1
|
Schrier I, Slotki-Itzchakov O, Elkis Y, Most-Menachem N, Adato O, Fitoussi-Allouche D, Shpungin S, Unger R, Nir U. Fer governs mTORC1 regulating pathways and sustains viability of pancreatic ductal adenocarcinoma cells. Front Oncol 2024; 14:1427029. [PMID: 39206154 PMCID: PMC11349523 DOI: 10.3389/fonc.2024.1427029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 07/29/2024] [Indexed: 09/04/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest cancers with a high percentage of morbidity. The deciphering and identification of novel targets and tools for intervening with its adverse progression are therefore of immense importance. To address this goal we adopted a specific inhibitor of the intracellular tyrosine kinase Fer, whose expression level is upregulated in PDAC tumors, and is associated with poor prognosis of patients. Subjecting PDAC cells to the E260-Fer inhibitor, unraveled its simultaneous effects on the mitochondria, and on a non-mitochondrial ERK1/2 regulatory cascade. E260 caused severe mitochondrial deformation, resulting in cellular- aspartate and ATP depletion, and followed by the activation of the metabolic sensor AMPK. This led to the phosphorylation and deactivation of the bona fide AMPK substrate, RAPTOR, which serves as a positive regulator of the mTORC1 metabolic hub. Accordingly, this resulted in the inhibition of the mTORC1 activity. In parallel, E260 downregulated the activation state of the ERK1/2 kinases, and their ability to neutralize the mTORC1 suppressor TSC2, thereby accentuating the inhibition of mTORC1. Importantly, both activation of AMPK and downregulation of ERK1/2 and mTORC1 were also achieved upon the knockdown of Fer, corroborating the regulatory role of Fer in these processes. Concomitantly, in PDAC tumors and not in healthy pancreatic tissues, the expression levels of Fer demonstrate moderate but statistically significant positive correlation with the expression levels of mTOR and its downstream effector LARP1. Finally, targeting the Fer driven activation of mTORC1, culminated in necrotic death of the treated PDAC cells, envisaging a new intervention tool for the challenging PDAC disease.
Collapse
Affiliation(s)
- Ilan Schrier
- Department of Surgery, Rabin Medical Center, Petah Tikva, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Orel Slotki-Itzchakov
- The Mina and Everard Goodman Faculty of Life-Sciences, Bar-Ilan University, Ramat-Gan, Israel
| | - Yoav Elkis
- The Mina and Everard Goodman Faculty of Life-Sciences, Bar-Ilan University, Ramat-Gan, Israel
| | - Nofar Most-Menachem
- The Mina and Everard Goodman Faculty of Life-Sciences, Bar-Ilan University, Ramat-Gan, Israel
| | - Orit Adato
- The Mina and Everard Goodman Faculty of Life-Sciences, Bar-Ilan University, Ramat-Gan, Israel
| | | | - Sally Shpungin
- The Mina and Everard Goodman Faculty of Life-Sciences, Bar-Ilan University, Ramat-Gan, Israel
| | - Ron Unger
- The Mina and Everard Goodman Faculty of Life-Sciences, Bar-Ilan University, Ramat-Gan, Israel
| | - Uri Nir
- The Mina and Everard Goodman Faculty of Life-Sciences, Bar-Ilan University, Ramat-Gan, Israel
| |
Collapse
|
2
|
Zhang Y, Xiong X, Sun R, Zhu X, Wang C, Jiang B, Yang X, Li D, Fan G. Development of the non-receptor tyrosine kinase FER-targeting PROTACs as a potential strategy for antagonizing ovarian cancer cell motility and invasiveness. J Biol Chem 2023:104825. [PMID: 37196766 DOI: 10.1016/j.jbc.2023.104825] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 04/23/2023] [Accepted: 05/07/2023] [Indexed: 05/19/2023] Open
Abstract
Aberrant overexpression of non-receptor tyrosine kinase FER has been reported in various ovarian carcinoma-derived tumor cells and is a poor prognosis factor for patient survival. It plays an essential role in tumor cell migration and invasion, acting concurrently in both kinase-dependent and -independent manners, which is not easily suppressed by conventional enzymatic inhibitors. Nevertheless, the proteolysis-targeting chimeras (PROTACs) technology offers superior efficacy over traditional activity-based inhibitors by simultaneously targeting enzymatic and scaffold functions. Hence in this study, we report the development of two PROTAC compounds that promote robust FER degradation in a cereblon-dependent manner. Both PROTAC degraders outperform an FDA-approved drug, Brigatinib, in ovarian cancer cell motility suppression. Importantly, these PROTAC compounds also degrade multiple oncogenic FER fusion proteins identified in human tumor samples. These results lay an experimental foundation to apply the PROTAC strategy to antagonize cell motility and invasiveness in ovarian and other types of cancers with aberrant expression of FER kinase and highlight PROTACs as a superior strategy for targeting proteins with multiple tumor-promoting functions.
Collapse
Affiliation(s)
- Yanchun Zhang
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Xuexue Xiong
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Renhong Sun
- Gluetacs Therapeutics (Shanghai) Co., Ltd., Shanghai, China
| | - Xiaotong Zhu
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Chen Wang
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Biao Jiang
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, China
| | - Xiaobao Yang
- Gluetacs Therapeutics (Shanghai) Co., Ltd., Shanghai, China.
| | - Dake Li
- Department of Gynecology, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, China.
| | - Gaofeng Fan
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China.
| |
Collapse
|
3
|
Yang CZ, Guo W, Wang YF, Hu LH, Wang J, Luo JM, Yao XH, Liu S, Tao LT, Sun LL, Lin LZ. Reduction in gefitinib resistance mediated by Yi-Fei San-Jie pill in non-small cell lung cancer through regulation of tyrosine metabolism, cell cycle, and the MET/EGFR signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2023; 314:116566. [PMID: 37169317 DOI: 10.1016/j.jep.2023.116566] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 04/16/2023] [Accepted: 04/29/2023] [Indexed: 05/13/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The Chinese herbal prescription Yi-Fei San-Jie pill (YFSJ) has been used for adjuvant treatment in patients with lung cancer for a long time. AIM OF THE STUDY Reports have indicated that the combination of gefitinib (Gef) with YFSJ inhibits the proliferation of EGFR-TKI-resistant cell lines by enhancing cellular apoptosis and autophagy in non-small cell lung cancer (NSCLC). However, the molecular mechanisms underlying the effect of YFSJ on EGFR-TKI resistance and related metabolic pathways remain to be explored. MATERIALS AND METHODS In our report, ultra-performance liquid chromatography-tandem mass spectrometry (UPLC-MS/MS), metabolomics, network pharmacology, bioinformatics, and biological analysis methods were used to investigate the mechanism. RESULTS The UPLC-MS/MS data identified 42 active compounds of YFSJ extracts. YFSJ extracts can enhance the antitumor efficacy of Gef without hepatic and renal toxicity in vivo. The analysis of the metabolomics pathway enrichment revealed that YFSJ mainly affected the tyrosine metabolism pathway in rat models. Moreover, YFSJ has been shown to reverse Gef resistance and improve the effects of Gef on the cellular viability, migration capacity, and cell cycle arrest of NSCLC cell lines with EGFR mutations. The results of network pharmacology and molecular docking analyses revealed that tyrosine metabolism-related active compounds of YFSJ affect EGFR-TKIs resistance in NSCLC by targeting cell cycle and the MET/EGFR signaling pathway; these findings were validated by western blotting and immunohistochemistry. CONCLUSIONS YFSJ inhibits NSCLC by inducing cell cycle arrest in the G1/S phase to suppress tumor growth, cell viability, and cell migration through synergistic effects with Gef via the tyrosine metabolic pathway and the EGFR/MET signaling pathway. To summarize, the findings of the current study indicate that YFSJ is a prospective complementary treatment for Gef-resistant NSCLC.
Collapse
Affiliation(s)
- Cai-Zhi Yang
- The First School of Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
| | - Wei Guo
- Department of Oncology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
| | - Yi-Fan Wang
- The First School of Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
| | - Lei-Hao Hu
- The First School of Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
| | - Jing Wang
- State Key Laboratory of Quality Research in Chinese Medicines, Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, 999078, China.
| | - Jia-Min Luo
- The First School of Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
| | - Xiao-Hui Yao
- Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
| | - Shan Liu
- The First School of Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
| | - Lan-Ting Tao
- Department of Oncology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, 510120, China.
| | - Ling-Ling Sun
- Department of Oncology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
| | - Li-Zhu Lin
- Department of Oncology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
| |
Collapse
|
4
|
Yasir M, Park J, Han ET, Park WS, Han JH, Kwon YS, Lee HJ, Hassan M, Kloczkowski A, Chun W. Exploration of Flavonoids as Lead Compounds against Ewing Sarcoma through Molecular Docking, Pharmacogenomics Analysis, and Molecular Dynamics Simulations. MOLECULES (BASEL, SWITZERLAND) 2023; 28:molecules28010414. [PMID: 36615603 PMCID: PMC9823950 DOI: 10.3390/molecules28010414] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 12/28/2022] [Accepted: 12/28/2022] [Indexed: 01/05/2023]
Abstract
Ewing sarcoma (ES) is a highly malignant carcinoma prevalent in children and most frequent in the second decade of life. It mostly occurs due to t(11;22) (q24;q12) translocation. This translocation encodes the oncogenic fusion protein EWS/FLI (Friend leukemia integration 1 transcription factor), which acts as an aberrant transcription factor to deregulate target genes essential for cancer. Traditionally, flavonoids from plants have been investigated against viral and cancerous diseases and have shown some promising results to combat these disorders. In the current study, representative flavonoid compounds from various subclasses are selected and used to disrupt the RNA-binding motif of EWS, which is required for EWS/FLI fusion. By blocking the RNA-binding motif of EWS, it might be possible to combat ES. Therefore, molecular docking experiments validated the binding interaction patterns and structural behaviors of screened flavonoid compounds within the active region of the Ewing sarcoma protein (EWS). Furthermore, pharmacogenomics analysis was used to investigate potential drug interactions with Ewing sarcoma-associated genes. Finally, molecular dynamics simulations were used to investigate the stability of the best selected docked complexes. Taken together, daidzein, kaempferol, and genistein exhibited a result comparable to ifosfamide in the proposed in silico study and can be further analyzed as possible candidate compounds in biological in vitro studies against ES.
Collapse
Affiliation(s)
- Muhammad Yasir
- Department of Pharmacology, Kangwon National University School of Medicine, Chuncheon 24341, Republic of Korea
| | - Jinyoung Park
- Department of Pharmacology, Kangwon National University School of Medicine, Chuncheon 24341, Republic of Korea
| | - Eun-Taek Han
- Department of Medical Environmental Biology and Tropical Medicine, Kangwon National University School of Medicine, Chuncheon 24341, Republic of Korea
| | - Won Sun Park
- Department of Physiology, Kangwon National University School of Medicine, Chuncheon 24341, Republic of Korea
| | - Jin-Hee Han
- Department of Medical Environmental Biology and Tropical Medicine, Kangwon National University School of Medicine, Chuncheon 24341, Republic of Korea
| | - Yong-Soo Kwon
- College of Pharmacy, Kangwon National University School of Medicine, Chuncheon 24341, Republic of Korea
| | - Hee-Jae Lee
- Department of Pharmacology, Kangwon National University School of Medicine, Chuncheon 24341, Republic of Korea
| | - Mubashir Hassan
- The Steve and Cindy Rasmussen Institute for Genomic Medicine at Nationwide Children’s Hospital, Columbus, OH 43205, USA
| | - Andrzej Kloczkowski
- The Steve and Cindy Rasmussen Institute for Genomic Medicine at Nationwide Children’s Hospital, Columbus, OH 43205, USA
| | - Wanjoo Chun
- Department of Pharmacology, Kangwon National University School of Medicine, Chuncheon 24341, Republic of Korea
- Correspondence: ; Tel.: +82-33-250-8853
| |
Collapse
|
5
|
Elsocht M, Giron P, De Grève J, Ballet S. Second generation Spautin-1 analogues targeting EGFR-mutant non-small cell lung cancer cells. Bioorg Med Chem Lett 2023; 79:129066. [PMID: 36410591 DOI: 10.1016/j.bmcl.2022.129066] [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: 06/21/2022] [Revised: 09/29/2022] [Accepted: 11/09/2022] [Indexed: 11/19/2022]
Abstract
Treatment of advanced stage epidermal growth factor receptor (EGFR)-mutant non-small cell lung cancer (NSCLC) is often complicated by the occurrence of acquired resistance, which emphasizes the need for improved treatment options. Based on a previously reported structure-activity relationship (SAR) study of Spautin-1, which resulted in the discovery of 10a, the search for more potent analogues was envisaged through optimization of the amine substituent. Our search led to the discovery of analogue 15b, harbouring the 2-[4-(4-fluoro-phenoxy)-phenyl]ethylamine substituent, among other potent and original analogues, with nanomolar activity towards EGFR-mutant NSCLC cells. Moreover, this compound 15b showed good selectivity for cancer cells over healthy lung epithelial cells and provides additive effects with food and drug administration (FDA) approved EGFR-tyrosine kinase inhibitors (TKIs), as proven by the co-administration of 15b with Afatinib. Altogether, we report promising lead compounds which show the potential to improve current treatment options.
