1
|
Tanaka M, Chuaychob S, Homme M, Yamazaki Y, Lyu R, Yamashita K, Ae K, Matsumoto S, Kumegawa K, Maruyama R, Qu W, Miyagi Y, Yokokawa R, Nakamura T. ASPSCR1::TFE3 orchestrates the angiogenic program of alveolar soft part sarcoma. Nat Commun 2023; 14:1957. [PMID: 37029109 PMCID: PMC10082046 DOI: 10.1038/s41467-023-37049-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 03/01/2023] [Indexed: 04/09/2023] Open
Abstract
Alveolar soft part sarcoma (ASPS) is a soft part malignancy affecting adolescents and young adults. ASPS is characterized by a highly integrated vascular network, and its high metastatic potential indicates the importance of ASPS's prominent angiogenic activity. Here, we find that the expression of ASPSCR1::TFE3, the fusion transcription factor causatively associated with ASPS, is dispensable for in vitro tumor maintenance; however, it is required for in vivo tumor development via angiogenesis. ASPSCR1::TFE3 is frequently associated with super-enhancers (SEs) upon its DNA binding, and the loss of its expression induces SE-distribution dynamic modification related to genes belonging to the angiogenesis pathway. Using epigenomic CRISPR/dCas9 screening, we identify Pdgfb, Rab27a, Sytl2, and Vwf as critical targets associated with reduced enhancer activities due to the ASPSCR1::TFE3 loss. Upregulation of Rab27a and Sytl2 promotes angiogenic factor-trafficking to facilitate ASPS vascular network construction. ASPSCR1::TFE3 thus orchestrates higher ordered angiogenesis via modulating the SE activity.
Collapse
Affiliation(s)
- Miwa Tanaka
- Division of Carcinogenesis, The Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan.
- Department of Experimental Pathology, Institute of Medical Science, Tokyo Medical University, Tokyo, Japan.
- Project for Cancer Epigenomics, The Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan.
| | - Surachada Chuaychob
- Department of Micro Engineering, Graduate School of Engineering, Kyoto University, Kyoto, Japan
| | - Mizuki Homme
- Division of Carcinogenesis, The Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan
- Division of Cell Biology, The Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Yukari Yamazaki
- Division of Carcinogenesis, The Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan
- Department of Experimental Pathology, Institute of Medical Science, Tokyo Medical University, Tokyo, Japan
| | - Ruyin Lyu
- Department of Micro Engineering, Graduate School of Engineering, Kyoto University, Kyoto, Japan
| | - Kyoko Yamashita
- Division of Pathology, The Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Keisuke Ae
- Department of Orthopedic Oncology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Seiichi Matsumoto
- Department of Orthopedic Oncology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Kohei Kumegawa
- Project for Cancer Epigenomics, The Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Reo Maruyama
- Project for Cancer Epigenomics, The Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Wei Qu
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Japan
| | - Yohei Miyagi
- Molecular Pathology and Genetics Division, Kanagawa Cancer Center Research Institute, Yokohama, Japan
| | - Ryuji Yokokawa
- Department of Micro Engineering, Graduate School of Engineering, Kyoto University, Kyoto, Japan
| | - Takuro Nakamura
- Division of Carcinogenesis, The Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan.
- Department of Experimental Pathology, Institute of Medical Science, Tokyo Medical University, Tokyo, Japan.
| |
Collapse
|
2
|
Kohl C, Aung T, Haerteis S, Ignatov A, Ortmann O, Papathemelis T. The 3D in vivo chorioallantoic membrane model and its role in breast cancer research. J Cancer Res Clin Oncol 2022; 148:1033-1043. [PMID: 35122110 DOI: 10.1007/s00432-022-03936-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 01/24/2022] [Indexed: 02/06/2023]
Abstract
PURPOSE We aimed to evaluate the role of the chorioallantoic membrane model (CAM) in breast cancer research. METHODS The following is an overview of the use of the CAM in the field of breast cancer research based on a PubMed literature query. RESULTS The CAM is a 3D in vivo model that can be used for the analysis of tumor growth, biology and angiogenesis of primary tumor tissue or tumor cell lines. The CAM model has been used in breast cancer research for drug testing, migration assays and the evaluation of vascularization, amongst others. The CAM model is a valuable method that offers a better imitation of the physiological phenomena compared to 2D or 3D in vitro models. CONCLUSION The CAM model has primarily and successfully been utilized for the assessment of the tumor biology of established breast cancer cell lines. Further, the CAM model is a promising method to analyze patient derived primary tumor material and could be used as a "patient-specific 3D-tumor-therapy-model" for the cost-efficient evaluation of anti-cancer drugs to find the optimal treatment for breast cancer patients.
Collapse
Affiliation(s)
- Cynthia Kohl
- Department of Gynecology and Obstetrics, St. Marien Hospital Amberg, 92224, Amberg, Germany.
| | - Thiha Aung
- Institute for Molecular and Cellular Anatomy, University of Regensburg, 93053, Regensburg, Germany.,Faculty of Applied Healthcare Science, Deggendorf Institute of Technology, 94469, Deggendorf, Germany
| | - Silke Haerteis
- Institute for Molecular and Cellular Anatomy, University of Regensburg, 93053, Regensburg, Germany
| | - Atanas Ignatov
- Department of Gynecology and Obstetrics, University Hospital Magdeburg, 39120, Magdeburg, Germany
| | - Olaf Ortmann
- Department of Gynecology and Obstetrics, University Medical Center Regensburg, 93053, Regensburg, Germany
| | - Thomas Papathemelis
- Department of Gynecology and Obstetrics, St. Marien Hospital Amberg, 92224, Amberg, Germany
| |
Collapse
|
3
|
Liu Z, Xiong J, Gao S, Zhu MX, Sun K, Li M, Zhang G, Li YP. Ameliorating cancer cachexia by inhibiting cancer cell release of Hsp70 and Hsp90 with omeprazole. J Cachexia Sarcopenia Muscle 2022; 13:636-647. [PMID: 34729960 PMCID: PMC8818607 DOI: 10.1002/jcsm.12851] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 09/27/2021] [Accepted: 09/30/2021] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Cancer cachexia, characterized by muscle and fat tissue wasting, is a major determinant of cancer-related mortality without established treatment. Recent animal data revealed that cancer cells induce muscle wasting by releasing Hsp70 and Hsp90 as surface proteins on extracellular vesicles (EVs). Here, we test a therapeutic strategy for ameliorating cancer cachexia by inhibiting the release of Hsp70 and Hsp90 using proton pump inhibitor omeprazole. METHODS Omeprazole effect on Hsp70/90 release through EVs by Lewis lung carcinoma (LLC) cells in vitro, serum levels of Hsp70/90 and Hsp70/90-carrying EVs in LLC tumour-bearing mice, and LLC-induced muscle protein degradation pathways in C2C12 myotubes and mice were determined. Omeprazole effect on endolysosomal pH and Rab27b expression in LLC cells were analysed. RESULTS Omeprazole treatment of LLC cells inhibited Hsp70/90 and Hsp70/90-carrying EV release in a dose-dependent manner (1 to 10 μM) and attenuated the catabolic activity of LLC cell-conditioned medium on C2C12 myotubes. Systemic omeprazole administration to LLC tumour-bearing mice (5 mg/kg/day subcutaneously) for 2 weeks blocked elevation of serum Hsp70, Hsp90, and Hsp70/90-carrying EVs, abrogated skeletal muscle catabolism, and prevented loss of muscle function as well as muscle and epididymal fat mass without altering tumour growth. Consequently, median survival increased by 23.3%. Mechanistically, omeprazole increased cancer cell endolysosomal pH level dose-dependently (0.1 to 1 μM) by inhibiting vacuolar H+ -ATPase. Further, omeprazole suppressed the highly elevated expression of Rab27b, a key regulator of EV release, in LLC cells. CONCLUSIONS Omeprazole ameliorates cancer cachexia by inhibiting cancer cell release of Hsp70 and Hsp90.
Collapse
Affiliation(s)
- Zhelong Liu
- Department of Integrative Biology and Pharmacology, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Jian Xiong
- Department of Integrative Biology and Pharmacology, The University of Texas Health Science Center at Houston, Houston, TX, USA.,Biochemistry and Cell Biology Program, MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Song Gao
- Department of Integrative Biology and Pharmacology, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Michael X Zhu
- Department of Integrative Biology and Pharmacology, The University of Texas Health Science Center at Houston, Houston, TX, USA.,Biochemistry and Cell Biology Program, MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Kai Sun
- Department of Integrative Biology and Pharmacology, The University of Texas Health Science Center at Houston, Houston, TX, USA.,Biochemistry and Cell Biology Program, MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA.,The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, University of Texas Health Science Center, Houston, TX, USA
| | - Min Li
- Department of Integrative Biology and Pharmacology, The University of Texas Health Science Center at Houston, Houston, TX, USA.,The Vivian L. Smith Department of Neurosurgery, The University of Texas Health Science Center at Houston, Houston, TX, USA.,Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.,Department of Surgery, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Guohua Zhang
- Department of Integrative Biology and Pharmacology, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Yi-Ping Li
- Department of Integrative Biology and Pharmacology, The University of Texas Health Science Center at Houston, Houston, TX, USA.,Biochemistry and Cell Biology Program, MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| |
Collapse
|
4
|
Chen F, Kang R, Liu J, Tang D. The V-ATPases in cancer and cell death. Cancer Gene Ther 2022; 29:1529-1541. [PMID: 35504950 PMCID: PMC9063253 DOI: 10.1038/s41417-022-00477-y] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 04/07/2022] [Accepted: 04/21/2022] [Indexed: 02/04/2023]
Abstract
Transmembrane ATPases are membrane-bound enzyme complexes and ion transporters that can be divided into F-, V-, and A-ATPases according to their structure. The V-ATPases, also known as H+-ATPases, are large multi-subunit protein complexes composed of a peripheral domain (V1) responsible for the hydrolysis of ATP and a membrane-integrated domain (V0) that transports protons across plasma membrane or organelle membrane. V-ATPases play a fundamental role in maintaining pH homeostasis through lysosomal acidification and are involved in modulating various physiological and pathological processes, such as macropinocytosis, autophagy, cell invasion, and cell death (e.g., apoptosis, anoikis, alkaliptosis, ferroptosis, and lysosome-dependent cell death). In addition to participating in embryonic development, V-ATPase pathways, when dysfunctional, are implicated in human diseases, such as neurodegenerative diseases, osteopetrosis, distal renal tubular acidosis, and cancer. In this review, we summarize the structure and regulation of isoforms of V-ATPase subunits and discuss their context-dependent roles in cancer biology and cell death. Updated knowledge about V-ATPases may enable us to design new anticancer drugs or strategies.
Collapse
Affiliation(s)
- Fangquan Chen
- grid.417009.b0000 0004 1758 4591DAMP Laboratory, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510120 China
| | - Rui Kang
- grid.267313.20000 0000 9482 7121Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX 75390 USA
| | - Jiao Liu
- grid.417009.b0000 0004 1758 4591DAMP Laboratory, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510120 China
| | - Daolin Tang
- grid.267313.20000 0000 9482 7121Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX 75390 USA
| |
Collapse
|
5
|
Ghazy E, Taghi HS. The Autophagy-Inducing Mechanisms of Vitexin, Cinobufacini, and Physalis alkekengi Hydroalcoholic Extract against Breast Cancer in vitro and in vivo. J Gastrointest Cancer 2021; 53:592-596. [PMID: 34287803 DOI: 10.1007/s12029-021-00668-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/08/2021] [Indexed: 10/20/2022]
Abstract
OBJECTIVES Owing to inefficiency of chemotherapy towards cancer treatment, formulation and application of herbal drug compounds will open new avenues with this regard. In this study, the anticancer effects of itexin, cinobufacini, and Physalis alkekengi (P. alkekengi) were assessed. METHODS Herein, synergistic effects of vitexin, cinobufacini, and P. alkekengi hydroalcoholic extract were assessed against estrogen-receptor (EGFR2)-positive breast cancer mouse model. Sixty ER + breast cancer BALB/c mice (six groups each including ten members) were included. The anticancer effects of P. alkekengi hydroalcoholic extract, vitexin, and cinobufacini were administered against EGFR2 cancerous cells for 14 days. The tumor size, cytotoxic effects, and expression of Beclin-1, LC3-II, and ATG5 autophagy-related genes were investigated using RT-qPCR technique. The data was analyzed using chi-square, ANOVA, and multinomial logistic regression tests. KEY FINDINGS The 50% lethal dose (LD50) of P. alkekengi and vitexin against the breast cancer cells included 12 mg/kg, respectively, while cinobufacini LD50 was 24 mg/kg but had no toxicity against CRL7242 breast normal cells. Furthermore, 24 mg/kg of the P. alkekengi, vitexin, and cinobufacini significantly increased the ATG5, Beclin-1, and LC3-II gene expression. CONCLUSION Considering anticancer effects of P. alkekengi, vitexin, and cinobufacini against breast cancer through induction of the autophagy pathway, the compound formulations can be applied as anticancer therapies.
