1
|
Chen YJN, Shi RC, Xiang YC, Fan L, Tang H, He G, Zhou M, Feng XZ, Tan JD, Huang P, Ye X, Zhao K, Fu WY, Li LL, Bian XT, Chen H, Wang F, Wang T, Zhang CK, Zhou BH, Chen W, Liang TT, Lv JT, Kang X, Shi YX, Kim E, Qin YH, Hettinghouse A, Wang KD, Zhao XL, Yang MY, Tang YZ, Piao HL, Guo L, Liu CJ, Miao HM, Tang KL. Malate initiates a proton-sensing pathway essential for pH regulation of inflammation. Signal Transduct Target Ther 2024; 9:367. [PMID: 39737965 DOI: 10.1038/s41392-024-02076-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 11/14/2024] [Accepted: 11/15/2024] [Indexed: 01/01/2025] Open
Abstract
Metabolites can double as a signaling modality that initiates physiological adaptations. Metabolism, a chemical language encoding biological information, has been recognized as a powerful principle directing inflammatory responses. Cytosolic pH is a regulator of inflammatory response in macrophages. Here, we found that L-malate exerts anti-inflammatory effect via BiP-IRF2BP2 signaling, which is a sensor of cytosolic pH in macrophages. First, L-malate, a TCA intermediate upregulated in pro-inflammatory macrophages, was identified as a potent anti-inflammatory metabolite through initial screening. Subsequent screening with DARTS and MS led to the isolation of L-malate-BiP binding. Further screening through protein‒protein interaction microarrays identified a L-malate-restrained coupling of BiP with IRF2BP2, a known anti-inflammatory protein. Interestingly, pH reduction, which promotes carboxyl protonation of L-malate, facilitates L-malate and carboxylate analogues such as succinate to bind BiP, and disrupt BiP-IRF2BP2 interaction in a carboxyl-dependent manner. Both L-malate and acidification inhibit BiP-IRF2BP2 interaction, and protect IRF2BP2 from BiP-driven degradation in macrophages. Furthermore, both in vitro and in vivo, BiP-IRF2BP2 signal is required for effects of both L-malate and pH on inflammatory responses. These findings reveal a previously unrecognized, proton/carboxylate dual sensing pathway wherein pH and L-malate regulate inflammatory responses, indicating the role of certain carboxylate metabolites as adaptors in the proton biosensing by interactions between macromolecules.
Collapse
Affiliation(s)
- Yu-Jia-Nan Chen
- Department of Orthopedic Surgery/Sports Medicine Center, Southwest Hospital, Army Medical University, Chongqing, 400038, China.
- Department of Pathophysiology, College of High Altitude Military Medicine, Army Medical University, Chongqing, 400038, China.
- Department of Orthopedic Surgery, NYU Grossman School of Medicine, New York, NY, 10003, USA.
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases & Department of Neurology, The First Affiliated Hospital, Chongqing Medical University, 400016, Chongqing, China.
- Department of Biochemistry and Molecular Biology, Army Medical University, Chongqing, 400038, China.
| | - Rong-Chen Shi
- Department of Pathophysiology, College of High Altitude Military Medicine, Army Medical University, Chongqing, 400038, China
- Department of Biochemistry and Molecular Biology, Army Medical University, Chongqing, 400038, China
| | - Yuan-Cai Xiang
- Department of Pathophysiology, College of High Altitude Military Medicine, Army Medical University, Chongqing, 400038, China
- Department of Biochemistry and Molecular Biology, Army Medical University, Chongqing, 400038, China
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Li Fan
- Department of Orthopedic Surgery/Sports Medicine Center, Southwest Hospital, Army Medical University, Chongqing, 400038, China
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases & Department of Neurology, The First Affiliated Hospital, Chongqing Medical University, 400016, Chongqing, China
| | - Hong Tang
- Department of Orthopedic Surgery/Sports Medicine Center, Southwest Hospital, Army Medical University, Chongqing, 400038, China
| | - Gang He
- Department of Orthopedic Surgery/Sports Medicine Center, Southwest Hospital, Army Medical University, Chongqing, 400038, China
| | - Mei Zhou
- Department of Orthopedic Surgery/Sports Medicine Center, Southwest Hospital, Army Medical University, Chongqing, 400038, China
| | - Xin-Zhe Feng
- Department of Orthopedic Surgery, NYU Grossman School of Medicine, New York, NY, 10003, USA
| | - Jin-Dong Tan
- Department of Orthopedic Surgery/Sports Medicine Center, Southwest Hospital, Army Medical University, Chongqing, 400038, China
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Pan Huang
- Department of Orthopedic Surgery/Sports Medicine Center, Southwest Hospital, Army Medical University, Chongqing, 400038, China
| | - Xiao Ye
- Department of Orthopedic Surgery/Sports Medicine Center, Southwest Hospital, Army Medical University, Chongqing, 400038, China
| | - Kun Zhao
- Department of Pathophysiology, College of High Altitude Military Medicine, Army Medical University, Chongqing, 400038, China
- Department of Biochemistry and Molecular Biology, Army Medical University, Chongqing, 400038, China
| | - Wen-Yu Fu
- Department of Orthopedic Surgery, NYU Grossman School of Medicine, New York, NY, 10003, USA
- Department of Orthopedics and Rehabilitations, Yale University School of Medicine, New Haven, CT, 06519, USA
| | - Liu-Li Li
- Department of Pathophysiology, College of High Altitude Military Medicine, Army Medical University, Chongqing, 400038, China
| | - Xu-Ting Bian
- Department of Orthopedic Surgery/Sports Medicine Center, Southwest Hospital, Army Medical University, Chongqing, 400038, China
| | - Huan Chen
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
| | - Feng Wang
- Department of Orthopedic Surgery/Sports Medicine Center, Southwest Hospital, Army Medical University, Chongqing, 400038, China
| | - Teng Wang
- Department of Pathophysiology, College of High Altitude Military Medicine, Army Medical University, Chongqing, 400038, China
- Department of Biochemistry and Molecular Biology, Army Medical University, Chongqing, 400038, China
| | - Chen-Ke Zhang
- Department of Orthopedic Surgery/Sports Medicine Center, Southwest Hospital, Army Medical University, Chongqing, 400038, China
| | - Bing-Hua Zhou
- Department of Orthopedic Surgery/Sports Medicine Center, Southwest Hospital, Army Medical University, Chongqing, 400038, China
| | - Wan Chen
- Department of Orthopedic Surgery/Sports Medicine Center, Southwest Hospital, Army Medical University, Chongqing, 400038, China
| | - Tao-Tao Liang
- Department of Orthopedic Surgery/Sports Medicine Center, Southwest Hospital, Army Medical University, Chongqing, 400038, China
| | - Jing-Tong Lv
- Department of Orthopedic Surgery/Sports Medicine Center, Southwest Hospital, Army Medical University, Chongqing, 400038, China
| | - Xia Kang
- Department of Orthopedic Surgery/Sports Medicine Center, Southwest Hospital, Army Medical University, Chongqing, 400038, China
- Department of Pathophysiology, College of High Altitude Military Medicine, Army Medical University, Chongqing, 400038, China
- Department of Biochemistry and Molecular Biology, Army Medical University, Chongqing, 400038, China
| | - You-Xing Shi
- Department of Orthopedic Surgery/Sports Medicine Center, Southwest Hospital, Army Medical University, Chongqing, 400038, China
| | - Ellen Kim
- Department of Orthopedic Surgery, NYU Grossman School of Medicine, New York, NY, 10003, USA
| | - Yin-Hua Qin
- Department of Anatomy, Engineering Research Center for Organ Intelligent Biological Manufacturing of Chongqing, Key Lab for Biomechanics and Tissue Engineering of Chongqing, Army Medical University, Chongqing, 400038, China
| | - Aubryanna Hettinghouse
- Department of Orthopedic Surgery, NYU Grossman School of Medicine, New York, NY, 10003, USA
| | - Kai-di Wang
- Department of Orthopedic Surgery, NYU Grossman School of Medicine, New York, NY, 10003, USA
- Department of Medical Experimental Center, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, 266000, China
| | - Xiang-Li Zhao
- Department of Orthopedic Surgery, NYU Grossman School of Medicine, New York, NY, 10003, USA
- Department of Orthopedics and Rehabilitations, Yale University School of Medicine, New Haven, CT, 06519, USA
| | - Ming-Yu Yang
- Department of Orthopedic Surgery/Sports Medicine Center, Southwest Hospital, Army Medical University, Chongqing, 400038, China
| | - Yu-Zhen Tang
- Department of Orthopedic Surgery/Sports Medicine Center, Southwest Hospital, Army Medical University, Chongqing, 400038, China
| | - Hai-Long Piao
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
| | - Lin Guo
- Department of Orthopedic Surgery/Sports Medicine Center, Southwest Hospital, Army Medical University, Chongqing, 400038, China.
| | - Chuan-Ju Liu
- Department of Orthopedic Surgery, NYU Grossman School of Medicine, New York, NY, 10003, USA.
- Department of Orthopedics and Rehabilitations, Yale University School of Medicine, New Haven, CT, 06519, USA.
| | - Hong-Ming Miao
- Department of Pathophysiology, College of High Altitude Military Medicine, Army Medical University, Chongqing, 400038, China.
- Jinfeng Laboratory, Chongqing, 401329, China.
| | - Kang-Lai Tang
- Department of Orthopedic Surgery/Sports Medicine Center, Southwest Hospital, Army Medical University, Chongqing, 400038, China.
| |
Collapse
|
2
|
Cohen BE. The Role of the Swollen State in Cell Proliferation. J Membr Biol 2024:10.1007/s00232-024-00328-x. [PMID: 39482485 DOI: 10.1007/s00232-024-00328-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 10/17/2024] [Indexed: 11/03/2024]
Abstract
Cell swelling is known to be involved in various stages of the growth of plant cells and microorganisms but in mammalian cells how crucial a swollen state is for determining the fate of the cellular proliferation remains unclear. Recent evidence has increased our understanding of how the loss of the cell surface interactions with the extracellular matrix at early mitosis decreases the membrane tension triggering curvature changes in the plasma membrane and the activation of the sodium/hydrogen (Na +/H +) exchanger (NHE1) that drives osmotic swelling. Such a swollen state is temporary, but it is critical to alter essential membrane biophysical parameters that are required to activate Ca2 + channels and modulate the opening of K + channels involved in setting the membrane potential. A decreased membrane potential across the mitotic cell membrane enhances the clustering of Ras proteins involved in the Ca2 + and cytoskeleton-driven events that lead to cell rounding. Changes in the external mechanical and osmotic forces also have an impact on the lipid composition of the plasma membrane during mitosis.
Collapse
|
3
|
Kisor KP, Ruiz DG, Jacobson MP, Barber DL. A role for pH dynamics regulating transcription factor DNA binding selectivity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.21.595212. [PMID: 38826444 PMCID: PMC11142074 DOI: 10.1101/2024.05.21.595212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
Intracellular pH (pHi) dynamics regulates diverse cell processes such as proliferation, dysplasia, and differentiation, often mediated by the protonation state of a functionally critical histidine residue in endogenous pH sensing proteins. How pHi dynamics can directly regulate gene expression and whether transcription factors can function as pH sensors has received limited attention. We tested the prediction that transcription factors with a histidine in their DNA binding domain (DBD) that forms hydrogen bonds with nucleotides can have pH-regulated activity, which is relevant to more than 85 transcription factors in distinct families, including FOX, KLF, SOX and MITF/Myc. Focusing on FOX family transcription factors, we used unbiased SELEX-seq to identify pH-dependent DNA binding motif preferences, then confirm pH-regulated binding affinities for FOXC2, FOXM1, and FOXN1 to a canonical FkhP DNA motif that are 2.5 to 7.5 greater at pH 7.0 compared with pH 7.5. For FOXC2, we also find greater activity for an FkhP motif at lower pHi in cells and that pH-regulated binding and activity are dependent on a conserved histidine (His122) in the DBD. RNA-seq with FOXC2 also reveals pH-dependent differences in enriched promoter motifs. Our findings identify pH-regulated transcription factor-DNA binding selectivity with relevance to how pHi dynamics can regulate gene expression for myriad cell behaviours.
Collapse
|
4
|
Axelsen TV, Olesen C, Khan D, Mohammadi A, Bouzinova EV, Nielsen CJF, Mele M, Hauerslev KR, Pedersen HL, Balling E, Vahl P, Tramm T, Christiansen PM, Boedtkjer E. Antibodies toward Na +,HCO 3--cotransporter NBCn1/SLC4A7 block net acid extrusion and cause pH-dependent growth inhibition and apoptosis in breast cancer. Br J Cancer 2024; 130:1206-1220. [PMID: 38310186 PMCID: PMC10991555 DOI: 10.1038/s41416-024-02591-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 01/09/2024] [Accepted: 01/17/2024] [Indexed: 02/05/2024] Open
Abstract
BACKGROUND Na+,HCO3--cotransporter NBCn1/Slc4a7 accelerates murine breast carcinogenesis. Lack of specific pharmacological tools previously restricted therapeutic targeting of NBCn1 and identification of NBCn1-dependent functions in human breast cancer. METHODS We develop extracellularly-targeted anti-NBCn1 antibodies, screen for functional activity on cells, and evaluate (a) mechanisms of intracellular pH regulation in human primary breast carcinomas, (b) proliferation, cell death, and tumor growth consequences of NBCn1 in triple-negative breast cancer, and (c) association of NBCn1-mediated Na+,HCO3--cotransport with human breast cancer metastasis. RESULTS We identify high-affinity (KD ≈ 0.14 nM) anti-NBCn1 antibodies that block human NBCn1-mediated Na+,HCO3--cotransport in cells, without cross-reactivity towards human NBCe1 or murine NBCn1. These anti-NBCn1 antibodies abolish Na+,HCO3--cotransport activity in freshly isolated primary organoids from human breast carcinomas and lower net acid extrusion effectively in primary breast cancer tissue from patients with macrometastases in axillary lymph nodes. Inhibitory anti-NBCn1 antibodies decelerate tumor growth in vivo by ~50% in a patient-derived xenograft model of triple-negative breast cancer and pH-dependently reduce colony formation, cause G2/M-phase cell cycle accumulation, and increase apoptosis of metastatic triple-negative breast cancer cells in vitro. CONCLUSIONS Inhibitory anti-NBCn1 antibodies block net acid extrusion in human breast cancer tissue, particularly from patients with disseminated disease, and pH-dependently limit triple-negative breast cancer growth.
