Skip to main content

OPINION article

Front. Physiol., 07 September 2021
Sec. Membrane Physiology and Membrane Biophysics
This article is part of the Research Topic ASICs: Structure, Function, and Pharmacology, Volume I View all 11 articles

Acid-Sensing Ion Channel 1 Contributes to Weak Acid-Induced Migration of Human Malignant Glioma Cells

\nSareena ShahSareena Shah1Yuyang ChuYuyang Chu2Victoria CegielskiVictoria Cegielski1Xiang-Ping Chu
Xiang-Ping Chu1*
  • 1Department of Biomedical Sciences, School of Medicine, University of Missouri-Kansas City, Kansas City, MO, United States
  • 2Feinberg School of Medicine, Northwestern University, Chicago, IL, United States

Introduction

Glioblastoma multiform (GBM), also referred to as glioblastoma, is the most common malignant tumor in the brain, known for its resistance to therapeutic agents and poor prognosis (Shergalis et al., 2018). Recent studies have seen success of some target compounds in combating GBMs in laboratory settings; however, these compounds failed in clinical trials (Mandel et al., 2018). Multiple factors may have contributed to this drug failure in GBM treatment, including difficulty penetrating the blood-brain barrier (BBB), an immunosuppressive microenvironment, and complex intratumoral heterogeneity (Khaddour et al., 2020; Ou et al., 2020). Difficulty in treating GBMs has led to a 5-year survival rate of <7.2% in humans. This is the lowest long-term rate of malignant brain tumors (Ostrom et al., 2020). Current treatment options for patients with GBM include surgical resection and a combination of chemotherapy, radiotherapy, and immunotherapy (Carlsson et al., 2014; Suter et al., 2020; Wen et al., 2020; Medikonda et al., 2021). Surgical resection is the most effective way to increase short-term survival rate, with research demonstrating a higher 1-year survival rate in patients with at least a 90% surgical resection compared to those without one (Medikonda et al., 2021). Yet, the long-term survival rate remains the same, suggesting that tumor recurrence is very likely (Yu et al., 2021).

GBMs, like many other tumors, have an acidic microenvironment due to the tumor's high rate of metabolism and limited blood supply (Tian et al., 2017). Acidosis may result from an accumulation of acid or an increase of partial pressure of carbon dioxide in tissues, which can cause lactic acid build-up in the brain (Chesler, 2003). Excessive lactic acid in the brain can damage metabolic functionality, hindering recirculation and reoxygenation throughout the body (Siesjö, 1982; Siesj et al., 1993; Rehncrona, 2005). Studies have demonstrated that acidosis can activate a family of ligand-gated ion channels: acid-sensing ion channels (ASICs), which are widely expressed in neurons (Waldmann et al., 1997; Xiong et al., 2004; Krishtal, 2015). ASICs are formed through different combinations of subunits, either homotrimeric or heterotrimeric, constituting different electrophysiological and pharmacological properties (Gründer and Pusch, 2015; Vullo and Kellenberger, 2020). The activation of ASICs induces neuronal depolarization and generates action potentials mostly due to the influx of Na+ ions (Jiang et al., 2009; Boscardin et al., 2016). ASICs play critical roles in several physiological processes including synaptic plasticity, pain sensation, and fear conditioning (Wemmie et al., 2013; Huang et al., 2015; Uchitel et al., 2019; Storozhuka et al., 2021). They also contribute to several neurodegenerative disorders such as Parkinson's disease and multiple sclerosis (Chu and Xiong, 2012; Chu et al., 2014; Ortega-Ramírez et al., 2017). Additionally, ASICs are expressed in glial cells (Lin et al., 2010), particularly ASIC1, whose activation in glial cell lines has been linked to migration and proliferation of the GBMs (Berdiev et al., 2003; Bubien et al., 2004; Kapoor et al., 2009, 2011; Rooj et al., 2012). Glioma tumors aggressively destroy surrounding tissues while growing in the brain, displaying acidosis and resulting in pathophysiological consequences (Honasoge and Sontheimer, 2013). ASIC expression in glioma cells generates a Na+ current which contributes to its volume and migration (Rooj et al., 2012). Active conductance has been observed in gliomas due to the presence of ASICs and epithelial sodium channels, which can impact cell cycle progression (Kapoor et al., 2009). The impact of ASICs, particularly ASIC1, in glial cell lines is being further investigated in efforts to reduce glial cell migration, study the potential proliferation effects, and to understand how ASIC1 inhibitors can regulate apoptosis of glioma cells (Sun et al., 2013; Tian et al., 2017). ASIC1 gene expression in glioma cells can predict outcomes of carcinomas as they act as genomic biomarkers; therefore, finding more inhibitors to ASICs may provide potential therapeutic agents to inhibit glioma cell activity (Sun et al., 2013; Bychkov et al., 2020; Griffin et al., 2020).