Collapse
Affiliation(s)
- Mathias Elsocht
- Research Group of Organic Chemistry, Faculty of Sciences and Bioengineering Sciences, Vrije Universiteit Brussel, Pleinlaan 2, 1050 Brussels, Belgium.
| | - Philippe Giron
- Laboratory of Medical and Molecular Oncology and Center of Medical Genetics, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium.
| | - Jacques De Grève
- Laboratory of Medical and Molecular Oncology and Center of Medical Genetics, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium.
| | - Steven Ballet
- Research Group of Organic Chemistry, Faculty of Sciences and Bioengineering Sciences, Vrije Universiteit Brussel, Pleinlaan 2, 1050 Brussels, Belgium.
| |
Collapse
|
6
|
Wang M, Banik I, Shain AH, Yeh I, Bastian BC. Integrated genomic analyses of acral and mucosal melanomas nominate novel driver genes. Genome Med 2022; 14:65. [PMID: 35706047 PMCID: PMC9202124 DOI: 10.1186/s13073-022-01068-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 06/03/2022] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND Acral and mucosal melanomas are aggressive subtypes of melanoma, which have a significantly lower burden of somatic mutations than cutaneous melanomas, but more frequent copy number variations, focused gene amplifications, and structural alterations. The landscapes of their genomic alterations remain to be fully characterized. METHODS We compiled sequencing data of 240 human acral and mucosal melanoma samples from 11 previously published studies and applied a uniform pipeline to call tumor cell content, ploidy, somatic and germline mutations, as well as CNVs, LOH, and SVs. We identified genes that are significantly mutated or recurrently affected by CNVs and implicated in oncogenesis. We further examined the difference in the frequency of recurrent pathogenic alterations between the two melanoma subtypes, correlation between pathogenic alterations, and their association with clinical features. RESULTS We nominated PTPRJ, mutated and homozygously deleted in 3.8% (9/240) and 0.8% (2/240) of samples, respectively, as a probable tumor suppressor gene, and FER and SKP2, amplified in 3.8% and 11.7% of samples, respectively, as probable oncogenes. We further identified a long tail of infrequent pathogenic alterations, involving genes such as CIC and LZTR1. Pathogenic germline mutations were observed on MITF, PTEN, ATM, and PRKN. We found BRAF V600E mutations in acral melanomas with fewer structural variations, suggesting that they are distinct and related to cutaneous melanomas. Amplifications of PAK1 and GAB2 were more commonly observed in acral melanomas, whereas SF3B1 R625 codon mutations were unique to mucosal melanomas (12.9%). Amplifications at 11q13-14 were frequently accompanied by fusion to a region on chromosome 6q12, revealing a recurrent novel structural rearrangement whose role remains to be elucidated. CONCLUSIONS Our meta-analysis expands the catalog of driver mutations in acral and mucosal melanomas, sheds new light on their pathogenesis and broadens the catalog of therapeutic targets for these difficult-to-treat cancers.
Collapse
Affiliation(s)
- Meng Wang
- Department of Dermatology, University of California San Francisco, San Francisco, CA, USA
- Helen Diller Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
| | - Ishani Banik
- Department of Dermatology, University of California San Francisco, San Francisco, CA, USA
- Helen Diller Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
| | - A Hunter Shain
- Department of Dermatology, University of California San Francisco, San Francisco, CA, USA
- Helen Diller Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
| | - Iwei Yeh
- Department of Dermatology, University of California San Francisco, San Francisco, CA, USA.
- Helen Diller Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA.
- Department of Pathology, University of California San Francisco, San Francisco, CA, USA.
| | - Boris C Bastian
- Department of Dermatology, University of California San Francisco, San Francisco, CA, USA.
- Helen Diller Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA.
- Department of Pathology, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
7
|
Tavares S, Liv N, Pasolli M, Opdam M, Rätze MAK, Saornil M, Sluimer LM, Hengeveld RCC, van Es R, van Werkhoven E, Vos H, Rehmann H, Burgering BMT, Oosterkamp HM, Lens SMA, Klumperman J, Linn SC, Derksen PWB. FER regulates endosomal recycling and is a predictor for adjuvant taxane benefit in breast cancer. Cell Rep 2022; 39:110584. [PMID: 35385742 DOI: 10.1016/j.celrep.2022.110584] [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: 04/12/2021] [Revised: 10/28/2021] [Accepted: 03/07/2022] [Indexed: 11/18/2022] Open
Abstract
Elevated expression of non-receptor tyrosine kinase FER is an independent prognosticator that correlates with poor survival of high-grade and basal/triple-negative breast cancer (TNBC) patients. Here, we show that high FER levels are also associated with improved outcomes after adjuvant taxane-based combination chemotherapy in high-risk, HER2-negative patients. In TNBC cells, we observe a causal relation between high FER levels and sensitivity to taxanes. Proteomics and mechanistic studies demonstrate that FER regulates endosomal recycling, a microtubule-dependent process that underpins breast cancer cell invasion. Using chemical genetics, we identify DCTN2 as a FER substrate. Our work indicates that the DCTN2 tyrosine 6 is essential for the development of tubular recycling domains in early endosomes and subsequent propagation of TNBC cell invasion in 3D. In conclusion, we show that high FER expression promotes endosomal recycling and represents a candidate predictive marker for the benefit of adjuvant taxane-containing chemotherapy in high-risk patients, including TNBC patients.
Collapse
Affiliation(s)
- Sandra Tavares
- Department of Pathology, University Medical Center Utrecht, 3584CX Utrecht, the Netherlands
| | - Nalan Liv
- Section Cell Biology, Center for Molecular Medicine, University Medical Center Utrecht, 3584CX Utrecht, the Netherlands
| | - Milena Pasolli
- Cell Biology, Neurobiology, and Biophysics, Department of Biology, Faculty of Science, Utrecht University, 3584CH Utrecht, the Netherlands
| | - Mark Opdam
- Department of Molecular Pathology, Netherlands Cancer Institute, 1066CX Amsterdam, the Netherlands
| | - Max A K Rätze
- Department of Pathology, University Medical Center Utrecht, 3584CX Utrecht, the Netherlands
| | - Manuel Saornil
- Department of Pathology, University Medical Center Utrecht, 3584CX Utrecht, the Netherlands
| | - Lilian M Sluimer
- Department of Pathology, University Medical Center Utrecht, 3584CX Utrecht, the Netherlands
| | - Rutger C C Hengeveld
- Oncode Institute, Department of Molecular Cancer Research, University Medical Center Utrecht, 3584CX Utrecht, the Netherlands
| | - Robert van Es
- Oncode Institute, Department of Molecular Cancer Research, University Medical Center Utrecht, 3584CX Utrecht, the Netherlands
| | - Erik van Werkhoven
- Department of Molecular Pathology, Netherlands Cancer Institute, 1066CX Amsterdam, the Netherlands
| | - Harmjan Vos
- Oncode Institute, Department of Molecular Cancer Research, University Medical Center Utrecht, 3584CX Utrecht, the Netherlands
| | - Holger Rehmann
- Flensburg University of Applied Sciences, 24943 Flensburg, Germany
| | - Boudewijn M T Burgering
- Oncode Institute, Department of Molecular Cancer Research, University Medical Center Utrecht, 3584CX Utrecht, the Netherlands
| | - Hendrika M Oosterkamp
- Department of Medical Oncology, Haaglanden Medisch Centrum, 2501 CK The Hague, the Netherlands
| | - Susanne M A Lens
- Oncode Institute, Department of Molecular Cancer Research, University Medical Center Utrecht, 3584CX Utrecht, the Netherlands
| | - Judith Klumperman
- Section Cell Biology, Center for Molecular Medicine, University Medical Center Utrecht, 3584CX Utrecht, the Netherlands
| | - Sabine C Linn
- Department of Pathology, University Medical Center Utrecht, 3584CX Utrecht, the Netherlands; Department of Molecular Pathology, Netherlands Cancer Institute, 1066CX Amsterdam, the Netherlands; Department of Medical Oncology, Netherlands Cancer Institute, 1066CX Amsterdam, the Netherlands
| | - Patrick W B Derksen
- Department of Pathology, University Medical Center Utrecht, 3584CX Utrecht, the Netherlands.
| |
Collapse
|
8
|
Ding W, Fan Y, Jia W, Pan X, Han G, Zhang Y, Chen Z, Lu Y, Wang J, Wu J, Wang X. FER Regulated by miR-206 Promotes Hepatocellular Carcinoma Progression via NF-κB Signaling. Front Oncol 2021; 11:683878. [PMID: 34295819 PMCID: PMC8289706 DOI: 10.3389/fonc.2021.683878] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 06/21/2021] [Indexed: 12/09/2022] Open
Abstract
Objectives Feline sarcoma-related protein (FER) is known to play a critical regulatory role in several carcinomas. However, the exact biological function of FER in hepatocellular carcinoma (HCC) still needs to be investigated. The primary objective of this research was to investigate the unknown function and molecular mechanisms of FER in HCC. Materials and Methods The expression level of FER in HCC tissue samples and cells was examined by RT-qPCR, immunohistochemistry and western blot. Cellular and animal experiments were used to explore the effect of FER on the proliferative and metastatic capacities of HCC cells. The crosstalk between FER and NF-κB signaling was explored by western blot. The upstream factors that regulate FER were evaluated through dual-luciferase experiments and western blot assays. Results FER was overexpressed in HCC specimens and HCC cell lines. FER expression levels were positively associated with unfavorable clinicopathological characteristics. The higher the expression of FER was, the worse the overall survival of HCC patients was. The results of loss-of-function and gain-of-function experiments indicated that knockdown of FER decreased, while overexpression of FER increased, the proliferation, invasion and metastasis of HCC cells in vitro and in vivo. Mechanistically, we found that FER activated the NF-κB signaling pathway and stimulated epithelial-to-mesenchymal transition (EMT). We also found that FER was directly regulated by miR-206, and the downregulation of miR-206 was associated with proliferation and metastatic progression in HCC. Conclusions The present research was the first to reveal that a decrease in miR-206 levels results in an increase in FER expression in HCC, leading to enhanced cell growth and metastatic abilities via activation of the NF-κB signaling pathway.
Collapse
Affiliation(s)
- Wenzhou Ding
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, National Health Commission (NHC) Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, China
| | - Ye Fan
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, National Health Commission (NHC) Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, China
| | - Wenbo Jia
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, National Health Commission (NHC) Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, China
| | - Xiongxiong Pan
- Department of Anesthesiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Guoyong Han
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, National Health Commission (NHC) Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, China
| | - Yao Zhang
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, National Health Commission (NHC) Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, China
| | - Zhiqiang Chen
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, National Health Commission (NHC) Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, China
| | - Yiwei Lu
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, National Health Commission (NHC) Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, China
| | - Jinyi Wang
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, National Health Commission (NHC) Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, China
| | - Jindao Wu
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, National Health Commission (NHC) Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, China
| | - Xuehao Wang
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, National Health Commission (NHC) Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, China
| |
Collapse
|
9
|
Loss of Fer Jeopardizes Metabolic Plasticity and Mitochondrial Homeostasis in Lung and Breast Carcinoma Cells. Int J Mol Sci 2021; 22:ijms22073387. [PMID: 33806191 PMCID: PMC8037256 DOI: 10.3390/ijms22073387] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 03/18/2021] [Accepted: 03/22/2021] [Indexed: 12/18/2022] Open
Abstract
Metabolic plasticity is a hallmark of the ability of metastatic cancer cells to survive under stressful conditions. The intracellular Fer kinase is a selective constituent of the reprogramed mitochondria and metabolic system of cancer cells. In the current work, we deciphered the modulatory roles of Fer in the reprogrammed metabolic systems of metastatic, lung (H358), non-small cell lung cancer (NSCLC), and breast (MDA-MB-231), triple-negative breast cancer (TNBC), carcinoma cells. We show that H358 cells devoid of Fer (H358ΔFer), strictly depend on glucose for their proliferation and growth, and fail to compensate for glucose withdrawal by oxidizing and metabolizing glutamine. Furthermore, glucose deficiency caused increased reactive oxygen species (ROS) production and induction of a DNA damage response (DDR), accompanied by the onset of apoptosis and attenuated cell-cycle progression. Analysis of mitochondrial function revealed impaired respiratory and electron transport chain (ETC) complex 1 (comp. I) activity in the Fer-deficient H358ΔFer cells. This was manifested by decreased levels of NAD+ and ATP and relatively low abundance of tricarboxylic acid (TCA) cycle metabolites. Impaired electron transport chain comp. I activity and dependence on glucose were also confirmed in Fer-deficient, MDA-MB-231ΔFer cells. Although both H358ΔFer and MDA-MB-231ΔFer cells showed a decreased aspartate level, this seemed to be compensated by the predominance of pyrimidines synthesis over the urea cycle progression. Notably, absence of Fer significantly impeded the growth of H358ΔFer and MDA-MB-231ΔFer xenografts in mice provided with a carb-deficient, ketogenic diet. Thus, Fer plays a key role in the sustention of metabolic plasticity of malignant cells. In compliance with this notion, targeting Fer attenuates the progression of H358 and MDA-MB-231 tumors, an effect that is potentiated by a glucose-restrictive diet.
