in a manner dependent on intracellular calcium levels. Indeed, overexpression of histone H3 markedly inhibited cell proliferation. Taken together, these findings suggest that NFATc1 prevents the detrimental effects of histone H3 accumulation by inhibiting expression of histone at the transcriptional level.
Collapse
Affiliation(s)
- Yuki Sato
- Department of Veterinary Biochemistry, Joint Faculty of Veterinary Medicine, Yamaguchi University, 1677-1 Yoshida, Yamaguchi, 753-8511, Japan
| | - Makoto Habara
- Department of Veterinary Biochemistry, Joint Faculty of Veterinary Medicine, Yamaguchi University, 1677-1 Yoshida, Yamaguchi, 753-8511, Japan
| | - Shunsuke Hanaki
- Department of Veterinary Biochemistry, Joint Faculty of Veterinary Medicine, Yamaguchi University, 1677-1 Yoshida, Yamaguchi, 753-8511, Japan
| | - Jafar Sharif
- Developmental Genetics Group, Center for Integrative Medical Sciences (IMS), RIKEN, 1-7-22 Suehiro, Tsurumi-ku, Yokohama, Kanagawa, 230-0045, Japan
| | - Haruki Tomiyasu
- Department of Veterinary Biochemistry, Joint Faculty of Veterinary Medicine, Yamaguchi University, 1677-1 Yoshida, Yamaguchi, 753-8511, Japan
| | - Yosei Miki
- Department of Veterinary Biochemistry, Joint Faculty of Veterinary Medicine, Yamaguchi University, 1677-1 Yoshida, Yamaguchi, 753-8511, Japan
| | - Midori Shimada
- Department of Veterinary Biochemistry, Joint Faculty of Veterinary Medicine, Yamaguchi University, 1677-1 Yoshida, Yamaguchi, 753-8511, Japan.
- Department of Molecular Biology, Graduate School of Medicine, Nagoya University, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan.
| |
Collapse
|
5
|
Hasani S, Fathabadi F, Saeidi S, Mohajernoei P, Hesari Z. The role of NFATc1 in the progression and metastasis of prostate cancer: A review on the molecular mechanisms and signaling pathways. Cell Biol Int 2023; 47:1895-1904. [PMID: 37814550 DOI: 10.1002/cbin.12094] [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: 04/17/2023] [Revised: 08/27/2023] [Accepted: 09/26/2023] [Indexed: 10/11/2023]
Abstract
A common type of cancer among men is the prostate cancer that kills many people every year. The multistage of this disease and the involvement of the vital organs of the body have reduced the life span and quality of life of the people involved and turned the treatment process into a complex one. NFATc1 biomarker contributes significantly in the diagnosis and treatment of this disease by increasing its expression in prostate cancer and helping the proliferation, differentiation, and invasion of cancer cells through different signaling pathways. NFATc1 is also able to target the metabolism of cancer cells by inserting specific oncogene molecules such as c-myc that it causes cell growth and proliferation. Bone is a common tissue where prostate cancer cells metastasize. In this regard, the activity of NFATc1, through the regulation of different signaling cascades, including the RANKL/RANK signaling pathway, in turn, increases the activity of osteoclasts, and as a result, bone tissue is gradually ruined. Using Silibinin as a medicinal plant extract can inhibit the activity of osteoclasts related to prostate cancer by targeting NFATc. Undoubtedly, NFATc1 is one of the effective oncogenes related to prostate cancer, which has the potential to put this cancer on the path of progression and metastasis. In this review, we will highlight the role of NFATc1 in the progression and metastasis of prostate cancer. Furthermore, we will summarize signaling pathways and molecular mechanism, through which NFATc1 regulates the process of prostate cancer.
Collapse
Affiliation(s)
- Samaneh Hasani
- Department of Nursing, Faculty of Medical Sciences, Khalkhal University of Medical Sciences, Khalkhal, Iran
| | - Farshid Fathabadi
- Laboratory Sciences Research Center, Golestan University of Medical Sciences, Department of Laboratory Sciences, Faculty of Paramedicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Saman Saeidi
- Laboratory Sciences Research Center, Golestan University of Medical Sciences, Department of Laboratory Sciences, Faculty of Paramedicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Pouya Mohajernoei
- Department of Medicine and Surgery, Università degli Studi di Padova, Padua, Italy
| | - Zahra Hesari
- Laboratory Sciences Research Center, Golestan University of Medical Sciences, Department of Laboratory Sciences, Faculty of Paramedicine, Golestan University of Medical Sciences, Gorgan, Iran
| |
Collapse
|
6
|
Gul S, Pang J, Yuan H, Chen Y, Yu Q, Wang H, Tang W. Stemness signature and targeted therapeutic drugs identification for Triple Negative Breast Cancer. Sci Data 2023; 10:815. [PMID: 37985782 PMCID: PMC10662149 DOI: 10.1038/s41597-023-02709-8] [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: 06/13/2023] [Accepted: 10/31/2023] [Indexed: 11/22/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is the most aggressive subtype of breast cancer and carries the worst prognosis, characterized by the lack of progesterone, estrogen, and HER2 gene expression. This study aimed to analyze cancer stemness-related gene signature to determine patients' risk stratification and prognosis feature with TNBC. Here one-class logistic regression (OCLR) algorithm was applied to compute the stemness index of TNBC patients. Cox and LASSO regression analysis was performed on stemness-index related genes to establish 16 genes-based prognostic signature, and their predictive performance was verified in TCGA and METABERIC merged data cohort. We diagnosed the expression level of prognostic genes signature in the tumor immune microenvironment, analyzed the TNBC scRNA-seq GSE176078 dataset, and further validated the expression level of prognostic genes using the HPA database. Finally, the small molecular compounds targeted at the anti-tumor effect of predictive genes were screened by molecular docking; this novel stemness-based prognostic genes signature study could facilitate the prognosis of patients with TNBC and thus provide a feasible therapeutic target for TNBC.
Collapse
Affiliation(s)
- Samina Gul
- Laboratory of Molecular Genetics of Aging & Tumor, Medical School, Kunming University of Science and Technology, 727 jingming south road, Kunming city, Yunnan province, 650500, China
| | - Jianyu Pang
- Laboratory of Molecular Genetics of Aging & Tumor, Medical School, Kunming University of Science and Technology, 727 jingming south road, Kunming city, Yunnan province, 650500, China
| | - Hongjun Yuan
- Laboratory of Molecular Genetics of Aging & Tumor, Medical School, Kunming University of Science and Technology, 727 jingming south road, Kunming city, Yunnan province, 650500, China
| | - Yongzhi Chen
- Laboratory of Molecular Genetics of Aging & Tumor, Medical School, Kunming University of Science and Technology, 727 jingming south road, Kunming city, Yunnan province, 650500, China
| | - Qian Yu
- Laboratory of Molecular Genetics of Aging & Tumor, Medical School, Kunming University of Science and Technology, 727 jingming south road, Kunming city, Yunnan province, 650500, China
| | - Hui Wang
- Laboratory of Molecular Genetics of Aging & Tumor, Medical School, Kunming University of Science and Technology, 727 jingming south road, Kunming city, Yunnan province, 650500, China
| | - Wenru Tang
- Laboratory of Molecular Genetics of Aging & Tumor, Medical School, Kunming University of Science and Technology, 727 jingming south road, Kunming city, Yunnan province, 650500, China.
| |
Collapse
|
7
|
Hanusz K, Domański P, Strojec K, Zapała P, Zapała Ł, Radziszewski P. Prostate Cancer in Transplant Receivers-A Narrative Review on Oncological Outcomes. Biomedicines 2023; 11:2941. [PMID: 38001942 PMCID: PMC10669184 DOI: 10.3390/biomedicines11112941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 10/19/2023] [Accepted: 10/27/2023] [Indexed: 11/26/2023] Open
Abstract
Prostate cancer (PCa) is a low tumor mutational burden (TMB) cancer with a poor response to immunotherapy. Nonetheless, immunotherapy can be useful, especially in metastatic castration-resistant PCa (mCRPC). Increased cytotoxic T lymphocytes (CTLs) density is correlated with a shorter overall survival (OS), an early biochemical relapse, and a generally poor PCa prognosis. An increased number of CCR4+ regulatory T cells (CCR4 + Tregs) relates to a higher Gleason score or earlier progression. The same therapeutic options are available for renal transplant recipients (RTRs) as for the population, with a comparable functional and oncological outcome. Radical retropubic prostatectomy (RRP) is the most common method of radical treatment in RTRs. Brachytherapy and robot-assisted radical prostatectomy (RARP) seem to be promising therapies. Further studies are needed to assess the need for prostatectomy in low-risk patients before transplantation. The rate of adverse pathological features in RTRs does not seem to differ from those observed in the non-transplant population and the achieved cancer control seems comparable. The association between PCa and transplantation is not entirely clear. Some researchers indicate a possible association between a more frequent occurrence of PCa and a worse prognosis in advanced or metastatic PCa. However, others claim that the risk and survival prognosis is comparable to the non-transplant population.
Collapse
Affiliation(s)
- Karolina Hanusz
- Department of General, Oncological and Functional Urology, Medical University of Warsaw, Poland Lindleya 4, 02-005 Warsaw, Poland
| | - Piotr Domański
- Department of General, Oncological and Functional Urology, Medical University of Warsaw, Poland Lindleya 4, 02-005 Warsaw, Poland
| | - Kacper Strojec
- Department of General, Oncological and Functional Urology, Medical University of Warsaw, Poland Lindleya 4, 02-005 Warsaw, Poland
| | - Piotr Zapała
- Department of General, Oncological and Functional Urology, Medical University of Warsaw, Poland Lindleya 4, 02-005 Warsaw, Poland
| | - Łukasz Zapała
- Department of General, Oncological and Functional Urology, Medical University of Warsaw, Poland Lindleya 4, 02-005 Warsaw, Poland
| | - Piotr Radziszewski
- Department of General, Oncological and Functional Urology, Medical University of Warsaw, Poland Lindleya 4, 02-005 Warsaw, Poland
| |
Collapse
|
8
|
Ren F, Zhu K, Wang Y, Zhou F, Pang S, Chen L. Proliferation, apoptosis and invasion of human lung cancer cells are associated with NFATc1. Exp Ther Med 2022; 25:49. [PMID: 36588811 PMCID: PMC9780516 DOI: 10.3892/etm.2022.11748] [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: 03/18/2022] [Accepted: 08/31/2022] [Indexed: 12/12/2022] Open
Abstract
The expression of nuclear factor of activated T cells c1 (NFATc1) is closely associated with the progression of numerous types of cancer. When NFATc1 expression becomes dysregulated in some types of cancer, this alteration can promote malignant transformation and thereby progression of cancer. NFATc1 expression has been demonstrated to be upregulated in lung cancer cells. This suggests that knockdown of NFATc1 in lung cancer cells may be a therapeutic marker for the treatment of cancer. In the present study, the effects of NFATc1 on the proliferation, apoptosis, invasion and migration of NCI-H1299 and A549 lung cancer cell lines were explored. Lentivirus infection was used to establish a cell model of NFATc1 knockdown in A549 and NCI-H1299 lung cancer cells. Reverse transcription-quantitative PCR was subsequently performed to detect NFATc1 expression in these human lung cancer cells. MTT, wound healing, colony formation and Transwell invasion assays, and flow cytometry were then performed to measure the proliferation, invasion, apoptosis and cell cycle of the cells. Finally, western blot analysis was performed to investigate the mechanism underlying the involvement of NFATc1 in these processes. NFATc1 knockdown was found to significantly inhibit the proliferation, clone formation, migration and invasion of the cells. Furthermore, the cell cycle was arrested at the G1 phase and the expression levels of the target proteins located downstream in the signaling pathway, namely CDK4, c-Myc, ERK, p38 and N-cadherin, were decreased. Following NFATc1 knockdown, the percentages of apoptotic cells were increased, and the expression levels of Bax, cleaved caspase-3 and E-cadherin were also increased. Taken together, the results of the present study suggested that NFATc1 serves an oncogenic role in lung cancer. In terms of the underlying mechanism, NFATc1 promoted the proliferation of lung cancer cells by inhibiting the MAPK and epithelial-to-mesenchymal transition signaling pathways, suggesting that NFATc1 may be a novel target for therapeutic intervention for the treatment of lung cancer.