Collapse
Affiliation(s)
- Trine V Axelsen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Claus Olesen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Danish Khan
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Ali Mohammadi
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | | | | | - Marco Mele
- Department of Surgery, Randers Regional Hospital, Randers, Denmark
| | - Katrine R Hauerslev
- Department of Plastic and Breast Surgery, Aarhus University Hospital, Aarhus, Denmark
| | - Helene L Pedersen
- Department of Pathology, Randers Regional Hospital, Randers, Denmark
| | - Eva Balling
- Department of Surgery, Randers Regional Hospital, Randers, Denmark
| | - Pernille Vahl
- Department of Pathology, Aarhus University Hospital, Aarhus, Denmark
| | - Trine Tramm
- Department of Pathology, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Peer M Christiansen
- Department of Surgery, Randers Regional Hospital, Randers, Denmark
- Department of Plastic and Breast Surgery, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Ebbe Boedtkjer
- Department of Biomedicine, Aarhus University, Aarhus, Denmark.
| |
Collapse
|
5
|
Pedersen SHF. Acid-base transporters in the context of tumor heterogeneity. Pflugers Arch 2024; 476:689-701. [PMID: 38332178 DOI: 10.1007/s00424-024-02918-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 01/20/2024] [Accepted: 01/29/2024] [Indexed: 02/10/2024]
Abstract
The copious metabolic acid production and -extrusion by cancer cells render poorly vascularized regions of solid tumors highly acidic. A growing list of proton - and bicarbonate transporters has been suggested to contribute to net acid extrusion from cancer cells, and/or been shown to be dysregulated and favor malignant development in various cancers. The great majority of these roles have been studied at the level of the cancer cells. However, recent advances in understanding of the cellular and physicochemical heterogeneity of solid tumors both enable and necessitate a reexamination of the regulation and roles of acid-base transporters in such malignancies. This review will briefly summarize the state-of-the-art, with a focus on the SLC9A and SLC4A families, for which most evidence is available. This is followed by a discussion of key concepts and open questions arising from recent insights and of the challenges that need to be tackled to address them. Finally, opportunities and challenges in therapeutic targeting of the acid-base transportome in cancers will be addressed.
Collapse
Affiliation(s)
- Stine Helene Falsig Pedersen
- Section for Cell Biology and Physiology, Department of Biology, Faculty of Science, University of Copenhagen, Universitetsparken 13, 2100, Copenhagen, Denmark.
| |
Collapse
|
6
|
Arnhold J. Inflammation-Associated Cytotoxic Agents in Tumorigenesis. Cancers (Basel) 2023; 16:81. [PMID: 38201509 PMCID: PMC10778456 DOI: 10.3390/cancers16010081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 12/16/2023] [Accepted: 12/20/2023] [Indexed: 01/12/2024] Open
Abstract
Chronic inflammatory processes are related to all stages of tumorigenesis. As inflammation is closely associated with the activation and release of different cytotoxic agents, the interplay between cytotoxic agents and antagonizing principles is highlighted in this review to address the question of how tumor cells overcome the enhanced values of cytotoxic agents in tumors. In tumor cells, the enhanced formation of mitochondrial-derived reactive species and elevated values of iron ions and free heme are antagonized by an overexpression of enzymes and proteins, contributing to the antioxidative defense and maintenance of redox homeostasis. Through these mechanisms, tumor cells can even survive additional stress caused by radio- and chemotherapy. Through the secretion of active agents from tumor cells, immune cells are suppressed in the tumor microenvironment and an enhanced formation of extracellular matrix components is induced. Different oxidant- and protease-based cytotoxic agents are involved in tumor-mediated immunosuppression, tumor growth, tumor cell invasion, and metastasis. Considering the special metabolic conditions in tumors, the main focus here was directed on the disturbed balance between the cytotoxic agents and protective mechanisms in late-stage tumors. This knowledge is mandatory for the implementation of novel anti-cancerous therapeutic approaches.
Collapse
Affiliation(s)
- Jürgen Arnhold
- Institute of Medical Physics and Biophysics, Medical Faculty, Leipzig University, Härtelstr. 16-18, 04107 Leipzig, Germany
| |
Collapse
|
7
|
Dhakan C, Anemone A, Ventura V, Carella A, Corrado A, Pirotta E, Villano D, Romdhane F, Gammaraccio F, Aime S, Longo DL. Assessing the Therapeutic Efficacy of Proton Transport Inhibitors in a Triple-Negative Breast Cancer Murine Model with Magnetic Resonance Imaging-Chemical Exchange Saturation Transfer Tumor pH Imaging. Metabolites 2023; 13:1161. [PMID: 37999256 PMCID: PMC10673543 DOI: 10.3390/metabo13111161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 11/06/2023] [Accepted: 11/14/2023] [Indexed: 11/25/2023] Open
Abstract
Proton transporters play a key role in maintaining the acidic tumor microenvironment; hence, their inhibition has been proposed as a new therapeutic treatment, although few methods can accurately assess their effect in vivo. In this study, we investigated whether MRI-CEST (Magnetic Resonance Imaging-Chemical Exchange Saturation Transfer) tumor pH imaging can be a useful tool to evaluate in vivo the therapeutic efficacy of several Proton Pump Inhibitors (PPIs) in breast cancer. Cell viability and extracellular pH assays were carried out in breast cancer cells cultured at physiological pH (7.4) or acid-adapted (pH of 6.5 and 6.8) following the exposure to inhibitors of V-ATPase (Lansoprazole, Esomeprazole) or NHE1 (Amiloride, Cariporide) at several concentrations. Next, triple-negative breast cancer 4T1 tumor-bearing mice were treated with Lansoprazole or Amiloride and MRI-CEST tumor pH imaging was utilized to assess the in vivo efficacy. Only Lansoprazole induced, in addition to breast cancer cell toxicity, a significant inhibition of proton extrusion. A significant reduction in tumor volume, prolonged survival, and increase in extracellular tumor pH after 1 and 2 weeks were observed after Lansoprazole treatment, whereas no significant changes were detected upon Amiloride treatment. Our results suggested that MRI-CEST tumor pH imaging can monitor the therapeutic efficacy of PPIs in breast cancer murine models.
Collapse
Affiliation(s)
- Chetan Dhakan
- Institute of Biostructures and Bioimaging (IBB), National Research Council of Italy (CNR), Via Nizza 52, 10126 Turin, Italy
| | - Annasofia Anemone
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Via Nizza 52, 10126 Turin, Italy
| | - Vittoria Ventura
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Via Nizza 52, 10126 Turin, Italy
| | - Antonella Carella
- Institute of Biostructures and Bioimaging (IBB), National Research Council of Italy (CNR), Via Nizza 52, 10126 Turin, Italy
| | - Alessia Corrado
- Institute of Biostructures and Bioimaging (IBB), National Research Council of Italy (CNR), Via Nizza 52, 10126 Turin, Italy
| | - Elisa Pirotta
- Institute of Biostructures and Bioimaging (IBB), National Research Council of Italy (CNR), Via Nizza 52, 10126 Turin, Italy
| | - Daisy Villano
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Via Nizza 52, 10126 Turin, Italy
| | - Feriel Romdhane
- Institute of Biostructures and Bioimaging (IBB), National Research Council of Italy (CNR), Via Nizza 52, 10126 Turin, Italy
| | - Francesco Gammaraccio
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Via Nizza 52, 10126 Turin, Italy
| | - Silvio Aime
- IRCCS SynLAB SDN, Via Gianturco 113, 80143 Naples, Italy
| | - Dario Livio Longo
- Institute of Biostructures and Bioimaging (IBB), National Research Council of Italy (CNR), Via Nizza 52, 10126 Turin, Italy
| |
Collapse
|
8
|
Wang BY, Shen HT, Lee YL, Chien PJ, Chang WW. Inhibition of Na+/H+ exchanger (NHE) 7 by 5-(N-ethyl-N-isopropyl)-Amiloride displays anti-cancer activity in non-small cell lung cancer by disrupting cancer stem cell activity and downregulating PD-L1 expression. Am J Cancer Res 2023; 13:4721-4733. [PMID: 37970357 PMCID: PMC10636675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 08/06/2023] [Indexed: 11/17/2023] Open
Abstract
The alkaline intracellular environment of cancer cells is critical for cell proliferation and controlled by various plasma membrane transporters including Na+/H+ exchangers (NHEs). NHEs can also mediate cell behavior by regulating signaling transduction. In this study, we investigated the role of NHE7 in cancer stem cell (CSC) activity in non-small cell lung cancer (NSCLC) cells and the potential therapeutic implications of targeting NHE7 and the associated immune checkpoint molecule PD-L1. By analyzing the database from The Cancer Genome Atlas, we found a positive correlation between SLC9A7 mRNA levels (the gene encoding NHE7) and poor overall survival in lung adenocarcinoma patients. Using 5-(N-ethyl-N-isopropyl)-Amiloride (EIPA) to inhibit NHE7 activity, we observed disrupted cell cycle progression and suppressed NSCLC cell proliferation without inducing apoptosis. Furthermore, EIPA demonstrated a suppressive effect on CSC activity, evidenced by decreased tumorsphere numbers and inhibition of CSC markers such as ALDH1A2, ABCG2, CD44, and CD133. Flow cytometric analysis revealed that EIPA treatment or NHE7 knockdown in NSCLC cells led to downregulated PD-L1 expression, associated with inhibited STAT3 activity. Interestingly, EIPA's CSC-targeting activity was preferentially observed in NSCLC cells overexpressing BMI1, while increased PD-L1 expression was detected in BMI1-overexpressing NSCLC cells. Our findings suggest that targeting NHE7 with inhibitors like EIPA may have therapeutic potential in NSCLC treatment by disrupting cell cycle progression and suppressing CSC activity. The observed increase in PD-L1 expression in BMI1-overexpressing NSCLC cells upon EIPA treatment highlights the potential benefit of combining NHE7 inhibitors with anti-PD-L1 agents as a promising new therapeutic strategy for NSCLC.
Collapse
Affiliation(s)
- Bing-Yen Wang
- Division of Thoracic Surgery, Department of Surgery, Changhua Christian HospitalNo. 135, Nanhsiao Street, Changhua 500209, Taiwan
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing UniversityNo. 145, Xingda Rd., South Dist., Taichung 402202, Taiwan
| | - Huan-Ting Shen
- Department of Pulmonary Medicine, Taichung Tzu Chi Hospital, Buddhist Tzu Chi Medical FoundationNo. 88, Sec. 1, Fengxing Rd., Tanzi Dist., Taichung 427, Taiwan
| | - Yu-Ling Lee
- Department of Biomedical Sciences, Chung Shan Medical UniversityNo. 110, Sec. 1, Jianguo N. Rd., Taichung 402306, Taiwan
| | - Peng-Ju Chien
- Department of Biomedical Sciences, Chung Shan Medical UniversityNo. 110, Sec. 1, Jianguo N. Rd., Taichung 402306, Taiwan
| | - Wen-Wei Chang
- Department of Biomedical Sciences, Chung Shan Medical UniversityNo. 110, Sec. 1, Jianguo N. Rd., Taichung 402306, Taiwan
- Department of Medical Research, Chung Shan Medical University HospitalNo. 110, Sec. 1, Jianguo N. Rd., Taichung 402306, Taiwan
| |
Collapse
|
9
|
Michl J, Monterisi S, White B, Blaszczak W, Hulikova A, Abdullayeva G, Bridges E, Yin Z, Bodmer WF, Swietach P. Acid-adapted cancer cells alkalinize their cytoplasm by degrading the acid-loading membrane transporter anion exchanger 2, SLC4A2. Cell Rep 2023; 42:112601. [PMID: 37270778 DOI: 10.1016/j.celrep.2023.112601] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 03/16/2023] [Accepted: 05/19/2023] [Indexed: 06/06/2023] Open
Abstract
Acidic environments reduce the intracellular pH (pHi) of most cells to levels that are sub-optimal for growth and cellular functions. Yet, cancers maintain an alkaline cytoplasm despite low extracellular pH (pHe). Raised pHi is thought to be beneficial for tumor progression and invasiveness. However, the transport mechanisms underpinning this adaptation have not been studied systematically. Here, we characterize the pHe-pHi relationship in 66 colorectal cancer cell lines and identify the acid-loading anion exchanger 2 (AE2, SLC4A2) as a regulator of resting pHi. Cells adapt to chronic extracellular acidosis by degrading AE2 protein, which raises pHi and reduces acid sensitivity of growth. Acidity inhibits mTOR signaling, which stimulates lysosomal function and AE2 degradation, a process reversed by bafilomycin A1. We identify AE2 degradation as a mechanism for maintaining a conducive pHi in tumors. As an adaptive mechanism, inhibiting lysosomal degradation of AE2 is a potential therapeutic target.
