1
|
Tafech A, Stéphanou A. On the Importance of Acidity in Cancer Cells and Therapy. BIOLOGY 2024; 13:225. [PMID: 38666837 PMCID: PMC11048434 DOI: 10.3390/biology13040225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 03/25/2024] [Accepted: 03/27/2024] [Indexed: 04/28/2024]
Abstract
Cancer cells are associated with high glycolytic activity, which results in acidification of the tumor microenvironment. The occurrence of this stressful condition fosters tumor aggressiveness, with the outcome of invasiveness and metastasis that are linked to a poor clinical prognosis. Acidosis can be both the cause or consequence of alterations in the functions and expressions of transporters involved in intracellular acidity regulation. This review aims to explore the origin of acidity in cancer cells and the various mechanisms existing in tumors to resist, survive, or thrive in the acidic environment. It highlights the difficulties in measuring the intracellular pH evolution that impedes our understanding of the many regulatory and feedback mechanisms. It finally presents the consequences of acidity on tumor development as well as the friend or foe role of acidity in therapy.
Collapse
Affiliation(s)
| | - Angélique Stéphanou
- Université Grenoble Alpes, CNRS, UMR 5525, VetAgro Sup, Grenoble INP, TIMC, 38000 Grenoble, France
| |
Collapse
|
2
|
Tejeda-Muñoz N, Mei KC, Sheladiya P, Monka J. Targeting Membrane Trafficking as a Strategy for Cancer Treatment. Vaccines (Basel) 2022; 10:vaccines10050790. [PMID: 35632546 PMCID: PMC9144176 DOI: 10.3390/vaccines10050790] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 05/07/2022] [Accepted: 05/15/2022] [Indexed: 12/17/2022] Open
Abstract
Membrane trafficking is emerging as an attractive therapeutic strategy for cancer. Recent reports have found a connection between Wnt signaling, receptor-mediated endocytosis, V-ATPase, lysosomal activity, and macropinocytosis through the canonical Wnt pathway. In macropinocytic cells, a massive internalization of the plasma membrane can lead to the loss of cell-surface cadherins, integrins, and other antigens that mediate cell–cell adhesion, favoring an invasive phenotype. V-ATPase is a key regulator in maintaining proper membrane trafficking, homeostasis, and the earliest developmental decisions in the Xenopus vertebrate development model system. Here, we review how the interference of membrane trafficking with membrane trafficking inhibitors might be clinically relevant in humans.
Collapse
Affiliation(s)
- Nydia Tejeda-Muñoz
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, CA 90095-1662, USA; (P.S.); (J.M.)
- Correspondence:
| | - Kuo-Ching Mei
- Division of Pharmacoengineering and Molecular School Pharmaceutics, Eshelman of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA;
| | - Pooja Sheladiya
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, CA 90095-1662, USA; (P.S.); (J.M.)
| | - Julia Monka
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, CA 90095-1662, USA; (P.S.); (J.M.)
| |
Collapse
|
3
|
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: 30] [Impact Index Per Article: 15.0] [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
|
4
|
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
|
5
|
Yao J, Zhang Z, Li S, Li B, Wang XH. Melittin inhibits proliferation, migration and invasion of bladder cancer cells by regulating key genes based on bioinformatics and experimental assays. J Cell Mol Med 2019; 24:655-670. [PMID: 31691530 PMCID: PMC6933335 DOI: 10.1111/jcmm.14775] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 09/05/2019] [Accepted: 09/30/2019] [Indexed: 12/12/2022] Open
Abstract
The antitumour effect of melittin (MEL) has recently attracted considerable attention. Nonetheless, information regarding the functional role of MEL in bladder cancer (BC) is currently limited. Herein, we investigated the effect of MEL on critical module genes identified in BC. In total, 2015 and 4679 differentially expressed genes (DEGs) associated with BC were identified from the GSE31189 set and The Cancer Genome Atlas database, respectively. GSE‐identified DEGs were mapped and analysed using Gene Ontology and Kyoto Encyclopaedia of Genes and Genomes analyses to determine BC‐involved crucial genes and signal pathways. Coupled with protein–protein interaction network and Molecular Complex Detection analyses, Modules 2 and 4 were highlighted in the progression of BC. In in‐vitro experiments, MEL inhibited the proliferation, migration, and invasion of UM‐UC‐3 and 5637 cells. The expression of NRAS, PAK2, EGFR and PAK1 in Module 4—enriched in the MAPK signalling pathway—was significantly reduced after treatment with MEL at concentrations of 4 or 6 μg/mL. Finally, quantitative reverse transcription‐polymerase chain reaction and Western blotting analyses revealed MEL inhibited the expression of genes at the mRNA (ERK1/2, ERK5, JNK and MEK5), protein (ERK5, MEK5, JNK and ERK1/2) and phosphorylation (p‐ERK1/2, p‐JNK, and p‐38) levels. This novel evidence indicates MEL exerts effects on the ERK5‐MAK pathway—a branch of MAPK signalling pathway. Collectively, these findings provide a theoretical basis for MEL application in BC treatment.
Collapse
Affiliation(s)
- Jie Yao
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China.,Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Zhan Zhang
- Department of Rehabilitation Medicine, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Sheng Li
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Bai Li
- Department of Rehabilitation Medicine, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Xing-Huan Wang
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
6
|
Targeting V-ATPase Isoform Restores Cisplatin Activity in Resistant Ovarian Cancer: Inhibition of Autophagy, Endosome Function, and ERK/MEK Pathway. JOURNAL OF ONCOLOGY 2019; 2019:2343876. [PMID: 31057611 PMCID: PMC6463777 DOI: 10.1155/2019/2343876] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 01/28/2019] [Accepted: 03/04/2019] [Indexed: 02/06/2023]
Abstract
Ovarian cancer (OVCA) patients often develop tolerance to standard platinum therapy that accounts for extensive treatment failures. Cisplatin resistant OVCA cells (cis-R) display enhanced survival mechanisms to cope with therapeutic stress. In these cells, increased autophagy process assists in chemoresistance by boosting the nutrient pool under stress. To improve the treatment response, both protective autophagy inhibition and its overactivation are showing efficacy in chemosensitization. Autophagy requires a tightly regulated intracellular pH. Vacuolar ATPases (V-ATPases) are proton extruding nanomotors present on cellular/vesicular membranes where they act as primary pH regulators. V-ATPase ‘a2' isoform (V0a2), the major pH sensing unit, is markedly overexpressed on the plasma membrane and the early endosomes of OVCA cells. Previously, V0a2 inhibition sensitized cis-R cells to platinum drugs by acidifying cytosolic pH that elevated DNA damage. Here, we examined how V0a2 inhibition affected endosomal function and the autophagy process as a possible factor for cisplatin sensitization. Clinically, V0a2 expression was significantly higher in tissues from drug nonresponder OVCA patients compared to treatment responders. In vitro V0a2 knockdown in cis-R cells (sh-V0a2-cisR) significantly reduced the tumor sphere-forming ability and caused complete disintegration of the spheres upon cisplatin treatment. The apoptotic capacity of sh-V0a2-cisR improved substantially with potentiation of both intrinsic and extrinsic apoptotic pathway when treated with cisplatin. Unlike the chemical V-ATPase inhibitors that acutely induce autophagy, here, the stable V0a2 inhibition dampened the protective autophagy process in sh-V0a2-cisR cells with downregulated expression of proteins beclin-1, ATG-7, and LC3B and low autophagosome numbers compared to control cis-R cells. These cells showed downregulated ERK/MEK pathway that is known to repress autophagy. Interestingly, upon cisplatin treatment of sh-V0a2-cisR, the autophagy initiation proteins (LC3B, ATG7, and Beclin 1) were found upregulated as a stress response compared to the untreated cells. However, there was a concomitant downstream autophagosome accumulation and an enhanced P62 protein levels indicating the overall block in autophagy flux. Mechanistically, V0a2 knockdown caused defects in early endosome function as the transferrin internalization was impaired. Taken together, this study provides a novel insight into the mechanism by which V-ATPase-isoform regulates autophagy that assists in chemoresistance in ovarian cancer. We conclude that V-ATPase-V0a2 is a potent target for developing an effective treatment to enhance patient survival rates in ovarian cancer.
