1
|
Yang Y, Qing L, You C, Li Q, Xu W, Dong Z. Methuosis key gene ARF6 as a diagnostic, prognostic and immunotherapeutic marker for prostate cancer: based on a comprehensive pan-cancer multi-omics analysis. Discov Oncol 2025; 16:882. [PMID: 40410613 PMCID: PMC12102050 DOI: 10.1007/s12672-025-02275-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Accepted: 04/01/2025] [Indexed: 05/25/2025] Open
Abstract
BACKGROUND Prostate cancer (PCa) is a leading cause of cancer-related mortality among men worldwide. Despite progress in the understanding of tumor biology, the prognosis for advanced prostate cancer remains poor, necessitating the identification of novel diagnostic, prognostic, and therapeutic biomarkers. Methuosis, a recently identified form of programmed cell death (PCD), is characterized by cytoplasmic vacuole accumulation and subsequent cell rupture, distinct from classical apoptosis and necrosis. The key regulatory gene in Methuosis, ARF6 (ADP-ribosylation factor 6), has emerged as a potential marker for cancer diagnosis and treatment. However, its role in prostate cancer and other malignancies remains insufficiently understood. METHODS In this study, we performed a comprehensive pan-cancer multi-omics analysis to investigate the role of ARF6 in Methuosis across multiple cancer types, with a specific focus on PCa as the primary context. Using data from public databases, including RNA sequencing, gene expression profiling, and clinical outcomes, we assessed the association between ARF6 expression and patient prognosis in PCa within this broader pan-cancer framework. Additionally, we employed functional enrichment analyses and survival analysis to explore the potential of ARF6 as a diagnostic and prognostic marker for prostate cancer. Immunotherapy-related gene expression signatures were also evaluated to determine the therapeutic relevance of ARF6. RESULTS ARF6 was significantly overexpressed in PCa tissues compared to normal tissues and was associated with poor prognosis (p < 0.05), particularly in advanced and metastatic stages. Receiver operating characteristic (ROC) analysis revealed a diagnostic AUC of 0.792 for ARF6. Functional analyses indicated that ARF6 regulates pathways critical to cell migration, invasion, and drug resistance. Moreover, ARF6 expression showed a strong negative correlation with immune checkpoint markers, such as PD-L1 (r = - 0.74), suggesting its potential as an immunotherapy target. These findings underscore ARF6's pivotal role in Methuosis and its promise as a biomarker in PCa. CONCLUSION ARF6 is a key regulator of Methuosis in prostate cancer, contributing to tumor progression, metastasis, and resistance to treatment. Our findings support the potential of ARF6 as a diagnostic, prognostic, and immunotherapeutic target in prostate cancer. Further experimental validation is needed to confirm these observations and to explore the therapeutic implications of targeting ARF6 in cancer treatment.
Collapse
Affiliation(s)
- Yongjin Yang
- Department of Urology, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, 730000, Gansu, China
- Gansu Province Clinical Research Center for Urinary System Disease, Lanzhou, 730000, Gansu, China
| | - Liangliang Qing
- Department of Urology, Zigong Fourth People's Hospital, No.19 Tanmulin Street, Ziliujing District, Zigong, 643000, China
| | - Chengyu You
- Department of Urology, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, 730000, Gansu, China
- Gansu Province Clinical Research Center for Urinary System Disease, Lanzhou, 730000, Gansu, China
| | - Qingchao Li
- Department of Urology, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, 730000, Gansu, China
- Gansu Province Clinical Research Center for Urinary System Disease, Lanzhou, 730000, Gansu, China
| | - Wenbo Xu
- Department of Urology, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, 730000, Gansu, China
- Gansu Province Clinical Research Center for Urinary System Disease, Lanzhou, 730000, Gansu, China
| | - Zhilong Dong
- Department of Urology, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, 730000, Gansu, China.
- Gansu Province Clinical Research Center for Urinary System Disease, Lanzhou, 730000, Gansu, China.
| |
Collapse
|
2
|
Jathal MK, Mudryj M, Dall'Era MA, Ghosh PM. Amiloride sensitizes prostate cancer cells to the reversible tyrosine kinase inhibitor lapatinib by modulating Erbb3 subcellular localization. Cell Mol Life Sci 2024; 82:24. [PMID: 39725713 PMCID: PMC11671466 DOI: 10.1007/s00018-024-05540-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Revised: 11/13/2024] [Accepted: 12/05/2024] [Indexed: 12/28/2024]
Abstract
Neoadjuvant therapy (NAT) has been studied in clinically localized prostate cancer (PCa) to improve the outcomes from radical prostatectomy (RP) by 'debulking' of high-risk PCa; however, using androgen deprivation therapy (ADT) at this point risks castration resistant PCa (CRPC) clonal proliferation. Our goal is to identify alternative NAT that reduce hormone sensitive PCa (HSPC) without affecting androgen receptor (AR) transcriptional activity. PCa is associated with increased expression and activation of the epidermal growth factor receptor (EGFR) family, including HER2 and ErbB3. The FDA-approved HER2 inhibitor lapatinib has been tested in PCa but was ineffective due to continued activation of ErbB3. We now demonstrate that this is due to ErbB3 being localized to the nucleus in HSPC and thus protected from lapatinib which affect membrane localized HER2/ErbB3 dimers. Here, we show that the well-established, well-tolerated potassium-sparing diuretic amiloride hydrochloride dose dependently prevented ErbB3 nuclear localization via formation of plasma membrane localized HER2/ErbB3 dimers. This in turn allowed lapatinib inactivation of these dimers via inhibition of its target HER2, which dephosphorylated ERK1/2 and inhibited survival. Amiloride combined with lapatinib significantly increased apoptosis at relatively low doses of both drugs but did not affect AR transcriptional activity. Thus, our data indicate that a combination of amiloride and lapatinib could target HSPC tumors without problems associated with using ADT as NAT in HSPC.
Collapse
Affiliation(s)
- Maitreyee K Jathal
- Research Service, VA Northern California Health Care System, Mather, CA, USA
- Department of Medical Microbiology and Immunology, University of California Davis, Davis, CA, USA
| | - Maria Mudryj
- Research Service, VA Northern California Health Care System, Mather, CA, USA
- Department of Medical Microbiology and Immunology, University of California Davis, Davis, CA, USA
| | - Marc A Dall'Era
- Department of Urologic Surgery, University of California Davis, 4860 Y Street, Suite 3500, Sacramento, CA, 95817, USA
| | - Paramita M Ghosh
- Research Service, VA Northern California Health Care System, Mather, CA, USA.
