1
|
Russell S, Xu L, Kam Y, Abrahams D, Ordway B, Lopez AS, Bui MM, Johnson J, Epstein T, Ruiz E, Lloyd MC, Swietach P, Verduzco D, Wojtkowiak J, Gillies RJ. Proton export upregulates aerobic glycolysis. BMC Biol 2022; 20:163. [PMID: 35840963 PMCID: PMC9287933 DOI: 10.1186/s12915-022-01340-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Accepted: 05/30/2022] [Indexed: 01/06/2023] Open
Abstract
INTRODUCTION Aggressive cancers commonly ferment glucose to lactic acid at high rates, even in the presence of oxygen. This is known as aerobic glycolysis, or the "Warburg Effect." It is widely assumed that this is a consequence of the upregulation of glycolytic enzymes. Oncogenic drivers can increase the expression of most proteins in the glycolytic pathway, including the terminal step of exporting H+ equivalents from the cytoplasm. Proton exporters maintain an alkaline cytoplasmic pH, which can enhance all glycolytic enzyme activities, even in the absence of oncogene-related expression changes. Based on this observation, we hypothesized that increased uptake and fermentative metabolism of glucose could be driven by the expulsion of H+ equivalents from the cell. RESULTS To test this hypothesis, we stably transfected lowly glycolytic MCF-7, U2-OS, and glycolytic HEK293 cells to express proton-exporting systems: either PMA1 (plasma membrane ATPase 1, a yeast H+-ATPase) or CA-IX (carbonic anhydrase 9). The expression of either exporter in vitro enhanced aerobic glycolysis as measured by glucose consumption, lactate production, and extracellular acidification rate. This resulted in an increased intracellular pH, and metabolomic analyses indicated that this was associated with an increased flux of all glycolytic enzymes upstream of pyruvate kinase. These cells also demonstrated increased migratory and invasive phenotypes in vitro, and these were recapitulated in vivo by more aggressive behavior, whereby the acid-producing cells formed higher-grade tumors with higher rates of metastases. Neutralizing tumor acidity with oral buffers reduced the metastatic burden. CONCLUSIONS Therefore, cancer cells which increase export of H+ equivalents subsequently increase intracellular alkalization, even without oncogenic driver mutations, and this is sufficient to alter cancer metabolism towards an upregulation of aerobic glycolysis, a Warburg phenotype. Overall, we have shown that the traditional understanding of cancer cells favoring glycolysis and the subsequent extracellular acidification is not always linear. Cells which can, independent of metabolism, acidify through proton exporter activity can sufficiently drive their metabolism towards glycolysis providing an important fitness advantage for survival.
Collapse
Affiliation(s)
- Shonagh Russell
- Cancer Physiology, Moffitt Cancer Center, 12902 USF Magnolia Dr, Tampa, FL 33612 USA
- Graduate School, University of South Florida, 4202 E Fowler Ave, Tampa, FL 33620 USA
| | - Liping Xu
- Cancer Physiology, Moffitt Cancer Center, 12902 USF Magnolia Dr, Tampa, FL 33612 USA
| | - Yoonseok Kam
- Agilent Technologies, 5301 Stevens Creek Blvd, Santa Clara, CA 9505 USA
| | - Dominique Abrahams
- Cancer Physiology, Moffitt Cancer Center, 12902 USF Magnolia Dr, Tampa, FL 33612 USA
| | - Bryce Ordway
- Cancer Physiology, Moffitt Cancer Center, 12902 USF Magnolia Dr, Tampa, FL 33612 USA
- Graduate School, University of South Florida, 4202 E Fowler Ave, Tampa, FL 33620 USA
| | - Alex S. Lopez
- Anatomic Pathology, Moffitt Cancer Center, 12902 USF Magnolia Dr, Tampa, FL 33612 USA
| | - Marilyn M. Bui
- Anatomic Pathology, Moffitt Cancer Center, 12902 USF Magnolia Dr, Tampa, FL 33612 USA
- Analytic Microscopy Core, Moffitt Cancer Center, 12902 USF Magnolia Dr, Tampa, FL 33612 USA
| | - Joseph Johnson
- Analytic Microscopy Core, Moffitt Cancer Center, 12902 USF Magnolia Dr, Tampa, FL 33612 USA
| | | | - Epifanio Ruiz
- Small Animal Imaging Department, Moffitt Cancer Center, 12902 USF Magnolia Dr, Tampa, FL 33612 USA
| | - Mark C. Lloyd
- Inspirata, Inc., One North Dale Mabry Hwy. Suite 600, Tampa, FL 33609 USA
| | - Pawel Swietach
- Department of Physiology, Anatomy and Genetics, University of Oxford, Parks Road, Oxford, OX1 3PT UK
| | - Daniel Verduzco
- Cancer Physiology, Moffitt Cancer Center, 12902 USF Magnolia Dr, Tampa, FL 33612 USA
| | - Jonathan Wojtkowiak
- Cancer Physiology, Moffitt Cancer Center, 12902 USF Magnolia Dr, Tampa, FL 33612 USA
| | - Robert J. Gillies
- Cancer Physiology, Moffitt Cancer Center, 12902 USF Magnolia Dr, Tampa, FL 33612 USA
| |
Collapse
|
2
|
Zamay TN, Starkov AK, Kolovskaya OS, Kichkailo AS, Inzhevatkin EV, Zamay GS, Titova NM, Zamay SS, Patc YS. Reduction of the Cisplatin Toxicity by Its Conjugation with Arabinogalactan. BIOCHEMISTRY (MOSCOW), SUPPLEMENT SERIES A: MEMBRANE AND CELL BIOLOGY 2020. [DOI: 10.1134/s1990747820010092] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
3
|
Leslie TK, James AD, Zaccagna F, Grist JT, Deen S, Kennerley A, Riemer F, Kaggie JD, Gallagher FA, Gilbert FJ, Brackenbury WJ. Sodium homeostasis in the tumour microenvironment. Biochim Biophys Acta Rev Cancer 2019; 1872:188304. [PMID: 31348974 PMCID: PMC7115894 DOI: 10.1016/j.bbcan.2019.07.001] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 07/11/2019] [Accepted: 07/12/2019] [Indexed: 12/17/2022]
Abstract
The concentration of sodium ions (Na+) is raised in solid tumours and can be measured at the cellular, tissue and patient levels. At the cellular level, the Na+ gradient across the membrane powers the transport of H+ ions and essential nutrients for normal activity. The maintenance of the Na+ gradient requires a large proportion of the cell's ATP. Na+ is a major contributor to the osmolarity of the tumour microenvironment, which affects cell volume and metabolism as well as immune function. Here, we review evidence indicating that Na+ handling is altered in tumours, explore our current understanding of the mechanisms that may underlie these alterations and consider the potential consequences for cancer progression. Dysregulated Na+ balance in tumours may open opportunities for new imaging biomarkers and re-purposing of drugs for treatment.
Collapse
Affiliation(s)
- Theresa K Leslie
- Department of Biology, University of York, Heslington, York YO10 5DD, UK; York Biomedical Research Institute, University of York, Heslington, York YO10 5DD, UK
| | - Andrew D James
- Department of Biology, University of York, Heslington, York YO10 5DD, UK; York Biomedical Research Institute, University of York, Heslington, York YO10 5DD, UK
| | - Fulvio Zaccagna
- Department of Radiology, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK
| | - James T Grist
- Department of Radiology, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK
| | - Surrin Deen
- Department of Radiology, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK
| | - Aneurin Kennerley
- York Biomedical Research Institute, University of York, Heslington, York YO10 5DD, UK; Department of Chemistry, University of York, Heslington, York YO10 5DD, UK
| | - Frank Riemer
- Department of Radiology, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK
| | - Joshua D Kaggie
- Department of Radiology, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK
| | - Ferdia A Gallagher
- Department of Radiology, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK
| | - Fiona J Gilbert
- Department of Radiology, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK
| | - William J Brackenbury
- Department of Biology, University of York, Heslington, York YO10 5DD, UK; York Biomedical Research Institute, University of York, Heslington, York YO10 5DD, UK.
| |
Collapse
|
4
|
Ganapathy-Kanniappan S. Molecular intricacies of aerobic glycolysis in cancer: current insights into the classic metabolic phenotype. Crit Rev Biochem Mol Biol 2019; 53:667-682. [PMID: 30668176 DOI: 10.1080/10409238.2018.1556578] [Citation(s) in RCA: 128] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Aerobic glycolysis is the process of oxidation of glucose into pyruvate followed by lactate production under normoxic condition. Distinctive from its anaerobic counterpart (i.e. glycolysis that occurs under hypoxia), aerobic glycolysis is frequently witnessed in cancers, popularly known as the "Warburg effect", and it is one of the earliest known evidences of metabolic alteration in neoplasms. Intracellularly, aerobic glycolysis circumvents mitochondrial oxidative phosphorylation (OxPhos), facilitating an increased rate of glucose hydrolysis. This in turn enables cancer cells to successfully compete with normal cells for glucose uptake in order to maintain uninterrupted growth. In addition, evading OxPhos mitigates excessive generation/accumulation of reactive oxygen species that otherwise may be deleterious to cells. Emerging data indicate that aerobic glycolysis in cancer also promotes glutaminolysis to satisfy the precursor requirements of certain biosynthetic processes (e.g. nucleic acids). Next, the metabolic intermediates of aerobic glycolysis also feed the pentose phosphate pathway (PPP) to facilitate macromolecular biosynthesis necessary for cancer cell growth and proliferation. Extracellularly, the extrusion of the end-product of aerobic glycolysis, i.e. lactate, alters the tumor microenvironment, and impacts cancer-associated cells. Collectively, accumulating data unequivocally demonstrate that aerobic glycolysis implicates myriad of molecular and functional processes to support cancer progression. This review, in the light of recent research, dissects the molecular intricacies of its regulation, and also deliberates the emerging paradigms to target aerobic glycolysis in cancer therapy.
Collapse
Affiliation(s)
- Shanmugasundaram Ganapathy-Kanniappan
- a The Division of Interventional Radiology, Russell H. Morgan Department of Radiology & Radiological Science , The Johns Hopkins University School of Medicine , Baltimore , MD , USA
| |
Collapse
|
5
|
Flinck M, Kramer SH, Pedersen SF. Roles of pH in control of cell proliferation. Acta Physiol (Oxf) 2018; 223:e13068. [PMID: 29575508 DOI: 10.1111/apha.13068] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 02/17/2018] [Accepted: 03/19/2018] [Indexed: 02/06/2023]
Abstract
Precise spatiotemporal regulation of intracellular pH (pHi ) is a prerequisite for normal cell function, and changes in pHi or pericellular pH (pHe ) exert important signalling functions. It is well established that proliferation of mammalian cells is dependent on a permissive pHi in the slightly alkaline range (7.0-7.2). It is also clear that mitogen signalling in nominal absence of HCO3- is associated with an intracellular alkalinization (~0.3 pH unit above steady-state pHi ), which is secondary to activation of Na+ /H+ exchange. However, it remains controversial whether this increase in pHi is part of the mitogenic signal cascade leading to cell cycle entry and progression, and whether it is relevant under physiological conditions. Furthermore, essentially all studies of pHi in mammalian cell proliferation have focused on the mitogen-induced G0-G1 transition, and the regulation and roles of pHi during the cell cycle remain poorly understood. The aim of this review is to summarize and critically discuss the possible roles of pHi and pHe in cell cycle progression. While the focus is on the mammalian cell cycle, important insights from studies in lower eukaryotes are also discussed. We summarize current evidence of links between cell cycle progression and pHi and discuss possible pHi - and pHe sensors and signalling pathways relevant to mammalian proliferation control. The possibility that changes in pHi during cell cycle progression may be an integral part of the checkpoint control machinery is explored. Finally, we discuss the relevance of links between pH and proliferation in the context of the perturbed pH homoeostasis and acidic microenvironment of solid tumours.
Collapse
Affiliation(s)
- M. Flinck
- Section for Cell Biology and Physiology; Department of Biology; Faculty of Science; University of Copenhagen; Copenhagen Denmark
| | - S. H. Kramer
- Section for Cell Biology and Physiology; Department of Biology; Faculty of Science; University of Copenhagen; Copenhagen Denmark
| | - S. F. Pedersen
- Section for Cell Biology and Physiology; Department of Biology; Faculty of Science; University of Copenhagen; Copenhagen Denmark
| |
Collapse
|
6
|
Out of Warburg effect: An effective cancer treatment targeting the tumor specific metabolism and dysregulated pH. Semin Cancer Biol 2017; 43:134-138. [PMID: 28122260 DOI: 10.1016/j.semcancer.2017.01.005] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 01/17/2017] [Accepted: 01/17/2017] [Indexed: 11/20/2022]
Abstract
As stated by Otto Warburg nearly a century ago, cancer is a metabolic disease, a fermentation caused by malfunctioning mitochondria, resulting in increased anabolism and decreased catabolism. Treatment should, therefore, aim at restoring the energy yield. To decrease anabolism, glucose uptake should be reduced (ketogenic diet). To increase catabolism, the oxidative phosphorylation should be restored. Treatment with a combination of α-lipoic acid and hydroxycitrate has been shown to be effective in multiple animal models. This treatment, in combination with conventional chemotherapy, has yielded extremely encouraging results in glioblastoma, brain metastasis and lung cancer. Randomized trials are necessary to confirm these preliminary data. The major limitation is the fact that the combination of α-lipoic acid and hydroxycitrate can only be effective if the mitochondria are still present and/or functional. That may not be the case in the most aggressive tumors. The increased intracellular alkalosis is a strong mitogenic signal, which bypasses most inhibitory signals. Concomitant correction of this alkalosis may be a very effective treatment in case of mitochondrial failure.