Collapse
Affiliation(s)
- Esraa Ghazy
- Department of Pharmacy, Al-Rasheed University College, Baghdad, Iraq.
| | | |
Collapse
|
6
|
Li R, Dong C, Jiang K, Sun R, Zhou Y, Yin Z, Lv J, Zhang J, Wang Q, Wang L. Rab27B enhances drug resistance in hepatocellular carcinoma by promoting exosome-mediated drug efflux. Carcinogenesis 2021; 41:1583-1591. [PMID: 32390047 DOI: 10.1093/carcin/bgaa029] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 03/08/2020] [Accepted: 03/23/2020] [Indexed: 01/06/2023] Open
Abstract
Liver cancer is a major threat to human life and health, and chemotherapy has been the standard non-surgical treatment for liver cancer. However, the emergence of drug resistance of liver cancer cells has hindered the therapeutic effect of chemical drugs. The discovery of exosomes has provided new insights into the mechanisms underlying tumour cell resistance. In this study, we aimed to determine the proteins associated with drug resistance in tumour cells and to elucidate the underlying mechanisms. We found that Rab27B expression in drug (5-fluorouracil, 5Fu)-resistant Bel7402 (Bel/5Fu) cells increased significantly compared with that in drug-sensitive Bel7402 cells. In addition, Bel/5Fu cells secreted more exosomes under 5Fu stimulation. The number of exosomes secreted by Bel/5Fu cells significantly reduced after knocking down Rab27B, and the cellular concentration of 5Fu increased, enhancing its therapeutic effect. We also found that the administration of classical drug efflux pump (P-glycoprotein, P-gp) inhibitors together with knockdown of Rab27B further improved the therapeutic effects of chemotherapy drugs. In conclusion, our findings suggest that Rab27B could be a new therapeutic target in liver cancer.
Collapse
Affiliation(s)
- Rui Li
- Division of Hepatobiliary and Pancreatic Surgery, Department of General Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Chengyong Dong
- Division of Hepatobiliary and Pancreatic Surgery, Department of General Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Keqiu Jiang
- Division of Hepatobiliary and Pancreatic Surgery, Department of General Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Rui Sun
- Division of Hepatobiliary and Pancreatic Surgery, Department of General Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Yang Zhou
- Liaoning Clinical Research Center for Lung Cancer, The Second Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Zeli Yin
- Division of Hepatobiliary and Pancreatic Surgery, Department of General Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China.,Engineering Research Center for New Materials and Precision Treatment Technology of Malignant Tumors Therapy, Dalian Medical University, Dalian, Liaoning, China
| | - Jiaxin Lv
- Department of Anesthesiology, The First Affiliated Hospital of Dalian Medical University, Zhongshan Road, Dalian, Liaoning, China
| | - Junlin Zhang
- Division of Hepatobiliary and Pancreatic Surgery, Department of General Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Qi Wang
- Department of Respiratory Medicine, The Second Hospital, Dalian Medical University, Dalian, Liaoning, China
| | - Liming Wang
- Division of Hepatobiliary and Pancreatic Surgery, Department of General Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| |
Collapse
|
7
|
Bertolini I, Storaci AM, Terrasi A, Cristofori AD, Locatelli M, Caroli M, Ferrero S, Altieri DC, Vaira V. Interplay Between V-ATPase G1 and Small EV-miRNAs Modulates ERK1/2 Activation in GBM Stem Cells and Nonneoplastic Milieu. Mol Cancer Res 2020; 18:1744-1754. [PMID: 32753475 DOI: 10.1158/1541-7786.mcr-20-0078] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 04/17/2020] [Accepted: 07/24/2020] [Indexed: 12/15/2022]
Abstract
The ATP6V1G1 subunit (V1G1) of the vacuolar proton ATPase (V-ATPase) pump is crucial for glioma stem cells (GSC) maintenance and in vivo tumorigenicity. Moreover, V-ATPase reprograms the tumor microenvironment through acidification and release of extracellular vesicles (EV). Therefore, we investigated the role of V1G1 in GSC small EVs and their effects on primary brain cultures. To this end, small EVs were isolated from patients-derived GSCs grown as neurospheres (NS) with high (V1G1HIGH-NS) or low (V1G1LOW-NS) V1G1 expression and analyzed for V-ATPase subunits presence, miRNA contents, and cellular responses in recipient cultures. Our results show that NS-derived small EVs stimulate proliferation and motility of recipient cells, with small EV derived from V1G1HIGH-NS showing the most pronounced activity. This involved activation of ERK1/2 signaling, in a response reversed by V-ATPase inhibition in NS-producing small EV. The miRNA profile of V1G1HIGH-NS-derived small EVs differed significantly from that of V1G1LOW-NS, which included miRNAs predicted to target MAPK/ERK signaling. Mechanistically, forced expression of a MAPK-targeting pool of miRNAs in recipient cells suppressed MAPK/ERK pathway activation and blunted the prooncogenic effects of V1G1HIGH small EV. These findings propose that the GSC influences the brain milieu through a V1G1-coordinated EVs release of MAPK/ERK-targeting miRNAs. Interfering with V-ATPase activity could prevent ERK-dependent oncogenic reprogramming of the microenvironment, potentially hampering local GBM infiltration. IMPLICATIONS: Our data identify a novel molecular mechanism of gliomagenesis specific of the GBM stem cell niche, which coordinates a V-ATPase-dependent reprogramming of the brain microenvironment through the release of specialized EVs.
Collapse
Affiliation(s)
- Irene Bertolini
- Division of Pathology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Alessandra Maria Storaci
- Division of Pathology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.,Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Andrea Terrasi
- Division of Pathology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Andrea Di Cristofori
- Division of Neurosurgery, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Marco Locatelli
- Division of Neurosurgery, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Manuela Caroli
- Division of Neurosurgery, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Stefano Ferrero
- Division of Pathology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.,Department of Biomedical, Surgical and Dental Sciences University of Milan, Milan, Italy
| | - Dario C Altieri
- Immunology, Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, Pennsylvania
| | - Valentina Vaira
- Division of Pathology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.
| |
Collapse
|
8
|
Hraběta J, Belhajová M, Šubrtová H, Merlos Rodrigo MA, Heger Z, Eckschlager T. Drug Sequestration in Lysosomes as One of the Mechanisms of Chemoresistance of Cancer Cells and the Possibilities of Its Inhibition. Int J Mol Sci 2020; 21:ijms21124392. [PMID: 32575682 PMCID: PMC7352242 DOI: 10.3390/ijms21124392] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 06/17/2020] [Accepted: 06/18/2020] [Indexed: 12/12/2022] Open
Abstract
Resistance to chemotherapeutics and targeted drugs is one of the main problems in successful cancer therapy. Various mechanisms have been identified to contribute to drug resistance. One of those mechanisms is lysosome-mediated drug resistance. Lysosomes have been shown to trap certain hydrophobic weak base chemotherapeutics, as well as some tyrosine kinase inhibitors, thereby being sequestered away from their intracellular target site. Lysosomal sequestration is in most cases followed by the release of their content from the cell by exocytosis. Lysosomal accumulation of anticancer drugs is caused mainly by ion-trapping, but active transport of certain drugs into lysosomes was also described. Lysosomal low pH, which is necessary for ion-trapping is achieved by the activity of the V-ATPase. This sequestration can be successfully inhibited by lysosomotropic agents and V-ATPase inhibitors in experimental conditions. Clinical trials have been performed only with lysosomotropic drug chloroquine and their results were less successful. The aim of this review is to give an overview of lysosomal sequestration and expression of acidifying enzymes as yet not well known mechanism of cancer cell chemoresistance and about possibilities how to overcome this form of resistance.
Collapse
Affiliation(s)
- Jan Hraběta
- Department of Paediatric Haematology and Oncology, 2nd Faculty of Medicine, Charles University and Motol University Hospital, CZ-150 06 Prague, Czech Republic; (J.H.); (M.B.)
| | - Marie Belhajová
- Department of Paediatric Haematology and Oncology, 2nd Faculty of Medicine, Charles University and Motol University Hospital, CZ-150 06 Prague, Czech Republic; (J.H.); (M.B.)
| | - Hana Šubrtová
- Department of Chemistry and Biochemistry, Mendel University in Brno, CZ-613 00 Brno, Czech Republic; (H.Š.); (M.A.M.R.); (Z.H.)
| | - Miguel Angel Merlos Rodrigo
- Department of Chemistry and Biochemistry, Mendel University in Brno, CZ-613 00 Brno, Czech Republic; (H.Š.); (M.A.M.R.); (Z.H.)
- Central European Institute of Technologies, Brno University of Technology, CZ-612 00 Brno, Czech Republic
| | - Zbyněk Heger
- Department of Chemistry and Biochemistry, Mendel University in Brno, CZ-613 00 Brno, Czech Republic; (H.Š.); (M.A.M.R.); (Z.H.)
- Central European Institute of Technologies, Brno University of Technology, CZ-612 00 Brno, Czech Republic
| | - Tomáš Eckschlager
- Department of Paediatric Haematology and Oncology, 2nd Faculty of Medicine, Charles University and Motol University Hospital, CZ-150 06 Prague, Czech Republic; (J.H.); (M.B.)
- Correspondence: ; Tel.: +420-606-364-730
| |
Collapse
|
9
|
Almasi S, El Hiani Y. Exploring the Therapeutic Potential of Membrane Transport Proteins: Focus on Cancer and Chemoresistance. Cancers (Basel) 2020; 12:cancers12061624. [PMID: 32575381 PMCID: PMC7353007 DOI: 10.3390/cancers12061624] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 06/16/2020] [Indexed: 02/06/2023] Open
Abstract
Improving the therapeutic efficacy of conventional anticancer drugs represents the best hope for cancer treatment. However, the shortage of druggable targets and the increasing development of anticancer drug resistance remain significant problems. Recently, membrane transport proteins have emerged as novel therapeutic targets for cancer treatment. These proteins are essential for a plethora of cell functions ranging from cell homeostasis to clinical drug toxicity. Furthermore, their association with carcinogenesis and chemoresistance has opened new vistas for pharmacology-based cancer research. This review provides a comprehensive update of our current knowledge on the functional expression profile of membrane transport proteins in cancer and chemoresistant tumours that may form the basis for new cancer treatment strategies.
Collapse
Affiliation(s)
- Shekoufeh Almasi
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON KIH 8M5, Canada;
| | - Yassine El Hiani
- Department of Physiology and Biophysics, Faculty of Medicine, Dalhousie University, Halifax, NS B3H 4R2, Canada
- Correspondence:
| |
Collapse
|
10
|
Avagliano A, Fiume G, Pelagalli A, Sanità G, Ruocco MR, Montagnani S, Arcucci A. Metabolic Plasticity of Melanoma Cells and Their Crosstalk With Tumor Microenvironment. Front Oncol 2020; 10:722. [PMID: 32528879 PMCID: PMC7256186 DOI: 10.3389/fonc.2020.00722] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 04/16/2020] [Indexed: 12/21/2022] Open
Abstract
Cutaneous melanoma (CM) is a highly aggressive and drug resistant solid tumor, showing an impressive metabolic plasticity modulated by oncogenic activation. In particular, melanoma cells can generate adenosine triphosphate (ATP) during cancer progression by both cytosolic and mitochondrial compartments, although CM energetic request mostly relies on glycolysis. The upregulation of glycolysis is associated with constitutive activation of BRAF/MAPK signaling sustained by BRAFV600E kinase mutant. In this scenario, the growth and progression of CM are strongly affected by melanoma metabolic changes and interplay with tumor microenvironment (TME) that sustain tumor development and immune escape. Furthermore, CM metabolic plasticity can induce a metabolic adaptive response to BRAF/MEK inhibitors (BRAFi/MEKi), associated with the shift from glycolysis toward oxidative phosphorylation (OXPHOS). Therefore, in this review article we survey the metabolic alterations and plasticity of CM, its crosstalk with TME that regulates melanoma progression, drug resistance and immunosurveillance. Finally, we describe hallmarks of melanoma therapeutic strategies targeting the shift from glycolysis toward OXPHOS.
Collapse
Affiliation(s)
- Angelica Avagliano
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Giuseppe Fiume
- Department of Experimental and Clinical Medicine, University "Magna Graecia" of Catanzaro, Catanzaro, Italy
| | - Alessandra Pelagalli
- Department of Advanced Biomedical Sciences, University of Naples Federico II, Naples, Italy.,Institute of Biostructures and Bioimages, National Research Council, Naples, Italy
| | - Gennaro Sanità
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| | - Maria Rosaria Ruocco
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| | - Stefania Montagnani
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Alessandro Arcucci
- Department of Public Health, University of Naples Federico II, Naples, Italy
| |
Collapse
|
11
|
Ward C, Meehan J, Gray ME, Murray AF, Argyle DJ, Kunkler IH, Langdon SP. The impact of tumour pH on cancer progression: strategies for clinical intervention. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2020; 1:71-100. [PMID: 36046070 PMCID: PMC9400736 DOI: 10.37349/etat.2020.00005] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Accepted: 02/05/2020] [Indexed: 02/06/2023] Open
Abstract
Dysregulation of cellular pH is frequent in solid tumours and provides potential opportunities for therapeutic intervention. The acidic microenvironment within a tumour can promote migration, invasion and metastasis of cancer cells through a variety of mechanisms. Pathways associated with the control of intracellular pH that are under consideration for intervention include carbonic anhydrase IX, the monocarboxylate transporters (MCT, MCT1 and MCT4), the vacuolar-type H+-ATPase proton pump, and the sodium-hydrogen exchanger 1. This review will describe progress in the development of inhibitors to these targets.