Collapse
Affiliation(s)
- Johanna Michl
- Department of Physiology, Anatomy and Genetics, Parks Road, Oxford OX1 3PT, UK
| | - Stefania Monterisi
- Department of Physiology, Anatomy and Genetics, Parks Road, Oxford OX1 3PT, UK
| | - Bobby White
- Department of Physiology, Anatomy and Genetics, Parks Road, Oxford OX1 3PT, UK
| | - Wiktoria Blaszczak
- Department of Physiology, Anatomy and Genetics, Parks Road, Oxford OX1 3PT, UK
| | - Alzbeta Hulikova
- Department of Physiology, Anatomy and Genetics, Parks Road, Oxford OX1 3PT, UK
| | - Gulnar Abdullayeva
- MRC Weatherall Institute for Molecular Medicine, John Radcliffe Hospital, Headington, Oxford OX3 9DS, UK
| | - Esther Bridges
- Department of NDM Experimental Medicine, MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, JR Hospital, Headington, Oxford OX3 9DS, UK
| | - Zinan Yin
- Department of NDM Experimental Medicine, MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, JR Hospital, Headington, Oxford OX3 9DS, UK
| | - Walter F Bodmer
- MRC Weatherall Institute for Molecular Medicine, John Radcliffe Hospital, Headington, Oxford OX3 9DS, UK
| | - Pawel Swietach
- Department of Physiology, Anatomy and Genetics, Parks Road, Oxford OX1 3PT, UK.
| |
Collapse
|
10
|
Liu S, Liu Z, Shang A, Xun J, Lv Z, Zhou S, Liu C, Zhang Q, Yang Y. CD44 is a potential immunotherapeutic target and affects macrophage infiltration leading to poor prognosis. Sci Rep 2023; 13:9657. [PMID: 37316699 DOI: 10.1038/s41598-023-33915-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 04/20/2023] [Indexed: 06/16/2023] Open
Abstract
CD44 plays a key role in the communication of CSCs with the microenvironment and the regulation of stem cell properties. UALCAN was used to analyze the expression of CD44 in bladder cancer (BLCA) and normal tissue. The UALCAN was utilized to analyze the prognostic value of CD44 in BLCA. The TIMER database was used to explore the relationship between CD44 and PD-L1; CD44 and tumor-infiltrating immune cells. The regulatory effect of CD44 on PD-L1 was verified by cell experiments in vitro. IHC confirmed the results of the bioinformatics analysis. GeneMania and Metascape were used to analyze protein-protein interaction (PPI) investigations and functional enrichment analysis. We found that BLCA patients with high CD44 expression had worse survival than those with low CD44 expression (P < 0.05). IHC and the TIMER database results showed that CD44 expression was positively correlated with PD-L1 expression (P < 0.05). At the cellular level, the expression of PD-L1 was significantly inhibited after CD44 expression was inhibited by siRNA. Immune infiltration analysis showed that CD44 expression levels in BLCA were significantly correlated with immune infiltration levels of different immune cells. IHC staining results further confirmed that the expression of CD44 in tumor cells was positively associated with the number of CD68+ macrophages and CD163+ macrophages (P < 0.05). Our results suggest that CD44 is a positive regulator of PD-L1 in BLCA and may be a key regulator of tumor macrophages infiltration and may be involved in M2 macrophage polarization. Our study provided new insights into the prognosis and immunotherapy of BLCA patients through macrophage infiltration and immune checkpoints.
Collapse
Affiliation(s)
- Shuangqing Liu
- Tianjin Medical University Nankai Hospital, Tianjin, 300070, China
| | - Zehan Liu
- Tianjin Medical University Nankai Hospital, Tianjin, 300070, China
- Section for HepatoPancreatoBiliary Surgery, Department of General Surgery, The Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University and The Second Affiliated Hospital of Chengdu, Chongqing Medical University, Chengdu, 610031, China
| | - Aichen Shang
- Tianjin Medical University Nankai Hospital, Tianjin, 300070, China
- Department of Pathology, Sino-Singapore Eco-City Hospital of Tianjin Medical University, Tianjin, 300456, China
| | - Jing Xun
- Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Institute of Acute Abdominal Diseases, Tianjin Nankai Hospital, Tianjin, China
| | - Zongjing Lv
- Tianjin Medical University Nankai Hospital, Tianjin, 300070, China
| | - Siying Zhou
- Tianjin Medical University Nankai Hospital, Tianjin, 300070, China
| | - Cui Liu
- Tianjin Medical University Nankai Hospital, Tianjin, 300070, China
| | - Qi Zhang
- Tianjin Medical University Nankai Hospital, Tianjin, 300070, China.
- Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Institute of Acute Abdominal Diseases, Tianjin Nankai Hospital, Tianjin, China.
| | - Yuming Yang
- Tianjin Medical University Nankai Hospital, Tianjin, 300070, China.
- Department of Pathology, Tianjin Nankai Hospital, Tianjin, China.
| |
Collapse
|
11
|
Rolver MG, Holland LKK, Ponniah M, Prasad NS, Yao J, Schnipper J, Kramer S, Elingaard‐Larsen L, Pedraz‐Cuesta E, Liu B, Pardo LA, Maeda K, Sandelin A, Pedersen SF. Chronic acidosis rewires cancer cell metabolism through PPARα signaling. Int J Cancer 2023; 152:1668-1684. [PMID: 36533672 PMCID: PMC10108231 DOI: 10.1002/ijc.34404] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 11/25/2022] [Accepted: 12/06/2022] [Indexed: 12/23/2022]
Abstract
The mechanisms linking tumor microenvironment acidosis to disease progression are not understood. Here, we used mammary, pancreatic, and colon cancer cells to show that adaptation to growth at an extracellular pH (pHe ) mimicking acidic tumor niches is associated with upregulated net acid extrusion capacity and elevated intracellular pH at physiological pHe , but not at acidic pHe . Using metabolic profiling, shotgun lipidomics, imaging and biochemical analyses, we show that the acid adaptation-induced phenotype is characterized by a shift toward oxidative metabolism, increased lipid droplet-, triacylglycerol-, peroxisome content and mitochondrial hyperfusion. Peroxisome proliferator-activated receptor-α (PPARA, PPARα) expression and activity are upregulated, at least in part by increased fatty acid uptake. PPARα upregulates genes driving increased mitochondrial and peroxisomal mass and β-oxidation capacity, including mitochondrial lipid import proteins CPT1A, CPT2 and SLC25A20, electron transport chain components, peroxisomal proteins PEX11A and ACOX1, and thioredoxin-interacting protein (TXNIP), a negative regulator of glycolysis. This endows acid-adapted cancer cells with increased capacity for utilizing fatty acids for metabolic needs, while limiting glycolysis. As a consequence, the acid-adapted cells exhibit increased sensitivity to PPARα inhibition. We conclude that PPARα is a key upstream regulator of metabolic changes favoring cancer cell survival in acidic tumor niches.
Collapse
Affiliation(s)
- Michala G. Rolver
- Section for Cell Biology and Physiology, Department of BiologyUniversity of CopenhagenCopenhagenDenmark
| | - Lya K. K. Holland
- Cell Death and Metabolism, Center for Autophagy, Recycling and Disease, Danish Cancer Society Research CenterCopenhagenDenmark
| | - Muthulakshmi Ponniah
- Section for Cell Biology and Physiology, Department of BiologyUniversity of CopenhagenCopenhagenDenmark
| | - Nanditha S. Prasad
- Section for Cell Biology and Physiology, Department of BiologyUniversity of CopenhagenCopenhagenDenmark
| | - Jiayi Yao
- The Bioinformatics Center, Department of BiologyUniversity of CopenhagenCopenhagenDenmark
- Biotech Research and Innovation Center, University of CopenhagenCopenhagenDenmark
| | - Julie Schnipper
- Laboratory of Cellular and Molecular Physiology, University of Picardie Jules VerneAmiensFrance
| | - Signe Kramer
- Section for Cell Biology and Physiology, Department of BiologyUniversity of CopenhagenCopenhagenDenmark
| | | | - Elena Pedraz‐Cuesta
- Section for Cell Biology and Physiology, Department of BiologyUniversity of CopenhagenCopenhagenDenmark
| | - Bin Liu
- Cell Death and Metabolism, Center for Autophagy, Recycling and Disease, Danish Cancer Society Research CenterCopenhagenDenmark
| | - Luis A. Pardo
- Oncophysiology Group, Max‐Planck‐Institute for Multidisciplinary SciencesGöttingenGermany
| | - Kenji Maeda
- Cell Death and Metabolism, Center for Autophagy, Recycling and Disease, Danish Cancer Society Research CenterCopenhagenDenmark
| | - Albin Sandelin
- The Bioinformatics Center, Department of BiologyUniversity of CopenhagenCopenhagenDenmark
- Biotech Research and Innovation Center, University of CopenhagenCopenhagenDenmark
| | - Stine Falsig Pedersen
- Section for Cell Biology and Physiology, Department of BiologyUniversity of CopenhagenCopenhagenDenmark
| |
Collapse
|
12
|
Rajan S, Terman JR, Reisler E. MICAL-mediated oxidation of actin and its effects on cytoskeletal and cellular dynamics. Front Cell Dev Biol 2023; 11:1124202. [PMID: 36875759 PMCID: PMC9982024 DOI: 10.3389/fcell.2023.1124202] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 02/02/2023] [Indexed: 02/19/2023] Open
Abstract
Actin and its dynamic structural remodelings are involved in multiple cellular functions, including maintaining cell shape and integrity, cytokinesis, motility, navigation, and muscle contraction. Many actin-binding proteins regulate the cytoskeleton to facilitate these functions. Recently, actin's post-translational modifications (PTMs) and their importance to actin functions have gained increasing recognition. The MICAL family of proteins has emerged as important actin regulatory oxidation-reduction (Redox) enzymes, influencing actin's properties both in vitro and in vivo. MICALs specifically bind to actin filaments and selectively oxidize actin's methionine residues 44 and 47, which perturbs filaments' structure and leads to their disassembly. This review provides an overview of the MICALs and the impact of MICAL-mediated oxidation on actin's properties, including its assembly and disassembly, effects on other actin-binding proteins, and on cells and tissue systems.
Collapse
Affiliation(s)
- Sudeepa Rajan
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, CA, United States
| | - Jonathan R. Terman
- Departments of Neuroscience and Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Emil Reisler
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, CA, United States
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
13
|
Hu J, Li G, Liu Z, Ma H, Yuan W, Lu Z, Zhang D, Ling H, Zhang F, Liu Y, Liu C, Qiu Y. Bicarbonate transporter SLC4A7 promotes EMT and metastasis of HNSCC by activating the PI3K/AKT/mTOR signaling pathway. Mol Carcinog 2023; 62:628-640. [PMID: 36727616 DOI: 10.1002/mc.23511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 10/30/2022] [Accepted: 10/31/2022] [Indexed: 02/03/2023]
Abstract
Head and neck squamous cell carcinoma (HNSCC) is the sixth most common cancer worldwide. Currently, therapeutic modalities such as surgery, chemotherapy, radiotherapy, and immunotherapy are being used to treat HNSCC. However, the treatment outcomes of most patients are dismal because they are already in middle or advanced stage by the time of diagnosis and poorly responsive to treatments. It is therefore of great interest to clarify mechanisms that contribute to the metastasis of cells to identify possible targets for therapy. In this study, we identified the Na+ -coupled bicarbonate transporter, SLC4A7, play essential roles in the metastasis of HNSCC. Our results showed that the relative expression of SLC4A7 messenger RNA was highly expressed in HNSCCs samples from TCGA, and compared with precancerous cells of human oral mucosa (DOK), SLC4A7 was highly expressed in HNSCC cell lines. In vitro and in vivo experiments showed that dysregulation of SLC4A7 had minor influence on the proliferation of HNSCC but impacted HNSCC's migration and invasion. Meanwhile, SLC4A7 could promote epithelial-mesenchymal transition (EMT) in HNSCC. RNA-seq, KEGG pathway enrichment analysis and Western blot further revealed that downregulation of SLC4A7 in HNSCC cells inhibited the PI3K/AKT pathway. These findings were further validated via rescue experiments using a small molecule inhibitor of PI3K/mTOR (GDC-0980). Our findings suggest that SLC4A7 promotes EMT and metastasis of HNSCC through the PI3K/AKT/mTOR signaling pathway, which may be a valuable predictive biomarker and potential therapeutic target in HNSCC.
