1
|
Lv B, Zhang X, Wang Y, Wu W, Li D, Hu Z. Discovery of the Chlorinated and Ammoniated Derivatives of Vanillin as Potential Insecticidal Candidates Targeting V-ATPase: Structure-Based Virtual Screening, Synthesis, and Bioassay. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:20872-20881. [PMID: 39259043 DOI: 10.1021/acs.jafc.4c05174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2024]
Abstract
Vacuolar-type H+-ATPases (V-ATPases) play a crucial role in the life cycle of agricultural pests and represent a promising target for the development of novel insecticides. In this study, S18, a derivative of vanillin acquired from Specs database using a structure-based virtual screening methodology, was first identified as a V-ATPase inhibitor. It binds to subunit A of the enzyme with a Kd of 1 nM and exhibits insecticidal activity against M. separata. Subsequently, using S18 as the lead compound, a new series of vanillin derivatives were rationally designed and efficiently synthesized. and their biological activities were assessed. Among them, compound 3b-03 showed the strongest insecticidal activity against M. separata by effectively targeting the V-ATPase subunit A with Kd of 0.803 μM. Isothermal titration calorimetric measurements and docking results provided insights into its interaction with subunit A of V-ATPase, which could facilitate future research aimed at the development of novel chemical insecticides.
Collapse
Affiliation(s)
- Bo Lv
- Shaanxi Key Laboratory of Botanical Pesticide R & D, Institute of Pesticide Science, Northwest A&F University, Yangling, Shaanxi 712100, PR China
- Key Laboratory of Integrated Pest Management on the Loess Plateau of Ministry of Agriculture and Rural Affairs, Key Laboratory of Plant Protection Resources and Pest Management of Ministry of Education, College of Plant Protection, Northwest A&F University, Yangling, Shaanxi 712100, PR China
| | - Xianxia Zhang
- Shaanxi Key Laboratory of Botanical Pesticide R & D, Institute of Pesticide Science, Northwest A&F University, Yangling, Shaanxi 712100, PR China
- Key Laboratory of Integrated Pest Management on the Loess Plateau of Ministry of Agriculture and Rural Affairs, Key Laboratory of Plant Protection Resources and Pest Management of Ministry of Education, College of Plant Protection, Northwest A&F University, Yangling, Shaanxi 712100, PR China
| | - Yu Wang
- Shaanxi Key Laboratory of Botanical Pesticide R & D, Institute of Pesticide Science, Northwest A&F University, Yangling, Shaanxi 712100, PR China
- Key Laboratory of Integrated Pest Management on the Loess Plateau of Ministry of Agriculture and Rural Affairs, Key Laboratory of Plant Protection Resources and Pest Management of Ministry of Education, College of Plant Protection, Northwest A&F University, Yangling, Shaanxi 712100, PR China
| | - Wenjun Wu
- Shaanxi Key Laboratory of Botanical Pesticide R & D, Institute of Pesticide Science, Northwest A&F University, Yangling, Shaanxi 712100, PR China
| | - Ding Li
- Shaanxi Key Laboratory of Botanical Pesticide R & D, Institute of Pesticide Science, Northwest A&F University, Yangling, Shaanxi 712100, PR China
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, College of Chemistry & Pharmacy, Northwest A&F University, Yangling, Shaanxi 712100, PR China
| | - Zhaonong Hu
- Shaanxi Key Laboratory of Botanical Pesticide R & D, Institute of Pesticide Science, Northwest A&F University, Yangling, Shaanxi 712100, PR China
- Key Laboratory of Integrated Pest Management on the Loess Plateau of Ministry of Agriculture and Rural Affairs, Key Laboratory of Plant Protection Resources and Pest Management of Ministry of Education, College of Plant Protection, Northwest A&F University, Yangling, Shaanxi 712100, PR China
| |
Collapse
|
2
|
Thenuwara G, Javed B, Singh B, Tian F. Biosensor-Enhanced Organ-on-a-Chip Models for Investigating Glioblastoma Tumor Microenvironment Dynamics. SENSORS (BASEL, SWITZERLAND) 2024; 24:2865. [PMID: 38732975 PMCID: PMC11086276 DOI: 10.3390/s24092865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 04/19/2024] [Accepted: 04/27/2024] [Indexed: 05/13/2024]
Abstract
Glioblastoma, an aggressive primary brain tumor, poses a significant challenge owing to its dynamic and intricate tumor microenvironment. This review investigates the innovative integration of biosensor-enhanced organ-on-a-chip (OOC) models as a novel strategy for an in-depth exploration of glioblastoma tumor microenvironment dynamics. In recent years, the transformative approach of incorporating biosensors into OOC platforms has enabled real-time monitoring and analysis of cellular behaviors within a controlled microenvironment. Conventional in vitro and in vivo models exhibit inherent limitations in accurately replicating the complex nature of glioblastoma progression. This review addresses the existing research gap by pioneering the integration of biosensor-enhanced OOC models, providing a comprehensive platform for investigating glioblastoma tumor microenvironment dynamics. The applications of this combined approach in studying glioblastoma dynamics are critically scrutinized, emphasizing its potential to bridge the gap between simplistic models and the intricate in vivo conditions. Furthermore, the article discusses the implications of biosensor-enhanced OOC models in elucidating the dynamic features of the tumor microenvironment, encompassing cell migration, proliferation, and interactions. By furnishing real-time insights, these models significantly contribute to unraveling the complex biology of glioblastoma, thereby influencing the development of more accurate diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Gayathree Thenuwara
- School of Food Science and Environmental Health, Technological University Dublin, Grangegorman Lower, D07 H6K8 Dublin, Ireland; (G.T.); (B.J.)
- Institute of Biochemistry, Molecular Biology, and Biotechnology, University of Colombo, Colombo 00300, Sri Lanka
| | - Bilal Javed
- School of Food Science and Environmental Health, Technological University Dublin, Grangegorman Lower, D07 H6K8 Dublin, Ireland; (G.T.); (B.J.)
- Nanolab Research Centre, FOCAS Research Institute, Technological University Dublin, Camden Row, D08 CKP1 Dublin, Ireland
| | - Baljit Singh
- MiCRA Biodiagnostics Technology Gateway, Technological University Dublin (TU Dublin), D24 FKT9 Dublin, Ireland;
| | - Furong Tian
- School of Food Science and Environmental Health, Technological University Dublin, Grangegorman Lower, D07 H6K8 Dublin, Ireland; (G.T.); (B.J.)
- Nanolab Research Centre, FOCAS Research Institute, Technological University Dublin, Camden Row, D08 CKP1 Dublin, Ireland
| |
Collapse
|
3
|
Xu J, Jiang J, Yin C, Wang Y, Shi B. Identification of ATP6V0A4 as a potential biomarker in renal cell carcinoma using integrated bioinformatics analysis. Oncol Lett 2023; 26:366. [PMID: 37559594 PMCID: PMC10407721 DOI: 10.3892/ol.2023.13952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 06/16/2023] [Indexed: 08/11/2023] Open
Abstract
Clear cell renal cell carcinoma (ccRCC) is the most common pathological type of renal cancer, and is associated with a high mortality rate, which is related to high rates of tumor recurrence and metastasis. The aim of the present study was to identify reliable molecular biomarkers with high specificity and sensitivity for ccRCC. A total of eight ccRCC-related expression profiles were downloaded from Gene Expression Omnibus for integrated bioinformatics analysis to screen for significantly differentially expressed genes (DEGs). Reverse transcription-quantitative (RT-q)PCR, western blotting and immunohistochemistry staining assays were performed to evaluate the expression levels of candidate biomarkers in ccRCC tissues and cell lines. In total, 255 ccRCC specimens and 165 adjacent normal kidney specimens were analyzed, and 344 significant DEGs, consisting of 115 upregulated DEGs and 229 downregulated DEGs, were identified. The results of Gene Ontology analysis suggested a significant enrichment of DEGs in 'organic anion transport' and 'small molecule catabolic process' in biological processes, in 'apical plasma membrane' and 'apical part of the cell' in cell components, and in 'anion transmembrane transporter activity' and 'active transmembrane transporter activity' in molecular functions. Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis indicated that the DEGs were significantly enriched in the 'phagosome', the 'PPAR signaling pathway', 'complement and coagulation cascades', the 'HIF-1 signaling pathway' and 'carbon metabolism'. Next, 7 hub genes (SUCNR1, CXCR4, VCAN, CASR, ATP6V0A4, VEGFA and SERPINE1) were identified and validated using The Cancer Genome Atlas database. Survival analysis showed that low expression of ATP6V0A4 was associated with a poor prognosis in patients with ccRCC. Additionally, received operating characteristic curves indicated that ATP6V0A4 could distinguish ccRCC samples from normal kidney samples. Furthermore, RT-qPCR, western blotting and immunohistochemistry staining results showed that ATP6V0A4 was significantly downregulated in ccRCC tissues and cell lines. In conclusion, ATP6V0A4 may be involved in tumor progression and regarded as a potential therapeutic target for the recurrence and metastasis of ccRCC.
Collapse
Affiliation(s)
- Jinming Xu
- Department of Urology, Shenzhen Second People's Hospital/First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong 518035, P.R. China
- Department of Urology, Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Jiahao Jiang
- Department of Urology, Shenzhen Second People's Hospital, Clinical College of Anhui Medical University, Shenzhen, Guangdong 518035, P.R. China
| | - Cong Yin
- Department of Urology, Shenzhen Second People's Hospital/First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong 518035, P.R. China
| | - Yan Wang
- Department of Urology, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, P.R. China
| | - Bentao Shi
- Department of Urology, Shenzhen Second People's Hospital/First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong 518035, P.R. China
| |
Collapse
|
4
|
Su K, Collins MP, McGuire CM, Alshagawi MA, Alamoudi MK, Li Z, Forgac M. Isoform a4 of the vacuolar ATPase a subunit promotes 4T1-12B breast cancer cell-dependent tumor growth and metastasis in vivo. J Biol Chem 2022; 298:102395. [PMID: 35988642 PMCID: PMC9508560 DOI: 10.1016/j.jbc.2022.102395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 08/11/2022] [Accepted: 08/15/2022] [Indexed: 12/24/2022] Open
Abstract
The vacuolar H+-ATPase (V-ATPase) is an ATP-dependent proton pump that governs the pH of various intracellular compartments and also functions at the plasma membrane in certain cell types, including cancer cells. Membrane targeting of the V-ATPase is controlled by isoforms of subunit a, and we have previously shown that isoforms a3 and a4 are important for the migration and invasion of several breast cancer cell lines in vitro. Using CRISPR-mediated genome editing to selectively disrupt each of the four a subunit isoforms, we also recently showed that a4 is critical to plasma membrane V-ATPase localization, as well as in vitro migration and invasion of 4T1-12B murine breast cancer cells. We now report that a4 is important for the growth of 4T1-12B tumors in vivo. We found that BALB/c mice bearing a4-/- 4T1-12B allografts had significantly smaller tumors than mice in the control group. In addition, we determined that a4-/- allografts showed dramatically reduced metastases to the lung and reduced luminescence intensity of metastases to bone relative to the control group. Taken together, these results suggest that the a4 isoform of the V-ATPase represents a novel potential therapeutic target to limit breast cancer growth and metastasis.
Collapse
Affiliation(s)
- Kevin Su
- Department of Pharmacology and Drug Development, Graduate School of Biomedical Sciences, Tufts University, Boston, Massachusetts, USA
| | - Michael P Collins
- Department of Cellular, Molecular and Developmental Biology, Graduate School of Biomedical Sciences, Tufts University, Boston, Massachusetts, USA
| | - Christina M McGuire
- Department of Biochemistry, Graduate School of Biomedical Sciences, Tufts University, Boston, Massachusetts, USA
| | - Mohammed A Alshagawi
- Department of Pharmacology and Drug Development, Graduate School of Biomedical Sciences, Tufts University, Boston, Massachusetts, USA
| | - Mariam K Alamoudi
- Department of Developmental, Molecular, and Chemical Biology, Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Zhen Li
- Department of Pharmacology and Drug Development, Graduate School of Biomedical Sciences, Tufts University, Boston, Massachusetts, USA
| | - Michael Forgac
- Department of Pharmacology and Drug Development, Graduate School of Biomedical Sciences, Tufts University, Boston, Massachusetts, USA; Department of Cellular, Molecular and Developmental Biology, Graduate School of Biomedical Sciences, Tufts University, Boston, Massachusetts, USA; Department of Biochemistry, Graduate School of Biomedical Sciences, Tufts University, Boston, Massachusetts, USA; Department of Developmental, Molecular, and Chemical Biology, Tufts University School of Medicine, Boston, Massachusetts, USA.
| |
Collapse
|
5
|
Keon KA, Benlekbir S, Kirsch SH, Müller R, Rubinstein JL. Cryo-EM of the Yeast V O Complex Reveals Distinct Binding Sites for Macrolide V-ATPase Inhibitors. ACS Chem Biol 2022; 17:619-628. [PMID: 35148071 DOI: 10.1021/acschembio.1c00894] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Vacuolar-type adenosine triphosphatases (V-ATPases) are proton pumps found in almost all eukaryotic cells. These enzymes consist of a soluble catalytic V1 region that hydrolyzes ATP and a membrane-embedded VO region responsible for proton translocation. V-ATPase activity leads to acidification of endosomes, phagosomes, lysosomes, secretory vesicles, and the trans-Golgi network, with extracellular acidification occurring in some specialized cells. Small-molecule inhibitors of V-ATPase have played a crucial role in elucidating numerous aspects of cell biology by blocking acidification of intracellular compartments, while therapeutic use of V-ATPase inhibitors has been proposed for the treatment of cancer, osteoporosis, and some infections. Here, we determine structures of the isolated VO complex from Saccharomyces cerevisiae bound to two well-known macrolide inhibitors: bafilomycin A1 and archazolid A. The structures reveal different binding sites for the inhibitors on the surface of the proton-carrying c ring, with only a small amount of overlap between the two sites. Binding of both inhibitors is mediated primarily through van der Waals interactions in shallow pockets and suggests that the inhibitors block rotation of the ring. Together, these structures indicate the existence of a large chemical space available for V-ATPase inhibitors that block acidification by binding the c ring.