Collapse
Affiliation(s)
- Carol Ward
- Cancer Research UK Edinburgh Centre and Edinburgh Pathology, Institute of Genetics and Molecular Medicine, University of Edinburgh, Crewe Road South, EH4 2XU Edinburgh, UK
| | - James Meehan
- Cancer Research UK Edinburgh Centre and Edinburgh Pathology, Institute of Genetics and Molecular Medicine, University of Edinburgh, Crewe Road South, EH4 2XU Edinburgh, UK
| | - Mark E Gray
- Royal (Dick) School of Veterinary Studies and Roslin Institute, University of Edinburgh, Easter Bush, EH25 9RG Midlothian, UK
| | - Alan F Murray
- School of Engineering, Institute for Integrated Micro and Nano Systems, EH9 3JL Edinburgh, UK
| | - David J Argyle
- Royal (Dick) School of Veterinary Studies and Roslin Institute, University of Edinburgh, Easter Bush, EH25 9RG Midlothian, UK
| | - Ian H Kunkler
- Cancer Research UK Edinburgh Centre and Edinburgh Pathology, Institute of Genetics and Molecular Medicine, University of Edinburgh, Crewe Road South, EH4 2XU Edinburgh, UK
| | - Simon P Langdon
- Cancer Research UK Edinburgh Centre and Edinburgh Pathology, Institute of Genetics and Molecular Medicine, University of Edinburgh, Crewe Road South, EH4 2XU Edinburgh, UK
| |
Collapse
|
12
|
Wang J, Chen D, Song W, Liu Z, Ma W, Li X, Zhang C, Wang X, Wang Y, Yang Y, Cao W, Qi L. ATP6L promotes metastasis of colorectal cancer by inducing epithelial-mesenchymal transition. Cancer Sci 2020; 111:477-488. [PMID: 31840304 PMCID: PMC7004526 DOI: 10.1111/cas.14283] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 12/04/2019] [Accepted: 12/08/2019] [Indexed: 12/22/2022] Open
Abstract
ATP6L, the C subunit of the V-ATPase V0 domain, is involved in regulating the acidic tumor micro-environment and may promote tumor progression. However, the expression and functional role of ATP6L in tumors have not yet been well explored. In this study, we found that ATP6L protein overexpression was related to colorectal cancer histological differentiation (P < 0.001), presence of metastasis (P < 0.001) and recurrence (P = 0.02). ATP6L expression in the liver metastatic foci was higher than in the primary foci (P = 0.04). ATP6L expression was notably concomitant with epithelial-mesenchymal transition (EMT) immunohistochemical features, such as reduced expression of the epithelial marker E-cadherin (P = 0.021) and increased expression of the mesenchymal marker vimentin (P = 0.004). Results of in vitro and in vivo experiments showed that ATP6L expression could alter cell morphology, regulate EMT-associated protein expression, and enhance migration and invasion. The effect of ATP6L on metastasis was further demonstrated in a tail vein injection mice model. In addition, the mouse xenograft model showed that ATP6L-overexpressing HCT116 cells grew into larger tumor masses, showed less necrosis and formed more micro-vessels than the control cells. Taken together, our results suggest that ATP6L promotes metastasis of colorectal cancer by inducing EMT and angiogenesis, and is a potential target for tumor therapy.
Collapse
Affiliation(s)
- Jingyi Wang
- Department of Pathology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy,Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Dandan Chen
- Department of Pathology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy,Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Wangzhao Song
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Zhiyong Liu
- Department of Pathology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy,Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Wenjuan Ma
- Department of Breast Imaging, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Xiaofeng Li
- Department of Molecular Imaging and Nuclear Medicine, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Chao Zhang
- Department of Bone and Soft Tissue Tumors, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Xin Wang
- Department of Epidemiology and Biostatistics, First Affiliated Hospital, Army Medical University, Chongqing, China
| | - Yalei Wang
- Department of Pathology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy,Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Ye Yang
- Department of Pathology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy,Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Wenfeng Cao
- Department of Pathology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy,Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Lisha Qi
- Department of Pathology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy,Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| |
Collapse
|
13
|
Inactivation of Rab27B-dependent signaling pathway by calycosin inhibits migration and invasion of ER-negative breast cancer cells. Gene 2019; 709:48-55. [DOI: 10.1016/j.gene.2019.04.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2019] [Revised: 03/25/2019] [Accepted: 04/02/2019] [Indexed: 01/08/2023]
|
14
|
Cashikar AG, Hanson PI. A cell-based assay for CD63-containing extracellular vesicles. PLoS One 2019; 14:e0220007. [PMID: 31339911 PMCID: PMC6655660 DOI: 10.1371/journal.pone.0220007] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2019] [Accepted: 07/05/2019] [Indexed: 02/06/2023] Open
Abstract
Extracellular vesicles (EVs) are thought to be important in cell-cell communication and have elicited extraordinary interest as potential biomarkers of disease. However, quantitative methods to enable elucidation of mechanisms underlying release are few. Here, we describe a cell-based assay for monitoring EV release using the EV-enriched tetraspanin CD63 fused to the small, ATP-independent reporter enzyme, Nanoluciferase. Release of CD63-containing EVs from stably expressing cell lines was monitored by comparing luciferase activity in culture media to that remaining in cells. HEK293, U2OS, U87 and SKMel28 cells released 0.3%-0.6% of total cellular CD63 in the form of EVs over 5 hrs, varying by cell line. To identify cellular machinery important for secretion of CD63-containing EVs, we performed a screen of biologically active chemicals in HEK293 cells. While a majority of compounds did not significantly affect EV release, treating cells with the plecomacrolides bafilomycin or concanamycin, known to inhibit the V-ATPase, dramatically increased EV release. Interestingly, alkalization of the endosomal lumen using weak bases had no effect, suggesting a pH-independent enhancement of EV release by V-ATPase inhibitors. The ability to quantify EVs in small samples will enable future detailed studies of release kinetics as well as further chemical and genetic screening to define pathways involved in EV secretion.
Collapse
Affiliation(s)
- Anil G. Cashikar
- Department of Cell Biology & Physiology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Phyllis I. Hanson
- Department of Cell Biology & Physiology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| |
Collapse
|
15
|
McGuire CM, Collins MP, Sun-Wada G, Wada Y, Forgac M. Isoform-specific gene disruptions reveal a role for the V-ATPase subunit a4 isoform in the invasiveness of 4T1-12B breast cancer cells. J Biol Chem 2019; 294:11248-11258. [PMID: 31167791 PMCID: PMC6643023 DOI: 10.1074/jbc.ra119.007713] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 05/30/2019] [Indexed: 12/17/2022] Open
Abstract
The vacuolar H+-ATPase (V-ATPase) is an ATP-driven proton pump present in various intracellular membranes and at the plasma membrane of specialized cell types. Previous work has reported that plasma membrane V-ATPases are key players in breast cancer cell invasiveness. The two subunit a-isoforms known to target the V-ATPase to the plasma membrane are a3 and a4, and expression of a3 has been shown to correlate with plasma membrane localization of the V-ATPase in various invasive human breast cancer cell lines. Here we analyzed the role of subunit a-isoforms in the invasive mouse breast cancer cell line, 4T1-12B. Quantitation of mRNA levels for each isoform by quantitative RT-PCR revealed that a4 is the dominant isoform expressed in these cells. Using a CRISPR/Cas9-based approach to disrupt the genes encoding each of the four V-ATPase subunit a-isoforms, we found that ablation of only the a4-encoding gene significantly inhibits invasion and migration of 4T1-12B cells. Additionally, cells with disrupted a4 exhibited reduced V-ATPase expression at the leading edge, suggesting that the a4 isoform is primarily responsible for targeting the V-ATPase to the plasma membrane in 4T1-12B cells. These findings suggest that different subunit a-isoforms may direct V-ATPases to the plasma membrane of different invasive breast cancer cell lines. They further suggest that expression of V-ATPases at the cell surface is the primary factor that promotes an invasive cancer cell phenotype.
Collapse
Affiliation(s)
- Christina M McGuire
- Department of Developmental, Molecular, and Chemical Biology, Tufts University School of Medicine, Tufts University, Boston, Massachusetts 02111
- Program in Biochemistry, Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, Massachusetts 02111
| | - Michael P Collins
- Department of Developmental, Molecular, and Chemical Biology, Tufts University School of Medicine, Tufts University, Boston, Massachusetts 02111
- Program in Cell, Molecular, and Developmental Biology, Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, Massachusetts 02111
| | - GeHong Sun-Wada
- Department of Biochemistry, Faculty of Pharmaceutical Sciences, Doshisha Women's College, Kyotanabe, Kyoto 610-0395, Japan
| | - Yoh Wada
- Division of Biological Science, Institute of Scientific and Industrial Research, Osaka University, Ibaraki, Osaka 567-0047, Japan
| | - Michael Forgac
- Department of Developmental, Molecular, and Chemical Biology, Tufts University School of Medicine, Tufts University, Boston, Massachusetts 02111
- Program in Biochemistry, Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, Massachusetts 02111
- Program in Cell, Molecular, and Developmental Biology, Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, Massachusetts 02111
| |
Collapse
|
16
|
Yang J, Zhang Z, Zhang Y, Ni X, Zhang G, Cui X, Liu M, Xu C, Zhang Q, Zhu H, Yan J, Zhu VF, Luo Y, Hagan JP, Li Z, Fang J, Jatoi A, Fernandez-Zapico ME, Zheng L, Edil BH, Bronze MS, Houchen CW, Li YP, Li M. ZIP4 Promotes Muscle Wasting and Cachexia in Mice With Orthotopic Pancreatic Tumors by Stimulating RAB27B-Regulated Release of Extracellular Vesicles From Cancer Cells. Gastroenterology 2019; 156:722-734.e6. [PMID: 30342032 PMCID: PMC6878980 DOI: 10.1053/j.gastro.2018.10.026] [Citation(s) in RCA: 96] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2018] [Revised: 09/13/2018] [Accepted: 10/09/2018] [Indexed: 12/22/2022]
Abstract
BACKGROUND & AIMS Cachexia, which includes muscle wasting, is a frequent complication of pancreatic cancer. There are no therapies that reduce cachexia and increase patient survival, so it is important to learn more about its mechanisms. The zinc transporter ZIP4 promotes growth and metastasis of pancreatic tumors. We investigated its effects on muscle catabolism via extracellular vesicle (EV)-mediated stimulation of mitogen-activated protein kinase 14 (p38 MAPK). METHODS We studied nude mice with orthotopic tumors grown from human pancreatic cancer cell lines (AsPC-1 and BxPC-3); tumors were removed 8 days after cell injection and analyzed by histology. Mouse survival was analyzed by Kaplan-Meier curves. ZIP4 was knocked down in AsPC-1 and BxPC-3 cells with small hairpin RNAs; cells with empty vectors were used as controls. Muscle tissues were collected from mice and analyzed by histology and immunohistochemistry. Conditioned media from cell lines and 3-dimensional spheroid/organoid cultures of cancer cells were applied to C2C12 myotubes. The myotubes and the media were analyzed by immunoblots, enzyme-linked immunosorbent assays, and immunofluorescence microscopy. EVs were isolated from conditioned media and analyzed by immunoblots. RESULTS Mice with orthotopic tumors grown from pancreatic cancer cells with knockdown of ZIP4 survived longer and lost less body weight and muscle mass than mice with control tumors. Conditioned media from cancer cells activated p38 MAPK, induced expression of F-box protein 32 and UBR2 in C2C12 myotubes, and also led to loss of myofibrillar protein myosin heavy chain and myotube thinning. Knockdown of ZIP4 in cancer cells reduced these effects. ZIP4 knockdown also reduced pancreatic cancer cell release of heat shock protein (HSP) 70 and HSP90, which are associated with EVs, by decreasing CREB-regulated expression of RAB27B. CONCLUSIONS ZIP4 promotes growth of orthotopic pancreatic tumors in mice and loss of muscle mass by activating CREB-regulated expression of RAB27B, required for release of EVs from pancreatic cancer cells. These EVs activate p38 MAPK and induce expression of F-box protein 32 and UBR2 in myotubes, leading to loss of myofibrillar myosin heavy chain and myotube thinning. Strategies to disrupt these pathways might be developed to reduce pancreatic cancer progression and accompanying cachexia.