Collapse
Affiliation(s)
- Fenghai Ren
- Department of Thoracic Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang 150081, P.R. China
| | - Kaibin Zhu
- Department of Thoracic Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang 150081, P.R. China
| | - Yanbo Wang
- Department of Thoracic Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang 150081, P.R. China
| | - Fucheng Zhou
- Department of Thoracic Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang 150081, P.R. China
| | - Sainan Pang
- Department of Thoracic Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang 150081, P.R. China
| | - Lantao Chen
- Department of Thoracic Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang 150081, P.R. China,Correspondence to: Dr Lantao Chen, Department of Thoracic Surgery, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, Heilongjiang 150081, P.R. China
| |
Collapse
|
9
|
Kushwaha PP, Verma S, Kumar S, Gupta S. Role of prostate cancer stem-like cells in the development of antiandrogen resistance. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2022; 5:459-471. [PMID: 35800367 PMCID: PMC9255247 DOI: 10.20517/cdr.2022.07] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 02/16/2022] [Accepted: 03/24/2022] [Indexed: 12/22/2022]
Abstract
Androgen deprivation therapy (ADT) is the standard of care treatment for advance stage prostate cancer. Treatment with ADT develops resistance in multiple ways leading to the development of castration-resistant prostate cancer (CRPC). Present research establishes that prostate cancer stem-like cells (CSCs) play a central role in the development of treatment resistance followed by disease progression. Prostate CSCs are capable of self-renewal, differentiation, and regenerating tumor heterogeneity. The stemness properties in prostate CSCs arise due to various factors such as androgen receptor mutation and variants, epigenetic and genetic modifications leading to alteration in the tumor microenvironment, changes in ATP-binding cassette (ABC) transporters, and adaptations in molecular signaling pathways. ADT reprograms prostate tumor cellular machinery leading to the expression of various stem cell markers such as Aldehyde Dehydrogenase 1 Family Member A1 (ALDH1A1), Prominin 1 (PROM1/CD133), Indian blood group (CD44), SRY-Box Transcription Factor 2 (Sox2), POU Class 5 Homeobox 1(POU5F1/Oct4), Nanog and ABC transporters. These markers indicate enhanced self-renewal and stemness stimulating CRPC evolution, metastatic colonization, and resistance to antiandrogens. In this review, we discuss the role of ADT in prostate CSCs differentiation and acquisition of CRPC, their isolation, identification and characterization, as well as the factors and pathways contributing to CSCs expansion and therapeutic opportunities.
Collapse
Affiliation(s)
- Prem Prakash Kushwaha
- Department of Urology, Case Western Reserve University, Cleveland, OH 44106, USA.,The Urology Institute, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
| | - Shiv Verma
- Department of Urology, Case Western Reserve University, Cleveland, OH 44106, USA.,The Urology Institute, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
| | - Shashank Kumar
- Molecular Signaling and Drug Discovery Laboratory, Department of Biochemistry, Central University of Punjab, Bathinda 151401, India
| | - Sanjay Gupta
- Department of Urology, Case Western Reserve University, Cleveland, OH 44106, USA.,The Urology Institute, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA.,Department of Pathology, Case Western Reserve University, Cleveland, OH 44106, USA.,Department of Pharmacology, Case Western Reserve University, Cleveland, OH 44106, USA.,Department of Nutrition, Case Western Reserve University, Cleveland, OH 44106, USA.,Divison of General Medical Sciences, Case Comprehensive Cancer Center, Cleveland, OH 44106, USA
| |
Collapse
|
10
|
Nie R, Zheng X, Zhang W, Zhang B, Ling Y, Zhang H, Wu C. Morphological Characteristics and Transcriptome Landscapes of Chicken Follicles during Selective Development. Animals (Basel) 2022; 12:ani12060713. [PMID: 35327110 PMCID: PMC8944860 DOI: 10.3390/ani12060713] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 03/07/2022] [Accepted: 03/09/2022] [Indexed: 01/27/2023] Open
Abstract
Ovarian follicle selection largely depends on the transition of granulosa cells from an undifferentiated to a fully differentiated state, which is accompanied by morphological and functional changes in follicles. The processes and transcriptional regulation of follicles during follicle selection are unclear; we thus used follicles from the prehierarchal to the hierarchal stage to investigate histology, reproductive endocrinology, and transcription. The morphology of follicles changed markedly during follicle selection. The numbers of large white, small yellow, and large yellow follicles (LWF, SYF, and LYF, respectively) were 11.83 ± 2.79, 6.83 ± 2.23, and 1.00, respectively, per ovary. LYF showed thicker granulosa cell layers than those of other prehierarchal follicles. Progesterone concentrations were significantly higher in LYF than that in LWF and SYF. In total, 16,823 genes were positively expressed in LWF, SYF, and LYF. Among follicle types, 1,290 differentially expressed genes were enriched regarding cell differentiation, blood vessel morphogenesis, and response to steroid hormones. Candidate genes associated with follicle selection participated in the Wnt signaling pathway, steroid hormone biosynthesis, and the TGF-β signaling pathway. We produced insights into crucial morphological characteristics of transcriptional regulation in follicle development. Our results provide an important basis for revealing the mechanism of follicle selection and potential impact on the poultry industry.
Collapse
Affiliation(s)
- Ruixue Nie
- National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (R.N.); (X.Z.); (W.Z.); (B.Z.); (Y.L.); (C.W.)
| | - Xiaotong Zheng
- National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (R.N.); (X.Z.); (W.Z.); (B.Z.); (Y.L.); (C.W.)
- School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China
| | - Wenhui Zhang
- National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (R.N.); (X.Z.); (W.Z.); (B.Z.); (Y.L.); (C.W.)
| | - Bo Zhang
- National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (R.N.); (X.Z.); (W.Z.); (B.Z.); (Y.L.); (C.W.)
| | - Yao Ling
- National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (R.N.); (X.Z.); (W.Z.); (B.Z.); (Y.L.); (C.W.)
| | - Hao Zhang
- National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (R.N.); (X.Z.); (W.Z.); (B.Z.); (Y.L.); (C.W.)
- Correspondence:
| | - Changxin Wu
- National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (R.N.); (X.Z.); (W.Z.); (B.Z.); (Y.L.); (C.W.)
| |
Collapse
|
11
|
Decoding the Phosphatase Code: Regulation of Cell Proliferation by Calcineurin. Int J Mol Sci 2022; 23:ijms23031122. [PMID: 35163061 PMCID: PMC8835043 DOI: 10.3390/ijms23031122] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 01/18/2022] [Accepted: 01/18/2022] [Indexed: 02/06/2023] Open
Abstract
Calcineurin, a calcium-dependent serine/threonine phosphatase, integrates the alterations in intracellular calcium levels into downstream signaling pathways by regulating the phosphorylation states of several targets. Intracellular Ca2+ is essential for normal cellular physiology and cell cycle progression at certain critical stages of the cell cycle. Recently, it was reported that calcineurin is activated in a variety of cancers. Given that abnormalities in calcineurin signaling can lead to malignant growth and cancer, the calcineurin signaling pathway could be a potential target for cancer treatment. For example, NFAT, a typical substrate of calcineurin, activates the genes that promote cell proliferation. Furthermore, cyclin D1 and estrogen receptors are dephosphorylated and stabilized by calcineurin, leading to cell proliferation. In this review, we focus on the cell proliferative functions and regulatory mechanisms of calcineurin and summarize the various substrates of calcineurin. We also describe recent advances regarding dysregulation of the calcineurin activity in cancer cells. We hope that this review will provide new insights into the potential role of calcineurin in cancer development.
Collapse
|
12
|
Lucchetta M, Pellegrini M. Drug repositioning by merging active subnetworks validated in cancer and COVID-19. Sci Rep 2021; 11:19839. [PMID: 34615934 PMCID: PMC8494853 DOI: 10.1038/s41598-021-99399-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Accepted: 09/23/2021] [Indexed: 02/08/2023] Open
Abstract
Computational drug repositioning aims at ranking and selecting existing drugs for novel diseases or novel use in old diseases. In silico drug screening has the potential for speeding up considerably the shortlisting of promising candidates in response to outbreaks of diseases such as COVID-19 for which no satisfactory cure has yet been found. We describe DrugMerge as a methodology for preclinical computational drug repositioning based on merging multiple drug rankings obtained with an ensemble of disease active subnetworks. DrugMerge uses differential transcriptomic data on drugs and diseases in the context of a large gene co-expression network. Experiments with four benchmark diseases demonstrate that our method detects in first position drugs in clinical use for the specified disease, in all four cases. Application of DrugMerge to COVID-19 found rankings with many drugs currently in clinical trials for COVID-19 in top positions, thus showing that DrugMerge can mimic human expert judgment.
Collapse
Affiliation(s)
- Marta Lucchetta
- Institute of Informatics and Telematics (IIT), CNR, Pisa, 56124, Italy
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, 53100, Italy
| | - Marco Pellegrini
- Institute of Informatics and Telematics (IIT), CNR, Pisa, 56124, Italy.
| |
Collapse
|
13
|
The Effect of Cyclosporine A on Proteins Controlling Intracellular Calcium Concentration in Breast Cancer Cells. J Membr Biol 2021; 255:33-39. [PMID: 34580765 DOI: 10.1007/s00232-021-00201-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 08/31/2021] [Indexed: 01/11/2023]
Abstract
Cyclosporine A (CsA) is an immunosuppressive drug commonly used to prevent autoimmune diseases. At the same time, CsA is a calcineurin (CaN) inhibitor. It affects the intracellular calcium signaling pathway. The effect of CsA on breast cancer cells, MDA-MB-231, plasma membrane calcium pump 1 (PMCA1), calmodulin (CaM), calcineurin (CaN), and cMyc, which are proteins that affect calcium signaling, were investigated. CsA inhibited the proliferation of MDA-MB-231 cells but did not affect the migration of the cells. After 24 h of incubation, CsA suppressed the PMCA1 protein, which pumps intracellular calcium out of the cell. At the same time, calcium started to accumulate inside the cell and CaM protein was expressed, while PMCA1 was suppressed. The CaN protein was suppressed 72 h after the administration of CsA, but the cMyc protein was expressed. Interestingly, 24 h incubation when the PMCA1 protein is down-regulated after the duration of time, the cMyc protein is also down-regulated. Although the indirect effect of CaN and cMyc is known, this relationship between PMCA1 and cMyc was not known. As a result, it has been shown that CsA affects the PMCA pump by disrupting the intracellular calcium pathway in breast cancer cells.