Collapse
|
7
|
Caruso JP, Koch BJ, Benson PD, Varughese E, Monterey MD, Lee AE, Dave AM, Kiousis S, Sloan AE, Mathupala SP. pH, Lactate, and Hypoxia: Reciprocity in Regulating High-Affinity Monocarboxylate Transporter Expression in Glioblastoma. Neoplasia 2017; 19:121-134. [PMID: 28092823 PMCID: PMC5238458 DOI: 10.1016/j.neo.2016.12.011] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Revised: 12/15/2016] [Accepted: 12/20/2016] [Indexed: 02/04/2023] Open
Abstract
Highly malignant brain tumors harbor the aberrant propensity for aerobic glycolysis, the excessive conversion of glucose to lactic acid even in the presence of ample tissue oxygen. Lactic acid is rapidly effluxed to the tumor microenvironment via a group of plasma-membrane transporters denoted monocarboxylate transporters (MCTs) to prevent “self-poisoning.” One isoform, MCT2, has the highest affinity for lactate and thus should have the ability to respond to microenvironment conditions such as hypoxia, lactate, and pH to help maintain high glycolytic flux in the tumor. Yet, MCT2 is considered to not respond to hypoxia, which is counterintuitive. Its response to tumor lactate has not been reported. In this report, we experimentally identify the transcription initiation site/s for MCT2 in astrocytes (normal) and glioma (tumor). We then use a BACmid library to isolate a 4.2-kbp MCT2 promoter-exon I region and examine promoter response to glycolysis-mediated stimuli in glioma cells. Reporter analysis of nested-promoter constructs indicated response of MCT2 to hypoxia, pH, lactate, and glucose, the major physiological “players” that facilitate a tumor's growth and proliferation. Immunoblot analysis of native MCT2 expression under altered pH and hypoxia reflected the reporter data. The pH-mediated gene-regulation studies we describe are the first to record H+-based reporter studies for any mammalian system and demonstrate the exquisite response of the MCT2 gene to minute changes in tumor pH. Identical promoter usage also provides the first evidence of astrocytes harnessing the same gene regulatory regions to facilitate astrocyte-neuron lactate shuttling, a metabolic feature of normal brain.
Collapse
Affiliation(s)
- James P Caruso
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI 48201; Program in Biophysics, University of Michigan, Ann Arbor, MI 48109
| | - Brandon J Koch
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI 48201; Department of Biochemistry and Molecular Biology, Rollins College, Winter Park, FL 32789
| | - Philip D Benson
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI 48201
| | - Elsa Varughese
- Department of Physics & Astronomy, Wayne State University College of Liberal Arts & Sciences, Detroit, MI, 48201
| | - Michael D Monterey
- Department of Neurosurgery & Basic Medical Sciences Program, Wayne State University School of Medicine, Detroit, MI 48201
| | - Amy E Lee
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI 48201
| | - Ajal M Dave
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI 48201
| | - Sam Kiousis
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI 48201
| | - Andrew E Sloan
- Department of Neurological Surgery, University Hospitals-Case Medical Center & Case Comprehensive Cancer Center, Cleveland, OH 44106
| | - Saroj P Mathupala
- Department of Neurosurgery and Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48201.
| |
Collapse
|
8
|
Shin JJ, Aftab Q, Austin P, McQueen JA, Poon T, Li SC, Young BP, Roskelley CD, Loewen CJR. Systematic identification of genes involved in metabolic acid stress resistance in yeast and their potential as cancer targets. Dis Model Mech 2016; 9:1039-49. [PMID: 27519690 PMCID: PMC5047693 DOI: 10.1242/dmm.023374] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Accepted: 07/18/2016] [Indexed: 12/12/2022] Open
Abstract
A hallmark of all primary and metastatic tumours is their high rate of glucose uptake and glycolysis. A consequence of the glycolytic phenotype is the accumulation of metabolic acid; hence, tumour cells experience considerable intracellular acid stress. To compensate, tumour cells upregulate acid pumps, which expel the metabolic acid into the surrounding tumour environment, resulting in alkalization of intracellular pH and acidification of the tumour microenvironment. Nevertheless, we have only a limited understanding of the consequences of altered intracellular pH on cell physiology, or of the genes and pathways that respond to metabolic acid stress. We have used yeast as a genetic model for metabolic acid stress with the rationale that the metabolic changes that occur in cancer that lead to intracellular acid stress are likely fundamental. Using a quantitative systems biology approach we identified 129 genes required for optimal growth under conditions of metabolic acid stress. We identified six highly conserved protein complexes with functions related to oxidative phosphorylation (mitochondrial respiratory chain complex III and IV), mitochondrial tRNA biosynthesis [glutamyl-tRNA(Gln) amidotransferase complex], histone methylation (Set1C-COMPASS), lysosome biogenesis (AP-3 adapter complex), and mRNA processing and P-body formation (PAN complex). We tested roles for two of these, AP-3 adapter complex and PAN deadenylase complex, in resistance to acid stress using a myeloid leukaemia-derived human cell line that we determined to be acid stress resistant. Loss of either complex inhibited growth of Hap1 cells at neutral pH and caused sensitivity to acid stress, indicating that AP-3 and PAN complexes are promising new targets in the treatment of cancer. Additionally, our data suggests that tumours may be genetically sensitized to acid stress and hence susceptible to acid stress-directed therapies, as many tumours accumulate mutations in mitochondrial respiratory chain complexes required for their proliferation.
Collapse
Affiliation(s)
- John J Shin
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada V6T 1Z3
| | - Qurratulain Aftab
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada V6T 1Z3
| | - Pamela Austin
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada V6T 1Z3
| | - Jennifer A McQueen
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada V6T 1Z3
| | - Tak Poon
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada V6T 1Z3
| | - Shu Chen Li
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada V6T 1Z3
| | - Barry P Young
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada V6T 1Z3
| | - Calvin D Roskelley
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada V6T 1Z3
| | - Christopher J R Loewen
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada V6T 1Z3
| |
Collapse
|
9
|
Messeha SS, Zarmouh NO, Taka E, Gendy SG, Shokry GR, Kolta MG, Soliman KFA. The Role of Monocarboxylate Transporters and Their Chaperone CD147 in Lactate Efflux Inhibition and the Anticancer Effects of Terminalia chebula in Neuroblastoma Cell Line N2-A. ACTA ACUST UNITED AC 2016; 12. [PMID: 27158628 PMCID: PMC4857771 DOI: 10.9734/ejmp/2016/23992] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
AIMS In the presence of oxygen, most of the synthesized pyruvate during glycolysis in the cancer cell of solid tumors is released away from the mitochondria to form lactate (Warburg Effect). To maintain cell homeostasis, lactate is transported across the cell membrane by monocarboxylate transporters (MCTs). The major aim of the current investigation is to identify novel compounds that inhibit lactate efflux that may lead to identifying effective targets for cancer treatment. STUDY DESIGN In this study, 900 ethanol plant extracts were screened for their lactate efflux inhibition using neuroblastoma (N2-A) cell line. Additionally, we investigated the mechanism of inhibition for the most potent plant extract regarding monocarboxylate transporters expression, and consequences effects on viability, growth, and apoptosis. METHODOLOGY The potency of lactate efflux inhibition of ethanol plant extracts was evaluated in N2-A cells by measuring extracellular lactate levels. Caspase 3- activity and acridine orange/ethidium bromide staining were performed to assess the apoptotic effect. The antiproliferative effect was measured using WST assay. Western blotting was performed to quantify protein expression of MCTs and their chaperone CD147 in treated cells lysates. RESULTS Terminalia chebula plant extract was the most potent lactate efflux inhibitor in N2-A cells among the 900 - tested plant extracts. The results obtained show that extract of Terminalia chebula fruits (TCE) significantly (P = 0.05) reduced the expression of the MCT1, MCT3, MCT4 and the chaperone CD147. The plant extract was more potent (IC50 of 3.59 ± 0.26 μg/ml) than the MCT standard inhibitor phloretin (IC50 76.54 ± 3.19 μg/ml). The extract also showed more potency and selective cytotoxicity in cancer cells than DI-TNC1 primary cell line (IC50 7.37 ± 0.28 vs. 17.35 ± 0.19 μg/ml). Moreover, TCE Inhibited N2-A cell growth (IG50 = 5.20 ± 0.30 μg/ml) and induced apoptosis at the 7.5 μg/ml concentration. CONCLUSION Out of the 900 plant extracts screened, Terminalia chebula ethanol extract was found to be the most potent lactate efflux inhibitor with the ability to inhibit chaperone CD147 expression and impact the function of monocarboxylate transporters. Furthermore, TCE was found to have growth inhibition and apoptotic effects. The results obtained indicate that Terminalia chebula constituent(s) may contain promising compounds that can be useful in the management of neuroblastoma cancer.
Collapse
Affiliation(s)
- S S Messeha
- College of Pharmacy and Pharmaceutical Sciences, Florida A & M University, Tallahassee, Florida 32307, USA
| | - N O Zarmouh
- College of Pharmacy and Pharmaceutical Sciences, Florida A & M University, Tallahassee, Florida 32307, USA
| | - E Taka
- College of Pharmacy and Pharmaceutical Sciences, Florida A & M University, Tallahassee, Florida 32307, USA
| | - S G Gendy
- College of Pharmacy and Pharmaceutical Sciences, Florida A & M University, Tallahassee, Florida 32307, USA
| | - G R Shokry
- College of Pharmacy and Pharmaceutical Sciences, Florida A & M University, Tallahassee, Florida 32307, USA
| | - M G Kolta
- College of Pharmacy and Pharmaceutical Sciences, Florida A & M University, Tallahassee, Florida 32307, USA
| | - K F A Soliman
- College of Pharmacy and Pharmaceutical Sciences, Florida A & M University, Tallahassee, Florida 32307, USA
| |
Collapse
|
10
|
Smith GA, Howell GJ, Phillips C, Muench SP, Ponnambalam S, Harrison MA. Extracellular and Luminal pH Regulation by Vacuolar H+-ATPase Isoform Expression and Targeting to the Plasma Membrane and Endosomes. J Biol Chem 2016; 291:8500-15. [PMID: 26912656 PMCID: PMC4861423 DOI: 10.1074/jbc.m116.723395] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Indexed: 01/02/2023] Open
Abstract
Plasma membrane vacuolar H+-ATPase (V-ATPase) activity of tumor cells is a major factor in control of cytoplasmic and extracellular pH and metastatic potential, but the isoforms involved and the factors governing plasma membrane recruitment remain uncertain. Here, we examined expression, distribution, and activity of V-ATPase isoforms in invasive prostate adenocarcinoma (PC-3) cells. Isoforms 1 and 3 were the most highly expressed forms of membrane subunit a, with a1 and a3 the dominant plasma membrane isoforms. Correlation between plasma membrane V-ATPase activity and invasiveness was limited, but RNAi knockdown of either a isoform did slow cell proliferation and inhibit invasion in vitro. Isoform a1 was recruited to the cell surface from the early endosome-recycling complex pathway, its knockdown arresting transferrin receptor recycling. Isoform a3 was associated with the late endosomal/lysosomal compartment. Both a isoforms associated with accessory protein Ac45, knockdown of which stalled transit of a1 and transferrin-transferrin receptor, decreased proton efflux, and reduced cell growth and invasiveness; this latter effect was at least partly due to decreased delivery of the membrane-bound matrix metalloproteinase MMP-14 to the plasma membrane. These data indicate that in prostatic carcinoma cells, a1 and a3 isoform populations predominate in different compartments where they maintain different luminal pH. Ac45 plays a central role in navigating the V-ATPase to the plasma membrane, and hence it is an important factor in expression of the invasive phenotype.
Collapse
Affiliation(s)
- Gina A Smith
- From the Endothelial Cell Biology Unit, School of Molecular and Cellular Biology and
| | - Gareth J Howell
- From the Endothelial Cell Biology Unit, School of Molecular and Cellular Biology and
| | - Clair Phillips
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Stephen P Muench
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, United Kingdom
| | | | - Michael A Harrison
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, United Kingdom
| |
Collapse
|
11
|
Amith SR, Wilkinson JM, Fliegel L. Assessing Na +/H + exchange and cell effector functionality in metastatic breast cancer. BIOCHIMIE OPEN 2016; 2:16-23. [PMID: 29632834 PMCID: PMC5889484 DOI: 10.1016/j.biopen.2016.01.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Accepted: 01/10/2016] [Indexed: 11/28/2022]
Abstract
Metastasis is the leading cause of mortality in patients with breast cancer. In triple-negative breast cancer, high recurrence rates, increased invasive capacity of cells, and their aggressive ability to metastasize at secondary sites dictate patient survival. The Na+/H+ exchanger isoform 1 (NHE1) plays a critical role in controlling the metastatic potential of these cells. Its activity results in an elevation of intracellular pH and in extracellular acidification, a key step in the establishment of the tumor microenvironment. Here, we describe assays for characterization of Na+/H+ exchanger activity and its related downstream physiological effects on triple-negative breast cancer cells. Na+/H+ exchanger activity can be routinely and rapidly measured in live cells with a fluorometric assay that assesses changes in intracellular pH. Characterization of downstream cell effector function as a result of Na+/H+ exchanger activation can be evaluated by measuring directed cell migration and invasion. Cell migration is assessed with wound-healing assays, where a gap is introduced in a confluent monolayer of cells and the rate of gap closure is measured over time. Cell invasion is assessed in the short-term by transwell invasion assays that track cell movement through an extracellular matrix. Long-term invasiveness, growth and proliferation can be assessed with 3-D invasion assays using transwell inserts fitted with specialized scaffolds optimized for 3-D cell culture. Taken together these assays provide powerful tools for testing the effects of altering Na+/H+ exchanger activity with chemical inhibition on the metastatic capacity of breast cancer cells. The Na+/H+ exchanger in breast cancer cells is assayed by rapid fluorometric assay. Migration of triple negative breast cancer cells is assessed by wound healing. Cell invasion is measured via transwell 3D invasion assays.