Collapse
Affiliation(s)
- Kristine A. Keon
- Molecular Medicine Program, The Hospital for Sick Children, Toronto, Canada M5G0A4
- Department of Medical Biophysics, The University of Toronto, Toronto, Canada M5G1L7
| | - Samir Benlekbir
- Molecular Medicine Program, The Hospital for Sick Children, Toronto, Canada M5G0A4
| | - Susanne H. Kirsch
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Saarland University Campus, 66123 Saarbrücken, Germany
| | - Rolf Müller
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI) and Department of Pharmacy, Saarland University Campus, 66123 Saarbrücken, Germany
| | - John L. Rubinstein
- Molecular Medicine Program, The Hospital for Sick Children, Toronto, Canada M5G0A4
- Department of Medical Biophysics, The University of Toronto, Toronto, Canada M5G1L7
- Department of Biochemistry, The University of Toronto, Toronto, Canada M5S1A8
| |
Collapse
|
6
|
Worsley CM, Veale RB, Mayne ES. The acidic tumour microenvironment: Manipulating the immune response to elicit escape. Hum Immunol 2022; 83:399-408. [PMID: 35216847 DOI: 10.1016/j.humimm.2022.01.014] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 01/25/2022] [Accepted: 01/25/2022] [Indexed: 12/18/2022]
Abstract
The success of cancer treatment relies on the composition of the tumour microenvironment which is comprised of tumour cells, blood vessels, stromal cells, immune cells, and extracellular matrix components. Barriers to effective cancer treatment need to be overcome, and the acidic microenvironment of the tumour provides a key target for treatment. This review intends to provide an overview of the effects that low extracellular pH has on components of the tumour microenvironment and how they contribute to immune escape. Further, potential therapeutic targets will be discussed.
Collapse
Affiliation(s)
- Catherine M Worsley
- Department of Immunology, Faculty of Health Sciences, University of Pretoria, South Africa; Department of Haematology and Molecular Medicine, Faculty of Health Sciences, University of the Witwatersrand, South Africa; National Health Laboratory Service, South Africa.
| | - Rob B Veale
- School of Molecular and Cell Biology, Faculty of Science, University of the Witwatersrand, South Africa
| | - Elizabeth S Mayne
- Department of Haematology and Molecular Medicine, Faculty of Health Sciences, University of the Witwatersrand, South Africa; Department of Immunology Faculty of Health Sciences, University of the Witwatersrand, South Africa; Division of Immunology, Department of Pathology, Faculty of Health Sciences, University of Cape Town, South Africa
| |
Collapse
|
7
|
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: 42] [Impact Index Per Article: 14.0] [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
|
8
|
Targeting lysosomes in human disease: from basic research to clinical applications. Signal Transduct Target Ther 2021; 6:379. [PMID: 34744168 PMCID: PMC8572923 DOI: 10.1038/s41392-021-00778-y] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 09/26/2021] [Indexed: 01/18/2023] Open
Abstract
In recent years, accumulating evidence has elucidated the role of lysosomes in dynamically regulating cellular and organismal homeostasis. Lysosomal changes and dysfunction have been correlated with the development of numerous diseases. In this review, we interpreted the key biological functions of lysosomes in four areas: cellular metabolism, cell proliferation and differentiation, immunity, and cell death. More importantly, we actively sought to determine the characteristic changes and dysfunction of lysosomes in cells affected by these diseases, the causes of these changes and dysfunction, and their significance to the development and treatment of human disease. Furthermore, we outlined currently available targeting strategies: (1) targeting lysosomal acidification; (2) targeting lysosomal cathepsins; (3) targeting lysosomal membrane permeability and integrity; (4) targeting lysosomal calcium signaling; (5) targeting mTOR signaling; and (6) emerging potential targeting strategies. Moreover, we systematically summarized the corresponding drugs and their application in clinical trials. By integrating basic research with clinical findings, we discussed the current opportunities and challenges of targeting lysosomes in human disease.
Collapse
|
9
|
Li Z, Ning F, Wang C, Yu H, Ma Q, Sun Y. Normalization of the tumor microvasculature based on targeting and modulation of the tumor microenvironment. NANOSCALE 2021; 13:17254-17271. [PMID: 34651623 DOI: 10.1039/d1nr03387e] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Angiogenesis is an essential process for tumor development. Owing to the imbalance between pro- and anti-angiogenic factors, the tumor vasculature possesses the characteristics of tortuous, hyperpermeable vessels and compressive force, resulting in a reduction in the effect of traditional chemotherapy and radiotherapy. Anti-angiogenesis has emerged as a promising strategy for cancer treatment. Tumor angiogenesis, however, has been proved to be a complex process in which the tumor microenvironment (TME) plays a vital role in the initiation and development of the tumor microvasculature. The host stromal cells in the TME, such as cancer associated fibroblasts (CAFs), tumor associated macrophages (TAMs) and Treg cells, contribute to angiogenesis. Furthermore, the abnormal metabolic environment, such as hypoxia and acidosis, leads to the up-regulated expression of angiogenic factors. Indeed, normalization of the tumor microvasculature via targeting and modulating the TME has become a promising strategy for anti-angiogenesis and anti-tumor therapy. In this review, we summarize the abnormalities of the tumor microvasculature, tumor angiogenesis induced by an abnormal metabolic environment and host stromal cells, as well as drug delivery therapies to restore the balance between pro- and anti-angiogenic factors by targeting and normalizing the tumor vasculature in the TME.
Collapse
Affiliation(s)
- Zhipeng Li
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao 266021, China.
| | - Fang Ning
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao 266021, China.
| | - Changduo Wang
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao 266021, China.
| | - Hongli Yu
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao 266021, China.
| | - Qingming Ma
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao 266021, China.
| | - Yong Sun
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao 266021, China.
| |
Collapse
|
10
|
Scheeff S, Rivière S, Ruiz J, Dedenbach S, Menche D. Modular Total Synthesis of iso-Archazolids and Archazologs. J Org Chem 2021; 86:10190-10223. [PMID: 34293866 DOI: 10.1021/acs.joc.1c00946] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Full details on the design, development, and successful implementation of suitable synthetic strategies directed toward the total synthesis of iso-archazolids and archazologs are reported. Both a biomimetic and a multistep total synthesis of iso-archazolid B, the most potent and least abundant archazolid, are described. The bioinspired conversion from archazolid B was realized by a high-yielding 1,8-Diazabicyclo[5.4.0]undec-7-ene catalyzed one-step double-bond shift. A highly stereoselective total synthesis was accomplished in 25 steps, involving a sequence of highly stereoselective aldol reactions, an efficient aldol condensation to forge two elaborate fragments, and a challenging ring-closing metathesis macrocyclization with an unusual Stewart-Grubbs catalyst. These strategies proved to be generally useful and could be successfully implemented for the preparation of three novel iso-archazolids as well as five novel archazologs, lacking the thiazole side chain. A wide variety of further archazolids and archazologs may now be targeted for exploration of the promising anticancer potential of these polyketide macrolides.
Collapse
Affiliation(s)
- Stephan Scheeff
- Kekulé-Institut für Organische Chemie und Biochemie, Universität Bonn, Gerhard-Domagk-Strasse 1, 53121 Bonn, Germany
| | - Solenne Rivière
- Kekulé-Institut für Organische Chemie und Biochemie, Universität Bonn, Gerhard-Domagk-Strasse 1, 53121 Bonn, Germany
| | - Johal Ruiz
- Kekulé-Institut für Organische Chemie und Biochemie, Universität Bonn, Gerhard-Domagk-Strasse 1, 53121 Bonn, Germany
| | - Simon Dedenbach
- Kekulé-Institut für Organische Chemie und Biochemie, Universität Bonn, Gerhard-Domagk-Strasse 1, 53121 Bonn, Germany
| | - Dirk Menche
- Kekulé-Institut für Organische Chemie und Biochemie, Universität Bonn, Gerhard-Domagk-Strasse 1, 53121 Bonn, Germany
| |
Collapse
|
11
|
Menche D. Design and Synthesis of Simplified Polyketide Analogs: New Modalities beyond the Rule of 5. ChemMedChem 2021; 16:2068-2074. [PMID: 33755304 PMCID: PMC8360190 DOI: 10.1002/cmdc.202100150] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Indexed: 12/29/2022]
Abstract
Natural products provide important lead structures for development of pharmaceutical agents or present attractive tools for medicinal chemistry. However, structurally complex and thus less accessible metabolites defying conventional drug-like properties, as expressed by Pfizer's rule of five, have received less attention as medicinal leads. Traditionally, research focus has been on realizing total syntheses rather than developing more readily available analogs to resolve the critical supply issue. However, very recent studies with complex myxobacterial polyketides have demonstrated that considerable structural simplification may be realized with retention of biological potencies. The context, underlying rationale and importance of tailored synthetic strategies of three such case studies are presented, which may inspire further related activities and may eventually help exploiting the largely untapped biological potential of complex metabolites in general.
Collapse
Affiliation(s)
- Dirk Menche
- Kekulé-Institut für Organische Chemie und BiochemieUniversität BonnGerhard-Domagk-Strasse 153121BonnGermany
| |
Collapse
|
12
|
Xie X, Lu S, Pan X, Zou M, Li F, Lin H, Hu J, Fan S, He J. Antiviral Bafilomycins from a Feces-Inhabiting Streptomyces sp. JOURNAL OF NATURAL PRODUCTS 2021; 84:537-543. [PMID: 33631936 DOI: 10.1021/acs.jnatprod.0c01243] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
A new bafilomycin derivative (1) and another seven known bafilomycins (2-8) were isolated from feces-derived Streptomyces sp. HTL16. The structure of 1 was elucidated by 1D and 2D NMR spectroscopic analysis. Biological testing demonstrated that these bafilomycins exhibited potent antiviral activities against the influenza A and SARS-CoV-2 viruses, with IC50 values in the nanomolar range, by inhibiting the activity of endosomal ATP-driven proton pumps.
Collapse
Affiliation(s)
- Xi Xie
- Group of Peptides and Natural Products Research, School of Pharmaceutical Sciences, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou 510515, People's Republic of China
| | - Shengsheng Lu
- Group of Peptides and Natural Products Research, School of Pharmaceutical Sciences, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou 510515, People's Republic of China
| | - Xiaoyan Pan
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan 430071, People's Republic of China
| | - Min Zou
- Group of Peptides and Natural Products Research, School of Pharmaceutical Sciences, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou 510515, People's Republic of China
| | - Fangfang Li
- Guangdong Provincial Key Laboratory of Emergency Test for Dangerous Chemicals, China National Analytical Center, Guangzhou 510070, People's Republic of China
| | - Haixing Lin
- Group of Peptides and Natural Products Research, School of Pharmaceutical Sciences, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou 510515, People's Republic of China
| | - Jianan Hu
- Group of Peptides and Natural Products Research, School of Pharmaceutical Sciences, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou 510515, People's Republic of China
| | - Sheng Fan
- Group of Peptides and Natural Products Research, School of Pharmaceutical Sciences, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou 510515, People's Republic of China
| | - Jian He
- Group of Peptides and Natural Products Research, School of Pharmaceutical Sciences, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou 510515, People's Republic of China
| |
Collapse
|
13
|
Role of the V1G1 subunit of V-ATPase in breast cancer cell migration. Sci Rep 2021; 11:4615. [PMID: 33633298 PMCID: PMC7907067 DOI: 10.1038/s41598-021-84222-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 01/18/2021] [Indexed: 12/26/2022] Open
Abstract
V-ATPase is a large multi-subunit complex that regulates acidity of intracellular compartments and of extracellular environment. V-ATPase consists of several subunits that drive specific regulatory mechanisms. The V1G1 subunit, a component of the peripheral stalk of the pump, controls localization and activation of the pump on late endosomes and lysosomes by interacting with RILP and RAB7. Deregulation of some subunits of the pump has been related to tumor invasion and metastasis formation in breast cancer. We observed a decrease of V1G1 and RAB7 in highly invasive breast cancer cells, suggesting a key role of these proteins in controlling cancer progression. Moreover, in MDA-MB-231 cells, modulation of V1G1 affected cell migration and matrix metalloproteinase activation in vitro, processes important for tumor formation and dissemination. In these cells, characterized by high expression of EGFR, we demonstrated that V1G1 modulates EGFR stability and the EGFR downstream signaling pathways that control several factors required for cell motility, among which RAC1 and cofilin. In addition, we showed a key role of V1G1 in the biogenesis of endosomes and lysosomes. Altogether, our data describe a new molecular mechanism, controlled by V1G1, required for cell motility and that promotes breast cancer tumorigenesis.