Collapse
|
14
|
Pontrelli P, Gigante M, Spadaccino F, Netti GS, Saldarelli M, Balducci L, Gigante M, Battaglia M, Storkus WJ, Castellano G, Stallone G, Gesualdo L, Ranieri E. CD40 Cross-Linking Induces Migration of Renal Tumor Cell through Nuclear Factor of Activated T Cells (NFAT) Activation. Int J Mol Sci 2021; 22:ijms22168871. [PMID: 34445576 PMCID: PMC8396205 DOI: 10.3390/ijms22168871] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 08/10/2021] [Accepted: 08/13/2021] [Indexed: 12/11/2022] Open
Abstract
CD40 crosslinking plays an important role in regulating cell migration, adhesion and proliferation in renal cell carcinoma (RCC). CD40/CD40L interaction on RCC cells activates different intracellular pathways but the molecular mechanisms leading to cell scattering are not yet clearly defined. Aim of our study was to investigate the main intracellular pathways activated by CD40 ligation and their specific involvement in RCC cell migration. CD40 ligation increased the phosphorylation of extracellular signal-regulated kinase (ERK), c-Jun NH (2)-terminal kinase (JNK) and p38 MAPK. Furthermore, CD40 crosslinking activated different transcriptional factors on RCC cell lines: AP-1, NFkB and some members of the Nuclear Factor of Activated T cells (NFAT) family. Interestingly, the specific inhibition of NFAT factors by cyclosporine A, completely blocked RCC cell motility induced by CD40 ligation. In tumor tissue, we observed a higher expression of NFAT factors and in particular an increased activation and nuclear migration of NFATc4 on RCC tumor tissues belonging to patients that developed metastases when compared to those who did not. Moreover, CD40-CD40L interaction induced a cytoskeleton reorganization and increased the expression of integrin β1 on RCC cell lines, and this effect was reversed by cyclosporine A and NFAT inhibition. These data suggest that CD40 ligation induces the activation of different intracellular signaling pathways, in particular the NFATs factors, that could represent a potential therapeutic target in the setting of patients with metastatic RCC.
Collapse
Affiliation(s)
- Paola Pontrelli
- Department of Emergency and Organ Transplantation, Divisions of Nephrology and Urology, University of Bari, Piazza G. Cesare 11, 70124 Bari, Italy; (P.P.); (M.B.); (L.G.)
| | - Margherita Gigante
- Department of Medical and Surgical Sciences, Divisions of Clinical Pathology and Nephrology, University of Foggia, Policlinico Riuniti, Viale L. Pinto, 71100 Foggia, Italy; (M.G.); (F.S.); (G.S.N.); (M.S.); (L.B.); (M.G.); (G.C.); (G.S.)
| | - Federica Spadaccino
- Department of Medical and Surgical Sciences, Divisions of Clinical Pathology and Nephrology, University of Foggia, Policlinico Riuniti, Viale L. Pinto, 71100 Foggia, Italy; (M.G.); (F.S.); (G.S.N.); (M.S.); (L.B.); (M.G.); (G.C.); (G.S.)
| | - Giuseppe Stefano Netti
- Department of Medical and Surgical Sciences, Divisions of Clinical Pathology and Nephrology, University of Foggia, Policlinico Riuniti, Viale L. Pinto, 71100 Foggia, Italy; (M.G.); (F.S.); (G.S.N.); (M.S.); (L.B.); (M.G.); (G.C.); (G.S.)
| | - Marilisa Saldarelli
- Department of Medical and Surgical Sciences, Divisions of Clinical Pathology and Nephrology, University of Foggia, Policlinico Riuniti, Viale L. Pinto, 71100 Foggia, Italy; (M.G.); (F.S.); (G.S.N.); (M.S.); (L.B.); (M.G.); (G.C.); (G.S.)
| | - Luigi Balducci
- Department of Medical and Surgical Sciences, Divisions of Clinical Pathology and Nephrology, University of Foggia, Policlinico Riuniti, Viale L. Pinto, 71100 Foggia, Italy; (M.G.); (F.S.); (G.S.N.); (M.S.); (L.B.); (M.G.); (G.C.); (G.S.)
| | - Maddalena Gigante
- Department of Medical and Surgical Sciences, Divisions of Clinical Pathology and Nephrology, University of Foggia, Policlinico Riuniti, Viale L. Pinto, 71100 Foggia, Italy; (M.G.); (F.S.); (G.S.N.); (M.S.); (L.B.); (M.G.); (G.C.); (G.S.)
| | - Michele Battaglia
- Department of Emergency and Organ Transplantation, Divisions of Nephrology and Urology, University of Bari, Piazza G. Cesare 11, 70124 Bari, Italy; (P.P.); (M.B.); (L.G.)
| | - Walter J. Storkus
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA;
| | - Giuseppe Castellano
- Department of Medical and Surgical Sciences, Divisions of Clinical Pathology and Nephrology, University of Foggia, Policlinico Riuniti, Viale L. Pinto, 71100 Foggia, Italy; (M.G.); (F.S.); (G.S.N.); (M.S.); (L.B.); (M.G.); (G.C.); (G.S.)
| | - Giovanni Stallone
- Department of Medical and Surgical Sciences, Divisions of Clinical Pathology and Nephrology, University of Foggia, Policlinico Riuniti, Viale L. Pinto, 71100 Foggia, Italy; (M.G.); (F.S.); (G.S.N.); (M.S.); (L.B.); (M.G.); (G.C.); (G.S.)
| | - Loreto Gesualdo
- Department of Emergency and Organ Transplantation, Divisions of Nephrology and Urology, University of Bari, Piazza G. Cesare 11, 70124 Bari, Italy; (P.P.); (M.B.); (L.G.)
| | - Elena Ranieri
- Department of Medical and Surgical Sciences, Divisions of Clinical Pathology and Nephrology, University of Foggia, Policlinico Riuniti, Viale L. Pinto, 71100 Foggia, Italy; (M.G.); (F.S.); (G.S.N.); (M.S.); (L.B.); (M.G.); (G.C.); (G.S.)
- Correspondence: ; Tel.: +39-0881-732611; Fax: +39-0881-732627
| |
Collapse
|
15
|
Mahmoud SS, Hussein S, Rashed H, Abdelghany EMA, Ali AI. Anticancer Effects of Tacrolimus on Induced Hepatocellular Carcinoma in Mice. Curr Mol Pharmacol 2021; 15:434-445. [PMID: 34061012 DOI: 10.2174/1874467214666210531164546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 02/12/2021] [Accepted: 02/17/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Tacrolimus is a calcineurin inhibitor widely used for immunological disorders. However, there is a significant controversy regarding its effect on the liver. The present study was conducted to evaluate the anticancer effects of tacrolimus on an induced murine hepatocellular carcinoma (HCC) model and its possible hepatotoxicity at standard therapeutic doses. METHODS Fifty-four male mice were divided into five groups: a control healthy group, control HCC group, tacrolimus-treated group, doxorubicin (DOXO)-treated group, and combined tacrolimus- and DOXO-treated group. The activity of liver enzymes, including alkaline phosphatase, gamma-glutamyl transferase, lactate dehydrogenase, alanine transaminase, and aspartate transaminase, was determined. Serum vascular endothelial growth factor (VEGF) was measured using an enzyme-linked immunosorbent assay. A quantitative real-time polymerase chain reaction (qRT-PCR) was conducted to measure the expression of proliferating cell nuclear antigen (PCNA), Bax, and p53 mRNA. Immunohistochemical staining for cyclin D1 and VEGF was performed. RESULTS Mice that received combined treatment with tacrolimus and DOXO exhibited the best improvement in all parameters when compared with the groups that received DOXO or tacrolimus alone (p < 0.001). CONCLUSION The combination of DOXO and tacrolimus was more effective in the management of HCC compared with either agent alone. This improvement was detected by the reduction of liver enzymes and the improvement of the histopathological picture. The involved mechanisms included significant apoptosis induction demonstrated by upregulation of bax along with a reduction in angiogenesis demonstrated by downregulation of VEGF. This was accompanied by inhibition of cell cycle progression mediated by upregulated p53 and downregulated PCNA and cyclin D1.
Collapse
Affiliation(s)
- Shireen Sami Mahmoud
- Clinical Pharmacology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Samia Hussein
- Medical Biochemistry & Molecular Biology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Hayam Rashed
- Pathology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Eman M A Abdelghany
- Anatomy and Embryology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Alaa I Ali
- Clinical Pharmacology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| |
Collapse
|
16
|
Kawahara T, Teramoto Y, Li Y, Ishiguro H, Gordetsky J, Yang Z, Miyamoto H. Impact of Vasectomy on the Development and Progression of Prostate Cancer: Preclinical Evidence. Cancers (Basel) 2020; 12:E2295. [PMID: 32824199 PMCID: PMC7464827 DOI: 10.3390/cancers12082295] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 08/07/2020] [Accepted: 08/13/2020] [Indexed: 12/13/2022] Open
Abstract
Some observational studies have implied a link between vasectomy and an elevated risk of prostate cancer. We investigated the impact of vasectomy on prostate cancer outgrowth, mainly using preclinical models. Neoplastic changes in the prostate were compared in transgenic TRAMP mice that underwent vasectomy vs. sham surgery performed at 4 weeks of age. One of the molecules identified by DNA microarray (i.e., ZKSCAN3) was then assessed in radical prostatectomy specimens and human prostate cancer lines. At 24 weeks, gross tumor (p = 0.089) and poorly differentiated adenocarcinoma (p = 0.036) occurred more often in vasectomized mice. Vasectomy significantly induced ZKSCAN3 expression in prostate tissues from C57BL/6 mice and prostate cancers from TRAMP mice. Immunohistochemistry showed increased ZKSCAN3 expression in adenocarcinoma vs. prostatic intraepithelial neoplasia (PIN), PIN vs. non-neoplastic prostate, Grade Group ≥3 vs. ≤2 tumors, pT3 vs. pT2 tumors, pN1 vs. pN0 tumors, and prostate cancer from patients with a history of vasectomy. Additionally, strong (2+/3+) ZKSCAN3 expression (p = 0.002), as an independent prognosticator, or vasectomy (p = 0.072) was associated with the risk of tumor recurrence. In prostate cancer lines, ZKSCAN3 silencing resulted in significant decreases in cell proliferation/migration/invasion. These findings suggest that there might be an association between vasectomy and the development and progression of prostate cancer, with up-regulation of ZKSCAN3 expression as a potential underlying mechanism.