- Department of Urologic Surgery, University of California Davis, 4860 Y Street, Suite 3500, Sacramento, CA, 95817, USA.
- Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, CA, USA.
| |
Collapse
|
3
|
Jathal MK, Mudryj MM, Dall'Era M, Ghosh PM. Amiloride Sensitizes Prostate Cancer Cells to the Reversible Tyrosine Kinase Inhibitor Lapatinib by Modulating ERBB3 Subcellular Localization. RESEARCH SQUARE 2024:rs.3.rs-4844371. [PMID: 39257973 PMCID: PMC11384790 DOI: 10.21203/rs.3.rs-4844371/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2024]
Abstract
Neoadjuvant therapy (NAT) has been studied in clinically localized prostate cancer (PCa) to improve the outcomes from radical prostatectomy (RP) by 'debulking' of high-risk PCa; however, using androgen deprivation at this point risks castration resistant PCa (CRPC) clonal proliferation with potentially profound side effects such as fatigue, loss of libido, hot flashes, loss of muscle mass, and weight gain. Our goal is to identify alternative NAT that reduce hormone sensitive PCa (HSPC) without affecting androgen receptor (AR) transcriptional activity. PCa is associated with increased expression and activation of the epidermal growth factor receptor (EGFR) family, including HER2 and ErbB3. Dimerization between these receptors is required for activation of downstream targets involved in tumor progression. The FDA-approved HER2 inhibitor lapatinib has been tested in PCa but was ineffective due to continued activation of ErbB3. We now demonstrate that this is due to ErbB3 being localized to the nucleus in HSPC and thus protected from lapatinib which affect membrane localized HER2/ErbB3 dimers. Here, we show that the well-established, well-tolerated diuretic amiloride hydrochloride dose dependently prevented ErbB3 nuclear localization via formation of plasma membrane localized HER2/ErbB3 dimers. This in turn allowed lapatinib inactivation of these dimers via inhibition of its target HER2, which dephosphorylated downstream survival and proliferation regulators AKT and ERK1/2. Amiloride combined with lapatinib significantly increased apoptosis but did not affect AR transcriptional activity. Thus, our data indicate that a combination of amiloride and lapatinib could target HSPC tumors without problems associated with androgen deprivation therapy in localized PCa.
Collapse
|
4
|
Abstract
Macropinocytosis is a critical route of nutrient acquisition in pancreatic cancer cells. Constitutive macropinocytosis is promoted by mutant KRAS, which activates the PI3Kα lipid kinase and RAC1, to drive membrane ruffling, macropinosome uptake and processing. However, our recent study on the KRASG12R mutant indicated the presence of a KRAS-independent mode of macropinocytosis in pancreatic cancer cell lines, thereby increasing the complexity of this process. We found that KRASG12R-mutant cell lines promote macropinocytosis independent of KRAS activity using PI3Kγ and RAC1, highlighting the convergence of regulation on RAC signaling. While macropinocytosis has been proposed to be a therapeutic target for the treatment of pancreatic cancer, our studies have underscored how little we understand about the activation and regulation of this metabolic process. Therefore, this review seeks to highlight the differences in macropinocytosis regulation in the two cellular subtypes while also highlighting the features that make the KRASG12R mutant atypical.
Collapse
Affiliation(s)
- G Aaron Hobbs
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, SC, USA.
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA.
| | - Channing J Der
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Curriculum in Genetics and Molecular Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
5
|
Song S, Zhang Y, Ding T, Ji N, Zhao H. The Dual Role of Macropinocytosis in Cancers: Promoting Growth and Inducing Methuosis to Participate in Anticancer Therapies as Targets. Front Oncol 2021; 10:570108. [PMID: 33542897 PMCID: PMC7851083 DOI: 10.3389/fonc.2020.570108] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Accepted: 12/01/2020] [Indexed: 02/05/2023] Open
Abstract
Macropinocytosis is an important mechanism of internalizing extracellular materials and dissolved molecules in eukaryotic cells. Macropinocytosis has a dual effect on cancer cells. On the one hand, cells expressing RAS genes (such as K-RAS, H-RAS) under the stress of nutrient deficiency can spontaneously produce constitutive macropinocytosis to promote the growth of cancer cells by internalization of extracellular nutrients (like proteins), receptors, and extracellular vesicles(EVs). On the other hand, abnormal expression of RAS genes and drug treatment (such as MOMIPP) can induce a novel cell death associated with hyperactivated macropinocytosis: methuosis. Based on the dual effect, there is immense potential for designing anticancer therapies that target macropinocytosis in cancer cells. In view of the fact that there has been little review of the dual effect of macropinocytosis in cancer cells, herein, we systematically review the general process of macropinocytosis, its specific manifestation in cancer cells, and its application in cancer treatment, including anticancer drug delivery and destruction of macropinocytosis. This review aims to serve as a reference for studying macropinocytosis in cancers and designing macropinocytosis-targeting anticancer drugs in the future.
Collapse
Affiliation(s)
- Shaojuan Song
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yanan Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Tingting Ding
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Ning Ji
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Hang Zhao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
6
|
Lei H, Ma F, Jia R, Tan B. Effects of Arf6 downregulation on biological characteristics of human prostate cancer cells. Int Braz J Urol 2020; 46:950-961. [PMID: 32822124 PMCID: PMC7527080 DOI: 10.1590/s1677-5538.ibju.2019.0499] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 12/24/2019] [Indexed: 11/21/2022] Open
Abstract
Objective To evaluate the effects of Arf6 downregulation on human prostate cancer cells. Materials and Methods The effects of Arf6 downregulation on cell proliferation, migration, invasion and apoptosis were assessed by MTT, BrdU, scratch, Transwell assays and flow cytometry respectively. AKT, p-AKT, ERK1/2, p-ERK1/2 and Rac1 protein expressions were detected by Western blot. Results Downregulating Arf6 by siRNA interference suppressed the mRNA and protein expressions of Arf6. The proliferation capacities of siRNA group at 48h, 72h, and 96h were significantly lower than those of control group (P <0.05). The migration distance of siRNA group at 18h was significantly shorter than that of control group (P <0.01). The number of cells penetrating Transwell chamber membrane significantly decreased in siRNA group compared with that of control group (P <0.01). After 24h, negative control and normal control groups had similar apoptotic rates (P >0.05) which were both significantly lower than that of siRNA group (P <0.01). After Arf6 expression was downregulated, p-ERK1/2 and Rac1 protein expressions were significantly lower than those of control group (P <0.05). Conclusion Downregulating Arf6 expression can inhibit the proliferation, migration and invasion of prostate cancer cells in vitro, which may be related to ERK1/2 phosphorylation and Rac1 downregulation.