Collapse
|
7
|
Zhang M, Saad C, Le L, Halfter K, Bauer B, Mansmann UR, Li J. Computational modeling of methionine cycle-based metabolism and DNA methylation and the implications for anti-cancer drug response prediction. Oncotarget 2018; 9:22546-22558. [PMID: 29875994 PMCID: PMC5989406 DOI: 10.18632/oncotarget.24547] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Accepted: 07/29/2017] [Indexed: 12/14/2022] Open
Abstract
The relationship between metabolism and methylation is considered to be an important aspect of cancer development and drug efficacy. However, it remains poorly defined how to apply this aspect to improve preclinical disease characterization and clinical treatment outcome. Using available molecular information from Kyoto Encyclopedia of Genes and Genomes (KEGG) and literature, we constructed a large-scale knowledge-based metabolic in silico model. For the purpose of model validation, we applied data from the Cancer Cell Line Encyclopedia (CCLE) to investigate computationally the impact of metabolism on chemotherapy efficacy. In our model, different metabolic components such as MAT2A, ATP6V0E1, NNMT involved in methionine cycle correlate with biologically measured chemotherapy outcome (IC50) that are in agreement with findings of independent studies. These proteins are potentially also involved in cellular methylation processes. In addition, several components such as 3,4-dihydoxymandelate, PAPSS2, UPP1 from metabolic pathways involved in the production of purine and pyrimidine correlate with IC50. This study clearly demonstrates that complex computational approaches can reflect findings of biological experiments. This demonstrates their high potential to grasp complex issues within systems medicine such as response prediction, biomarker identification using available data resources.
Collapse
Affiliation(s)
- Mengying Zhang
- Institute for Medical Informatics, Biometry and Epidemiology, Ludwig-Maximilians University of München, Munich, Germany
| | - Christian Saad
- Department of Computational Science, University of Augsburg, Augsburg, Germany
| | - Lien Le
- Institute for Medical Informatics, Biometry and Epidemiology, Ludwig-Maximilians University of München, Munich, Germany
| | - Kathrin Halfter
- Institute for Medical Informatics, Biometry and Epidemiology, Ludwig-Maximilians University of München, Munich, Germany
| | - Bernhard Bauer
- Department of Computational Science, University of Augsburg, Augsburg, Germany
| | - Ulrich R Mansmann
- Institute for Medical Informatics, Biometry and Epidemiology, Ludwig-Maximilians University of München, Munich, Germany.,German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Jian Li
- Institute for Medical Informatics, Biometry and Epidemiology, Ludwig-Maximilians University of München, Munich, Germany.,German Cancer Consortium (DKTK), Heidelberg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
8
|
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
|
9
|
Kitazawa S, Nishizawa S, Nakagawa H, Funata M, Nishimura K, Soga T, Hara T. Cancer with low cathepsin D levels is susceptible to vacuolar (H + )-ATPase inhibition. Cancer Sci 2017; 108:1185-1193. [PMID: 28317223 PMCID: PMC5480082 DOI: 10.1111/cas.13240] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Revised: 03/09/2017] [Accepted: 03/13/2017] [Indexed: 01/17/2023] Open
Abstract
Vacuolar (H+)‐ATPases (V‐ATPases) have important roles in the supply of nutrients to tumors by mediating autophagy and the endocytic uptake of extracellular fluids. Accordingly, V‐ATPases are attractive therapeutic targets for cancer. However, the clinical use of V‐ATPase inhibitors as anticancer drugs has not been realized, possibly owing to their high toxicity in humans. Inhibition of V‐ATPase may be an appropriate strategy in highly susceptible cancers. In this study, we explored markers of V‐ATPase inhibitor sensitivity. V‐ATPase inhibitors led to pH impairment in acidic intracellular compartments, suppression of macropinocytosis, and decreased intracellular amino acid levels. The sensitivity of cells to V‐ATPase inhibitors was correlated with low cathepsin D expression, and cancer cells showed increased sensitivity to V‐ATPase inhibitors after pretreatment with a cathepsin D inhibitor and siRNA targeting the cathepsin D gene (CTSD). In addition, V‐ATPase inhibitor treatment led to the induction of the amino acid starvation response, upregulation of endoplasmic reticulum stress markers, and suppression of mammalian target of rapamycin (mTOR) signaling in cells expressing low levels of cathepsin D. Some colorectal cancer patients showed the downregulation of cathepsin D in tumor tissues compared with matched normal tissues. These findings indicate that V‐ATPase inhibitors are promising therapeutic options for cancers with downregulated cathepsin D.
Collapse
Affiliation(s)
- Satoshi Kitazawa
- Oncology Drug Discovery Unit, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Satoru Nishizawa
- Oncology Drug Discovery Unit, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Hideyuki Nakagawa
- Biomolecular Research Laboratories, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Masaaki Funata
- Biomolecular Research Laboratories, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Kazuho Nishimura
- Oncology Drug Discovery Unit, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Tomoyoshi Soga
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Japan
| | - Takahito Hara
- Oncology Drug Discovery Unit, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| |
Collapse
|
10
|
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
|
11
|
Zhang JL, Liu M, Yang Q, Lin SY, Shan HB, Wang HY, Xu GL. Effects of omeprazole in improving concurrent chemoradiotherapy efficacy in rectal cancer. World J Gastroenterol 2017; 23:2575-2584. [PMID: 28465642 PMCID: PMC5394521 DOI: 10.3748/wjg.v23.i14.2575] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Revised: 01/26/2017] [Accepted: 02/17/2017] [Indexed: 02/06/2023] Open
Abstract
AIM To explore the effects of omeprazole on chemoradiotherapy efficacy and tumor recurrence in rectal cancer.
METHODS The medical data of 125 rectal cancer patients who received the same neoadjuvant chemoradiotherapy (CRT) followed by surgery were retrospectively collected. Patients who received omeprazole (OME) orally at a dose of 20 mg at least once daily for six days and/or intravenously at 40 mg a day were recognized as eligible OME users (EOU). Otherwise, patients were regarded as non-eligible OME users (non-EOU). Moreover, a preferred OME dose cut-off of 200 mg on tumor recurrence was obtained by receiver operating characteristic (ROC) curves. Patients were divided into two groups: the effective OME group (EOG, OME ≥ 200 mg) and the non-effective OME group (non-EOG, OME < 200 mg).
RESULTS The good response rate of CRT efficacy (50.8%) in EOU was significantly increased compared with non-EOU (30.6%) (P = 0.02). The recurrence rate in the EOG was 10.3%, which was significantly lower compared with 31.3% in non-EOG (P = 0.025). The good response rate of CRT efficacy in EOG was 55.2%, which was obviously higher compared with 36.5% in non-EOG, with a significant difference (P = 0.072). Multivariate Cox analysis demonstrated that OME (non-EOG and EOG) was an independent and significant impact factor for DFS (P = 0.048, HR = 0.30, 95%CI: 0.09-0.99).
CONCLUSION When applied as an adjuvant drug in cancer treatment for relieving common side effects of chemotherapy, omeprazole has a synergetic effect in improving CRT efficacy and decreasing rectal cancer recurrence.
Collapse
|
12
|
Falcone R, Roberto M, D'Antonio C, Romiti A, Milano A, Onesti CE, Marchetti P, Fais S. High-doses of proton pump inhibitors in refractory gastro-intestinal cancer: A case series and the state of art. Dig Liver Dis 2016; 48:1503-1505. [PMID: 27651344 DOI: 10.1016/j.dld.2016.08.126] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Revised: 08/06/2016] [Accepted: 08/09/2016] [Indexed: 12/11/2022]
Abstract
BACKGROUND In recent years, proton pump inhibitors (PPIs) have been investigated at high-dose to modulate tumour microenvironment acidification thus restoring chemotherapeutic sensitivity. Moreover, several clinical data supports the role of cytotoxic drugs at low-dose continuously delivered as anticancer therapy. METHODS Clinical records of three patients affected with gastrointestinal cancer refractory to standard treatments, who had received a combination of high-dose rabeprazole and metronomic chemotherapy were reviewed. RESULTS The first case, a 78-year-old man was treated for lung metastasis from colon adenocarcinoma. The second case, a 73-year-old man was treated for metastatic rectal cancer to the liver. The third one, a 68-year-old man, underwent the combination regimen for colon cancer with lung, liver and peritoneal metastases. CONCLUSIONS Despite the failure of previous standard chemotherapy for metastatic disease, good clinical outcome was shown in these patients treated with an unconventional association of high-dose PPIs and metronomic chemotherapy.