Collapse
Affiliation(s)
- Takashi Kawahara
- Department of Pathology & Laboratory Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA; (T.K.); (Y.T.); (Y.L.); (H.I.); (J.G.); (Z.Y.)
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Departments of Urology and Renal Transplantation, Yokohama City University Medical Center, Yokohama 232-0024, Japan
| | - Yuki Teramoto
- Department of Pathology & Laboratory Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA; (T.K.); (Y.T.); (Y.L.); (H.I.); (J.G.); (Z.Y.)
- James P. Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Yi Li
- Department of Pathology & Laboratory Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA; (T.K.); (Y.T.); (Y.L.); (H.I.); (J.G.); (Z.Y.)
| | - Hitoshi Ishiguro
- Department of Pathology & Laboratory Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA; (T.K.); (Y.T.); (Y.L.); (H.I.); (J.G.); (Z.Y.)
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Kanagawa Institute of Industrial Science and Technology, Kawasaki 210-0821, Japan
- Department of Urology, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan
| | - Jennifer Gordetsky
- Department of Pathology & Laboratory Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA; (T.K.); (Y.T.); (Y.L.); (H.I.); (J.G.); (Z.Y.)
- Department of Urology, University of Rochester Medical Center, Rochester, NY 14642, USA
- Departments of Pathology and Urology, Vanderbilt University Medical Center, Nashville, TN 37212, USA
| | - Zhiming Yang
- Department of Pathology & Laboratory Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA; (T.K.); (Y.T.); (Y.L.); (H.I.); (J.G.); (Z.Y.)
| | - Hiroshi Miyamoto
- Department of Pathology & Laboratory Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA; (T.K.); (Y.T.); (Y.L.); (H.I.); (J.G.); (Z.Y.)
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- James P. Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY 14642, USA
- Department of Urology, University of Rochester Medical Center, Rochester, NY 14642, USA
| |
Collapse
|
17
|
Tacrolimus and ascomycin inhibit melanoma cell growth, migration and invasion via targeting nuclear factor of activated T-cell 3. Melanoma Res 2020; 30:325-335. [PMID: 32301802 DOI: 10.1097/cmr.0000000000000663] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Melanoma is the most malignant form of skin cancer with high metastatic potential. Nuclear factor of activated T-cells (NFATs) are discovered as transcription factors that regulate the expression of proinflammatory cytokines and other genes during the immune response. Among five NFAT members, NFAT3 is exclusively not expressed in immune cells and its role in progression of different types of cancer remains controversial. Our previous study showed that NFAT3 was highly expressed in skin cancer compared with normal skin tissues and critical for melanoma cell survival and tumor growth. Here, we reported that knockdown of NFAT3 expression, as well as treatment with the calcineurin (CaN) inhibitors, tacrolimus (FK506) or ascomycin (FK520) inhibits melanoma cell migration and invasion, and also proliferation and colony formation. Mechanistic studies revealed that FK506 or FK520 blocked the nuclear translocation and reduced the transcriptional activity of NFAT3. These data support that the antimelanoma effect of FK506 and FK520 is partially mediated by inhibiting the oncogenic factor NFAT3, suggesting that therapeutics based on NFAT3 inhibition may be effective in clinical melanoma treatment.
Collapse
|
18
|
HSF1 phosphorylation by cyclosporin A confers hyperthermia sensitivity through suppression of HSP expression. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2019; 1862:846-857. [DOI: 10.1016/j.bbagrm.2019.04.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 04/21/2019] [Accepted: 04/30/2019] [Indexed: 12/31/2022]
|
19
|
Gao L, Dong J, Zhang N, Le Z, Ren W, Li S, Li F, Song J, Wang Q, Dou Z, Park SY, Zhi K. Cyclosporine A Suppresses the Malignant Progression of Oral Squamous Cell Carcinoma in vitro. Anticancer Agents Med Chem 2019; 19:248-255. [PMID: 30378503 DOI: 10.2174/1871520618666181029170605] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2017] [Revised: 02/14/2018] [Accepted: 10/09/2018] [Indexed: 12/15/2022]
Abstract
Background:The Oral Squamous Cell Carcinoma (OSCC) is one of the most frequent cancer types. Failure of treatment of OSCC is potentially lethal because of local recurrence, regional lymph node metastasis, and distant metastasis. Chemotherapy plays a vital role through suppression of tumorigenesis. Cyclosporine A (CsA), an immunosuppressant drug, has been efficiently used in allograft organ transplant recipients to prevent rejection, and also has been used in a subset of patients with autoimmunity related disorders. The present study aims to investigate novel and effective chemotherapeutic drugs to overcome drug-resistance in the treatment of OSCC.Methods:Cells were incubated in the standard way. Cell viability was assayed using the MTT assay. Cell proliferation was determined using colony formation assay. The cell cycle assay was performed using flow cytometry. Apoptosis was assessed using fluorescence-activated cell sorting after stained by the Annexin V-fluorescein isothiocyanate (FITC). Cell migration and invasion were analyzed using wound healing assay and tranwell. The effect of COX-2, c-Myc, MMP-9, MMP-2, and NFATc1 protein expression was determined using Western blot analysis while NFATc1 mRNA expression was determined by RT-PCR.Results:In vitro studies indicated that CsA inhibited partial OSCC growth by inducing cell cycle arrest, apoptosis, and the migration and invasion of OSCC cells. We also demonstrated that CsA could inhibit the expression of NFATc1 and its downstream genes COX-2, c-Myc, MMP-9, and MMP-2 in OSCC cells. Furthermore, we analyzed the expression of NFATc1 in head and neck cancer through the Oncomine database. The data was consistent with the experimental findings.Conclusion:The present study initially demonstrated that CsA could inhibit the progression of OSCC cells and can mediate the signal molecules of NFATc1 signaling pathway, which has strong relationship with cancer development. That explains us CsA has potential to explore the possibilities as a novel chemotherapeutic drug for the treatment of OSCC.
Collapse
Affiliation(s)
- Ling Gao
- Key Lab of Oral Clinical Medicine, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Jianwei Dong
- Department of Stomatology, Shangluo Central Hospital, Shangluo, Shanxi, China
| | - Nanyang Zhang
- Key Lab of Oral Clinical Medicine, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Zhanxian Le
- Fujian Provincial Key Laboratory of Screening for Novel Microbial Products, Fujian Institute of Microbiology, Fuzhou, Fujian, China
| | - Wenhao Ren
- Key Lab of Oral Clinical Medicine, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Shaoming Li
- Key Lab of Oral Clinical Medicine, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Fan Li
- Key Lab of Oral Clinical Medicine, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Jianzhong Song
- Key Lab of Oral Clinical Medicine, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Qibo Wang
- Key Lab of Oral Clinical Medicine, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Zhichao Dou
- Key Lab of Oral Clinical Medicine, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Soo Y. Park
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Daegu, South Korea
| | - Keqian Zhi
- Key Lab of Oral Clinical Medicine, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| |
Collapse
|
20
|
Flores C, Fouquet G, Moura IC, Maciel TT, Hermine O. Lessons to Learn From Low-Dose Cyclosporin-A: A New Approach for Unexpected Clinical Applications. Front Immunol 2019; 10:588. [PMID: 30984176 PMCID: PMC6447662 DOI: 10.3389/fimmu.2019.00588] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 03/05/2019] [Indexed: 01/09/2023] Open
Abstract
Cyclosporin-A has been known and used for a long time, since its "fast track" approval in the early 80's. This molecule has rapidly demonstrated unexpected immunosuppressive properties, transforming the history of organ transplantation. Cyclosporin's key effect relies on modulation on T-lymphocyte activity, which explains its role in the prevention of graft rejection. However, whether cyclosporin-A exerts other effects on immune system remains to be determined. Until recently, cyclosporin-A was mainly used at a high-dose, but given the drug toxicity and despite the fear of losing its immunosuppressive effects, there is nowadays a tendency to decrease its dose. The literature has been reporting data revealing a paradoxical effect of low dosage of cyclosporin-A. These low-doses appear to have immunomodulatory properties, with different effects from high-doses on CD8+ T lymphocyte activation, auto-immune diseases, graft-vs.-host disease and cancer. The aim of this review is to discuss the role of cyclosporin-A, not only as a consecrated immunosuppressive agent, but also as an immunomodulatory drug when administrated at low-dose. The use of low-dose cyclosporin-A may become a new therapeutic strategy, particularly to treat cancer.
Collapse
Affiliation(s)
- Camila Flores
- INSERM UMR1163 and CNRS URL 8254, Imagine Institute, Paris, France
- Paris Descartes University-Sorbonne Paris Cité, Paris, France
| | - Guillemette Fouquet
- INSERM UMR1163 and CNRS URL 8254, Imagine Institute, Paris, France
- Paris Descartes University-Sorbonne Paris Cité, Paris, France
| | - Ivan Cruz Moura
- INSERM UMR1163 and CNRS URL 8254, Imagine Institute, Paris, France
- Paris Descartes University-Sorbonne Paris Cité, Paris, France
| | - Thiago Trovati Maciel
- INSERM UMR1163 and CNRS URL 8254, Imagine Institute, Paris, France
- Paris Descartes University-Sorbonne Paris Cité, Paris, France
| | - Olivier Hermine
- INSERM UMR1163 and CNRS URL 8254, Imagine Institute, Paris, France
- Paris Descartes University-Sorbonne Paris Cité, Paris, France
- Department of Hematology, Necker Children's Hospital, APHP, Paris, France
| |
Collapse
|
21
|
Ding N, Geng B, Li Z, Yang Q, Yan L, Wan L, Zhang B, Wang C, Xia Y. Fluid shear stress promotes osteoblast proliferation through the NFATc1-ERK5 pathway. Connect Tissue Res 2019; 60:107-116. [PMID: 29609502 DOI: 10.1080/03008207.2018.1459588] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
PURPOSE Extracellular-regulated kinase 5 (ERK5) is thought to regulate osteoblast proliferation. To further understand how ERK5 signaling regulates osteoblast proliferation induced by fluid shear stress (FSS), we examined some potential signaling targets associated with ERK5 in MC3T3-E1 cells. METHODS MC3T3-E1 cells were treated with XMD8-92 (an ERK5 inhibitor) or Cyclosporin A (CsA, a nuclear factor of activated T cells (NFAT) c1 inhibitor) and/or exposed to 12 dyn/cm2 FSS. Phosphorylated-ERK5 (p-ERK5) and expression levels of NFATc1, ERK5, E2F2, and cyclin E1 were analyzed by western blot. The mRNA levels of genes associated with cell proliferation were analyzed by Polymerase Chain Reaction (PCR) array. Subcellular localization of p-ERK5 and NFATc1 were determined by immunofluorescence. Cell proliferation was evaluated by MTT assay. RESULTS NFATc1 expression was up-regulated by FSS. XMD8-92 only blocked ERK5 activation; however, CsA decreased NFATc1 and p-ERK5 levels, including after FSS stimulation. Exposure to NFATc1 inhibitor or ERK5 inhibitor resulted in decreased E2F2 and cyclin E1 expression and proliferation by proliferative MC3T3-E1 cells. Furthermore, immunofluorescence results illustrated that NFATc1 induced ERK5 phosphorylation, resulting in p-ERK5 translocation to the nucleus. CONCLUSIONS Our results reveal that NFATc1 acts as an intermediate to promote the phosphorylation of ERK5 induced by FSS. Moreover, activated NFATc1-ERK5 signaling up-regulates the expression of E2F2 and cyclin E1, which promote osteoblast proliferation.