Collapse
|
10
|
Marciano O, Mehazri L, Shpungin S, Varvak A, Zacksenhaus E, Nir U. Fer and FerT Govern Mitochondrial Susceptibility to Metformin and Hypoxic Stress in Colon and Lung Carcinoma Cells. Cells 2021; 10:cells10010097. [PMID: 33430475 PMCID: PMC7826929 DOI: 10.3390/cells10010097] [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: 11/25/2020] [Revised: 12/26/2020] [Accepted: 01/05/2021] [Indexed: 12/25/2022] Open
Abstract
Aerobic glycolysis is an important metabolic adaptation of cancer cells. However, there is growing evidence that reprogrammed mitochondria also play an important metabolic role in metastatic dissemination. Two constituents of the reprogrammed mitochondria of cancer cells are the intracellular tyrosine kinase Fer and its cancer- and sperm-specific variant, FerT. Here, we show that Fer and FerT control mitochondrial susceptibility to therapeutic and hypoxic stress in metastatic colon (SW620) and non-small cell lung cancer (NSCLC-H1299) cells. Fer- and FerT-deficient SW620 and H1299 cells (SW∆Fer/FerT and H∆Fer/FerT cells, respectively) become highly sensitive to metformin treatment and to hypoxia under glucose-restrictive conditions. Metformin impaired mitochondrial functioning that was accompanied by ATP deficiency and robust death in SW∆Fer/FerT and H∆Fer/FerT cells compared to the parental SW620 and H1299 cells. Notably, selective knockout of the fer gene without affecting FerT expression reduced sensitivity to metformin and hypoxia seen in SW∆Fer/FerT cells. Thus, Fer and FerT modulate the mitochondrial susceptibility of metastatic cancer cells to hypoxia and metformin. Targeting Fer/FerT may therefore provide a novel anticancer treatment by efficient, selective, and more versatile disruption of mitochondrial function in malignant cells.
Collapse
Affiliation(s)
- Odeya Marciano
- The Mina and Everard Goodman Faculty of Life-Sciences, Bar-Ilan University, Ramat-Gan 5290002, Israel; (O.M.); (L.M.); (S.S.); (A.V.)
| | - Linoy Mehazri
- The Mina and Everard Goodman Faculty of Life-Sciences, Bar-Ilan University, Ramat-Gan 5290002, Israel; (O.M.); (L.M.); (S.S.); (A.V.)
| | - Sally Shpungin
- The Mina and Everard Goodman Faculty of Life-Sciences, Bar-Ilan University, Ramat-Gan 5290002, Israel; (O.M.); (L.M.); (S.S.); (A.V.)
| | - Alexander Varvak
- The Mina and Everard Goodman Faculty of Life-Sciences, Bar-Ilan University, Ramat-Gan 5290002, Israel; (O.M.); (L.M.); (S.S.); (A.V.)
| | - Eldad Zacksenhaus
- Laboratory of Medicine & Pathology, University of Toronto, Toronto, ON M5G 2M1, Canada;
| | - Uri Nir
- The Mina and Everard Goodman Faculty of Life-Sciences, Bar-Ilan University, Ramat-Gan 5290002, Israel; (O.M.); (L.M.); (S.S.); (A.V.)
- Correspondence: ; Tel.: +972-52-4416968
| |
Collapse
|
11
|
Zhong C, Yu J, Li D, Jiang K, Tang Y, Yang M, Shen H, Fang X, Ding K, Zheng S, Yuan Y. Zyxin as a potential cancer prognostic marker promotes the proliferation and metastasis of colorectal cancer cells. J Cell Physiol 2019; 234:15775-15789. [PMID: 30697742 DOI: 10.1002/jcp.28236] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 01/08/2019] [Accepted: 01/10/2019] [Indexed: 01/24/2023]
Abstract
Colorectal cancer (CRC) is a leading cause of cancer death. This study was conducted to investigate the functions and mechanisms of Zyxin (ZYX) in CRC. Multiomics analysis associated ZYX with CRC metastasis. ZYX expression levels were increased in human CRC tissues and related to shorter recurrence-free survival. Knockdown of ZYX expression resulted in inhibition of cell growth, invasion, and migration in vitro and in vivo. Comprehensive analysis of gene microarray analysis showed that ZYX may activate the pathway of NUPR1 and JNK, inhibit CST5, regulate focal adhesion (FA), and affect epithelial-mesenchymal transition in CRC cells. Results of gene microarray and membrane protein isobaric tags with relative and absolute quantitation labeling mass spectrometry found ten differentially expressed genes, which were associated with ZYX activity. Furthermore, real-time polymerase chain reaction was used to validate the expression patterns of selected genes in the integrative analysis. Taken together, our findings provide the first evidence that decreased expression level of ZYX impairs CRC cell proliferation and metastasis probably via the FA pathway.
Collapse
Affiliation(s)
- Chenhan Zhong
- Department of Medical Oncology, (Key Laboratory of Cancer Prevention and Intervention, Chinese National Ministry of Education; Key Laboratory of Molecular Biology in Medical Sciences) The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Jiekai Yu
- Cancer Institute, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Dan Li
- Department of Medical Oncology, (Key Laboratory of Cancer Prevention and Intervention, Chinese National Ministry of Education; Key Laboratory of Molecular Biology in Medical Sciences) The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Kai Jiang
- Department of Surgical Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yang Tang
- Department of Surgical Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Mengyuan Yang
- Department of Medical Oncology, (Key Laboratory of Cancer Prevention and Intervention, Chinese National Ministry of Education; Key Laboratory of Molecular Biology in Medical Sciences) The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Hong Shen
- Department of Medical Oncology, (Key Laboratory of Cancer Prevention and Intervention, Chinese National Ministry of Education; Key Laboratory of Molecular Biology in Medical Sciences) The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xuefeng Fang
- Department of Medical Oncology, (Key Laboratory of Cancer Prevention and Intervention, Chinese National Ministry of Education; Key Laboratory of Molecular Biology in Medical Sciences) The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Kefeng Ding
- Department of Surgical Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Shu Zheng
- Cancer Institute, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Research Center for Air Pollution and Health, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Ying Yuan
- Department of Medical Oncology, (Key Laboratory of Cancer Prevention and Intervention, Chinese National Ministry of Education; Key Laboratory of Molecular Biology in Medical Sciences) The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Cancer Institute, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| |
Collapse
|
12
|
Matsuura Y. High-resolution structural analysis shows how different crystallographic environments can induce alternative modes of binding of a phosphotyrosine peptide to the SH2 domain of Fer tyrosine kinase. Protein Sci 2019; 28:2011-2019. [PMID: 31441171 DOI: 10.1002/pro.3713] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 08/16/2019] [Accepted: 08/20/2019] [Indexed: 11/08/2022]
Abstract
Fes and Fes-related (Fer) protein tyrosine kinases (PTKs) comprise a subfamily of nonreceptor tyrosine kinases characterized by a unique multidomain structure composed of an N-terminal Fer/CIP4 homology-Bin/Amphiphysin/Rvs (F-BAR) domain, a central Src homology 2 (SH2) domain, and a C-terminal PTK domain. Fer is ubiquitously expressed, and upregulation of Fer has been implicated in various human cancers. The PTK activity of Fes has been shown to be positively regulated by the binding of phosphotyrosine-containing ligands to the SH2 domain. Here, the X-ray crystal structure of human Fer SH2 domain bound to a phosphopeptide that has D-E-pY-E-N-V-D sequence is reported at 1.37 å resolution. The asymmetric unit (ASU) contains six Fer-phosphopeptide complexes, and the structure reveals three distinct binding modes for the same phosphopeptide. At four out of the six binding sites in the ASU, the phosphopeptide binds to Fer SH2 domain in a type I β-turn conformation, and this could be the optimal binding mode of this phosphopeptide. At the other two binding sites in the ASU, it appears that spatial proximity of neighboring SH2 domains in the crystal induces alternative modes of binding of this phosphopeptide.
Collapse
Affiliation(s)
- Yoshiyuki Matsuura
- Division of Biological Science, Graduate School of Science, Nagoya University, Nagoya, Japan.,Structural Biology Research Center, Graduate School of Science, Nagoya University, Nagoya, Japan
| |
Collapse
|
13
|
YY1 inhibits the migration and invasion of pancreatic ductal adenocarcinoma by downregulating the FER/STAT3/MMP2 signaling pathway. Cancer Lett 2019; 463:37-49. [PMID: 31404611 DOI: 10.1016/j.canlet.2019.07.019] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 07/29/2019] [Accepted: 07/29/2019] [Indexed: 02/07/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) has a poor prognosis and a high mortality rate. The transcription factor YY1 acts as an inhibitor of many types of tumors. We found that YY1 knockdown promoted the invasion and migration of PANC-1 and BxPC-3 cells; FER knockdown partially restored the promotion of pancreatic cancer caused by YY1 knockdown. In vivo experiments yielded the same results. According to luciferase reporter gene, electrophoretic mobility shift (EMSA) and chromatin immunoprecipitation (ChIP) assays, YY1 directly binds to the FER promoter region. Moreover, higher level FER expression results in a worse TNM stage and prognosis for patients with PDAC. Furthermore, by downregulating FER, YY1 inhibits the formation of the STAT3-MMP2 complex, thereby suppressing expression of MMP2 and ultimately inhibiting the migration and invasion of pancreatic cancer. Our study demonstrates that the YY1/FER/STAT3/MMP2 axis is associated with the progression of pancreatic cancer and may provide a new therapeutic target for the treatment of pancreatic cancer.
Collapse
|
14
|
Hu X, Guo Z, Xu J, Mei X, Bi M, Jiang F, Yu D, Zhong C. Role of feline sarcoma‑related protein in the viability and apoptosis of bladder cancer cells. Mol Med Rep 2019; 19:5219-5226. [PMID: 31059042 DOI: 10.3892/mmr.2019.10204] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2017] [Accepted: 11/19/2018] [Indexed: 11/06/2022] Open
Abstract
Feline sarcoma‑related protein (Fer) is a type of nuclear and cytoplasmic non‑receptor protein tyrosine kinase, which is associated with the progression of numerous types of cancer. Previously, we identified that Fer is associated with the migration and invasion of bladder cancer. The present study aimed to investigate the role of Fer in bladder cancer cell viability and apoptosis. Reverse transcription‑quantitative polymerase chain reaction and western blot analysis were performed to detect the expression levels of Fer; short interference RNA (siRNA) and overexpression vectors were used to downregulate or upregulate Fer expression, respectively. The effects on cell proliferation ability and cell apoptosis were then tested by MTT assay and flow cytometry. The results revealed that Fer expression was upregulated in bladder cancer cell lines. Downregulation of Fer expression by siRNA significantly suppressed T24 cell viability and induced apoptosis, as well as inducing cell cycle arrest. Conversely, Fer overexpression in 5637 cells significantly promoted cell viability and cell cycle progression, but inhibited cell apoptosis. Furthermore, the suppression and overexpression of Fer significantly altered the expression of cleaved caspase‑3 and Bcl‑2, and dysregulated the P38 mitogen‑activated protein kinase signaling pathway. The findings of the present study indicate a possible molecular mechanism of Fer in bladder cancer and may be considered as a potential target in the treatment of this disease.