ASIC1 Contributes to Weak Acid-Induced Migration of Human Malignant Cells

A recent study from Dr. Xiong's laboratory published in the American Journal of Cancer Research described a combination of wet-lab techniques, including whole-cell patch-clamp recording, Western blotting, cell viability assay, wound-healing assay and trans-well migration assay, that were used to determine if the activation of ASIC1 by a weak acid (e.g., pH 7.0) had a correlation with the migration and proliferation of glial cell lines U87MG and A172 (Sheng et al., 2021). The study determined the expression of ASIC1 in glioblastoma cell lines, along with the induction of transient inward currents when the pH was dropped from 7.4 to 6.0, suggesting the existence of acid-activated currents in glioblastoma cell lines. When PcTx1, a selective ASIC1 inhibitor, was introduced, there was a decrease in acid-activated currents, indicating that ASIC1 was involved. The study also examined cell viability of A172 and U87MG cells when treated under different pH conditions (e.g., pH 7.4 and pH 7.0) to determine if weak acidosis could affect proliferation of the cells; however, they found that neither acidosis nor the addition of PcTx1 had an influence on cell viability of the two glioma cells, suggesting that weak acidosis has no effect on proliferation of human glioma cells. Using a wound healing assay, they investigated whether weak acid could promote migration of A172 and U87MG cells and found that weak acid did increase the migration rate by 26% in A172 cells and 67% in U87MG cells, respectively. When PcTx1 was introduced, it inhibited both glioma cells from migrating. Further investigation of this migration was conducted using a trans-well migration assay and revealed similar results. They also found that there was a heavier expression of the ASIC1 protein in U87MG cells compared to the expression in A172 cells, indicating that different cell lines of GBMs have different levels of ASIC1 expression. The study also introduced ASIC1-siRNA to U87MG cells, which silences ASIC1 expression, to better understand the influence of this protein on migration induced by treatment of weak acidosis. When compared to the control group, application of ASIC1-siRNA decreased the expression of the ASIC1 protein and did not affect the proliferation of U87MG cells, but it reduced the migration of U87MG cells under weak acidic conditions. Collectively, the results demonstrate that activation of ASIC1 by weak acidosis in glioma cells A172 and U87MG promotes migration of the cells but not proliferation, while PcTx1 inhibits migration of glioma cells. These findings suggest that ASIC1 could serve as a potential therapeutic target for GBMs.

Perspective

Although there are several studies of ASICs in the pathophysiology of glioma cells (Berdiev et al., 2003; Bubien et al., 2004; Kapoor et al., 2009, 2011; Rooj et al., 2012), this study uniquely dives deeper into the connection between weak acidosis activated ASIC1 and its effect on the migration of glioma cells (Sheng et al., 2021). A recent study from Dr. Grunder's group reported that glioblastoma stem cell (GSC) lines (R8 and R54) express functional ASIC1 and ASIC3, and their data suggest that expression of ASICs is associated with an improved survival in GSC lines (Tian et al., 2017). This result is inconsistent with other studies (Kapoor et al., 2009; Sheng et al., 2021). One possibility might be due to different glioblastoma cell lines used in different studies, suggesting the complexity of GBM. Therefore, additional studies might be necessary to further delineate the precise role of ASIC1 in high-grade glioblastoma. This study solely focuses on glial cells U87MG and A172, so it would be beneficial to utilize the same wet-lab techniques for cultured human glioma cells directly from the patients and see if parallel correlations are found. It would also be interesting to test PcTx1 on cultured human glioma cells to see whether it has any effects on glioma growth during a small pH drop (pH 7.0). In most whole-cell recordings, ASICs deactivate fast in response to brief pH stimulation (Gründer and Pusch, 2015). Even a small decrease in pH leads to profound steady state desensitization of ASICs. Therefore, it is intriguing how migration of high-grade gliomas could be promoted via ASIC continuously subjected to a slightly acidic pH, and the exact mechanism should be explored in the near future. Interestingly, a recent study demonstrates that the deactivation of ASIC1a is steeply dependent on the pH, spanning nearly three orders of magnitude from extremely fast (<1 ms) at pH 8.0 to very slow (more than 300 ms) at pH 7.0 (MacLean and Jayaraman, 2017). In addition, like most of ligand-gated ion channels, the desensitization of ASIC is also pH-dependent, with much slower desensitization occurs in response to a smaller pH drop (e.g., to 7.0) than a bigger pH drop (e.g., to 6.0). Although the current amplitude might be small with a small pH drop (Sheng et al., 2021), the current may last for a long period of time. Another possibility is that, in intact cells where cellular components are not washed out like in whole-cell recordings, the kinetics of ASIC1a current could be completely different. Additionally, the study from Dr. Xiong's group (Sheng et al., 2021) focuses on glial cells U87MG and A172 being introduced to a weak acid, but it would be insightful to compare the migration of these glial cell lines when introduced to a larger pH drop (e.g., pH 6.0). Further investigation may determine the consequences of cell migration under permanent inhibition of ASIC1 in glioma cells. Several existing studies have used PcTx1 or Mambalgin-2 as an ASIC1 inhibitor (Kellenberger and Schild, 2015; Dibas et al., 2019). However, it is necessary to explore other therapeutic agents since PcTx1 and Mambalgin-2 cannot be used in clinical settings due to their inability to cross the BBB (Sun et al., 2013; Bychkov et al., 2020). Therefore, determining effective delivery pathways for ASIC1 inhibitors is needed for the clinical treatment of GBMs. From a more malignant standpoint, late-stage glioma cells express high levels of ASIC1a, which is not portrayed in normal astrocytes. Thus, learning more about how and why ASIC1 expression changes in higher-grade gliomas may be vital in determining optimal treatment plans for patients with gliomas (Sun et al., 2013; Wang et al., 2015).