Collapse
Affiliation(s)
- Ning Ding
- a Department of Orthopaedics , Lanzhou University Second Hospital , Lanzhou , Gansu , China.,b Orthopaedics Key Laboratory of Gansu Province , Lanzhou , Gansu , China
| | - Bin Geng
- a Department of Orthopaedics , Lanzhou University Second Hospital , Lanzhou , Gansu , China.,b Orthopaedics Key Laboratory of Gansu Province , Lanzhou , Gansu , China
| | - Zhonghao Li
- a Department of Orthopaedics , Lanzhou University Second Hospital , Lanzhou , Gansu , China.,b Orthopaedics Key Laboratory of Gansu Province , Lanzhou , Gansu , China
| | - Quanzeng Yang
- a Department of Orthopaedics , Lanzhou University Second Hospital , Lanzhou , Gansu , China.,b Orthopaedics Key Laboratory of Gansu Province , Lanzhou , Gansu , China
| | - Liang Yan
- a Department of Orthopaedics , Lanzhou University Second Hospital , Lanzhou , Gansu , China.,b Orthopaedics Key Laboratory of Gansu Province , Lanzhou , Gansu , China
| | - Lang Wan
- a Department of Orthopaedics , Lanzhou University Second Hospital , Lanzhou , Gansu , China.,b Orthopaedics Key Laboratory of Gansu Province , Lanzhou , Gansu , China
| | - Bo Zhang
- a Department of Orthopaedics , Lanzhou University Second Hospital , Lanzhou , Gansu , China.,b Orthopaedics Key Laboratory of Gansu Province , Lanzhou , Gansu , China
| | - Cuifang Wang
- a Department of Orthopaedics , Lanzhou University Second Hospital , Lanzhou , Gansu , China.,b Orthopaedics Key Laboratory of Gansu Province , Lanzhou , Gansu , China
| | - Yayi Xia
- a Department of Orthopaedics , Lanzhou University Second Hospital , Lanzhou , Gansu , China.,b Orthopaedics Key Laboratory of Gansu Province , Lanzhou , Gansu , China
| |
Collapse
|
22
|
Liu Y, Liang T, Qiu X, Ye X, Li Z, Tian B, Yan D. Down-Regulation of Nfatc1 Suppresses Proliferation, Migration, Invasion, and Warburg Effect in Prostate Cancer Cells. Med Sci Monit 2019; 25:1572-1581. [PMID: 30817743 PMCID: PMC6404685 DOI: 10.12659/msm.910998] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2018] [Accepted: 10/04/2018] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Prostate cancer (PCa), accounting for 28% of all male cancer cases, is the second leading cause of cancer-related death among men. NFATc1, belonging to the NFAT family, is overexpressed in PCa and is correlated with the risk of recurrence after radical prostatectomy. MATERIAL AND METHODS In the present study, the expression of NFATc, c-myc, and PKM2 in PCa cells was regulated by lentiviruses and then detected by real-time PCR and Western blot analysis. Further, proliferation, invasion, and migration assays were performed. The glucose consumption and lactate production were assessed by biochemical detection. RESULTS We found that NFATc1 down-regulation significantly suppressed the proliferation and Warburg effect of PCa cells, concurrent with a decrease of c-myc and PKM2 expression. Likewise, the abilities of migration and invasion were also inhibited in NFATc1-silenced PCa cells. In addition, NFATc1 down-regulation-induced inhibition of cell proliferation, migration, invasion, and Warburg effect were counteracted by up-regulation of c-myc or PKM2. The expression of PKM2 was positively regulated by NFATc1 and c-myc expression. CONCLUSIONS These results indicate that NFATc1 down-regulation can suppress the proliferation, Warburg effect, and migration and invasion abilities of PCa cells, probably by regulating c-myc and PKM2 expression. NFATc1 may be a potential therapeutic target for PCa and could be used as a diagnosis or prognosis indicator of PCa.
Collapse
Affiliation(s)
- Yuanyuan Liu
- Department of Urology, Affiliated Hospital of Binzhou Medical University, Binzhou, Shandong, P.R. China
- Department of Urology, Shanghai Sixth People’s Hospital East Affiliated to Shanghai University of Medicine and Health Sciences, Shanghai, P.R. China
| | - Tao Liang
- Department of Urology, Shanghai Sixth People’s Hospital East Affiliated to Shanghai University of Medicine and Health Sciences, Shanghai, P.R. China
| | - Xinkai Qiu
- Department of Urology, Affiliated Hospital of Binzhou Medical University, Binzhou, Shandong, P.R. China
| | - Xuxiao Ye
- Department of Urology, Shanghai Sixth People’s Hospital East Affiliated to Shanghai University of Medicine and Health Sciences, Shanghai, P.R. China
| | - Zuowei Li
- Department of Urology, Shanghai Sixth People’s Hospital East Affiliated to Shanghai University of Medicine and Health Sciences, Shanghai, P.R. China
| | - Binqiang Tian
- Department of Urology, Shanghai Sixth People’s Hospital East Affiliated to Shanghai University of Medicine and Health Sciences, Shanghai, P.R. China
| | - Dongliang Yan
- Department of Urology, Shanghai Sixth People’s Hospital East Affiliated to Shanghai University of Medicine and Health Sciences, Shanghai, P.R. China
| |
Collapse
|
23
|
Kawahara T, Ishiguro Y, Ohtake S, Kato I, Ito Y, Ito H, Makiyama K, Kondo K, Miyoshi Y, Yumura Y, Hayashi N, Hasumi H, Osaka K, Muraoka K, Izumi K, Teranishi JI, Uemura H, Yao M, Nakaigawa N. PD-1 and PD-L1 are more highly expressed in high-grade bladder cancer than in low-grade cases: PD-L1 might function as a mediator of stage progression in bladder cancer. BMC Urol 2018; 18:97. [PMID: 30400941 PMCID: PMC6219206 DOI: 10.1186/s12894-018-0414-8] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 10/25/2018] [Indexed: 02/08/2023] Open
Abstract
Background Bladder cancers have been characterized as a tumor group in which the immunological response is relatively well preserved. Programmed death ligand 1 (PD-L1, B7-H1, CD274) has been shown to be expressed in several malignancies, including bladder cancer. However, the clinicopathological impact of this biomarker has not yet been established. In the present study, a quantitative real-time polymerase chain reaction (qPCR) was performed using paired normal and cancerous bladder cancer tissue to investigate PD-1/PD-L1 gene expression. Methods We examined the mRNA expression of PD-1/PD-L1 by a qPCR using 58 pairs of normal and cancerous human bladder tissue specimens. We also examined the correlation with the expressions of the STAT1 and NFAT genes, which are thought to be upstream and downstream of the PD-L1 pathway, respectively. Results There were no significant differences between normal and cancerous tissue in the expression of the PD-1 and PD-L1 genes (p = 0.724 and p = 0.102, respectively). However, PD-1 and PD-L1 were both more highly expressed in high-grade bladder cancer than in low-grade bladder cancer (p < 0.050 and p < 0.010). PD-L1 was positively correlated with the expressions of both the STAT1 (r = 0.681, p < 0.001) and the NFATc1 genes (r = 0.444. p < 0.001). Conclusions PD-1 and PD-L1 might be a new biomarker that correlates with the pathological grade of bladder cancer. PD-L1 might function as a mediator of stage progression in bladder cancer and STAT1-NFAT pathway might associate this function. Electronic supplementary material The online version of this article (10.1186/s12894-018-0414-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Takashi Kawahara
- Department of Urology, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, Kanagawa, 2360004, Japan
| | - Yukari Ishiguro
- Department of Urology, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, Kanagawa, 2360004, Japan
| | - Shinji Ohtake
- Department of Urology, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, Kanagawa, 2360004, Japan
| | - Ikuma Kato
- Department of Pathology, Yokohama City University Hospital, Yokohama, Japan
| | - Yusuke Ito
- Department of Urology, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, Kanagawa, 2360004, Japan
| | - Hiroki Ito
- Department of Urology, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, Kanagawa, 2360004, Japan
| | - Kazuhide Makiyama
- Department of Urology, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, Kanagawa, 2360004, Japan
| | - Keiichi Kondo
- Department of Urology, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, Kanagawa, 2360004, Japan
| | - Yasuhide Miyoshi
- Department of Urology, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, Kanagawa, 2360004, Japan
| | - Yasushi Yumura
- Department of Urology, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, Kanagawa, 2360004, Japan
| | - Narihiko Hayashi
- Department of Urology, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, Kanagawa, 2360004, Japan
| | - Hisashi Hasumi
- Department of Urology, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, Kanagawa, 2360004, Japan
| | - Kimito Osaka
- Department of Urology, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, Kanagawa, 2360004, Japan
| | - Kentaro Muraoka
- Department of Urology, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, Kanagawa, 2360004, Japan
| | - Koji Izumi
- Department of Urology, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, Kanagawa, 2360004, Japan
| | - Jun-Ichi Teranishi
- Department of Urology, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, Kanagawa, 2360004, Japan
| | - Hiroji Uemura
- Department of Urology, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, Kanagawa, 2360004, Japan.,Department of Urology and Renal Transplantation, Yokohama City University Medical Center, Yokohama, Japan
| | - Masahiro Yao
- Department of Urology, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, Kanagawa, 2360004, Japan
| | - Noboru Nakaigawa
- Department of Urology, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, Kanagawa, 2360004, Japan.
| |
Collapse
|
24
|
Qin JJ, Li X, Wang W, Zi X, Zhang R. Targeting the NFAT1-MDM2-MDMX Network Inhibits the Proliferation and Invasion of Prostate Cancer Cells, Independent of p53 and Androgen. Front Pharmacol 2017; 8:917. [PMID: 29311926 PMCID: PMC5735069 DOI: 10.3389/fphar.2017.00917] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 11/30/2017] [Indexed: 12/31/2022] Open
Abstract
The MDM2 and MDMX oncogenes are overexpressed in various types of human cancer and are highly associated with the initiation, progression, metastasis and chemotherapeutic resistance of these diseases, including prostate cancer. The present study was designed to test a natural MDM2 inhibitor, Inulanolide A (InuA), for anti-prostate cancer activity and to determine the underlying mechanism(s) of action. InuA directly bound to the RING domains of both MDM2 and MDMX with high affinity and specificity and disrupted MDM2-MDMX binding, markedly enhancing MDM2 protein degradation. We further discovered that InuA bound to the DNA binding domain of NFAT1, resulting in marked inhibition of MDM2 transcription. InuA inhibited the proliferation, migration, and invasion of prostate cancer cells, regardless of their p53 status and AR responsiveness. Double knockdown of MDM2 and NFAT1 also revealed that the expression of both of these molecules is important for InuA’s inhibitory effects on the proliferation and invasion of prostate cancer cells. In summary, InuA represents a novel class of bifunctional MDM2 inhibitors, and should be further investigated as a candidate lead compound for prostate cancer prevention and therapy.