Collapse
Affiliation(s)
- Rosa Falcone
- Department of Molecular and Clinical Medicine, "Sapienza" University of Rome, Italy.
| | - Michela Roberto
- Department of Molecular and Clinical Medicine, "Sapienza" University of Rome, Italy
| | - Chiara D'Antonio
- Department of Molecular and Clinical Medicine, "Sapienza" University of Rome, Italy
| | - Adriana Romiti
- Department of Molecular and Clinical Medicine, "Sapienza" University of Rome, Italy
| | - Annalisa Milano
- Department of Molecular and Clinical Medicine, "Sapienza" University of Rome, Italy
| | | | - Paolo Marchetti
- Department of Molecular and Clinical Medicine, "Sapienza" University of Rome, Italy
| | | |
Collapse
|
13
|
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: 55] [Impact Index Per Article: 6.9] [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
|
14
|
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
|
15
|
Kazami S, Takaine M, Itoh H, Kubota T, Kobayashi J, Usui T. Iejimalide C is a potent V-ATPase inhibitor, and induces actin disorganization. Biol Pharm Bull 2015; 37:1944-7. [PMID: 25451843 DOI: 10.1248/bpb.b14-00548] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Iejimalides (IEJLs) A-D are 24-membered macrolides isolated from a tunicate Eudistoma cf. rigida, and exhibit potent cytotoxicity in vitro and antitumor activity in vivo. We previously reported that the molecular target of IEJL-A and -B was the vacuolar-type H(+)-ATPases (V-ATPases). However IEJL-C and -D, which are sulfonylated IEJL-A and -B, respectively, show more potent antitumor activity, and their molecular targets remain to be discovered. Here, we report that IEJL-C is also a potent V-ATPase inhibitor by binding in a site similar to the bafilomycin-binding site. Two-hour treatment with IEJL-C resulted in the complete disappearance of acidic organelles in HeLa cells. Interestingly, after 24-h treatment, small actin aggregates were observed instead of actin fibers. The same actin reorganization was also observed in cells treated with another V-ATPase inhibitor, bafilomycin A1. Because IEJLs did not inhibit actin polymerization in vitro, these results suggest that the primary target of IEJL-C, as well as IEJL-A and -B, is V-ATPase, and actin reorganizations are probably caused by the disruption of pH homeostasis via V-ATPase inhibition.
Collapse
Affiliation(s)
- Sayaka Kazami
- Tsukuba Research Laboratory, Hamamatsu Photonics K.K
| | | | | | | | | | | |
Collapse
|
16
|
Katara GK, Kulshrestha A, Jaiswal MK, Pamarthy S, Gilman-Sachs A, Beaman KD. Inhibition of vacuolar ATPase subunit in tumor cells delays tumor growth by decreasing the essential macrophage population in the tumor microenvironment. Oncogene 2015; 35:1058-65. [PMID: 25961933 DOI: 10.1038/onc.2015.159] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Revised: 02/17/2015] [Accepted: 03/11/2015] [Indexed: 01/04/2023]
Abstract
In cancer cells, vacuolar ATPase (V-ATPase), a multi-subunit enzyme, is expressed on the plasma as well as vesicular membranes and critically influences metastatic behavior. The soluble, cleaved N-terminal domain of V-ATPase a2 isoform is associated with in vitro induction of tumorigenic characteristics in macrophages. This activity led us to further investigate its in vivo role in cancer progression by inhibition of a2 isoform (a2V) in tumor cells and the concomitant effect on tumor microenvironment in the mouse 4T-1 breast cancer model. Results showed that macrophages cocultivated with a2V knockdown (sh-a2) 4T-1 cells produce lower amounts of tumorigenic factors in vitro and have reduced ability to suppress T-cell activation and proliferation compared with control 4T-1 cells. Data analysis showed a delayed mammary tumor growth in Balb/c mice inoculated with sh-a2 4T-1 cells compared with control. The purified CD11b(+) macrophages from sh-a2 tumors showed a reduced expression of mannose receptor-1 (CD206), interleukin-10, transforming growth factor-β, arginase-1, matrix metalloproteinase and vascular endothelial growth factor. Flow cytometric analysis of tumor-infiltrated macrophages showed a significantly low number of F4/80(+)CD11c(+)CD206(+) macrophages in sh-a2 tumors compared with control. In sh-a2 tumors, most of the macrophages were F4/80(+)CD11c(+) (antitumor M1 macrophages) suggesting it to be the reason behind delayed tumor growth. Additionally, tumor-infiltrating macrophages from sh-a2 tumors showed a reduced expression of CD206 compared with control whereas CD11c expression was unaffected. These findings demonstrate that in the absence of a2V in tumor cells, the resident macrophage population in the tumor microenvironment is altered which affects in vivo tumor growth. We suggest that by involving the host immune system, tumor growth can be controlled through targeting of a2V on tumor cells.
Collapse
Affiliation(s)
- G K Katara
- Department of Microbiology and Immunology, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - A Kulshrestha
- Department of Microbiology and Immunology, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - M K Jaiswal
- Department of Microbiology and Immunology, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - S Pamarthy
- Department of Microbiology and Immunology, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - A Gilman-Sachs
- Department of Microbiology and Immunology, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - K D Beaman
- Department of Microbiology and Immunology, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| |
Collapse
|
17
|
Kazami S, Muroi M, Kawatani M, Kubota T, Usui T, Kobayashi J, Osada H. Iejimalides Show Anti-Osteoclast ActivityviaV-ATPase Inhibition. Biosci Biotechnol Biochem 2014; 70:1364-70. [PMID: 16794315 DOI: 10.1271/bbb.50644] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Iejimalides (IEJLs), 24-membered macrolides, are potent antitumor compounds, but their molecular targets remain to be revealed. In the course of screening, we identified IEJLs as potent osteoclast inhibitors. Since it is known that osteoclasts are sensitive to vacuolar H(+)-ATPase (V-ATPase) inhibitor, we investigated the effect of IEJLs on V-ATPases. IEJLs inhibited the V-ATPases of both mammalian and yeast cells in situ, and of yeast V-ATPases in vitro. A bafilomycin-resistant yeast mutant conferred IEJL resistance, suggesting that IEJLs bind a site similar to the bafilomycins/concanamycins-binding site. These results indicate that IEJLs are novel V-ATPase inhibitors, and that antitumor and antiosteporotic activities are exerted via V-ATPase inhibition.
Collapse
Affiliation(s)
- Sayaka Kazami
- Antibiotics Laboratory, RIKEN Discovery Research Institute, Hirosawa, Saitama
| | | | | | | | | | | | | |
Collapse
|
18
|
Katara GK, Jaiswal MK, Kulshrestha A, Kolli B, Gilman-Sachs A, Beaman KD. Tumor-associated vacuolar ATPase subunit promotes tumorigenic characteristics in macrophages. Oncogene 2013; 33:5649-54. [DOI: 10.1038/onc.2013.532] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Revised: 10/21/2013] [Accepted: 11/06/2013] [Indexed: 01/01/2023]
|
19
|
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
|
20
|
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
|
21
|
Huang S, Chen M, Ding X, Zhang X, Zou X. Proton pump inhibitor selectively suppresses proliferation and restores the chemosensitivity of gastric cancer cells by inhibiting STAT3 signaling pathway. Int Immunopharmacol 2013; 17:585-92. [PMID: 23973653 DOI: 10.1016/j.intimp.2013.07.021] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2013] [Revised: 07/23/2013] [Accepted: 07/30/2013] [Indexed: 01/05/2023]
Abstract
BACKGROUNDS AND AIMS Recent studies reported that pretreatment of proton pump inhibitors (PPIs) induced sensitization to chemotherapeutic agents in several cancer cells. The devastating effects of PPIs on tumor cells were discovered and raised great interests; therefore we designed the following experiments to fully explain the direct antitumor effects of PPIs. METHODS We compared the viability of gastric cancer cells and epithelia cells in buffered and unbuffered culture conditions with PPZ treatment by WST-8 assay. The sensitivity to cisplatin of gastric cancer cells with/without PPZ pretreatment was assessed by IC50 calculation and Annexin V/PI assay. The secretion of IL-6 was detected by ELISA. Western blot analysis and real time RT-PCR were used to evaluate the expression and activation of STAT3 and its downstream targets. RESULTS PPZ selectively exhibited a dose-dependent cytotoxic effect of gastric cancer cells in acidic unbuffered medium. Low dose of PPZ pretreatment (20 μg/mL) enhanced the sensitivity to cisplatin in gastric cancer cells. PPZ induced cell apoptosis and reduced the secretion of the pro-inflammatory cytokine IL-6 specifically in gastric cancer cells, but had no effect on the epithelia cells. Consequently, the activation of STAT3, not the total amount, was suppressed by PPZ in gastric cancer cells. The downstream targets of STAT3, c-Myc, cyclin D1 and Bcl-2 were also down-regulated. CONCLUSION PPZ causes gastric cancer cell death by induction of apoptosis and its mechanism of action is mediated in part via the inhibition of IL-6/STAT3 pathway.