Collapse
Affiliation(s)
- Haiming Lei
- School of Clinical Medicine, Jiangsu Vocational College of Medicine, Yancheng, Jiangsu Province, China
| | - Fujun Ma
- Department of Urology, Shengli Oilfield Central Hospital, Dongying, Shandong Province, China
| | - Renfeng Jia
- Department of Urology, Shengli Oilfield Central Hospital, Dongying, Shandong Province, China
| | - Bo Tan
- Department of Urology, Shengli Oilfield Central Hospital, Dongying, Shandong Province, China
| |
Collapse
|
7
|
Bobrin VA, Lin Y, He J, Qi Y, Gu W, Monteiro MJ. Therapeutic Delivery of Polymeric Tadpole Nanostructures with High Selectivity to Triple Negative Breast Cancer Cells. Biomacromolecules 2020; 21:4457-4468. [PMID: 32212644 DOI: 10.1021/acs.biomac.0c00302] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Targeted delivery of therapeutic drugs using nanoparticles to the highly aggressive triple negative breast cancer cells has the potential to reduce side effects and drug resistance. Cell entry into triple negative cells can be enhanced by incorporating cell binding receptor molecules on the surface of the nanoparticles to enhance receptor-mediated entry pathways, including clatherin or caveolae endocytosis. However, for highly aggressive cancer cells, these pathways may not be effective, with the more rapid and high volume uptake from macropinocytosis or phagocytosis being significantly more advantageous. Here we show, in the absence of attached cell binding receptor molecules, that asymmetric polymer tadpole nanostructure coated with a thermoresponsive poly(N-isopropylacrylamide) polymer with approximately 50% of this polymer in a globular conformation resulted in both high selectivity and rapid uptake into the triple breast cancer cell line MDA-MB-231. We found that the poly(N-isopropylacrylamide) surface coating in combination with the tadpole's unique shape had an almost 15-fold increase in cell uptake compared to spherical particles with the same polymer coating, and that the mode of entry was most likely through phagocytosis. Delivery of the tadpole attached with doxorubicin (a prodrug, which can be released at pHs < 6) showed a remarkable 10-fold decrease in the IC50 compared to free doxorubicin. It was further observed that cell death was primarily through late apoptosis, which may allow further protection from the body's own immune system. Our results demonstrate that by tuning the chemical composition, polymer conformation and using an asymmetric-shaped nanoparticle, both selectivity and effective delivery and release of therapeutics can be achieved, and such insights will allow the design of nanoparticles for optimal cancer outcomes.
Collapse
Affiliation(s)
- Valentin A Bobrin
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane Queensland 4072, Australia
| | - Yanling Lin
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane Queensland 4072, Australia
| | - Jianwei He
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane Queensland 4072, Australia.,Department of Clinical Lab Diagnosis, First Affiliated Hospital of Shihezi University School of Medicine, Shihezhi University, Xinjiang, China
| | - Yan Qi
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane Queensland 4072, Australia.,Department of Pathology and Key Laboratory for Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Shihezhi University, Xinjiang, China
| | - Wenyi Gu
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane Queensland 4072, Australia
| | - Michael J Monteiro
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane Queensland 4072, Australia
| |
Collapse
|
8
|
Wang L, Wang B, Quan Z. Identification of aberrantly methylated‑differentially expressed genes and gene ontology in prostate cancer. Mol Med Rep 2019; 21:744-758. [PMID: 31974616 PMCID: PMC6947816 DOI: 10.3892/mmr.2019.10876] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 09/10/2019] [Indexed: 12/24/2022] Open
Abstract
Prostate cancer (PCa) is the most frequent urological malignancy in men worldwide. DNA methylation has an essential role in the etiology and pathogenesis of PCa. The purpose of the present study was to identify the aberrantly methylated-differentially expressed genes and to determine their potential roles in PCa. The important node genes identified were screened by integrated analysis. Gene expression microarrays and gene methylation microarrays were downloaded and aberrantly methylated-differentially expressed genes were obtained. Enrichment analysis and protein-protein interactions (PPI) were obtained, their interactive and visual networks were created, and the node genes in the PPI network were validated. A total of 105 hypomethylation-high expression genes and 561 hypermethylation-low expression genes along with their biological processes were identified. The top 10 node genes obtained from the PPI network were identified for each of the two gene groups. The methylation and gene expression status of node genes in TCGA database, GEPIA tool, and the HPA database were generally consistent with those of our results. In conclusion, the present study identified 20 aberrantly methylated-differentially expressed genes in PCa by combining bioinformatics analyses of gene expression and gene methylation microarrays, and concurrently, the survival of these genes was analyzed. Notably, methylation is a reversible biological process, which makes it of great biological significance for the diagnosis and treatment of prostate cancer using bioinformatics technology to determine abnormal methylation gene markers. The present study provided novel therapeutic targets for the treatment of PCa.