Collapse
Affiliation(s)
- Schammim Ray Amith
- Department of Biochemistry, University of Alberta, Edmonton, Alberta, T6G 2H7, Canada
| | - Jodi Marie Wilkinson
- Department of Biochemistry, University of Alberta, Edmonton, Alberta, T6G 2H7, Canada
| | - Larry Fliegel
- Department of Biochemistry, University of Alberta, Edmonton, Alberta, T6G 2H7, Canada
| |
Collapse
|
12
|
Abstract
Cation-coupled HCO3(-) transport was initially identified in the mid-1970s when pioneering studies showed that acid extrusion from cells is stimulated by CO2/HCO3(-) and associated with Na(+) and Cl(-) movement. The first Na(+)-coupled bicarbonate transporter (NCBT) was expression-cloned in the late 1990s. There are currently five mammalian NCBTs in the SLC4-family: the electrogenic Na,HCO3-cotransporters NBCe1 and NBCe2 (SLC4A4 and SLC4A5 gene products); the electroneutral Na,HCO3-cotransporter NBCn1 (SLC4A7 gene product); the Na(+)-driven Cl,HCO3-exchanger NDCBE (SLC4A8 gene product); and NBCn2/NCBE (SLC4A10 gene product), which has been characterized as an electroneutral Na,HCO3-cotransporter or a Na(+)-driven Cl,HCO3-exchanger. Despite the similarity in amino acid sequence and predicted structure among the NCBTs of the SLC4-family, they exhibit distinct differences in ion dependency, transport function, pharmacological properties, and interactions with other proteins. In epithelia, NCBTs are involved in transcellular movement of acid-base equivalents and intracellular pH control. In nonepithelial tissues, NCBTs contribute to intracellular pH regulation; and hence, they are crucial for diverse tissue functions including neuronal discharge, sensory neuron development, performance of the heart, and vascular tone regulation. The function and expression levels of the NCBTs are generally sensitive to intracellular and systemic pH. Animal models have revealed pathophysiological roles of the transporters in disease states including metabolic acidosis, hypertension, visual defects, and epileptic seizures. Studies are being conducted to understand the physiological consequences of genetic polymorphisms in the SLC4-members, which are associated with cancer, hypertension, and drug addiction. Here, we describe the current knowledge regarding the function, structure, and regulation of the mammalian cation-coupled HCO3(-) transporters of the SLC4-family.
Collapse
Affiliation(s)
- Christian Aalkjaer
- Department of Biomedicine, and the Water and Salt Research Center, Aarhus University, Aarhus, Denmark; Department of Physiology, Emory University School of Medicine, Atlanta, USA
| | | | | | | |
Collapse
|
13
|
Fais S, Venturi G, Gatenby B. Microenvironmental acidosis in carcinogenesis and metastases: new strategies in prevention and therapy. Cancer Metastasis Rev 2015; 33:1095-108. [PMID: 25376898 PMCID: PMC4244550 DOI: 10.1007/s10555-014-9531-3] [Citation(s) in RCA: 122] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Much effort is currently devoted to developing patient-specific cancer therapy based on molecular characterization of tumors. In particular, this approach seeks to identify driver mutations that can be blocked through small molecular inhibitors. However, this approach is limited by extensive intratumoral genetic heterogeneity, and, not surprisingly, even dramatic initial responses are typically of limited duration as resistant tumor clones rapidly emerge and proliferate. We propose an alternative approach based on observations that while tumor evolution produces genetic divergence, it is also associated with striking phenotypic convergence that loosely correspond to the well-known cancer “hallmarks”. These convergent properties can be described as driver phenotypes and may be more consistently and robustly expressed than genetic targets. To this purpose, it is necessary to identify strategies that are critical for cancer progression and metastases, and it is likely that these driver phenotypes will be closely related to cancer “hallmarks”. It appears that an antiacidic approach, by targetting a driver phenotype in tumors, may be thought as a future strategy against tumors in either preventing the occurrence of cancer or treating tumor patients with multiple aims, including the improvement of efficacy of existing therapies, possibly reducing their systemic side effects, and controlling tumor growth, progression, and metastasis. This may be achieved with existing molecules such as proton pump inhibitors (PPIs) and buffers such as sodium bicarbonate, citrate, or TRIS.
Collapse
Affiliation(s)
- Stefano Fais
- Department of Therapeutic Research and Medicines Evaluation, Unit of Antitumor Drugs, Istituto Superiore di Sanità, Viale Regina Elena 299, Rome, Italy
- Department of Drug Research and Medicines Evaluation, Istituto Superiore di Sanità (National Institute of Health), Viale Regina Elena 299, 00161 Rome, Italy
| | - Giulietta Venturi
- Department of Therapeutic Research and Medicines Evaluation, Unit of Antitumor Drugs, Istituto Superiore di Sanità, Viale Regina Elena 299, Rome, Italy
| | - Bob Gatenby
- Radiology Department, Cancer Biology and Evolution Program Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL 33612 USA
| |
Collapse
|
14
|
Marchiq I, Pouysségur J. Hypoxia, cancer metabolism and the therapeutic benefit of targeting lactate/H(+) symporters. J Mol Med (Berl) 2015; 94:155-71. [PMID: 26099350 PMCID: PMC4762928 DOI: 10.1007/s00109-015-1307-x] [Citation(s) in RCA: 209] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Revised: 06/03/2015] [Accepted: 06/05/2015] [Indexed: 12/15/2022]
Abstract
Since Otto Warburg reported the 'addiction' of cancer cells to fermentative glycolysis, a metabolic pathway that provides energy and building blocks, thousands of studies have shed new light on the molecular mechanisms contributing to altered cancer metabolism. Hypoxia, through hypoxia-inducible factors (HIFs), in addition to oncogenes activation and loss of tumour suppressors constitute major regulators of not only the "Warburg effect" but also many other metabolic pathways such as glutaminolysis. Enhanced glucose and glutamine catabolism has become a recognised feature of cancer cells, leading to accumulation of metabolites in the tumour microenvironment, which offers growth advantages to tumours. Among these metabolites, lactic acid, besides imposing an acidic stress, is emerging as a key signalling molecule that plays a pivotal role in cancer cell migration, angiogenesis, immune escape and metastasis. Although interest in lactate for cancer development only appeared recently, pharmacological molecules blocking its metabolism are already in phase I/II clinical trials. Here, we review the metabolic pathways generating lactate, and we discuss the rationale for targeting lactic acid transporter complexes for the development of efficient and selective anticancer therapies.
Collapse
Affiliation(s)
- Ibtissam Marchiq
- Institute for Research on Cancer and Aging of Nice (IRCAN), University of Nice Sophia Antipolis, Centre A. Lacassagne, 33 Avenue, 06189, Nice, France
| | - Jacques Pouysségur
- Institute for Research on Cancer and Aging of Nice (IRCAN), University of Nice Sophia Antipolis, Centre A. Lacassagne, 33 Avenue, 06189, Nice, France.
- Medical Biology Department (CSM), Centre Scientifique de Monaco, Quai Antoine 1er, Monaco.
| |
Collapse
|
15
|
Epstein T, Xu L, Gillies RJ, Gatenby RA. Separation of metabolic supply and demand: aerobic glycolysis as a normal physiological response to fluctuating energetic demands in the membrane. Cancer Metab 2014; 2:7. [PMID: 24982758 PMCID: PMC4060846 DOI: 10.1186/2049-3002-2-7] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2014] [Accepted: 05/07/2014] [Indexed: 01/05/2023] Open
Abstract
Background Cancer cells, and a variety of normal cells, exhibit aerobic glycolysis, high rates of glucose fermentation in the presence of normal oxygen concentrations, also known as the Warburg effect. This metabolism is considered abnormal because it violates the standard model of cellular energy production that assumes glucose metabolism is predominantly governed by oxygen concentrations and, therefore, fermentative glycolysis is an emergency back-up for periods of hypoxia. Though several hypotheses have been proposed for the origin of aerobic glycolysis, its biological basis in cancer and normal cells is still not well understood. Results We examined changes in glucose metabolism following perturbations in membrane activity in different normal and tumor cell lines and found that inhibition or activation of pumps on the cell membrane led to reduction or increase in glycolysis, respectively, while oxidative phosphorylation remained unchanged. Computational simulations demonstrated that these findings are consistent with a new model of normal physiological cellular metabolism in which efficient mitochondrial oxidative phosphorylation supplies chronic energy demand primarily for macromolecule synthesis and glycolysis is necessary to supply rapid energy demands primarily to support membrane pumps. A specific model prediction was that the spatial distribution of ATP-producing enzymes in the glycolytic pathway must be primarily localized adjacent to the cell membrane, while mitochondria should be predominantly peri-nuclear. The predictions were confirmed experimentally. Conclusions Our results show that glycolytic metabolism serves a critical physiological function under normoxic conditions by responding to rapid energetic demand, mainly from membrane transport activities, even in the presence of oxygen. This supports a new model for glucose metabolism in which glycolysis and oxidative phosphorylation supply different types of energy demand. Cells use efficient but slow-responding aerobic metabolism to meet baseline, steady energy demand and glycolytic metabolism, which is inefficient but can rapidly increase adenosine triphosphate (ATP) production, to meet short-timescale energy demands, mainly from membrane transport activities. In this model, the origin of the Warburg effect in cancer cells and aerobic glycolysis in general represents a normal physiological function due to enhanced energy demand for membrane transporters activity required for cell division, growth, and migration.
Collapse
Affiliation(s)
- Tamir Epstein
- Program of Cancer Biology and Evolution, Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL 33612, USA
| | - Liping Xu
- Department of Cancer Imaging and Metabolism, Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL 33612, USA
| | - Robert J Gillies
- Department of Cancer Imaging and Metabolism, Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL 33612, USA
| | - Robert A Gatenby
- Department of Radiology and Program of Cancer Biology and Evolution, Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL 33612, USA
| |
Collapse
|
16
|
Hobbs GA, Zhou B, Cox AD, Campbell SL. Rho GTPases, oxidation, and cell redox control. Small GTPases 2014; 5:e28579. [PMID: 24809833 DOI: 10.4161/sgtp.28579] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
While numerous studies support regulation of Ras GTPases by reactive oxygen and nitrogen species, the Rho subfamily has received considerably less attention. Over the last few years, increasing evidence is emerging that supports the redox sensitivity of Rho GTPases. Moreover, as Rho GTPases regulate the cellular redox state by controlling enzymes that generate and convert reactive oxygen and nitrogen species, redox feedback loops likely exist. Here, we provide an overview of cellular oxidants, Rho GTPases, and their inter-dependence.
Collapse
Affiliation(s)
- G Aaron Hobbs
- Department of Biochemistry and Biophysics; University of North Carolina; Chapel Hill, NC USA
| | - Bingying Zhou
- Department of Pharmacology; University of North Carolina; Chapel Hill, NC USA
| | - Adrienne D Cox
- Department of Pharmacology; University of North Carolina; Chapel Hill, NC USA; Department of Radiation Oncology, University of North Carolina; Chapel Hill, NC USA; Lineberger Comprehensive Cancer Center; University of North Carolina; Chapel Hill, NC USA
| | - Sharon L Campbell
- Department of Biochemistry and Biophysics; University of North Carolina; Chapel Hill, NC USA; Lineberger Comprehensive Cancer Center; University of North Carolina; Chapel Hill, NC USA
| |
Collapse
|
17
|
Swietach P, Vaughan-Jones RD, Harris AL, Hulikova A. The chemistry, physiology and pathology of pH in cancer. Philos Trans R Soc Lond B Biol Sci 2014; 369:20130099. [PMID: 24493747 PMCID: PMC3917353 DOI: 10.1098/rstb.2013.0099] [Citation(s) in RCA: 377] [Impact Index Per Article: 37.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Cell survival is conditional on the maintenance of a favourable acid-base balance (pH). Owing to intensive respiratory CO2 and lactic acid production, cancer cells are exposed continuously to large acid-base fluxes, which would disturb pH if uncorrected. The large cellular reservoir of H(+)-binding sites can buffer pH changes but, on its own, is inadequate to regulate intracellular pH. To stabilize intracellular pH at a favourable level, cells control trans-membrane traffic of H(+)-ions (or their chemical equivalents, e.g. ) using specialized transporter proteins sensitive to pH. In poorly perfused tumours, additional diffusion-reaction mechanisms, involving carbonic anhydrase (CA) enzymes, fine-tune control extracellular pH. The ability of H(+)-ions to change the ionization state of proteins underlies the exquisite pH sensitivity of cellular behaviour, including key processes in cancer formation and metastasis (proliferation, cell cycle, transformation, migration). Elevated metabolism, weakened cell-to-capillary diffusive coupling, and adaptations involving H(+)/H(+)-equivalent transporters and extracellular-facing CAs give cancer cells the means to manipulate micro-environmental acidity, a cancer hallmark. Through genetic instability, the cellular apparatus for regulating and sensing pH is able to adapt to extracellular acidity, driving disease progression. The therapeutic potential of disturbing this sequence by targeting H(+)/H(+)-equivalent transporters, buffering or CAs is being investigated, using monoclonal antibodies and small-molecule inhibitors.