Collapse
Affiliation(s)
- Shuling Huang
- Department of Gastroenterology, Drum Tower Hospital, Affiliated to Medical School of Nanjing University, No. 22, Hankou Road, Gulou District, Nanjing 210008, China
| | | | | | | | | |
Collapse
|
22
|
Kallunki T, Olsen OD, Jäättelä M. Cancer-associated lysosomal changes: friends or foes? Oncogene 2013; 32:1995-2004. [PMID: 22777359 DOI: 10.1038/onc.2012.292] [Citation(s) in RCA: 213] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2012] [Accepted: 06/01/2012] [Indexed: 12/28/2022]
Abstract
Rapidly dividing and invasive cancer cells are strongly dependent on effective lysosomal function. Accordingly, transformation and cancer progression are characterized by dramatic changes in lysosomal volume, composition and cellular distribution. Depending on one's point of view, the cancer-associated changes in the lysosomal compartment can be regarded as friends or foes. Most of them are clearly transforming as they promote invasive growth, angiogenesis and drug resistance. The same changes can, however, strongly sensitize cells to lysosomal membrane permeabilization and thereby to lysosome-targeting anti-cancer drugs. In this review we compile our current knowledge on cancer-associated changes in lysosomal composition and discuss the consequences of these alterations to cancer progression and the possibilities they can bring to cancer therapy.
Collapse
Affiliation(s)
- T Kallunki
- Cell Death and Metabolism and Centre for Genotoxic Stress Research, Danish Cancer Society Research Center, Copenhagen, Denmark
| | | | | |
Collapse
|
23
|
Kulkarni H, Göring HHH, Curran JE, Diego V, Dyer TD, Cole S, Walder KR, Collier GR, Blangero J, Carless MA. Genetic basis for the increased expression of vacuolar H+ translocating ATPase genes upon imatinib treatment in human lymphoblastoid cells. Cancer Chemother Pharmacol 2013; 71:1095-100. [PMID: 23420437 DOI: 10.1007/s00280-013-2110-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2012] [Accepted: 02/05/2013] [Indexed: 12/01/2022]
Abstract
PURPOSE The role of v-ATPases in cancer biology is being increasingly recognized. Yeast studies indicate that the tyrosine kinase inhibitor imatinib may interact with the v-ATPase genes and alter the course of cancer progression. Data from humans in this regard are lacking. METHODS We constructed 55 lymphoblastoid cell lines from pedigreed, cancer-free human subjects and treated them with IC20 concentration of imatinib mesylate. Using these cell lines, we (i) estimated the heritability and differential expression of 19 genes encoding several subunits of the v-ATPase protein in response to imatinib treatment; (ii) estimated the genetic similarity among these genes; and (iii) conducted a high-density scan to find cis-regulating genetic variation associated with differential expression of these genes. RESULTS We found that the imatinib response of the genes encoding v-ATPase subunits is significantly heritable and can be clustered to identify novel drug targets in imatinib therapy. Further, five of these genes were significantly cis-regulated and together represented nearly half-log fold change in response to imatinib (p = 0.0107) that was homogenous (p = 0.2598). CONCLUSIONS Our results proffer support to the growing view that personalized regimens using proton pump inhibitors or v-ATPase inhibitors may improve outcomes of imatinib therapy in various cancers.
Collapse
Affiliation(s)
- Hemant Kulkarni
- Department of Genetics, Texas Biomedical Research Institute, San Antonio, TX 78227, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Xu J, Xie R, Liu X, Wen G, Jin H, Yu Z, Jiang Y, Zhao Z, Yang Y, Ji B, Dong H, Tuo B. Expression and functional role of vacuolar H(+)-ATPase in human hepatocellular carcinoma. Carcinogenesis 2012; 33:2432-40. [PMID: 22962303 DOI: 10.1093/carcin/bgs277] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Tumor cells often exist in a hypoxic microenvironment, which produces acidic metabolites. To survive in this harsh environment, tumor cells must exhibit a dynamic cytosolic pH regulatory system. Vacuolar H(+)-adenosine triphosphatase (V-ATPase) is considered to play an important role in the regulation of the acidic microenvironment of some tumors. In this study, we made an investigation on the expression and functional role of V-ATPase in native human hepatocellular carcinoma (HCC). The results showed that the messenger RNA and protein expression levels of V-ATPase subunit ATP6L in native human HCC tissues were markedly increased, compared with normal liver tissues. Immunohistochemical analysis further confirmed the enhanced expression of V-ATPase ATP6L in human HCC cells and revealed that V-ATPase ATP6L was distributed in the cytoplasm and plasma membrane of HCC cells. The results from immunofluorescence and biotinylation of cell surface protein showed that V-ATPase ATP6L was conspicuously located in the plasma membrane of human HCC cells. Bafilomycin A1, a specific V-ATPase inhibitor, markedly slowed the intracellular pH (pHi) recovery after acid load in human HCC cells and retarded the growth of human HCC in orthotopic xenograft model. These results demonstrated that V-ATPase is up-regulated in human HCC and involved in the regulation of pHi of human HCC cells. The inhibition of V-ATPase can effectively retard the growth of HCC, indicating that V-ATPase may play an important role in the development and progression of human HCC, and targeting V-ATPase may be a promising therapeutic strategy against human HCC.
Collapse
Affiliation(s)
- Jingyu Xu
- Department of Gastroenterology, Affiliated Hospital, Zunyi Medical College, Zunyi 563003, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
LASS2 enhances chemosensitivity of breast cancer by counteracting acidic tumor microenvironment through inhibiting activity of V-ATPase proton pump. Oncogene 2012; 32:1682-90. [PMID: 22580606 DOI: 10.1038/onc.2012.183] [Citation(s) in RCA: 77] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
A main obstacle to overcome during the treatment of tumors is drug resistance to chemotherapy; emerging studies indicate that a key factor contributing to this problem is the acidic tumor microenvironment. Here, we found that LASS2 expression was significantly lower in drug-resistant Michigan Cancer Foundation-7/adriamycin (MCF-7/ADR) human breast cancer cells than the drug-sensitive MCF-7 cells, and low expression of LASS2 was associated with poor prognosis in patients with breast cancer. Our results showed that the overexpression of LASS2 in MCF-7/ADR cells increased the chemosensitivity to multiple chemotherapeutic agents, including doxorubicin (Dox), whereas LASS2 knockdown in MCF-7 cells decreased the chemosensitivity. Cell-cycle analysis revealed a corresponding increase in apoptosis in the LASS2-overexpressing cells following Dox exposure, showing that the overexpression of LASS2 increased the susceptibility to Dox cytotoxicity. This effect was mediated by a significant increase in pHe (extracellular pH) and lysosomal pH, and more Dox entered the cells and stayed in the nuclei of cells. In nude mice, the combination of LASS2 overexpression and Dox significantly inhibited the growth of xenografts. Our findings suggest that LASS2 is involved in chemotherapeutic outcomes and low LASS2 expression may predict chemoresistance.
Collapse
|
26
|
Spugnini EP, Baldi A, Buglioni S, Carocci F, de Bazzichini GM, Betti G, Pantaleo I, Menicagli F, Citro G, Fais S. Lansoprazole as a rescue agent in chemoresistant tumors: a phase I/II study in companion animals with spontaneously occurring tumors. J Transl Med 2011; 9:221. [PMID: 22204495 PMCID: PMC3264547 DOI: 10.1186/1479-5876-9-221] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2011] [Accepted: 12/28/2011] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND The treatment of human cancer has been seriously hampered for decades by resistance to chemotherapeutic drugs. Mechanisms underlying this resistance are far from being entirely known. A very efficient mechanism of tumor resistance to drugs is related to the modification of tumour microenvironment through changes in the extracellular and intracellular pH. The acidification of tumor microenvironment depends on proton pumps that actively pump protons outside the cells, mostly to avoid intracellular acidification. In fact, we have shown in pre-clinical settings as pre-treatment with proton-pumps inhibitors (PPI) increase tumor cell and tumor responsiveness to chemotherapeutics. In this study pet with spontaneously occurring cancer proven refractory to conventional chemotherapy have been recruited in a compassionate study. METHODS Thirty-four companion animals (27 dogs and 7 cats) were treated adding to their chemotherapy protocols the pump inhibitor lansoprazole at high dose, as suggested by pre-clinical experiments. Their responses have been compared to those of seventeen pets (10 dogs and 7 cats) whose owners did not pursue any other therapy than continuing the currently ongoing chemotherapy protocols. RESULTS The drug was overall well tolerated, with only four dogs experiencing side effects due to gastric hypochlorhydria consisting with vomiting and or diarrhea. In terms of overall response twenty-three pets out of 34 had partial or complete responses (67.6%) the remaining patients experienced no response or progressive disease however most owners reported improved quality of life in most of the non responders. On the other hand, only three animals in the control group (17%) experienced short lived partial responses (1-3 months duration) while all the others died of progressive disease within two months. CONCLUSIONS high dose proton pump inhibitors have been shown to induce reversal of tumor chemoresistance as well as improvement of the quality of life in pets with down staged cancer and in the majority of the treated animals PPI were well tolerated. Further studies are warranted to assess the efficacy of this strategy in patients with advanced cancers in companion animals as well as in humans.