Collapse
Affiliation(s)
- Linbang Wang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Bing Wang
- Laboratory of Environmental Monitoring, Shaanxi Province Health Inspection Institution, Xi'an, Shaanxi 710077, P.R. China
| | - Zhengxue Quan
- Department of Orthopedic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| |
Collapse
|
9
|
Yu Q, Gratzke C, Wang R, Li B, Kuppermann P, Herlemann A, Tamalunas A, Wang Y, Rutz B, Ciotkowska A, Wang X, Strittmatter F, Waidelich R, Stief CG, Hennenberg M. A NAV2729-sensitive mechanism promotes adrenergic smooth muscle contraction and growth of stromal cells in the human prostate. J Biol Chem 2019; 294:12231-12249. [PMID: 31243101 DOI: 10.1074/jbc.ra119.007958] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 06/19/2019] [Indexed: 12/25/2022] Open
Abstract
Voiding symptoms in benign prostatic hyperplasia (BPH) are driven by prostate smooth muscle contraction and prostate growth. Smooth muscle contraction in the prostate and other organs critically depends on activation of the small monomeric GTPase RhoA and probably Rac1. A role of another GTPase, ADP-ribosylation factor 6 (ARF6), for smooth muscle contraction has been recently suggested by indirect evidence but remains to be proven for any organ. Here, we report effects of NAV2729, an inhibitor with assumed specificity for ARF6, in human prostate tissues and cultured prostate stromal cells (WPMY-1). NAV2729 (5 μm) inhibited neurogenic and α1-adrenergic contractions of human prostate tissues. Contractions induced by endothelin-1, by the thromboxane A2 agonist U46619, or by high molar KCl were not inhibited. Correlation analyses suggested up-regulation of prostatic ARF6 expression with increasing degree of BPH, as ARF6 expression increased with the content of prostate-specific antigen (PSA) of prostate tissues. NAV2729 inhibited ARF6 activity but not other GTPases (ARF1, RhoA, Rac1) in prostate tissues and in WPMY-1 cells. Proliferation of WPMY-1 cells was inhibited concentration-dependently by NAV2726, as reflected by decreased viability, 5-ethynyl-2'-deoxyuridine (EdU) assay, colony formation assay, and expression of Ki-67. Silencing of ARF6 expression mimicked effects of NAV2729 on viability and in the EdU assay. Effects of NAV2729 on viability and proliferation were attenuated in cells with silenced ARF6 expression. Our findings suggest that a NAV2729-sensitive mechanism promotes adrenergic contraction and stromal cell growth in the human prostate, which is probably ARF6-mediated. Similar actions in other organs and urodynamic effects of NAV2729 appear possible.
Collapse
Affiliation(s)
- Qingfeng Yu
- Department of Urology, University Hospital, LMU Munich, 81377 Munich, Germany; Department of Urology and Guangdong Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510230, China
| | - Christian Gratzke
- Department of Urology, University Hospital, LMU Munich, 81377 Munich, Germany; Department of Urology, University of Freiburg, Freiburg 79106, Germany
| | - Ruixiao Wang
- Department of Urology, University Hospital, LMU Munich, 81377 Munich, Germany
| | - Bingsheng Li
- Department of Urology, University Hospital, LMU Munich, 81377 Munich, Germany
| | - Paul Kuppermann
- Department of Urology, University Hospital, LMU Munich, 81377 Munich, Germany
| | - Annika Herlemann
- Department of Urology, University Hospital, LMU Munich, 81377 Munich, Germany
| | - Alexander Tamalunas
- Department of Urology, University Hospital, LMU Munich, 81377 Munich, Germany
| | - Yiming Wang
- Department of Urology, University Hospital, LMU Munich, 81377 Munich, Germany
| | - Beata Rutz
- Department of Urology, University Hospital, LMU Munich, 81377 Munich, Germany
| | - Anna Ciotkowska
- Department of Urology, University Hospital, LMU Munich, 81377 Munich, Germany
| | - Xiaolong Wang
- Department of Urology, University Hospital, LMU Munich, 81377 Munich, Germany
| | - Frank Strittmatter
- Department of Urology, University Hospital, LMU Munich, 81377 Munich, Germany
| | - Raphaela Waidelich
- Department of Urology, University Hospital, LMU Munich, 81377 Munich, Germany
| | - Christian G Stief
- Department of Urology, University Hospital, LMU Munich, 81377 Munich, Germany
| | - Martin Hennenberg
- Department of Urology, University Hospital, LMU Munich, 81377 Munich, Germany.
| |
Collapse
|
10
|
Lang L, Shay C, Zhao X, Teng Y. Combined targeting of Arf1 and Ras potentiates anticancer activity for prostate cancer therapeutics. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2017; 36:112. [PMID: 28830537 PMCID: PMC5568197 DOI: 10.1186/s13046-017-0583-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Accepted: 08/16/2017] [Indexed: 02/07/2023]
Abstract
Background Although major improvements have been made in surgical management, chemotherapeutic, and radiotherapeutic of prostate cancer, many prostate cancers remain refractory to treatment with standard agents. Therefore, the identification of new molecular targets in cancer progression and development of novel therapeutic strategies to target them are very necessary for achieving better survival for patients with prostate cancer. Activation of small GTPases such as Ras and Arf1 is a critical component of the signaling pathways for most of the receptors shown to be upregulated in advanced prostate cancer. Methods The drug effects on cell proliferation were measured by CellTiter 96® AQueous One Solution Cell Proliferation Assay. The drug effects on cell migration and invasion were determined by Radius™ 24-well and Matrigel-coated Boyden chambers. The drug effects on apoptosis were assessed by FITC Annexin V Apoptosis Detection Kit with 7-AAD and Western blot with antibodies against cleaved PARP and Caspase 3. A NOD/SCID mouse model generated by subcutaneous injection was used to assess the in vivo drug efficacy in tumor growth. ERK activation and tumor cell proliferation in xenografts were examined by immunohistochemistry. Results We show that Exo2, a small-molecule inhibitor that reduces Arf1 activation, effectively suppresses prostate cancer cell proliferation by blocking ERK1/2 activation. Exo2 also has other effects, inhibiting migration and invasion of PCa cells and inducing apoptosis. The Ras inhibitor salirasib augments Exo2-induced cytotoxicity in prostate cancer cells partially by enhancing the suppression of ERK1/2 phosphorylation. In a xenograft mouse model of prostate cancer, Exo2 reduces prostate tumor burden and inhibits ERK1/2 activation at a dose of 20 mg/kg. Synergistic treatment of salirasib and Exo2 exhibits a superior inhibitory effect on prostate tumor growth compared with either drug alone, which may be attributed to the more efficient inhibition of ERK1/2 phosphorylation. Conclusion This study suggests that simultaneous blockade of Arf1 and Ras activation in prostate cancer cells is a potential targeted therapeutic strategy for preventing prostate cancer development. Electronic supplementary material The online version of this article (doi:10.1186/s13046-017-0583-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Liwei Lang
- Department of Oral Biology, Augusta University, Augusta, GA, 30912, USA
| | - Chloe Shay
- Department of Pediatrics, Emory Children's Center, Emory University, Atlanta, GA, 30322, USA
| | - Xiangdong Zhao
- Department of Oral Biology, Augusta University, Augusta, GA, 30912, USA
| | - Yong Teng
- Department of Oral Biology, Augusta University, Augusta, GA, 30912, USA. .,Georgia Cancer Center, Augusta University, 1120 15th Street, Augusta, GA, 30912, USA. .,Department of Biochemistry and Molecular Biology, Augusta University, Augusta, GA, 30912, USA.