Collapse
Affiliation(s)
- Jiang-Jiang Qin
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, United States.,Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, United States
| | - Xin Li
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, United States.,Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, United States
| | - Wei Wang
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, United States.,Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, United States.,Center for Drug Discovery, University of Houston, Houston, TX, United States
| | - Xiaolin Zi
- Department of Urology, University of California, Irvine, Irvine, CA, United States.,Department of Pharmacology, University of California, Irvine, Irvine, CA, United States
| | - Ruiwen Zhang
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, United States.,Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, United States.,Center for Drug Discovery, University of Houston, Houston, TX, United States
| |
Collapse
|
25
|
Santio NM, Koskinen PJ. PIM kinases: From survival factors to regulators of cell motility. Int J Biochem Cell Biol 2017; 93:74-85. [DOI: 10.1016/j.biocel.2017.10.016] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 10/26/2017] [Accepted: 10/31/2017] [Indexed: 01/01/2023]
|
26
|
Jin H, Wang C, Jin G, Ruan H, Gu D, Wei L, Wang H, Wang N, Arunachalam E, Zhang Y, Deng X, Yang C, Xiong Y, Feng H, Yao M, Fang J, Gu J, Cong W, Qin W. Regulator of Calcineurin 1 Gene Isoform 4, Down-regulated in Hepatocellular Carcinoma, Prevents Proliferation, Migration, and Invasive Activity of Cancer Cells and Metastasis of Orthotopic Tumors by Inhibiting Nuclear Translocation of NFAT1. Gastroenterology 2017; 153:799-811.e33. [PMID: 28583823 DOI: 10.1053/j.gastro.2017.05.045] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Revised: 05/27/2017] [Accepted: 05/27/2017] [Indexed: 01/09/2023]
Abstract
BACKGROUND & AIMS Individuals with Down syndrome have a low risk for many solid tumors, prompting the search for tumor suppressor genes on human chromosome 21 (HSA21). We aimed to identify and explore potential mechanisms of tumor suppressors on HSA21 in hepatocellular carcinoma (HCC). METHODS We compared expression of HSA21 genes in 14 pairs of primary HCC and adjacent noncancer liver tissues using the Affymetrix HG-U133 Plus 2.0 array (Affymetrix, Santa Clara, CA). HCC tissues and adjacent normal liver tissues were collected from 108 patients at a hospital in China for real-time polymerase chain reaction and immunohistochemical analyses; expression levels of regulator of calcineurin 1 (RCAN1) isoform 4 (RCAN1.4) were associated with clinical features. We overexpressed RCAN1.4 from lentiviral vectors in MHCC97H and HCCLM3 cells and knocked expression down using small interfering RNAs in SMMC7721 and Huh7 cells. Cells were analyzed in proliferation, migration, and invasion assays. HCC cells that overexpressed RCAN1.4 or with RCAN1.4 knockdown were injected into livers or tail veins of nude mice; tumor growth and numbers of lung metastases were quantified. We performed bisulfite pyrosequencing and methylation-specific polymerase chain reaction analyses to analyze CpG island methylation. We measured phosphatase activity of calcineurin in HCC cells. RESULTS RCAN1.4 mRNA and protein levels were significantly decreased in primary HCC compared with adjacent noncancer liver tissues. Reduced levels of RCAN1.4 mRNA were significantly associated with advanced tumor stages, poor differentiation, larger tumor size, and vascular invasion. Kaplan-Meier survival analysis showed that patients with HCCs with lower levels of RCAN1.4 mRNA had shorter time of overall survival and time to recurrence than patients whose tumors had high levels of RCAN1.4 mRNA. In HCC cell lines, expression of RCAN1.4 significantly reduced proliferation, migration, and invasive activity. HCC cells that overexpressed RCAN1.4 formed smaller xenograft tumors, with fewer metastases and blood vessels, than control HCC cells. In HCC cells, RCAN1.4 inhibited expression of insulin-like growth factor 1 and vascular endothelial growth factor A by reducing calcineurin activity and blocking nuclear translocation of nuclear factor of activated T cells (NFAT1). HCC cells incubated with the calcineurin inhibitor cyclosporin A had decreased nuclear level of NFAT1. HCC cells had hypermethylation of a CpG island in the 5' regulatory region of RCAN1.4, which reduced its expression. CONCLUSIONS RCAN1.4 is down-regulated in HCC tissues, compared with non-tumor liver tissues. RCAN1.4 prevents cell proliferation, migration, and invasion in vitro; overexpressed RCAN1.4 in HCC cells prevents growth, angiogenesis, and metastases of xenograft tumors by inhibiting calcineurin activity and nuclear translocation of NFAT1.
Collapse
Affiliation(s)
- Haojie Jin
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Cun Wang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guangzhi Jin
- Department of Pathology, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| | - Haoyu Ruan
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dishui Gu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Department of Pathophysiology, School of Basic Medical Sciences, Guangdong Medical University, Dongguan, Guangdong, China
| | - Lin Wei
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hui Wang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ning Wang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Einthavy Arunachalam
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; The School of Biosciences and Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, Surry, UK
| | - Yurong Zhang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xuan Deng
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chen Yang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yi Xiong
- State Key Laboratory of Microbial Metabolism and College of Life Science and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Hugang Feng
- Department of Life Science, Imperial College, London, UK
| | - Ming Yao
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jingyuan Fang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jianren Gu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wenming Cong
- Department of Pathology, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China.
| | - Wenxin Qin
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
27
|
Li Y, Zhang H, Li Y, Zhao C, Li W, Liu H, Wen J, Chen J. [Study on the Effect of Immunosuppressive Agent FK506 on Growth and Migration of Lung Cancer Cell]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2017; 20:446-451. [PMID: 28738959 PMCID: PMC5972949 DOI: 10.3779/j.issn.1009-3419.2017.07.02] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
背景与目的 FK506(他克莫司,tacrolimus)是一种大环内酯类的新型免疫抑制剂。研究报道FK506对多种肿瘤细胞具有增殖抑制作用。本研究旨在观察FK506对肺癌细胞增殖和迁移的作用,并探讨其可能的机制。 方法 体外培养A549和H1299细胞,采用CCK-8法测定FK506对A549和H1299细胞增殖的作用;EDU标记法检测DNA合成;流式细胞术检测细胞的周期分布情况;Transwell和细胞划痕实验检测细胞的体外迁移能力;Western blot技术检测P27、RB1、CDK4、CDK6和MMP9蛋白的表达。 结果 FK506可抑制A549和H1299细胞的增殖、诱导细胞周期G0期/G1期阻滞;与对照组比较,FK506处理后A549和H1299细胞迁移能力明显降低,且呈剂量依赖性。此外,与对照组相比,FK506处理组中P27和RB1表达上调,而CDK4、CDK6和MMP9表达显著下降。 结论 FK506对人肺癌A549和H1299细胞的增殖和迁移能力有明显的抑制作用,其机制可能与上调p27表达、抑制CDK4、CDK6和MMP-9表达有关。
Collapse
Affiliation(s)
- Yongwen Li
- Department of Biological Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, China;Tianjin Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Hongbing Zhang
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Ying Li
- Tianjin Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Chenlong Zhao
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Weiting Li
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Hongyu Liu
- Tianjin Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Jianping Wen
- Department of Biological Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, China
| | - Jun Chen
- Tianjin Lung Cancer Institute;Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin 300052, China
| |
Collapse
|
28
|
Yokomizo Y, Kawahara T, Nagashima Y, Ishiguro H, Kato I, Yao M, Miyamoto H, Uemura H. Lack of an association between the aPKCλ/ι expression in prostate cancer and the patient outcomes. Int J Surg Case Rep 2017; 37:180-182. [PMID: 28697433 PMCID: PMC5504077 DOI: 10.1016/j.ijscr.2017.06.047] [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: 04/04/2017] [Revised: 06/20/2017] [Accepted: 06/20/2017] [Indexed: 11/20/2022] Open
Abstract
This is the first study to assess aPKCλ/ι expression in primary prostate cancer with metastatic disease. A total of 43 patients with prostate cancer and its metastasis to the lymph node and/or bone were analyzed in this study. We found no strong association between aPKCλ/ι expression and the prognosis of the patients.
Collapse
Affiliation(s)
- Yumiko Yokomizo
- Department of Urology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Takashi Kawahara
- Department of Urology, Yokohama City University Graduate School of Medicine, Yokohama, Japan; Departments of Urology and Renal Transplantation, Yokohama City Medical Center, Yokohama, Japan.
| | - Yoji Nagashima
- Department of Pathology, Tokyo Woman Medical University, Tokyo, Japan
| | - Hitoshi Ishiguro
- Department of Urology, Yokohama City University Graduate School of Medicine, Yokohama, Japan; Photocatalyst Group, Kanagawa Academy of Science and Technology, Kawasaki, Japan
| | - Ikuma Kato
- Department of Pathology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Masahiro Yao
- Department of Urology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Hiroshi Miyamoto
- Departments of Pathology and Urology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Hiroji Uemura
- Departments of Urology and Renal Transplantation, Yokohama City Medical Center, Yokohama, Japan
| |
Collapse
|
29
|
Daunting but Worthy Goal: Reducing the De Novo Cancer Incidence After Transplantation. Transplantation 2017; 100:2569-2583. [PMID: 27861286 DOI: 10.1097/tp.0000000000001428] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Solid-organ transplant recipients are at increased risk of developing de novo malignancies compared with the general population, and malignancies become a major limitation in achieving optimal outcomes. The prevention and the management of posttransplant malignancies must be considered as a main goal in our transplant programs. For these patients, immunosuppression plays a major role in oncogenesis by both impairement of immunosurveillance, enhancement of chronic viral infection, and by direct prooncogenic effects. It is essential to manage the recipient with a long-term adapted screening program beginning before transplantation to use a prophylaxis to decrease infection-related cancer, to propose a viral monitoring, and to modulate the immunosuppression toward lower doses especially for calcineurin inhibitors. Indeed, strategies to induce tolerance or to allow a dramatic reduction of the immunosuppression burden are the more promising approaches for the reduction of the posttransplant malignancies.