Collapse
|
27
|
Chung C, Mader CC, Schmitz J, Atladottir J, Fitchev P, Cornwell M, Koleske AJ, Crawford SE, Gorelick F. The vacuolar-ATPase modulates matrix metalloproteinase isoforms in human pancreatic cancer. J Transl Med 2011; 91:732-43. [PMID: 21339745 PMCID: PMC3084324 DOI: 10.1038/labinvest.2011.8] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
The vacuolar-ATPase (v-ATPase) is a proton transporter found on many intracellular organelles and the plasma membrane (PM). The v-ATPase on PMs of cancer cells may contribute to their invasive properties in vitro. Its relevance to human cancer tissues remains unclear. We investigated whether the expression and cellular localization of v-ATPase corresponded to the stage of human pancreatic cancer, and its effect on matrix metalloproteinase (MMP) activation in vitro. The intensity of v-ATPase staining increased significantly across the range of pancreatic histology from normal ducts to pancreatic intraepithelial neoplasms (PanIN), and finally pancreatic ductal adenocarcinoma (PDAC). Low-grade PanIN lesions displayed polarized staining confined to the basal aspect of the cell in the majority (86%) of fields examined. High-grade PanIN lesions and PDAC showed intense and diffuse v-ATPase localization. In pancreatic cancer cells, PM-associated v-ATPase colocalized with cortactin, a component of the leading edge that helps direct MMP release. Blockade of the v-ATPase with concanamycin or short-hairpin RNA targeting the V₁E subunit reduced MMP-9 activity; this effect was greatest in cells with prominent PM-associated v-ATPase. In cells with detectable MMP-2 activities, however, treatment with concanamycin markedly increased MMP-2's most activated forms. V-ATPase blockade inhibited functional migration and invasion in those cells with predominantly MMP-9 activity. These results indicate that human PDAC specimens show loss of v-ATPase polarity and increased expression that correlates with increasing invasive potential. Thus, v-ATPase selectively modulates specific MMPs that may be linked to an invasive cancer phenotype.
Collapse
Affiliation(s)
- Chuhan Chung
- Department of Medicine, Section of Digestive Diseases, VA CT Research, VA CT Healthcare System, Yale University School of Medicine, West Haven, CT 06516, USA.
| | | | | | | | - Phillip Fitchev
- Department of Surgery, NorthShore Research Institute, University of Chicago Pritzker School of Medicine
| | - Mona Cornwell
- Department of Surgery, NorthShore Research Institute, University of Chicago Pritzker School of Medicine
| | | | - Susan E Crawford
- Department of Surgery, NorthShore Research Institute, University of Chicago Pritzker School of Medicine
| | - Fred Gorelick
- Section of Digestive Diseases, Department of Medicine, VA CT Healthcare System, Yale University School of Medicine
| |
Collapse
|
28
|
Pérez-Sayáns M, Somoza-Martín JM, Barros-Angueira F, Diz PG, Rey JMG, García-García A. Multidrug resistance in oral squamous cell carcinoma: The role of vacuolar ATPases. Cancer Lett 2010; 295:135-43. [DOI: 10.1016/j.canlet.2010.03.019] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2009] [Revised: 03/23/2010] [Accepted: 03/25/2010] [Indexed: 10/19/2022]
|
29
|
Spugnini EP, Citro G, Fais S. Proton pump inhibitors as anti vacuolar-ATPases drugs: a novel anticancer strategy. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2010; 29:44. [PMID: 20459683 PMCID: PMC2876100 DOI: 10.1186/1756-9966-29-44] [Citation(s) in RCA: 89] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/03/2010] [Accepted: 05/08/2010] [Indexed: 12/25/2022]
Abstract
The vacuolar ATPases are ATP-dependent proton pumps whose functions include the acidification of intracellular compartments and the extrusion of protons through the cell cytoplasmic membrane. These pumps play a pivotal role in the regulation of cell pH in normal cells and, to a much greater extent, in tumor cells. In fact, the glucose metabolism in hypoxic conditions by the neoplasms leads to an intercellular pH drift towards acidity. The acid microenvironment is modulated through the over-expression of H+ transporters that are also involved in tumor progression, invasiveness, distant spread and chemoresistance. Several strategies to block/downmodulate the efficiency of these transporters are currently being investigated. Among them, proton pump inhibitors have shown to successfully block the H+ transporters in vitro and in vivo, leading to apoptotic death. Furthermore, their action seems to synergize with conventional chemotherapy protocols, leading to chemosensitization and reversal of chemoresistance. Aim of this article is to critically revise the current knowledge of this cellular machinery and to summarize the therapeutic strategies developed to counter this mechanism.
Collapse
|
30
|
Abstract
This review presents a possible new approach against cancer, as represented by inhibition of proton pumps, a mechanism used by tumour cells to avoid intracellular accumulation of toxic substances. Proton pump inhibitors (PPIs) belong to a family of pro-drugs that are currently used in the treatment of peptic diseases needing acidity to be activated. PPIs target the acidic tumour mass, where they are metabolized, thus blocking proton traffic. Proton pump inhibition triggers a rapid cell death as a result of intracellular acidification, caspase activation and early accumulation of reactive oxygen species into tumour cells. As a whole, the devastating effect of PPIs on tumour cells suggest the triggering of a fatal cell toxification. Many human tumours, including melanoma, osteosarcoma, lymphomas and various adenocarcinomas are responsive to PPIs. This appears highly conceivable, in as much as almost all human tumours are acidic and express high levels of proton pumps. Paradoxically, metastatic tumours appear to be more responsive to PPIs being more acidic than the majority of primary tumours. However, two clinical trials test the effectiveness of PPIs in chemosensitizing melanoma and osteosarcoma patients. Indeed, tumour acidity represents a very potent mechanism of chemoresistance. A majority of cytotoxic agents, being weak bases, are quickly protonated outside and do not enter the cells, thus preventing drugs to reach specific cellular targets. Clinical data will provide the proof of concept on the use of PPIs as a new class of antitumour agent with a very low level of systemic toxicity as compared with standard chemotherapeutic agents.
Collapse
Affiliation(s)
- S Fais
- Anti-Tumour Drugs Section, Department of Therapeutic Research and Medicines Evaluation, Istituo Superiore di Sanità (National Institute of Health), Rome, Italy.
| |
Collapse
|
31
|
Pérez-Sayáns M, Somoza-Martín JM, Barros-Angueira F, Rey JMG, García-García A. V-ATPase inhibitors and implication in cancer treatment. Cancer Treat Rev 2009; 35:707-13. [PMID: 19758758 DOI: 10.1016/j.ctrv.2009.08.003] [Citation(s) in RCA: 144] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2009] [Revised: 08/03/2009] [Accepted: 08/06/2009] [Indexed: 01/08/2023]
Abstract
Acidity is one of the main features of the tumors. The V-ATPase is the primary responsible for the control of tumor microenvironment by proton extrusion to the extracellular medium. The acid environment favors tissue damage, activation of destructive enzymes in the extracellular matrix, the acquisition of metastatic cell phenotypes as well as increasing the destructive capacity. The application of specific inhibitors of V-ATPases, can decrease the acidity of tumor and may allow the reduction of tumor metastasis, acting on the survival of tumor cells and prevent the phenomena of chemoresistance. Among the most important inhibitors can be distinguished benzolactone enamides (salicylihalamide), lobatamide A and B, apicularen, indolyls, oximidine, macrolactone archazolid, lobatamide C, and cruentaren. The latest generation of inhibitors includes NiK12192, FR202126, and PPI SB 242784. The purpose of this paper is to describe the latest advances in the field of V-ATPase inhibitors, describe further developments related to the classic inhibitors, and discuss new potential applications of these drugs in cancer treatment.