Collapse
Affiliation(s)
- Junli Hu
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, China.,Otolaryngology Major Disease Research Key Laboratory of Hunan Province, Changsha, China.,Clinical Research Center for Pharyngolaryngeal Diseases and Voice Disorders in Hunan Province, Changsha, China.,Department of Otolaryngology Head and Neck Surgery, Yantian District People's Hospital, Shenzhen, Guangdong, China
| | - Guo Li
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, China.,Otolaryngology Major Disease Research Key Laboratory of Hunan Province, Changsha, China.,Clinical Research Center for Pharyngolaryngeal Diseases and Voice Disorders in Hunan Province, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Zhifeng Liu
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, China.,Otolaryngology Major Disease Research Key Laboratory of Hunan Province, Changsha, China.,Clinical Research Center for Pharyngolaryngeal Diseases and Voice Disorders in Hunan Province, Changsha, China
| | - Huiling Ma
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, China.,Otolaryngology Major Disease Research Key Laboratory of Hunan Province, Changsha, China.,Clinical Research Center for Pharyngolaryngeal Diseases and Voice Disorders in Hunan Province, Changsha, China
| | - Wenhui Yuan
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, China.,Otolaryngology Major Disease Research Key Laboratory of Hunan Province, Changsha, China.,Clinical Research Center for Pharyngolaryngeal Diseases and Voice Disorders in Hunan Province, Changsha, China
| | - Zhaoyi Lu
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, China.,Otolaryngology Major Disease Research Key Laboratory of Hunan Province, Changsha, China.,Clinical Research Center for Pharyngolaryngeal Diseases and Voice Disorders in Hunan Province, Changsha, China
| | - Diekuo Zhang
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, China.,Otolaryngology Major Disease Research Key Laboratory of Hunan Province, Changsha, China.,Clinical Research Center for Pharyngolaryngeal Diseases and Voice Disorders in Hunan Province, Changsha, China
| | - Hang Ling
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, China.,Otolaryngology Major Disease Research Key Laboratory of Hunan Province, Changsha, China.,Clinical Research Center for Pharyngolaryngeal Diseases and Voice Disorders in Hunan Province, Changsha, China
| | - Fengyu Zhang
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, China.,Otolaryngology Major Disease Research Key Laboratory of Hunan Province, Changsha, China.,Clinical Research Center for Pharyngolaryngeal Diseases and Voice Disorders in Hunan Province, Changsha, China
| | - Yong Liu
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, China.,Otolaryngology Major Disease Research Key Laboratory of Hunan Province, Changsha, China.,Clinical Research Center for Pharyngolaryngeal Diseases and Voice Disorders in Hunan Province, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Chao Liu
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, China.,Otolaryngology Major Disease Research Key Laboratory of Hunan Province, Changsha, China.,Clinical Research Center for Pharyngolaryngeal Diseases and Voice Disorders in Hunan Province, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Yuanzheng Qiu
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, China.,Otolaryngology Major Disease Research Key Laboratory of Hunan Province, Changsha, China.,Clinical Research Center for Pharyngolaryngeal Diseases and Voice Disorders in Hunan Province, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
14
|
Onufriev AV. Biologically relevant small variations of intra-cellular pH can have significant effect on stability of protein-DNA complexes, including the nucleosome. Front Mol Biosci 2023; 10:1067787. [PMID: 37143824 PMCID: PMC10151541 DOI: 10.3389/fmolb.2023.1067787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 01/16/2023] [Indexed: 05/06/2023] Open
Abstract
Stability of a protein-ligand complex may be sensitive to pH of its environment. Here we explore, computationally, stability of a set of protein-nucleic acid complexes using fundamental thermodynamic linkage relationship. The nucleosome, as well as an essentially random selection of 20 protein complexes with DNA or RNA, are included in the analysis. An increase in intra-cellular/intra-nuclear pH destabilizes most complexes, including the nucleosome. We propose to quantify the effect by ΔΔG0.3-the change in the binding free energy due to pH increase of 0.3 units, corresponding to doubling of the H + activity; variations of pH of this amplitude can occur in living cells, including in the course of the cell cycle, and in cancer cells relative to normal ones. We suggest, based on relevant experimental findings, a threshold of biological significance of 1 2 k B T ( ∼ 0.3 k c a l / m o l ) for changes of stability of chromatin-related protein-DNA complexes: a change in the binding affinity above the threshold may have biological consequences. We find that for 70% of the examined complexes, Δ Δ G 0.3 > 1 2 k B T (for 10%, ΔΔG0.3 is between 3 and 4 k B T). Thus, small but relevant variations of intra-nuclear pH of 0.3 may have biological consequences for many protein-nucleic acid complexes. The binding affinity between the histone octamer and its DNA, which directly affects the DNA accessibility in the nucleosome, is predicted to be highly sensitive to intra-nuclear pH. A variation of 0.3 units results in ΔΔG0.3 ∼ 10k B T ( ∼ 6 k c a l / m o l ) ; for spontaneous unwrapping of 20 bp long entry/exit fragments of the nucleosomal DNA, ΔΔG0.3 = 2.2k B T; partial disassembly of the nucleosome into the tetrasome is characterized by ΔΔG0.3 = 5.2k B T. The predicted pH -induced modulations of the nucleosome stability are significant enough to suggest that they may have consequences relevant to the biological function of the nucleosome. Accessibility of the nucleosomal DNA is predicted to positively correlate with pH variations during the cell cycle; an increase in intra-cellular pH seen in cancer cells is predicted to lead to a more accessible nucleosomal DNA; a drop in pH associated with apoptosis is predicted to make nucleosomal DNA less accessible. We speculate that processes that depend on accessibility to the DNA in the nucleosomes, such as transcription or DNA replication, might become upregulated due to relatively small, but nevertheless realistic increases of intra-nuclear pH.
Collapse
Affiliation(s)
- Alexey V. Onufriev
- Department of Physics, Virginia Tech, Blacksburg, Blacksburg, VA, United States
- Department of Computer Science, Virginia Tech, Blacksburg, Blacksburg, VA, United States
- Center from Soft Matter and Biological Physics, Virginia Tech, Blacksburg, VA, United States
- *Correspondence: Alexey V. Onufriev,
| |
Collapse
|
15
|
Acid Adaptation Promotes TRPC1 Plasma Membrane Localization Leading to Pancreatic Ductal Adenocarcinoma Cell Proliferation and Migration through Ca 2+ Entry and Interaction with PI3K/CaM. Cancers (Basel) 2022; 14:cancers14194946. [PMID: 36230869 PMCID: PMC9563726 DOI: 10.3390/cancers14194946] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 10/03/2022] [Accepted: 10/07/2022] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest cancers globally, with a 5-year overall survival of less than 10%. The development and progression of PDAC are linked to its fluctuating acidic tumor microenvironment. Ion channels act as important sensors of this acidic tumor microenvironment. They transduce extracellular signals and regulate signaling pathways involved in all hallmarks of cancer. In this study, we evaluated the interplay between a pH-sensitive ion channel, the calcium (Ca2+) channel transient receptor potential C1 (TRPC1), and three different stages of the tumor microenvironment, normal pH, acid adaptation, and acid recovery, and its impact on PDAC cell migration, proliferation, and cell cycle progression. In acid adaptation and recovery conditions, TRPC1 localizes to the plasma membrane, where it interacts with PI3K and calmodulin, and permits Ca2+ entry, which results in downstream signaling, leading to proliferation and migration. Thus, TRPC1 exerts a more aggressive role after adaptation to the acidic tumor microenvironment. Abstract Pancreatic ductal adenocarcinoma (PDAC) remains one of the most lethal malignancies, with a low overall survival rate of less than 10% and limited therapeutic options. Fluctuations in tumor microenvironment pH are a hallmark of PDAC development and progression. Many ion channels are bona fide cellular sensors of changes in pH. Yet, the interplay between the acidic tumor microenvironment and ion channel regulation in PDAC is poorly understood. In this study, we show that acid adaption increases PANC-1 cell migration but attenuates proliferation and spheroid growth, which are restored upon recovery. Moreover, acid adaptation and recovery conditions favor the plasma membrane localization of the pH-sensitive calcium (Ca2+) channel transient receptor potential C1 (TRPC1), TRPC1-mediated Ca2+ influx, channel interaction with the PI3K p85α subunit and calmodulin (CaM), and AKT and ERK1/2 activation. Knockdown (KD) of TRPC1 suppresses cell migration, proliferation, and spheroid growth, notably in acid-recovered cells. KD of TRPC1 causes the accumulation of cells in G0/G1 and G2/M phases, along with reduced expression of CDK6, −2, and −1, and cyclin A, and increased expression of p21CIP1. TRPC1 silencing decreases the basal Ca2+ influx in acid-adapted and -recovered cells, but not in normal pH conditions, and Ca2+ chelation reduces cell migration and proliferation solely in acid adaptation and recovery conditions. In conclusion, acid adaptation and recovery reinforce the involvement of TRPC1 in migration, proliferation, and cell cycle progression by permitting Ca2+ entry and forming a complex with the PI3K p85α subunit and CaM.
Collapse
|
16
|
Li X, Yang Y, Zhang B, Lin X, Fu X, An Y, Zou Y, Wang JX, Wang Z, Yu T. Lactate metabolism in human health and disease. Signal Transduct Target Ther 2022; 7:305. [PMID: 36050306 PMCID: PMC9434547 DOI: 10.1038/s41392-022-01151-3] [Citation(s) in RCA: 351] [Impact Index Per Article: 117.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Revised: 07/17/2022] [Accepted: 08/09/2022] [Indexed: 12/29/2022] Open
Abstract
The current understanding of lactate extends from its origins as a byproduct of glycolysis to its role in tumor metabolism, as identified by studies on the Warburg effect. The lactate shuttle hypothesis suggests that lactate plays an important role as a bridging signaling molecule that coordinates signaling among different cells, organs and tissues. Lactylation is a posttranslational modification initially reported by Professor Yingming Zhao’s research group in 2019. Subsequent studies confirmed that lactylation is a vital component of lactate function and is involved in tumor proliferation, neural excitation, inflammation and other biological processes. An indispensable substance for various physiological cellular functions, lactate plays a regulatory role in different aspects of energy metabolism and signal transduction. Therefore, a comprehensive review and summary of lactate is presented to clarify the role of lactate in disease and to provide a reference and direction for future research. This review offers a systematic overview of lactate homeostasis and its roles in physiological and pathological processes, as well as a comprehensive overview of the effects of lactylation in various diseases, particularly inflammation and cancer.
Collapse
Affiliation(s)
- Xiaolu Li
- Center for Regenerative Medicine, Institute for Translational Medicine, The Affiliated Hospital of Qingdao University; Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao, 266000, China
| | - Yanyan Yang
- Department of Immunology, School of Basic Medicine, Qingdao University, Qingdao, 266071, China
| | - Bei Zhang
- Department of Immunology, School of Basic Medicine, Qingdao University, Qingdao, 266071, China
| | - Xiaotong Lin
- Department of Respiratory Medicine, Qingdao Municipal Hospital, Qingdao, 266011, China
| | - Xiuxiu Fu
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao, 266000, China
| | - Yi An
- Department of Cardiology, The Affiliated Hospital of Qingdao University, No. 1677 Wutaishan Road, Qingdao, 266555, China
| | - Yulin Zou
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao, 266000, China
| | - Jian-Xun Wang
- Department of Immunology, School of Basic Medicine, Qingdao University, Qingdao, 266071, China
| | - Zhibin Wang
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao, 266000, China.
| | - Tao Yu
- Center for Regenerative Medicine, Institute for Translational Medicine, The Affiliated Hospital of Qingdao University; Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao, 266000, China.
| |
Collapse
|
17
|
Karmazyn M, Pierce GN, Fliegel L. The Remaining Conundrum of the Role of the Na +/H + Exchanger Isoform 1 (NHE1) in Cardiac Physiology and Pathology: Can It Be Rectified? Rev Cardiovasc Med 2022; 23:284. [PMID: 39076631 PMCID: PMC11266974 DOI: 10.31083/j.rcm2308284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 06/29/2022] [Accepted: 07/08/2022] [Indexed: 07/31/2024] Open
Abstract
The mammalian Na + /H + exchanger (NHE) is a family of ubiquitous membrane proteins present in humans. Isoform one (NHE1) is present on the plasma membrane and regulates intracellular pH by removal of one intracellular proton in exchange for one extracellular sodium thus functioning as an electroneutral process. Human NHE1 has a 500 amino acid membrane domain plus a C-terminal 315 amino acid, regulatory cytosolic tail. It is regulated through a cytosolic regulatory C-terminal tail which is subject to phosphorylation and is modulated by proteins and lipids. Substantial evidence has implicated NHE1 activity in both myocardial ischemia and reperfusion damage and myocardial remodeling resulting in heart failure. Experimental data show excellent cardioprotection with NHE1 inhibitors although results from clinical results have been mixed. In cardiac surgery patients receiving the NHE1 inhibitor cariporide, subgroups showed beneficial effects of treatment. However, in one trial this was associated with a significantly increased incidence of ischemic strokes. This likely reflected both inappropriate dosing regimens as well as overly high drug doses. We suggest that further progress towards NHE1 inhibition as a treatment for cardiovascular disease is warranted through the development of novel compounds to inhibit NHE1 that are structurally different than those previously used in compromised clinical trials. Some novel pyrazinoyl guanidine inhibitors of NHE1 are already in development and the recent elucidation of the three-dimensional structure of the NHE1 protein and identity of the inhibitor binding site may facilitate development. An alternative approach may also be to control the endogenous regulation of activity of NHE1, which is activated in disease.
Collapse
Affiliation(s)
- Morris Karmazyn
- Department of Physiology and Pharmacology, University of Western Ontario, London, ON N6A 5C1, Canada
| | - Grant N. Pierce
- Institute of Cardiovascular Sciences, Albrechtsen Research Centre, St. Boniface Hospital, and Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R2H 2A6, Canada
| | - Larry Fliegel
- Department of Biochemistry, University Alberta, Edmonton, AB T6G 2H7, Canada
| |
Collapse
|
18
|
Sloth RA, Axelsen TV, Espejo MS, Toft NJ, Voss NCS, Burton M, Thomassen M, Vahl P, Boedtkjer E. Loss of RPTPγ primes breast tissue for acid extrusion, promotes malignant transformation and results in early tumour recurrence and shortened survival. Br J Cancer 2022; 127:1226-1238. [PMID: 35821297 DOI: 10.1038/s41416-022-01911-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 06/22/2022] [Accepted: 06/29/2022] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND While cellular metabolism and acidic waste handling accelerate during breast carcinogenesis, temporal patterns of acid-base regulation and underlying molecular mechanisms responding to the tumour microenvironment remain unclear. METHODS We explore data from human cohorts and experimentally investigate transgenic mice to evaluate the putative extracellular HCO3--sensor Receptor Protein Tyrosine Phosphatase (RPTP)γ during breast carcinogenesis. RESULTS RPTPγ expression declines during human breast carcinogenesis and particularly in high-malignancy grade breast cancer. Low RPTPγ expression associates with poor prognosis in women with Luminal A or Basal-like breast cancer. RPTPγ knockout in mice favours premalignant changes in macroscopically normal breast tissue, accelerates primary breast cancer development, promotes malignant breast cancer histopathologies, and shortens recurrence-free survival. In RPTPγ knockout mice, expression of Na+,HCO3--cotransporter NBCn1-a breast cancer susceptibility protein-is upregulated in normal breast tissue but, contrary to wild-type mice, shows no further increase during breast carcinogenesis. Associated augmentation of Na+,HCO3--cotransport in normal breast tissue from RPTPγ knockout mice elevates steady-state intracellular pH, which has known pro-proliferative effects. CONCLUSIONS Loss of RPTPγ accelerates cellular net acid extrusion and elevates NBCn1 expression in breast tissue. As these effects precede neoplastic manifestations in histopathology, we propose that RPTPγ-dependent enhancement of Na+,HCO3--cotransport primes breast tissue for cancer development.