Collapse
Affiliation(s)
- Pawel Swietach
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | | | - Adrian L. Harris
- Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Alzbeta Hulikova
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| |
Collapse
|
18
|
Poehlmann A, Reissig K, Schönfeld P, Walluscheck D, Schinlauer A, Hartig R, Lessel W, Guenther T, Silver A, Roessner A. Repeated H2 O2 exposure drives cell cycle progression in an in vitro model of ulcerative colitis. J Cell Mol Med 2013; 17:1619-31. [PMID: 24118792 PMCID: PMC3914643 DOI: 10.1111/jcmm.12150] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2013] [Accepted: 09/04/2013] [Indexed: 12/20/2022] Open
Abstract
The production of hydrogen peroxide (H2O2) drives tumourigenesis in ulcerative colitis (UC). Recently, we showed that H2O2 activates DNA damage checkpoints in human colonic epithelial cells (HCEC) through c-Jun N-terminal Kinases (JNK) that induces p21WAF1. Moreover, caspases circumvented the G1/S and intra-S checkpoints, and cells accumulated in G2/M. The latter observation raised the question of whether repeated H2O2 exposures alter JNK activation, thereby promoting a direct passage of cells from G2/M arrest to driven cell cycle progression. Here, we report that increased proliferation of repeatedly H2O2-exposed HCEC cells (C-cell cultures) was associated with (i) increased phospho-p46 JNK, (ii) decreased total JNK and phospho-p54 JNK and (iii) p21WAF1 down-regulation. Altered JNK activation and p21WAF1 down-regulation were accompanied by defects in maintaining G2/M and mitotic spindle checkpoints through adaptation, as well as by apoptosis resistance following H2O2 exposure. This may cause increased proliferation of C-cell cultures, a defining initiating feature in the inflammation-carcinoma pathway in UC. We further suggest that dysregulated JNK activation is attributed to a non-apoptotic function of caspases, causing checkpoint adaptation in C-cell cultures. Additionally, loss of cell-contact inhibition and the overcoming of senescence, hallmarks of cancer, contributed to increased proliferation. Furthermore, there was evidence that p54 JNK inactivation is responsible for loss of cell-contact inhibition. We present a cellular model of UC and suggest a sinusoidal pattern of proliferation, which is triggered by H2O2-induced reactive oxygen species generation, involving an interplay between JNK activation/inactivation, p21WAF1, c-Fos, c-Jun/phospho-c-Jun, ATF2/phospho-ATF2, β-catenin/TCF4-signalling, c-Myc, CDK6 and Cyclin D2, leading to driven cell cycle progression.
Collapse
Affiliation(s)
- Angela Poehlmann
- Department of Pathology, Otto-von-Guericke University, Magdeburg, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Abstract
Heterogeneous microenvironmental conditions play critical roles in cancer pathogenesis and therapy resistance and arise from changes in tissue dimensionality, cell-extracellular matrix (ECM) interactions, soluble factor signaling, oxygen as well as metabolic gradients, and exogeneous biomechanical cues. Traditional cell culture approaches are restricted in their ability to mimic this complexity with physiological relevance, offering only partial explanation as to why novel therapeutic compounds are frequently efficacious in vitro but disappoint in preclinical and clinical studies. In an effort to overcome these limitations, physical sciences-based strategies have been employed to model specific aspects of the cancer microenvironment. Although these strategies offer promise to reveal the contributions of microenvironmental parameters on tumor initiation, progression, and therapy resistance, they, too, frequently suffer from limitations. This review highlights physicochemical and biological key features of the tumor microenvironment, critically discusses advantages and limitations of current engineering strategies, and provides a perspective on future opportunities for engineered tumor models.
Collapse
Affiliation(s)
- David W Infanger
- Department of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | | | | |
Collapse
|
20
|
Martinez-Zaguilan R, Tompkins LS, Gillies RJ, Lynch RM. Simultaneous analysis of intracellular pH and Ca²⁺ from cell populations. Methods Mol Biol 2013; 937:253-71. [PMID: 23007592 DOI: 10.1007/978-1-62703-086-1_16] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Although changes in both pH(in) and [Ca(2+)](i) have been observed in response to a variety of agonists, it is not clear whether these ionic events work independently or are coordinated to lead to a specific physiological response. One of the fundamental problems in studying these ionic events is that changes in pH(in) modify Ca(2+) regulatory mechanisms and changes in Ca(2+) may modify pH regulation. It is desirable to use a technique that allows concomitant monitoring of these two ions in cell populations with high time resolution. Furthermore, like many Ca(2+) binding proteins, all Ca(2+)-sensitive fluoroprobes are inherently sensitive to pH owing to competition of H(+) for the Ca(2+)-binding sites. This chapter describes experimental paradigms that provide optimum conditions for simultaneous measurement of pH from the fluorescence emission of snarf-1, and Ca(2+) using fura-2. The fluorescence spectra of these compounds are sufficiently different to allow simultaneous measurement of pH and Ca(2+) both in vitro and in vivo. Moreover, the ratio of the H(+)-sensitive wavelengths of snarf-1 is unaffected by Ca(2+), or the concomitant presence of fura-2 in cells. Although the fluorescence ratio of fura-2 is insensitive to the presence of snarf-1, it is affected by pH, as indicated above. We describe procedures to correct for this effect and to obtain calibration parameters for fura-2 and snarf-1 required to facilitate analysis of pH and Ca(2+) concentrations within cell populations.
Collapse
Affiliation(s)
- Raul Martinez-Zaguilan
- Department of Physiology, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | | | | | | |
Collapse
|
21
|
Xu J, Xie R, Liu X, Wen G, Jin H, Yu Z, Jiang Y, Zhao Z, Yang Y, Ji B, Dong H, Tuo B. Expression and functional role of vacuolar H(+)-ATPase in human hepatocellular carcinoma. Carcinogenesis 2012; 33:2432-40. [PMID: 22962303 DOI: 10.1093/carcin/bgs277] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Tumor cells often exist in a hypoxic microenvironment, which produces acidic metabolites. To survive in this harsh environment, tumor cells must exhibit a dynamic cytosolic pH regulatory system. Vacuolar H(+)-adenosine triphosphatase (V-ATPase) is considered to play an important role in the regulation of the acidic microenvironment of some tumors. In this study, we made an investigation on the expression and functional role of V-ATPase in native human hepatocellular carcinoma (HCC). The results showed that the messenger RNA and protein expression levels of V-ATPase subunit ATP6L in native human HCC tissues were markedly increased, compared with normal liver tissues. Immunohistochemical analysis further confirmed the enhanced expression of V-ATPase ATP6L in human HCC cells and revealed that V-ATPase ATP6L was distributed in the cytoplasm and plasma membrane of HCC cells. The results from immunofluorescence and biotinylation of cell surface protein showed that V-ATPase ATP6L was conspicuously located in the plasma membrane of human HCC cells. Bafilomycin A1, a specific V-ATPase inhibitor, markedly slowed the intracellular pH (pHi) recovery after acid load in human HCC cells and retarded the growth of human HCC in orthotopic xenograft model. These results demonstrated that V-ATPase is up-regulated in human HCC and involved in the regulation of pHi of human HCC cells. The inhibition of V-ATPase can effectively retard the growth of HCC, indicating that V-ATPase may play an important role in the development and progression of human HCC, and targeting V-ATPase may be a promising therapeutic strategy against human HCC.
Collapse
Affiliation(s)
- Jingyu Xu
- Department of Gastroenterology, Affiliated Hospital, Zunyi Medical College, Zunyi 563003, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Yeung CH, Wang K, Cooper TG. Why are epididymal tumours so rare? Asian J Androl 2012; 14:465-75. [PMID: 22522502 DOI: 10.1038/aja.2012.20] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Epididymal tumour incidence is at most 0.03% of all male cancers. It is an enigma why the human epididymis does not often succumb to cancer, when it expresses markers of stem and cancer cells, and constitutively expresses oncogenes, pro-proliferative and pro-angiogenic factors that allow tumour cells to escape immunosurveillance in cancer-prone tissues. The privileged position of the human epididymis in evading tumourigenicity is reflected in transgenic mouse models in which induction of tumours in other organs is not accompanied by epididymal neoplasia. The epididymis appears to: (i) prevent tumour initiation (it probably lacks stem cells and has strong anti-oxidative mechanisms, active tumour suppressors and inactive oncogene products); (ii) foster tumour monitoring and destruction (by strong immuno-surveillance and -eradication, and cellular senescence); (iii) avert proliferation and angiogenesis (with persistent tight junctions, the presence of anti-angiogenic factors and misplaced pro-angiogenic factors), which together (iv) promote dormancy and restrict dividing cells to hyperplasia. Epididymal cells may be rendered non-responsive to oncogenic stimuli by the constitutive expression of factors generally inducible in tumours, and resistant to the normal epididymal environment, which mimics that of a tumour niche promoting tumour growth. The threshold for tumour initiation may thus be higher in the epididymis than in other organs. Several anti-tumour mechanisms are those that maintain spermatozoa quiescent and immunologically silent, so the low incidence of cancer in the epididymis may be a consequence of its role in sperm maturation and storage. Understanding these mechanisms may throw light on cancer prevention and therapy in general.
Collapse
Affiliation(s)
- Ching-Hei Yeung
- Shandong Stem Cell Engineering and Technology Research Centre, YuHuangDing Hospital, Yantai, China
| | | | | |
Collapse
|
23
|
Rothberg JM, Sameni M, Moin K, Sloane BF. Live-cell imaging of tumor proteolysis: impact of cellular and non-cellular microenvironment. BIOCHIMICA ET BIOPHYSICA ACTA 2012; 1824:123-32. [PMID: 21854877 PMCID: PMC3232330 DOI: 10.1016/j.bbapap.2011.07.025] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2011] [Revised: 07/28/2011] [Accepted: 07/29/2011] [Indexed: 01/26/2023]
Abstract
Our laboratory has had a longstanding interest in how the interactions between tumors and their microenvironment affect malignant progression. Recently, we have focused on defining the proteolytic pathways that function in the transition of breast cancer from the pre-invasive lesions of ductal carcinoma in situ (DCIS) to invasive ductal carcinomas (IDCs). We use live-cell imaging to visualize, localize and quantify proteolysis as it occurs in real-time and thereby have established roles for lysosomal cysteine proteases both pericellularly and intracellularly in tumor proteolysis. To facilitate these studies, we have developed and optimized 3D organotypic co-culture models that recapitulate the in vivo interactions of mammary epithelial cells or tumor cells with stromal and inflammatory cells. Here we will discuss the background that led to our present studies as well as the techniques and models that we employ. This article is part of a Special Issue entitled: Proteolysis 50 years after the discovery of lysosome.
Collapse
MESH Headings
- Animals
- Breast Neoplasms/metabolism
- Breast Neoplasms/pathology
- Carcinoma, Ductal, Breast/metabolism
- Carcinoma, Ductal, Breast/pathology
- Carcinoma, Intraductal, Noninfiltrating/metabolism
- Carcinoma, Intraductal, Noninfiltrating/pathology
- Cells, Cultured
- Cellular Microenvironment/physiology
- Diagnostic Imaging/methods
- Female
- Humans
- Microscopy, Video
- Models, Biological
- Neoplasms/diagnosis
- Neoplasms/metabolism
- Neoplasms/pathology
- Proteolysis
- Single-Cell Analysis/methods
- Tumor Microenvironment/physiology
Collapse
Affiliation(s)
- Jennifer M Rothberg
- Barbara Ann Karmanos Cancer Institute, Wayne State University, Detroit, MI 48201, USA.
| | | | | | | |
Collapse
|
24
|
Suk J, Kwak SS, Lee JH, Choi JH, Lee SH, Lee DH, Byun B, Lee GH, Joe CO. Alkaline stress-induced autophagy is mediated by mTORC1 inactivation. J Cell Biochem 2011; 112:2566-73. [PMID: 21590709 DOI: 10.1002/jcb.23181] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The activation of autophagic pathway by alkaline stress was investigated. Various types of mammalian cells were subjected to alkaline stress by incubation in bicarbonate buffered media in humidified air containing atmospheric 0.04% CO(2) . The induction of autophagy following alkaline stress was evaluated by assessing the conversion of cytosolic LC3-I into lipidated LC3-II, the accumulation of autophagosomes, and the formation of autolysosomes. Colocalization of GFP-LC3 with endolysosomal marker in HeLa GFP-LC3 cells undergoing autophagic process by alkaline stress further demonstrates that autophagosomes triggered by alkaline stress matures into autolysosomes for the lysosome dependent degradation. We found that the inactivation of mTORC1 is important for the pathway leading to the induction of autophagy by alkaline stress since the expression of RhebQ64L, a constitutive activator of mTORC1, downregulates the induction of autophagy after alkaline stress in transfected human 293T cells. These results imply that activation of autophagic pathway following the inactivation of mTORC1 is important cellular events governing alkaline stress-induced cytotoxicity and clinical symptoms associated with alkalosis.