Collapse
Affiliation(s)
- Jingxuan Yang
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA,Department of Surgery, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA,The Vivian L. Smith Department of Neurosurgery, the University of Texas Health Science Center at Houston, Houston, Texas 77030, USA
| | - Zicheng Zhang
- Department of Integrative Biology & Pharmacology, the University of Texas Health Science Center at Houston, Houston, Texas 77030, USA
| | - Yuqing Zhang
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA,Department of Surgery, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Xiaoling Ni
- The Vivian L. Smith Department of Neurosurgery, the University of Texas Health Science Center at Houston, Houston, Texas 77030, USA,Department of General Surgery, Zhongshan Hospital, Shanghai Medical College, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
| | - Guohua Zhang
- Department of Integrative Biology & Pharmacology, the University of Texas Health Science Center at Houston, Houston, Texas 77030, USA
| | - Xiaobo Cui
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA,Department of Surgery, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA,The Vivian L. Smith Department of Neurosurgery, the University of Texas Health Science Center at Houston, Houston, Texas 77030, USA
| | - Mingyang Liu
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA,Department of Surgery, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Can Xu
- The Vivian L. Smith Department of Neurosurgery, the University of Texas Health Science Center at Houston, Houston, Texas 77030, USA,Department of Gastroenterology, Changhai Hospital, Shanghai, China
| | - Qiang Zhang
- The Vivian L. Smith Department of Neurosurgery, the University of Texas Health Science Center at Houston, Houston, Texas 77030, USA
| | - Huiyun Zhu
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA,Department of Surgery, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA,Department of Gastroenterology, Changhai Hospital, Shanghai, China
| | - Jie Yan
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA,Department of Surgery, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Vivian F. Zhu
- The Vivian L. Smith Department of Neurosurgery, the University of Texas Health Science Center at Houston, Houston, Texas 77030, USA
| | - Yusheng Luo
- The Vivian L. Smith Department of Neurosurgery, the University of Texas Health Science Center at Houston, Houston, Texas 77030, USA
| | - John P. Hagan
- The Vivian L. Smith Department of Neurosurgery, the University of Texas Health Science Center at Houston, Houston, Texas 77030, USA
| | - Zhaoshen Li
- Department of Gastroenterology, Changhai Hospital, Shanghai, China
| | - Jing Fang
- The Key Lab of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China,Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao, 266061, China
| | - Aminah Jatoi
- Department of Oncology, Mayo Clinic, Rochester, MN 55905, USA
| | | | - Lei Zheng
- The Sidney Kimmel Comprehensive Cancer Center and Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Barish H. Edil
- Department of Surgery, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Michael S. Bronze
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Courtney W. Houchen
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Yi-Ping Li
- Department of Integrative Biology and Pharmacology, the University of Texas Health Science Center at Houston, Houston, Texas.
| | - Min Li
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma; Department of Surgery, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma; The Vivian L. Smith Department of Neurosurgery, the University of Texas Health Science Center at Houston, Houston, Texas; Department of Integrative Biology and Pharmacology, the University of Texas Health Science Center at Houston, Houston, Texas.
| |
Collapse
|
17
|
Li Z, Fang R, Fang J, He S, Liu T. Functional implications of Rab27 GTPases in Cancer. Cell Commun Signal 2018; 16:44. [PMID: 30081925 PMCID: PMC6080553 DOI: 10.1186/s12964-018-0255-9] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Accepted: 07/27/2018] [Indexed: 12/14/2022] Open
Abstract
Background The Rab27 family of small GTPases promotes the progression of breast cancer, melanoma, and other human cancers. In this review, we discuss the role of Rab27 GTPases in cancer progression and the potential applications of these targets in cancer treatment. Main body Elevated expression of Rab27 GTPases is associated with poor prognosis and cancer metastasis. Moreover, these GTPases govern a variety of oncogenic functions, including cell proliferation, cell motility, and chemosensitivity. In addition, small GTPases promote tumor growth and metastasis by enhancing exosome secretion, which alters intracellular microRNA levels, signaling molecule expression, and the tumor microenvironment. Conclusion Rab27 GTPases may have applications as prognostic markers and therapeutic targets in cancer treatment.
Collapse
Affiliation(s)
- Zhihong Li
- Department of Orthopaedics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Rui Fang
- Department of Orthopaedics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jia Fang
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Shasha He
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China.
| | - Tang Liu
- Department of Orthopaedics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
18
|
Whitton B, Okamoto H, Packham G, Crabb SJ. Vacuolar ATPase as a potential therapeutic target and mediator of treatment resistance in cancer. Cancer Med 2018; 7:3800-3811. [PMID: 29926527 PMCID: PMC6089187 DOI: 10.1002/cam4.1594] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 05/07/2018] [Accepted: 05/08/2018] [Indexed: 01/10/2023] Open
Abstract
Vacuolar ATPase (V-ATPase) is an ATP-dependent H+ -transporter that pumps protons across intracellular and plasma membranes. It consists of a large multi-subunit protein complex and influences a wide range of cellular processes. This review focuses on emerging evidence for the roles for V-ATPase in cancer. This includes how V-ATPase dysregulation contributes to cancer growth, metastasis, invasion and proliferation, and the potential link between V-ATPase and the development of drug resistance.
Collapse
Affiliation(s)
- Bradleigh Whitton
- Southampton Cancer Research UK CentreUniversity of SouthamptonSouthamptonUK
- Biological SciencesFaculty of Natural and Environmental SciencesUniversity of SouthamptonSouthamptonUK
| | - Haruko Okamoto
- Biological SciencesFaculty of Natural and Environmental SciencesUniversity of SouthamptonSouthamptonUK
| | - Graham Packham
- Southampton Cancer Research UK CentreUniversity of SouthamptonSouthamptonUK
| | - Simon J. Crabb
- Southampton Cancer Research UK CentreUniversity of SouthamptonSouthamptonUK
| |
Collapse
|
19
|
Son SW, Kim SH, Moon EY, Kim DH, Pyo S, Um SH. Prognostic significance and function of the vacuolar H+-ATPase subunit V1E1 in esophageal squamous cell carcinoma. Oncotarget 2018; 7:49334-49348. [PMID: 27384996 PMCID: PMC5226512 DOI: 10.18632/oncotarget.10340] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Accepted: 06/13/2016] [Indexed: 01/05/2023] Open
Abstract
Vacuolar H+-ATPase (V-ATPase), a hetero-multimeric ATP-driven proton pump has recently emerged as a critical regulator of mTOR-induced amino acid sensing for cell growth. Although dysregulated activity of cell growth regulators is often associated with cancer, the prognostic significance and metabolic roles of V-ATPase in esophageal cancer progression remain unclear. Here, we show that high levels of V-ATPase subunit V1E1 (V-ATPase V1E1) were significantly associated with shortened disease-free survival in patients with esophageal squamous cell carcinoma (ESCC). Multivariate analysis identified the V-ATPase V1E1 as an independent adverse prognostic factor (hazard ratio;1.748, P = 0.018). In addition, depletion of V-ATPase V1E1 resulted in reduced cell motility, decreased glucose uptake, diminished levels of lactate, and decreased ATP production, as well as inhibition of glycolytic enzyme expression in TE8 esophageal cancer cells. Consistent with these results, the Cancer Genome Atlas (TCGA) data and Gene Set Enrichment Analysis (GSEA) showed a high frequency of copy number alterations of the V-ATPase V1E1 gene, and identified a correlation between levels of V-ATPase V1E1 mRNA and Pyruvate Kinase M2 (PKM2) in ESCC. High expression levels of both V-ATPase V1E1 and phosphorylated PKM2 (p-PKM2), a key player in cancer metabolism, were associated with poorer prognosis in ESCC. Collectively, our findings suggest that expression of the V-ATPase V1E1 has prognostic significance in ESCC, and is closely linked to migration, invasion, and aerobic glycolysis in esophageal cancer cells.
Collapse
Affiliation(s)
- Sung Wook Son
- Department of Molecular Cell Biology, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Gyeonggi-do, 16419, Korea
| | - Seok-Hyung Kim
- Department of Pathology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, 06351, Korea.,Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Samsung Medical Center, Sungkyunkwan University, Seoul, 06351, Korea
| | - Eun-Yi Moon
- Department of Bioscience and Biotechnology, Sejong University, Seoul, 05006, Korea
| | - Dong-Hoon Kim
- Department of Pharmacology, Korea University College of Medicine, Seoul, 02841, Korea
| | - Suhkneung Pyo
- Division of Immunopharmacology, School of Pharmacy, Sungkyunkwan University, Gyeonggi-do, 16419, Korea
| | - Sung Hee Um
- Department of Molecular Cell Biology, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Gyeonggi-do, 16419, Korea.,Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Samsung Medical Center, Sungkyunkwan University, Seoul, 06351, Korea
| |
Collapse
|
20
|
The a3 isoform of subunit a of the vacuolar ATPase localizes to the plasma membrane of invasive breast tumor cells and is overexpressed in human breast cancer. Oncotarget 2018; 7:46142-46157. [PMID: 27323815 PMCID: PMC5216787 DOI: 10.18632/oncotarget.10063] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Accepted: 06/02/2016] [Indexed: 12/11/2022] Open
Abstract
The vacuolar (H+)-ATPases (V-ATPases) are a family of ATP-driven proton pumps that acidify intracellular compartments and transport protons across the plasma membrane. Previous work has demonstrated that plasma membrane V-ATPases are important for breast cancer invasion in vitro and that the V-ATPase subunit a isoform a3 is upregulated in and critical for MDA-MB231 and MCF10CA1a breast cancer cell invasion. It has been proposed that subunit a3 is present on the plasma membrane of invasive breast cancer cells and is overexpressed in human breast cancer. To test this, we used an a3-specific antibody to assess localization in breast cancer cells. Subunit a3 localizes to the leading edge of migrating breast cancer cells, but not the plasma membrane of normal breast epithelial cells. Furthermore, invasive breast cancer cells express a3 throughout all intracellular compartments tested, including endosomes, the Golgi, and lysosomes. Moreover, subunit a3 knockdown in MB231 breast cancer cells reduces in vitro migration. This reduction is not enhanced upon addition of a V-ATPase inhibitor, suggesting that a3-containing V-ATPases are critical for breast cancer migration. Finally, we have tested a3 expression in human breast cancer tissue and mRNA prepared from normal and cancerous breast tissue. a3 mRNA was upregulated 2.5-47 fold in all breast tumor cDNA samples tested relative to normal tissue, with expression generally correlated to cancer stage. Furthermore, a3 protein expression was increased in invasive breast cancer tissue relative to noninvasive cancer and normal breast tissue. These studies suggest that subunit a3 plays an important role in invasive human breast cancer.
Collapse
|
21
|
Tumor cell cholesterol depletion and V-ATPase inhibition as an inhibitory mechanism to prevent cell migration and invasiveness in melanoma. Biochim Biophys Acta Gen Subj 2017; 1862:684-691. [PMID: 29253593 DOI: 10.1016/j.bbagen.2017.12.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Revised: 11/18/2017] [Accepted: 12/13/2017] [Indexed: 01/01/2023]
Abstract
BACKGROUND V-ATPase interactions with cholesterol enriched membrane microdomains have been related to metastasis in a variety of cancers, but the underlying mechanism remains at its beginnings. It has recently been reported that the inhibition of this H+ pump affects cholesterol mobilization to the plasma membrane. METHODS Inhibition of melanoma cell migration and invasiveness was assessed by wound healing and Transwell assays in murine cell lines (B16F10 and Melan-A). V-ATPase activity was measured in vitro by ATP hydrolysis and H+ transport in membrane vesicles, and intact cell H+ fluxes were measured by using a non-invasive Scanning Ion-selective Electrode Technique (SIET). RESULTS Cholesterol depletion by 5mM MβCD was found to be inhibitory to the hydrolytic and H+ pumping activities of the V-ATPase of melanoma cell lines, as well as to the migration and invasiveness capacities of these cells. Nearly the same effects were obtained using concanamycin A, a specific inhibitor of V-ATPase, which also promoted a decrease of the H+ efflux in live cells at the same extent of MβCD. CONCLUSIONS We found that cholesterol depletion significantly affects the V-ATPase activity and the initial metastatic processes following a profile similar to those observed in the presence of the V-ATPase specific inhibitor, concanamycin. GENERAL SIGNIFICANCE The results shed new light on the functional role of the interactions between V-ATPases and cholesterol-enriched microdomains of cell membranes that contribute with malignant phenotypes in melanoma.
Collapse
|
22
|
Aasebø E, Bartaula-Brevik S, Hernandez-Valladares M, Bruserud Ø. Vacuolar ATPase as a possible therapeutic target in human acute myeloid leukemia. Expert Rev Hematol 2017; 11:13-24. [PMID: 29168399 DOI: 10.1080/17474086.2018.1407239] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
INTRODUCTION V-ATPase is a proton pump expressed both in the membrane of intracellular organelles (e.g. endosomes, lysosomes, Golgi structures) and the plasma membrane. It is an important regulator of organellar functions, intracellular molecular trafficking, intercellular communication and intracellular signaling. It is therefore considered as a possible therapeutic target in the treatment of human malignancies. Areas covered: Relevant publications were identified through literature searches in the PubMed database. We searched for original articles and reviews describing the possible importance of V-ATPase for leukemogenesis and chemosensitivity in human myeloid cells, especially acute myeloid leukemia (AML) cells. Expert commentary: The expression of V-ATPase in the primary human AML cells varies between patients, and high levels are associated with high constitutive release of a wide range of soluble mediators. Several of the molecules included in the V-ATPase interactome may also be important in leukemogenesis and/or development of chemoresistance in human AML. Therapeutic targeting of V-ATPase should therefore be regarded as a possible therapeutic strategy in human AML, but the efficiency of such targeting will probably differ between patients. The possibility of toxicity, especially hematological toxicity and immunosuppression, also has to be clarified.