| |
Collapse
|
11
|
Hayes DA, Kunde DA, Taylor RL, Pyecroft SB, Sohal SS, Snow ET. ERBB3: A potential serum biomarker for early detection and therapeutic target for devil facial tumour 1 (DFT1). PLoS One 2017; 12:e0177919. [PMID: 28591206 PMCID: PMC5462353 DOI: 10.1371/journal.pone.0177919] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 05/05/2017] [Indexed: 12/13/2022] Open
Abstract
Devil Facial Tumour 1 (DFT1) is one of two transmissible neoplasms of Tasmanian devils (Sarcophilus harrisii) predominantly affecting their facial regions. DFT1's cellular origin is that of Schwann cell lineage where lesions are evident macroscopically late in the disease. Conversely, the pre-clinical timeframe from cellular transmission to appearance of DFT1 remains uncertain demonstrating the importance of an effective pre-clinical biomarker. We show that ERBB3, a marker expressed normally by the developing neural crest and Schwann cells, is immunohistohemically expressed by DFT1, therefore the potential of ERBB3 as a biomarker was explored. Under the hypothesis that serum ERBB3 levels may increase as DFT1 invades local and distant tissues our pilot study determined serum ERBB3 levels in normal Tasmanian devils and Tasmanian devils with DFT1. Compared to the baseline serum ERBB3 levels in unaffected Tasmanian devils, Tasmanian devils with DFT1 showed significant elevation of serum ERBB3 levels. Interestingly Tasmanian devils with cutaneous lymphoma (CL) also showed elevation of serum ERBB3 levels when compared to the baseline serum levels of Tasmanian devils without DFT1. Thus, elevated serum ERBB3 levels in otherwise healthy looking devils could predict possible DFT1 or CL in captive or wild devil populations and would have implications on the management, welfare and survival of Tasmanian devils. ERBB3 is also a therapeutic target and therefore the potential exists to consider modes of administration that may eradicate DFT1 from the wild.
Collapse
Affiliation(s)
- Dane A. Hayes
- Department of Primary Industries, Parks Water and Environment, Animal Health Laboratory, Launceston, Tasmania, Australia
- Save the Tasmanian Devil Program, University of Tasmania, Hobart, Tasmania, Australia
- School of Health Sciences, Faculty of Health, University of Tasmania, Launceston, Tasmania, Australia
| | - Dale A. Kunde
- School of Health Sciences, Faculty of Health, University of Tasmania, Launceston, Tasmania, Australia
| | - Robyn L. Taylor
- Save the Tasmanian Devil Program, University of Tasmania, Hobart, Tasmania, Australia
- Department of Primary Industries, Parks Water and Environment, Resource Management and Conservation, Hobart, Tasmania, Australia
| | - Stephen B. Pyecroft
- School of Animal & Veterinary Sciences, Faculty of Science, University of Adelaide, Roseworthy Campus, Roseworthy, South Australia
| | - Sukhwinder Singh Sohal
- School of Health Sciences, Faculty of Health, University of Tasmania, Launceston, Tasmania, Australia
| | - Elizabeth T. Snow
- School of Health Sciences, Faculty of Health, University of Tasmania, Launceston, Tasmania, Australia
| |
Collapse
|
12
|
Ha KD, Bidlingmaier SM, Liu B. Macropinocytosis Exploitation by Cancers and Cancer Therapeutics. Front Physiol 2016; 7:381. [PMID: 27672367 PMCID: PMC5018483 DOI: 10.3389/fphys.2016.00381] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Accepted: 08/22/2016] [Indexed: 12/19/2022] Open
Abstract
Macropinocytosis has long been known as a primary method for cellular intake of fluid-phase and membrane-bound bulk cargo. This review seeks to re-examine the latest studies to emphasize how cancers exploit macropinocytosis to further their tumorigenesis, including details in how macropinocytosis can be adapted to serve diverse functions. Furthermore, this review will also cover the latest endeavors in targeting macropinocytosis as an avenue for novel therapeutics.
Collapse
Affiliation(s)
- Kevin D Ha
- Department of Anesthesia, UCSF Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco San Francisco, CA, USA
| | - Scott M Bidlingmaier
- Department of Anesthesia, UCSF Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco San Francisco, CA, USA
| | - Bin Liu
- Department of Anesthesia, UCSF Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco San Francisco, CA, USA
| |
Collapse
|
13
|
He Y, Su Z, Xue L, Xu H, Zhang C. Co-delivery of erlotinib and doxorubicin by pH-sensitive charge conversion nanocarrier for synergistic therapy. J Control Release 2016; 229:80-92. [DOI: 10.1016/j.jconrel.2016.03.001] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Revised: 02/04/2016] [Accepted: 03/01/2016] [Indexed: 12/29/2022]
|
14
|
Fei F, Joo EJ, Tarighat SS, Schiffer I, Paz H, Fabbri M, Abdel-Azim H, Groffen J, Heisterkamp N. B-cell precursor acute lymphoblastic leukemia and stromal cells communicate through Galectin-3. Oncotarget 2016; 6:11378-94. [PMID: 25869099 PMCID: PMC4484463 DOI: 10.18632/oncotarget.3409] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Accepted: 02/19/2015] [Indexed: 12/11/2022] Open
Abstract
The molecular interactions between B-cell precursor acute lymphoblastic leukemia (pre-B ALL) cells and stromal cells in the bone marrow that provide microenvironmentally-mediated protection against therapeutic drugs are not well-defined. Galectin-3 (Lgals3) is a multifunctional galactose-binding lectin with reported location in the nucleus, cytoplasm and extracellular space in different cell types. We previously reported that ALL cells co-cultured with stroma contain high levels of Galectin-3. We here establish that, in contrast to more mature B-lineage cancers, Galectin-3 detected in and on the ALL cells originates from stromal cells, which express it on their surface, secrete it as soluble protein and also in exosomes. Soluble and stromal-bound Galectin-3 is internalized by ALL cells, transported to the nucleus and stimulates transcription of endogenous LGALS3 mRNA. When human and mouse ALL cells develop tolerance to different drugs while in contact with protective stromal cells, Galectin-3 protein levels are consistently increased. This correlates with induction of Galectin-3 transcription in the ALL cells. Thus Galectin-3 sourced from stroma becomes supplemented by endogenous Galectin-3 production in the pre-B ALL cells that are under continuous stress from drug treatment. Our data suggest that stromal Galectin-3 may protect ALL cells through auto-induction of Galectin-3 mRNA and tonic NFκB pathway activation. Since endogenously synthesized Galectin-3 protects pre-B ALL cells against drug treatment, we identify Galectin-3 as one possible target to counteract the protective effects of stroma.