Collapse
|
14
|
Santos-Pereira C, Rodrigues LR, Côrte-Real M. Emerging insights on the role of V-ATPase in human diseases: Therapeutic challenges and opportunities. Med Res Rev 2021; 41:1927-1964. [PMID: 33483985 DOI: 10.1002/med.21782] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 12/05/2020] [Accepted: 01/05/2021] [Indexed: 12/13/2022]
Abstract
The control of the intracellular pH is vital for the survival of all organisms. Membrane transporters, both at the plasma and intracellular membranes, are key players in maintaining a finely tuned pH balance between intra- and extracellular spaces, and therefore in cellular homeostasis. V-ATPase is a housekeeping ATP-driven proton pump highly conserved among prokaryotes and eukaryotes. This proton pump, which exhibits a complex multisubunit structure based on cell type-specific isoforms, is essential for pH regulation and for a multitude of ubiquitous and specialized functions. Thus, it is not surprising that V-ATPase aberrant overexpression, mislocalization, and mutations in V-ATPase subunit-encoding genes have been associated with several human diseases. However, the ubiquitous expression of this transporter and the high toxicity driven by its off-target inhibition, renders V-ATPase-directed therapies very challenging and increases the need for selective strategies. Here we review emerging evidence linking V-ATPase and both inherited and acquired human diseases, explore the therapeutic challenges and opportunities envisaged from recent data, and advance future research avenues. We highlight the importance of V-ATPases with unique subunit isoform molecular signatures and disease-associated isoforms to design selective V-ATPase-directed therapies. We also discuss the rational design of drug development pipelines and cutting-edge methodological approaches toward V-ATPase-centered drug discovery. Diseases like cancer, osteoporosis, and even fungal infections can benefit from V-ATPase-directed therapies.
Collapse
Affiliation(s)
- Cátia Santos-Pereira
- Department of Biology, Centre of Molecular and Environmental Biology (CBMA), University of Minho, Braga, Portugal.,Department of Biological Engineering, Centre of Biological Engineering (CEB), University of Minho, Braga, Portugal
| | - Lígia R Rodrigues
- Department of Biological Engineering, Centre of Biological Engineering (CEB), University of Minho, Braga, Portugal
| | - Manuela Côrte-Real
- Department of Biology, Centre of Molecular and Environmental Biology (CBMA), University of Minho, Braga, Portugal
| |
Collapse
|
15
|
Shear Stress Increases V-H + -ATPase and Acidic Vesicle Number Density, and p-mTORC2 Activation in Prostate Cancer Cells. Cell Mol Bioeng 2020; 13:591-604. [PMID: 33281989 DOI: 10.1007/s12195-020-00632-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 06/18/2020] [Indexed: 02/06/2023] Open
Abstract
Introduction Cells in the tumor microenvironment experience mechanical stresses, such as compression generated by uncontrolled cell growth within a tissue, increased substrate stiffness due to tumor cell extracellular matrix (ECM) remodeling, and leaky angiogenic vessels which involve low fluid shear stress. With our hypothesis that shear stress increases V-H + -ATPase number density in prostate cancer cells via activation of the mTORC1 and mTORC2 pathways, we demonstrated and quantified such a mechanism in prostate cancer cells. Methods Moderately metastatic DU145 and highly metastatic PC3 prostate cancer cells were subjected to 0.05 dynes cm - 2 wall shear stress for 24 h, followed by immunocytochemistry and fluorescence measurements of β 1 integrin, endosome, lysosome, V-H + -ATPase proton pump, mTORC1, and p-mTORC2 antibodies. Post shear stress migration assays, and the effects of vacuolar proton pump inhibitor Bafilomycin A1 (60 nM, 24 h) as well as shear stress on the ICC fluorescence intensity of the proteins of interest were conducted with DU145 cells. Results Low fluid shear stress increases the fluorescence intensity of β 1 integrin, endosome, lysosome, V-H + -ATPase, mTORC1, and p-mTORC2 antibodies in PC3 and DU145 cells, and also increased cell migration. However, Bafilomycin A1 decreased fluorescence intensity of all of these proteins in DU145 cells exposed to shear stress, revealing that V-H + -ATPase controls the expression of these proteins. Conclusions Prostate cancer cell mechanotransduction increases endosomes, lysosomes, and proton pumps-where increases have been associated with enhanced cancer aggressiveness. We also show that the prostate cancer cell's response to force promotes the cancer drivers mTORC1 and mTORC2.
Collapse
|
16
|
Rivière S, Vielmuth C, Ennenbach C, Abdelrahman A, Lemke C, Gütschow M, Müller CE, Menche D. Design, Synthesis and Biological Evaluation of Highly Potent Simplified Archazolids. ChemMedChem 2020; 15:1348-1363. [PMID: 32363789 PMCID: PMC7496434 DOI: 10.1002/cmdc.202000154] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Indexed: 01/25/2023]
Abstract
The archazolids are potent antiproliferative compounds that have recently emerged as a novel class of promising anticancer agents. Their complex macrolide structures and scarce natural supply make the development of more readily available analogues highly important. Herein, we report the design, synthesis and biological evaluation of four simplified and partially saturated archazolid derivatives. We also reveal important structure-activity relationship data as well as insights into the pharmacophore of these complex polyketides.
Collapse
Affiliation(s)
- Solenne Rivière
- Kekulé-Institut für Organische Chemie und BiochemieUniversität BonnGerhard-Domagk-Strasse 153121BonnGermany
| | - Christin Vielmuth
- Pharmazeutische & Medizinische ChemiePharmazeutisches InstitutUniversität BonnAn der Immenburg 453121BonnGermany
| | - Christiane Ennenbach
- Pharmazeutische & Medizinische ChemiePharmazeutisches InstitutUniversität BonnAn der Immenburg 453121BonnGermany
| | - Aliaa Abdelrahman
- Pharmazeutische & Medizinische ChemiePharmazeutisches InstitutUniversität BonnAn der Immenburg 453121BonnGermany
| | - Carina Lemke
- Pharmazeutische & Medizinische ChemiePharmazeutisches InstitutUniversität BonnAn der Immenburg 453121BonnGermany
| | - Michael Gütschow
- Pharmazeutische & Medizinische ChemiePharmazeutisches InstitutUniversität BonnAn der Immenburg 453121BonnGermany
| | - Christa E. Müller
- Pharmazeutische & Medizinische ChemiePharmazeutisches InstitutUniversität BonnAn der Immenburg 453121BonnGermany
| | - Dirk Menche
- Kekulé-Institut für Organische Chemie und BiochemieUniversität BonnGerhard-Domagk-Strasse 153121BonnGermany
| |
Collapse
|
17
|
Peng W, Casey AK, Fernandez J, Carpinone EM, Servage KA, Chen Z, Li Y, Tomchick DR, Starai VJ, Orth K. A distinct inhibitory mechanism of the V-ATPase by Vibrio VopQ revealed by cryo-EM. Nat Struct Mol Biol 2020; 27:589-597. [PMID: 32424347 DOI: 10.1038/s41594-020-0429-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 04/01/2020] [Indexed: 12/18/2022]
Abstract
The Vibrio parahaemolyticus T3SS effector VopQ targets host-cell V-ATPase, resulting in blockage of autophagic flux and neutralization of acidic compartments. Here, we report the cryo-EM structure of VopQ bound to the Vo subcomplex of the V-ATPase. VopQ inserts into membranes and forms an unconventional pore while binding directly to subunit c of the V-ATPase membrane-embedded subcomplex Vo. We show that VopQ arrests yeast growth in vivo by targeting the immature Vo subcomplex in the endoplasmic reticulum (ER), thus providing insight into the observation that VopQ kills cells in the absence of a functional V-ATPase. VopQ is a bacterial effector that has been discovered to inhibit a host-membrane megadalton complex by coincidentally binding its target, inserting into a membrane and disrupting membrane potential. Collectively, our results reveal a mechanism by which bacterial effectors modulate host cell biology and provide an invaluable tool for future studies on V-ATPase-mediated membrane fusion and autophagy.
Collapse
Affiliation(s)
- Wei Peng
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Amanda K Casey
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jessie Fernandez
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | | | - Kelly A Servage
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Zhe Chen
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Yang Li
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Diana R Tomchick
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Vincent J Starai
- Department of Microbiology, University of Georgia, Athens, GA, USA
- Department of Infectious Diseases, University of Georgia, Athens, GA, USA
| | - Kim Orth
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA.
- Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA.
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
18
|
Collins MP, Forgac M. Regulation and function of V-ATPases in physiology and disease. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2020; 1862:183341. [PMID: 32422136 DOI: 10.1016/j.bbamem.2020.183341] [Citation(s) in RCA: 90] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 04/30/2020] [Accepted: 05/03/2020] [Indexed: 02/07/2023]
Abstract
The vacuolar H+-ATPases (V-ATPases) are essential, ATP-dependent proton pumps present in a variety of eukaryotic cellular membranes. Intracellularly, V-ATPase-dependent acidification functions in such processes as membrane traffic, protein degradation, autophagy and the coupled transport of small molecules. V-ATPases at the plasma membrane of certain specialized cells function in such processes as bone resorption, sperm maturation and urinary acidification. V-ATPases also function in disease processes such as pathogen entry and cancer cell invasiveness, while defects in V-ATPase genes are associated with disorders such as osteopetrosis, renal tubular acidosis and neurodegenerative diseases. This review highlights recent advances in our understanding of V-ATPase structure, mechanism, function and regulation, with an emphasis on the signaling pathways controlling V-ATPase assembly in mammalian cells. The role of V-ATPases in cancer and other human pathologies, and the prospects for therapeutic intervention, are also discussed.
Collapse
Affiliation(s)
- Michael P Collins
- Cell, Molecular and Developmental Biology, Tufts University Graduate School of Biomedical Sciences, United States of America
| | - Michael Forgac
- Cell, Molecular and Developmental Biology, Tufts University Graduate School of Biomedical Sciences, United States of America; Dept. of Developmental, Molecular and Chemical Biology, Tufts University School of Medicine, Boston, MA 02111, United States of America.
| |
Collapse
|
19
|
Scheeff S, Rivière S, Ruiz J, Abdelrahman A, Schulz-Fincke AC, Köse M, Tiburcy F, Wieczorek H, Gütschow M, Müller CE, Menche D. Synthesis of Novel Potent Archazolids: Pharmacology of an Emerging Class of Anticancer Drugs. J Med Chem 2020; 63:1684-1698. [DOI: 10.1021/acs.jmedchem.9b01887] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Affiliation(s)
- Stephan Scheeff
- Kekulé-Institut für Organische Chemie und Biochemie, Universität Bonn, Gerhard-Domagk-Str. 1, D-53121 Bonn, Germany
| | - Solenne Rivière
- Kekulé-Institut für Organische Chemie und Biochemie, Universität Bonn, Gerhard-Domagk-Str. 1, D-53121 Bonn, Germany
| | - Johal Ruiz
- Kekulé-Institut für Organische Chemie und Biochemie, Universität Bonn, Gerhard-Domagk-Str. 1, D-53121 Bonn, Germany
| | - Aliaa Abdelrahman
- Pharmazeutisches Institut, Universität Bonn, An der Immenburg 4, D-53121 Bonn, Germany
| | | | - Meryem Köse
- Pharmazeutisches Institut, Universität Bonn, An der Immenburg 4, D-53121 Bonn, Germany
| | - Felix Tiburcy
- Fachbereich Biologie/Chemie, Universität Osnabrück, D-49069 Osnabrück, Germany
| | - Helmut Wieczorek
- Fachbereich Biologie/Chemie, Universität Osnabrück, D-49069 Osnabrück, Germany
| | - Michael Gütschow
- Pharmazeutisches Institut, Universität Bonn, An der Immenburg 4, D-53121 Bonn, Germany
| | - Christa E. Müller
- Pharmazeutisches Institut, Universität Bonn, An der Immenburg 4, D-53121 Bonn, Germany
| | - Dirk Menche
- Kekulé-Institut für Organische Chemie und Biochemie, Universität Bonn, Gerhard-Domagk-Str. 1, D-53121 Bonn, Germany
| |
Collapse
|
20
|
Cellular and Extracellular Components in Tumor Microenvironment and Their Application in Early Diagnosis of Cancers. Anal Cell Pathol (Amst) 2020; 2020:6283796. [PMID: 32377504 PMCID: PMC7199555 DOI: 10.1155/2020/6283796] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 12/11/2019] [Accepted: 12/23/2019] [Indexed: 12/11/2022] Open
Abstract
Tumors are surrounded by complex environmental components, including blood and lymph vessels, fibroblasts, endothelial cells, immune cells, cytokines, extracellular vesicles, and extracellular matrix. All the stromal components together with the tumor cells form the tumor microenvironment (TME). In addition, extracellular physical and chemical factors, including extracellular pH, hypoxia, elevated interstitial fluid pressure, and fibrosis, are closely associated with tumor progression, metastasis, immunosuppression, and drug resistance. Cellular and extracellular components in TME contribute to nearly all procedures of carcinogenesis. By summarizing the recent work in this field, we make a comprehensive review on the role of cellular and extracellular components in the process of carcinogenesis and their potential application in early diagnosis of cancer. We hope that a systematic review of the diverse aspects of TME will help both research scientists and clinicians in this field.