Collapse
Affiliation(s)
- Elise Aasebø
- a Section for Hematology, Department of Clinical Science , University of Bergen , Bergen , Norway.,b Proteomics Unit (PROBE), Department of Biomedicine , University of Bergen , Bergen , Norway
| | - Sushma Bartaula-Brevik
- a Section for Hematology, Department of Clinical Science , University of Bergen , Bergen , Norway
| | - Maria Hernandez-Valladares
- a Section for Hematology, Department of Clinical Science , University of Bergen , Bergen , Norway.,b Proteomics Unit (PROBE), Department of Biomedicine , University of Bergen , Bergen , Norway
| | - Øystein Bruserud
- a Section for Hematology, Department of Clinical Science , University of Bergen , Bergen , Norway.,c Department of Medicine , Haukeland University Hospital , Bergen , Norway
| |
Collapse
|
23
|
Abstract
It has been recognized that cancer-associated mortality is more often a result of the disrupted physiological functions in multiple organs following metastatic dissemination of cancer cells, rather than the presence and growth of the primary tumor. Despite advances in our understanding of the events leading to cancer initiation, growth, and acquisition of invasive properties, we are still unable to effectively treat metastatic disease. It is now being accepted that the secretion of extracellular vesicles, such as exosomes from cancer cells, has a profound impact on the initiation and propagation of metastatic breast cancer. These cancer-secreted vesicles differ from other means of cellular communication due to their capability of bulk delivery and organotropism. Here, we provide an overview of the role of extracellular vesicles in breast cancer metastasis and discuss key areas that may facilitate our understanding of metastatic breast cancer to guide our efforts towards providing better therapies.
Collapse
Affiliation(s)
- Andrew R Chin
- Department of Pathology, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA
- City of Hope Irell & Manella Graduate School of Biological Sciences, Duarte, CA, 91010, USA
| | - Shizhen Emily Wang
- Department of Pathology, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA.
| |
Collapse
|
24
|
Asgharzadeh MR, Barar J, Pourseif MM, Eskandani M, Jafari Niya M, Mashayekhi MR, Omidi Y. Molecular machineries of pH dysregulation in tumor microenvironment: potential targets for cancer therapy. BIOIMPACTS : BI 2017; 7:115-133. [PMID: 28752076 PMCID: PMC5524986 DOI: 10.15171/bi.2017.15] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Revised: 05/28/2017] [Accepted: 06/06/2017] [Indexed: 12/30/2022]
Abstract
Introduction: Cancer is an intricate disorder/dysfunction of cells that can be defined as a genetic heterogeneity in human disease. Therefore, it is characterized by several adaptive complex hallmarks. Among them, the pH dysregulation appears as a symbol of aberrant functions within the tumor microenvironment (TME). In comparison with normal tissues, in the solid tumors, we face with an irregular acidification and alkalinization of the extracellular and intracellular fluids. Methods: In this study, we comprehensively discussed the most recent reports on the hallmarks of solid tumors to provide deep insights upon the molecular machineries involved in the pH dysregulation of solid tumors and their impacts on the initiation and progression of cancer. Results: The dysregulation of pH in solid tumors is fundamentally related to the Warburg effect and hypoxia, leading to expression of a number of molecular machineries, including: NHE1, H+ pump V-ATPase, CA-9, CA-12, MCT-1, GLUT-1. Activation of proton exchangers and transporters (PETs) gives rise to formation of TME. This condition favors the cancer cells to evade from the anoikis and apoptosis, granting them aggressive and metastasis phenotype, as well as resistance to chemotherapy and radiation therapy. This review aimed to discuss the key molecular changes of tumor cells in terms of bio-energetics and cancer metabolism in relation with pH dysregulation. During this phenomenon, the intra- and extracellular metabolites are altered and/or disrupted. Such molecular alterations provide molecular hallmarks for direct targeting of the PETs by potent relevant inhibitors in combination with conventional cancer therapies as ultimate therapy against solid tumors. Conclusion: Taken all, along with other treatment strategies, targeting the key molecular machineries related to intra- and extracellular metabolisms within the TME is proposed as a novel strategy to inhibit or block PETs that are involved in the pH dysregulation of solid tumors.
Collapse
Affiliation(s)
- Mohammad Reza Asgharzadeh
- Department of Biology, Fars Science and Research Branch, Islamic Azad University, Marvdasht, Iran
- Department of Biology, Marvdasht Branch, Islamic Azad University, Marvdasht, Iran
| | - Jaleh Barar
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad M. Pourseif
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Morteza Eskandani
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mojtaba Jafari Niya
- Department of Biology, Fars Science and Research Branch, Islamic Azad University, Marvdasht, Iran
- Department of Biology, Marvdasht Branch, Islamic Azad University, Marvdasht, Iran
| | | | - Yadollah Omidi
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
25
|
Abstract
The vacuolar ATPases (V-ATPases) are a family of proton pumps that couple ATP hydrolysis to proton transport into intracellular compartments and across the plasma membrane. They function in a wide array of normal cellular processes, including membrane traffic, protein processing and degradation, and the coupled transport of small molecules, as well as such physiological processes as urinary acidification and bone resorption. The V-ATPases have also been implicated in a number of disease processes, including viral infection, renal disease, and bone resorption defects. This review is focused on the growing evidence for the important role of V-ATPases in cancer. This includes functions in cellular signaling (particularly Wnt, Notch, and mTOR signaling), cancer cell survival in the highly acidic environment of tumors, aiding the development of drug resistance, as well as crucial roles in tumor cell invasion, migration, and metastasis. Of greatest excitement is evidence that at least some tumors express isoforms of V-ATPase subunits whose disruption is not lethal, leading to the possibility of developing anti-cancer therapeutics that selectively target V-ATPases that function in cancer cells.
Collapse
Affiliation(s)
- Laura Stransky
- Department of Developmental, Molecular and Chemical Biology, Tufts University School of Medicine, and Program in Cellular and Molecular Physiology, Program in Biochemistry, and Program in Cell, Molecular and Developmental Biology, Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, Massachusetts
| | - Kristina Cotter
- Department of Developmental, Molecular and Chemical Biology, Tufts University School of Medicine, and Program in Cellular and Molecular Physiology, Program in Biochemistry, and Program in Cell, Molecular and Developmental Biology, Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, Massachusetts
| | - Michael Forgac
- Department of Developmental, Molecular and Chemical Biology, Tufts University School of Medicine, and Program in Cellular and Molecular Physiology, Program in Biochemistry, and Program in Cell, Molecular and Developmental Biology, Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, Massachusetts
| |
Collapse
|
26
|
Huang D, Bian G, Pan Y, Han X, Sun Y, Wang Y, Shen G, Cheng M, Fang X, Hu S. MiR-20a-5p promotes radio-resistance by targeting Rab27B in nasopharyngeal cancer cells. Cancer Cell Int 2017; 17:32. [PMID: 28265202 PMCID: PMC5333421 DOI: 10.1186/s12935-017-0389-7] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Accepted: 01/28/2017] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND MicroRNAs (miRNAs) was reported to be involved in cancer radio-resistance, which remains a major obstacle for effective cancer therapy. METHODS The differently expressed miRNAs were detected by RNA-seq experiment in nasopharyngeal cancer (NPC) cells. MiR-20a-5p was selected as our target, which was subject to finding its target gene Rab27B via bioinformatics analysis. The qRT-PCR, western blot and the luciferase reporter assays were performed to confirm Rab27B as the target of miR-20a-5p. In addition, the roles of miR-20a-5p in NPC radio-resistance were detected by transfection of either miR-20a-5p-mimic or miR-20a-5p-antagomiR. The involvement of Rab27B with NPC radio-resistance was also detected by the experiments with siRNA-mediated repression of Rab27B or over-expression of GFP-Rab27B. Wound healing and invasion assays were performed to detect the roles of both miR-20a-5p and Rab27B. RESULTS MiR-20a-5p promotes NPC radio-resistance. We identified that its target gene Rab27B negatively correlates with miR-20a-5p-mediated NPC radio-resistance by systematic studies of a radio-sensitive (CNE-2) and resistant (CNE-1) NPC cell lines. Repression of Rab27B by siRNA suppresses cell apoptosis and passivates CNE-2 cells, whereas over-expression of Rab27B triggered cell apoptosis and sensitizes CNE-1 cells. CONCLUSIONS MiR-20a-5p and its target gene Rab27B might be involved in the NPC radio-resistance. Thus the key players and regulators involved in this pathway might be the potential targets for developing effective therapeutic strategies against NPC.
Collapse
Affiliation(s)
- Dabing Huang
- Shandong University School of Medicine, Jinan, 250012 China.,Department of Geriatrics, Anhui Provincial Hospital, Anhui Medical University, Hefei, 230031 Anhui China.,Department of Oncology, Anhui Provincial Hospital, Anhui Medical University, Hefei, 230001 China
| | - Geng Bian
- Department of Geriatrics, Anhui Provincial Hospital, Anhui Medical University, Hefei, 230031 Anhui China
| | - Yueyin Pan
- Department of Oncology, Anhui Provincial Hospital, Anhui Medical University, Hefei, 230001 China
| | - Xinghua Han
- Department of Oncology, Anhui Provincial Hospital, Anhui Medical University, Hefei, 230001 China
| | - Yubei Sun
- Department of Oncology, Anhui Provincial Hospital, Anhui Medical University, Hefei, 230001 China
| | - Yong Wang
- Department of Oncology, Anhui Provincial Hospital, Anhui Medical University, Hefei, 230001 China
| | - Guodong Shen
- Anhui Provincial Key Laboratory of Tumor Immunotherapy and Nutrition Therapy, Hefei, 230001 China
| | - Min Cheng
- Anhui Provincial Key Laboratory of Tumor Immunotherapy and Nutrition Therapy, Hefei, 230001 China
| | - Xiang Fang
- Department of Geriatrics, Anhui Provincial Hospital, Anhui Medical University, Hefei, 230031 Anhui China
| | - Shilian Hu
- Shandong University School of Medicine, Jinan, 250012 China.,Department of Geriatrics, Anhui Provincial Hospital, Anhui Medical University, Hefei, 230031 Anhui China.,Anhui Provincial Key Laboratory of Tumor Immunotherapy and Nutrition Therapy, Hefei, 230001 China
| |
Collapse
|
27
|
The asialoglycoprotein receptor suppresses the metastasis of hepatocellular carcinoma via LASS2-mediated inhibition of V-ATPase activity. Cancer Lett 2016; 379:107-16. [PMID: 27241665 DOI: 10.1016/j.canlet.2016.05.030] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 05/25/2016] [Accepted: 05/25/2016] [Indexed: 12/28/2022]
Abstract
The asialoglycoprotein receptor (ASGR), which is expressed mainly in hepatocytes, is downregulated in poorly differentiated hepatocellular carcinoma (HCC). Here we investigated the role of ASGR1 in HCC metastasis as well as the possible underlying molecular mechanisms. We found that ASGR1 was downregulated in HCC tissue compared with adjacent non-tumorous liver tissue and that lower ASGR1 expression was associated with higher TNM stage and poorer prognosis in HCC patients. ASGR1 overexpression inhibited hepatoma cell migration and invasion in vitro and in vivo, while ASGR1 knockdown had the opposite effects. Furthermore, ASGR1 interacted directly with human longevity assurance homolog 2 of yeast LAG1 (LASS2). Knockdown of LASS2 attenuated the inhibitory effects of ASGR1 on hepatoma cell migration and invasion in vitro. ASGR1 decreased V-ATPase activity in hepatoma cells, and this was reversed by LASS2 knockdown. Finally, HCC patients with low LASS2 levels had poor prognosis, while those with high ASGR1 and LASS2 levels had better prognosis. Thus, ASGR1 may act as a potential metastasis suppressor in HCC, and the combination of ASGR1 and LASS2 may help predict the prognosis of HCC patients.