Collapse
Affiliation(s)
- Fei Fei
- Section of Molecular Carcinogenesis, Division of Hematology/Oncology and Bone Marrow Transplant, The Saban Research Institute of Children's Hospital Los Angeles, Los Angeles, CA, USA.,Division of Hematology/Oncology and Bone Marrow Transplant, Children's Hospital Los Angeles, Los Angeles, CA, USA
| | - Eun Ji Joo
- Section of Molecular Carcinogenesis, Division of Hematology/Oncology and Bone Marrow Transplant, The Saban Research Institute of Children's Hospital Los Angeles, Los Angeles, CA, USA.,Division of Hematology/Oncology and Bone Marrow Transplant, Children's Hospital Los Angeles, Los Angeles, CA, USA
| | - Somayeh S Tarighat
- Section of Molecular Carcinogenesis, Division of Hematology/Oncology and Bone Marrow Transplant, The Saban Research Institute of Children's Hospital Los Angeles, Los Angeles, CA, USA.,Division of Hematology/Oncology and Bone Marrow Transplant, Children's Hospital Los Angeles, Los Angeles, CA, USA
| | - Isabelle Schiffer
- Section of Molecular Carcinogenesis, Division of Hematology/Oncology and Bone Marrow Transplant, The Saban Research Institute of Children's Hospital Los Angeles, Los Angeles, CA, USA.,Division of Hematology/Oncology and Bone Marrow Transplant, Children's Hospital Los Angeles, Los Angeles, CA, USA
| | - Helicia Paz
- Section of Molecular Carcinogenesis, Division of Hematology/Oncology and Bone Marrow Transplant, The Saban Research Institute of Children's Hospital Los Angeles, Los Angeles, CA, USA.,Division of Hematology/Oncology and Bone Marrow Transplant, Children's Hospital Los Angeles, Los Angeles, CA, USA
| | - Muller Fabbri
- Department of Pediatrics, Molecular Microbiology and Immunology, Keck School of Medicine, Norris Comprehensive Cancer Center, Children's Center for Cancer and Blood Diseases, Children's Hospital Los Angeles, University of Southern California, Los Angeles, CA, USA
| | - Hisham Abdel-Azim
- Division of Hematology/Oncology and Bone Marrow Transplant, Children's Hospital Los Angeles, Los Angeles, CA, USA
| | - John Groffen
- Section of Molecular Carcinogenesis, Division of Hematology/Oncology and Bone Marrow Transplant, The Saban Research Institute of Children's Hospital Los Angeles, Los Angeles, CA, USA.,Division of Hematology/Oncology and Bone Marrow Transplant, Children's Hospital Los Angeles, Los Angeles, CA, USA.,Leukemia and Lymphoma Program, Norris Comprehensive Cancer Center, Los Angeles, CA, USA.,Departments of Pediatrics and Pathology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Nora Heisterkamp
- Section of Molecular Carcinogenesis, Division of Hematology/Oncology and Bone Marrow Transplant, The Saban Research Institute of Children's Hospital Los Angeles, Los Angeles, CA, USA.,Division of Hematology/Oncology and Bone Marrow Transplant, Children's Hospital Los Angeles, Los Angeles, CA, USA.,Leukemia and Lymphoma Program, Norris Comprehensive Cancer Center, Los Angeles, CA, USA.,Departments of Pediatrics and Pathology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
15
|
Chen MK, Hung MC. Proteolytic cleavage, trafficking, and functions of nuclear receptor tyrosine kinases. FEBS J 2015; 282:3693-721. [PMID: 26096795 DOI: 10.1111/febs.13342] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Revised: 05/18/2015] [Accepted: 06/09/2015] [Indexed: 01/18/2023]
Abstract
Intracellular localization has been reported for over three-quarters of receptor tyrosine kinase (RTK) families in response to environmental stimuli. Internalized RTK may bind to non-canonical substrates and affect various cellular processes. Many of the intracellular RTKs exist as fragmented forms that are generated by γ-secretase cleavage of the full-length receptor, shedding, alternative splicing, or alternative translation initiation. Soluble RTK fragments are stabilized and intracellularly transported into subcellular compartments, such as the nucleus, by binding to chaperone or transcription factors, while membrane-bound RTKs (full-length or truncated) are transported from the plasma membrane to the ER through the well-established Rab- or clathrin adaptor protein-coated vesicle retrograde trafficking pathways. Subsequent nuclear transport of membrane-bound RTK may occur via two pathways, INFS or INTERNET, with the former characterized by release of receptors from the ER into the cytosol and the latter characterized by release of membrane-bound receptor from the ER into the nucleoplasm through the inner nuclear membrane. Although most non-canonical intracellular RTK signaling is related to transcriptional regulation, there may be other functions that have yet to be discovered. In this review, we summarize the proteolytic processing, intracellular trafficking and nuclear functions of RTKs, and discuss how they promote cancer progression, and their clinical implications.