Collapse
|
21
|
Bartel K, Müller R, von Schwarzenberg K. Differential regulation of AMP-activated protein kinase in healthy and cancer cells explains why V-ATPase inhibition selectively kills cancer cells. J Biol Chem 2019; 294:17239-17248. [PMID: 31604821 DOI: 10.1074/jbc.ra119.010243] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 09/16/2019] [Indexed: 01/02/2023] Open
Abstract
The cellular energy sensor AMP-activated protein kinase (AMPK) is a metabolic hub regulating various pathways involved in tumor metabolism. Here we report that vacuolar H+-ATPase (V-ATPase) inhibition differentially affects regulation of AMPK in tumor and nontumor cells and that this differential regulation contributes to the selectivity of V-ATPase inhibitors for tumor cells. In nonmalignant cells, the V-ATPase inhibitor archazolid increased phosphorylation and lysosomal localization of AMPK. We noted that AMPK localization has a prosurvival role, as AMPK silencing decreased cellular growth rates. In contrast, in cancer cells, we found that AMPK is constitutively active and that archazolid does not affect its phosphorylation and localization. Moreover, V-ATPase-independent AMPK induction in tumor cells protected them from archazolid-induced cytotoxicity, further underlining the role of AMPK as a prosurvival mediator. These observations indicate that AMPK regulation is uncoupled from V-ATPase activity in cancer cells and that this makes them more susceptible to cell death induction by V-ATPase inhibitors. In both tumor and healthy cells, V-ATPase inhibition induced a distinct metabolic regulatory cascade downstream of AMPK, affecting ATP and NADPH levels, glucose uptake, and reactive oxygen species production. We could attribute the prosurvival effects to AMPK's ability to maintain redox homeostasis by inhibiting reactive oxygen species production and maintaining NADPH levels. In summary, the results of our work indicate that V-ATPase inhibition has differential effects on AMPK-mediated metabolic regulation in cancer and healthy cells and explain the tumor-specific cytotoxicity of V-ATPase inhibition.
Collapse
Affiliation(s)
- Karin Bartel
- Department of Pharmacy, Pharmaceutical Biology, Ludwig Maximilians University, 81377 Munich, Germany
| | - Rolf Müller
- Helmholtz Center for Pharmaceutical Research Saarland, Helmholtz Centre for Infection Research, and Department of Pharmacy at Saarland University, Saarland University Campus, 66123 Saarbrücken, Germany
| | - Karin von Schwarzenberg
- Department of Pharmacy, Pharmaceutical Biology, Ludwig Maximilians University, 81377 Munich, Germany
| |
Collapse
|
22
|
Bartel K, Pein H, Popper B, Schmitt S, Janaki-Raman S, Schulze A, Lengauer F, Koeberle A, Werz O, Zischka H, Müller R, Vollmar AM, von Schwarzenberg K. Connecting lysosomes and mitochondria - a novel role for lipid metabolism in cancer cell death. Cell Commun Signal 2019; 17:87. [PMID: 31358011 PMCID: PMC6664539 DOI: 10.1186/s12964-019-0399-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 07/15/2019] [Indexed: 12/29/2022] Open
Abstract
Background The understanding of lysosomes has been expanded in recent research way beyond their view as cellular trash can. Lysosomes are pivotal in regulating metabolism, endocytosis and autophagy and are implicated in cancer. Recently it was discovered that the lysosomal V-ATPase, which is known to induce apoptosis, interferes with lipid metabolism in cancer, yet the interplay between these organelles is poorly understood. Methods LC-MS/MS analysis was performed to investigate lipid distribution in cells. Cell survival and signaling pathways were analyzed by means of cell biological methods (qPCR, Western Blot, flow cytometry, CellTiter-Blue). Mitochondrial structure was analyzed by confocal imaging and electron microscopy, their function was determined by flow cytometry and seahorse measurements. Results Our data reveal that interfering with lysosomal function changes composition and subcellular localization of triacylglycerids accompanied by an upregulation of PGC1α and PPARα expression, master regulators of energy and lipid metabolism. Furthermore, cardiolipin content is reduced driving mitochondria into fission, accompanied by a loss of membrane potential and reduction in oxidative capacity, which leads to a deregulation in cellular ROS and induction of mitochondria-driven apoptosis. Additionally, cells undergo a metabolic shift to glutamine dependency, correlated with the fission phenotype and sensitivity to lysosomal inhibition, most prominent in Ras mutated cells. Conclusion This study sheds mechanistic light on a largely uninvestigated triangle between lysosomes, lipid metabolism and mitochondrial function. Insight into this organelle crosstalk increases our understanding of mitochondria-driven cell death. Our findings furthermore provide a first hint on a connection of Ras pathway mutations and sensitivity towards lysosomal inhibitors. Graphical Abstract ![]()
Electronic supplementary material The online version of this article (10.1186/s12964-019-0399-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Karin Bartel
- Department of Pharmacy, Pharmaceutical Biology, Ludwig-Maximilians-Universität München, Butenandtstr. 5-13, 81377, Munich, Germany.
| | - Helmut Pein
- Chair of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, Philosophenweg 14, 07743, Jena, Germany
| | - Bastian Popper
- Department of Anatomy and Cell Biology, Biomedical Center, Ludwig-Maximilians-Universität München, Grosshaderner Strasse 9, 82152, Planegg-Martinsried, Germany
| | - Sabine Schmitt
- Institute of Toxicology and Environmental Hygiene, Technical University Munich, School of Medicine, 80802, Munich, Germany
| | - Sudha Janaki-Raman
- Department of Biochemistry and Molecular Biology, Theodor-Boveri-Institute, Biocenter, Am Hubland, 97074, Würzburg, Germany
| | - Almut Schulze
- Department of Biochemistry and Molecular Biology, Theodor-Boveri-Institute, Biocenter, Am Hubland, 97074, Würzburg, Germany
| | - Florian Lengauer
- Department of Pharmacy, Pharmaceutical Biology, Ludwig-Maximilians-Universität München, Butenandtstr. 5-13, 81377, Munich, Germany
| | - Andreas Koeberle
- Chair of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, Philosophenweg 14, 07743, Jena, Germany
| | - Oliver Werz
- Chair of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, Philosophenweg 14, 07743, Jena, Germany
| | - Hans Zischka
- Institute of Toxicology and Environmental Hygiene, Technical University Munich, School of Medicine, 80802, Munich, Germany.,Institute of Molecular Toxicology and Pharmacology, Helmholtz Center Munich, German Research Center for Environmental Health, 85764, Neuherberg, Germany
| | - Rolf Müller
- Helmholtz Centre for Infection Research and Department of Pharmaceutical Biotechnology, Helmholtz Institute for Pharmaceutical Research Saarland, Saarland University, PO 151150, Universitätscampus E8 1, 66123, Saarbrücken, Germany
| | - Angelika M Vollmar
- Department of Pharmacy, Pharmaceutical Biology, Ludwig-Maximilians-Universität München, Butenandtstr. 5-13, 81377, Munich, Germany
| | - Karin von Schwarzenberg
- Department of Pharmacy, Pharmaceutical Biology, Ludwig-Maximilians-Universität München, Butenandtstr. 5-13, 81377, Munich, Germany.
| |
Collapse
|
23
|
Kocik J, Machula M, Wisniewska A, Surmiak E, Holak TA, Skalniak L. Helping the Released Guardian: Drug Combinations for Supporting the Anticancer Activity of HDM2 (MDM2) Antagonists. Cancers (Basel) 2019; 11:E1014. [PMID: 31331108 PMCID: PMC6678622 DOI: 10.3390/cancers11071014] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Revised: 07/13/2019] [Accepted: 07/16/2019] [Indexed: 01/22/2023] Open
Abstract
The protein p53, known as the "Guardian of the Genome", plays an important role in maintaining DNA integrity, providing protection against cancer-promoting mutations. Dysfunction of p53 is observed in almost every cancer, with 50% of cases bearing loss-of-function mutations/deletions in the TP53 gene. In the remaining 50% of cases the overexpression of HDM2 (mouse double minute 2, human homolog) protein, which is a natural inhibitor of p53, is the most common way of keeping p53 inactive. Disruption of HDM2-p53 interaction with the use of HDM2 antagonists leads to the release of p53 and expression of its target genes, engaged in the induction of cell cycle arrest, DNA repair, senescence, and apoptosis. The induction of apoptosis, however, is restricted to only a handful of p53wt cells, and, generally, cancer cells treated with HDM2 antagonists are not efficiently eliminated. For this reason, HDM2 antagonists were tested in combinations with multiple other therapeutics in a search for synergy that would enhance the cancer eradication. This manuscript aims at reviewing the recent progress in developing strategies of combined cancer treatment with the use of HDM2 antagonists.
Collapse
Affiliation(s)
- Justyna Kocik
- Department of Organic Chemistry, Faculty of Chemistry, Jagiellonian University, ul. Gronostajowa 2, 30-387 Krakow, Poland
| | - Monika Machula
- Department of Organic Chemistry, Faculty of Chemistry, Jagiellonian University, ul. Gronostajowa 2, 30-387 Krakow, Poland
| | - Aneta Wisniewska
- Department of Organic Chemistry, Faculty of Chemistry, Jagiellonian University, ul. Gronostajowa 2, 30-387 Krakow, Poland
| | - Ewa Surmiak
- Department of Organic Chemistry, Faculty of Chemistry, Jagiellonian University, ul. Gronostajowa 2, 30-387 Krakow, Poland
| | - Tad A Holak
- Department of Organic Chemistry, Faculty of Chemistry, Jagiellonian University, ul. Gronostajowa 2, 30-387 Krakow, Poland
| | - Lukasz Skalniak
- Department of Organic Chemistry, Faculty of Chemistry, Jagiellonian University, ul. Gronostajowa 2, 30-387 Krakow, Poland.
| |
Collapse
|
24
|
McGuire CM, Collins MP, Sun-Wada G, Wada Y, Forgac M. Isoform-specific gene disruptions reveal a role for the V-ATPase subunit a4 isoform in the invasiveness of 4T1-12B breast cancer cells. J Biol Chem 2019; 294:11248-11258. [PMID: 31167791 PMCID: PMC6643023 DOI: 10.1074/jbc.ra119.007713] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 05/30/2019] [Indexed: 12/17/2022] Open
Abstract
The vacuolar H+-ATPase (V-ATPase) is an ATP-driven proton pump present in various intracellular membranes and at the plasma membrane of specialized cell types. Previous work has reported that plasma membrane V-ATPases are key players in breast cancer cell invasiveness. The two subunit a-isoforms known to target the V-ATPase to the plasma membrane are a3 and a4, and expression of a3 has been shown to correlate with plasma membrane localization of the V-ATPase in various invasive human breast cancer cell lines. Here we analyzed the role of subunit a-isoforms in the invasive mouse breast cancer cell line, 4T1-12B. Quantitation of mRNA levels for each isoform by quantitative RT-PCR revealed that a4 is the dominant isoform expressed in these cells. Using a CRISPR/Cas9-based approach to disrupt the genes encoding each of the four V-ATPase subunit a-isoforms, we found that ablation of only the a4-encoding gene significantly inhibits invasion and migration of 4T1-12B cells. Additionally, cells with disrupted a4 exhibited reduced V-ATPase expression at the leading edge, suggesting that the a4 isoform is primarily responsible for targeting the V-ATPase to the plasma membrane in 4T1-12B cells. These findings suggest that different subunit a-isoforms may direct V-ATPases to the plasma membrane of different invasive breast cancer cell lines. They further suggest that expression of V-ATPases at the cell surface is the primary factor that promotes an invasive cancer cell phenotype.
Collapse
Affiliation(s)
- Christina M McGuire
- Department of Developmental, Molecular, and Chemical Biology, Tufts University School of Medicine, Tufts University, Boston, Massachusetts 02111
- Program in Biochemistry, Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, Massachusetts 02111
| | - Michael P Collins
- Department of Developmental, Molecular, and Chemical Biology, Tufts University School of Medicine, Tufts University, Boston, Massachusetts 02111
- Program in Cell, Molecular, and Developmental Biology, Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, Massachusetts 02111
| | - GeHong Sun-Wada
- Department of Biochemistry, Faculty of Pharmaceutical Sciences, Doshisha Women's College, Kyotanabe, Kyoto 610-0395, Japan
| | - Yoh Wada
- Division of Biological Science, Institute of Scientific and Industrial Research, Osaka University, Ibaraki, Osaka 567-0047, Japan
| | - Michael Forgac
- Department of Developmental, Molecular, and Chemical Biology, Tufts University School of Medicine, Tufts University, Boston, Massachusetts 02111
- Program in Biochemistry, Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, Massachusetts 02111
- Program in Cell, Molecular, and Developmental Biology, Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, Massachusetts 02111
| |
Collapse
|
25
|
Specific V-ATPase expression sub-classifies IDHwt lower-grade gliomas and impacts glioma growth in vivo. EBioMedicine 2019; 41:214-224. [PMID: 30737087 PMCID: PMC6441867 DOI: 10.1016/j.ebiom.2019.01.052] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 01/24/2019] [Accepted: 01/25/2019] [Indexed: 12/27/2022] Open
Abstract
Background Cancer cells use specific V-ATPase subunits to activate oncogenic pathways. Therefore, we investigated V-ATPase deregulation in aggressive gliomas and associated signaling. Methods V-ATPase genes expression and associated pathways were analyzed in different series of glioma available from public databases, as well as in patients' cohort. Activation of pathways was analyzed at gene and protein expression levels. A genetic model of glioma in Drosophila melanogaster and mice with GBM patients-derived orthotopic xenografts were used as in vivo models of disease. Findings GBM and recurrent gliomas display a specific V-ATPase signature. Such signature resolves the heterogeneous class of IDH-wild type lower-grade gliomas, identifying the patients with worse prognosis independently from clinical and molecular features (p = 0·03, by Cox proportional-hazards model). In vivo, V-ATPase subunits deregulation significantly impacts tumor growth and proliferation. At the molecular level, GBM-like V-ATPase expression correlates with upregulation of Homeobox genes. Interpretation Our data identify a V-ATPase signature that accompanies glioma aggressiveness and suggest new entry points for glioma stratification and follow-up. Fund This work was supported by Fondazione Cariplo (2014–1148 to VV), Fondazione IRCCS Ca' Granda, and Fondazione INGM Grant in Molecular Medicine 2014 (to VV).