Collapse
Affiliation(s)
- Xudong Hu
- Department of Urology, Anqing First People's Hospital, Anqing, Anhui 246000, P.R. China
| | - Zhiwen Guo
- Department of Urology, Anqing First People's Hospital, Anqing, Anhui 246000, P.R. China
| | - Jifei Xu
- Department of Urology, Anqing First People's Hospital, Anqing, Anhui 246000, P.R. China
| | - Xiangbao Mei
- Department of Urology, Anqing First People's Hospital, Anqing, Anhui 246000, P.R. China
| | - Manhua Bi
- Department of Urology, Anqing First People's Hospital, Anqing, Anhui 246000, P.R. China
| | - Fang Jiang
- Department of Urology, Anqing First People's Hospital, Anqing, Anhui 246000, P.R. China
| | - Dexin Yu
- Department of Urology, The Second Hospital of Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Caiyun Zhong
- Department of Nutrition and Food Safety, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, P.R. China
| |
Collapse
|
15
|
Targeting FER Kinase Inhibits Melanoma Growth and Metastasis. Cancers (Basel) 2019; 11:cancers11030419. [PMID: 30909648 PMCID: PMC6468679 DOI: 10.3390/cancers11030419] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 03/12/2019] [Accepted: 03/21/2019] [Indexed: 12/28/2022] Open
Abstract
Melanoma is one of the most aggressive types of tumors and exhibits high metastatic potential. Fes-related (FER) kinase is a non-receptor tyrosine kinase that has been implicated in growth and metastasis of various epithelial tumors. In this study, we have examined the role that FER kinase plays in melanoma at the molecular level. FER-depleted melanoma cells exhibit impaired Wnt/β-catenin pathway activity, as well as multiple proteomic changes, which include decreased abundance of L1-cell adhesion molecule (L1-CAM). Consistent with the pro-metastatic functions of these pathways, we demonstrate that depletion of FER kinase decreases melanoma growth and formation of distant metastases in a xenograft model. These findings indicate that FER is an important positive regulator of melanoma metastasis and a potential target for innovative therapies.
Collapse
|
16
|
Watt K, Newsted D, Voorand E, Gooding RJ, Majewski A, Truesdell P, Yao B, Tuschl T, Renwick N, Craig AW. MicroRNA-206 suppresses TGF-β signalling to limit tumor growth and metastasis in lung adenocarcinoma. Cell Signal 2018; 50:25-36. [PMID: 29935234 DOI: 10.1016/j.cellsig.2018.06.008] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 06/18/2018] [Accepted: 06/18/2018] [Indexed: 12/19/2022]
Abstract
MicroRNA-206 (miR-206) has demonstrated tumor suppressive effects in a variety of cancers. Numerous studies have identified aberrantly expressed targets of miR-206 that contribute to tumor progression and metastasis, however, the broader gene-networks and pathways regulated by miR-206 remain poorly defined. Here, we have ectopically expressed miR-206 in lung adenocarcinoma cell lines and tumors to identify differentially expressed genes, and study the effects on tumor growth and metastasis. In H1299 tumor xenograft assays, stable expression of miR-206 suppressed both tumor growth and metastasis in mice. Profiling of xenograft tumors using small RNA sequencing and a targeted panel of tumor progression and metastasis-related genes revealed a network of genes involved in TGF-β signalling that were regulated by miR-206. Among these were the TGFB1 ligand, as well as direct transcriptional targets of Smad3. Other differentially expressed genes included components of the extracellular matrix involved in TGF-β activation and signalling, including Thrombospondin-1, which is responsible for the activation of latent TGF-β in the stroma. In cultured lung adenocarcinoma cells treated with recombinant TGF-β, ectopic expression of miR-206 impaired canonical signalling, and expression of TGF-β target genes linked to epithelial-mesenchymal transition. This was due at least in part to the suppression of Smad3 protein levels in lung adenocarcinoma cells with ectopic miR-206 expression. Together, these findings indicate that miR-206 can suppress tumor progression and metastasis by limiting autocrine production of TGF-β, and highlight the potential utility of TGF-β inhibitors for the treatment of lung adenocarcinomas.
Collapse
Affiliation(s)
- Kathleen Watt
- Cancer Biology & Genetics Division, Queen's Cancer Research Institute, Kingston, Canada; Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Canada
| | - Daniel Newsted
- Cancer Biology & Genetics Division, Queen's Cancer Research Institute, Kingston, Canada; Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Canada
| | - Elena Voorand
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Canada
| | - Robert J Gooding
- Cancer Biology & Genetics Division, Queen's Cancer Research Institute, Kingston, Canada; Department of Physics, Queen's University, Kingston, Canada; Department of Pathology and Molecular Medicine, Queen's University, Kingston, Canada
| | - Adrianna Majewski
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Canada; Department of Pathology and Molecular Medicine, Queen's University, Kingston, Canada
| | - Peter Truesdell
- Cancer Biology & Genetics Division, Queen's Cancer Research Institute, Kingston, Canada; Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Canada
| | - Binchen Yao
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Canada
| | - Thomas Tuschl
- HHMI Laboratory of RNA Molecular Biology, The Rockefeller University, New York, USA
| | - Neil Renwick
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, Canada; HHMI Laboratory of RNA Molecular Biology, The Rockefeller University, New York, USA
| | - Andrew W Craig
- Cancer Biology & Genetics Division, Queen's Cancer Research Institute, Kingston, Canada; Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Canada.
| |
Collapse
|
17
|
Bhattacharya P, Shetake NG, Pandey BN, Kumar A. Receptor tyrosine kinase signaling in cancer radiotherapy and its targeting for tumor radiosensitization. Int J Radiat Biol 2018; 94:628-644. [DOI: 10.1080/09553002.2018.1478160] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Poushali Bhattacharya
- Radiation Signaling and Cancer Biology Section, Radiation Biology and Health Sciences Division, Bhabha Atomic Research Centre, Mumbai, India
| | - Neena G. Shetake
- Radiation Signaling and Cancer Biology Section, Radiation Biology and Health Sciences Division, Bhabha Atomic Research Centre, Mumbai, India
| | - Badri N. Pandey
- Radiation Signaling and Cancer Biology Section, Radiation Biology and Health Sciences Division, Bhabha Atomic Research Centre, Mumbai, India
| | - Amit Kumar
- Radiation Signaling and Cancer Biology Section, Radiation Biology and Health Sciences Division, Bhabha Atomic Research Centre, Mumbai, India
| |
Collapse
|
18
|
Dolgachev V, Panicker S, Balijepalli S, McCandless LK, Yin Y, Swamy S, Suresh MV, Delano MJ, Hemmila MR, Raghavendran K, Machado-Aranda D. Electroporation-mediated delivery of FER gene enhances innate immune response and improves survival in a murine model of pneumonia. Gene Ther 2018; 25:359-375. [PMID: 29907877 PMCID: PMC6195832 DOI: 10.1038/s41434-018-0022-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 05/02/2018] [Accepted: 05/11/2018] [Indexed: 12/23/2022]
Abstract
Previously, we reported that electroporation-mediated (EP) delivery of the FER gene improved survival in a combined trauma-pneumonia model. The mechanism of this protective effect is unknown. In this paper, we performed a pneumonia model in C57/BL6 mice with 500 CFU of Klebsiella pneumoniae. After inoculation, a plasmid encoding human FER was delivered by EP into the lung (PNA/pFER-EP). Survival of FER-treated vs. controls (PNA; PNA/EP-pcDNA) was recorded. In parallel cohorts, bronchial alveolar lavage (BAL) and lung were harvested at 24 and 72 h with markers of infection measured. FER-EP-treated animals reduced bacterial counts and had better 5-day survival compared to controls (80 vs. 20 vs. 25%; p < 0.05). Pre-treatment resulted in 100% survival. With FER, inflammatory monocytes were quickly recruited into BAL. These cells had increased surface expression for Toll-receptor 2 and 4, and increased phagocytic and myeloperoxidase activity at 24 h. Samples from FER electroporated animals had increased phosphorylation of STAT transcription factors, varied gene expression of IL1β, TNFα, Nrf2, Nlrp3, Cxcl2, HSP90 and increased cytokine production of TNF-α, CCL-2, KC, IFN-γ, and IL-1RA. In a follow-up experiment, using Methicillin-resistant Staphylococcus aureus (MRSA) similar bacterial reduction effects were obtained with FER gene delivery. We conclude that FER overexpression improves survival through STAT activation enhancing innate immunity and accelerating bacterial clearance in the lung. This constitutes a novel mechanism of inflammatory regulation with therapeutic potential in the setting of hospital-acquired pneumonia.
Collapse
Affiliation(s)
- Vladislav Dolgachev
- Division of Acute Care Surgery, University of Michigan, 1500 E Medical Center Dr, UH-1C421, SPC 5033, Ann Arbor, MI, 48109-5033, USA
| | - Sreehari Panicker
- Division of Acute Care Surgery, University of Michigan, 1500 E Medical Center Dr, UH-1C421, SPC 5033, Ann Arbor, MI, 48109-5033, USA
| | - Sanjay Balijepalli
- Division of Acute Care Surgery, University of Michigan, 1500 E Medical Center Dr, UH-1C421, SPC 5033, Ann Arbor, MI, 48109-5033, USA
| | - Lane Kelly McCandless
- Division of Acute Care Surgery, University of Michigan, 1500 E Medical Center Dr, UH-1C421, SPC 5033, Ann Arbor, MI, 48109-5033, USA
| | - Yue Yin
- Division of Acute Care Surgery, University of Michigan, 1500 E Medical Center Dr, UH-1C421, SPC 5033, Ann Arbor, MI, 48109-5033, USA
| | - Samantha Swamy
- Division of Acute Care Surgery, University of Michigan, 1500 E Medical Center Dr, UH-1C421, SPC 5033, Ann Arbor, MI, 48109-5033, USA
| | - M V Suresh
- Division of Acute Care Surgery, University of Michigan, 1500 E Medical Center Dr, UH-1C421, SPC 5033, Ann Arbor, MI, 48109-5033, USA
| | - Matthew J Delano
- Division of Acute Care Surgery, University of Michigan, 1500 E Medical Center Dr, UH-1C421, SPC 5033, Ann Arbor, MI, 48109-5033, USA
| | - Mark R Hemmila
- Division of Acute Care Surgery, University of Michigan, 1500 E Medical Center Dr, UH-1C421, SPC 5033, Ann Arbor, MI, 48109-5033, USA
| | - Krishnan Raghavendran
- Division of Acute Care Surgery, University of Michigan, 1500 E Medical Center Dr, UH-1C421, SPC 5033, Ann Arbor, MI, 48109-5033, USA
| | - David Machado-Aranda
- Division of Acute Care Surgery, University of Michigan, 1500 E Medical Center Dr, UH-1C421, SPC 5033, Ann Arbor, MI, 48109-5033, USA.
| |
Collapse
|
19
|
Hu J, Meng Y, Zeng J, Zeng B, Jiang X. Ubiquitin E3 Ligase MARCH7 promotes proliferation and invasion of cervical cancer cells through VAV2-RAC1-CDC42 pathway. Oncol Lett 2018; 16:2312-2318. [PMID: 30008934 PMCID: PMC6036418 DOI: 10.3892/ol.2018.8908] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 10/19/2017] [Indexed: 12/25/2022] Open
Abstract
Ubiquitin E3 Ligase MARCH7 is involved in T cell proliferation and neuronal development. In our previous study, we demonstrated MARCH7 promoted malignant behavior of ovarian cancer via the nuclear factor (NF)-κB and Wnt/β-catenin signaling pathway. However, the expression and function of MARCH7 in cervical cancer remains unknown. The present study aimed to unravel the expression and function of MARCH7 in cervical cancer to elucidate its potential role in the diagnosis and pathogenesis of cervical cancer. Results indicated that the expression of MARCH7 was abnormally high in cervical cancer tissues than normal cervical tissues. However, silencing the expression of MARCH7 in HeLa cells resulted in decreased cell proliferation and invasion. Mechanistic investigations revealed that MARCH7 interacted with VAV2. Silencing the expression of MARCH7 in HeLa cells inhibited the VAV2-RAC1-CDC42 signaling pathway. Overall, the results of the present study identified MARCH7 as a candidate oncogene in cervical cancer, and a potential target for cervical cancer therapy.
Collapse
Affiliation(s)
- Jianguo Hu
- Department of Obstetrics and Gynecology, Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, P.R. China
| | - Ying Meng
- Department of Obstetrics and Gynecology, Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, P.R. China
| | - Jianhua Zeng
- Department of Obstetrics and Gynecology, Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, P.R. China
| | - Biao Zeng
- Department of Obstetrics and Gynecology, Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, P.R. China
| | - Xingwei Jiang
- Department of Obstetrics and Gynecology, Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, P.R. China
| |
Collapse
|
20
|
Stanicka J, Rieger L, O’Shea S, Cox O, Coleman M, O’Flanagan C, Addario B, McCabe N, Kennedy R, O’Connor R. FES-related tyrosine kinase activates the insulin-like growth factor-1 receptor at sites of cell adhesion. Oncogene 2018. [DOI: 10.1038/s41388-017-0113-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
21
|
Fan G. FER mediated HGF-independent regulation of HGFR/MET activates RAC1-PAK1 pathway to potentiate metastasis in ovarian cancer. Small GTPases 2018; 11:155-159. [PMID: 29099290 DOI: 10.1080/21541248.2017.1379931] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Abstract
Uncontrolled metastasis significantly contributes to high lethality of patients suffering from ovarian cancer. To date, the detailed molecular mechanisms which account for ovarian tumor cell spreading and metastasis remain largely unknown. In a recent study, we have demonstrated that aberrantly high expression of the non-receptor tyrosine kinase FER is responsible for ovarian tumor cell metastasis both in vitro and in vivo. Mechanistically, we indentified Hepatocyte Growth Factor Receptor HGFR/MET as a novel substrate of FER, and through which the kinase FER modulates ovarian cancer cell motility and invasiveness in a ligand-independent manner. We also observed aberrantly high expression of PAK1 kinase in cancer cells, and RNAi-mediated knockdown of FER kinase inactivated the RAC1-PAK1 signaling pathway and decreased metastatic potential of CAOV4 ovarian cancer cells. Overall, our study revealed a previously uncharacterized, pro-metastatic role of the kinase FER in ovarian cancer through the MET-RAC1-PAK1 pathway. Further efforts are essential to investigating beneficial outcomes towards targeting the RAC1-PAK1 signaling pathway in reducing metastatic burden of this deadly disease.