Collapse
Affiliation(s)
- Jinkyu Suk
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 305-701, South Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Alvarez-Baron CP, Jonsson P, Thomas C, Dryer SE, Williams C. The two-pore domain potassium channel KCNK5: induction by estrogen receptor alpha and role in proliferation of breast cancer cells. Mol Endocrinol 2011; 25:1326-36. [PMID: 21680658 DOI: 10.1210/me.2011-0045] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The growth of many human breast tumors requires the proliferative effect of estrogen acting via the estrogen receptor α (ERα). ERα signaling is therefore a clinically important target for breast cancer prevention and therapeutics. Although extensively studied, the mechanism by which ERα promotes proliferation remains to be fully established. We observed an up-regulation of transcript encoding the pH-sensitive two-pore domain potassium channel KCNK5 in a screen for genes stimulated by 17β-estradiol (E2) in the ERα(+) breast cancer cell lines MCF-7 and T47D. KCNK5 mRNA increased starting 1 h after the onset of E2 treatment, and protein levels followed after 12 h. Estrogen-responsive elements are found in the enhancer region of KCNK5, and chromatin immunoprecipitation assays revealed binding of ERα to the KCNK5 enhancer in E2-treated MCF-7 cells. Cells treated with E2 also showed increases in the amplitude of pH-sensitive potassium currents, as assessed by whole-cell recordings. These currents are blocked by clofilium. Although confocal microscopy suggested that most of the channels are located in intracellular compartments, the increase in macroscopic currents suggests that E2 treatment increases the number of active channels at the cell surface. Application of small interfering RNA specific for KCNK5 decreased pH-sensitive potassium currents and also reduced the estrogen-induced proliferation of T47D cells. We conclude that E2 induces the expression of KCNK5 via ERα(+) in breast cancer cells, and this channel plays a role in regulating proliferation in these cell lines. KCNK5 may therefore represent a useful target for treatment, for example, of tamoxifen-resistant breast cancer.
Collapse
Affiliation(s)
- Claudia P Alvarez-Baron
- Department of Biology and Biochemistry, University of Houston, Houston, Texas 77204-5001, USA
| | | | | | | | | |
Collapse
|
26
|
Hulikova A, Vaughan-Jones RD, Swietach P. Dual role of CO2/HCO3(-) buffer in the regulation of intracellular pH of three-dimensional tumor growths. J Biol Chem 2011; 286:13815-26. [PMID: 21345798 PMCID: PMC3077582 DOI: 10.1074/jbc.m111.219899] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Intracellular pH (pHi), a major modulator of cell function, is regulated by acid/base transport across membranes. Excess intracellular H+ ions (e.g. produced by respiration) are extruded by transporters such as Na+/H+ exchange, or neutralized by HCO3− taken up by carriers such as Na+-HCO3− cotransport. Using fluorescence pHi imaging, we show that cancer-derived cell lines (colorectal HCT116 and HT29, breast MDA-MB-468, pancreatic MiaPaca2, and cervical HeLa) extrude acid by H+ efflux and HCO3− influx, largely sensitive to dimethylamiloride and 4,4′-diisothiocyanatostilbene-2,2′-disulfonate (DIDS), respectively. The magnitude of HCO3− influx was comparable among the cell lines and may represent a constitutive element of tumor pHi regulation. In contrast, H+ efflux varied considerably (MDA-MB-468 > HCT116 > HT29 > MiaPaca2 > HeLa). When HCO3− flux was pharmacologically inhibited, acid extrusion in multicellular HT29 and HCT116 spheroids (∼10,000 cells) was highly non-uniform and produced low pHi at the core. With depth, acid extrusion became relatively more DIDS-sensitive because the low extracellular pH at the spheroid core inhibits H+ flux more than HCO3− flux. HCO3− flux inhibition also decelerated HCT116 spheroid growth. In the absence of CO2/HCO3−, acid extrusion by H+ flux in HCT116 and MDA-MB-468 spheroids became highly non-uniform and inadequate at the core. This is because H+ transporters require extracellular mobile pH buffers, such as CO2/HCO3−, to overcome low H+ ion mobility and chaperone H+ ions away from cells. CO2/HCO3− exerts a dual effect: as substrate for membrane-bound HCO3− transporters and as a mobile buffer for facilitating extracellular diffusion of H+ ions extruded from cells. These processes can be augmented by carbonic anhydrase activity. We conclude that CO2/HCO3− is important for maintaining uniformly alkaline pHi in small, non-vascularized tumor growths and may be important for cancer disease progression.
Collapse
Affiliation(s)
- Alzbeta Hulikova
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3PT, United Kingdom
| | | | | |
Collapse
|
27
|
Rachakatla RS, Balivada S, Seo GM, Myers CB, Wang H, Samarakoon TN, Dani R, Pyle M, Kroh FO, Walker B, Leaym X, Koper OB, Chikan V, Bossmann SH, Tamura M, Troyer DL. Attenuation of mouse melanoma by A/C magnetic field after delivery of bi-magnetic nanoparticles by neural progenitor cells. ACS NANO 2010; 4:7093-104. [PMID: 21058696 PMCID: PMC3011034 DOI: 10.1021/nn100870z] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
Localized magnetic hyperthermia as a treatment modality for cancer has generated renewed interest, particularly if it can be targeted to the tumor site. We examined whether tumor-tropic neural progenitor cells (NPCs) could be utilized as cell delivery vehicles for achieving preferential accumulation of core/shell iron/iron oxide magnetic nanoparticles (MNPs) within a mouse model of melanoma. We developed aminosiloxane-porphyrin functionalized MNPs, evaluated cell viability and loading efficiency, and transplanted neural progenitor cells loaded with this cargo into mice with melanoma. NPCs were efficiently loaded with core/shell Fe/Fe(3)O(4) MNPs with minimal cytotoxicity; the MNPs accumulated as aggregates in the cytosol. The NPCs loaded with MNPs could travel to subcutaneous melanomas, and after A/C (alternating current) magnetic field (AMF) exposure, the targeted delivery of MNPs by the cells resulted in a measurable regression of the tumors. The tumor attenuation was significant (p < 0.05) a short time (24 h) after the last of three AMF exposures.
Collapse
Affiliation(s)
- Raja Shekar Rachakatla
- Deryl Troyer, Department of Anatomy and Physiology, 228 Coles Hall, Kansas State University, Manhattan, KS 66506, USA
| | - Sivasai Balivada
- Deryl Troyer, Department of Anatomy and Physiology, 228 Coles Hall, Kansas State University, Manhattan, KS 66506, USA
| | - Gwi-Moon Seo
- Deryl Troyer, Department of Anatomy and Physiology, 228 Coles Hall, Kansas State University, Manhattan, KS 66506, USA
| | - Carl B Myers
- Department of Diagnostic Pathobiology, 223 Mosier Hall, Kansas State University, Manhattan, KS 66506
| | - Hongwang Wang
- Department of Chemistry, 213 CBC Building, Kansas State University, Manhattan, KS 66506, USA, , phone: 785-532-6817, fax: 785-532-6666, http://www.k-state.edu/chem/
| | - Thilani N. Samarakoon
- Department of Chemistry, 213 CBC Building, Kansas State University, Manhattan, KS 66506, USA, , phone: 785-532-6817, fax: 785-532-6666, http://www.k-state.edu/chem/
| | - Raj Dani
- Department of Chemistry, 213 CBC Building, Kansas State University, Manhattan, KS 66506, USA, , phone: 785-532-6817, fax: 785-532-6666, http://www.k-state.edu/chem/
| | - Marla Pyle
- Deryl Troyer, Department of Anatomy and Physiology, 228 Coles Hall, Kansas State University, Manhattan, KS 66506, USA
| | - Franklin O. Kroh
- NanoScale Corporation, 1310 Research Park Drive, Manhattan, KS 66502, USA, , phone: 785-537-0179, fax: 785-537-0226
| | - Brandon Walker
- NanoScale Corporation, 1310 Research Park Drive, Manhattan, KS 66502, USA, , phone: 785-537-0179, fax: 785-537-0226
| | - Xiaoxuan Leaym
- NanoScale Corporation, 1310 Research Park Drive, Manhattan, KS 66502, USA, , phone: 785-537-0179, fax: 785-537-0226
| | - Olga B. Koper
- NanoScale Corporation, 1310 Research Park Drive, Manhattan, KS 66502, USA, , phone: 785-537-0179, fax: 785-537-0226
| | - Viktor Chikan
- Department of Chemistry, 213 CBC Building, Kansas State University, Manhattan, KS 66506, USA, , phone: 785-532-6817, fax: 785-532-6666, http://www.k-state.edu/chem/
| | - Stefan H. Bossmann
- Department of Chemistry, 213 CBC Building, Kansas State University, Manhattan, KS 66506, USA, , phone: 785-532-6817, fax: 785-532-6666, http://www.k-state.edu/chem/
| | - Masaaki Tamura
- Deryl Troyer, Department of Anatomy and Physiology, 228 Coles Hall, Kansas State University, Manhattan, KS 66506, USA
| | - Deryl L. Troyer
- Deryl Troyer, Department of Anatomy and Physiology, 228 Coles Hall, Kansas State University, Manhattan, KS 66506, USA
- Corresponding author: Deryl Troyer, Department of Anatomy and Physiology, 228 Coles Hall, Kansas State University, Manhattan, KS 66506, USA , phone: 785-532-4509, fax: 785-532-4557
| |
Collapse
|
28
|
Merchan S, Pedelini L, Hueso G, Calzada A, Serrano R, Yenush L. Genetic alterations leading to increases in internal potassium concentrations are detrimental for DNA integrity in Saccharomyces cerevisiae. Genes Cells 2010; 16:152-65. [PMID: 21143561 DOI: 10.1111/j.1365-2443.2010.01472.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
We have investigated the effects of alterations in potassium homeostasis on cell cycle progression and genome stability in Saccharomyces cerevisiae. Yeast strains lacking the PPZ1 and PPZ2 phosphatase genes, which aberrantly accumulate potassium, are sensitive to agents causing replicative stress or DNA damage and present a cell cycle delay in the G(1) /S phase. A synthetic slow growth phenotype was identified in a subset of DNA repair mutants upon inhibition of Ppz activity. Moreover, we observe that this slow growth phenotype observed in cdc7(ts) mutants with reduced Ppz activity is reverted by disrupting the TRK1 potassium transporter gene. As over-expression of a mammalian potassium transporter leads to similar phenotypes, we conclude that these defects can be attributed to potassium accumulation. As we reported previously, internal potassium accumulation activates the Slt2 MAP kinase pathway. We show that the removal of SLT2 in ppz1 ppz2 mutants ameliorates sensitivity to agents causing replication stress and DNA damage, whereas over-activation of the pathway leads to similar cell cycle-related defects. Taken together, these results are consistent with inappropriate potassium accumulation reducing DNA replication efficiency, negatively influencing DNA integrity and leading to the requirement of mismatch repair, the MRX complex, or homologous recombination pathways for normal growth.
Collapse
Affiliation(s)
- Stephanie Merchan
- Instituto de Biología Molecular y Celular de Plantas, Universidad Politécnica de Valencia-Consejo Superior de Investigaciones Científicas, Ciudad Politécnica de Innovación, 46022 Valencia, Spain
| | | | | | | | | | | |
Collapse
|
29
|
New insights into the physiological role of carbonic anhydrase IX in tumour pH regulation. Oncogene 2010; 29:6509-21. [PMID: 20890298 DOI: 10.1038/onc.2010.455] [Citation(s) in RCA: 175] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
In this review, we discuss the role of the tumour-associated carbonic anhydrase isoform IX (CAIX) in the context of pH regulation. We summarise recent experimental findings on the effect of CAIX on cell growth and survival, and present a diffusion-reaction model to help in the assessment of CAIX function under physiological conditions. CAIX emerges as an important facilitator of acid diffusion and acid transport, helping to overcome large cell-to-capillary distances that are characteristic of solid tumours. The source of substrate for CAIX catalysis is likely to be CO₂, generated by adequately oxygenated mitochondria or from the titration of metabolic acids with HCO₃⁻ taken up from the extracellular milieu. The relative importance of these pathways will depend on oxygen and metabolite availability, the spatiotemporal patterns of the cell's exposure to hypoxia and on the regulation of metabolism by genes. This is now an important avenue for further investigation. The importance of CAIX in regulating tumour pH highlights the protein as a potential target for cancer therapy.