Collapse
Affiliation(s)
- Rasmus A Sloth
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Trine V Axelsen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | | | - Nicolai J Toft
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Ninna C S Voss
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Mark Burton
- Department of Clinical Genetics, University of Southern Denmark, Odense, Denmark.,Clinical Genome Center, University and Region of Southern Denmark, Odense, Denmark.,Department of Clinical Medicine, University of Southern Denmark, Odense, Denmark
| | - Mads Thomassen
- Department of Clinical Genetics, University of Southern Denmark, Odense, Denmark.,Clinical Genome Center, University and Region of Southern Denmark, Odense, Denmark
| | - Pernille Vahl
- Department of Pathology, Aarhus University Hospital, Aarhus, Denmark
| | - Ebbe Boedtkjer
- Department of Biomedicine, Aarhus University, Aarhus, Denmark.
| |
Collapse
|
19
|
Audero MM, Prevarskaya N, Fiorio Pla A. Ca 2+ Signalling and Hypoxia/Acidic Tumour Microenvironment Interplay in Tumour Progression. Int J Mol Sci 2022; 23:7377. [PMID: 35806388 PMCID: PMC9266881 DOI: 10.3390/ijms23137377] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 06/26/2022] [Accepted: 06/27/2022] [Indexed: 01/18/2023] Open
Abstract
Solid tumours are characterised by an altered microenvironment (TME) from the physicochemical point of view, displaying a highly hypoxic and acidic interstitial fluid. Hypoxia results from uncontrolled proliferation, aberrant vascularization and altered cancer cell metabolism. Tumour cellular apparatus adapts to hypoxia by altering its metabolism and behaviour, increasing its migratory and metastatic abilities by the acquisition of a mesenchymal phenotype and selection of aggressive tumour cell clones. Extracellular acidosis is considered a cancer hallmark, acting as a driver of cancer aggressiveness by promoting tumour metastasis and chemoresistance via the selection of more aggressive cell phenotypes, although the underlying mechanism is still not clear. In this context, Ca2+ channels represent good target candidates due to their ability to integrate signals from the TME. Ca2+ channels are pH and hypoxia sensors and alterations in Ca2+ homeostasis in cancer progression and vascularization have been extensively reported. In the present review, we present an up-to-date and critical view on Ca2+ permeable ion channels, with a major focus on TRPs, SOCs and PIEZO channels, which are modulated by tumour hypoxia and acidosis, as well as the consequent role of the altered Ca2+ signals on cancer progression hallmarks. We believe that a deeper comprehension of the Ca2+ signalling and acidic pH/hypoxia interplay will break new ground for the discovery of alternative and attractive therapeutic targets.
Collapse
Affiliation(s)
- Madelaine Magalì Audero
- U1003—PHYCEL—Laboratoire de Physiologie Cellulaire, Inserm, University of Lille, Villeneuve d’Ascq, 59000 Lille, France; (M.M.A.); (N.P.)
- Laboratory of Cellular and Molecular Angiogenesis, Department of Life Sciences and Systems Biology, University of Turin, 10123 Turin, Italy
| | - Natalia Prevarskaya
- U1003—PHYCEL—Laboratoire de Physiologie Cellulaire, Inserm, University of Lille, Villeneuve d’Ascq, 59000 Lille, France; (M.M.A.); (N.P.)
| | - Alessandra Fiorio Pla
- U1003—PHYCEL—Laboratoire de Physiologie Cellulaire, Inserm, University of Lille, Villeneuve d’Ascq, 59000 Lille, France; (M.M.A.); (N.P.)
- Laboratory of Cellular and Molecular Angiogenesis, Department of Life Sciences and Systems Biology, University of Turin, 10123 Turin, Italy
| |
Collapse
|
20
|
Hagelund S, Trauzold A. Impact of Extracellular pH on Apoptotic and Non-Apoptotic TRAIL-Induced Signaling in Pancreatic Ductal Adenocarcinoma Cells. Front Cell Dev Biol 2022; 10:768579. [PMID: 35281089 PMCID: PMC8907891 DOI: 10.3389/fcell.2022.768579] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 01/20/2022] [Indexed: 12/24/2022] Open
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is an important mediator of tumor immune surveillance. In addition, its potential to kill cancer cells without harming healthy cells led to the development of TRAIL receptor agonists, which however did not show the desired effects in clinical trials. This is caused mainly by apoptosis resistance mechanisms operating in primary cancer cells. Meanwhile, it has been realized that in addition to cell death, TRAIL also induces non-apoptotic pro-inflammatory pathways that may enhance tumor malignancy. Due to its late detection and resistance to current therapeutic options, pancreatic ductal adenocarcinoma (PDAC) is still one of the deadliest types of cancer worldwide. A dysregulated pH microenvironment contributes to PDAC development, in which the cancer cells become highly dependent on to maintain their metabolism. The impact of extracellular pH (pHe) on TRAIL-induced signaling in PDAC cells is poorly understood so far. To close this gap, we analyzed the effects of acidic and alkaline pHe, both in short-term and long-term settings, on apoptotic and non-apoptotic TRAIL-induced signaling. We found that acidic and alkaline pHe differentially impact TRAIL-induced responses, and in addition, the duration of the pHe exposition also represents an important parameter. Thus, adaptation to acidic pHe increases TRAIL sensitivity in two different PDAC cell lines, Colo357 and Panc1, one already TRAIL-sensitive and the other TRAIL-resistant, respectively. However, the latter became highly TRAIL-sensitive only by concomitant inhibition of Bcl-xL. None of these effects was observed under other pHe conditions studied. Both TRAIL-induced non-apoptotic signaling pathways, as well as constitutively expressed anti-apoptotic proteins, were regulated by acidic pHe. Whereas the non-apoptotic pathways were differently affected in Colo357 than in Panc1 cells, the impact on the anti-apoptotic protein levels was similar in both cell lines. In Panc1 cells, adaptation to either acidic or alkaline pHe blocked the activation of the most of TRAIL-induced non-apoptotic pathways. Interestingly, under these conditions, significant downregulation of the plasma membrane levels of TRAIL-R1 and TRAIL-R2 was observed. Summing up, extracellular pH influences PDAC cells’ response to TRAIL with acidic pHe adaptation, showing the ability to strongly increase TRAIL sensitivity and in addition to inhibit TRAIL-induced pro-inflammatory signaling.
Collapse
|
21
|
Mo L, Xu L, Jia M, Su B, Hu Y, Hu Z, Li H, Zhao C, Zhao Z, Li J. Shikonin suppresses the epithelial-to-mesenchymal transition by downregulating NHE1 in bladder cancer cells. J Cancer 2021; 12:6814-6824. [PMID: 34659570 PMCID: PMC8518005 DOI: 10.7150/jca.63429] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 09/19/2021] [Indexed: 12/01/2022] Open
Abstract
Shikonin (SK) is the major bioactive component extracted from the roots of Lithospermum erythrorhizon with anticancer activity. SK could inhibit the epithelial-to-mesenchymal transition (EMT) of cancer cells. However, the underlying mechanism is elusive. In the present study, the inhibitory activities of SK on proliferation, invasion and migration were examined in bladder cancer (BC) cells. SK potently decreased the viabilities of BC cells but showed less cytotoxicity to normal bladder epithelial cells. Moreover, SK reversed the EMT, suppressed the migration and invasion of BC cells. Intriguingly, NHE1, the major proton efflux pump, was dramatically down-regulated by SK. The EMT-inhibitory effect of SK was mediated by NHE1 down-regulation, as NHE1-overexpress alleviated while Cariporide (NHE1 inhibitor) enhanced this effect. Further, enforced alkalinization of intracellular pH (pHi) reversed the EMT-inhibitory effect of SK, indicating a key role of acidic pHi in this process. Finally, elevated NHE1 expression was observed in human bladder cancer tissues. Collectively, this research reveals a supportive effect of NHE1 and alkaline pHi on EMT. SK can suppress EMT through inhibiting NHE1 and hence inducing an acidic pHi.
Collapse
Affiliation(s)
- Lijun Mo
- Institute of Biotherapy, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, Guangdong, China.,Shenzhen Ruipuxun Academy for Stem Cell & Regenerative Medicine, 14 Jinhui Road, Shenzhen 518118, People's Republic of China
| | - Lili Xu
- Institute of Biotherapy, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, Guangdong, China.,Shenzhen Ruipuxun Academy for Stem Cell & Regenerative Medicine, 14 Jinhui Road, Shenzhen 518118, People's Republic of China
| | - Min Jia
- Shenzhen Ruipuxun Academy for Stem Cell & Regenerative Medicine, 14 Jinhui Road, Shenzhen 518118, People's Republic of China.,Department of Clinical Laboratory, Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Bijia Su
- Institute of Biotherapy, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, Guangdong, China.,Shenzhen Ruipuxun Academy for Stem Cell & Regenerative Medicine, 14 Jinhui Road, Shenzhen 518118, People's Republic of China
| | - Yaolong Hu
- Institute of Biotherapy, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, Guangdong, China
| | - Zhiming Hu
- Institute of Biotherapy, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, Guangdong, China
| | - Hongwei Li
- Institute of Biotherapy, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, Guangdong, China
| | - Chenye Zhao
- Shenzhen Ruipuxun Academy for Stem Cell & Regenerative Medicine, 14 Jinhui Road, Shenzhen 518118, People's Republic of China
| | - Zhenlin Zhao
- Shenzhen Ruipuxun Academy for Stem Cell & Regenerative Medicine, 14 Jinhui Road, Shenzhen 518118, People's Republic of China
| | - Jinlong Li
- Institute of Biotherapy, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, Guangdong, China.,Shenzhen Ruipuxun Academy for Stem Cell & Regenerative Medicine, 14 Jinhui Road, Shenzhen 518118, People's Republic of China
| |
Collapse
|
22
|
Pizzagalli MD, Bensimon A, Superti‐Furga G. A guide to plasma membrane solute carrier proteins. FEBS J 2021; 288:2784-2835. [PMID: 32810346 PMCID: PMC8246967 DOI: 10.1111/febs.15531] [Citation(s) in RCA: 186] [Impact Index Per Article: 46.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 08/07/2020] [Accepted: 08/17/2020] [Indexed: 12/13/2022]
Abstract
This review aims to serve as an introduction to the solute carrier proteins (SLC) superfamily of transporter proteins and their roles in human cells. The SLC superfamily currently includes 458 transport proteins in 65 families that carry a wide variety of substances across cellular membranes. While members of this superfamily are found throughout cellular organelles, this review focuses on transporters expressed at the plasma membrane. At the cell surface, SLC proteins may be viewed as gatekeepers of the cellular milieu, dynamically responding to different metabolic states. With altered metabolism being one of the hallmarks of cancer, we also briefly review the roles that surface SLC proteins play in the development and progression of cancer through their influence on regulating metabolism and environmental conditions.
Collapse
Affiliation(s)
- Mattia D. Pizzagalli
- CeMM, Research Center for Molecular Medicine of the Austrian Academy of SciencesViennaAustria
| | - Ariel Bensimon
- CeMM, Research Center for Molecular Medicine of the Austrian Academy of SciencesViennaAustria
| | - Giulio Superti‐Furga
- CeMM, Research Center for Molecular Medicine of the Austrian Academy of SciencesViennaAustria
- Center for Physiology and PharmacologyMedical University of ViennaAustria
| |
Collapse
|
23
|
Environmental control of mammary carcinoma cell expansion by acidification and spheroid formation in vitro. Sci Rep 2020; 10:21959. [PMID: 33319820 PMCID: PMC7738540 DOI: 10.1038/s41598-020-78989-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Accepted: 12/01/2020] [Indexed: 12/31/2022] Open
Abstract
Breast cancer is the leading cause of cancer death among women worldwide. Like other cancers, mammary carcinoma progression involves acidification of the tumor microenvironment, which is an important factor for cancer detection and treatment strategies. However, the effects of acidity on mammary carcinoma cell morphology and phenotype have not been thoroughly characterized. Here, we evaluated fundamental effects of environmental acidification on mammary carcinoma cells in standard two-dimensional cultures and three-dimensional spheroids. Acidification decreased overall mammary carcinoma cell viability, while increasing their resistance to the anthracycline doxorubicin. Environmental acidification also increased extracellular vesicle production by mammary carcinoma cells. Conditioned media containing these vesicles appeared to increase fibroblast motility. Acidification also increased mammary carcinoma cell motility when cultured with fibroblasts in spheroids. Taken together, results from this study suggest that environmental acidification induces drug resistance and extracellular vesicle production by mammary carcinoma cells that promote tumor expansion.
Collapse
|
24
|
Koltai T. Targeting the pH Paradigm at the Bedside: A Practical Approach. Int J Mol Sci 2020; 21:E9221. [PMID: 33287221 PMCID: PMC7730959 DOI: 10.3390/ijms21239221] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 11/27/2020] [Accepted: 11/28/2020] [Indexed: 02/07/2023] Open
Abstract
The inversion of the pH gradient in malignant tumors, known as the pH paradigm, is increasingly becoming accepted by the scientific community as a hallmark of cancer. Accumulated evidence shows that this is not simply a metabolic consequence of a dysregulated behavior, but rather an essential process in the physiopathology of accelerated proliferation and invasion. From the over-simplification of increased lactate production as the cause of the paradigm, as initially proposed, basic science researchers have arrived at highly complex and far-reaching knowledge, that substantially modified that initial belief. These new developments show that the paradigm entails a different regulation of membrane transporters, electrolyte exchangers, cellular and membrane enzymes, water trafficking, specialized membrane structures, transcription factors, and metabolic changes that go far beyond fermentative glycolysis. This complex world of dysregulations is still shuttered behind the walls of experimental laboratories and has not yet reached bedside medicine. However, there are many known pharmaceuticals and nutraceuticals that are capable of targeting the pH paradigm. Most of these products are well known, have low toxicity, and are also inexpensive. They need to be repurposed, and this would entail shorter clinical studies and enormous cost savings if we compare them with the time and expense required for the development of a new molecule. Will targeting the pH paradigm solve the "cancer problem"? Absolutely not. However, reversing the pH inversion would strongly enhance standard treatments, rendering them more efficient, and in some cases permitting lower doses of toxic drugs. This article's goal is to describe how to reverse the pH gradient inversion with existing drugs and nutraceuticals that can easily be used in bedside medicine, without adding toxicity to established treatments. It also aims at increasing awareness among practicing physicians that targeting the pH paradigm would be able to improve the results of standard therapies. Some clinical cases will be presented as well, showing how the pH gradient inversion can be treated at the bedside in a simple manner with repurposed drugs.