Collapse
Affiliation(s)
- Gaofeng Fan
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| |
Collapse
|
22
|
Sponges: A Reservoir of Genes Implicated in Human Cancer. Mar Drugs 2018; 16:md16010020. [PMID: 29320389 PMCID: PMC5793068 DOI: 10.3390/md16010020] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 12/21/2017] [Accepted: 01/04/2018] [Indexed: 12/22/2022] Open
Abstract
Recently, it was shown that the majority of genes linked to human diseases, such as cancer genes, evolved in two major evolutionary transitions—the emergence of unicellular organisms and the transition to multicellularity. Therefore, it has been widely accepted that the majority of disease-related genes has already been present in species distantly related to humans. An original way of studying human diseases relies on analyzing genes and proteins that cause a certain disease using model organisms that belong to the evolutionary level at which these genes have emerged. This kind of approach is supported by the simplicity of the genome/proteome, body plan, and physiology of such model organisms. It has been established for quite some time that sponges are an ideal model system for such studies, having a vast variety of genes known to be engaged in sophisticated processes and signalling pathways associated with higher animals. Sponges are considered to be the simplest multicellular animals and have changed little during evolution. Therefore, they provide an insight into the metazoan ancestor genome/proteome features. This review compiles current knowledge of cancer-related genes/proteins in marine sponges.
Collapse
|
23
|
Elkis Y, Cohen M, Yaffe E, Satmary-Tusk S, Feldman T, Hikri E, Nyska A, Feiglin A, Ofran Y, Shpungin S, Nir U. A novel Fer/FerT targeting compound selectively evokes metabolic stress and necrotic death in malignant cells. Nat Commun 2017; 8:940. [PMID: 29038547 PMCID: PMC5643328 DOI: 10.1038/s41467-017-00832-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Accepted: 07/28/2017] [Indexed: 12/31/2022] Open
Abstract
Disruption of the reprogrammed energy management system of malignant cells is a prioritized goal of targeted cancer therapy. Two regulators of this system are the Fer kinase, and its cancer cell specific variant, FerT, both residing in subcellular compartments including the mitochondrial electron transport chain. Here, we show that a newly developed inhibitor of Fer and FerT, E260, selectively evokes metabolic stress in cancer cells by imposing mitochondrial dysfunction and deformation, and onset of energy-consuming autophagy which decreases the cellular ATP level. Notably, Fer was also found to associate with PARP-1 and E260 disrupted this association thereby leading to PARP-1 activation. The cooperative intervention with these metabolic pathways leads to energy crisis and necrotic death in malignant, but not in normal human cells, and to the suppression of tumors growth in vivo. Thus, E260 is a new anti-cancer agent which imposes metabolic stress and cellular death in cancer cells. The tyrosine-kinases Fer/FerT associate with the mitochondrial electron transport chain in cancer cells supporting their metabolic reprogramming. Here the authors discover a compound that disrupts Fer /FerT activity and selectively induces cell death of cancer cell lines displaying anti-tumor activity in vivo.
Collapse
Affiliation(s)
- Yoav Elkis
- The Mina and Everard Goodman Faculty of Life-Sciences, Bar-Ilan University, Ramat-Gan, 52900, Israel
| | - Moshe Cohen
- The Mina and Everard Goodman Faculty of Life-Sciences, Bar-Ilan University, Ramat-Gan, 52900, Israel
| | - Etai Yaffe
- The Mina and Everard Goodman Faculty of Life-Sciences, Bar-Ilan University, Ramat-Gan, 52900, Israel
| | - Shirly Satmary-Tusk
- The Mina and Everard Goodman Faculty of Life-Sciences, Bar-Ilan University, Ramat-Gan, 52900, Israel
| | - Tal Feldman
- The Mina and Everard Goodman Faculty of Life-Sciences, Bar-Ilan University, Ramat-Gan, 52900, Israel
| | - Elad Hikri
- The Mina and Everard Goodman Faculty of Life-Sciences, Bar-Ilan University, Ramat-Gan, 52900, Israel
| | - Abraham Nyska
- Consultant in Toxicological Pathology, Timrat, and Sackler School of Medicine, Tel Aviv University, Tel Aviv, 36576, Israel
| | - Ariel Feiglin
- The Mina and Everard Goodman Faculty of Life-Sciences, Bar-Ilan University, Ramat-Gan, 52900, Israel
| | - Yanay Ofran
- The Mina and Everard Goodman Faculty of Life-Sciences, Bar-Ilan University, Ramat-Gan, 52900, Israel
| | - Sally Shpungin
- The Mina and Everard Goodman Faculty of Life-Sciences, Bar-Ilan University, Ramat-Gan, 52900, Israel
| | - Uri Nir
- The Mina and Everard Goodman Faculty of Life-Sciences, Bar-Ilan University, Ramat-Gan, 52900, Israel.
| |
Collapse
|
24
|
Chen ZH, Yu YP, Tao J, Liu S, Tseng G, Nalesnik M, Hamilton R, Bhargava R, Nelson JB, Pennathur A, Monga SP, Luketich JD, Michalopoulos GK, Luo JH. MAN2A1-FER Fusion Gene Is Expressed by Human Liver and Other Tumor Types and Has Oncogenic Activity in Mice. Gastroenterology 2017; 153:1120-1132.e15. [PMID: 28245430 PMCID: PMC5572118 DOI: 10.1053/j.gastro.2016.12.036] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Revised: 12/16/2016] [Accepted: 12/23/2016] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Human tumors and liver cancer cell lines express the product of a fusion between the first 13 exons in the mannosidase α class 2A member 1 gene (MAN2A1) and the last 6 exons in the FER tyrosine kinase gene (FER), called MAN2A1-FER. We investigated whether MAN2A1-FER is expressed by human liver tumors and its role in liver carcinogenesis. METHODS We performed reverse transcription polymerase chain reaction analyses of 102 non-small cell lung tumors, 61 ovarian tumors, 70 liver tumors, 156 glioblastoma multiform samples, 27 esophageal adenocarcinomas, and 269 prostate cancer samples, as well as 10 nontumor liver tissues and 20 nontumor prostate tissues, collected at the University of Pittsburgh. We also measured expression by 15 human cancer cell lines. We expressed a tagged form of MAN2A1-FER in NIH3T3 and HEP3B (liver cancer) cells; Golgi were isolated for analysis. MAN2A1-FER was also overexpressed in PC3 or DU145 (prostate cancer), NIH3T3 (fibroblast), H23 (lung cancer), and A-172 (glioblastoma multiforme) cell lines and knocked out in HUH7 (liver cancer) cells. Cells were analyzed for proliferation and in invasion assays, and/or injected into flanks of severe combined immunodeficient mice; xenograft tumor growth and metastasis were assessed. Mice with hepatic deletion of PTEN were given tail-vein injections of MAN2A1-FER. RESULTS We detected MAN2A1-FER messenger RNA and fusion protein (114 kD) in the hepatocellular carcinoma cell line HUH7, as well as in liver tumors, esophageal adenocarcinoma, glioblastoma multiforme, prostate tumors, non-small cell lung tumors, and ovarian tumors, but not nontumor prostate or liver tissues. MAN2A1-FER protein retained the signal peptide for Golgi localization from MAN2A1 and translocated from the cytoplasm to Golgi in cancer cell lines. MAN2A1-FER had tyrosine kinase activity almost 4-fold higher than that of wild-type FER, and phosphorylated the epidermal growth factor receptor at tyrosine 88 in its N-terminus. Expression of MAN2A1-FER in 4 cell lines led to epidermal growth factor receptor activation of BRAF, MEK, and AKT; HUH7 cells with MAN2A1-FER knockout had significant decreases in phosphorylation of these proteins. Cell lines that expressed MAN2A1-FER had increased proliferation, colony formation, and invasiveness and formed larger (>2-fold) xenograft tumors in mice, with more metastases, than cells not expressing the fusion protein. HUH7 cells with MAN2A1-FER knockout formed smaller xenograft tumors, with fewer metastases, than control HUH7 cells. HUH7, A-172, and PC3 cells that expressed MAN2A1-FER were about 2-fold more sensitive to the FER kinase inhibitor crizotinib and the epidermal growth factor receptor kinase inhibitor canertinib; these drugs slowed growth of xenograft tumors from MAN2A1-FER cells and prevented their metastasis in mice. Hydrodynamic tail-vein injection of MAN2A1-FER resulted in rapid development of liver cancer in mice with hepatic disruption of Pten. CONCLUSIONS Many human tumor types and cancer cell lines express the MAN2A1-FER fusion, which increases proliferation and invasiveness of cancer cell lines and has liver oncogenic activity in mice.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents/pharmacology
- Cell Line, Tumor
- Cell Movement
- Cell Proliferation
- Cell Transformation, Neoplastic/genetics
- Cell Transformation, Neoplastic/metabolism
- Cell Transformation, Neoplastic/pathology
- Crizotinib
- Dose-Response Relationship, Drug
- Enzyme Activation
- ErbB Receptors/genetics
- ErbB Receptors/metabolism
- Gene Expression Regulation, Enzymologic
- Gene Expression Regulation, Neoplastic
- Gene Fusion
- Golgi Apparatus/enzymology
- Humans
- Liver Neoplasms/drug therapy
- Liver Neoplasms/enzymology
- Liver Neoplasms/genetics
- Liver Neoplasms/pathology
- Mice
- Mice, Knockout
- Mice, SCID
- Morpholines/pharmacology
- NIH 3T3 Cells
- Neoplasm Invasiveness
- Neoplasm Transplantation
- Oncogene Proteins, Fusion/antagonists & inhibitors
- Oncogene Proteins, Fusion/genetics
- Oncogene Proteins, Fusion/metabolism
- Oncogenes
- PTEN Phosphohydrolase/deficiency
- PTEN Phosphohydrolase/genetics
- Phosphorylation
- Protein Kinase Inhibitors/pharmacology
- Protein-Tyrosine Kinases/antagonists & inhibitors
- Protein-Tyrosine Kinases/genetics
- Protein-Tyrosine Kinases/metabolism
- Pyrazoles/pharmacology
- Pyridines/pharmacology
- RNA Interference
- Time Factors
- Transfection
- Tumor Burden
- alpha-Mannosidase/antagonists & inhibitors
- alpha-Mannosidase/genetics
- alpha-Mannosidase/metabolism
Collapse
Affiliation(s)
- Zhang-Hui Chen
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Yan P Yu
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Junyan Tao
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Silvia Liu
- Department of Biostatistics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - George Tseng
- Department of Biostatistics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Michael Nalesnik
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Ronald Hamilton
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Rohit Bhargava
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Joel B Nelson
- Department of Urology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Arjun Pennathur
- Department of Cardiothoracic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Satdarshan P Monga
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - James D Luketich
- Department of Cardiothoracic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - George K Michalopoulos
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Jian-Hua Luo
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| |
Collapse
|
25
|
Fan G, Zhang S, Gao Y, Greer PA, Tonks NK. HGF-independent regulation of MET and GAB1 by nonreceptor tyrosine kinase FER potentiates metastasis in ovarian cancer. Genes Dev 2017; 30:1542-57. [PMID: 27401557 PMCID: PMC4949327 DOI: 10.1101/gad.284166.116] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Accepted: 06/07/2016] [Indexed: 12/29/2022]
Abstract
In this study, Fan et al. report a novel ligand- and autophosphorylation-independent activation of MET through the nonreceptor tyrosine kinase FER. The findings show that levels of FER were elevated in ovarian cancer cell lines and that loss of FER impaired the metastasis of ovarian cancer cells in vivo, providing new insights into signaling events that underlie metastasis in ovarian cancer cells. Ovarian cancer cells disseminate readily within the peritoneal cavity, which promotes metastasis, and are often resistant to chemotherapy. Ovarian cancer patients tend to present with advanced disease, which also limits treatment options; consequently, new therapies are required. The oncoprotein tyrosine kinase MET, which is the receptor for hepatocyte growth factor (HGF), has been implicated in ovarian tumorigenesis and has been the subject of extensive drug development efforts. Here, we report a novel ligand- and autophosphorylation-independent activation of MET through the nonreceptor tyrosine kinase feline sarcoma-related (FER). We demonstrated that the levels of FER were elevated in ovarian cancer cell lines relative to those in immortalized normal surface epithelial cells and that suppression of FER attenuated the motility and invasive properties of these cancer cells. Furthermore, loss of FER impaired the metastasis of ovarian cancer cells in vivo. Mechanistically, we demonstrated that FER phosphorylated a signaling site in MET: Tyr1349. This enhanced activation of RAC1/PAK1 and promoted a kinase-independent scaffolding function that led to recruitment and phosphorylation of GAB1 and the specific activation of the SHP2–ERK signaling pathway. Overall, this analysis provides new insights into signaling events that underlie metastasis in ovarian cancer cells, consistent with a prometastatic role of FER and highlighting its potential as a novel therapeutic target for metastatic ovarian cancer.