Collapse
Affiliation(s)
- Mei-Kuang Chen
- The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, TX, USA.,Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mien-Chie Hung
- The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, TX, USA.,Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Center of Molecular Medicine and Graduate Institute of Cancer Biology, China Medical University, Taichung, Taiwan.,Department of Biotechnology, Asia University, Taichung, Taiwan
| |
Collapse
|
16
|
O'Kelly I. Endocytosis as a mode to regulate functional expression of two-pore domain potassium (K₂p) channels. Pflugers Arch 2014; 467:1133-42. [PMID: 25413469 PMCID: PMC4428836 DOI: 10.1007/s00424-014-1641-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2014] [Revised: 10/22/2014] [Accepted: 10/24/2014] [Indexed: 11/06/2022]
Abstract
Two-pore domain potassium (K2P) channels are implicated in an array of physiological and pathophysiological roles. As a result of their biophysical properties, these channels produce a background leak K+ current which has a direct effect on cellular membrane potential and activity. The regulation of potassium leak from cells through K2P channels is of critical importance to cell function, development and survival. Controlling the cell surface expression of these channels is one mode to regulate their function and is achieved through a balance between regulated channel delivery to and retrieval from the cell surface. Here, we explore the modes of retrieval of K2P channels from the plasma membrane and observe that K2P channels are endocytosed in both a clathrin-mediated and clathrin-independent manner. K2P channels use a variety of pathways and show altered internalisation and sorting in response to external cues. These pathways working in concert, equip the cell with a range of approaches to maintain steady state levels of channels and to respond rapidly should changes in channel density be required.
Collapse
Affiliation(s)
- Ita O'Kelly
- Human Development and Health, Centre for Human Development, Stem Cells and Regeneration, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD, UK, I.M.O'
| |
Collapse
|
17
|
Bertelsen V, Stang E. The Mysterious Ways of ErbB2/HER2 Trafficking. MEMBRANES 2014; 4:424-46. [PMID: 25102001 PMCID: PMC4194043 DOI: 10.3390/membranes4030424] [Citation(s) in RCA: 127] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/27/2014] [Revised: 07/01/2014] [Accepted: 07/22/2014] [Indexed: 12/14/2022]
Abstract
The EGFR- or ErbB-family of receptor tyrosine kinases consists of EGFR/ErbB1, ErbB2/HER2, ErbB3/HER3 and ErbB4/HER4. Receptor activation and downstream signaling are generally initiated upon ligand-induced receptor homo- or heterodimerization at the plasma membrane, and endocytosis and intracellular membrane transport are crucial for regulation of the signaling outcome. Among the receptors, ErbB2 is special in several ways. Unlike the others, ErbB2 has no known ligand, but is still the favored dimerization partner. Furthermore, while the other receptors are down-regulated either constitutively or upon ligand-binding, ErbB2 is resistant to down-regulation, and also inhibits down-regulation of its partner upon heterodimerization. The reason(s) why ErbB2 is resistant to down-regulation are the subject of debate. Contrary to other ErbB-proteins, mature ErbB2 needs Hsp90 as chaperone. Several data suggest that Hsp90 is an important regulator of factors like ErbB2 stability, dimerization and/or signaling. Hsp90 inhibitors induce degradation of ErbB2, but whether Hsp90 directly makes ErbB2 endocytosis resistant is unclear. Exposure to anti-ErbB2 antibodies can also induce down-regulation of ErbB2. Down-regulation induced by Hsp90 inhibitors or antibodies does at least partly involve internalization and endosomal sorting to lysosomes for degradation, but also retrograde trafficking to the nucleus has been reported. In this review, we will discuss different molecular mechanisms suggested to be important for making ErbB2 resistant to down-regulation, and review how membrane trafficking is involved when down-regulation and/or relocalization of ErbB2 is induced.
Collapse
Affiliation(s)
- Vibeke Bertelsen
- Department of Pathology, Oslo University Hospital, Rikshospitalet, Post Box 4950 Nydalen, 0424 Oslo, Norway.
| | - Espen Stang
- Department of Pathology, Oslo University Hospital, Rikshospitalet, Post Box 4950 Nydalen, 0424 Oslo, Norway.
| |
Collapse
|
18
|
Song S, Rosen KM, Corfas G. Biological function of nuclear receptor tyrosine kinase action. Cold Spring Harb Perspect Biol 2013; 5:5/7/a009001. [PMID: 23818495 DOI: 10.1101/cshperspect.a009001] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Receptor tyrosine kinases (RTKs) were believed until recently to act at the cell membrane in a singular fashion (i.e., binding of ligands on the extracellular domain would activate the intrinsic tyrosine kinase activity in the intracellular domain), which would then start a cascade involving other intracellular signaling molecules that would act as effectors. However, new evidence indicates that some RTKs can signal through a different modality; they can move into the nucleus where they directly exert their actions. Although some studies have showed that the proteolytically released intracellular domain of several RTKs can move to the nucleus where they influence gene expression and cell function, others suggest that RTKs can also move to the nucleus as holoproteins. The identification of this novel signaling mechanism calls for a critical reevaluation of the mechanisms of action of RTKs and their biological roles.
Collapse
Affiliation(s)
- Sungmin Song
- FM Kirby Neurobiology Center, Children's Hospital Boston, Massachusetts 02115, USA
| | | | | |
Collapse
|
19
|
Hiscox S, Baruha B, Smith C, Bellerby R, Goddard L, Jordan N, Poghosyan Z, Nicholson RI, Barrett-Lee P, Gee J. Overexpression of CD44 accompanies acquired tamoxifen resistance in MCF7 cells and augments their sensitivity to the stromal factors, heregulin and hyaluronan. BMC Cancer 2012; 12:458. [PMID: 23039365 PMCID: PMC3517483 DOI: 10.1186/1471-2407-12-458] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2012] [Accepted: 09/20/2012] [Indexed: 12/20/2022] Open
Abstract
Background Acquired resistance to endocrine therapy in breast cancer is a significant problem with relapse being associated with local and/or regional recurrence and frequent distant metastases. Breast cancer cell models reveal that endocrine resistance is accompanied by a gain in aggressive behaviour driven in part through altered growth factor receptor signalling, particularly involving erbB family receptors. Recently we identified that CD44, a transmembrane cell adhesion receptor known to interact with growth factor receptors, is upregulated in tamoxifen-resistant (TamR) MCF7 breast cancer cells. The purpose of this study was to explore the consequences of CD44 upregulation in an MCF7 cell model of acquired tamoxifen resistance, specifically with respect to the hypothesis that CD44 may influence erbB activity to promote an adverse phenotype. Methods CD44 expression in MCF7 and TamR cells was assessed by RT-PCR, Western blotting and immunocytochemistry. Immunofluorescence and immunoprecipitation studies revealed CD44-erbB associations. TamR cells (± siRNA-mediated CD44 suppression) or MCF7 cells (± transfection with the CD44 gene) were treated with the CD44 ligand, hyaluronon (HA), or heregulin and their in vitro growth (MTT), migration (Boyden chamber and wound healing) and invasion (Matrigel transwell migration) determined. erbB signalling was assessed using Western blotting. The effect of HA on erbB family dimerisation in TamR cells was determined by immunoprecipitation in the presence or absence of CD44 siRNA. Results TamR cells overexpressed CD44 where it was seen to associate with erbB2 at the cell surface. siRNA-mediated suppression of CD44 in TamR cells significantly attenuated their response to heregulin, inhibiting heregulin-induced cell migration and invasion. Furthermore, TamR cells exhibited enhanced sensitivity to HA, with HA treatment resulting in modulation of erbB dimerisation, ligand-independent activation of erbB2 and EGFR and induction of cell migration. Overexpression of CD44 in MCF7 cells, which lack endogenous CD44, generated an HA-sensitive phenotype, with HA-stimulation promoting erbB/EGFR activation and migration. Conclusions These data suggest an important role for CD44 in the context of tamoxifen-resistance where it may augment cellular response to erbB ligands and HA, factors that are reported to be present within the tumour microenvironment in vivo. Thus CD44 may present an important determinant of breast cancer progression in the setting of endocrine resistance.