Collapse
|
26
|
Ardelt MA, Fröhlich T, Martini E, Müller M, Kanitz V, Atzberger C, Cantonati P, Meßner M, Posselt L, Lehr T, Wojtyniak J, Ulrich M, Arnold GJ, König L, Parazzoli D, Zahler S, Rothenfußer S, Mayr D, Gerbes A, Scita G, Vollmar AM, Pachmayr J. Inhibition of Cyclin-Dependent Kinase 5: A Strategy to Improve Sorafenib Response in Hepatocellular Carcinoma Therapy. Hepatology 2019; 69:376-393. [PMID: 30033593 PMCID: PMC6590289 DOI: 10.1002/hep.30190] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 07/08/2018] [Indexed: 12/29/2022]
Abstract
Therapeutic options for patients with advanced-stage hepatocellular carcinoma (HCC) are very limited. The only approved first-line treatment is the multi-tyrosine kinase inhibitor sorafenib, which shows low response rates and severe side effects. In particular, the compensatory activation of growth factor receptors leads to chemoresistance and limits the clinical impact of sorafenib. However, combination approaches to improve sorafenib have failed. Here we investigate the inhibition of cyclin-dependent kinase 5 (Cdk5) as a promising combination strategy to improve sorafenib response in HCC. Combination of sorafenib with Cdk5 inhibition (genetic knockdown by short hairpin RNA or CRISPR/Cas9 and pharmacologic inhibition) synergistically impaired HCC progression in vitro and in vivo by inhibiting both tumor cell proliferation and migration. Importantly, these effects were mediated by a mechanism for Cdk5: A liquid chromatography-tandem mass spectrometry-based proteomic approach revealed that Cdk5 inhibition interferes with intracellular trafficking, a process crucial for cellular homeostasis and growth factor receptor signaling. Cdk5 inhibition resulted in an accumulation of enlarged vesicles and respective cargos in the perinuclear region, considerably impairing the extent and quality of growth factor receptor signaling. Thereby, Cdk5 inhibition offers a comprehensive approach to globally disturb growth factor receptor signaling that is superior to specific inhibition of individual growth factor receptors. Conclusion: Cdk5 inhibition represents an effective approach to improve sorafenib response and to prevent sorafenib treatment escape in HCC. Notably, Cdk5 is an addressable target frequently overexpressed in HCC, and with Dinaciclib, a clinically tested Cdk5 inhibitor is readily available. Thus, our study provides evidence for clinically evaluating the combination of sorafenib and Dinaciclib to improve the therapeutic situation for patients with advanced-stage HCC.
Collapse
Affiliation(s)
- Maximilian A. Ardelt
- Department of Pharmacy, Pharmaceutical BiologyLMU MunichMunichGermany,Institute of PharmacyParacelsus Medical UniversitySalzburgAustria
| | - Thomas Fröhlich
- Laboratory for Functional Genome Analysis, LAFUGA, Gene CentreUniversity of MunichMunichGermany
| | - Emanuele Martini
- IFOM‐FIRC Institute of Molecular OncologyDepartment of Oncology and Hemato‐OncologyUniversity of MilanMilanItalyMilanItaly
| | - Martin Müller
- Department of Pharmacy, Pharmaceutical BiologyLMU MunichMunichGermany
| | - Veronika Kanitz
- Institute of PathologyLudwig Maximilians University of MunichMunichGermany
| | - Carina Atzberger
- Department of Pharmacy, Pharmaceutical BiologyLMU MunichMunichGermany
| | - Petra Cantonati
- Institute of PharmacyParacelsus Medical UniversitySalzburgAustria
| | - Martina Meßner
- Department of Pharmacy, Pharmaceutical BiologyLMU MunichMunichGermany,Institute of PharmacyParacelsus Medical UniversitySalzburgAustria
| | - Laura Posselt
- Center of Integrated Protein Science Munich (CIPS‐M) and Division of Clinical Pharmacology, Department of Internal Medicine IVKlinikum der Universität MünchenMunichGermany
| | - Thorsten Lehr
- Clinical PharmacySaarland UniversitySaarbrückenGermany
| | | | - Melanie Ulrich
- Department of Pharmacy, Pharmaceutical BiologyLMU MunichMunichGermany
| | - Georg J. Arnold
- Laboratory for Functional Genome Analysis, LAFUGA, Gene CentreUniversity of MunichMunichGermany
| | - Lars König
- Center of Integrated Protein Science Munich (CIPS‐M) and Division of Clinical Pharmacology, Department of Internal Medicine IVKlinikum der Universität MünchenMunichGermany
| | - Dario Parazzoli
- IFOM‐FIRC Institute of Molecular OncologyDepartment of Oncology and Hemato‐OncologyUniversity of MilanMilanItalyMilanItaly
| | - Stefan Zahler
- Department of Pharmacy, Pharmaceutical BiologyLMU MunichMunichGermany
| | - Simon Rothenfußer
- Center of Integrated Protein Science Munich (CIPS‐M) and Division of Clinical Pharmacology, Department of Internal Medicine IVKlinikum der Universität MünchenMunichGermany
| | - Doris Mayr
- Institute of PathologyLudwig Maximilians University of MunichMunichGermany
| | - Alexander Gerbes
- Department of Medicine 2, Liver Center MunichUniversity Hospital, LMU MunichMunichGermany
| | - Giorgio Scita
- IFOM‐FIRC Institute of Molecular OncologyDepartment of Oncology and Hemato‐OncologyUniversity of MilanMilanItalyMilanItaly
| | | | - Johanna Pachmayr
- Department of Pharmacy, Pharmaceutical BiologyLMU MunichMunichGermany,Institute of PharmacyParacelsus Medical UniversitySalzburgAustria
| |
Collapse
|
27
|
Affiliation(s)
- Stephan Scheeff
- Kekulé-Institut für Organische Chemie und Biochemie, Universität Bonn, Gerhard-Domagk-Strasse 1, D-53121 Bonn, Germany
| | - Dirk Menche
- Kekulé-Institut für Organische Chemie und Biochemie, Universität Bonn, Gerhard-Domagk-Strasse 1, D-53121 Bonn, Germany
| |
Collapse
|
28
|
The vacuolar-type ATPase inhibitor archazolid increases tumor cell adhesion to endothelial cells by accumulating extracellular collagen. PLoS One 2018; 13:e0203053. [PMID: 30204757 PMCID: PMC6133348 DOI: 10.1371/journal.pone.0203053] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Accepted: 08/14/2018] [Indexed: 11/19/2022] Open
Abstract
The vacuolar-type H+-ATPase (v-ATPase) is the major proton pump that acidifies intracellular compartments of eukaryotic cells. Since the inhibition of v-ATPase resulted in anti-tumor and anti-metastatic effects in different tumor models, this enzyme has emerged as promising strategy against cancer. Here, we used the well-established v-ATPase inhibitor archazolid, a natural product first isolated from the myxobacterium Archangium gephyra, to study the consequences of v-ATPase inhibition in endothelial cells (ECs), in particular on the interaction between ECs and cancer cells, which has been neglected so far. Human endothelial cells treated with archazolid showed an increased adhesion of tumor cells, whereas the transendothelial migration of tumor cells was reduced. The adhesion process was independent from the EC adhesion molecules ICAM-1, VCAM-1, E-selectin and N-cadherin. Instead, the adhesion was mediated by β1-integrins expressed on tumor cells, as blocking of the integrin β1 subunit reversed this process. Tumor cells preferentially adhered to the β1-integrin ligand collagen and archazolid led to an increase in the amount of collagen on the surface of ECs. The accumulation of collagen was accompanied by a strong decrease of the expression and activity of the protease cathepsin B. Overexpression of cathepsin B in ECs prevented the capability of archazolid to increase the adhesion of tumor cells onto ECs. Our study demonstrates that the inhibition of v-ATPase by archazolid induces a pro-adhesive phenotype in endothelial cells that promotes their interaction with cancer cells, whereas the transmigration of tumor cells was reduced. These findings further support archazolid as a promising anti-metastatic compound.
Collapse
|
29
|
Whitton B, Okamoto H, Packham G, Crabb SJ. Vacuolar ATPase as a potential therapeutic target and mediator of treatment resistance in cancer. Cancer Med 2018; 7:3800-3811. [PMID: 29926527 PMCID: PMC6089187 DOI: 10.1002/cam4.1594] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 05/07/2018] [Accepted: 05/08/2018] [Indexed: 01/10/2023] Open
Abstract
Vacuolar ATPase (V-ATPase) is an ATP-dependent H+ -transporter that pumps protons across intracellular and plasma membranes. It consists of a large multi-subunit protein complex and influences a wide range of cellular processes. This review focuses on emerging evidence for the roles for V-ATPase in cancer. This includes how V-ATPase dysregulation contributes to cancer growth, metastasis, invasion and proliferation, and the potential link between V-ATPase and the development of drug resistance.
Collapse
Affiliation(s)
- Bradleigh Whitton
- Southampton Cancer Research UK CentreUniversity of SouthamptonSouthamptonUK
- Biological SciencesFaculty of Natural and Environmental SciencesUniversity of SouthamptonSouthamptonUK
| | - Haruko Okamoto
- Biological SciencesFaculty of Natural and Environmental SciencesUniversity of SouthamptonSouthamptonUK
| | - Graham Packham
- Southampton Cancer Research UK CentreUniversity of SouthamptonSouthamptonUK
| | - Simon J. Crabb
- Southampton Cancer Research UK CentreUniversity of SouthamptonSouthamptonUK
| |
Collapse
|
30
|
Collins MP, Forgac M. Regulation of V-ATPase Assembly in Nutrient Sensing and Function of V-ATPases in Breast Cancer Metastasis. Front Physiol 2018; 9:902. [PMID: 30057555 PMCID: PMC6053528 DOI: 10.3389/fphys.2018.00902] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Accepted: 06/21/2018] [Indexed: 12/20/2022] Open
Abstract
V-ATPases are proton pumps that function to acidify intracellular compartments in all eukaryotic cells, and to transport protons across the plasma membrane of certain specialized cells. V-ATPases function in many normal and disease processes, including membrane traffic, protein degradation, pathogen entry, and cancer cell invasion. An important mechanism of regulating V-ATPase activity in vivo is regulated assembly, which is the reversible dissociation of the ATP-hydrolytic V1 domain from the proton-conducting V0 domain. Regulated assembly is highly conserved and occurs in response to various nutrient cues, suggesting that it plays an important role in cellular homeostasis. We have recently found that starvation of mammalian cells for either amino acids or glucose increases V-ATPase assembly on lysosomes, possibly to increase protein degradation (for amino acid homeostasis) or for the utilization of alternative energy sources (during glucose starvation). While regulation of assembly in response to amino acid starvation does not involve PI3K or mTORC1, glucose-regulated assembly involves both PI3K and AMPK. Another important form of V-ATPase regulation is the targeting of the enzyme to different cellular membranes, which is controlled by isoforms of subunit a. We have shown that V-ATPases are localized to the plasma membrane of highly invasive breast cancer cells, where they promote cell migration and invasion. Furthermore, overexpression of the a3 isoform is responsible for plasma membrane targeting of V-ATPases in breast tumor cells leading to their increased invasiveness.