Collapse
Affiliation(s)
- Gaofeng Fan
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724, USA
| | - Siwei Zhang
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724, USA; Department of Molecular Genetics and Microbiology, Stony Brook University, Stony Brook, New York 11794, USA
| | - Yan Gao
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario K7L3N6, Canada
| | - Peter A Greer
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario K7L3N6, Canada
| | - Nicholas K Tonks
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724, USA
| |
Collapse
|
26
|
Rosenberg BJ, Gil-Henn H, Mader CC, Halo T, Yin T, Condeelis J, Machida K, Wu YI, Koleske AJ. Phosphorylated cortactin recruits Vav2 guanine nucleotide exchange factor to activate Rac3 and promote invadopodial function in invasive breast cancer cells. Mol Biol Cell 2017; 28:1347-1360. [PMID: 28356423 PMCID: PMC5426849 DOI: 10.1091/mbc.e16-12-0885] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Revised: 03/20/2017] [Accepted: 03/22/2017] [Indexed: 12/12/2022] Open
Abstract
Phosphorylation of cortactin downstream of the EGF receptor–Src-Arg kinase cascade triggers maturation of invadopodia, actin-rich protrusions that breast cancer cells use to invade the extracellular matrix. Phosphocortactin recruits Vav2 to invadopodia to activate Rac3 and support actin polymerization, matrix degradation, and invasion. Breast carcinoma cells use specialized, actin-rich protrusions called invadopodia to degrade and invade through the extracellular matrix. Phosphorylation of the actin nucleation–promoting factor and actin-stabilizing protein cortactin downstream of the epidermal growth factor receptor–Src-Arg kinase cascade is known to be a critical trigger for invadopodium maturation and subsequent cell invasion in breast cancer cells. The functions of cortactin phosphorylation in this process, however, are not completely understood. We identify the Rho-family guanine nucleotide exchange factor Vav2 in a comprehensive screen for human SH2 domains that bind selectively to phosphorylated cortactin. We demonstrate that the Vav2 SH2 domain binds selectively to phosphotyrosine-containing peptides corresponding to cortactin tyrosines Y421 and Y466 but not to Y482. Mutation of the Vav2 SH2 domain disrupts its recruitment to invadopodia, and an SH2-domain mutant form of Vav2 cannot support efficient matrix degradation in invasive MDA-MB-231 breast cancer cells. We show that Vav2 function is required for promoting invadopodium maturation and consequent actin polymerization, matrix degradation, and invasive migratory behavior. Using biochemical assays and a novel Rac3 biosensor, we show that Vav2 promotes Rac3 activation at invadopodia. Rac3 knockdown reduces matrix degradation by invadopodia, whereas a constitutively active Rac3 can rescue the deficits in invadopodium function in Vav2-knockdown cells. Together these data indicate that phosphorylated cortactin recruits Vav2 to activate Rac3 and promote invadopodial maturation in invasive breast cancer cells.
Collapse
Affiliation(s)
| | - Hava Gil-Henn
- Faculty of Medicine in the Galilee, Bar-Ilan University, Safed 1311520, Israel
| | | | - Tiffany Halo
- Department of Chemistry, Yale University, New Haven, CT 06520
| | - Taofei Yin
- Raymond and Beverly Sackler Laboratory of Genetics and Molecular Medicine, Department of Genetics and Genome Sciences and Center for Cell Analysis and Modeling, University of Connecticut School of Medicine, Farmington, CT 06030
| | - John Condeelis
- Department of Anatomy and Structural Biology and Gruss Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY 10461
| | - Kazuya Machida
- Raymond and Beverly Sackler Laboratory of Genetics and Molecular Medicine, Department of Genetics and Genome Sciences and Center for Cell Analysis and Modeling, University of Connecticut School of Medicine, Farmington, CT 06030
| | - Yi I Wu
- Raymond and Beverly Sackler Laboratory of Genetics and Molecular Medicine, Department of Genetics and Genome Sciences and Center for Cell Analysis and Modeling, University of Connecticut School of Medicine, Farmington, CT 06030
| | - Anthony J Koleske
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06520 .,Department of Neuroscience, Yale University, New Haven, CT 06520
| |
Collapse
|
27
|
Mitsunari K, Miyata Y, Watanabe SI, Asai A, Yasuda T, Kanda S, Sakai H. Stromal expression of Fer suppresses tumor progression in renal cell carcinoma and is a predictor of survival. Oncol Lett 2016; 13:834-840. [PMID: 28356966 DOI: 10.3892/ol.2016.5481] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2015] [Accepted: 10/21/2016] [Indexed: 11/06/2022] Open
Abstract
Fps/Fes related (Fer) is a non-receptor tyrosine kinase that is expressed in fibroblasts, immune cells and endothelial cells. Fer serves an important pathological role in cell survival, angiogenesis and the immune system. However, the pathological role of Fer expression in the stromal cells surrounding renal cell carcinoma (RCC) has not been previously investigated. In the present study, immunohistochemical analysis of Fer was performed using the formalin-fixed tissue samples of 152 patients with RCC. The proliferative and apoptotic indices were used to represent the percentage of proliferation marker protein Ki-67- and cleaved caspase-3-positive cells, respectively. The microvessel density was defined as the number of cluster of differentiation (CD) 31-positively stained vessels/mm2. In addition, CD57+ and CD68+ cells were counted using semi-quantification of natural killer (NK) cells and macrophages. Fer expression in stromal cells was negatively associated with Fuhrman grade, pathological tumor stage and metastasis (P<0.001). Fer expression in stromal cells was negatively associated with CD68+ macrophage density, whereas it was positively associated with CD57+ NK cell density. Kaplan-Meier estimators indicated that decreased stromal Fer expression was a predictive marker of decreased cause-specific survival rate (P<0.001). Furthermore, low expression of Fer was identified as being an independent marker of decreased cause-specific survival using multivariate analysis (hazard ratio, 7.4; 95% confidence interval, 1.7-33.0; P<0.001). The results of the present study suggested that low Fer expression in stromal cells is associated with increased malignant aggressiveness and decreased survival in patients with RCC. CD57+ NK cell and CD68+ macrophage regulation in cancer-stromal tissue is considered to affect RCC pathology.
Collapse
Affiliation(s)
- Kensuke Mitsunari
- Department of Urology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| | - Yasuyoshi Miyata
- Department of Urology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| | - Shin-Ichi Watanabe
- Department of Urology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| | - Akihiro Asai
- Department of Urology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| | - Takuji Yasuda
- Department of Urology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| | - Shigeru Kanda
- Department of Urology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| | - Hideki Sakai
- Department of Urology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| |
Collapse
|
28
|
Hu X, Zhang Z, Liang Z, Xie D, Zhang T, Yu D, Zhong C. Downregulation of feline sarcoma-related protein inhibits cell migration, invasion and epithelial-mesenchymal transition via the ERK/AP-1 pathway in bladder urothelial cell carcinoma. Oncol Lett 2016; 13:686-694. [PMID: 28356947 PMCID: PMC5351348 DOI: 10.3892/ol.2016.5459] [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: 04/23/2015] [Accepted: 09/13/2016] [Indexed: 11/17/2022] Open
Abstract
Feline sarcoma-related protein (Fer) is a nuclear and cytoplasmic non-receptor protein tyrosine kinase and Fer overexpression is associated with various biological processes. However, the clinicopathological characteristics and molecular mechanisms of Fer expression in bladder urothelial cell carcinoma (UCC) have yet to be elucidated. The present study demonstrated that Fer was significantly upregulated in bladder UCC tissues and cell lines. A clinicopathological analysis suggested that Fer expression was significantly associated with tumor stage, histological grade and lymph node status, and Fer expression was a prognostic factor for overall survival in a multivariate analysis. Furthermore, small interfering RNA (siRNA) was used to silence the expression of the Fer gene in human bladder UCC T24 cells, and was shown to significantly reduce the migration and invasion of the cells. It was also observed that Fer-siRNA caused the T24 cells to acquire an epithelial cobblestone phenotype, and was able to reverse the epithelial-mesenchymal transition of the cells. Subsequently, Fer-knockdown was shown to deactivate the extracellular signal-regulated kinase/activator protein-1 signaling pathway in T24 cells. These results indicated, for the first time, that Fer has a critical role in bladder UCC progression and may be a potential therapeutic target for bladder UCC metastasis.
Collapse
Affiliation(s)
- Xudong Hu
- Department of Urology, Anqing First People's Hospital, Anqing, Anhui 246000, P.R. China
| | - Zhiqiang Zhang
- Department of Urology, The Second Hospital of Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Zhaofeng Liang
- Department of Nutrition and Food Safety, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, P.R. China
| | - Dongdong Xie
- Department of Urology, The Second Hospital of Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Tao Zhang
- Department of Urology, The Second Hospital of Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Dexin Yu
- Department of Urology, The Second Hospital of Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Caiyun Zhong
- Department of Nutrition and Food Safety, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, P.R. China
| |
Collapse
|
29
|
Electroporation-mediated delivery of the FER gene in the resolution of trauma-related fatal pneumonia. Gene Ther 2016; 23:785-796. [PMID: 27454317 PMCID: PMC5096957 DOI: 10.1038/gt.2016.58] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Revised: 06/19/2016] [Accepted: 07/11/2016] [Indexed: 12/18/2022]
Abstract
Injured patients with lung contusion (LC) are at risk of developing bacterial pneumonia (PNA) followed by sepsis and death. A recent genome-wide association study (GWAS) showed FER gene expression positively correlating with survival rates among individuals with above conditions. We sought to determine whether electroporation (EP)-mediated delivery of FER gene could indeed improve survival, in a lethal model of combined LC and PNA. C57BL/6 mice sustained unilateral LC, which preceded a 500 Klebsiella colony forming unit (CFU) inoculation by 6 h. In-between these insults, human FER plasmid (pFER) was introduced into the lungs followed by eight EP pulses applied externally (10 ms at 200 V cm-1). Control groups included EP of empty vector (pcDNA3) or Na+/K+-ATPase genes (pPump) and no treatment (LC+PNA). We recorded survival, histology, lung mechanics, bronchial alveolar lavage (BAL) fluid, FER and inflammatory gene expression and bacteriology. The data show that 7-day survival was significantly improved by pFER compared with control groups. pFER increased BAL monocytes and activated antibacterial response genes (nitric oxide synthase (NOS), Fizz). pFER treatment showed decreased lung and blood Klebsiella counts reaching, in some cases, complete sterilization. In conclusion, FER gene delivery promoted survival in LC+PNA mice via recruitment of activated immune cells, improving efficiency of bacterial clearance within contused lung.
Collapse
|
30
|
Cho CY, Lee KT, Chen WC, Wang CY, Chang YS, Huang HL, Hsu HP, Yen MC, Lai MZ, Lai MD. MST3 promotes proliferation and tumorigenicity through the VAV2/Rac1 signal axis in breast cancer. Oncotarget 2016; 7:14586-604. [PMID: 26910843 PMCID: PMC4924737 DOI: 10.18632/oncotarget.7542] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 01/02/2016] [Indexed: 12/15/2022] Open
Abstract
MST3 (mammalian STE20-like kinase 3) belongs to the Ste20 serine/threonine protein kinase family. The role of MST3 in tumor growth is less studied; therefore, we investigates the function of MST3 in breast cancer. Here, we demonstrate that MST3 is overexpressed in human breast tumors. Online Kaplan-Meier plotter analysis reveals that overexpression of MST3 predicts poor prognosis in breast cancer patients. Knockdown of MST3 with shRNA inhibits proliferation and anchorage-independent growth in vitro. Downregulation of MST3 in triple-negative MDA-MB-231 and MDA-MB-468 breast cancer cells decreases tumor formation in NOD/SCID mice. MST3 interacts with VAV2, but not VAV3, as demonstrated by co-immunoprecipitation and confocal microscopy. By domain mapping of MST3, we determine that the proline-rich region of MST3 (353KDIPKRP359) interacts with the SH3 domain of VAV2. Mutation of the two proline residues in this domain significantly attenuates the interaction between MST3 and VAV2. Overexpression of wild-type MST3 (WT-MST3), but not proline-rich-deleted MST3 (âP-MST3), enhances the proliferation rate and anchorage-independent growth of MDA-MB-468 cells. Overexpression of MST3 increases VAV2 phosphorylation and GTP-Rac1, whereas downregulation of MST3 or delivery of âP-MST3 results in a reduction of VAV2 and Rac1 activation. Knockdown of MST3 inhibits cyclin D1 protein expression. The Rac1 inhibitor EHop-016 attenuates cell proliferation induced by WT-MST3. Finally, Knockdown of MST3 or Rac1 inhibitor decreases cyclin D protein expression, which is important for tumor growth. These results indicate that MST3 interacts with VAV2 to activate Rac1 and promote the tumorigenicity of breast cancer.