Collapse
Affiliation(s)
- Stephen Hiscox
- Welsh School of Pharmacy, Cardiff University, Wales, UK.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Cetuximab in combination with anti-human IgG antibodies efficiently down-regulates the EGF receptor by macropinocytosis. Exp Cell Res 2012; 318:2578-91. [PMID: 22975728 DOI: 10.1016/j.yexcr.2012.09.001] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2012] [Revised: 08/31/2012] [Accepted: 09/04/2012] [Indexed: 02/05/2023]
Abstract
The monoclonal antibody C225 (Cetuximab) blocks binding of ligand to the epidermal growth factor receptor (EGFR). In addition, it is known that incubation with C225 induces endocytosis of the EGFR. This endocytosis has previously been shown to be increased when C225 is combined with an additional monoclonal anti-EGFR antibody. However, the effects of antibody combinations on EGFR activation, endocytosis, trafficking and degradation have been unclear. By binding a secondary antibody to the C225-EGFR complex, we here demonstrate that a combination of antibodies can efficiently internalize and degrade the EGFR. Although the combination of antibodies activated the EGFR kinase and induced ubiquitination of the EGFR, the kinase activity was not required for internalization of the EGFR. In contrast to EGF-induced EGFR down-regulation, the antibody combination efficiently degraded the EGFR without initiating downstream proliferative signaling. The antibody-induced internalization of EGFR was found not to depend on clathrin and/or dynamin, but depended on actin polymerization, suggesting induction of macropinocytosis. Macropinocytosis may cause internalization of large membrane areas, and this could explain the highly efficient internalization of the EGFR induced by combination of antibodies.
Collapse
|
21
|
Schmees C, Villaseñor R, Zheng W, Ma H, Zerial M, Heldin CH, Hellberg C. Macropinocytosis of the PDGF β-receptor promotes fibroblast transformation by H-RasG12V. Mol Biol Cell 2012; 23:2571-82. [PMID: 22573884 PMCID: PMC3386220 DOI: 10.1091/mbc.e11-04-0317] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Fibroblast transformation by H-RasG12V induces internalization of PDGFRβ by macropinocytosis, enhancing its signaling activity and increasing anchorage-independent proliferation. It is proposed that H-Ras transformation promotes tumor progression by enhancing growth factor receptor signaling through increased receptor macropinocytosis. Receptor tyrosine kinase (RTK) signaling is frequently increased in tumor cells, sometimes as a result of decreased receptor down-regulation. The extent to which the endocytic trafficking routes can contribute to such RTK hyperactivation is unclear. Here, we show for the first time that fibroblast transformation by H-RasG12V induces the internalization of platelet-derived growth factor β-receptor (PDGFRβ) by macropinocytosis, enhancing its signaling activity and increasing anchorage-independent proliferation. H-RasG12V transformation and PDGFRβ activation were synergistic in stimulating phosphatidylinositol (PI) 3-kinase activity, leading to receptor macropinocytosis. PDGFRβ macropinocytosis was both necessary and sufficient for enhanced receptor activation. Blocking macropinocytosis by inhibition of PI 3-kinase prevented the increase in receptor activity in transformed cells. Conversely, increasing macropinocytosis by Rabankyrin-5 overexpression was sufficient to enhance PDGFRβ activation in nontransformed cells. Simultaneous stimulation with PDGF-BB and epidermal growth factor promoted macropinocytosis of both receptors and increased their activation in nontransformed cells. We propose that H-Ras transformation promotes tumor progression by enhancing growth factor receptor signaling as a result of increased receptor macropinocytosis.
Collapse
Affiliation(s)
- C Schmees
- Ludwig Institute for Cancer Research, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | | | | | | | | | | | | |
Collapse
|
22
|
De Angelis Campos AC, Rodrigues MA, de Andrade C, de Goes AM, Nathanson MH, Gomes DA. Epidermal growth factor receptors destined for the nucleus are internalized via a clathrin-dependent pathway. Biochem Biophys Res Commun 2011; 412:341-6. [PMID: 21821003 DOI: 10.1016/j.bbrc.2011.07.100] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2011] [Accepted: 07/22/2011] [Indexed: 10/17/2022]
Abstract
The epidermal growth factor (EGF) transduces its actions via the EGF receptor (EGFR), which can traffic from the plasma membrane to either the cytoplasm or the nucleus. However, the mechanism by which EGFR reaches the nucleus is unclear. To investigate these questions, liver cells were analyzed by immunoblot of cell fractions, confocal immunofluorescence and real time confocal imaging. Cell fractionation studies showed that EGFR was detectable in the nucleus after EGF stimulation with a peak in nuclear receptor after 10 min. Movement of EGFR to the nucleus was confirmed by confocal immunofluorescence and labeled EGF moved with the receptor to the nucleus. Small interference RNA (siRNA) was used to knockdown clathrin in order to assess the first endocytic steps of EGFR nuclear translocation in liver cells. A mutant dynamin (dynamin K44A) was also used to determine the pathways for this traffic. Movement of labeled EGF or EGFR to the nucleus depended upon dynamin and clathrin. This identifies the pathway that mediates the first steps for EGFR nuclear translocation in liver cells.
Collapse
|