Collapse
Affiliation(s)
- Michael P Collins
- Program in Cell, Molecular and Developmental Biology, Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, MA, United States
| | - Michael Forgac
- Program in Cell, Molecular and Developmental Biology, Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, MA, United States.,Department of Developmental, Molecular and Chemical Biology, School of Medicine, Tufts University, Boston, MA, United States
| |
Collapse
|
31
|
Licon-Munoz Y, Fordyce CA, Hayek SR, Parra KJ. V-ATPase-dependent repression of androgen receptor in prostate cancer cells. Oncotarget 2018; 9:28921-28934. [PMID: 29988966 PMCID: PMC6034745 DOI: 10.18632/oncotarget.25641] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Accepted: 06/04/2018] [Indexed: 12/17/2022] Open
Abstract
Prostate Cancer (PCa) is the most commonly diagnosed cancer and the third leading cause of death for men in the United States. Suppression of androgen receptor (AR) expression is a desirable mechanism to manage PCa. Our studies showed that AR expression was reduced in LAPC4 and LNCaP PCa cell lines treated with nanomolar concentrations of the V-ATPase inhibitor concanamycin A (CCA). This treatment decreased PSA mRNA levels, indicative of reduced AR activity. V-ATPase-dependent repression of AR expression was linked to defective endo-lysosomal pH regulation and reduced AR expression at the transcriptional level. CCA treatment increased the protein level and nuclear localization of the alpha subunit of the transcription factor HIF-1 (HIF-1α) in PCa cells via decreased hydroxylation and degradation of HIF-1α. The addition of iron (III) citrate restored HIF-1α hydroxylation and decreased total HIF-1α levels in PCa cells treated with CCA. Moreover, iron treatment partially rescued CCA-mediated AR repression. Dimethyloxalylglycine (DMOG), which prevents HIF-1α degradation independently of V-ATPase, also decreased AR levels, supporting our hypothesis that HIF-1α serves as a downstream mediator in the V-ATPase-AR axis. We propose a new V-ATPase-dependent mechanism to inhibit androgen receptor expression in prostate cancer cells involving defective endosomal trafficking of iron and the inhibition of HIF-1 α-subunit turnover.
Collapse
Affiliation(s)
- Yamhilette Licon-Munoz
- Department of Biochemistry and Molecular Biology, School of Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, 87131, USA
| | - Colleen A Fordyce
- Department of Biochemistry and Molecular Biology, School of Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, 87131, USA
| | - Summer Raines Hayek
- Department of Biochemistry and Molecular Biology, School of Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, 87131, USA
| | - Karlett J Parra
- Department of Biochemistry and Molecular Biology, School of Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, 87131, USA
| |
Collapse
|
32
|
Bartel K, Winzi M, Ulrich M, Koeberle A, Menche D, Werz O, Müller R, Guck J, Vollmar AM, von Schwarzenberg K. V-ATPase inhibition increases cancer cell stiffness and blocks membrane related Ras signaling - a new option for HCC therapy. Oncotarget 2018; 8:9476-9487. [PMID: 28036299 PMCID: PMC5354746 DOI: 10.18632/oncotarget.14339] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 12/12/2016] [Indexed: 11/25/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the fifth most frequent cancer worldwide and the third leading cause of cancer-related death. However, therapy options are limited leaving an urgent need to develop new strategies. Currently, targeting cancer cell lipid and cholesterol metabolism is gaining interest especially regarding HCC. High cholesterol levels support proliferation, membrane-related mitogenic signaling and increase cell softness, leading to tumor progression, malignancy and invasive potential. However, effective ways to target cholesterol metabolism for cancer therapy are still missing. The V-ATPase inhibitor archazolid was recently shown to interfere with cholesterol metabolism. In our study, we report a novel therapeutic potential of V-ATPase inhibition in HCC by altering the mechanical phenotype of cancer cells leading to reduced proliferative signaling. Archazolid causes cellular depletion of free cholesterol leading to an increase in cell stiffness and membrane polarity of cancer cells, while hepatocytes remain unaffected. The altered membrane composition decreases membrane fluidity and leads to an inhibition of membrane-related Ras signaling resulting decreased proliferation in vitro and in vivo. V-ATPase inhibition represents a novel link between cell biophysical properties and proliferative signaling selectively in malignant HCC cells, providing the basis for an attractive and innovative strategy against HCC.
Collapse
Affiliation(s)
- Karin Bartel
- Department of Pharmacy, Pharmaceutical Biology, Ludwig-Maximilians-University of Munich, 81377 Munich, Germany
| | - Maria Winzi
- Biotechnology Center, Technische Universität Dresden, 01307 Dresden, Germany
| | - Melanie Ulrich
- Department of Pharmacy, Pharmaceutical Biology, Ludwig-Maximilians-University of Munich, 81377 Munich, Germany
| | - Andreas Koeberle
- Chair of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, 07743 Jena, Germany
| | - Dirk Menche
- Kekulé Institute of Organic Chemistry and Biochemistry, University of Bonn, 53121 Bonn, Germany
| | - Oliver Werz
- Chair of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, 07743 Jena, Germany
| | - Rolf Müller
- Department of Microbial Natural Products, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) - Helmholtz Centre for Infection Research (HZI), Saarland University, 66123 Saarbrücken, Germany
| | - Jochen Guck
- Biotechnology Center, Technische Universität Dresden, 01307 Dresden, Germany
| | - Angelika M Vollmar
- Department of Pharmacy, Pharmaceutical Biology, Ludwig-Maximilians-University of Munich, 81377 Munich, Germany
| | - Karin von Schwarzenberg
- Department of Pharmacy, Pharmaceutical Biology, Ludwig-Maximilians-University of Munich, 81377 Munich, Germany
| |
Collapse
|
33
|
Pamarthy S, Kulshrestha A, Katara GK, Beaman KD. The curious case of vacuolar ATPase: regulation of signaling pathways. Mol Cancer 2018; 17:41. [PMID: 29448933 PMCID: PMC5815226 DOI: 10.1186/s12943-018-0811-3] [Citation(s) in RCA: 121] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Accepted: 02/07/2018] [Indexed: 02/06/2023] Open
Abstract
The Vacuolar ATPase (V-ATPase) is a proton pump responsible for controlling the intracellular and extracellular pH of cells. The structure of V-ATPase has been highly conserved among all eukaryotic cells and is involved in diverse functions across species. V-ATPase is best known for its acidification of endosomes and lysosomes and is also important for luminal acidification of specialized cells. Several reports have suggested the involvement of V-ATPase in maintaining an alkaline intracellular and acidic extracellular pH thereby aiding in proliferation and metastasis of cancer cells respectively. Increased expression of V-ATPase and relocation to the plasma membrane aids in cancer modulates key tumorigenic cell processes like autophagy, Warburg effect, immunomoduation, drug resistance and most importantly cancer cell signaling. In this review, we discuss the direct role of V-ATPase in acidification and indirect regulation of signaling pathways, particularly Notch Signaling.
Collapse
Affiliation(s)
- Sahithi Pamarthy
- Department of Microbiology and Immunology, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL, 60064, USA
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL, 60611, USA
| | - Arpita Kulshrestha
- Department of Microbiology and Immunology, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL, 60064, USA
| | - Gajendra K Katara
- Department of Microbiology and Immunology, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL, 60064, USA
| | - Kenneth D Beaman
- Department of Microbiology and Immunology, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL, 60064, USA.
| |
Collapse
|
34
|
Pereira CS, Guedes JP, Gonçalves M, Loureiro L, Castro L, Gerós H, Rodrigues LR, Côrte-Real M. Lactoferrin selectively triggers apoptosis in highly metastatic breast cancer cells through inhibition of plasmalemmal V-H+-ATPase. Oncotarget 2018; 7:62144-62158. [PMID: 27556694 PMCID: PMC5308717 DOI: 10.18632/oncotarget.11394] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Accepted: 08/08/2016] [Indexed: 01/12/2023] Open
Abstract
Breast cancer is the most common type of cancer affecting women. Despite the good prognosis when detected early, significant challenges remain in the treatment of metastatic breast cancer. The recruitment of the vacuolar H+-ATPase (V-H+-ATPase) to the plasma membrane, where it mediates the acidification of the tumor microenvironment (TME), is a recognized feature involved in the acquisition of a metastatic phenotype in breast cancer. Therefore, inhibitors of this pump have emerged as promising anticancer drugs. Lactoferrin (Lf) is a natural pro-apoptotic iron-binding glycoprotein with strong anticancer activity whose mechanism of action is not fully understood. Here, we show that bovine Lf (bLf) preferentially induces apoptosis in the highly metastatic breast cancer cell lines Hs 578T and MDA-MB-231, which display a prominent localisation of V-H+-ATPase at the plasma membrane, but not in the lowly metastatic T-47D or in the non-tumorigenic MCF-10-2A cell lines. We also demonstrate that bLf decreases the extracellular acidification rate and causes intracellular acidification in metastatic breast cancer cells and, much like the well-known proton pump inhibitors concanamycin A and bafilomycin A1, inhibits V-H+-ATPase in sub-cellular fractions. These data further support that bLf targets V-H+-ATPase and explain the selectivity of bLf for cancer cells, especially for highly metastatic breast cancer cells. Altogether, our results pave the way for more rational in vivo studies aiming to explore this natural non-toxic compound for metastatic breast cancer therapy.
Collapse
Affiliation(s)
- Cátia S Pereira
- Centre of Molecular and Environmental Biology (CBMA), Department of Biology, University of Minho, Braga, Portugal.,Centre of Biological Engineering (CEB), Department of Biological Engineering, University of Minho, Braga, Portugal
| | - Joana P Guedes
- Centre of Molecular and Environmental Biology (CBMA), Department of Biology, University of Minho, Braga, Portugal.,Centre of Biological Engineering (CEB), Department of Biological Engineering, University of Minho, Braga, Portugal
| | - Marília Gonçalves
- Centre of Molecular and Environmental Biology (CBMA), Department of Biology, University of Minho, Braga, Portugal
| | - Luís Loureiro
- Centre of Molecular and Environmental Biology (CBMA), Department of Biology, University of Minho, Braga, Portugal
| | - Lisandra Castro
- Centre of Molecular and Environmental Biology (CBMA), Department of Biology, University of Minho, Braga, Portugal
| | - Hernâni Gerós
- Centre of Molecular and Environmental Biology (CBMA), Department of Biology, University of Minho, Braga, Portugal.,Centre for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB), Department of Biology, University of Minho, Braga, Portugal
| | - Lígia R Rodrigues
- Centre of Biological Engineering (CEB), Department of Biological Engineering, University of Minho, Braga, Portugal
| | - Manuela Côrte-Real
- Centre of Molecular and Environmental Biology (CBMA), Department of Biology, University of Minho, Braga, Portugal
| |
Collapse
|
35
|
The a3 isoform of subunit a of the vacuolar ATPase localizes to the plasma membrane of invasive breast tumor cells and is overexpressed in human breast cancer. Oncotarget 2018; 7:46142-46157. [PMID: 27323815 PMCID: PMC5216787 DOI: 10.18632/oncotarget.10063] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Accepted: 06/02/2016] [Indexed: 12/11/2022] Open
Abstract
The vacuolar (H+)-ATPases (V-ATPases) are a family of ATP-driven proton pumps that acidify intracellular compartments and transport protons across the plasma membrane. Previous work has demonstrated that plasma membrane V-ATPases are important for breast cancer invasion in vitro and that the V-ATPase subunit a isoform a3 is upregulated in and critical for MDA-MB231 and MCF10CA1a breast cancer cell invasion. It has been proposed that subunit a3 is present on the plasma membrane of invasive breast cancer cells and is overexpressed in human breast cancer. To test this, we used an a3-specific antibody to assess localization in breast cancer cells. Subunit a3 localizes to the leading edge of migrating breast cancer cells, but not the plasma membrane of normal breast epithelial cells. Furthermore, invasive breast cancer cells express a3 throughout all intracellular compartments tested, including endosomes, the Golgi, and lysosomes. Moreover, subunit a3 knockdown in MB231 breast cancer cells reduces in vitro migration. This reduction is not enhanced upon addition of a V-ATPase inhibitor, suggesting that a3-containing V-ATPases are critical for breast cancer migration. Finally, we have tested a3 expression in human breast cancer tissue and mRNA prepared from normal and cancerous breast tissue. a3 mRNA was upregulated 2.5-47 fold in all breast tumor cDNA samples tested relative to normal tissue, with expression generally correlated to cancer stage. Furthermore, a3 protein expression was increased in invasive breast cancer tissue relative to noninvasive cancer and normal breast tissue. These studies suggest that subunit a3 plays an important role in invasive human breast cancer.
Collapse
|
36
|
Endolysosomal Cation Channels and Cancer-A Link with Great Potential. Pharmaceuticals (Basel) 2018; 11:ph11010004. [PMID: 29303993 PMCID: PMC5874700 DOI: 10.3390/ph11010004] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 01/01/2018] [Accepted: 01/04/2018] [Indexed: 12/21/2022] Open
Abstract
The endolysosomal system (ES) consists of lysosomes; early, late, and recycling endosomes; and autophagosomes. It is a key regulator not only of macromolecule degradation and recycling, plasma membrane repair, homeostasis, and lipid storage, but also of antigen presentation, immune defense, cell motility, cell death signaling, tumor growth, and cancer progression. In addition, it plays a critical role in autophagy, and the autophagy-lysosome pathway is intimately associated with the hallmarks of cancer, such as escaping cell death pathways, evading immune surveillance, and deregulating metabolism. The function of endolysosomes is critically dependent on both soluble and endolysosomal membrane proteins such as ion channels and transporters. Cation channels found in the ES include members of the TRP (transient receptor potential) channel superfamily, namely TRPML channels (mucolipins) as well as two-pore channels (TPCs). In recent studies, these channels have been found to play crucial roles in endolysosomal trafficking, lysosomal exocytosis, and autophagy. Mutation or loss of these channel proteins can impact multiple endolysosomal trafficking pathways. A role for TPCs in cancer cell migration and metastasis, linked to distinct defects in endolysosomal trafficking such as integrin trafficking, has been recently established. In this review, we give an overview on the function of lysosomes in cancer with a particular focus on the roles which TPCs and TRPML channels play in the ES and how this can affect cancer cells.