Collapse
|
28
|
Di Cristofori A, Ferrero S, Bertolini I, Gaudioso G, Russo MV, Berno V, Vanini M, Locatelli M, Zavanone M, Rampini P, Vaccari T, Caroli M, Vaira V. The vacuolar H+ ATPase is a novel therapeutic target for glioblastoma. Oncotarget 2016; 6:17514-31. [PMID: 26020805 PMCID: PMC4627325 DOI: 10.18632/oncotarget.4239] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Accepted: 05/02/2015] [Indexed: 01/01/2023] Open
Abstract
The vacuolar H+ ATPase (V-ATPase) is a proton pump responsible for acidification of cellular microenvironments, an activity exploited by tumors to survive, proliferate and resist to therapy. Despite few observations, the role of V-ATPase in human tumorigenesis remains unclear. We investigated the expression of ATP6V0C, ATP6V0A2, encoding two subunits belonging to the V-ATPase V0 sector and ATP6V1C, ATP6V1G1, ATPT6V1G2, ATP6V1G3, which are part of the V1 sector, in series of adult gliomas and in cancer stem cell-enriched neurospheres isolated from glioblastoma (GBM) patients. ATP6V1G1 expression resulted significantly upregulated in tissues of patients with GBM and correlated with shorter patients' overall survival independent of clinical variables. ATP6V1G1 knockdown in GBM neurospheres hampered sphere-forming ability, induced cell death, and decreased matrix invasion, a phenotype not observed in GBM monolayer cultures. Treating GBM organotypic cultures or neurospheres with the selective V-ATPase inhibitor bafilomycin A1 reproduced the effects of ATP6V1G1 siRNA and strongly suppressed expression of the stem cell markers Nestin, CD133 and transcription factors SALL2 and POU3F2 in neurospheres. These data point to ATP6V1G1 as a novel marker of poor prognosis in GBM patients and identify V-ATPase inhibition as an innovative therapeutic strategy for GBM.
Collapse
Affiliation(s)
- Andrea Di Cristofori
- Department of Pathophysiology and Organ Transplantation, University of Milan, Milan, Italy.,Division of Neurosurgery, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Stefano Ferrero
- Division of Pathology, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy.,Department of Biomedical, Surgical and Dental Sciences, University of Milan, Milan, Italy
| | - Irene Bertolini
- Department of Pathophysiology and Organ Transplantation, University of Milan, Milan, Italy.,Division of Pathology, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Gabriella Gaudioso
- Department of Pathophysiology and Organ Transplantation, University of Milan, Milan, Italy.,Division of Pathology, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Maria Veronica Russo
- Department of Pathophysiology and Organ Transplantation, University of Milan, Milan, Italy.,Division of Pathology, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Valeria Berno
- Istituto Nazionale Genetica Molecolare "Romeo ed Enrica Invernizzi", Milan, Italy
| | - Marco Vanini
- Surgical Pathology Unit, St. Anna Hospital, Como, Italy
| | - Marco Locatelli
- Division of Neurosurgery, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Mario Zavanone
- Department of Pathophysiology and Organ Transplantation, University of Milan, Milan, Italy.,Division of Neurosurgery, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Paolo Rampini
- Division of Neurosurgery, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Thomas Vaccari
- IFOM - The FIRC Institute of Molecular Oncology, Milan, Italy
| | - Manuela Caroli
- Division of Neurosurgery, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Valentina Vaira
- Division of Pathology, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy.,Istituto Nazionale Genetica Molecolare "Romeo ed Enrica Invernizzi", Milan, Italy
| |
Collapse
|
29
|
Cotter K, Stransky L, McGuire C, Forgac M. Recent Insights into the Structure, Regulation, and Function of the V-ATPases. Trends Biochem Sci 2016; 40:611-622. [PMID: 26410601 DOI: 10.1016/j.tibs.2015.08.005] [Citation(s) in RCA: 215] [Impact Index Per Article: 26.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Revised: 08/06/2015] [Accepted: 08/07/2015] [Indexed: 10/23/2022]
Abstract
The vacuolar (H(+))-ATPases (V-ATPases) are ATP-dependent proton pumps that acidify intracellular compartments and are also present at the plasma membrane. They function in such processes as membrane traffic, protein degradation, virus and toxin entry, bone resorption, pH homeostasis, and tumor cell invasion. V-ATPases are large multisubunit complexes, composed of an ATP-hydrolytic domain (V1) and a proton translocation domain (V0), and operate by a rotary mechanism. This review focuses on recent insights into their structure and mechanism, the mechanisms that regulate V-ATPase activity (particularly regulated assembly and trafficking), and the role of V-ATPases in processes such as cell signaling and cancer. These developments have highlighted the potential of V-ATPases as a therapeutic target in a variety of human diseases.
Collapse
Affiliation(s)
- Kristina Cotter
- Program in Cellular and Molecular Physiology, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA 02111, USA
| | - Laura Stransky
- Program in Cellular and Molecular Physiology, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA 02111, USA
| | - Christina McGuire
- Program in Biochemistry, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA 02111, USA
| | - Michael Forgac
- Program in Cellular and Molecular Physiology, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA 02111, USA; Program in Biochemistry, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA 02111, USA; Department of Developmental, Molecular, and Chemical Biology, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA 02111, USA.
| |
Collapse
|
30
|
Fan SH, Wang YY, Lu J, Zheng YL, Wu DM, Zhang ZF, Shan Q, Hu B, Li MQ, Cheng W. CERS2 suppresses tumor cell invasion and is associated with decreased V-ATPase and MMP-2/MMP-9 activities in breast cancer. J Cell Biochem 2016; 116:502-13. [PMID: 25213553 DOI: 10.1002/jcb.24978] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2013] [Accepted: 09/05/2014] [Indexed: 02/04/2023]
Abstract
Ceramide synthase 2 (CERS2) is the gene identified from a human liver cDNA library in 2001. Our previous studies have shown higher expression of CERS2 in the breast cancer patients was associated with fewer lymph node metastases. However, the molecular mechanism of CERS2 involved is unknown. Here, we found CERS2 was heterogeneously expressed in various breast cancer cells. The mRNA and protein expression levels of CERS2 in MCF7 cells, which are poorly invasive breast cancer cells, were obviously higher than that in the highly invasive cells MDA-MB-231. Results showed overexpression of CERS2 in MDA-MB-231 cells could significantly inhibit the migration and invasion ability, whereas CERS2 knockdown in MCF7 cells could significantly increase the migration and invasion ability. Overexpression of CERS2 in MDA-MB-231 cells significantly reduced the V-ATPase activity, increased the extracellular pH and decreased the pH-dependent activity of MMP-2 and MMP-9 matrix metalloproteinases (MMPs). CERS2 knockdown in MCF7 cells significantly increased the V-ATPase activity, decreased the extracellular pH and increased the activity of MMP-2 and MMP-9. Taken together, CERS2 can significantly inhibit breast cancer cell invasion and is associated with the decrease of the V-ATPase activity and extracellular hydrogen ion concentration, and in turn the activation of secreted MMP-2/MMP-9 and degradation of extracellular matrix (ECM), which ultimately suppressed tumor's invasion. Thus, CERS2 may represent a novel target for selectively disrupting V-ATPase activity and the invasive potential of cancer cells.
Collapse
Affiliation(s)
- Shao-hua Fan
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou, Jiangsu, 221116, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
MiR-193a-3p and miR-193a-5p suppress the metastasis of human osteosarcoma cells by down-regulating Rab27B and SRR, respectively. Clin Exp Metastasis 2016; 33:359-72. [PMID: 26913720 PMCID: PMC4799803 DOI: 10.1007/s10585-016-9783-0] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Accepted: 02/17/2016] [Indexed: 11/01/2022]
Abstract
MicroRNAs have been identified as key players in the development and progression of osteosarcoma, which is the most common primary malignancy of bone. Sequencing-based miR-omic and quantitative real-time PCR analyses suggested that the expression of miR-193a-3p and miR-193a-5p was decreased by DNA methylation at their promoter region in a highly metastatic osteosarcoma cell line (MG63.2) relative to their expression in the less metastatic MG63 cell line. Further wound-healing and invasion assays demonstrated that both miR-193a-3p and miR-193a-5p suppressed osteosarcoma cell migration and invasion. Moreover, introducing miR-193a-3p and miR-193a-5p mimics into MG63.2 cells or antagomiRs into MG63 cells confirmed their critical roles in osteosarcoma metastasis. Additionally, bioinformatics prediction along with biochemical assay results clearly suggested that the secretory small GTPase Rab27B and serine racemase (SRR) were direct targets of miR-193a-3p and miR-193a-5p, respectively. These two targets are indeed involved in the miR-193a-3p- and miR-193a-5p-induced suppression of osteosarcoma cell migration and invasion. MiR-193a-3p and miR-193a-5p play important roles in osteosarcoma metastasis through down-regulation of the Rab27B and SRR genes and therefore may serve as useful biomarkers for the diagnosis of osteosarcoma and as potential candidates for the treatment of metastatic osteosarcoma.
Collapse
|
32
|
Zhao H, Wang Q, Wang X, Zhu H, Zhang S, Wang W, Wang Z, Huang J. Correlation Between RAB27B and p53 Expression and Overall Survival in Pancreatic Cancer. Pancreas 2016; 45:204-10. [PMID: 26418905 PMCID: PMC4714634 DOI: 10.1097/mpa.0000000000000453] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OBJECTIVES RAB27B is a member of the Rab family GTPases involved in vesicle trafficking, and p53 has recently been implicated in regulating the exosome secretion pathway. Because exosome secretion plays an important role in modulating tumor microenvironment and invasive growth, we hypothesized that RAB27B and p53 expression might be associated with the aggressive behavior in pancreatic ductal adenocarcinoma, one of the most deadly human malignancies. METHODS We determined protein expression of RAB27B and p53 in 260 pancreatic tissues (186 malignant and 74 normal or benign) by immunohistochemistry analysis on tissue microarray and their correlation with patients' clinical parameters and overall survival. RESULTS We found that a high RAB27B protein expression (RAB27B) was significantly associated with perineural and vascular invasion, as well as distant metastasis. Patients with a high RAB27B expression had significantly poorer overall survival in both univariate and multivariate analyses. A significant correlation between RAB27B and p53 expression was observed. CONCLUSIONS Our data indicate that RAB27B expression is an independent prognostic marker for pancreatic ductal adenocarcinoma and suggest that RAB27B-regulated exosome secretion pathway represents a novel therapeutic target in pancreatic cancer.
Collapse
Affiliation(s)
- Hui Zhao
- From the *Departments of Interventional Radiology, †General Surgery, ‡Laboratory Medicine, and §Pathology, and ∥Surgical Comprehensive Laboratory, Nantong University Affiliated Hospital, Jiangsu, China
| | - Qingqing Wang
- From the *Departments of Interventional Radiology, †General Surgery, ‡Laboratory Medicine, and §Pathology, and ∥Surgical Comprehensive Laboratory, Nantong University Affiliated Hospital, Jiangsu, China
| | - Xudong Wang
- From the *Departments of Interventional Radiology, †General Surgery, ‡Laboratory Medicine, and §Pathology, and ∥Surgical Comprehensive Laboratory, Nantong University Affiliated Hospital, Jiangsu, China
| | - Huijun Zhu
- From the *Departments of Interventional Radiology, †General Surgery, ‡Laboratory Medicine, and §Pathology, and ∥Surgical Comprehensive Laboratory, Nantong University Affiliated Hospital, Jiangsu, China
| | - Shu Zhang
- From the *Departments of Interventional Radiology, †General Surgery, ‡Laboratory Medicine, and §Pathology, and ∥Surgical Comprehensive Laboratory, Nantong University Affiliated Hospital, Jiangsu, China
| | - Wei Wang
- From the *Departments of Interventional Radiology, †General Surgery, ‡Laboratory Medicine, and §Pathology, and ∥Surgical Comprehensive Laboratory, Nantong University Affiliated Hospital, Jiangsu, China
| | - Zhiwei Wang
- From the *Departments of Interventional Radiology, †General Surgery, ‡Laboratory Medicine, and §Pathology, and ∥Surgical Comprehensive Laboratory, Nantong University Affiliated Hospital, Jiangsu, China
| | - Jianfei Huang
- From the *Departments of Interventional Radiology, †General Surgery, ‡Laboratory Medicine, and §Pathology, and ∥Surgical Comprehensive Laboratory, Nantong University Affiliated Hospital, Jiangsu, China
| |
Collapse
|
33
|
Kulshrestha A, Katara GK, Ginter J, Pamarthy S, Ibrahim SA, Jaiswal MK, Sandulescu C, Periakaruppan R, Dolan J, Gilman-Sachs A, Beaman KD. Selective inhibition of tumor cell associated Vacuolar-ATPase 'a2' isoform overcomes cisplatin resistance in ovarian cancer cells. Mol Oncol 2016; 10:789-805. [PMID: 26899534 DOI: 10.1016/j.molonc.2016.01.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Revised: 01/07/2016] [Accepted: 01/09/2016] [Indexed: 12/31/2022] Open
Abstract
Development of resistance to platinum compounds significantly hinders successful ovarian cancer (OVCA) treatment. In tumor cells, dysregulated pH gradient across cell membranes is a key physiological mechanism of metastasis/chemo-resistance. These pH alterations are mediated by aberrant activation of key multi-subunit proton pumps, Vacuolar-ATPases (V-ATPases). In tumor cells, its 'a2' isoform (V-ATPase-V0a2) is a component of functional plasma-membrane complex and promotes tumor invasion through tumor-acidification and immuno-modulation. Its involvement in chemo-resistance has not been studied. Here, we show that V-ATPase-V0a2 is over-expressed in acquired-cisplatin resistant OVCA cells (cis-A2780/cis-TOV112D). Of all the 'a' subunit isoforms, V-ATPase-V0a2 exhibited an elevated expression on plasma membrane of cisplatin-resistant cells compared to sensitive counterparts. Immuno-histochemistry revealed V-ATPase-V0a2 expression in both low grade (highly drug-resistant) and high grade (highly recurrent) human OVCA tissues indicating its role in a centralized mechanism of tumor resistance. In cisplatin resistant cells, shRNA mediated inhibition of V-ATPase-V0a2 enhanced sensitivity towards both cisplatin and carboplatin. This improved cytotoxicity was mediated by enhanced cisplatin-DNA-adduct formation and suppressed DNA-repair pathway, leading to enhanced apoptosis. Suppression of V0a2 activity strongly reduced cytosolic pH in resistant tumor cells, which is known to enhance platinum-associated DNA-damage. As an indicator of reduced metastasis and chemo-resistance, in contrast to plasma membrane localization, a diffused cytoplasmic localization of acidic vacuoles was observed in V0a2-knockdown resistant cells. Interestingly, pre-treatment with monoclonal V0a2-inhibitory antibody enhanced cisplatin cytotoxicity in resistant cells. Taken together, our findings suggest that the isoform specific inhibition of V-ATPase-V0a2 could serve as a therapeutic strategy for chemo-resistant ovarian carcinoma and improve efficacy of platinum drugs.