Collapse
|
30
|
BAX inhibitor-1 enhances cancer metastasis by altering glucose metabolism and activating the sodium-hydrogen exchanger: the alteration of mitochondrial function. Oncogene 2010; 29:2130-41. [PMID: 20118983 DOI: 10.1038/onc.2009.491] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The anti-apoptotic protein, BAX inhibitor-1 (BI-1), has a role in cancer/tumor progression. BI-1-overexpressing HT1080 and B16F10 cells produced higher lung weights and tumor volumes after injection into the tail veins of mice. Transfection of BI-1 siRNA into cells before injection blocked lung metastasis. in vitro, the overexpression of BI-1 increased cell mobility and invasiveness, with highly increased glucose consumption and cytosolic accumulation of lactate and pyruvate, but decreased mitochondrial O(2) consumption and ATP production. Glucose metabolism-associated extracellular pH also decreased as cells excreted more H(+), and sodium hydrogen exchanger (NHE) activity increased, probably as a homeostatic mechanism for intracellular pH. These alterations activated MMP 2/9 and cell mobility and invasiveness, which were reversed by the NHE inhibitor, 5-(N-ethyl-N-isopropyl) amiloride (EIPA), suggesting a role for NHE in cancer metastasis. In both in vitro and in vivo experiments, C-terminal deleted (CDeltaBI-1) cells showed similar results to control cells, suggesting that the C-terminal motif is required for BI-1-associated alterations of glucose metabolism, NHE activation and cancer metastasis. These findings strongly suggest that BI-1 reduces extracellular pH and regulates metastasis by altering glucose metabolism and activating NHE, with the C-terminal tail having a pivotal role in these processes.
Collapse
|
31
|
Chiche J, Brahimi-Horn MC, Pouysségur J. Tumour hypoxia induces a metabolic shift causing acidosis: a common feature in cancer. J Cell Mol Med 2009; 14:771-94. [PMID: 20015196 PMCID: PMC3823111 DOI: 10.1111/j.1582-4934.2009.00994.x] [Citation(s) in RCA: 459] [Impact Index Per Article: 30.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Maintenance of cellular pH homeostasis is fundamental to life. A number of key intracellular pH (pHi) regulating systems including the Na+/H+ exchangers, the proton pump, the monocarboxylate transporters, the HCO3− transporters and exchangers and the membrane-associated and cytosolic carbonic anhydrases cooperate in maintaining a pHi that is permissive for cell survival. A common feature of tumours is acidosis caused by hypoxia (low oxygen tension). In addition to oncogene activation and transformation, hypoxia is responsible for inducing acidosis through a shift in cellular metabolism that generates a high acid load in the tumour microenvironment. However, hypoxia and oncogene activation also allow cells to adapt to the potentially toxic effects of an excess in acidosis. Hypoxia does so by inducing the activity of a transcription factor the hypoxia-inducible factor (HIF), and particularly HIF-1, that in turn enhances the expression of a number of pHi-regulating systems that cope with acidosis. In this review, we will focus on the characterization and function of some of the hypoxia-inducible pH-regulating systems and their induction by hypoxic stress. It is essential to understand the fundamentals of pH regulation to meet the challenge consisting in targeting tumour metabolism and acidosis as an anti-tumour approach. We will summarize strategies that take advantage of intracellular and extracellular pH regulation to target the primary tumour and metastatic growth, and to turn around resistance to chemotherapy and radiotherapy.
Collapse
Affiliation(s)
- Johanna Chiche
- Institute of Developmental Biology and Cancer Research, University of Nice, CNRS UMR, Centre A. Lacassagne, Nice, France
| | | | | |
Collapse
|
32
|
Resveratrol regulates the expression of NHE-1 by repressing its promoter activity: critical involvement of intracellular H2O2 and caspases 3 and 6 in the absence of cell death. Int J Biochem Cell Biol 2008; 41:945-56. [PMID: 18951995 DOI: 10.1016/j.biocel.2008.09.028] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2008] [Revised: 09/16/2008] [Accepted: 09/16/2008] [Indexed: 01/19/2023]
Abstract
Na(+)/H(+) exchanger-1 (NHE-1) overexpression is associated with carcinogenesis and is an attractive target for intervention. We report that the chemopreventive agent resveratrol (RSV) downregulates NHE-1 in a caspase-dependent manner without inducing cell death. Resveratrol triggered early activation of caspase 3 and late activation of caspase 6, which were not inter-dependent. Whereas, caspase 3 activation appeared to be a direct effect of resveratrol, caspase 6 activation was mediated via intracellular hydrogen peroxide production and iron. Moreover, downregulation of NHE-1 expression was a function of resveratrol-induced repression of NHE-1 gene promoter activity. RNAi-mediated silencing of caspase 3 or 6 blocked the effect of resveratrol on NHE-1 expression, however the effect on NHE-1 promoter was observed at different phases of promoter repression with caspase 3 controlling the early phase (4-12 h) and caspase 6 regulating the late phase (12-24 h). Scavenging hydrogen peroxide or iron only reversed the late phase of resveratrol-induced NHE-1 promoter repression. Finally, an AP2 binding region within NHE-1 gene promoter was identified as the target of resveratrol. Collectively, these data could explain the anti-cancer activity of resveratrol in the light of the association of increased NHE-1 expression with carcinogenesis.
Collapse
|
33
|
Sennoune SR, Martinez-Zaguilan R. Plasmalemmal vacuolar H+-ATPases in angiogenesis, diabetes and cancer. J Bioenerg Biomembr 2008; 39:427-33. [PMID: 18058006 DOI: 10.1007/s10863-007-9108-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Angiogenesis, i.e., new blood vessel formation, is required in normal and pathological states. A dysfunction in the microvascular endothelium occurs in diabetes, leading to decreased blood flow and limb amputation. In cancer, angiogenesis is increased to allow for growth, invasion, and metastasis of tumor cells. Better understanding of the molecular events that cause or are associated with either of these diseases is needed to develop therapies. The tumor and angiogenic cells micro-environment is acidic and not permissive for growth. We have shown that to survive this environment, highly metastatic and angiogenic cells employ vacuolar H+-ATPase at their plasma membranes (pmV-ATPases) to maintain an alkaline pHcyt. However, in lowly metastatic and in microvascular endothelial cells from diabetic model, the density of pmV-ATPase and the cell invasiveness are decreased. Therefore, the overexpression of the pmV-ATPase is important for cell invasion, and essential for tumor progression, angiogenesis and metastasis. Both, cancer and diabetes are heterogenous diseases that involve many different proteins and signaling pathways. Changes in pHcyt have been associated with the regulation of a myriad of proteins, signaling molecules and pathways affecting many if not all cellular functions. Since changes in pHcyt are pleiotropic, we hypothesize that alteration in a single protein, pmV-ATPase, that can regulate pHcyt may explain the dysfunction of many proteins and cellular pathways in diabetes and cancer. Our long term goal is to determine the molecular mechanisms by which pmV-ATPase expression regulates tumor angiogenesis and metastasis. Such knowledge would be useful to identify targets for cancer therapy.
Collapse
Affiliation(s)
- Souad R Sennoune
- Department of Cellular Physiology and Molecular Biophysics, Health Sciences Center, Texas Tech University, 3601 4th Street, Lubbock, TX 79430-6551, USA.
| | | |
Collapse
|
34
|
Kokkonen N, Ulibarri IF, Kauppila A, Luosujärvi H, Rivinoja A, Pospiech H, Kellokumpu I, Kellokumpu S. Hypoxia upregulates carcinoembryonic antigen expression in cancer cells. Int J Cancer 2007; 121:2443-50. [PMID: 17657737 DOI: 10.1002/ijc.22965] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Carcinoembryonic antigen (CEA, ceacam5) is an important tumor-associated antigen with reported roles, e.g., in immunological defense, cell adhesion, cell survival and metastasis. Its overexpression in cancer cells is known to involve transcriptional activation of the CEA gene, but the underlying molecular details remain unclear. Here, we show that hypoxia and intracellular alkalinization, 2 factors commonly found in solid tumors, increase CEA protein expression in breast (MCF-7) and colorectal (CaCo-2 and HT-29) cancer cells. The increase was comparable (2-3-fold) to that observed in colorectal carcinomas in vivo. CEA promoter analyses further revealed that this upregulation involves a known binding site for HIF-1 transcription factor (5'-ACGTG-3') within one of the CEA promoter's positive regulatory elements (the FP1 site; the E-box). Accordingly, deletion or targeted mutagenesis of this motif rendered the CEA promoter unresponsive to hypoxia. Our chromatin immunoprecipitation data confirmed that endogenous HIF-1alpha binds to the CEA promoter in hypoxic cells but not in normoxic cells. Moreover, overexpression of the hypoxia-inducible factor (HIF-1alpha) was sufficient to increase CEA protein expression in the cells. In contrast, c-Myc, which is known to bind to the overlapping E-box, did not potentiate HIF-1alpha-induced CEA expression. CEA overexpression in vivo was also found to coincide with the expression of carbonic anhydrase IX, a well-known hypoxia marker. Collectively, these results define CEA as a hypoxia-inducible protein and suggest an important role for the tumor microenvironmental factors in CEA overexpression during tumorigenesis.
Collapse
Affiliation(s)
- Nina Kokkonen
- Department of Biochemistry, University of Oulu, Oulu, Finland
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Mathupala SP, Colen CB, Parajuli P, Sloan AE. Lactate and malignant tumors: a therapeutic target at the end stage of glycolysis. J Bioenerg Biomembr 2007; 39:73-7. [PMID: 17354062 PMCID: PMC3385854 DOI: 10.1007/s10863-006-9062-x] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Metabolic aberrations in the form of altered flux through key metabolic pathways are primary hallmarks of many malignant tumors. Primarily the result of altered isozyme expression, these adaptations enhance the survival and proliferation of the tumor at the expense of surrounding normal tissue. Consequently, they also expose a unique set of targets for tumor destruction while sparing healthy tissues. Despite this fact, development of drugs to directly target such altered metabolic pathways of malignant tumors has been under-investigated until recently. One such target is the ultimate step of glycolysis, which, as expected, presents itself as a metabolic aberration in most malignant tumors. Termed "aerobic glycolysis" due to abnormal conversion of pyruvic acid to lactic acid even under normoxia, the altered metabolism requires these tumors to rapidly efflux lactic acid to the microenvironment in order to prevent poisoning themselves. Thus, exposed is a prime "choke-point" to target these highly malignant, frequently chemo- and radio- resistant tumors. This review will focus on current outcomes in targeting lactate efflux in such tumors using glioma as a model, an ongoing project in our laboratory for the past half-decade, as well as supporting evidence from recent studies by others on targeting this "tail-end" of glycolysis in other tumor models.
Collapse
Affiliation(s)
- Saroj P Mathupala
- Department of Neurological Surgery and Karmanos Cancer Institute, Wayne State University School of Medicine, 808 HWCRC, 4100 John R. Road, Detroit, MI 48201, USA.
| | | | | | | |
Collapse
|
36
|
Abstract
The high metabolic rate required for tumor growth often leads to hypoxia in poorly-perfused regions. Hypoxia activates a complex gene expression program, mediated by hypoxia inducible factor 1 (HIF1alpha). One of the consequences of HIF1alpha activation is up-regulation of glycolysis and hence the production of lactic acid. In addition to the lactic acid-output, intracellular titration of acid with bicarbonate and the engagement of the pentose phosphate shunt release CO(2) from cells. Expression of the enzyme carbonic anhydrase 9 on the tumor cell surface catalyses the extracellular trapping of acid by hydrating cell-generated CO(2) into [see text] and H(+). These mechanisms contribute towards an acidic extracellular milieu favoring tumor growth, invasion and development. The lactic acid released by tumor cells is further metabolized by the tumor stroma. Low extracellular pH may adversely affect the intracellular milieu, possibly triggering apoptosis. Therefore, primary and secondary active transporters operate in the tumor cell membrane to protect the cytosol from acidosis. We review mechanisms regulating tumor intracellular and extracellular pH, with a focus on carbonic anhydrase 9. We also review recent evidence that may suggest a role for CA9 in coordinating pH(i) among cells of large, unvascularized cell-clusters.
Collapse
Affiliation(s)
- Pawel Swietach
- Department of Physiology, Anatomy and Genetics, Burdon Sanderson Cardiac Science Centre, Oxford OX1 3PT, UK
| | | | | |
Collapse
|
37
|
Navrátilová J, Horváth V, Kozubík A, Lojek A, Lipsick J, Smarda J. p53 arrests growth and induces differentiation of v-Myb-transformed monoblasts. Differentiation 2007; 75:592-604. [PMID: 17309603 DOI: 10.1111/j.1432-0436.2006.00158.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The p53 protein can control cell cycle progression, programmed cell death, and differentiation of many cell types. Ectopic expression of p53 can resume capability of cell cycle arrest, differentiation, and apoptosis in various leukemic cell lines. In this work, we expressed human p53 protein in v-Myb-transformed chicken monoblasts. We found that even this protein possessing only 53% amino acid homology to its avian counterpart can significantly alter morphology and physiology of these cells causing the G2-phase cell cycle arrest and early monocytic differentiation. Our results document that the species-specific differences of the p53 molecules, promoters/enhancers, and co-factors in avian and human cells do not interfere with differentiation- and cell cycle arrest promoting capabilites of the p53 tumor suppressor even in the presence of functional v-Myb oncoprotein. The p53-induced differentiation and cell cycle arrest of v-Myb-transformed monoblasts are not associated with apoptosis suggesting that the p53-driven pathways controlling apoptosis and differentiation/proliferation are independent.