Collapse
|
37
|
Rethinking the Combination of Proton Exchanger Inhibitors in Cancer Therapy. Metabolites 2017; 8:metabo8010002. [PMID: 29295495 PMCID: PMC5875992 DOI: 10.3390/metabo8010002] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 12/16/2017] [Accepted: 12/21/2017] [Indexed: 12/22/2022] Open
Abstract
Microenvironmental acidity is becoming a key target for the new age of cancer treatment. In fact, while cancer is characterized by genetic heterogeneity, extracellular acidity is a common phenotype of almost all cancers. To survive and proliferate under acidic conditions, tumor cells up-regulate proton exchangers and transporters (mainly V-ATPase, Na+/H+ exchanger (NHE), monocarboxylate transporters (MCTs), and carbonic anhydrases (CAs)), that actively extrude excess protons, avoiding intracellular accumulation of toxic molecules, thus becoming a sort of survival option with many similarities compared with unicellular microorganisms. These systems are also involved in the unresponsiveness or resistance to chemotherapy, leading to the protection of cancer cells from the vast majority of drugs, that when protonated in the acidic tumor microenvironment, do not enter into cancer cells. Indeed, as usually occurs in the progression versus malignancy, resistant tumor clones emerge and proliferate, following a transient initial response to a therapy, thus giving rise to more malignant behavior and rapid tumor progression. Recent studies are supporting the use of a cocktail of proton exchanger inhibitors as a new strategy against cancer.
Collapse
|
38
|
Tumor cell cholesterol depletion and V-ATPase inhibition as an inhibitory mechanism to prevent cell migration and invasiveness in melanoma. Biochim Biophys Acta Gen Subj 2017; 1862:684-691. [PMID: 29253593 DOI: 10.1016/j.bbagen.2017.12.006] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Revised: 11/18/2017] [Accepted: 12/13/2017] [Indexed: 01/01/2023]
Abstract
BACKGROUND V-ATPase interactions with cholesterol enriched membrane microdomains have been related to metastasis in a variety of cancers, but the underlying mechanism remains at its beginnings. It has recently been reported that the inhibition of this H+ pump affects cholesterol mobilization to the plasma membrane. METHODS Inhibition of melanoma cell migration and invasiveness was assessed by wound healing and Transwell assays in murine cell lines (B16F10 and Melan-A). V-ATPase activity was measured in vitro by ATP hydrolysis and H+ transport in membrane vesicles, and intact cell H+ fluxes were measured by using a non-invasive Scanning Ion-selective Electrode Technique (SIET). RESULTS Cholesterol depletion by 5mM MβCD was found to be inhibitory to the hydrolytic and H+ pumping activities of the V-ATPase of melanoma cell lines, as well as to the migration and invasiveness capacities of these cells. Nearly the same effects were obtained using concanamycin A, a specific inhibitor of V-ATPase, which also promoted a decrease of the H+ efflux in live cells at the same extent of MβCD. CONCLUSIONS We found that cholesterol depletion significantly affects the V-ATPase activity and the initial metastatic processes following a profile similar to those observed in the presence of the V-ATPase specific inhibitor, concanamycin. GENERAL SIGNIFICANCE The results shed new light on the functional role of the interactions between V-ATPases and cholesterol-enriched microdomains of cell membranes that contribute with malignant phenotypes in melanoma.
Collapse
|
39
|
Kessler SM, Lederer E, Laggai S, Golob-Schwarzl N, Hosseini K, Petzold J, Schweiger C, Reihs R, Keil M, Hoffmann J, Mayr C, Kiesslich T, Pichler M, Kim KS, Rhee H, Park YN, Lax S, Obrist P, Kiemer AK, Haybaeck J. IMP2/IGF2BP2 expression, but not IMP1 and IMP3, predicts poor outcome in patients and high tumor growth rate in xenograft models of gallbladder cancer. Oncotarget 2017; 8:89736-89745. [PMID: 29163784 PMCID: PMC5685705 DOI: 10.18632/oncotarget.21116] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Accepted: 09/04/2017] [Indexed: 12/24/2022] Open
Abstract
Overexpression of the oncofetal insulin-like growth factor 2 mRNA-binding protein 2 (IMP2/IGF2BP2) has been described in different cancer types. Gallbladder carcinoma (GBC) is a rare but highly aggressive cancer entity with late clinical detection and poor prognosis. The aim of this study was to investigate the role of IMP2 in human GBC. Tissue microarrays (TMAs) of an international multi-center GBC sample collection from n = 483 patients were analyzed by immunohistochemistry. IMP2 immunoreactivity was found in 74.3% of the tumor samples on TMA, of which 14.0% showed strong and 86.0% low staining intensity. 72.4% of the tumor samples were IMP1 positive, but IMP1 showed lower expression in tumor tissue compared to control tissues. IMP3 immunoreactivity was observed in 92.7% of all tumors, of which 53.6% revealed strong IMP3 expression. Kaplan-Meier analysis linked high IMP2 expression to shorter survival time (p = 0.033), whereas neither IMP1 nor IMP3 expression was linked to a decreased survival time. Eight different human biliary tract cancer (BTC) cell lines were evaluated for tumor growth kinetics in mouse xenografts. Cell lines with high IMP2 expression levels showed the fastest increase in tumor volumes in murine xenografts. Furthermore, IMP2 expression in these cells correlated with the generation of reactive oxygen species (ROS) and RAC1 expression in BTC cells, suggesting RAC1-induced ROS generation as a potential mechanism of IMP2-promoted progression of GBC. In conclusion, IMP2 is frequently overexpressed in GBC and significantly associated with poor prognosis and growth rates in vivo. IMP2 might therefore represent a new target for the treatment of advanced GBC.
Collapse
Affiliation(s)
- Sonja M Kessler
- Institute of Pathology, Medical University of Graz, Graz, Austria.,Department of Pharmacy, Pharmaceutical Biology, Saarland University, Saarbrücken, Germany
| | - Eva Lederer
- Institute of Pathology, Medical University of Graz, Graz, Austria
| | - Stephan Laggai
- Department of Pharmacy, Pharmaceutical Biology, Saarland University, Saarbrücken, Germany
| | - Nicole Golob-Schwarzl
- Institute of Pathology, Medical University of Graz, Graz, Austria.,Center for Biomarker Research in Medicine, Graz, Austria
| | - Kevan Hosseini
- Department of Pharmacy, Pharmaceutical Biology, Saarland University, Saarbrücken, Germany
| | - Johannes Petzold
- Institute of Pathology, Medical University of Graz, Graz, Austria
| | | | - Robert Reihs
- Institute of Pathology, Medical University of Graz, Graz, Austria
| | - Marlen Keil
- Experimental Pharmacology and Oncology Berlin-Buch GmbH, Berlin, Germany
| | - Jens Hoffmann
- Experimental Pharmacology and Oncology Berlin-Buch GmbH, Berlin, Germany
| | - Christian Mayr
- Laboratory for Tumor Biology and Experimental Therapies, Institute of Physiology and Pathophysiology, Department of Internal Medicine I, Paracelsus Medical University, Salzburg, Austria
| | - Tobias Kiesslich
- Laboratory for Tumor Biology and Experimental Therapies, Institute of Physiology and Pathophysiology, Department of Internal Medicine I, Paracelsus Medical University, Salzburg, Austria
| | - Martin Pichler
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, Graz, Austria.,Research Unit for Non-coding RNAs and Genome Editing in Cancer, Medical University of Graz, Graz, Austria.,Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - Kyung Sik Kim
- Department of Surgery, Yonsei University College of Medicine, Seoul, South Korea
| | - Hyungjin Rhee
- Department of Radiology, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Young Nyun Park
- Department of Pathology, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Sigurd Lax
- Department of Pathology, General Hospital Graz Sued-West, Graz, Austria
| | - Peter Obrist
- Laboratory of Pathology, Dr. Obrist and Dr. Brunhuber OG, Zams, Austria
| | - Alexandra K Kiemer
- Department of Pharmacy, Pharmaceutical Biology, Saarland University, Saarbrücken, Germany
| | - Johannes Haybaeck
- Institute of Pathology, Medical University of Graz, Graz, Austria.,Department of Pathology, Medical Faculty, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| |
Collapse
|
40
|
Pan S, Cai H, Gu L, Cao S. Cleistanthin A inhibits the invasion and metastasis of human melanoma cells by inhibiting the expression of matrix metallopeptidase-2 and -9. Oncol Lett 2017; 14:6217-6223. [PMID: 29113270 DOI: 10.3892/ol.2017.6917] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Accepted: 07/07/2017] [Indexed: 01/01/2023] Open
Abstract
It has been demonstrated that numerous types of metastatic cancer overexpress vacuolar-type H+ (V)-ATPases. It may be possible to inhibit the growth and metastasis of human cancer cells by inhibiting V-ATPases. It was previously reported that diphyllin, a novel V-ATPase inhibitor, can inhibit the migration and invasion of SGC7901 human gastric cancer cells; however, the effects of cleistanthin A (CA), a diphyllin glycoside, on melanoma cells has not been demonstrated. The present study aimed to investigate the effect of CA as a V-ATPase inhibitor and its effects on the invasion and metastasis of A375 cells. The results of an MTT assay in the present study indicated that the growth inhibition of A375 cells by CA was induced in a dose- and time-dependent manner; however, A375 cell viability was not significantly affected by low concentrations (0.03, 0.1 and 0.3 µM) after 24 h. Similar results were obtained by viable cell counting with trypan blue. Therefore, these concentrations of CA were selected for the treatment of A375 cells in further experiments. It was demonstrated that CA inhibited the expression of V-ATPases in a dose-dependent manner and decreased the internal pH level of A375 cells. Alterations to the lysosomal pH were associated with the CA concentration. Furthermore, CA treatment induced a significant decrease in cell migration and invasion, as demonstrated with wound-healing and Transwell assays. Gelatin zymography and western blot analysis demonstrated that the expression levels of matrix metallopeptidase (MMP)-2 and -9 decreased following CA treatment. Therefore, CA can be characterized as a novel V-ATPase inhibitor for the treatment of melanoma that may inhibit invasion and metastasis by downregulating the expression of MMP-2 and -9.
Collapse
Affiliation(s)
- Sheng Pan
- Department of Dermatology and Venereology, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China.,Institute of Nautical Medicine, Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Hengji Cai
- Department of Dermatology and Venereology, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Lixiong Gu
- Department of Dermatology and Venereology, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Shuanglin Cao
- Department of Dermatology and Venereology, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| |
Collapse
|
41
|
Lu ZN, Tian B, Guo XL. Repositioning of proton pump inhibitors in cancer therapy. Cancer Chemother Pharmacol 2017; 80:925-937. [PMID: 28861639 DOI: 10.1007/s00280-017-3426-2] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Accepted: 08/24/2017] [Indexed: 12/12/2022]
Abstract
Drug repositioning, as a smart way to exploit new molecular targets of a known drug, has been gaining increasing attention in the discovery of anti-cancer drugs. Proton pump inhibitors (PPIs) as benzimidazole derivatives, which are essentially H+-K+-ATPases inhibitors, are commonly used in the treatment of acid-related diseases such as gastric ulcer. In recent years, exploring the new application of PPIs in anti-cancer field has become a hot research topic. Interestingly, cancer cells display an alkaline intracellular pH and an acidic extracellular pH. The extracellular acidity of tumors can be corrected by PPIs that are selectively activated in an acid milieu. It is generally believed that PPIs might provoke disruption of pH homeostasis by targeting V-ATPase on cancer cells, which is the theoretical basis for PPIs to play an anti-cancer role. Numerous studies have shown specialized effects of the PPIs on tumor cell growth, metastasis, chemoresistance, and autophagy. PPIs may really represent new anti-cancer drugs due to better safety and tolerance, the potential selectivity in targeting tumor acidity, and the ability to inhibit mechanism pivotal for cancer homeostasis. In this review, we focus on the new therapeutic applications of PPIs in multiple cancers, explaining the rationale behind this approach and providing practical evidence.