Collapse
Affiliation(s)
- Arpita Kulshrestha
- Department of Microbiology and Immunology, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Gajendra K Katara
- Department of Microbiology and Immunology, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Jordyn Ginter
- Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Sahithi Pamarthy
- Department of Microbiology and Immunology, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Safaa A Ibrahim
- Department of Microbiology and Immunology, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Mukesh K Jaiswal
- Department of Microbiology and Immunology, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Corina Sandulescu
- Department of Obstetrics & Gynecology, Advocate Lutheran General Hospital, Park Ridge, IL, USA
| | - Ramayee Periakaruppan
- Department of Obstetrics & Gynecology, Advocate Lutheran General Hospital, Park Ridge, IL, USA
| | - James Dolan
- Department of Obstetrics & Gynecology, Advocate Lutheran General Hospital, Park Ridge, IL, USA
| | - Alice Gilman-Sachs
- Department of Microbiology and Immunology, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Kenneth D Beaman
- Department of Microbiology and Immunology, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA.
| |
Collapse
|
34
|
Kulshrestha A, Katara GK, Ibrahim S, Pamarthy S, Jaiswal MK, Gilman Sachs A, Beaman KD. Vacuolar ATPase 'a2' isoform exhibits distinct cell surface accumulation and modulates matrix metalloproteinase activity in ovarian cancer. Oncotarget 2016; 6:3797-810. [PMID: 25686833 PMCID: PMC4414154 DOI: 10.18632/oncotarget.2902] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Accepted: 12/14/2014] [Indexed: 01/25/2023] Open
Abstract
Tumor associated vacuolar H+-ATPases (V-ATPases) are multi-subunit proton pumps that acidify tumor microenvironment, thereby promoting tumor invasion. Subunit ‘a’ of its V0 domain is the major pH sensing unit that additionally controls sub-cellular targeting of V-ATPase and exists in four different isoforms. Our study reports an elevated expression of the V-ATPase-V0a2 isoform in ovarian cancer(OVCA) tissues and cell lines(A2780, SKOV-3 and TOV-112D). Among all V0’a’ isoforms, V0a2 exhibited abundant expression on OVCA cell surface while normal ovarian epithelia did not. Sub-cellular distribution of V-ATPase-V0a2 confirmed its localization on plasma-membrane, where it was also co-associated with cortactin, an F-actin stabilizing protein at leading edges of cancer cells. Additionally, V0a2 was also localized in early and late endosomal compartments that are sites for modulations of several signaling pathways in cancer. Targeted inhibition of V-ATPase-V0a2 suppressed matrix metalloproteinase activity(MMP-9 & MMP-2) in OVCA cells. In conclusion, V-ATPase-V0a2 isoform is abundantly expressed on ovarian tumor cell surface in association with invasion assembly related proteins and plays critical role in tumor invasion by modulating the activity of matrix-degrading proteases. This study highlights for the first time, the importance of V-ATPase-V0a2 isoform as a distinct biomarker and possible therapeutic target for treatment of ovarian carcinoma.
Collapse
Affiliation(s)
- Arpita Kulshrestha
- Department of Microbiology and Immunology, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Gajendra K Katara
- Department of Microbiology and Immunology, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Safaa Ibrahim
- Department of Microbiology and Immunology, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA.,Department of Microbiology and Immunology, Faculty of Pharmacy, Cairo University, Egypt
| | - Sahithi Pamarthy
- Department of Microbiology and Immunology, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Mukesh K Jaiswal
- Department of Microbiology and Immunology, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Alice Gilman Sachs
- Department of Microbiology and Immunology, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Kenneth D Beaman
- Department of Microbiology and Immunology, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| |
Collapse
|
35
|
Technical innovation in adjuvant radiotherapy: Evolution and evaluation of new treatments for today and tomorrow. Breast 2015; 24 Suppl 2:S114-9. [PMID: 26429399 DOI: 10.1016/j.breast.2015.07.026] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Recent innovations in breast cancer radiotherapy include intensity modulated radiotherapy, brachytherapy and intraoperative radiotherapy and current trials are seeking to evaluate their value in optimizing local control while maintaining cosmetic effects. Future clinical dividends in local control and survival may come from the identification of molecular signatures of breast cancer radiosensitivity, the development of predictive signatures and identification of immunohistochemical markers of risk of local recurrence. The importance of tumour heterogeneity is being increasingly recognized as an important factor in determining radiotherapy response and an improved understanding of the biology of the tumour microenvironment may identify targets that allow enhanced radiosensitisation or reversal of radioresistance when inhibited. This review describes recent developments in these areas.
Collapse
|
36
|
Lu X, Chen L, Chen Y, Shao Q, Qin W. Bafilomycin A1 inhibits the growth and metastatic potential of the BEL-7402 liver cancer and HO-8910 ovarian cancer cell lines and induces alterations in their microRNA expression. Exp Ther Med 2015; 10:1829-1834. [PMID: 26640557 DOI: 10.3892/etm.2015.2758] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Accepted: 09/01/2015] [Indexed: 11/06/2022] Open
Abstract
The vacuolar H+-ATPase (V-ATPase) is commonly highly activated in cancer cells and is a potential target of anti-cancer therapy. Bafilomycin A1 is a specific inhibitor of the c subunit of V-ATPase. In the present study, the effects of bafilomycin A1 on the BEL-7402 hepatocellular carcinoma and HO-8910 ovarian cancer cell lines were respectively studied. In addition, the bafilomycin A1-induced alterations in the mRNAs and microRNAs (miRNAs) in the cells were detected using microarray methods. The results demonstrated that the growth of the two cell lines was retarded and the metastatic potential was inhibited by bafilomycin A1. Transmission electron microscopy and assays of capsase-3 and -9 suggested that bafilomycin A1 induced apoptosis. Gene Ontology analysis of the microarrays of mRNA-miRNA integrity showed altered pathways following bafilomycin A1 treatment, including pathways regulating glucose or lipid metabolism, DNA repair or duplication and lysosomes. Quantitative polymerase chain reaction analysis confirmed that miR-923, miR-1246, miR-149*, miR-638 and miR-210 were upregulated and miR-99a, miR-181a-2* and miR-339-5p were downregulated following bafilomycin A1 treatment. The overlapped altered miRs may be effective targets for the two types of solid tumor, and may have potential for application to the treatment of other types of solid tumor.
Collapse
Affiliation(s)
- Xiaodong Lu
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China ; State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Shanghai 200032, P.R. China
| | - Lufang Chen
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Yuanyuan Chen
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Qixiang Shao
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Wenxin Qin
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Shanghai 200032, P.R. China
| |
Collapse
|
37
|
Graham RM, Thompson JW, Webster KA. Inhibition of the vacuolar ATPase induces Bnip3-dependent death of cancer cells and a reduction in tumor burden and metastasis. Oncotarget 2015; 5:1162-73. [PMID: 24811485 PMCID: PMC4012732 DOI: 10.18632/oncotarget.1699] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
The pro-apoptotic protein Bnip3 is induced by hypoxia and is present in the core regions of most solid tumors. Bnip3 induces programmed necrosis by an intrinsic caspase independent mitochondrial pathway. Many tumor cells have evolved pathways to evade Bnip3-mediated death attesting to the physiological relevance of the survival threat imposed by Bnip3. We have reported that acidosis can trigger the Bnip3 death pathway in hypoxic cells therefore we hypothesized that manipulation of intracellular pH by pharmacological inhibition of the vacuolar (v)ATPase proton pump, a significant pH control pathway, may activate Bnip3 and promote death of hypoxic cells within the tumor. Here we confirm that bafilomycin A1 (BafA1), a selective vATPase inhibitor, significantly increased death of breast cancer cells in a hypoxia and Bnip3-dependent manner and significantly reduced tumor growth in MCF7 and MDA-MB-231 mouse xenografts. Combined treatment of cells with BafA1 and the ERK1/2 inhibitor U0126 further augmented cell death. Combined treatment of mice containing MDA-MB-231 xenografts with BafA1 and the ERK1/2 inhibitor sorafenib was superior to either treatment alone and supported tumor regression. BafA1 and sorafenib treatments alone reduced MDA-MB-231 cell metastasis and again the combination was significantly more effective than either treatment alone and was without apparent side effects. These results present a novel mechanism to destroy hypoxic tumor cells that may help reverse the resistance of hypoxic tumors to radiation and chemotherapy and perhaps target tumor stem cells.
Collapse
|
38
|
Cotter K, Capecci J, Sennoune S, Huss M, Maier M, Martinez-Zaguilan R, Forgac M. Activity of plasma membrane V-ATPases is critical for the invasion of MDA-MB231 breast cancer cells. J Biol Chem 2014; 290:3680-92. [PMID: 25505184 DOI: 10.1074/jbc.m114.611210] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The vacuolar (H(+))-ATPases (V-ATPases) are a family of ATP-driven proton pumps that couple ATP hydrolysis with translocation of protons across membranes. Previous studies have implicated V-ATPases in cancer cell invasion. It has been proposed that V-ATPases participate in invasion by localizing to the plasma membrane and causing acidification of the extracellular space. To test this hypothesis, we utilized two separate approaches to specifically inhibit plasma membrane V-ATPases. First, we stably transfected highly invasive MDA-MB231 cells with a V5-tagged construct of the membrane-embedded c subunit of the V-ATPase, allowing for extracellular expression of the V5 epitope. We evaluated the effect of addition of a monoclonal antibody directed against the V5 epitope on both V-ATPase-mediated proton translocation across the plasma membrane and invasion using an in vitro Matrigel assay. The addition of anti-V5 antibody resulted in acidification of the cytosol and a decrease in V-ATPase-dependent proton flux across the plasma membrane in transfected but not control (untransfected) cells. These results demonstrate that the anti-V5 antibody inhibits activity of plasma membrane V-ATPases in transfected cells. Addition of the anti-V5 antibody also inhibited in vitro invasion of transfected (but not untransfected) cells. Second, we utilized a biotin-conjugated form of the specific V-ATPase inhibitor bafilomycin. When bound to streptavidin, this compound cannot cross the plasma membrane. Addition of this compound to MDA-MB231 cells also inhibited in vitro invasion. These studies suggest that plasma membrane V-ATPases play an important role in invasion of breast cancer cells.
Collapse
Affiliation(s)
- Kristina Cotter
- From the Department of Developmental, Molecular and Chemical Biology, Tufts University School of Medicine and the Program in Cellular and Molecular Physiology, Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, Massachusetts 02111
| | - Joseph Capecci
- From the Department of Developmental, Molecular and Chemical Biology, Tufts University School of Medicine and the Program in Cellular and Molecular Physiology, Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, Massachusetts 02111
| | - Souad Sennoune
- the Department of Cell Physiology and Molecular Biophysics, Texas Tech University, Lubbock, Texas 79430
| | - Markus Huss
- the Department of Biology/Chemistry, Division of Animal Physiology, University of Osnabrück, 49069 Osnabrück, Germany, and
| | - Martin Maier
- the Institute of Organic Chemistry, University of Tuebingen, 72076 Tuebingen, Germany
| | - Raul Martinez-Zaguilan
- the Department of Cell Physiology and Molecular Biophysics, Texas Tech University, Lubbock, Texas 79430
| | - Michael Forgac
- From the Department of Developmental, Molecular and Chemical Biology, Tufts University School of Medicine and the Program in Cellular and Molecular Physiology, Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, Massachusetts 02111,
| |
Collapse
|
39
|
Yang B, He XB. Tumor metastasis suppressor gene-1 and tumors. Shijie Huaren Xiaohua Zazhi 2014; 22:5291-5297. [DOI: 10.11569/wcjd.v22.i34.5291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Tumor metastasis suppressor gene-1 (TMSG-1) is a newly discovered tumor metastasis suppressor gene that plays important roles in promoting apoptosis and inhibiting invasion and metastasis of tumor cells. The inhibitory function of TMSG-1 in tumor cells may be related to vacuolar H+-ATPase and ceramide, but the underlying mechanism remains unknown. This review discusses the discovery of TMSG-1, the structure of TMSG-1 protein, as well as its possible mechanisms of action, expression in tumors and applications.