Collapse
|
30
|
Kawahara T, Inoue S, Fujita K, Mizushima T, Ide H, Yamaguchi S, Fushimi H, Nonomura N, Miyamoto H. NFATc1 Expression as a Prognosticator in Urothelial Carcinoma of the Upper Urinary Tract. Transl Oncol 2017; 10:318-323. [PMID: 28327458 PMCID: PMC5358926 DOI: 10.1016/j.tranon.2017.01.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Revised: 01/30/2017] [Accepted: 01/31/2017] [Indexed: 11/30/2022] Open
Abstract
We recently found that NFATc1, a member of the NFAT family and a key regulator of the immune response, could induce bladder carcinogenesis and cancer progression. In this study, we immunohistochemically stained for NFATc1 in upper urinary tract urothelial carcinoma (UUTUC) specimens and paired nonneoplastic urothelial tissues. NFATc1 was positive in 51 [52%; 40 (40%) weak (1+), 9 (9%) moderate (2+), and 2 (2%) strong (3+)] of 99 UUTUCs, which was significantly higher than in benign urothelium [30 (36%) of 83; 28 (34%) weak and 2 (2%) moderate] (0 vs 1+/2+/3+, P=.038; 0/1+ vs 2+/3+, P=.023). There were no significant associations between NFATc1 expression pattern and tumor grade or pT stage. However, the positive rates of NFATc1 expression tended to be higher in renal pelvic tumors (60%) than in ureteral tumors (42%; P=.080) as well as in pN+ tumors (75%) than in pN0 tumors (49%; P=.089). Kaplan-Meier and log-rank tests revealed that moderate (2+) to strong (3+) NFATc1 expression correlated with lower progression-free survival (P=.032) and cancer-specific survival (P=.005) rates in the 99 cases. Patients with high (2+/3+) NFATc1 muscle-invasive tumor (n=9) also had a significantly higher risk of cancer-specific mortality (P=.021) compared to those with low (0/1+) NFATc1 muscle-invasive tumor (n=53). Thus, compared with nonneoplastic urothelium, a significant increase in the expression of NFATc1 in UUTUC was seen, implying the involvement of NFATc1 signals in the development of UUTUC. The current results further suggest that NFATc1 overexpression serves as a predictor of poor prognosis in patients with UUTUC.
Collapse
Affiliation(s)
- Takashi Kawahara
- Departments of Pathology and Urology, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Pathology & Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA; Departments of Urology and Renal Transplantation, Yokohama City University Medical Center, Yokohama, Japan
| | - Satoshi Inoue
- Departments of Pathology and Urology, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Pathology & Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA; James P. Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY, USA
| | - Kazutoshi Fujita
- Department of Urology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Taichi Mizushima
- Departments of Pathology and Urology, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Pathology & Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA; James P. Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY, USA
| | - Hiroki Ide
- Departments of Pathology and Urology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Seiji Yamaguchi
- Department of Urology, Osaka General Medical Center, Osaka, Japan
| | - Hiroaki Fushimi
- Department of Pathology, Osaka General Medical Center, Osaka, Japan
| | - Norio Nonomura
- Department of Urology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Hiroshi Miyamoto
- Departments of Pathology and Urology, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Pathology & Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA; James P. Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY, USA; Department of Urology, University of Rochester Medical Center, Rochester, NY, USA.
| |
Collapse
|
31
|
Ohtaka M, Kawahara T, Mochizuki T, Takamoto D, Hattori Y, Teranishi JI, Miyoshi Y, Yumura Y, Hasumi H, Yokomizo Y, Hayashi N, Kondo K, Yao M, Miyamoto H, Uemura H. RANK/RANKL expression in prostate cancer. Int J Surg Case Rep 2016; 30:106-107. [PMID: 28012322 PMCID: PMC5192013 DOI: 10.1016/j.ijscr.2016.11.042] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2016] [Accepted: 11/21/2016] [Indexed: 01/21/2023] Open
Abstract
Expression of RANK and RANKL genes in prostate cancer is higher than non-neoplastic prostate. RANK/RANKL expression is not related to pathological features. There is no significant correlation of RANK/RANKL expression with biochemical recurrence after radical prostatectomy.
Collapse
Affiliation(s)
- Mari Ohtaka
- Departments of Urology and Renal Transplantation, Yokohama City University Medical Center, Yokohama, Japan.
| | - Takashi Kawahara
- Departments of Urology and Renal Transplantation, Yokohama City University Medical Center, Yokohama, Japan; Department of Urology, Yokohama City University Graduate School of Medicine, Yokohama, Japan.
| | - Taku Mochizuki
- Departments of Urology and Renal Transplantation, Yokohama City University Medical Center, Yokohama, Japan.
| | - Daiji Takamoto
- Departments of Urology and Renal Transplantation, Yokohama City University Medical Center, Yokohama, Japan.
| | - Yusuke Hattori
- Departments of Urology and Renal Transplantation, Yokohama City University Medical Center, Yokohama, Japan.
| | - Jun-Ichi Teranishi
- Departments of Urology and Renal Transplantation, Yokohama City University Medical Center, Yokohama, Japan.
| | - Yasuhide Miyoshi
- Departments of Urology and Renal Transplantation, Yokohama City University Medical Center, Yokohama, Japan.
| | - Yasushi Yumura
- Departments of Urology and Renal Transplantation, Yokohama City University Medical Center, Yokohama, Japan.
| | - Hisashi Hasumi
- Department of Urology, Yokohama City University Graduate School of Medicine, Yokohama, Japan.
| | - Yumiko Yokomizo
- Department of Urology, Yokohama City University Graduate School of Medicine, Yokohama, Japan.
| | - Narihiko Hayashi
- Department of Urology, Yokohama City University Graduate School of Medicine, Yokohama, Japan.
| | - Keiichi Kondo
- Department of Urology, Yokohama City University Graduate School of Medicine, Yokohama, Japan.
| | - Masahiro Yao
- Department of Urology, Yokohama City University Graduate School of Medicine, Yokohama, Japan.
| | - Hiroshi Miyamoto
- Departments of Pathology and Urology, Johns Hopkins University School of Medicine, Baltimore, USA.
| | - Hiroji Uemura
- Departments of Urology and Renal Transplantation, Yokohama City University Medical Center, Yokohama, Japan.
| |
Collapse
|
32
|
Liu X, Zhang Y, Wang Z, Wang X, Zhu G, Han G, Chen G, Hou C, Wang T, Shen B, Li Y, Ma N, Xiao H, Wang R. Metabotropic glutamate receptor 3 is involved in B-cell-related tumor apoptosis. Int J Oncol 2016; 49:1469-78. [DOI: 10.3892/ijo.2016.3623] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 07/11/2016] [Indexed: 11/05/2022] Open
|
33
|
Tracking Cancer Genetic Evolution using OncoTrack. Sci Rep 2016; 6:29647. [PMID: 27412732 PMCID: PMC4944131 DOI: 10.1038/srep29647] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Accepted: 06/20/2016] [Indexed: 02/07/2023] Open
Abstract
It is difficult for existing methods to quantify, and track the constant evolution of cancers due to high heterogeneity of mutations. However, structural variations associated with nucleotide number changes show repeatable patterns in localized regions of the genome. Here we introduce SPKMG, which generalizes nucleotide number based properties of genes, in statistical terms, at the genome-wide scale. It is measured from the normalized amount of aligned NGS reads in exonic regions of a gene. SPKMG values are calculated within OncoTrack. SPKMG values being continuous numeric variables provide a statistical metric to track DNA level changes. We show that SPKMG measures of cancer DNA show a normative pattern at the genome-wide scale. The analysis leads to the discovery of core cancer genes and also provides novel dynamic insights into the stage of cancer, including cancer development, progression, and metastasis. This technique will allow exome data to also be used for quantitative LOH/CNV analysis for tracking tumour progression and evolution with a higher efficiency.
Collapse
|
34
|
Li L, Duan Z, Yu J, Dang HX. NFATc1 regulates cell proliferation, migration, and invasion of ovarian cancer SKOV3 cells in vitro and in vivo. Oncol Rep 2016; 36:918-28. [PMID: 27350254 DOI: 10.3892/or.2016.4904] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Accepted: 02/20/2016] [Indexed: 11/06/2022] Open
Abstract
NFATc1 (nuclear factor of activated T‑cells c1) is associated with malignancy in several cancer models. However, the expression and function of NFATc1 in ovarian cancer remain elusive. In the present study, we investigated the role of NFATc1 in human epithelial ovarian cancer (EOC) using human ovarian adenocarcinoma SKOV3 cells and patient characteristics. NFATc1 expression was silenced by siRNA in the SKOV3 ovarian cancer cell line and in human ovarian cancer nude mouse xenografts. Real‑time PCR, western blotting, immunohistochemical staining, MTT, flow cytometry, transwell, erasion trace and mouse assays were used to detect NFATc1 expression, cell proliferation, apoptosis, cell invasion and migration, tumor growth and angiogenesis. Survival analysis was performed to assess the correlation between NFATc1 expression and survival. NFATc1 was overexpressed in the SKOV3 ovarian cancer cell line and in human serous/mucinous ovarian cancer tissues. The silencing of NFATc1 expression by siRNA reduced cell proliferation and migration and promoted apoptosis in vitro and decreased the ovarian cancer cell tumorigenesis in vivo in nude mice. NFATc1 overexpression in high‑grade serous ovarian carcinomas was an independent prognostic factor of poor overall survival and of early relapse (P<0.01) in a univariate analysis. Our present data provide evidence that NFATc1 is overexpressed in human serous/mucinous ovarian cancer and is associated with a poor prognosis. NFATc1 silencing regulates the cell cycle, apoptosis, invasion and migration. NFATc1 thus has the potential to be a therapeutic target and to be used in EOC diagnosis and prognosis.
Collapse
Affiliation(s)
- Long Li
- Department of Physical Examination, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Zhaoning Duan
- Department of Gynecology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Jihui Yu
- Department of Physical Examination, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Hong-Xing Dang
- Department of PICU, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders; China International Science and Technology Cooperation base of Child development and Critical Disorders; Chongqing Engineering Research Center of Stem Cell Therapy, Chongqing 400016, P.R. China
| |
Collapse
|
35
|
Kawahara T, Aljarah AK, Shareef HK, Inoue S, Ide H, Patterson JD, Kashiwagi E, Han B, Li Y, Zheng Y, Miyamoto H. Silodosin inhibits prostate cancer cell growth via ELK1 inactivation and enhances the cytotoxic activity of gemcitabine. Prostate 2016; 76:744-56. [PMID: 26864615 DOI: 10.1002/pros.23164] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 01/22/2016] [Indexed: 11/06/2022]
Abstract
BACKGROUND Biological significance of ELK1, a transcriptional factor whose phosphorylation is necessary for c-fos proto-oncogene activation, in prostate cancer remains far from fully understood. In this study, we aim to investigate the role of ELK1 in tumor growth as well as the efficacy of a selective α1A-adrenergic blocker, silodosin, in ELK1 activity in prostate cancer cells. METHODS We first immunohistochemically determined the levels of phospho-ELK1 (p-ELK1) expression in radical prostatectomy specimens. We then assessed the effects of ELK1 knockdown via short hairpin RNA and silodosin on cell proliferation, migration, and invasion in prostate cancer lines. RESULTS The levels of p-ELK1 expression were significantly higher in carcinoma than in benign (P < 0.001) or high-grade prostatic intraepithelial neoplasia (HGPIN) (P = 0.002) as well as in HGPIN than in benign (P < 0.001). Kaplan-Meier and log-rank tests revealed that moderate-strong positivity of p-ELK1 in carcinomas tended to correlate with biochemical recurrence after radical prostatectomy (P = 0.098). In PC3 and DU145 expressing ELK1 (mRNA/protein) but no androgen receptor (AR), ELK1 silencing resulted in considerable decreases in the expression of c-fos as well as in cell migration/invasion and matrix metalloproteinase-2 expression, but not in cell viability. Silodosin treatment reduced the expression/activity of ELK1 in these cells as well as the viability of AR-positive LNCaP and C4-2 cells and the migration of both AR-positive and AR-negative cells, but not the viability of AR-negative or ELK1-negative cells. Interestingly, silodosin significantly increased sensitivity to gemcitabine, but not to cisplatin or docetaxel, even in AR-negative cells. CONCLUSIONS ELK1 is likely to be activated in prostate cancer cells and promote tumor progression. Furthermore, silodosin that inactivates ELK1 in prostate cancer cells not only inhibits their growth but also enhances the cytotoxic activity of gemcitabine. Thus, ELK1 inhibition has the potential of being a therapeutic approach for prostate cancer.