Collapse
Affiliation(s)
- Zhen-Ning Lu
- Department of Pharmacology, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, No. 44 Wen Hua Xi Road, Jinan, 250012, People's Republic of China
| | - Bing Tian
- Department of Pharmacology, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, No. 44 Wen Hua Xi Road, Jinan, 250012, People's Republic of China
| | - Xiu-Li Guo
- Department of Pharmacology, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, No. 44 Wen Hua Xi Road, Jinan, 250012, People's Republic of China.
| |
Collapse
|
42
|
Merk H, Messer P, Ardelt MA, Lamb DC, Zahler S, Müller R, Vollmar AM, Pachmayr J. Inhibition of the V-ATPase by Archazolid A: A New Strategy to Inhibit EMT. Mol Cancer Ther 2017; 16:2329-2339. [DOI: 10.1158/1535-7163.mct-17-0129] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Revised: 06/21/2017] [Accepted: 06/26/2017] [Indexed: 11/16/2022]
|
43
|
Asgharzadeh MR, Barar J, Pourseif MM, Eskandani M, Jafari Niya M, Mashayekhi MR, Omidi Y. Molecular machineries of pH dysregulation in tumor microenvironment: potential targets for cancer therapy. BIOIMPACTS : BI 2017; 7:115-133. [PMID: 28752076 PMCID: PMC5524986 DOI: 10.15171/bi.2017.15] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Revised: 05/28/2017] [Accepted: 06/06/2017] [Indexed: 12/30/2022]
Abstract
Introduction: Cancer is an intricate disorder/dysfunction of cells that can be defined as a genetic heterogeneity in human disease. Therefore, it is characterized by several adaptive complex hallmarks. Among them, the pH dysregulation appears as a symbol of aberrant functions within the tumor microenvironment (TME). In comparison with normal tissues, in the solid tumors, we face with an irregular acidification and alkalinization of the extracellular and intracellular fluids. Methods: In this study, we comprehensively discussed the most recent reports on the hallmarks of solid tumors to provide deep insights upon the molecular machineries involved in the pH dysregulation of solid tumors and their impacts on the initiation and progression of cancer. Results: The dysregulation of pH in solid tumors is fundamentally related to the Warburg effect and hypoxia, leading to expression of a number of molecular machineries, including: NHE1, H+ pump V-ATPase, CA-9, CA-12, MCT-1, GLUT-1. Activation of proton exchangers and transporters (PETs) gives rise to formation of TME. This condition favors the cancer cells to evade from the anoikis and apoptosis, granting them aggressive and metastasis phenotype, as well as resistance to chemotherapy and radiation therapy. This review aimed to discuss the key molecular changes of tumor cells in terms of bio-energetics and cancer metabolism in relation with pH dysregulation. During this phenomenon, the intra- and extracellular metabolites are altered and/or disrupted. Such molecular alterations provide molecular hallmarks for direct targeting of the PETs by potent relevant inhibitors in combination with conventional cancer therapies as ultimate therapy against solid tumors. Conclusion: Taken all, along with other treatment strategies, targeting the key molecular machineries related to intra- and extracellular metabolisms within the TME is proposed as a novel strategy to inhibit or block PETs that are involved in the pH dysregulation of solid tumors.
Collapse
Affiliation(s)
- Mohammad Reza Asgharzadeh
- Department of Biology, Fars Science and Research Branch, Islamic Azad University, Marvdasht, Iran
- Department of Biology, Marvdasht Branch, Islamic Azad University, Marvdasht, Iran
| | - Jaleh Barar
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad M. Pourseif
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Morteza Eskandani
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mojtaba Jafari Niya
- Department of Biology, Fars Science and Research Branch, Islamic Azad University, Marvdasht, Iran
- Department of Biology, Marvdasht Branch, Islamic Azad University, Marvdasht, Iran
| | | | - Yadollah Omidi
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
44
|
Scheeff S, Menche D. Total syntheses of the archazolids: an emerging class of novel anticancer drugs. Beilstein J Org Chem 2017; 13:1085-1098. [PMID: 28684988 PMCID: PMC5480361 DOI: 10.3762/bjoc.13.108] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 05/09/2017] [Indexed: 12/19/2022] Open
Abstract
V-ATPase has recently emerged as a promising novel anticancer target based on extensive in vitro and in vivo studies with the archazolids, complex polyketide macrolides which present the most potent V-ATPase inhibitors known to date, rendering these macrolides important lead structures for the development of novel anticancer agents. The limited natural supply of these metabolites from their myxobacterial source renders total synthesis of vital importance for the further preclinical development. This review describes in detail the various tactics and strategies employed so far in archazolid syntheses that culminated in three total syntheses and discusses the future synthetic challenges that have to be addressed.
Collapse
Affiliation(s)
- Stephan Scheeff
- Kekulé-Institut für Organische Chemie und Biochemie, Universität Bonn, Gerhard-Domagk-Strasse 1, 53121 Bonn, Germany
| | - Dirk Menche
- Kekulé-Institut für Organische Chemie und Biochemie, Universität Bonn, Gerhard-Domagk-Strasse 1, 53121 Bonn, Germany
| |
Collapse
|
45
|
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
|
46
|
Selective upregulation of TNFα expression in classically-activated human monocyte-derived macrophages (M1) through pharmacological interference with V-ATPase. Biochem Pharmacol 2017; 130:71-82. [DOI: 10.1016/j.bcp.2017.02.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 02/01/2017] [Indexed: 11/21/2022]
|
47
|
Nguyen ONP, Grimm C, Schneider LS, Chao YK, Atzberger C, Bartel K, Watermann A, Ulrich M, Mayr D, Wahl-Schott C, Biel M, Vollmar AM. Two-Pore Channel Function Is Crucial for the Migration of Invasive Cancer Cells. Cancer Res 2017; 77:1427-1438. [PMID: 28108508 DOI: 10.1158/0008-5472.can-16-0852] [Citation(s) in RCA: 105] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Revised: 12/22/2016] [Accepted: 12/22/2016] [Indexed: 11/16/2022]
Abstract
Metastatic invasion is the major cause of cancer-related deaths. In this study, we introduce two-pore channels (TPC), a recently described class of NAADP- and PI(3,5)P2-sensitive Ca2+-permeable cation channels in the endolysosomal system of cells, as candidate targets for the treatment of invasive cancers. Inhibition of the channel abrogated migration of metastatic cancer cells in vitro Silencing or pharmacologic inhibition of the two-pore channel TPC2 reduced lung metastasis of mammary mouse cancer cells. Disrupting TPC function halted trafficking of β1-integrin, leading to its accumulation in EEA1-positive early endosomes. As a consequence, invasive cancer cells were no longer able to form leading edges, which are required for adequate migration. Our findings link TPC to cancer cell migration and provide a preclinical proof of concept for their candidacy as targets to treat metastatic cancers. Cancer Res; 77(6); 1427-38. ©2017 AACR.
Collapse
Affiliation(s)
- Ong Nam Phuong Nguyen
- Department of Pharmacy, Pharmaceutical Biology, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Christian Grimm
- Department of Pharmacy, Center for Drug Research and Center for Integrated Protein Science Munich (CIPSM), Ludwig-Maximilians-University Munich, Munich, Germany
| | - Lina S Schneider
- Department of Pharmacy, Pharmaceutical Biology, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Yu-Kai Chao
- Department of Pharmacy, Center for Drug Research and Center for Integrated Protein Science Munich (CIPSM), Ludwig-Maximilians-University Munich, Munich, Germany
| | - Carina Atzberger
- Department of Pharmacy, Pharmaceutical Biology, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Karin Bartel
- Department of Pharmacy, Pharmaceutical Biology, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Anna Watermann
- Department of Pharmacy, Pharmaceutical Biology, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Melanie Ulrich
- Department of Pharmacy, Pharmaceutical Biology, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Doris Mayr
- Pathological Institute, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Christian Wahl-Schott
- Department of Pharmacy, Center for Drug Research and Center for Integrated Protein Science Munich (CIPSM), Ludwig-Maximilians-University Munich, Munich, Germany
| | - Martin Biel
- Department of Pharmacy, Center for Drug Research and Center for Integrated Protein Science Munich (CIPSM), Ludwig-Maximilians-University Munich, Munich, Germany.
| | - Angelika M Vollmar
- Department of Pharmacy, Pharmaceutical Biology, Ludwig-Maximilians-University Munich, Munich, Germany.
| |
Collapse
|
48
|
Herrmann J, Fayad AA, Müller R. Natural products from myxobacteria: novel metabolites and bioactivities. Nat Prod Rep 2016; 34:135-160. [PMID: 27907217 DOI: 10.1039/c6np00106h] [Citation(s) in RCA: 152] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Covering: 2011-July 2016Myxobacteria are a rich source for structurally diverse secondary metabolites with intriguing biological activities. Here we report on new natural products that were isolated from myxobacteria in the period of 2011 to July 2016. Some examples of recent advances on modes-of-action are also summarised along with a more detailed overview on five compound classes currently assessed in preclinical studies.
Collapse
Affiliation(s)
- J Herrmann
- Helmholtz Institute for Pharmaceutical Research Saarland, Department of Microbial Natural Products, Helmholtz Centre for Infection Research and Department of Pharmaceutical Biotechnology, Saarland University, Campus E8.1, 66123 Saarbrücken, Germany.
| | | | | |
Collapse
|
49
|
Zhang S, Schneider LS, Vick B, Grunert M, Jeremias I, Menche D, Müller R, Vollmar AM, Liebl J. Anti-leukemic effects of the V-ATPase inhibitor Archazolid A. Oncotarget 2016; 6:43508-28. [PMID: 26496038 PMCID: PMC4791247 DOI: 10.18632/oncotarget.6180] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Accepted: 10/07/2015] [Indexed: 12/31/2022] Open
Abstract
Prognosis for patients suffering from T-ALL is still very poor and new strategies for T-ALL treatment are urgently needed. Our study shows potent anti-leukemic effects of the myxobacterial V-ATPase inhibitor Archazolid A. Archazolid A reduced growth and potently induced death of leukemic cell lines and human leukemic samples. By inhibiting lysosomal acidification, Archazolid A blocked activation of the Notch pathway, however, this was not the mechanism of V-ATPase inhibition relevant for cell death induction. In fact, V-ATPase inhibition by Archazolid A decreased the anti-apoptotic protein survivin. As underlying mode of action, this work is in line with recent studies from our group demonstrating that Archazolid A induced S-phase cell cycle arrest by interfering with the iron metabolism in leukemic cells. Our study provides evidence for V-ATPase inhibition as a potential new therapeutic option for T-ALL.
Collapse
Affiliation(s)
- Siwei Zhang
- Department of Pharmacy, Pharmaceutical Biology, Ludwig-Maximilians-University, Munich, Germany
| | - Lina S Schneider
- Department of Pharmacy, Pharmaceutical Biology, Ludwig-Maximilians-University, Munich, Germany
| | - Binje Vick
- Department of Gene Vectors, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich, Germany.,German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Michaela Grunert
- Department of Gene Vectors, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich, Germany
| | - Irmela Jeremias
- Department of Gene Vectors, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich, Germany.,Department of Oncology/Hematology, Dr. von Haunersches Kinderspital, Munich, Germany.,German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Dirk Menche
- Kekulé-Institut für Organische Chemie und Biochemie der Universität Bonn, Bonn, Germany
| | - Rolf Müller
- Helmholtz Institute for Pharmaceutical Research Saarland, Helmholtz Centre for Infection Research and Department of Pharmaceutical Biotechnology, Saarland University, Saarbrücken, Germany
| | - Angelika M Vollmar
- Department of Pharmacy, Pharmaceutical Biology, Ludwig-Maximilians-University, Munich, Germany
| | - Johanna Liebl
- Department of Pharmacy, Pharmaceutical Biology, Ludwig-Maximilians-University, Munich, Germany
| |
Collapse
|
50
|
Ibrahim SA, Katara GK, Kulshrestha A, Jaiswal MK, Amin MA, Beaman KD. Breast cancer associated a2 isoform vacuolar ATPase immunomodulates neutrophils: potential role in tumor progression. Oncotarget 2016; 6:33033-45. [PMID: 26460736 PMCID: PMC4741747 DOI: 10.18632/oncotarget.5439] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Accepted: 09/29/2015] [Indexed: 12/22/2022] Open
Abstract
In invasive breast cancer, tumor associated neutrophils (TAN) represent a significant portion of the tumor mass and are associated with increased angiogenesis and metastasis. Identifying the regulatory factors that control TAN behavior will help in developing ideal immunotherapies. Vacuolar ATPases (V-ATPases), multi-subunit proton pumps, are highly expressed in metastatic breast cancer cells. A cleaved peptide from a2 isoform V-ATPase (a2NTD) has immunomodulatory role in tumor microenvironment. Here, we report for the first time the role of V-ATPase in neutrophils modulation. In invasive breast cancer cells, a2NTD was detected and a2V was highly expressed on the surface. Immunohistochemical analysis of invasive breast cancer tissues revealed that increased neutrophil recruitment and blood vessel density correlated with increased a2NTD levels. In order to determine the direct regulatory role of a2NTD on neutrophils, recombinant a2NTD was used for the treatment of neutrophils isolated from the peripheral blood of healthy volunteers. Neutrophils treated with a2NTD (a2Neuɸ) showed increased secretion of IL-1RA, IL-10, CCL-2 and IL-6 that are important mediators in cancer related inflammation. Moreover, a2Neuɸ exhibited an increased production of protumorigenic factors including IL-8, matrix metaloprotinase-9 and vascular endothelial growth factor. Further, functional characterization of a2Neuɸ revealed that a2Neuɸ derived products induce in vitro angiogenesis as well as increase the invasiveness of breast cancer cells. This study establishes the modulatory effect of breast cancer associated a2V on neutrophils, by the action of a2NTD, which has a positive impact on tumor progression, supporting that a2V can be a potential selective target for breast cancer therapy.
Collapse
Affiliation(s)
- Safaa A Ibrahim
- Department of Microbiology and Immunology, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA.,Department of Microbiology and Immunology, Faculty of Pharmacy, Cairo University, Giza, Egypt
| | - Gajendra K Katara
- Department of Microbiology and Immunology, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Arpita Kulshrestha
- 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
| | - Magdy A Amin
- Department of Microbiology and Immunology, Faculty of Pharmacy, Cairo University, Giza, Egypt
| | - Kenneth D Beaman
- Department of Microbiology and Immunology, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| |
Collapse
|