Collapse
|
32
|
dos Santos SC, Sá-Correia I. Genome-Wide Identification of Genes Required for Yeast Growth Under Imatinib Stress: Vacuolar H+-ATPase Function Is an Important Target of This Anticancer Drug. OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2009; 13:185-98. [DOI: 10.1089/omi.2008.0086] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Affiliation(s)
- Sandra C. dos Santos
- IBB—Institute for Biotechnology and Bioengineering, Centre for Biological and Chemical Engineering, Instituto Superior Técnico, Lisboa, Portugal
| | - Isabel Sá-Correia
- IBB—Institute for Biotechnology and Bioengineering, Centre for Biological and Chemical Engineering, Instituto Superior Técnico, Lisboa, Portugal
| |
Collapse
|
33
|
Hinton A, Sennoune SR, Bond S, Fang M, Reuveni M, Sahagian GG, Jay D, Martinez-Zaguilan R, Forgac M. Function of a subunit isoforms of the V-ATPase in pH homeostasis and in vitro invasion of MDA-MB231 human breast cancer cells. J Biol Chem 2009; 284:16400-16408. [PMID: 19366680 DOI: 10.1074/jbc.m901201200] [Citation(s) in RCA: 151] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
It has previously been shown that highly invasive MDA-MB231 human breast cancer cells express vacuolar proton-translocating ATPase (V-ATPases) at the cell surface, whereas the poorly invasive MCF7 cell line does not. Bafilomycin, a specific V-ATPase inhibitor, reduces the in vitro invasion of MB231 cells but not MCF7 cells. Targeting of V-ATPases to different cellular membranes is controlled by isoforms of subunit a. mRNA levels for a subunit isoforms were measured in MB231 and MCF7 cells using quantitative reverse transcription-PCR. The results show that although all four isoforms are detectable in both cell types, levels of a3 and a4 are much higher in MB231 than in MCF7 cells. Isoform-specific small interfering RNAs (siRNA) were employed to selectively reduce mRNA levels for each isoform in MB231 cells. V-ATPase function was assessed using the fluorescent indicators SNARF-1 and pyranine to monitor the pH of the cytosol and endosomal/lysosomal compartments, respectively. Cytosolic pH was decreased only on knockdown of a3, whereas endosome/lysosome pH was increased on knockdown of a1, a2, and a3. Treatment of cells with siRNA to a4 did not affect either cytosolic or endosome/lysosome pH. Measurement of invasion using an in vitro transwell assay revealed that siRNAs to both a3 and a4 significantly inhibited invasion of MB231 cells. Immunofluorescence staining of MB231 cells for V-ATPase distribution revealed extensive intracellular staining, with plasma membrane staining observed in approximately 18% of cells. Knockdown of a4 had the greatest effect on plasma membrane staining, leading to a 32% reduction. These results suggest that the a4 isoform may be responsible for targeting V-ATPases to the plasma membrane of MB231 cells and that cell surface V-ATPases play a significant role in invasion. However, other V-ATPases affecting the pH of the cytosol and intracellular compartments, particularly those containing a3, are also involved in invasion.
Collapse
Affiliation(s)
- Ayana Hinton
- From the Department of Physiology, Tufts University School of Medicine, Boston, Massachusetts 02111
| | - Souad R Sennoune
- Department of Cell Physiology and Molecular Biophysics, Texas Tech University, Lubbock, Texas 79430
| | - Sarah Bond
- From the Department of Physiology, Tufts University School of Medicine, Boston, Massachusetts 02111
| | - Min Fang
- From the Department of Physiology, Tufts University School of Medicine, Boston, Massachusetts 02111
| | - Moshe Reuveni
- Department of Ornamental Horticulture, ARO Volcani Center, Bet Dagan 50250, Israel
| | - G Gary Sahagian
- From the Department of Physiology, Tufts University School of Medicine, Boston, Massachusetts 02111
| | - Daniel Jay
- From the Department of Physiology, Tufts University School of Medicine, Boston, Massachusetts 02111
| | - Raul Martinez-Zaguilan
- Department of Cell Physiology and Molecular Biophysics, Texas Tech University, Lubbock, Texas 79430
| | - Michael Forgac
- From the Department of Physiology, Tufts University School of Medicine, Boston, Massachusetts 02111.
| |
Collapse
|
34
|
Small interfering RNA targeting the subunit ATP6L of proton pump V-ATPase overcomes chemoresistance of breast cancer cells. Cancer Lett 2009; 280:110-9. [PMID: 19299075 DOI: 10.1016/j.canlet.2009.02.023] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2008] [Revised: 02/05/2009] [Accepted: 02/09/2009] [Indexed: 11/21/2022]
Abstract
One of the mechanisms of multiple drug resistance (MDR) is inappropriate sequestration of basic chemotherapeutic agents in acidic endo-lysosomes of cells. The protonation, sequestration, and secretion (PSS) model indicates that drug distribution can be affected by intracellular pH such as lysosomal pH. The vacuolar-H(+)-ATPase (V-ATPase) plays an important role in regulation of intracellular pH by pumping protons into acidic endosomes via an ATP-driven process. In this study, ATP6L, the 16kDa subunit of V-ATPase, was knocked-down by anti-ATP6L small interfering RNA (siRNA) to study the effect on chemosensitivity in the human drug-resistant breast cancer cells MCF-7/ADR. Introduction of anti-ATP6L small interfering RNA duplex into drug-resistant cancer cells significantly inhibited the expression of ATP6L mRNA and protein, as detected by qRT-PCR and Western blot. Inhibition of ATP6L expression by siRNA in MCF-7/ADR sensitized the cells to the cytotoxicity of basic chemotherapeutic agents like doxorobicin, 5-fluorourocil and vincristine. This effect was mediated by a significant increase in lysosomal pH and retention of anticancer drugs into nuclei of cells. These results support the role of tumor acidity in resistance to chemotherapy and provide a rationale for the use of tumor pH modifier agents as coadjuvants in novel anticancer therapies.
Collapse
|
35
|
Guillard M, Dimopoulou A, Fischer B, Morava E, Lefeber DJ, Kornak U, Wevers RA. Vacuolar H+-ATPase meets glycosylation in patients with cutis laxa. Biochim Biophys Acta Mol Basis Dis 2009; 1792:903-14. [PMID: 19171192 DOI: 10.1016/j.bbadis.2008.12.009] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2008] [Revised: 12/22/2008] [Accepted: 12/29/2008] [Indexed: 02/08/2023]
Abstract
Glycosylation of proteins is one of the most important post-translational modifications. Defects in the glycan biosynthesis result in congenital malformation syndromes, also known as congenital disorders of glycosylation (CDG). Based on the iso-electric focusing patterns of plasma transferrin and apolipoprotein C-III a combined defect in N- and O-glycosylation was identified in patients with autosomal recessive cutis laxa type II (ARCL II). Disease-causing mutations were identified in the ATP6V0A2 gene, encoding the a2 subunit of the vacuolar H(+)-ATPase (V-ATPase). The V-ATPases are multi-subunit, ATP-dependent proton pumps located in membranes of cells and organels. In this article, we describe the structure, function and regulation of the V-ATPase and the phenotypes currently known to result from V-ATPase mutations. A clinical overview of cutis laxa syndromes is presented with a focus on ARCL II. Finally, the relationship between ATP6V0A2 mutations, the glycosylation defect and the ARCLII phenotype is discussed.
Collapse
Affiliation(s)
- Mailys Guillard
- Laboratory of Pediatrics and Neurology, Radboud University Nijmegen Medical Centre, PO Box 9101, 6500 HB, Nijmegen, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
36
|
Sennoune SR, Martinez-Zaguilan R. Plasmalemmal vacuolar H+-ATPases in angiogenesis, diabetes and cancer. J Bioenerg Biomembr 2008; 39:427-33. [PMID: 18058006 DOI: 10.1007/s10863-007-9108-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Angiogenesis, i.e., new blood vessel formation, is required in normal and pathological states. A dysfunction in the microvascular endothelium occurs in diabetes, leading to decreased blood flow and limb amputation. In cancer, angiogenesis is increased to allow for growth, invasion, and metastasis of tumor cells. Better understanding of the molecular events that cause or are associated with either of these diseases is needed to develop therapies. The tumor and angiogenic cells micro-environment is acidic and not permissive for growth. We have shown that to survive this environment, highly metastatic and angiogenic cells employ vacuolar H+-ATPase at their plasma membranes (pmV-ATPases) to maintain an alkaline pHcyt. However, in lowly metastatic and in microvascular endothelial cells from diabetic model, the density of pmV-ATPase and the cell invasiveness are decreased. Therefore, the overexpression of the pmV-ATPase is important for cell invasion, and essential for tumor progression, angiogenesis and metastasis. Both, cancer and diabetes are heterogenous diseases that involve many different proteins and signaling pathways. Changes in pHcyt have been associated with the regulation of a myriad of proteins, signaling molecules and pathways affecting many if not all cellular functions. Since changes in pHcyt are pleiotropic, we hypothesize that alteration in a single protein, pmV-ATPase, that can regulate pHcyt may explain the dysfunction of many proteins and cellular pathways in diabetes and cancer. Our long term goal is to determine the molecular mechanisms by which pmV-ATPase expression regulates tumor angiogenesis and metastasis. Such knowledge would be useful to identify targets for cancer therapy.