Collapse
Affiliation(s)
- Takashi Kawahara
- Departments of Pathology and Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, New York
- Department of Urology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Ali Kadhim Aljarah
- Departments of Pathology and Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Biology, University of Baghdad College of Science, Baghdad, Iraq
| | - Hasanain Khaleel Shareef
- Departments of Pathology and Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Biology, University of Babylon College of Science for Women, Babylon, Iraq
| | - Satoshi Inoue
- Departments of Pathology and Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Hiroki Ide
- Departments of Pathology and Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - John D Patterson
- Departments of Pathology and Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Eiji Kashiwagi
- Departments of Pathology and Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Bin Han
- Departments of Pathology and Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Yi Li
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, New York
| | - Yichun Zheng
- Departments of Pathology and Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, New York
| | - Hiroshi Miyamoto
- Departments of Pathology and Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, New York
| |
Collapse
|
36
|
Cell cycle and apoptosis regulation by NFAT transcription factors: new roles for an old player. Cell Death Dis 2016; 7:e2199. [PMID: 27100893 PMCID: PMC4855676 DOI: 10.1038/cddis.2016.97] [Citation(s) in RCA: 140] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Revised: 03/13/2016] [Accepted: 03/16/2016] [Indexed: 12/11/2022]
Abstract
The NFAT (nuclear factor of activated T cells) family of transcription factors consists of four Ca2+-regulated members (NFAT1–NFAT4), which were first described in T lymphocytes. In addition to their well-documented role in T lymphocytes, where they control gene expression during cell activation and differentiation, NFAT proteins are also expressed in a wide range of cells and tissue types and regulate genes involved in cell cycle, apoptosis, angiogenesis and metastasis. The NFAT proteins share a highly conserved DNA-binding domain (DBD), which allows all NFAT members to bind to the same DNA sequence in enhancers or promoter regions. The same DNA-binding specificity suggests redundant roles for the NFAT proteins, which is true during the regulation of some genes such as IL-2 and p21. However, it has become increasingly clear that different NFAT proteins and even isoforms can have unique functions. In this review, we address the possible reasons for these distinct roles, particularly regarding N- and C-terminal transactivation regions (TADs) and the partner proteins that interact with these TADs. We also discuss the genes regulated by NFAT during cell cycle regulation and apoptosis and the role of NFAT during tumorigenesis.
Collapse
|
37
|
NFATC1 promotes cell growth and tumorigenesis in ovarian cancer up-regulating c-Myc through ERK1/2/p38 MAPK signal pathway. Tumour Biol 2015; 37:4493-500. [DOI: 10.1007/s13277-015-4245-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2015] [Accepted: 10/12/2015] [Indexed: 12/11/2022] Open
|
38
|
Manda KR, Tripathi P, Hsi AC, Ning J, Ruzinova MB, Liapis H, Bailey M, Zhang H, Maher CA, Humphrey PA, Andriole GL, Ding L, You Z, Chen F. NFATc1 promotes prostate tumorigenesis and overcomes PTEN loss-induced senescence. Oncogene 2015; 35:3282-92. [PMID: 26477312 PMCID: PMC5012433 DOI: 10.1038/onc.2015.389] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Revised: 08/25/2015] [Accepted: 09/08/2015] [Indexed: 02/06/2023]
Abstract
Despite recent insights into prostate cancer (PCa)-associated genetic changes, full understanding of prostate tumorigenesis remains elusive due to complexity of interactions among various cell types and soluble factors present in prostate tissue. We found upregulation of Nuclear Factor of Activated T Cells c1 (NFATc1) in human PCa and cultured PCa cells, but not in normal prostates and non-tumorigenic prostate cells. To understand the role of NFATc1 in prostate tumorigenesis in situ, we temporally and spatially controlled the activation of NFATc1 in mouse prostate and showed that such activation resulted in prostatic adenocarcinoma with features similar to those seen in human PCa. Our results indicate that the activation of a single transcription factor, NFATc1 in prostatic luminal epithelium to PCa can affect expression of diverse factors in both cells harboring the genetic changes and in neighboring cells through microenvironmental alterations. In addition to the activation of oncogenes c-MYC and STAT3 in tumor cells, a number of cytokines and growth factors, such as IL1β, IL6, and SPP1 (Osteopontin, a key biomarker for PCa), were upregulated in NFATc1-induced PCa, establishing a tumorigenic microenvironment involving both NFATc1 positive and negative cells for prostate tumorigenesis. To further characterize interactions between genes involved in prostate tumorigenesis, we generated mice with both NFATc1 activation and Pten inactivation in prostate. We showed that NFATc1 activation led to acceleration of Pten-null–driven prostate tumorigenesis by overcoming the PTEN loss–induced cellular senescence through inhibition of p21 activation. This study provides direct in vivo evidence of an oncogenic role of NFATc1 in prostate tumorigenesis and reveals multiple functions of NFATc1 in activating oncogenes, in inducing proinflammatory cytokines, in oncogene addiction, and in overcoming cellular senescence, which suggests calcineurin-NFAT signaling as a potential target in preventing PCa.
Collapse
Affiliation(s)
- K R Manda
- Department of Medicine, Washington University, School of Medicine, St Louis, MO, USA
| | - P Tripathi
- Department of Pathology and Immunology, Washington University, St Louis, MO, USA
| | - A C Hsi
- The Genome Institute, Washington University, St Louis, MO, USA
| | - J Ning
- Department of Medicine, Washington University, School of Medicine, St Louis, MO, USA.,The Genome Institute, Washington University, St Louis, MO, USA
| | - M B Ruzinova
- Department of Pathology and Immunology, Washington University, St Louis, MO, USA
| | - H Liapis
- Department of Pathology and Immunology, Washington University, St Louis, MO, USA
| | - M Bailey
- The Genome Institute, Washington University, St Louis, MO, USA
| | - H Zhang
- Department of Cell and Developmental Biology, University of Massachusetts Medical School, Worcester, MA, USA
| | - C A Maher
- Department of Medicine, Washington University, School of Medicine, St Louis, MO, USA.,The Genome Institute, Washington University, St Louis, MO, USA.,Siteman Cancer Center, Washington University, St Louis, MO, USA
| | - P A Humphrey
- Department of Pathology, Yale University, New Haven, CT, USA
| | - G L Andriole
- Siteman Cancer Center, Washington University, St Louis, MO, USA.,Department of Surgery, Washington University, St Louis, MO, USA
| | - L Ding
- Department of Medicine, Washington University, School of Medicine, St Louis, MO, USA.,The Genome Institute, Washington University, St Louis, MO, USA.,Siteman Cancer Center, Washington University, St Louis, MO, USA
| | - Z You
- Department of Structural and Cellular Biology, Tulane University, New Orleans, LA, USA
| | - F Chen
- Department of Medicine, Washington University, School of Medicine, St Louis, MO, USA.,Siteman Cancer Center, Washington University, St Louis, MO, USA.,Department of Cell Biology and Physiology, Washington University, St Louis, MO, USA
| |
Collapse
|
39
|
Liersch-Löhn B, Slavova N, Buhr HJ, Bennani-Baiti IM. Differential protein expression and oncogenic gene network link tyrosine kinase ephrin B4 receptor to aggressive gastric and gastroesophageal junction cancers. Int J Cancer 2015; 138:1220-31. [PMID: 26414866 DOI: 10.1002/ijc.29865] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Revised: 08/29/2015] [Accepted: 09/16/2015] [Indexed: 12/18/2022]
Abstract
Transmembrane tyrosine-kinase Ephrin receptors promote tumor progression and/or metastasis of several malignancies including leukemia, follicular lymphoma, glioma, malignant pleural mesothelioma, papillary thyroid carcinoma, sarcomas and ovarian, breast, bladder and non-small cell lung cancers. They also drive intestinal stem cell proliferation and positioning, control intestinal tissue boundaries and are involved in liver, pancreatic and colorectal cancers, indicating involvement in additional digestive system malignancies. We investigated the role of Ephrin-B4 receptor (EPHB4), and its ligand EFNB2, in gastric and gastroesophageal junction cancers in patient cohorts through computational, mathematical, molecular and immunohistochemical analyses. We show that EPHB4 is upregulated in preneoplastic gastroesophageal lesions and its expression further increased in gastroesophageal cancers in several independent cohorts. The closely related EPHB6 receptor, which also binds EFNB2, was downregulated in all tested cohorts, consistent with its tumor-suppressive properties in other cancers. EFNB2 expression is induced in esophageal cells by acidity, suggesting that gastroesophageal reflux disease (GERD) may constitute an early triggering event in activating EFNB2-EPHB4 signaling. Association of EPHB4 to both Barrett's esophagus and to advanced tumor stages, and its overexpression at the tumor invasion front and vascular endothelial cells intimate the notion that EPHB4 may be associated with multiple steps of gastroesophageal tumorigenesis. Analysis of oncogenomic signatures uncovered the first EPHB4-associated gene network (false discovery rate: 7 × 10(-90) ) composed of a five-transcription factor interconnected gene network that drives proliferation, angiogenesis and invasiveness. The EPHB4 oncogenomic network provides a molecular basis for its role in tumor progression and points to EPHB4 as a potential tumor aggressiveness biomarker and drug target in gastroesophageal cancers.
Collapse
Affiliation(s)
- Britta Liersch-Löhn
- Department of Surgery, Sana Klinikum Lichtenberg Berlin, Berlin, Germany.,Department of General, Vascular and Thoracic Surgery, Charité Universitätsmedizin Berlin, Campus Benjamin Franklin, Berlin, Germany
| | - Nadia Slavova
- Department of General, Vascular and Thoracic Surgery, Charité Universitätsmedizin Berlin, Campus Benjamin Franklin, Berlin, Germany
| | - Heinz J Buhr
- Department of General, Vascular and Thoracic Surgery, Charité Universitätsmedizin Berlin, Campus Benjamin Franklin, Berlin, Germany.,German Society for General and Visceral Surgery, Haus Der Bundespressekonferenz, Berlin, Germany
| | | |
Collapse
|