Collapse
Affiliation(s)
- Tomas Koltai
- Centro de Diagnostico y Tratamiento de la Obra Social del Personal de la Alimentacion, Talar de Pacheco, Buenos Aires 1617, Argentina
| |
Collapse
|
25
|
Role of Genetic Mutations of the Na +/H + Exchanger Isoform 1, in Human Disease and Protein Targeting and Activity. Mol Cell Biochem 2020; 476:1221-1232. [PMID: 33201382 DOI: 10.1007/s11010-020-03984-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 11/06/2020] [Indexed: 01/22/2023]
Abstract
The mammalian Na+/H+ exchanger isoform one (NHE1) is a plasma membrane protein that is ubiquitously present in human cells. It functions to regulate intracellular pH removing an intracellular proton in exchange for one extracellular sodium and is involved in heart disease and in promoting metastasis in cancer. It is made of a 500 amino acid membrane domain plus a 315 amino acid, regulatory cytosolic tail. The membrane domain is thought to have 12 transmembrane segments and a large membrane-associated extracellular loop. Early studies demonstrated that in mice, disruption of the NHE1 gene results in locomotor ataxia and a phenotype of slow-wave epilepsy. Defects included a progressive neuronal degeneration. Growth and reproductive ability were also reduced. Recent studies have identified human autosomal homozygous recessive mutations in the NHE1 gene (SLC9A1) that result in impaired development, ataxia and other severe defects, and explain the cause of the human disease Lichtenstein-Knorr syndrome. Other human mutations have been identified that are stop codon polymorphisms. These cause short non-functional NHE1 proteins, while other genetic polymorphisms in the NHE1 gene cause impaired expression of the NHE1 protein, reduced activity, enhanced protein degradation or altered kinetic activation of the protein. Since NHE1 plays a key role in many human physiological functions and in human disease, genetic polymorphisms of the protein that significantly alter its function and are likely play significant roles in varying human phenotypes and be involved in disease.
Collapse
|
26
|
Venkateswaran G, Dedhar S. Interplay of Carbonic Anhydrase IX With Amino Acid and Acid/Base Transporters in the Hypoxic Tumor Microenvironment. Front Cell Dev Biol 2020; 8:602668. [PMID: 33240897 PMCID: PMC7680889 DOI: 10.3389/fcell.2020.602668] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 10/09/2020] [Indexed: 01/13/2023] Open
Abstract
Solid tumors are challenged with a hypoxic and nutrient-deprived microenvironment. Hence, hypoxic tumor cells coordinatively increase the expression of nutrient transporters and pH regulators to adapt and meet their bioenergetic and biosynthetic demands. Carbonic Anhydrase IX (CAIX) is a membrane-bound enzyme that plays a vital role in pH regulation in the tumor microenvironment (TME). Numerous studies have established the importance of CAIX in mediating tumor progression and metastasis. To understand the mechanism of CAIX in mediating tumor progression, we performed an unbiased proteomic screen to identify the potential interactors of CAIX in the TME using the proximity-dependent biotin identification (BioID) technique. In this review, we focus on the interactors from this BioID screen that are crucial for nutrient and metabolite transport in the TME. We discuss the role of transport metabolon comprising CAIX and bicarbonate transporters in regulating intra- and extracellular pH of the tumor. We also discuss the role of amino acid transporters that are high confidence interactors of CAIX, in optimizing favorable metabolic state for tumor progression, and give our perspective on the coordinative interplay of CAIX with the amino acid transporters in the hypoxic TME.
Collapse
Affiliation(s)
- Geetha Venkateswaran
- Department of Integrative Oncology, British Columbia Cancer Research Centre, Vancouver, BC, Canada.,Interdisciplinary Oncology Program, The University of British Columbia, Vancouver, BC, Canada
| | - Shoukat Dedhar
- Department of Integrative Oncology, British Columbia Cancer Research Centre, Vancouver, BC, Canada.,Interdisciplinary Oncology Program, The University of British Columbia, Vancouver, BC, Canada.,Department of Biochemistry and Molecular Biology, The University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
27
|
Harguindey S, Alfarouk K, Polo Orozco J, Fais S, Devesa J. Towards an Integral Therapeutic Protocol for Breast Cancer Based upon the New H +-Centered Anticancer Paradigm of the Late Post-Warburg Era. Int J Mol Sci 2020; 21:E7475. [PMID: 33050492 PMCID: PMC7589677 DOI: 10.3390/ijms21207475] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 10/05/2020] [Accepted: 10/06/2020] [Indexed: 12/13/2022] Open
Abstract
A brand new approach to the understanding of breast cancer (BC) is urgently needed. In this contribution, the etiology, pathogenesis, and treatment of this disease is approached from the new pH-centric anticancer paradigm. Only this unitarian perspective, based upon the hydrogen ion (H+) dynamics of cancer, allows for the understanding and integration of the many dualisms, confusions, and paradoxes of the disease. The new H+-related, wide-ranging model can embrace, from a unique perspective, the many aspects of the disease and, at the same time, therapeutically interfere with most, if not all, of the hallmarks of cancer known to date. The pH-related armamentarium available for the treatment of BC reviewed here may be beneficial for all types and stages of the disease. In this vein, we have attempted a megasynthesis of traditional and new knowledge in the different areas of breast cancer research and treatment based upon the wide-ranging approach afforded by the hydrogen ion dynamics of cancer. The concerted utilization of the pH-related drugs that are available nowadays for the treatment of breast cancer is advanced.
Collapse
Affiliation(s)
- Salvador Harguindey
- Department of Oncology, Institute of Clinical Biology and Metabolism, 01004 Vitoria, Spain;
| | - Khalid Alfarouk
- Department of Pharmacology, Al-Ghad International Colleges for Applied Medical Sciences, Al-Madinah Al-Munawarah 42316, Saudi Arabia and Alfarouk Biomedical Research LLC, Tampa, FL 33617, USA;
| | - Julián Polo Orozco
- Department of Oncology, Institute of Clinical Biology and Metabolism, 01004 Vitoria, Spain;
| | - Stefano Fais
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità (National Institute of Health), 00161 Rome, Italy;
| | - Jesús Devesa
- Scientific Direction, Foltra Medical Centre, 15886 Teo, Spain;
| |
Collapse
|
28
|
Liu Y, White KA, Barber DL. Intracellular pH Regulates Cancer and Stem Cell Behaviors: A Protein Dynamics Perspective. Front Oncol 2020; 10:1401. [PMID: 32983969 PMCID: PMC7479815 DOI: 10.3389/fonc.2020.01401] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 07/02/2020] [Indexed: 12/12/2022] Open
Abstract
The International Society of Cancer Metabolism (ISCaM) meeting on Cancer Metabolic Rewiring, held in Braga Portugal in October 2019, provided an outstanding forum for investigators to present current findings and views, and discuss ideas and future directions on fundamental biology as well as clinical translations. The first session on Cancer pH Dynamics was preceded by the opening keynote presentation from our group entitled Intracellular pH Regulation of Protein Dynamics: From Cancer to Stem Cell Behaviors. In this review we introduce a brief background on intracellular pH (pHi) dynamics, including how it is regulated as well as functional consequences, summarize key findings included in our presentation, and conclude with perspectives on how understanding the role of pHi dynamics in stem cells can be relevant for understanding how pHi dynamics enables cancer progression.
Collapse
Affiliation(s)
- Yi Liu
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA, United States
| | - Katharine A White
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA, United States
| | - Diane L Barber
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA, United States
| |
Collapse
|
29
|
Bagdonaite I, Pallesen EM, Ye Z, Vakhrushev SY, Marinova IN, Nielsen MI, Kramer SH, Pedersen SF, Joshi HJ, Bennett EP, Dabelsteen S, Wandall HH. O-glycan initiation directs distinct biological pathways and controls epithelial differentiation. EMBO Rep 2020; 21:e48885. [PMID: 32329196 PMCID: PMC7271655 DOI: 10.15252/embr.201948885] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 03/03/2020] [Accepted: 03/16/2020] [Indexed: 12/17/2022] Open
Abstract
Post-translational modifications (PTMs) greatly expand the function and potential for regulation of protein activity, and O-glycosylation is among the most abundant and diverse PTMs. Initiation of O-GalNAc glycosylation is regulated by 20 distinct GalNAc-transferases (GalNAc-Ts), and deficiencies in individual GalNAc-Ts are associated with human disease, causing subtle but distinct phenotypes in model organisms. Here, we generate a set of isogenic keratinocyte cell lines lacking either of the three dominant and differentially expressed GalNAc-Ts. Through the ability of keratinocytes to form epithelia, we investigate the phenotypic consequences of the loss of individual GalNAc-Ts. Moreover, we probe the cellular responses through global transcriptomic, differential glycoproteomic, and differential phosphoproteomic analyses. We demonstrate that loss of individual GalNAc-T isoforms causes distinct epithelial phenotypes through their effect on specific biological pathways; GalNAc-T1 targets are associated with components of the endomembrane system, GalNAc-T2 targets with cell-ECM adhesion, and GalNAc-T3 targets with epithelial differentiation. Thus, GalNAc-T isoforms serve specific roles during human epithelial tissue formation.
Collapse
Affiliation(s)
- Ieva Bagdonaite
- Copenhagen Center for Glycomics, Institute of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Emil Mh Pallesen
- Copenhagen Center for Glycomics, Institute of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Zilu Ye
- Copenhagen Center for Glycomics, Institute of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Sergey Y Vakhrushev
- Copenhagen Center for Glycomics, Institute of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Irina N Marinova
- Copenhagen Center for Glycomics, Institute of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Mathias I Nielsen
- Copenhagen Center for Glycomics, Institute of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Signe H Kramer
- Cell Biology and Physiology, Department of Science, University of Copenhagen, Copenhagen, Denmark
| | - Stine F Pedersen
- Cell Biology and Physiology, Department of Science, University of Copenhagen, Copenhagen, Denmark
| | - Hiren J Joshi
- Copenhagen Center for Glycomics, Institute of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Eric P Bennett
- Copenhagen Center for Glycomics, Institute of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark.,School of Dentistry, University of Copenhagen, Copenhagen, Denmark
| | - Sally Dabelsteen
- School of Dentistry, University of Copenhagen, Copenhagen, Denmark
| | - Hans H Wandall
- Copenhagen Center for Glycomics, Institute of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
30
|
Targeting the Acidic Tumor Microenvironment: Unexpected Pro-Neoplastic Effects of Oral NaHCO 3 Therapy in Murine Breast Tissue. Cancers (Basel) 2020; 12:cancers12040891. [PMID: 32268614 PMCID: PMC7226235 DOI: 10.3390/cancers12040891] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 03/31/2020] [Accepted: 03/31/2020] [Indexed: 11/16/2022] Open
Abstract
The acidic tumor microenvironment modifies malignant cell behavior. Here, we study consequences of the microenvironment in breast carcinomas. Beginning at carcinogen-based breast cancer induction, we supply either regular or NaHCO3-containing drinking water to female C57BL/6j mice. We evaluate urine and blood acid-base status, tumor metabolism (microdialysis sampling), and tumor pH (pH-sensitive microelectrodes) in vivo. Based on freshly isolated epithelial organoids from breast carcinomas and normal breast tissue, we assess protein expression (immunoblotting, mass spectrometry), intracellular pH (fluorescence microscopy), and cell proliferation (bromodeoxyuridine incorporation). Oral NaHCO3 therapy increases breast tumor pH in vivo from 6.68 ± 0.04 to 7.04 ± 0.09 and intracellular pH in breast epithelial organoids by ~0.15. Breast tumors develop with median latency of 85.5 ± 8.2 days in NaHCO3-treated mice vs. 82 ± 7.5 days in control mice. Oral NaHCO3 therapy does not affect tumor growth, histopathology or glycolytic metabolism. The capacity for cellular net acid extrusion is increased in NaHCO3-treated mice and correlates negatively with breast tumor latency. Oral NaHCO3 therapy elevates proliferative activity in organoids from breast carcinomas. Changes in protein expression patterns-observed by high-throughput proteomics analyses-between cancer and normal breast tissue and in response to oral NaHCO3 therapy reveal complex influences on metabolism, cytoskeleton, cell-cell and cell-matrix interaction, and cell signaling pathways. We conclude that oral NaHCO3 therapy neutralizes the microenvironment of breast carcinomas, elevates the cellular net acid extrusion capacity, and accelerates proliferation without net effect on breast cancer development or tumor growth. We demonstrate unexpected pro-neoplastic consequences of oral NaHCO3 therapy that in breast tissue cancel out previously reported anti-neoplastic effects.
Collapse
|
31
|
Abstract
Acidic metabolic waste products accumulate in the tumor microenvironment because of high metabolic activity and insufficient perfusion. In tumors, the acidity of the interstitial space and the relatively well-maintained intracellular pH influence cancer and stromal cell function, their mutual interplay, and their interactions with the extracellular matrix. Tumor pH is spatially and temporally heterogeneous, and the fitness advantage of cancer cells adapted to extracellular acidity is likely particularly evident when they encounter less acidic tumor regions, for instance, during invasion. Through complex effects on genetic stability, epigenetics, cellular metabolism, proliferation, and survival, the compartmentalized pH microenvironment favors cancer development. Cellular selection exacerbates the malignant phenotype, which is further enhanced by acid-induced cell motility, extracellular matrix degradation, attenuated immune responses, and modified cellular and intercellular signaling. In this review, we discuss how the acidity of the tumor microenvironment influences each stage in cancer development, from dysplasia to full-blown metastatic disease.