Collapse
|
40
|
Prognostic value of Rab27B nuclear expression in gastrointestinal stromal tumors. DISEASE MARKERS 2014; 2014:942181. [PMID: 25382899 PMCID: PMC4213986 DOI: 10.1155/2014/942181] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Accepted: 09/22/2014] [Indexed: 12/31/2022]
Abstract
Rab proteins of the endocytosis and exocytosis pathways both play critical roles in cancer progression, and Rab27B has a significant relationship with several types of human cancer. However, the association between Rab27B expression and clinical features to determine its clinicopathological significance in gastrointestinal tumor (GIST) has not been investigated. To examine the expression of Rab27B in GIST and investigate the association between its expression and patient prognosis, immunohistochemistry analysis with tissue microarray was used to evaluate expression of Rab27B in 162 patients with GIST. The relationship between Rab27B expression and patient prognosis was analyzed. High nuclear staining of Rab27B was detected in 88 of 162 (54.32%) GIST tissues. Positive staining of Rab27B was significantly associated with tumor size (P = 0.006), mitotic index (P = 0.013), Armed Forces Institute of Pathology Miettinen risk classification (P = 0.002), and tumor grade (P = 0.021). Kaplan-Meier survival curves showed that GIST patients with low Rab27B nuclear expression (P = 0.038) and mitotic index <5 per 50 high-power fields (P = 0.029) had a more favorable prognosis. These findings indicate that Rab27B nuclear expression is correlated with several clinicopathological features of GIST patients, and it may serve as an unfavorable prognostic marker.
Collapse
|
41
|
Schempp CM, von Schwarzenberg K, Schreiner L, Kubisch R, Müller R, Wagner E, Vollmar AM. V-ATPase inhibition regulates anoikis resistance and metastasis of cancer cells. Mol Cancer Ther 2014; 13:926-37. [PMID: 24482380 DOI: 10.1158/1535-7163.mct-13-0484] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Fighting metastasis is a major challenge in cancer therapy and novel therapeutic targets and drugs are highly appreciated. Resistance of invasive cells to anoikis, a particular type of apoptosis induced by loss of cell-matrix contact, is a major prerequisite for their metastatic spread. Inducing anoikis in metastatic cancer cells is therefore a promising therapeutic approach. The vacuolar-ATPase (V-ATPase), a proton pump located at the membrane of acidic organelles, has recently come to focus as an antimetastatic cancer target. As V-ATPase inhibitors have shown to prevent invasion of tumor cells and are able to induce apoptosis, we proposed that V-ATPase inhibition induces anoikis-related pathways in invasive cancer cells. We used the V-ATPase inhibitor archazolid to investigate the mechanism of anoikis induction in various metastatic cancer cells (T24, MDA-MB-231, 4T1, 5637) in vitro. Anoikis induction by archazolid was characterized by decreased c-FLIP expression and caspase-8 activation as well as reduction of active integrin-β1 and an early increase of the proapoptotic protein BIM. However, we observed that archazolid also induces mechanisms opposing anoikis such as degradation of BIM mediated by extracellular signal-regulated kinase (ERK), Akt and Src kinases at later time points and induction of reactive oxygen species. Still, intravenous injection of archazolid-treated 4T1-Luc2 mouse breast cancer cells resulted in reduced metastasis in mouse lungs. Thus, V-ATPase inhibition is not only an interesting option to reduce cancer metastasis, but also to better understand anoikis resistance and to find choices to fight against it.
Collapse
Affiliation(s)
- Christina M Schempp
- Authors' Affiliations: Department of Pharmacy, Pharmaceutical Biology and Department of Pharmacy, Pharmaceutical Biotechnology, University of Munich, Munich; and Helmholtz Institute for Pharmaceutical Research Saarland, Helmholtz Centre for Infection Research and Department of Pharmaceutical Biotechnology, Saarland University, Saarbrücken, Germany
| | | | | | | | | | | | | |
Collapse
|
42
|
Liu Q, Tang H, Liu X, Liao Y, Li H, Zhao Z, Yuan X, Jiang W. miR-200b as a prognostic factor targets multiple members of RAB family in glioma. Med Oncol 2014; 31:859. [DOI: 10.1007/s12032-014-0859-x] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2013] [Accepted: 01/22/2014] [Indexed: 12/23/2022]
|
43
|
Rath S, Liebl J, Fürst R, Vollmar AM, Zahler S. Regulation of endothelial signaling and migration by v-ATPase. Angiogenesis 2013; 17:587-601. [PMID: 24254321 DOI: 10.1007/s10456-013-9408-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2013] [Accepted: 11/12/2013] [Indexed: 01/04/2023]
Abstract
The vacuolar ATPase (v-ATPase) is a proton pump, able to acidify intracellular compartments and the pericellular space. v-ATPase has extensively been studied in various functional contexts, e.g., migration of tumor cells, and inhibition of v-ATPase has been proven as intriguing novel therapeutic concept. Since the role of v-ATPase in endothelial cell migration and angiogenesis has scarcely been investigated, we examined the consequences of pharmacological inhibition of v-ATPase (by concanamycin) on proliferation, migration, VEGF-receptor 2 (VEGFR2) trafficking and signaling, as well as Notch-mediated transcription in endothelial cells [human microvascular endothelial cells (HMEC-1) and human umbilical vein endothelial cells (HUVEC)] Treatment of the cells with 3 or 10 nM of the v-ATPase inhibitor concanamycin for 48 h or longer inhibited proliferation and arrested cell cycle in the G2/M phase in HMEC-1, while a G1 phase arrest occurred in HUVEC. Already after 24 h these concentrations reduced migration (scratch assay, chemotactic gradient). Activation of the small GTPase Rac1 in freshly adherent cells was reduced by concanamycin. Downstream signaling of the VEGFR2 (phosphorylation of ERK1/2 and AKT), as well as autophosphorylation of VEGFR2 were inhibited. VEGFR2 on the cell surface was reduced, and sequestered in a lysosomal compartment. In addition, concanamycin blocked transcription of the Notch target genes Hey1 and Hey2 after stimulation with DLL4. Since the impaired signaling pathways (Rac-1, VEGFR2, Notch) all depend on vesicular recycling circuits, we conclude that the disturbance of these is the main mode of action of v-ATPase inhibition in endothelial cells, offering an attractive multi-factorial anti-angiogenic approach.
Collapse
Affiliation(s)
- Sebastian Rath
- Department of Pharmacy, Ludwig-Maximilians-Universität München, Butenandtstr. 5-13, 81377, Munich, Germany
| | | | | | | | | |
Collapse
|
44
|
Capecci J, Forgac M. The function of vacuolar ATPase (V-ATPase) a subunit isoforms in invasiveness of MCF10a and MCF10CA1a human breast cancer cells. J Biol Chem 2013; 288:32731-32741. [PMID: 24072707 DOI: 10.1074/jbc.m113.503771] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The vacuolar H(+) ATPases (V-ATPases) are ATP-driven proton pumps that transport protons across both intracellular and plasma membranes. Previous studies have implicated V-ATPases in the invasiveness of various cancer cell lines. In this study, we evaluated the role of V-ATPases in the invasiveness of two closely matched human breast cancer lines. MCF10a cells are a non-invasive, immortalized breast epithelial cell line, and MCF10CA1a cells are a highly invasive, H-Ras-transformed derivative of MCF10a cells selected for their metastatic potential. Using an in vitro Matrigel assay, MCF10CA1a cells showed a much higher invasion than the parental MCF10a cells. Moreover, this increased invasion was completely sensitive to the specific V-ATPase inhibitor concanamycin. MCF10CA1a cells expressed much higher levels of both a1 and a3 subunit isoforms relative to the parental line. Isoforms of subunit a are responsible for subcellular localization of V-ATPases, with a3 and a4 targeting V-ATPases to the plasma membrane of specialized cells. Knockdown of either a3 alone or a3 and a4 together using isoform-specific siRNAs inhibited invasion by MCF10CA1a cells. Importantly, overexpression of a3 but not the other a subunit isoforms greatly increased the invasiveness of the parental MCF10a cells. Similarly, overexpression of a3 significantly increased expression of V-ATPases at the plasma membrane. These studies suggest that breast tumor cells employ particular a subunit isoforms to target V-ATPases to the plasma membrane, where they function in tumor cell invasion.
Collapse
Affiliation(s)
- Joseph Capecci
- From the Department of Developmental, Molecular, and Chemical Biology, Tufts University School of Medicine and the Program in Cellular and Molecular Physiology, Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, Massachusetts 02111
| | - Michael Forgac
- From the Department of Developmental, Molecular, and Chemical Biology, Tufts University School of Medicine and the Program in Cellular and Molecular Physiology, Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, Massachusetts 02111.
| |
Collapse
|
45
|
Lu Q, Lu S, Huang L, Wang T, Wan Y, Zhou CX, Zhang C, Zhang Z, Li X. The expression of V-ATPase is associated with drug resistance and pathology of non-small-cell lung cancer. Diagn Pathol 2013; 8:145. [PMID: 23984887 PMCID: PMC3846167 DOI: 10.1186/1746-1596-8-145] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2013] [Accepted: 08/26/2013] [Indexed: 11/28/2022] Open
Abstract
Objective This article aims to investigate the expression of vacuolar-H + −ATPase (V-ATPase) in non-small cell lung cancer (NSCLC) and its variations with pathological type and grade. Furthermore, to evaluate the chemotherapy drug sensitivity of different cancer tissues as well as its correlation with V-ATPase expression in NSCLC. Methods V-ATPase expression was examined in 92 NSCLC tissue samples using the immunohistochemical Envision method and immunofluorescence assay. The location of V-ATPase expression was observed by confocal laser scanning microscopy and the difference of its expression rate was evaluated. The sensitivity of cancer tissues to chemotherapy drug was examined using MTT assay and its correlation with the V-ATPase expression was tested in NSCLC by Spearman rank correlation analysis. Results V-ATPase expression was mainly localized in the cell membrane and cytoplasm. The expression rate of V-ATPase was 71.43% in squamous cell lung cancer, significantly lower than that of the lung adenocarcinoma (83.72%, P = 0.000). In different pathological grades of squamous cell lung cancer, the expression rate of V-ATPase was 58.33% in grade II, significantly lower than that of the grade III (84.00%, P = 0.014). The expression rate of V-ATPase in grade II lung adenocarcinoma was 76.67%, significantly lower than that of the grade ΙΙΙ adenocarcinoma (100.0%, P = 0.012). Correlation analysis showed that the sensitivity of NSCLC tissues to cyclophosphamide, gemcitabine, doxorubicin, paclitaxel and cisplatin was significantly correlated with the V-ATPase expression rate (P < 0.05). Conclusions V-ATPase was overexpressed in NSCLC. The expression of V-ATPase was related to the pathological type and grade of cancer and was likely associated with chemotherapy drug resistance in NSCLC. Virtual slides The virtual slide(s) for this article can be found here: http://www.diagnosticpathology.diagnomx.eu/vs/7515811511020000
Collapse
Affiliation(s)
- Qiang Lu
- Department of Thoracic Surgery, Tangdu Hospital, Fourth Military Medical University, 710038 Xi'an, PR China.
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Hendrix A, De Wever O. Rab27 GTPases distribute extracellular nanomaps for invasive growth and metastasis: implications for prognosis and treatment. Int J Mol Sci 2013; 14:9883-92. [PMID: 23665896 PMCID: PMC3676819 DOI: 10.3390/ijms14059883] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2013] [Revised: 04/19/2013] [Accepted: 05/03/2013] [Indexed: 12/27/2022] Open
Abstract
The Rab27 family of small GTPases regulates exocytosis of distinct vesicle types including multivesicular endosomes, which results in the release of exosomes. Exosomes are nanometer-sized membrane vesicles that enclose soluble factors such as proteins and nucleic acids within a lipid bilayer and can travel toward distant tissues to influence multiple aspects of cell behavior. In our view that tumors are endocrine organs producing exosomes, Rab27 GTPases and their effector proteins are critical determinants for invasive growth and metastasis. Rab27 proteins and their effectors may serve as prognostic biomarkers or as targets for patient-tailored therapy.
Collapse
Affiliation(s)
- An Hendrix
- Laboratory of Experimental Cancer Research, Department of Radiation Oncology and Experimental Cancer Research, Ghent University Hospital, De Pintelaan 185, 9000 Ghent, Belgium.
| | | |
Collapse
|