Collapse
Affiliation(s)
- Souad R Sennoune
- Department of Cellular Physiology and Molecular Biophysics, Health Sciences Center, Texas Tech University, 3601 4th Street, Lubbock, TX 79430-6551, USA.
| | | |
Collapse
|
37
|
Fais S, De Milito A, You H, Qin W. Targeting vacuolar H+-ATPases as a new strategy against cancer. Cancer Res 2007; 67:10627-30. [PMID: 18006801 DOI: 10.1158/0008-5472.can-07-1805] [Citation(s) in RCA: 194] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Growing evidence suggests a key role of tumor acidic microenvironment in cancer development, progression, and metastasis. As a consequence, the need for compounds that specifically target the mechanism(s) responsible for the low pH of tumors is increasing. Among the key regulators of the tumor acidic microenvironment, vacuolar H(+)-ATPases (V-ATPases) play an important role. These proteins cover a number of functions in a variety of normal as well as tumor cells, in which they pump ions across the membranes. We discuss here some recent results showing that a molecular inhibition of V-ATPases by small interfering RNA in vivo as well as a pharmacologic inhibition through proton pump inhibitors led to tumor cytotoxicity and marked inhibition of human tumor growth in xenograft models. These results propose V-ATPases as a key target for new strategies in cancer treatment.
Collapse
Affiliation(s)
- Stefano Fais
- Department of Drug Research and Evaluation, Istituto Superiore di Sanità, Rome, Italy.
| | | | | | | |
Collapse
|
38
|
V-ATPase functions in normal and disease processes. Pflugers Arch 2007; 457:589-98. [DOI: 10.1007/s00424-007-0382-4] [Citation(s) in RCA: 152] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2007] [Accepted: 10/26/2007] [Indexed: 01/30/2023]
|
39
|
Byun YJ, Lee SB, Kim DJ, Lee HO, Son MJ, Yang CW, Sung KW, Kim HS, Kwon OJ, Kim IK, Jeong SW. Protective effects of vacuolar H+-ATPase c on hydrogen peroxide-induced cell death in C6 glioma cells. Neurosci Lett 2007; 425:183-7. [PMID: 17845832 DOI: 10.1016/j.neulet.2007.08.027] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2007] [Revised: 08/14/2007] [Accepted: 08/14/2007] [Indexed: 01/27/2023]
Abstract
We have isolated a gene, the c subunit (ATP6L) of vacuolar H(+)-ATPase, involved in oxidative stress response. In this study, we examined the role of ATP6L and its molecular mechanisms in glial cell death induced by H(2)O(2). Expression of the ATP6L gene was increased by H(2)O(2) treatment in C6 glial cells. ATP6L siRNA-transfected C6 cells treated with H(2)O(2) showed a significant decrease in viability. ATP6L siRNA-transfected cells that were pretreated with MEK1/2 inhibitor completely recovered cell viability. Pretreatment of the transfected cells with zVAD-fmk, a pan-specific caspase inhibitor, did not result in the recovery of cell viability, as determined by a H(2)O(2)-induced cytotoxicity assay. The ultrastructural morphology of the transfected cells as seen by the use of transmission electron microscopy showed numerous cytoplasmic autophagic vacuoles with double membrane. These results suggest that ATP6L has a protective role against H(2)O(2)-induced cytotoxicity via an inhibition of the Erk1/2 signaling pathway, leading to inhibition of autophagic cell death.
Collapse
Affiliation(s)
- Yu Jeong Byun
- Department of Biochemistry, College of Medicine, The Catholic University of Korea, 505 Banpo-dong, Socho-gu, Seoul 137-701, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Wang L, Sun J, Li YQ, Chen ZS, Akiyama SI, Xian LJ. Reversal effect of BM-cyclin 1 on multidrug resistance in C-A120 cells. Anticancer Drugs 2007; 18:1015-21. [PMID: 17704651 DOI: 10.1097/cad.0b013e328223f14d] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
In this study, multidrug-resistant human epidermoid C-A120 cells and the sensitive parental KB cells were used as experimental models. BM-cyclin 1, a traditional antimycoplasma drug, was tested to explore the reversal effect of multidrug resistance and its mechanisms in these cell lines. The MTT analysis showed that BM-cyclin 1 could reverse multidrug resistance effectively in C-A120 cells; the sensitivity of C-A120 cells to adriamycin, etoposide and cisplatin was enhanced by 6.0, 8.2 and 1.7 times, respectively. Immunoblotting analysis and reverse transcription-polymerase chain reaction were used to study the BM-cyclin 1-induced changes in topoisomerase IIalpha. The results showed that the expression of topoisomerase IIalpha in treated C-A120 cells increased significantly. Topoisomerase II catalytic activity increased by 30% compared with the untreated cells, as measured by decatenation of kinetopolast DNA. Immunoblotting analysis also indicated the transcription factor levels of specificity: those of protein 1 (Sp1) and nuclear factor-YA increased after treatment with BM-cyclin 1, whereas the mRNA and protein expression of multidrug resistance protein 2 was significantly downregulated. These results demonstrated that BM-cyclin 1 could effectively reverse the multidrug resistance of C-A120 cells by increasing the expression of topoisomerase IIalpha and by suppressing the expression of multidrug resistance protein 2, strongly suggesting that BM-cyclin 1 is a potential multidrug resistance reversal agent.
Collapse
Affiliation(s)
- Lin Wang
- State Key Laboratory of Oncology in Southern China
| | | | | | | | | | | |
Collapse
|
41
|
Yao G, Feng H, Cai Y, Qi W, Kong K. Characterization of vacuolar-ATPase and selective inhibition of vacuolar-H(+)-ATPase in osteoclasts. Biochem Biophys Res Commun 2007; 357:821-7. [PMID: 17462591 DOI: 10.1016/j.bbrc.2007.04.082] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2007] [Accepted: 04/07/2007] [Indexed: 02/05/2023]
Abstract
V-ATPase plays important roles in controlling the extra- and intra-cellular pH in eukaryotic cell, which is most crucial for cellular processes. V-ATPases are composed of a peripheral V(1) domain responsible for ATP hydrolysis and integral V(0) domain responsible for proton translocation. Osteoclasts are multinucleated cells responsible for bone resorption and relate to many common lytic bone disorders such as osteoporosis, bone aseptic loosening, and tumor-induced bone loss. This review summarizes the structure and function of V-ATPase and its subunit, the role of V-ATPase subunits in osteoclast function, V-ATPase inhibitors for osteoclast function, and highlights the importance of V-ATPase as a potential prime target for anti-resorptive agents.
Collapse
Affiliation(s)
- GuanFeng Yao
- Department of Orthopedics, The Second Affiliated Hospital, ShanTou University Medical College, ShanTou, GuangDong 515041, China
| | | | | | | | | |
Collapse
|
42
|
Niina I, Uchiumi T, Izumi H, Torigoe T, Wakasugi T, Igarashi T, Miyamoto N, Onitsuka T, Shiota M, Okayasu R, Chijiiwa K, Kohno K. DNA topoisomerase inhibitor, etoposide, enhances GC-box-dependent promoter activity via Sp1 phosphorylation. Cancer Sci 2007; 98:858-63. [PMID: 17441964 PMCID: PMC11158573 DOI: 10.1111/j.1349-7006.2007.00476.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Modification of transcription factors by anticancer agents plays an important role in both apoptotic and survival signaling. Here we report that both DNA topoisomerase I and II inhibitors such as SN-38 and etoposide, but not cisplatin, 5-fluorouracil or actinomycin D, can induce phosphorylation of the transcription factor Sp1. Furthermore, DNA topoisomerase inhibitors were shown to transactivate GC-box-dependent promoters such as the SV40 and vascular endothelial growth factor promoters. The phosphorylated form of Sp1 was detectable within 30 min of etoposide treatment and was greatly diminished by the presence of the PI3K inhibitor wortmannin and by DNA-dependent protein kinase (DNA-PK) knockdown. We also confirmed that the phosphorylated form of DNA-PK was increased by treatment with both etoposide and SN-38. Taken together, these findings demonstrate a novel genomic response to anticancer agents that induce Sp1 phosphorylation, and might contribute to tumor progression and drug resistance.