Collapse
Affiliation(s)
- Ebbe Boedtkjer
- Department of Biomedicine, Aarhus University, DK-8000 Aarhus C, Denmark
| | - Stine F. Pedersen
- Department of Biology, University of Copenhagen, DK-2100 Copenhagen, Denmark
| |
Collapse
|
32
|
Harguindey S, Alfarouk K, Polo Orozco J, Hardonnière K, Stanciu D, Fais S, Devesa J. A New and Integral Approach to the Etiopathogenesis and Treatment of Breast Cancer Based upon Its Hydrogen Ion Dynamics. Int J Mol Sci 2020; 21:E1110. [PMID: 32046158 PMCID: PMC7036897 DOI: 10.3390/ijms21031110] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 02/04/2020] [Accepted: 02/06/2020] [Indexed: 12/11/2022] Open
Abstract
Despite all efforts, the treatment of breast cancer (BC) cannot be considered to be a success story. The advances in surgery, chemotherapy and radiotherapy have not been sufficient at all. Indeed, the accumulated experience clearly indicates that new perspectives and non-main stream approaches are needed to better characterize the etiopathogenesis and treatment of this disease. This contribution deals with how the new pH-centric anticancer paradigm plays a fundamental role in reaching a more integral understanding of the etiology, pathogenesis, and treatment of this multifactorial disease. For the first time, the armamentarium available for the treatment of the different types and phases of BC is approached here from a Unitarian perspective-based upon the hydrogen ion dynamics of cancer. The wide-ranged pH-related molecular, biochemical and metabolic model is able to embrace most of the fields and subfields of breast cancer etiopathogenesis and treatment. This single and integrated approach allows advancing towards a unidirectional, concerted and synergistic program of treatment. Further efforts in this line are likely to first improve the therapeutics of each subtype of this tumor and every individual patient in every phase of the disease.
Collapse
Affiliation(s)
- Salvador Harguindey
- Institute of Clinical Biology and Metabolism, Postas 13, 01004 Vitoria, Spain;
| | - Khalid Alfarouk
- Al-Ghad International Colleges for Applied Medical Sciences, Al-Madinah Al-Munawarah, Saudi Arabia and Alfarouk Biomedical Research LLC, Tampa, FL 33617, USA;
| | - Julián Polo Orozco
- Institute of Clinical Biology and Metabolism, Postas 13, 01004 Vitoria, Spain;
| | - Kévin Hardonnière
- Université Paris-Saclay, Inserm, Inflammation, Microbiome and Immunosurveillance, 92290 Châtenay-Malabry, France;
| | - Daniel Stanciu
- Scientific Direction, MCS Foundation For Life, 5623KR Eindhoven, The Netherlands;
| | - Stefano Fais
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità (National Institute of Health), Viale Regina Elena, 299, 00161 Rome, Italy;
| | - Jesús Devesa
- Scientific Direction, Foltra Medical Centre, Travesía de Montouto 24, 15886 Teo, Spain;
| |
Collapse
|
33
|
Cao L, Yuan Z, Liu M, Stock C. (Patho-)Physiology of Na +/H + Exchangers (NHEs) in the Digestive System. Front Physiol 2020; 10:1566. [PMID: 32009977 PMCID: PMC6974801 DOI: 10.3389/fphys.2019.01566] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Accepted: 12/12/2019] [Indexed: 02/06/2023] Open
Abstract
Na+/H+ exchangers (NHEs) are expressed in virtually all human tissues and organs. Two major tasks of those NHE isoforms that are located in plasma membranes are cell volume control by Na+-uptake and cellular pH regulation by H+-extrusion. Several NHEs, particularly NHE 1–4 and 8, are involved in the pathogenesis of diseases of the digestive system such as inflammatory bowel disease (ulcerative colitis, Crohn’s disease) and gastric and colorectal tumorigenesis. In the present review, we describe the physiological purposes, possible malfunctions and pathophysiological effects of the different NHE isoforms along the alimentary canal from esophagus to colon, including pancreas, liver and gallbladder. Particular attention is paid to the functions of NHEs in injury repair and to the role of NHE1 in Barrett’s esophagus. The impact of NHEs on gut microbiota and intestinal mucosal integrity is also dealt with. As the hitherto existing findings are not always consistent, sometimes even controversial, they are compared and critically discussed.
Collapse
Affiliation(s)
- Li Cao
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhenglin Yuan
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Mei Liu
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Christian Stock
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hanover, Germany
| |
Collapse
|
34
|
3D multicellular models to study the regulation and roles of acid-base transporters in breast cancer. Biochem Soc Trans 2019; 47:1689-1700. [PMID: 31803922 DOI: 10.1042/bst20190131] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 11/01/2019] [Accepted: 11/12/2019] [Indexed: 12/24/2022]
Abstract
As a result of elevated metabolic rates and net acid extrusion in the rapidly proliferating cancer cells, solid tumours are characterized by a highly acidic microenvironment, while cancer cell intracellular pH is normal or even alkaline. Two-dimensional (2D) cell monocultures, which have been used extensively in breast cancer research for decades, cannot precisely recapitulate the rich environment and complex processes occurring in tumours in vivo. The use of such models can consequently be misleading or non-predictive for clinical applications. Models mimicking the tumour microenvironment are particularly pivotal for studying tumour pH homeostasis, which is profoundly affected by the diffusion-limited conditions in the tumour. To advance the understanding of the mechanisms and consequences of dysregulated acid-base homeostasis in breast cancer, clinically relevant models that incorporate the unique microenvironment of these tumours are required. The development of three-dimensional (3D) cell cultures has provided new tools for basic research and pre-clinical approaches, allowing the culture of breast cancer cells under conditions that closely resemble tumour growth in a living organism. Here we provide an overview of the main 3D techniques relevant for breast cancer cell culture. We discuss the advantages and limitations of the classical 3D models as well as recent advances in 3D culture techniques, focusing on how these culture methods have been used to study acid-base transport in breast cancer. Finally, we outline future directions of 3D culture technology and their relevance for studies of acid-base transport.
Collapse
|
35
|
Keurhorst D, Liashkovich I, Frontzek F, Nitzlaff S, Hofschröer V, Dreier R, Stock C. MMP3 activity rather than cortical stiffness determines NHE1-dependent invasiveness of melanoma cells. Cancer Cell Int 2019; 19:285. [PMID: 31728131 PMCID: PMC6842528 DOI: 10.1186/s12935-019-1015-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 11/01/2019] [Indexed: 12/31/2022] Open
Abstract
Background Both cell adhesion and matrix metalloproteinase (MMP) activity depend on pH at the cell surface. By regulating extracellular juxtamembrane pH, the Na+/H+ exchanger NHE1 plays a significant part in human melanoma (MV3) cell migration and invasion. Because NHE1, besides its pH-regulatory transport function, also serves as a structural element tying the cortical actin cytoskeleton to the plasma membrane, we investigated whether NHE1 affects cortical stiffness of MV3 cells, and how this makes an impact on their invasiveness. Methods NHE1 overexpressing MV3 cells were compared to the corresponding mock-transfected control cells. NHE1 expression was verified by Western blotting, cariporide (HOE642) was used to inhibit NHE1 activity, cell stiffness was determined by atomic force microscopy, and F-actin was visualized by phalloidin-staining. Migration on, and invasion of, native and glutaraldehyde-fixed collagen I substrates were analyzed using time-lapse video microscopy and Boyden-chamber assays, respectively. MMP secretion and activity were detected by Western blot and zymography, respectively. MMP activity was inhibited with NNGH. Results The cortical, but not the bulk stiffness, was significantly higher in NHE1 overexpressing cells. This increase in cortical stiffness was accompanied by a reorganization of the cortical cytoskeleton, i.e. a condensation of F-actin underneath and along the plasma membrane. However, it was not affected by NHE1 inhibition. Nevertheless, actin dynamics is required for cell invasion as demonstrated with the application of cytochalasin D. NHE1 overexpression was associated with an elevated MMP3 secretion and an increase in the invasion of a native matrix. This increase in invasiveness could be antagonized by the MMP inhibitor NNGH. Transmigration through a glutaraldehyde-fixed, indigestible substrate was not affected by NHE1 overexpression. Conclusion NHE1, as a structural element and independently of its transport activity, contributes to the organization of the cortical F-actin meshwork and thus impacts cortical stiffness. Since NHE1 overexpression stimulates MMP3 secretion but does not change transmigration through a fixed substrate, MV3 cell invasion of a native substrate depends on MMP activity rather than on a modifiable cortical stiffness.
Collapse
Affiliation(s)
- Dennis Keurhorst
- 1Institute of Physiology II, University of Münster, Robert-Koch-Str. 27b, 48149 Münster, Germany
| | - Ivan Liashkovich
- 1Institute of Physiology II, University of Münster, Robert-Koch-Str. 27b, 48149 Münster, Germany
| | - Fabian Frontzek
- 2Department of Oncology and Hematology, University Hospital of Münster, Albert-Schweitzer-Campus 1, 48149 Münster, Germany
| | - Svenja Nitzlaff
- 3Institute of Animal Physiology, University of Münster, Schlossplatz 8, 48143 Münster, Germany
| | - Verena Hofschröer
- 1Institute of Physiology II, University of Münster, Robert-Koch-Str. 27b, 48149 Münster, Germany
| | - Rita Dreier
- 4Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Waldeyer-Str. 15, 48149 Münster, Germany
| | - Christian Stock
- 1Institute of Physiology II, University of Münster, Robert-Koch-Str. 27b, 48149 Münster, Germany.,5Department of Gastroenterology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| |
Collapse
|
36
|
Pedersen SF, Counillon L. The SLC9A-C Mammalian Na +/H + Exchanger Family: Molecules, Mechanisms, and Physiology. Physiol Rev 2019; 99:2015-2113. [PMID: 31507243 DOI: 10.1152/physrev.00028.2018] [Citation(s) in RCA: 112] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Na+/H+ exchangers play pivotal roles in the control of cell and tissue pH by mediating the electroneutral exchange of Na+ and H+ across cellular membranes. They belong to an ancient family of highly evolutionarily conserved proteins, and they play essential physiological roles in all phyla. In this review, we focus on the mammalian Na+/H+ exchangers (NHEs), the solute carrier (SLC) 9 family. This family of electroneutral transporters constitutes three branches: SLC9A, -B, and -C. Within these, each isoform exhibits distinct tissue expression profiles, regulation, and physiological roles. Some of these transporters are highly studied, with hundreds of original articles, and some are still only rudimentarily understood. In this review, we present and discuss the pioneering original work as well as the current state-of-the-art research on mammalian NHEs. We aim to provide the reader with a comprehensive view of core knowledge and recent insights into each family member, from gene organization over protein structure and regulation to physiological and pathophysiological roles. Particular attention is given to the integrated physiology of NHEs in the main organ systems. We provide several novel analyses and useful overviews, and we pinpoint main remaining enigmas, which we hope will inspire novel research on these highly versatile proteins.
Collapse
Affiliation(s)
- S F Pedersen
- Section for Cell Biology and Physiology, Department of Biology, University of Copenhagen, Copenhagen, Denmark; and Université Côte d'Azur, CNRS, Laboratoire de Physiomédecine Moléculaire, LP2M, France, and Laboratories of Excellence Ion Channel Science and Therapeutics, Nice, France
| | - L Counillon
- Section for Cell Biology and Physiology, Department of Biology, University of Copenhagen, Copenhagen, Denmark; and Université Côte d'Azur, CNRS, Laboratoire de Physiomédecine Moléculaire, LP2M, France, and Laboratories of Excellence Ion Channel Science and Therapeutics, Nice, France
| |
Collapse
|
37
|
Anderson KJ, Cormier RT, Scott PM. Role of ion channels in gastrointestinal cancer. World J Gastroenterol 2019; 25:5732-5772. [PMID: 31636470 PMCID: PMC6801186 DOI: 10.3748/wjg.v25.i38.5732] [Citation(s) in RCA: 113] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Revised: 07/26/2019] [Accepted: 09/27/2019] [Indexed: 02/06/2023] Open
Abstract
In their seminal papers Hanahan and Weinberg described oncogenic processes a normal cell undergoes to be transformed into a cancer cell. The functions of ion channels in the gastrointestinal (GI) tract influence a variety of cellular processes, many of which overlap with these hallmarks of cancer. In this review we focus on the roles of the calcium (Ca2+), sodium (Na+), potassium (K+), chloride (Cl-) and zinc (Zn2+) transporters in GI cancer, with a special emphasis on the roles of the KCNQ1 K+ channel and CFTR Cl- channel in colorectal cancer (CRC). Ca2+ is a ubiquitous second messenger, serving as a signaling molecule for a variety of cellular processes such as control of the cell cycle, apoptosis, and migration. Various members of the TRP superfamily, including TRPM8, TRPM7, TRPM6 and TRPM2, have been implicated in GI cancers, especially through overexpression in pancreatic adenocarcinomas and down-regulation in colon cancer. Voltage-gated sodium channels (VGSCs) are classically associated with the initiation and conduction of action potentials in electrically excitable cells such as neurons and muscle cells. The VGSC NaV1.5 is abundantly expressed in human colorectal CRC cell lines as well as being highly expressed in primary CRC samples. Studies have demonstrated that conductance through NaV1.5 contributes significantly to CRC cell invasiveness and cancer progression. Zn2+ transporters of the ZIP/SLC39A and ZnT/SLC30A families are dysregulated in all major GI organ cancers, in particular, ZIP4 up-regulation in pancreatic cancer (PC). More than 70 K+ channel genes, clustered in four families, are found expressed in the GI tract, where they regulate a range of cellular processes, including gastrin secretion in the stomach and anion secretion and fluid balance in the intestinal tract. Several distinct types of K+ channels are found dysregulated in the GI tract. Notable are hERG1 upregulation in PC, gastric cancer (GC) and CRC, leading to enhanced cancer angiogenesis and invasion, and KCNQ1 down-regulation in CRC, where KCNQ1 expression is associated with enhanced disease-free survival in stage II, III, and IV disease. Cl- channels are critical for a range of cellular and tissue processes in the GI tract, especially fluid balance in the colon. Most notable is CFTR, whose deficiency leads to mucus blockage, microbial dysbiosis and inflammation in the intestinal tract. CFTR is a tumor suppressor in several GI cancers. Cystic fibrosis patients are at a significant risk for CRC and low levels of CFTR expression are associated with poor overall disease-free survival in sporadic CRC. Two other classes of chloride channels that are dysregulated in GI cancers are the chloride intracellular channels (CLIC1, 3 & 4) and the chloride channel accessory proteins (CLCA1,2,4). CLIC1 & 4 are upregulated in PC, GC, gallbladder cancer, and CRC, while the CLCA proteins have been reported to be down-regulated in CRC. In summary, it is clear, from the diverse influences of ion channels, that their aberrant expression and/or activity can contribute to malignant transformation and tumor progression. Further, because ion channels are often localized to the plasma membrane and subject to multiple layers of regulation, they represent promising clinical targets for therapeutic intervention including the repurposing of current drugs.