Collapse
Affiliation(s)
- Jarmila Navrátilová
- Institute of Experimental Biology, Faculty of Science, Masaryk University, Kotlárská 2, 611 37 Brno, Czech Republic
| | | | | | | | | | | |
Collapse
|
38
|
Rojas JD, Sennoune SR, Maiti D, Bakunts K, Reuveni M, Sanka SC, Martinez GM, Seftor EA, Meininger CJ, Wu G, Wesson DE, Hendrix MJC, Martínez-Zaguilán R. Vacuolar-type H+-ATPases at the plasma membrane regulate pH and cell migration in microvascular endothelial cells. Am J Physiol Heart Circ Physiol 2006; 291:H1147-57. [PMID: 16679513 DOI: 10.1152/ajpheart.00166.2006] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Microvascular endothelial cells involved in angiogenesis are exposed to an acidic environment that is not conducive for growth and survival. These cells must exhibit a dynamic intracellular (cytosolic) pH (pHcyt) regulatory mechanism to cope with acidosis, in addition to the ubiquitous Na+/H+exchanger and HCO3−-based H+-transporting systems. We hypothesize that the presence of plasmalemmal vacuolar-type proton ATPases (pmV-ATPases) allows microvascular endothelial cells to better cope with this acidic environment and that pmV-ATPases are required for cell migration. This study indicates that microvascular endothelial cells, which are more migratory than macrovascular endothelial cells, express pmV-ATPases. Spectral imaging microscopy indicates a more alkaline pHcytat the leading than at the lagging edge of microvascular endothelial cells. Treatment of microvascular endothelial cells with V-ATPase inhibitors decreases the proton fluxes via pmV-ATPases and cell migration. These data suggest that pmV-ATPases are essential for pHcytregulation and cell migration in microvascular endothelial cells.
Collapse
Affiliation(s)
- J D Rojas
- Department of Physiology, Texas Tech University Health Sciences Center, 3601 4th St., Lubbock, TX 79430-6551, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Wu D, Doods H, Stassen JM. Inhibition of Human Pulmonary Artery Smooth Muscle Cell Proliferation and Migration by Sabiporide, a New Specific NHE-1 Inhibitor. J Cardiovasc Pharmacol 2006; 48:34-40. [PMID: 16954819 DOI: 10.1097/01.fjc.0000239691.69346.6a] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Abnormal growth of vascular smooth muscle cells is seen in various pathological conditions such as hypertension, atherosclerosis, and restenosis. Na/H exchanger (NHE) activation appears to play a permissive role in vascular smooth muscle cell proliferation and vascular remodeling. The present study investigated the effect of a new specific NHE-1 inhibitor, sabiporide, on human pulmonary artery smooth muscle cell proliferation and migration. Concentrations of sabiporide as low as 20 micromol/L in the culture medium containing growth factors inhibited cell proliferation, as measured by cell counting, and also inhibited the rate of DNA synthesis, as examined by measuring BrdU incorporation into DNA. Cell growth inhibition was not caused by cell death, as demonstrated by the measurement of intracellular lactate dehydrogenase release and by the reversibility of inhibition upon washing. By fluorescent-activated cell sorting analysis, we are the first to demonstrate that NHE-1 inhibition arrests the cell cycle progression at G0/G1 phase, suggesting that NHE activation plays a permissive role in entrance of cells into the cell cycle. Sabiporide also concentration-dependently inhibited human pulmonary artery smooth muscle cell migration. The present study showed that sabiporide inhibits vascular smooth muscle cell proliferation and migration by blocking the cell cycle progression at G0/G1 phase.
Collapse
Affiliation(s)
- Dongmei Wu
- Department of Research, Mount Sinai Medical Center, Miami Beach, FL 33140, USA.
| | | | | |
Collapse
|
40
|
Marín-Hernández A, Rodríguez-Enríquez S, Vital-González PA, Flores-Rodríguez FL, Macías-Silva M, Sosa-Garrocho M, Moreno-Sánchez R. Determining and understanding the control of glycolysis in fast-growth tumor cells. Flux control by an over-expressed but strongly product-inhibited hexokinase. FEBS J 2006; 273:1975-88. [PMID: 16640561 DOI: 10.1111/j.1742-4658.2006.05214.x] [Citation(s) in RCA: 148] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Control analysis of the glycolytic flux was carried out in two fast-growth tumor cell types of human and rodent origin (HeLa and AS-30D, respectively). Determination of the maximal velocity (V(max)) of the 10 glycolytic enzymes from hexokinase to lactate dehydrogenase revealed that hexokinase (153-306 times) and phosphofructokinase-1 (PFK-1) (22-56 times) had higher over-expression in rat AS-30D hepatoma cells than in normal freshly isolated rat hepatocytes. Moreover, the steady-state concentrations of the glycolytic metabolites, particularly those of the products of hexokinase and PFK-1, were increased compared with hepatocytes. In HeLa cells, V(max) values and metabolite concentrations for the 10 glycolytic enzyme were also significantly increased, but to a much lesser extent (6-9 times for both hexokinase and PFK-1). Elasticity-based analysis of the glycolytic flux in AS-30D cells showed that the block of enzymes producing Fru(1,6)P2 (i.e. glucose transporter, hexokinase, hexosephosphate isomerase, PFK-1, and the Glc6P branches) exerted most of the flux control (70-75%), whereas the consuming block (from aldolase to lactate dehydrogenase) exhibited the remaining control. The Glc6P-producing block (glucose transporter and hexokinase) also showed high flux control (70%), which indicated low flux control by PFK-1. Kinetic analysis of PFK-1 showed low sensitivity towards its allosteric inhibitors citrate and ATP, at physiological concentrations of the activator Fru(2,6)P2. On the other hand, hexokinase activity was strongly inhibited by high, but physiological, concentrations of Glc6P. Therefore, the enhanced glycolytic flux in fast-growth tumor cells was still controlled by an over-produced, but Glc6P-inhibited hexokinase.
Collapse
Affiliation(s)
- Alvaro Marín-Hernández
- Instituto Nacional de Cardiología, Departamento de Bioquímica, Juan Badiano no. 1, Colonia Sección XVI, Tlalpan, México, Mexico
| | | | | | | | | | | | | |
Collapse
|
41
|
Yenush L, Merchan S, Holmes J, Serrano R. pH-Responsive, posttranslational regulation of the Trk1 potassium transporter by the type 1-related Ppz1 phosphatase. Mol Cell Biol 2005; 25:8683-92. [PMID: 16166647 PMCID: PMC1265754 DOI: 10.1128/mcb.25.19.8683-8692.2005] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Intracellular pH and K+ concentrations must be tightly controlled because they affect many cellular activities, including cell growth and death. The mechanisms of homeostasis of H+ and K+ are only partially understood. In the yeast Saccharomyces cerevisiae, proton efflux is mediated by the Pma1 H+-ATPase. As this pump is electrogenic, the activity of the Trk1 and -2 K+ uptake system is crucial for sustained Pma1p operation. The coordinated activities of these two systems determine cell volume, turgor, membrane potential, and pH. Genetic evidence indicates that Trk1p is activated by the Hal4 and -5 kinases and inhibited by the Ppz1 and -2 phosphatases, which, in turn, are inhibited by their regulatory subunit, Hal3p. We show that Trk1p, present in plasma membrane "rafts", physically interacts with Ppz1p, that Trk1p is phosphorylated in vivo, and that its level of phosphorylation increases in ppz1 and -2 mutants. Interestingly, both the interaction with and inhibition of Ppz1p by Hal3p are pH dependent. These results are consistent with a model in which the Ppz1-Hal3 interaction is a sensor of intracellular pH that modulates H+ and K+ homeostasis through the regulation of Trk1p activity.
Collapse
Affiliation(s)
- Lynne Yenush
- Instituto de Biología Molecular y Celular de Plantas, Universidad Politécnica de Valencia CSIC, Camino de Vera s/n, 46022 Valencia, Spain.
| | | | | | | |
Collapse
|
42
|
Lin KA, Chen JH, Lee DF, Lin LY. Alkaline induces metallothionein gene expression and potentiates cell proliferation in Chinese hamster ovary cells. J Cell Physiol 2005; 205:428-36. [PMID: 15965962 DOI: 10.1002/jcp.20417] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Metallothionein (MT) gene expression is increased in cadmium resistant Chinese hamster ovary cells (CHO Cd(R)) upon medium (regular or serum-free) change during culturing. Among the major components of the medium, NaHCO3 was found to be able to induce MT gene expression in a dose- and time-dependent manner. The same effect was observed with other alkaline solutions, such as HEPES and NaOH. Using MT promoter-luciferase reporter gene constructs, we found that the presence of metal response elements (MREs) in the promoter region is necessary for NaHCO3-induced MT gene transcription. This finding is further supported by the observation that the binding activity between the metal-responsive transcription factor 1 (MTF-1) and the MRE were increased after NaHCO3 treatment. Following NaHCO3 treatment, an increase in cell proliferation was observed in CdR cells but not in the parental CHO K1 cells that do not express MT transcripts due to MT gene methylation. Using synchronized cells, an increase in cell proliferation was observed 9 h after NaHCO3 addition. Notably, proliferation of CHO K1 cells was increased when transfected with an MT gene. The effect of MT on cell growth was affirmed by treating CHO K1 cells with 5-azacytidine (Aza) to demethylate the MT gene. Proliferation increased in Aza-treated CHO K1 cells after NaHCO3 treatment. These results demonstrate that NaHCO3 stimulates MT gene expression and causes an enhancement of cell proliferation in CHO cells.
Collapse
Affiliation(s)
- Kuei-Ann Lin
- Institute of Radiation Biology and Department of Life Science, National Tsing Hua University, Hsinchu, Taiwan
| | | | | | | |
Collapse
|
43
|
Harguindey S, Orive G, Luis Pedraz J, Paradiso A, Reshkin SJ. The role of pH dynamics and the Na+/H+ antiporter in the etiopathogenesis and treatment of cancer. Two faces of the same coin--one single nature. Biochim Biophys Acta Rev Cancer 2005; 1756:1-24. [PMID: 16099110 DOI: 10.1016/j.bbcan.2005.06.004] [Citation(s) in RCA: 101] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2004] [Accepted: 06/30/2005] [Indexed: 12/01/2022]
Abstract
Looked at from the genetic point-of-view cancer represents a daunting and, frankly, confusing multiplicity of diseases (at least 100) that require an equally large variety of therapeutic strategies and substances designed to treat the particular tumor. However, when analyzed phenotypically cancer is a relatively uniform disease of very conserved 'hallmark' behaviors across the entire spectrum of tissue and genetic differences [D. Hanahan, R.A. Weinberg, Hallmarks of cancer, Cell 100 (2000) 57-70]. This suggests that cancers do, indeed, share common biochemical and physiological characteristics that are independent of the varied genetic backgrounds, and that there may be a common mechanism underlying both the neoplastic transformation/progression side and the antineoplastic/therapy side of oncology. The challenge of modern oncology is to integrate all the diverse experimental data to create a physiological/metabolic/energetic paradigm that can unite our thinking in order to understand how both neoplastic progression and therapies function. This reductionist view gives the hope that, as in chemistry and physics, it will possible to identify common underlying driving forces that define a tumor and will permit, for the first time, the actual calculated manipulation of their state. That is, a rational therapeutic design. In the present review, we present evidence, obtained from a great number of studies, for a fundamental, underlying mechanism involved in the initiation and evolution of the neoplastic process. There is an ever growing body of evidence that all the important neoplastic phenotypes are driven by an alkalization of the transformed cell, a process which seems specific for transformed cells since the same alkalinization has no effect in cells that have not been transformed. Seen in that light, different fields of cancer research, from etiopathogenesis, cancer cell metabolism and neovascularization, to multiple drug resistance (MDR), selective apoptosis, modern cancer chemotherapy and the spontaneous regression of cancer (SRC) all appear to have in common a pivotal characteristic, the aberrant regulation of hydrogen ion dynamics [S. Harguindey, J.L. Pedraz, R. García Cañero, J. Pérez de Diego, E.J. Cragoe Jr., Hydrogen ion-dependent oncogenesis and parallel new avenues to cancer prevention and treatment using a H+-mediated unifying approach: pH-related and pH-unrelated mechanisms, Crit. Rev. Oncog. 6 (1) (1995) 1-33]. Cancer cells have an acid-base disturbance that is completely different than observed in normal tissues and that increases in correspondence with increasing neoplastic state: an interstitial acid microenvironment linked to an intracellular alkalosis.