Collapse
Affiliation(s)
- Ichiro Niina
- Department of Molecular Biology, School of Medicine, University of Occupational and Environmental Health, Iseigaoka, Fukuoka, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Andley UP, Patel HC, XI JH, Bai F. Identification of Genes Responsive to UV-A Radiation in Human Lens Epithelial Cells Using Complementary DNA Microarrays¶. Photochem Photobiol 2007. [DOI: 10.1111/j.1751-1097.2004.tb00050.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
44
|
Liao C, Hu B, Arno MJ, Panaretou B. Genomic screening in vivo reveals the role played by vacuolar H+ ATPase and cytosolic acidification in sensitivity to DNA-damaging agents such as cisplatin. Mol Pharmacol 2006; 71:416-25. [PMID: 17093137 DOI: 10.1124/mol.106.030494] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Screening the Saccharomyces cerevisiae homozygous diploid deletion library against a sublethal concentration of cisplatin revealed 76 strains sensitive to the drug. As expected, the largest category of deletions, representing 40% of the sensitive strains, was composed of strains lacking genes involved in DNA replication and damage repair. Deletions lacking function of the highly conserved vacuolar H+ translocating ATPase (V-ATPase) composed the category representing the second largest number of sensitive strains. The effect on cell death exhibited by V-ATPase mutants was found to be a general response to various DNA damaging agents as opposed to being specific to cisplatin, as evidenced by sensitivity of the mutants to hydroxyurea (a DNA-alkylating agent) and UV irradiation. Loss of V-ATPase does not affect DNA repair, because double mutants defective for V-ATPase function and DNA repair pathways were more sensitive to cisplatin than the single mutants. V-ATPase mutants are more prone to DNA damage than wild-type cells, indicated by enhanced activation of the DNA damage checkpoint. Vacuole function per se is not cisplatin-sensitive, because vacuolar morphology and vacuolar acidification were unaffected by cisplatin in wild-type cells. V-ATPase also controls cytoplasmic pH, so the enhanced sensitivity to DNA damage may be associated with the drop in pHi associated with V-ATPase mutants. The increased loss in cell viability induced by cisplatin at lower pH in V-ATPase mutants supports this hypothesis. The loss in viability seen in wild-type cells under the same conditions was far less dramatic.
Collapse
Affiliation(s)
- Chunyan Liao
- Pharmaceutical Science Research Division, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, UK
| | | | | | | |
Collapse
|
45
|
|
46
|
|
47
|
Huang Y, Sadée W. Membrane transporters and channels in chemoresistance and -sensitivity of tumor cells. Cancer Lett 2006; 239:168-82. [PMID: 16169662 DOI: 10.1016/j.canlet.2005.07.032] [Citation(s) in RCA: 154] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2005] [Accepted: 07/30/2005] [Indexed: 12/14/2022]
Abstract
Membrane transporters play important roles in mediating chemosensitivity and -resistance of tumor cells. ABC transporters, such as ABCB1/MDR1, ABCC1/MRP1 and ABCG2/BCRP, are frequently associated with decreased cellular accumulation of anticancer drugs and multidrug resistance of tumors. SLC transporters, such as folate, nucleoside, and amino acid transporters, commonly increase chemosensitivity by mediating the cellular uptake of hydrophilic drugs. Ion channels and pumps variably affect sensitivity to anticancer therapy by modulating viability of tumor cells. A pharmacogenomic approach, using correlations between drug potency and transporter gene expression in multiple cancer cell lines, has shown promise for identifying potential drug-transporter relationships and predicting anticancer drug response, in an effort to optimize chemotherapy for individual patients.
Collapse
Affiliation(s)
- Ying Huang
- Food and Drug Administration, Division of Pharmacogenomics and Molecular Epidemiology, National Center for Toxicological Research, 3900 NCTR Road, Jefferson, AR 72079, USA.
| | | |
Collapse
|
48
|
Li G, Yang Q, Krishnan S, Alexander EA, Borkan SC, Schwartz JH. A novel cellular survival factor--the B2 subunit of vacuolar H+-ATPase inhibits apoptosis. Cell Death Differ 2006; 13:2109-17. [PMID: 16710359 DOI: 10.1038/sj.cdd.4401970] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
The ubiquitous vacuolar H(+)-ATPase, a multisubunit proton pump, is essential for intraorganellar acidification. Disruption of its function leads to disturbances of organelle function and cell death. Here, we report that overexpression of the B2 subunit of the H(+)-ATPase inhibits apoptosis. This antiapoptotic effect is not mediated by an increase in H(+)-ATPase activity but through activation of the Ras-mitogen-activated protein kinase (MAPK)-signaling pathway that results in the serine phosphorylation of Bad at residues 112 and 155. Increased Bad phosphorylation reduces its translocation to mitochondria, limits the release of mitochondrial cytochrome c and apoptosis-inducing factor and increases the resistance of the B2 overexpressing cells to apoptosis. Screening experiments of kinase inhibitors, including inhibitors of cAMP-activated protein kinase, protein kinase C, protein kinase B, (MAPK/extracellular signal-regulated (ERK) kinase) MEK and Ste-MEK1(13), a cell permeable ERK activation inhibitor peptide, revealed that the B2 subunit of H(+)-ATPase acts upstream of MEK activation in the MEK/ERK pathway to ameliorate apoptosis.
Collapse
Affiliation(s)
- G Li
- Renal Section, Department of Medicine, Boston University Medical Center, Boston University School of Medicine, Boston, MA 02118-2908, USA.
| | | | | | | | | | | |
Collapse
|
49
|
Petrangolini G, Supino R, Pratesi G, Dal Bo L, Tortoreto M, Croce AC, Misiano P, Belfiore P, Farina C, Zunino F. Effect of a Novel Vacuolar-H+-ATPase Inhibitor on Cell and Tumor Response to Camptothecins. J Pharmacol Exp Ther 2006; 318:939-46. [PMID: 16714402 DOI: 10.1124/jpet.106.103481] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
The vacuolar-H(+)-ATPase, functionally expressed in cell membranes, is known to play a relevant role in intracellular pH regulatory mechanisms, because it is implicated in pumping protons into the extracellular environment or in sequestrating excess protons into acidic vacuolar compartments. Because tumor cells exist in a hypoxic microenvironment and produce acidic metabolites, this regulatory mechanism is recognized as a protective function. This study was designed to investigate the effect of NiK-12192 [4-(5,6-dichloro-1H-indol-2-yl)-3-ethoxy-N-(2,2,6,6-tetramethyl-piperidin-4-yl)-benzamide], an indole derivative identified as an effective inhibitor of vacuolar-H(+)-ATPase, on the cytotoxic activity of two camptothecins, i.e., topotecan and SN-38 (7-ethyl-10-hydroxycamptothecin, the active metabolite of irinotecan). The cellular studies performed in two pairs of human colon carcinoma cell lines, i.e., LoVo and LoVo/DX (overexpressing P-glycoprotein) and HT29 and HT29/Mit (overexpressing breast cancer resistant protein), indicated an enhancement of the antiproliferative effect of camptothecins by concomitant exposure to subtoxic concentrations of NiK-12192. Studies of subcellular distribution indicated that whereas topotecan alone localized mainly in mitochondria and endoplasmic compartment, the simultaneous presence of NiK-12192 caused a cytoplasmic redistribution. In HT29/Mit cells, NiK-12192 reverted the pattern of acidification induced by topotecan. The potentiation of topotecan efficacy by NiK-12192 was documented by an increased efficacy of the combination in both the HT29 tumor xenografts, being more evident in the topotecan-resistant HT29/Mit tumor. In conclusion, the vacuolar-H(+)-ATPase inhibitor NiK-12192 was able to potentiate the cytotoxic/antitumor effects of camptothecins, either in in vitro or in in vivo systems. Such findings support a potential interest for the use of vacuolar-H(+)-ATPase inhibitors in combination therapy to improve camptothecin efficacy.
Collapse
|
50
|
Yoshida T, Izumi H, Uchiumi T, Sasaguri Y, Tanimoto A, Matsumoto T, Naito S, Kohno K. Expression and cellular localization of dbpC/Contrin in germ cell tumor cell lines. ACTA ACUST UNITED AC 2006; 1759:80-8. [PMID: 16624424 DOI: 10.1016/j.bbaexp.2006.02.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2005] [Revised: 01/25/2006] [Accepted: 02/27/2006] [Indexed: 12/31/2022]
Abstract
The transcriptional regulation of the germ cell-specific cold-shock domain protein dbpC/Contrin was investigated, and the promoter region between -272 and -253 relative to the transcription start site was shown to be critical for the manifestation of cell-type specific transcription. In vivo footprint analysis demonstrated that the E-box located between -272 and -253 is protected in the dbpC/Contrin-positive germ cell tumor cell lines NEC8 and TERA1, but not in the dbpC/Contrin-negative bladder cancer cell line T24 or ovarian cancer cell line A2780. The promoter activity of the dbpC/Contrin gene was transactivated by co-transfection with c-Myc and the N-Myc expression plasmid. Western blotting analysis clearly showed that N-Myc is highly expressed in both NEC8 and TERA1 cells, and that c-Myc is expressed in both T24 and A2780 cells. These data demonstrate that cell-type specific dbpC/Contrin expression in germ cells is regulated by N-Myc. In addition, dbpC/Contrin is localized mainly in the cytoplasm of NEC8 and TERA1 cells, but is translocated to the nucleus when its C-terminal region is partially deleted. Our findings also suggest that dbpC/Contrin can be used as a molecular tool for the detection of germ cell tumors.
Collapse
Affiliation(s)
- Takeshi Yoshida
- Department of Molecular Biology, University of Occupational and Environmental Health School of Medicine, Iseigaoka, Kitakyushu 807-8555, Japan
| | | | | | | | | | | | | | | |
Collapse
|