Collapse
Affiliation(s)
- Kyle J Anderson
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN 55812, United States
| | - Robert T Cormier
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN 55812, United States
| | - Patricia M Scott
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN 55812, United States
| |
Collapse
|
38
|
Ma Z, Yuan D, Cheng X, Tuo B, Liu X, Li T. Function of ion transporters in maintaining acid-base homeostasis of the mammary gland and the pathophysiological role in breast cancer. Am J Physiol Regul Integr Comp Physiol 2019; 318:R98-R111. [PMID: 31553634 DOI: 10.1152/ajpregu.00202.2019] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The incidence of breast cancer is increasing year by year, and the pathogenesis is still unclear. Studies have shown that the high metabolism of solid tumors leads to an increase in hypoxia, glycolysis, production of lactic acid and carbonic acid, and extracellular acidification; a harsh microenvironment; and ultimately to tumor cell death. Approximately 50% of locally advanced breast cancers exhibit hypoxia and/or local hypoxia, and acid-base regulatory proteins play an important role in regulating milk secretion and maintaining mammary gland physiological function. Therefore, ion transporters have gradually become a hot topic in mammary gland and breast cancer research. This review focuses on the research progress of ion transporters in mammary glands and breast cancer. We hope to provide new targets for the treatment and prognosis of breast cancer.
Collapse
Affiliation(s)
- Zhiyuan Ma
- Department of Thyroid and Breast Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Dumin Yuan
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical University, Zunyi, China.,Digestive Disease Institute of Guizhou Province, Zunyi, China
| | - Xiaoming Cheng
- Department of Thyroid and Breast Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Biguang Tuo
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical University, Zunyi, China.,Digestive Disease Institute of Guizhou Province, Zunyi, China
| | - Xuemei Liu
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical University, Zunyi, China.,Digestive Disease Institute of Guizhou Province, Zunyi, China
| | - Taolang Li
- Department of Thyroid and Breast Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| |
Collapse
|
39
|
Charafeddine RA, Cortopassi WA, Lak P, Tan R, McKenney RJ, Jacobson MP, Barber DL, Wittmann T. Tau repeat regions contain conserved histidine residues that modulate microtubule-binding in response to changes in pH. J Biol Chem 2019; 294:8779-8790. [PMID: 30992364 DOI: 10.1074/jbc.ra118.007004] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 04/13/2019] [Indexed: 12/20/2022] Open
Abstract
Tau, a member of the MAP2/tau family of microtubule-associated proteins, stabilizes and organizes axonal microtubules in healthy neurons. In neurodegenerative tauopathies, tau dissociates from microtubules and forms neurotoxic extracellular aggregates. MAP2/tau family proteins are characterized by three to five conserved, intrinsically disordered repeat regions that mediate electrostatic interactions with the microtubule surface. Here, we used molecular dynamics, microtubule-binding experiments, and live-cell microscopy, revealing that highly-conserved histidine residues near the C terminus of each microtubule-binding repeat are pH sensors that can modulate tau-microtubule interaction strength within the physiological intracellular pH range. We observed that at low pH (<7.5), these histidines are positively charged and interact with phenylalanine residues in a hydrophobic cleft between adjacent tubulin dimers. At higher pH (>7.5), tau deprotonation decreased binding to microtubules both in vitro and in cells. Electrostatic and hydrophobic characteristics of histidine were both required for tau-microtubule binding, as substitutions with constitutively and positively charged nonaromatic lysine or uncharged alanine greatly reduced or abolished tau-microtubule binding. Consistent with these findings, tau-microtubule binding was reduced in a cancer cell model with increased intracellular pH but was rapidly restored by decreasing the pH to normal levels. These results add detailed insights into the intracellular regulation of tau activity that may be relevant in both normal and pathological conditions.
Collapse
Affiliation(s)
- Rabab A Charafeddine
- From the Department of Cell and Tissue Biology, University of California San Francisco, San Francisco, California 94143
| | - Wilian A Cortopassi
- the Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, California 94158, and
| | - Parnian Lak
- the Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, California 94158, and
| | - Ruensern Tan
- the Department of Molecular and Cellular Biology, University of California Davis, Davis, California 95616
| | - Richard J McKenney
- the Department of Molecular and Cellular Biology, University of California Davis, Davis, California 95616
| | - Matthew P Jacobson
- the Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, California 94158, and
| | - Diane L Barber
- From the Department of Cell and Tissue Biology, University of California San Francisco, San Francisco, California 94143
| | - Torsten Wittmann
- From the Department of Cell and Tissue Biology, University of California San Francisco, San Francisco, California 94143,
| |
Collapse
|
40
|
|
41
|
Zhou ZH, Wang QL, Mao LH, Li XQ, Liu P, Song JW, Liu X, Xu F, Lei J, He S. Chromatin accessibility changes are associated with enhanced growth and liver metastasis capacity of acid-adapted colorectal cancer cells. Cell Cycle 2019; 18:511-522. [PMID: 30712429 PMCID: PMC6422493 DOI: 10.1080/15384101.2019.1578145] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2018] [Revised: 11/19/2018] [Accepted: 12/27/2018] [Indexed: 02/08/2023] Open
Abstract
The acidic extracellular microenvironment, namely acidosis, is a biochemical hallmark of solid tumors. However, the tumorigenicity, metastatic potential, gene expression profile and chromatin accessibility of acidosis-adapted colorectal cancer cells remain unknown. The colorectal cancer cell SW620 was cultured in acidic medium (pH 6.5) for more than 3 months to be acidosis-adapted (SW620-AA). In comparison to parental cells, SW620-AA cells exhibit enhanced tumorigenicity and liver metastatic potential in vivo. Following mRNA and lncRNA expression profiling, we validated that OLMF1, NFIB, SMAD9, DGKB are upregulated, while SESN2, MAP1B, UTRN, PCDH19, IL18, LMO2, CNKSR3, GXYLT2 are downregulated in SW620-AA cells. The differentially expressed mRNAs were significantly enriched in DNA remodeling-associated pathways including HDACs deacetylate histones, SIRT1 pathway, DNA methylation, DNA bending complex, and RNA polymerase 1 chain elongation. Finally, chromatin accessibility evaluation by ATAC-sequencing revealed that the differentially opened peaks were enriched in pathways such as small cell lung cancer, pathways in cancer, ErbB signaling, endometrial cancer, and chronic myeloid leukemia, which were mainly distributed in intergenic regions and introns. These results suggest that the chromatin accessibility changes are correlated with enhanced growth and liver metastasis capacity of acid-adapted colorectal cancer cells.
Collapse
Affiliation(s)
- Zhi-Hang Zhou
- Department of Gastroenterology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Pathology, the 309 hospital of PLA, Beijing, China
| | - Qing-Liang Wang
- Department of Pathology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lin-Hong Mao
- Department of Gastroenterology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiao-Qin Li
- Department of Gastroenterology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Peng Liu
- Department of Emergency, Chest Pain Center, General Hospital of Guangzhou Military Command, Guangzhou, China
| | - Jin-Wen Song
- Treatment and Research Center for Infectious Diseases, The 302nd hosptital of PLA, Beijing, China
| | - Xue Liu
- Department of Pathology, Basic Science School, Jining Medical University, Jining, Shandong, China
| | - Feng Xu
- Department of Gastroenterology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jing Lei
- Department of Gastroenterology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Song He
- Department of Gastroenterology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
42
|
Guan X, Luo L, Begum G, Kohanbash G, Song Q, Rao A, Amankulor N, Sun B, Sun D, Jia W. Elevated Na/H exchanger 1 (SLC9A1) emerges as a marker for tumorigenesis and prognosis in gliomas. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2018; 37:255. [PMID: 30333031 PMCID: PMC6192309 DOI: 10.1186/s13046-018-0923-z] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Accepted: 10/02/2018] [Indexed: 12/11/2022]
Abstract
Background Sodium/hydrogen exchanger 1 (NHE1), encoded by the SLC9A1 gene (SoLute Carrier family 9A1) in humans, is the main H+ efflux mechanism in maintaining alkaline intracellular pH (pHi) and Warburg effects in glioma. However, to date, there are no clinical studies exploring pharmacological inhibition of NHE1 protein in cancer treatment. In this study, we investigated NHE1 expression in gliomas and its relationship with glioma clinical outcome. Methods The Chinese Glioma Genome Atlas (CGGA) dataset containing transcriptome sequencing data of 325 glioma samples and the Cancer Genome Atlas (TCGA) with 698 glioma mRNAseq data were analyzed in this study. Mouse SB28 and GL26 intracranial syngeneic glioma models in C57BL/6 J mice were established to investigate NHE1 expression and impact of NHE1 protein inhibition with its inhibitor HOE642 on tumorigenesis and anti-PD1 therapy. Tumor angiogenesis, immunogenicity, and progression were assessed by immunofluorescence staining and flow cytometric profiling. Results Analysis of SLC9A1 mRNA expression in two data sets, CGGA and TCGA, reveals significantly higher SLC9A1 mRNA levels in higher grade gliomas. The SLC9A1 mRNA expression was especially enriched in isocitrate dehydrogenase (IDH)1/2 wild-type glioblastoma (GBM) and in mesenchymal glioma subtypes. Worsened survival probabilities were correlated with the elevated SLC9A1 mRNA levels in gliomas. The underlying mechanisms include promoting angiogenesis, and extracellular matrix remodeling. Increased SLC9A1 mRNA expression was also associated with tumor-associated macrophage accumulation. NHE1 inhibitor HOE642 reduced glioma volume, invasion, and prolonged overall survival in mouse glioma models. Blockade of NHE1 protein also stimulated immunogenic tumor microenvironment via activating CD8 T-cell accumulation, increasing expression of interferon-gamma (Ifng), and sensitized animals to anti-PD-1 therapy. Conclusion Our findings strongly suggest that NHE1 protein emerges as a marker for tumorigenesis and prognosis in glioma. Blocking NHE1 protein is a novel strategy for adjuvant anti-cancer therapies. Electronic supplementary material The online version of this article (10.1186/s13046-018-0923-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xiudong Guan
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No. 6 Tiantan Xili, Dongcheng District, Beijing, 100050, China.,Chinese National Clinical Research Center for Neurological Diseases, Beijing, China.,Beijing Neurosurgical Institute, Beijing, China.,Chinese Glioma Genome Atlas Network, Beijing, 100050, China.,Department of Neurology, University of Pittsburgh, 7016 Biomedical Science Tower 3 3501 Fifth Ave., Pittsburgh, PA, 15260, USA
| | - Lanxin Luo
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, China.,Department of Neurology, University of Pittsburgh, 7016 Biomedical Science Tower 3 3501 Fifth Ave., Pittsburgh, PA, 15260, USA
| | - Gulnaz Begum
- Department of Neurology, University of Pittsburgh, 7016 Biomedical Science Tower 3 3501 Fifth Ave., Pittsburgh, PA, 15260, USA.,Pittsburgh Institute for Neurodegenerative Disorders, University of Pittsburgh, Pittsburgh, PA, USA
| | - Gary Kohanbash
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Qingkun Song
- Department of Science and Technology, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Aparna Rao
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Nduka Amankulor
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Baoshan Sun
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, China.,Pólo Dois Portos, Instituto National de Investigação Agrária e Veterinária, I.P., Quinta da Almoinha, Dois Portos, Portugal
| | - Dandan Sun
- Department of Neurology, University of Pittsburgh, 7016 Biomedical Science Tower 3 3501 Fifth Ave., Pittsburgh, PA, 15260, USA. .,Pittsburgh Institute for Neurodegenerative Disorders, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Wang Jia
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No. 6 Tiantan Xili, Dongcheng District, Beijing, 100050, China. .,Chinese National Clinical Research Center for Neurological Diseases, Beijing, China. .,Beijing Neurosurgical Institute, Beijing, China. .,Chinese Glioma Genome Atlas Network, Beijing, 100050, China.
| |
Collapse
|
43
|
Phosphorylation of NHERF1 S279 and S301 differentially regulates breast cancer cell phenotype and metastatic organotropism. Biochim Biophys Acta Mol Basis Dis 2018; 1865:26-37. [PMID: 30326259 DOI: 10.1016/j.bbadis.2018.10.017] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 09/21/2018] [Accepted: 10/11/2018] [Indexed: 02/08/2023]
Abstract
Metastatic cancer cells are highly plastic for the expression of different tumor phenotype hallmarks and organotropism. This plasticity is highly regulated but the dynamics of the signaling processes orchestrating the shift from one cell phenotype and metastatic organ pattern to another are still largely unknown. The scaffolding protein NHERF1 has been shown to regulate the expression of different neoplastic phenotypes through its PDZ domains, which forms the mechanistic basis for metastatic organotropism. This reprogramming activity was postulated to be dependent on its differential phosphorylation patterns. Here, we show that NHERF1 phosphorylation on S279/S301 dictates several tumor phenotypes such as in vivo invasion, NHE1-mediated matrix digestion, growth and vasculogenic mimicry. Remarkably, injecting mice with cells having differential NHERF1 expression and phosphorylation drove a shift from the predominantly lung colonization (WT NHERF1) to predominately bone colonization (double S279A/S301A mutant), indicating that NHERF1 phosphorylation also acts as a signaling switch in metastatic organotropism.
Collapse
|
44
|
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: 12.0] [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
|