Collapse
|
44
|
Lu X, Qin W, Li J, Tan N, Pan D, Zhang H, Xie L, Yao G, Shu H, Yao M, Wan D, Gu J, Yang S. The growth and metastasis of human hepatocellular carcinoma xenografts are inhibited by small interfering RNA targeting to the subunit ATP6L of proton pump. Cancer Res 2005; 65:6843-9. [PMID: 16061667 DOI: 10.1158/0008-5472.can-04-3822] [Citation(s) in RCA: 131] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Extracellular pH is usually low in solid tumors, in contrast to the approximately neutral intracellular pH. V-ATPase, which overly functions in some cancers with metastatic potential, plays an important role in maintaining neutral cytosolic pH, very acidic luminal pH, and acidic extracellular pH. ATP6L, the 16 kDa subunit of proton pump V-ATPase, can provide proton hydrophilic transmembrane path. In this study, ATP6L in a human hepatocellular carcinoma cell line with highly metastatic potential (HCCLM3) was knocked down using DNA vector-based small interfering RNA (siRNA) to suppress the metastasis. The expression of ATP6L in stable siRNA transfectants, designated as si-HCCLM3 cells, was inhibited by approximately 60%. The proton secretion and the intracellular pH recovery from NH4Cl-prepulsed acidification were inhibited in si-HCCLM3 cells. The invasion of the si-HCCLM3 cells was suppressed in vitro; simultaneously, the expressions of matrix metalloproteinase-2 and gelatinase activity were reduced. In vivo, at 35th day after implantation of the si-HCCLM3 xenografts into the livers in BalB/c (nu+/nu+) mice, the size of liver tumor tissues was dramatically smaller in siRNA group than in the controlled group. The most impressing effect of ATP6L siRNA is its striking reduction of the metastatic potential of HCCLM3 cells. In control, all eight mice had the intrahepatic metastasis and six of eight the pulmonary metastasis, whereas in ATP6L siRNA-treated group, three of eight had the intrahepatic metastasis and only one of eight the pulmonary metastasis. The results suggest that the inhibition of V-ATPase function via knockdown of ATP6L expression using RNA interfering technology can effectively retard the cancer growth and suppress the cancer metastasis by the decrease of proton extrusion and the down-regulation of gelatinase activity.
Collapse
Affiliation(s)
- Xiaodong Lu
- National Laboratory for Oncogenes and Related Genes, WHO Collaborating Center for Research on Cancer, Shanghai Cancer Institute, Shanghai, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Cutaia M, Black AD, Cohen I, Cassai ND, Sidhu GS. Alkaline stress-induced apoptosis in human pulmonary artery endothelial cells. Apoptosis 2005; 10:1457-67. [PMID: 16215687 DOI: 10.1007/s10495-005-1402-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
The effect of alkaline stress, or an increase in extracellular pH (pHext), on cell viability is poorly defined. Human pulmonary artery endothelial cells (HPAEC) were subjected to alkaline stress using different methods of increasing pHext. Viability and mode of cell death following alkaline stress were determined by assessing nuclear morphology, ultrastructural features, and caspase-3 activity. Incubation of monolayers in media set to different pHext values (7.4-8.4) for 24-h induced morphological changes suggesting apoptosis (35-45% apoptotic cells) following severe alkaline stress. The magnitude of apoptosis was related to the severity of alkaline stress. These findings were confirmed with an assessment of ultrastructural changes and caspase-3 activation. While there was no difference in the intracellular calcium level ([Ca(2+)](i)) in monolayers set to pHext 7.4 versus 8.4 following the first hour of alkaline stress, blockade of calcium uptake with the chelator, EGTA, potentiated the magnitude of apoptosis under these conditions. Potentiation of apoptosis was reduced by calcium supplementation of the media. Finally, alkaline stress was associated with an increase in intracellular pH. This is the first report of apoptosis following alkaline stress in endothelial cells in the absence of other cell death stimuli.
Collapse
Affiliation(s)
- M Cutaia
- Pulmonary Disease Section, Department of Medicine, Veterans Administration Medical Center, Brooklyn Campus, SUNY/Downstate Health Sciences Center, Brooklyn, NY 11209-7104, USA.
| | | | | | | | | |
Collapse
|
46
|
Sánchez-Armáss S, Sennoune SR, Maiti D, Ortega F, Martínez-Zaguilán R. Spectral imaging microscopy demonstrates cytoplasmic pH oscillations in glial cells. Am J Physiol Cell Physiol 2005; 290:C524-38. [PMID: 16135543 DOI: 10.1152/ajpcell.00290.2005] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Glial cells exhibit distinct cellular domains, somata, and filopodia. Thus the cytoplasmic pH (pH(cyt)) and/or the behavior of the fluorescent ion indicator might be different in these cellular domains because of distinct microenvironments. To address these issues, we loaded C6 glial cells with carboxyseminaphthorhodafluor (SNARF)-1 and evaluated pH(cyt) using spectral imaging microscopy. This approach allowed us to study pH(cyt) in discrete cellular domains with high temporal, spatial, and spectral resolution. Because there are differences in the cell microenvironment that may affect the behavior of SNARF-1, we performed in situ titrations in discrete cellular regions of single cells encompassing the somata and filopodia. The in situ titration parameters apparent acid-base dissociation constant (pK'(a)), maximum ratio (R(max)), and minimum ratio (R(min)) had a mean coefficient of variation approximately six times greater than those measured in vitro. Therefore, the individual in situ titration parameters obtained from specific cellular domains were used to estimate the pH(cyt) of each region. These studies indicated that glial cells exhibit pH(cyt) heterogeneities and pH(cyt) oscillations in both the absence and presence of physiological HCO(3)(-). The amplitude and frequency of the pH(cyt) oscillations were affected by alkalosis, by acidosis, and by inhibitors of the ubiquitous Na(+)/H(+) exchanger- and HCO(3)(-)-based H(+)-transporting mechanisms. Optical imaging approaches used in conjunction with BCECF as a pH probe corroborated the existence of pH(cyt) oscillations in glial cells.
Collapse
Affiliation(s)
- Sergio Sánchez-Armáss
- Departamento de Fisiología y Farmacología, Facultad de Medicina, Universidad Autónoma de San Luis Potosí, San Luis Potosí, Mexico
| | | | | | | | | |
Collapse
|
47
|
Mathupala SP, Parajuli P, Sloan AE. Silencing of monocarboxylate transporters via small interfering ribonucleic acid inhibits glycolysis and induces cell death in malignant glioma: an in vitro study. Neurosurgery 2005; 55:1410-9; discussion 1419. [PMID: 15574223 DOI: 10.1227/01.neu.0000143034.62913.59] [Citation(s) in RCA: 110] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2004] [Accepted: 08/06/2004] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE Dependence on glycolysis is a hallmark of malignant tumors. As a consequence, these tumors generate more lactate, which is effluxed from cells by monocarboxylate transporters (MCTs). We hypothesized that 1) MCT expression in malignant tumors may differ from normal tissue in quantity, isoform, or both; and 2) silencing MCT expression would induce intracellular acidification, resulting in decreased proliferation and/or increased cell death. METHODS We quantified expression of MCT isoforms in human glioblastoma multiforme and glioma-derived cells lines by Western blot analysis. MCTs that were abundant or specific to glioma then were targeted in the model U-87 MG glioma cell line via small interfering ribonucleic acid-mediated gene silencing and tested for inhibition of lactate efflux, intracellular pH changes, reduced proliferation, and/or induction of cell death. RESULTS MCT 1 and 2 were the primary isoforms expressed in human glioblastoma multiforme and glioma-derived cell lines. In contrast, MCT 3 was the predominantly expressed isoform in normal brain. Small interfering ribonucleic acid specific for MCT 1 and 2 reduced expression of these isoforms in U-87 MG cells to barely detectable levels and reduced lactate efflux by 30% individually and 85% in combination, with a concomitant decrease of intracellular pH by 0.6 units (a fourfold increase in intracellular H(+)). Prolonged silencing of both MCTs reduced viability by 75% individually and 92% in combination, as measured by both phenotypic and flow cytometric analyses. CONCLUSION MCT targeting significantly reduced the viability of U-87 MG cells mediated by both apoptosis and necrosis. This indicates that the strategy may be a useful therapeutic avenue for treatment of patients with malignant glioma.
Collapse
Affiliation(s)
- Saroj P Mathupala
- Department of Neurological Surgery and Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, Michigan 48201, USA.
| | | | | |
Collapse
|
48
|
Raghunand N, Gatenby RA, Gillies RJ. Microenvironmental and cellular consequences of altered blood flow in tumours. Br J Radiol 2004; 76 Spec No 1:S11-22. [PMID: 15456710 DOI: 10.1259/bjr/12913493] [Citation(s) in RCA: 163] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Tumour angiogenesis is triggered by various signals characteristic of the tumour microenvironment, including low oxygen tension, low extracellular pH and low glucose concentration. Tumour microvasculature is chaotic, producing perfusion heterogeneities which can be visualized by MRI and other modalities. Inefficient perfusion in tumours produces regions of transient and chronic hypoxia. Tumour hypoxia is associated with adverse clinical outcomes and reduced patient survival. Hypoxia may be a factor in activation of extracellular matrix-degrading proteases, and some studies have correlated primary tumour hypoxia with likelihood of tumour cell dissemination. Exposure to hypoxia either induces or selects for cells that are hyperglycolytic, and this in turn produces local acidosis which is also a common feature of solid tumours. Increased glucose uptake in hyperglycolyzing tumour cells is the basis of lesion-visualization in positron emission tomography using 18F-fluorodeoxyglucose. Tumour acidity can reduce the effectiveness of weak-base drugs, but can be exploited to increase the anti-tumour activity of weak-acid chemotherapeutics. Evidence linking tumour acidity with increased activity of several extracellular matrix-degrading enzyme systems is examined. High levels of lactate, another end-product of glycolysis, in primary lesions have been correlated with increased likelihood of metastasis. In the numerous studies correlating hypoxia, acidity and lactate with metastasis, the direction of the causality has not been adequately established. We hypothesize that adoption of a hyperglycolytic phenotype is a necessary feature of carcinogenesis itself, and confers a survival and proliferative advantage to tumour cells over surrounding normal cells. Empirical evidence supporting this "acid-mediated tumour invasion" model is discussed.
Collapse
Affiliation(s)
- N Raghunand
- Department of Biochemistry and Molecular Biophysics, University of Arizona Health Sciences Center, Tucson, AZ 85724-5024, USA
| | | | | |
Collapse
|
49
|
Abstract
If carcinogenesis occurs by somatic evolution, then common components of the cancer phenotype result from active selection and must, therefore, confer a significant growth advantage. A near-universal property of primary and metastatic cancers is upregulation of glycolysis, resulting in increased glucose consumption, which can be observed with clinical tumour imaging. We propose that persistent metabolism of glucose to lactate even in aerobic conditions is an adaptation to intermittent hypoxia in pre-malignant lesions. However, upregulation of glycolysis leads to microenvironmental acidosis requiring evolution to phenotypes resistant to acid-induced cell toxicity. Subsequent cell populations with upregulated glycolysis and acid resistance have a powerful growth advantage, which promotes unconstrained proliferation and invasion.
Collapse
Affiliation(s)
- Robert A Gatenby
- Department of Radiology, University of Arizona, Tucson, Arizona 85721, USA.
| | | |
Collapse
|
50
|
Turturro F, Friday E, Fowler R, Surie D, Welbourne T. Troglitazone Acts on Cellular pH and DNA Synthesis through a Peroxisome Proliferator-Activated Receptor γ-Independent Mechanism in Breast Cancer-Derived Cell Lines. Clin Cancer Res 2004; 10:7022-30. [PMID: 15501982 DOI: 10.1158/1078-0432.ccr-04-0879] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE The purpose of this study was to assess whether troglitazone (TRO) would induce cellular acidosis by inhibiting Na(+)/H(+) exchanger (NHE) 1 in breast carcinoma-derived cell lines and, if so, whether cellular acidosis would be associated with a reduction in proliferation. EXPERIMENTAL DESIGN Intracellular pH (pH(i)) and acid extrusion capacity after an exogenous acid load were assayed using (2, 7)-biscarboxyethyl-5(6)-carboxyfluorescein in MCF-7 and MDA-MB-231 cells treated with TRO. Radiolabeled thymidine incorporation was used to assess DNA synthesis. Peroxisome proliferator-activated receptor (PPAR) gamma involvement was assessed using an antagonist and PPARgamma(-/-) NIH3T3 cells. RESULTS TRO induced a prompt (<4 minute) and severe cellular acidosis in both MCF-7 (7.54 +/- 0.23 to 6.77 +/- 0.06; P < 0.001) and MDA-MB-231 cells (7.38 +/- 0.18 to 6.89 +/- 0.25; P < 0.05) after 12 minutes, without increasing acid production. Acid extrusion as assessed by the response to an exogenous acid load (NH(4)Cl pulse) was markedly blunted (MDA-MB-231, P < 0.01) or eliminated (MCF-7, P < 0.001). Chronic exposure to TRO resulted in NHE1 activity reduction (P < 0.05) and a dose-dependent decrease in DNA synthesis (<75% inhibition at 100 micromol/L; P < 0.001 and P < 0.01 for MCF-7 and MDA-MB-231, respectively) associated with a decreased number of viable cells. TRO-mediated inhibition of proliferation was not reversed by the presence of the PPARgamma inhibitor GW9662 and was demonstrable in PPARgamma(-/-) NIH3T3 cells, consistent with a PPARgamma-independent mechanism. CONCLUSIONS TRO induces marked cellular acidosis in MCF-7 and MDA-MD-231 cells. Sustained acidosis is consonant with decreased proliferation and growth that is not reversed by a PPARgamma antagonist. Our results support a NHE-mediated action of TRO that exerts its effect independent of PPARgamma.
Collapse
Affiliation(s)
- Francesco Turturro
- Department of Medicine, Feist-Weiller Cancer Center, Gene Therapy Program, Louisiana State University Health Sciences Center, Shreveport, Louisiana 71130, USA.
| | | | | | | | | |
Collapse
|