Collapse
Affiliation(s)
- Chien-Yu Cho
- Department of Biochemistry and Molecular Biology, National Cheng Kung University, Tainan, Taiwan, ROC
- Institute of Basic Medical Sciences, National Cheng Kung University, Tainan, Taiwan, ROC
- Center for Infectious Diseases and Signaling Research, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC
| | - Kuo-Ting Lee
- Department of Surgery, National Cheng Kung University, Tainan, Taiwan, ROC
| | - Wei-Ching Chen
- Department of Biochemistry and Molecular Biology, National Cheng Kung University, Tainan, Taiwan, ROC
- Institute of Basic Medical Sciences, National Cheng Kung University, Tainan, Taiwan, ROC
| | - Chih-Yang Wang
- Department of Biochemistry and Molecular Biology, National Cheng Kung University, Tainan, Taiwan, ROC
- Institute of Basic Medical Sciences, National Cheng Kung University, Tainan, Taiwan, ROC
| | - Yung-Sheng Chang
- Department of Biochemistry and Molecular Biology, National Cheng Kung University, Tainan, Taiwan, ROC
- Institute of Basic Medical Sciences, National Cheng Kung University, Tainan, Taiwan, ROC
| | - Hau-Lun Huang
- Department of Biochemistry and Molecular Biology, National Cheng Kung University, Tainan, Taiwan, ROC
- Institute of Basic Medical Sciences, National Cheng Kung University, Tainan, Taiwan, ROC
| | - Hui-Ping Hsu
- Department of Surgery, National Cheng Kung University, Tainan, Taiwan, ROC
| | - Meng-Chi Yen
- Department of Emergency Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan, ROC
| | - Ming-Zong Lai
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan, ROC
- Graduate Institute of Immunology, National Taiwan University, Taipei, Taiwan, ROC
| | - Ming-Derg Lai
- Department of Biochemistry and Molecular Biology, National Cheng Kung University, Tainan, Taiwan, ROC
- Institute of Basic Medical Sciences, National Cheng Kung University, Tainan, Taiwan, ROC
- Center for Infectious Diseases and Signaling Research, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC
| |
Collapse
|
31
|
Phosphatidic Acid Increases Epidermal Growth Factor Receptor Expression by Stabilizing mRNA Decay and by Inhibiting Lysosomal and Proteasomal Degradation of the Internalized Receptor. Mol Cell Biol 2015; 35:3131-44. [PMID: 26124282 DOI: 10.1128/mcb.00286-15] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2015] [Accepted: 06/22/2015] [Indexed: 12/18/2022] Open
Abstract
Overexpression of epidermal growth factor receptor (EGFR) is one of the frequent mechanisms implicated in cancer progression, and so is the overexpression of the enzyme phospholipase D (PLD) and its reaction product, phosphatidic acid (PA). However, an understanding of how these signaling molecules interact at the level of gene expression is lacking. Catalytically active PLD enhanced expression of EGFR in human breast cancer cells. Overexpression of the PLD2 isoform increased EGFR mRNA and protein expression. It also negated an EGFR downregulation mediated by small interfering RNA targeting EGFR (siEGFR). Several mechanisms contributed to the alteration in EGFR expression. First was the stabilization of EGFR transcripts as PLD2 delayed mRNA decay, which prolonged their half-lives. Second, RNase enzymatic activity was inhibited by PA. Third, protein stabilization also occurred, as indicated by PLD resistance to cycloheximide-induced EGFR protein degradation. Fourth, PA inhibited lysosomal and proteasomal degradation of internalized EGFR. PLD2 and EGFR colocalized at the cell membrane, and JAK3 phosphorylation at Tyr980/Tyr981 followed receptor endocytosis. Further, the presence of PLD2 increased stabilization of intracellular EGFR in large recycling vesicles at ∼15 min of EGF stimulation. Thus, PLD2-mediated production of PA contributed to the control of EGFR exposure to ligand through a multipronged transcriptional and posttranscriptional program during the out-of-control accumulation of EGFR signaling in cancer cells.
Collapse
|
32
|
Kiss A, Gong X, Kowalewski JM, Shafqat-Abbasi H, Strömblad S, Lock JG. Non-monotonic cellular responses to heterogeneity in talin protein expression-level. Integr Biol (Camb) 2015; 7:1171-85. [PMID: 26000342 DOI: 10.1039/c4ib00291a] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Talin is a key cell-matrix adhesion component with a central role in regulating adhesion complex maturation, and thereby various cellular properties including adhesion and migration. However, knockdown studies have produced inconsistent findings regarding the functional influence of talin in these processes. Such discrepancies may reflect non-monotonic responses to talin expression-level variation that are not detectable via canonical "binary" comparisons of aggregated control versus knockdown cell populations. Here, we deployed an "analogue" approach to map talin influence across a continuous expression-level spectrum, which we extended with sub-maximal RNAi-mediated talin depletion. Applying correlative imaging to link live cell and fixed immunofluorescence data on a single cell basis, we related per cell talin levels to per cell measures quantitatively defining an array of cellular properties. This revealed both linear and non-linear correspondences between talin expression and cellular properties, including non-monotonic influences over cell shape, adhesion complex-F-actin association and adhesion localization. Furthermore, we demonstrate talin level-dependent changes in networks of correlations among adhesion/migration properties, particularly in relation to cell migration speed. Importantly, these correlation networks were strongly affected by talin expression heterogeneity within the natural range, implying that this endogenous variation has a broad, quantitatively detectable influence. Overall, we present an accessible analogue method that reveals complex dependencies on talin expression-level, thereby establishing a framework for considering non-linear and non-monotonic effects of protein expression-level heterogeneity in cellular systems.
Collapse
Affiliation(s)
- Alexa Kiss
- Center for Innovative Medicine, Department of Biosciences and Nutrition, Karolinska Institutet, Novum, Hälsov. 7-9, G-building floor 6, S-141 83 Huddinge, Sweden.
| | | | | | | | | | | |
Collapse
|
33
|
Luo JH, Liu S, Zuo ZH, Chen R, Tseng GC, Yu YP. Discovery and Classification of Fusion Transcripts in Prostate Cancer and Normal Prostate Tissue. THE AMERICAN JOURNAL OF PATHOLOGY 2015; 185:1834-45. [PMID: 25963990 DOI: 10.1016/j.ajpath.2015.03.008] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Revised: 03/03/2015] [Accepted: 03/12/2015] [Indexed: 12/20/2022]
Abstract
Fusion transcript formation is one of the fundamental mechanisms that drives the development of prostate cancer. Because of the advance of high-throughput parallel sequencing, many fusion transcripts have been discovered. However, the discovery rate of fusion transcripts specific for prostate cancer is lagging behind the discoveries made on chromosome abnormalities of prostate cancer. Recent analyses suggest that many fusion transcripts are present in both benign and cancerous tissues. Some of these fusion transcripts likely represent important components of normal gene expression in cells. It is necessary to identify the criteria and features of fusion transcripts that are specific for cancer. In this review, we discuss optimization of RNA sequencing depth for fusion transcript discovery and the characteristics of fusion transcripts in normal prostate tissues and prostate cancer. We also propose a new classification of cancer-specific fusion transcripts on the basis of their tail gene fusion protein product and the roles that these fusions may play in cancer development.
Collapse
Affiliation(s)
- Jian-Hua Luo
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania.
| | - Silvia Liu
- Department of Biostatistics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Ze-Hua Zuo
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Rui Chen
- Department of Biostatistics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - George C Tseng
- Department of Biostatistics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Yan P Yu
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| |
Collapse
|
34
|
Havel LS, Kline ER, Salgueiro AM, Marcus AI. Vimentin regulates lung cancer cell adhesion through a VAV2-Rac1 pathway to control focal adhesion kinase activity. Oncogene 2015; 34:1979-90. [PMID: 24858039 PMCID: PMC4245398 DOI: 10.1038/onc.2014.123] [Citation(s) in RCA: 115] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2013] [Revised: 03/21/2014] [Accepted: 04/06/2014] [Indexed: 12/14/2022]
Abstract
Vimentin is an intermediate filament protein whose expression correlates with increased metastatic disease, reduced patient survival and poor prognosis across multiple tumor types. Despite these well-characterized correlations, the molecular role of vimentin in cancer cell motility remains undefined. To approach this, we used an unbiased phosphoproteomics screen in lung cancer cell lines to discover cell motility proteins that show significant changes in phosphorylation upon vimentin depletion. We identified the guanine nucleotide exchange factor (GEF), VAV2, as having the greatest loss of phosphorylation owing to vimentin depletion. Since VAV2 serves as a GEF for the small Rho GTPase Rac1, a key player in cell motility and adhesion, we explored the vimentin-VAV2 pathway as a potential novel regulator of lung cancer cell motility. We show that VAV2 localizes to vimentin-positive focal adhesions (FAs) in lung cancer cells and complexes with vimentin and FA kinase (FAK). Vimentin loss impairs both pY142-VAV2 and downstream pY397-FAK activity showing that vimentin is critical for maintaining VAV2 and FAK activity. Importantly, vimentin depletion reduces the activity of the VAV2 target, Rac1, and a constitutively active Rac1 rescues defects in FAK and cell adhesion when vimentin or VAV2 is compromised. Based upon this data, we propose a model whereby vimentin promotes FAK stabilization through VAV2-mediated Rac1 activation. This model may explain why vimentin expressing metastatic lung cancer cells are more motile and invasive.
Collapse
Affiliation(s)
- L S Havel
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | - E R Kline
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | - A M Salgueiro
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | - A I Marcus
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA, USA
| |
Collapse
|
35
|
Baldassarre T, Watt K, Truesdell P, Meens J, Schneider MM, Sengupta SK, Craig AW. Endophilin A2 Promotes TNBC Cell Invasion and Tumor Metastasis. Mol Cancer Res 2015; 13:1044-55. [PMID: 25784716 DOI: 10.1158/1541-7786.mcr-14-0573] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Accepted: 03/08/2015] [Indexed: 11/16/2022]
Abstract
UNLABELLED Triple-negative breast cancers (TNBCs) are highly aggressive cancers that lack targeted therapies. However, EGFR is frequently activated in a subset of TNBCs and represents a viable clinical target. Because the endocytic adaptor protein Endophilin A2 (SH3GL1/Endo II) has been implicated in EGFR internalization, we investigated Endo II expression and function in human TNBCs. Endo II expression was high in several TNBC cells compared with normal breast epithelial cells. Stable knockdown (KD) of Endo II was achieved in two TNBC cell lines, and although cell viability was unaffected, defects in receptor-mediated endocytosis were observed. EGFR signaling to Erk and Akt kinases was impaired in Endo II KD cells, and this correlated with reduced rates of EGFR internalization and cell motility. Endo II KD cells also displayed defects in three dimensional (3D) cell invasion, and this correlated with impaired extracellular matrix degradation and internalization of MT1-MMP. Endo II silencing also caused a significant reduction in TNBC tumor growth and lung metastasis in mammary orthotopic tumor xenograft assays. In human breast tumor specimens, Endo II expression was highest in TNBC tumors compared with other subtypes, and at the level of gene expression, high Endo II was associated with reduced relapse-free survival in patients with basal-like breast cancers. Together, these results identify a positive role for Endo II in TNBC tumor metastasis and a potential link with poor prognosis. IMPLICATIONS Endophilin A2 and related adaptor proteins represent important signaling hubs to target in metastatic cancers.
Collapse
Affiliation(s)
- Tomas Baldassarre
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada. Cancer Biology and Genetics Division, Queen's Cancer Research Institute, Kingston, Ontario, Canada
| | - Kathleen Watt
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada. Cancer Biology and Genetics Division, Queen's Cancer Research Institute, Kingston, Ontario, Canada
| | - Peter Truesdell
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada. Cancer Biology and Genetics Division, Queen's Cancer Research Institute, Kingston, Ontario, Canada
| | - Jalna Meens
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada. Cancer Biology and Genetics Division, Queen's Cancer Research Institute, Kingston, Ontario, Canada
| | - Mark M Schneider
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, Ontario, Canada
| | - Sandip K Sengupta
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, Ontario, Canada
| | - Andrew W Craig
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada. Cancer Biology and Genetics Division, Queen's Cancer Research Institute, Kingston, Ontario, Canada.
| |
Collapse
|
36
|
Nizard P, Ezan F, Bonnier D, Le Meur N, Langouët S, Baffet G, Arlot-Bonnemains Y, Théret N. Integrative analysis of high-throughput RNAi screen data identifies the FER and CRKL tyrosine kinases as new regulators of the mitogenic ERK-dependent pathways in transformed cells. BMC Genomics 2014; 15:1169. [PMID: 25540073 PMCID: PMC4367906 DOI: 10.1186/1471-2164-15-1169] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2014] [Accepted: 12/12/2014] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Cell proliferation is a hallmark of cancer and depends on complex signaling networks that are chiefly supported by protein kinase activities. Therapeutic strategies have been used to target specific kinases but new methods are required to identify combined targets and improve treatment. Here, we propose a small interfering RNA genetic screen and an integrative approach to identify kinase networks involved in the proliferation of cancer cells. RESULTS The functional siRNA screen of 714 kinases in HeLa cells identified 91 kinases implicated in the regulation of cell growth, most of them never being reported in previous whole-genome siRNA screens. Based on gene ontology annotations, we have further discriminated between two classes of kinases that, when suppressed, result in alterations of the mitotic index and provoke cell-cycle arrest. Extinguished kinases that lead to a low mitotic index mostly include kinases implicated in cytosolic signaling. In contrast, extinguished kinases that result in a high mitotic index mostly include kinases implicated in cell division. By mapping hit kinases in the PhosphPOINT phosphoprotein database, we generated scale-free networks consisting of 449 and 661 protein-protein interactions for kinases from low MI and high MI groups, respectively. Further analyses of the kinase interactomes revealed specific modules such as FER- and CRKL-containing modules that connect three members of the epidermal growth factor receptor (EGFR) family, suggesting a tight control of the mitogenic EGF-dependent pathway. Based on experimental studies, we confirm the involvement of these two kinases in the regulation of tumor cell growth. CONCLUSION Based on a combined approach of large kinome-wide siRNA screens and ontology annotations, our study identifies for the first time two kinase groups differentially implicated in the control of cell proliferation. We further demonstrate that integrative analysis of the kinase interactome provides key information which can be used to facilitate or optimize target design for new therapeutic strategies. The complete list of protein-protein interactions from the two functional kinase groups will provide a useful database for future investigations.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Nathalie Théret
- INSERM UMR1085, Institut de Recherche sur la Santé l'Environnement et le Travail IRSET, Université Rennes 1, SFR Biosit, Rennes, France.
| |
Collapse
|
37
|
Yaffe E, Hikri E, Elkis Y, Cohen O, Segal A, Makovski A, Varvak A, Shpungin S, Nir U. Oncogenic properties of a spermatogenic meiotic variant of fer kinase expressed in somatic cells. Cancer Res 2014; 74:6474-85. [PMID: 25237066 DOI: 10.1158/0008-5472.can-14-0058] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The kinase Fer and its spermatogenic meiotic variant, FerT, are coexpressed in normal testes and cancerous tumors, but whether they exert related roles in spermatogenic or malignant cells has not been known. Here, we show that Fer and FerT reside in the mitochondria of spermatogenic cells and are harnessed to the reprogrammed mitochondria of colon carcinoma cells. Both kinases bound complex I of the mitochondrial electron transport chain (ETC) in spermatogenic and in colon carcinoma cells, and silencing of either Fer or FerT was sufficient to impair the activity of this complex. Directed mitochondrial accumulation of FerT in nonmalignant NIH3T3 cells increased their ETC complex I activity, ATP production, and survival, contingent upon stress conditions caused by nutrient and oxygen deprivation. Strikingly, directed mitochondrial accumulation of FerT endowed nonmalignant cells with tumor-forming ability. Thus, recruitment of a meiotic mitochondrial component to cancer cell mitochondria highlights a pivotal role for reprogrammed mitochondria in tumorigenesis.
Collapse
Affiliation(s)
- Etai Yaffe
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel
| | - Elad Hikri
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel
| | - Yoav Elkis
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel
| | - Ortal Cohen
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel
| | - Ariela Segal
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel
| | - Adar Makovski
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel
| | - Alexander Varvak
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel
| | - Sally Shpungin
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel
| | - Uri Nir
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel.
| |
Collapse
|
38
|
CIP4 promotes lung adenocarcinoma metastasis and is associated with poor prognosis. Oncogene 2014; 34:3527-35. [PMID: 25174397 PMCID: PMC4978543 DOI: 10.1038/onc.2014.280] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Revised: 07/21/2014] [Accepted: 07/23/2014] [Indexed: 12/28/2022]
Abstract
Aberrant Epidermal growth factor receptor (EGFR) signaling in non-small cell lung cancer (NSCLC) is linked to tumor progression, metastasis, and poor survival rates. Here, we report the role of Cdc42-interacting protein 4 (CIP4) in the regulation of NSCLC cell invasiveness and tumor metastasis. CIP4 was highly expressed in a panel of NSCLC cell lines and normal lung epithelial cell lines. Stable knock-down (KD) of CIP4 in lung adenocarcinoma H1299 cells, expressing wild-type EGFR, led to increased EGFR levels on the cell surface, and defects in sustained activation of Erk kinase in H1299 cells treated with EGF. CIP4 localized to leading edge projections in NSCLC cells, and CIP4 KD cells displayed defects in EGF-induced cell motility and invasion through extracellular matrix. This correlated with reduced expression and activity of matrix metalloproteinase-2 (MMP-2) in CIP4 KD cells compared to control. In xenograft assays, CIP4 silencing had no effect on tumor growth, but resulted in significant defects in spontaneous metastases to the lungs from these subcutaneous tumors. This correlated with reduced expression of the Erk target gene Zeb1, and the Zeb1 target gene MMP-2 in CIP4 KD tumors compared to control. CIP4 also enhanced rates of metastasis to the liver and lungs in an intrasplenic experimental metastasis model. In human NSCLC tumor sections, CIP4 expression was elevated ≥ 2-fold in 43% of adenocarcinomas and 32% of squamous carcinomas compared to adjacent normal lung tissues. Analysis of microarray data for NSCLC patients also revealed that high CIP4 transcript levels correlated with reduced overall survival. Together, these results identify CIP4 as a positive regulator of NSCLC metastasis, and a potential poor prognostic biomarker in lung adenocarcinoma.
Collapse
|
39
|
Jimbo M, Knudsen KE, Brody JR. Fusing transcriptomics to progressive prostate cancer. THE AMERICAN JOURNAL OF PATHOLOGY 2014; 184:2608-10. [PMID: 25128905 DOI: 10.1016/j.ajpath.2014.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Revised: 08/05/2014] [Accepted: 08/06/2014] [Indexed: 11/25/2022]
Abstract
This commentary highlights the article by Yu et al, describing a set of novel fusion transcripts strongly associated with prostate cancer prognosis.
Collapse
Affiliation(s)
- Masaya Jimbo
- Division of Surgical Research, Department of Surgery, The Jefferson Pancreas, Biliary, and Related Cancer Center, Sidney Kimmel Medical College, Philadelphia, Pennsylvania
| | - Karen E Knudsen
- Departments of Cancer Biology, Urology, and Radiation Oncology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Jonathan R Brody
- Division of Surgical Research, Department of Surgery, The Jefferson Pancreas, Biliary, and Related Cancer Center, Sidney Kimmel Medical College, Philadelphia, Pennsylvania.
| |
Collapse
|
40
|
Giansanti P, Preisinger C, Huber KVM, Gridling M, Superti-Furga G, Bennett KL, Heck AJR. Evaluating the promiscuous nature of tyrosine kinase inhibitors assessed in A431 epidermoid carcinoma cells by both chemical- and phosphoproteomics. ACS Chem Biol 2014; 9:1490-8. [PMID: 24804581 DOI: 10.1021/cb500116c] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Deregulation of protein tyrosine kinase signaling has been linked to many diseases, most notably cancer. As a consequence, small molecule inhibitors of protein tyrosine kinases may provide powerful strategies for treatment. Following the successful introduction of imatinib in the treatment of chronic myelogenous leukemia, such drugs are also now evaluated for other types of cancer. However, many developed kinase inhibitors are not very target-specific and therefore may induce side effects. The importance of such side effects is certainly cell-proteome dependent. Understanding the all-inclusive action of a tyrosine kinase inhibitor on each individual cell-type entails the identification of potential targets, combined with monitoring the downstream effects revealing the signaling networks involved. Here, we explored a multilevel quantitative mass spectrometry-based proteomic strategy to identify the direct targets and downstream signaling effect of four tyrosine kinase inhibitors (imatinib, dasatinib, bosutinib, and nilotinib) in epidermoid carcinoma cells, as a model system for skin-cancer. More than 25 tyrosine kinases showed affinity to the drugs, with imatinib and nilotinib displaying a high specificity, especially when compared to dasatinib and bosutinib. Consequently, the latter two drugs showed a larger effect on downstream phosphotyrosine signaling. Many of the proteins affected are key regulators in cell adhesion and invasion. Our data represents a multiplexed view on the promiscuous action of certain tyrosine kinase inhibitors that needs to be taking into consideration prior to the application of these drugs in the treatment of different forms of cancer.
Collapse
Affiliation(s)
- Piero Giansanti
- Biomolecular
Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular
Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Padualaan 8, 3584 CH Utrecht, The Netherlands
- Netherlands Proteomics
Centre, Padualaan 8, 3584 CH Utrecht, The Netherlands
| | - Christian Preisinger
- Biomolecular
Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular
Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Padualaan 8, 3584 CH Utrecht, The Netherlands
- Netherlands Proteomics
Centre, Padualaan 8, 3584 CH Utrecht, The Netherlands
| | - Kilian V. M. Huber
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria
| | - Manuela Gridling
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria
| | - Giulio Superti-Furga
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria
| | - Keiryn L. Bennett
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria
| | - Albert J. R. Heck
- Biomolecular
Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular
Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Padualaan 8, 3584 CH Utrecht, The Netherlands
- Netherlands Proteomics
Centre, Padualaan 8, 3584 CH Utrecht, The Netherlands
| |
Collapse
|
41
|
Szatmary AC, Stuelten CH, Nossal R. Improving the design of the agarose spot assay for eukaryotic cell chemotaxis. RSC Adv 2014; 4:57343-57349. [PMID: 25530845 DOI: 10.1039/c4ra08572h] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Migration of cells along gradients of effector molecules, i.e., chemotaxis, is necessary in immune response and is involved in development and cancer metastasis. The experimental assessment of chemotaxis thus is of high interest. The agarose spot assay is a simple tissue culture system used to analyze chemotaxis. Although direction sensing requires gradients to be sufficiently steep, how the chemical gradients developed in this assay change over time, and thus, under what conditions chemotaxis is plausible, has not yet been determined. Here, we use numerical solution of the diffusion equation to determine the chemoattractant gradient produced in the assay. Our analysis shows that, for the usual spot size, the lifetime of the assay is optimized if the chemoattractant concentration in the spot is initially 30 times the dissociation constant of the chemoattractant-receptor bond. This result holds regardless of the properties of the chemoattractant. With this initial concentration, the chemoattractant gradient falls to the minimum threshold for directional sensing at the same time that the concentration drops to the optimal level for detecting gradient direction. If a higher initial chemoattractant concentration is used, the useful lifetime of the assay is likely to be shortened because receptor saturation may decrease the cells' sensitivity to the gradient; lower initial concentrations would result in too little chemoattractant for the cells to detect. Moreover, chemoattractants with higher diffusion coefficients would sustain gradients for less time. Based on previous measurements of the diffusion coefficients of the chemoattractants EGF and CXCL12, we estimate that the assay will produce gradients that cells can sense for a duration of 10 h for EGF and 5 h for CXCL12. These gradient durations are comparable to what can be achieved with the Boyden chamber assay. The analysis presented in this work facilitates determination of suitable parameters for the assay, and can be used to assess whether observed cell motility is likely due to chemotaxis or chemokinesis.
Collapse
Affiliation(s)
- Alex C Szatmary
- Program in Physical Biology, Eunice Kennedy Shriver National Institute of Child Health, and Human Development, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Christina H Stuelten
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Ralph Nossal
- Program in Physical Biology, Eunice Kennedy Shriver National Institute of Child Health, and Human Development, National Institutes of Health, Bethesda, Maryland 20892, USA
| |
Collapse
|
42
|
Auld D, Lea W, Davis MI, Simeonov A. Literature Search and Review. Assay Drug Dev Technol 2013. [DOI: 10.1089/adt.2013.1107.lr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Doug Auld
- Novartis Institutes for BioMedical Research, Cambridge, Massachusetts
| | | | | | | |
Collapse
|