Author Contributions

All authors listed have made a substantial, direct and intellectual contribution to the work and approved it for publication.

Funding

The work was supported by grant from American Heart Association (19AIREA34470007) to X-PC.

Conflict of Interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher's Note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

Acknowledgments

We thank Dr. Zhi-Gang Xiong from Morehouse School of Medicine for his critical reading of this manuscript.

References

Berdiev, B. K., Xia, J., McLean, L. A., Markert, J. M., Gillespie, G. Y., Mapstone, T. B., et al. (2003). Acid-sensing ion channels in malignant gliomas. J. Biol. Chem. 278, 15023–15034. doi: 10.1074/jbc.M300991200

PubMed Abstract | CrossRef Full Text | Google Scholar

Boscardin, E., Alijevic, O., Hummler, E., Frateschi, S., and Kellenberger, S. (2016). The function and regulation of acid-sensing ion channels (ASICs) and the epithelial Na+ channel (ENaC): IUPHAR Review 19. Br. J. Pharmacol. 173, 2671–2701. doi: 10.1111/bph.13533

PubMed Abstract | CrossRef Full Text | Google Scholar

Bubien, J. K., Ji, H. L., Gillespie, G. Y., Fuller, C. M., Markert, J. M., Mapstone, T. B., et al. (2004). Cation selectivity and inhibition of malignant glioma Na+ channels by Psalmotoxin 1. Am. J. Physiol,. Cell Physiol. 287, C1282–1291. doi: 10.1152/ajpcell.00077.2004

PubMed Abstract | CrossRef Full Text | Google Scholar

Bychkov, M., Shulepko, M., Osmakov, D., Andreev, Y., Sudarikova, A., Vasileva, V., et al. (2020). Mambalgin-2 induces cell cycle arrest and apoptosis in glioma cells via interaction with ASIC1a. Cancers 12:1837. doi: 10.3390/cancers12071837

PubMed Abstract | CrossRef Full Text | Google Scholar

Carlsson, S. K., Brothers, S. P., and Wahlestedt, C. (2014). Emerging treatment strategies for glioblastoma multiforme. EMBO Mol. Med. 6, 1359–1370. doi: 10.15252/emmm.201302627

PubMed Abstract | CrossRef Full Text | Google Scholar

Chesler, M. (2003). Regulation and modulation of pH in the brain. Physiol. Rev. 83, 1183–1221. doi: 10.1152/physrev.00010.2003

PubMed Abstract | CrossRef Full Text | Google Scholar

Chu, X.-P., Grasing, K. A., and Wang, J. Q. (2014). Acid-sensing ion channels contribute to neurotoxicity. Transl. Stroke Res. 5, 69–78. doi: 10.1007/s12975-013-0305-y

PubMed Abstract | CrossRef Full Text | Google Scholar

Chu, X.-P., and Xiong, Z.-G. (2012). Physiological and pathological functions of acid-sensing ion channels in the central nervous system. Curr. Drug Targets. 13, 263–271. doi: 10.2174/138945012799201685

PubMed Abstract | CrossRef Full Text | Google Scholar

Dibas, J., Al-Saad, H., and Dibas, A. (2019). Basics on the use of acid-sensing ion channels' inhibitors as therapeutics. Neural Regen. Res. 14, 395–398. doi: 10.4103/1673-5374.245466

PubMed Abstract | CrossRef Full Text | Google Scholar

Griffin, M., Khan, R., Basu, S., and Smith, S. (2020). Ion channels as therapeutic targets in high grade gliomas. Cancers 12:3068. doi: 10.3390/cancers12103068

PubMed Abstract | CrossRef Full Text | Google Scholar

Gründer, S., and Pusch, M. (2015). Biophysical properties of acid-sensing ion channels (ASICs). Neuropharmacology 94, 9–18. doi: 10.1016/j.neuropharm.2014.12.016

PubMed Abstract | CrossRef Full Text | Google Scholar

Honasoge, A., and Sontheimer, H. (2013). Involvement of tumor acidification in brain cancer pathophysiology. Front. Physiol. 4:316. doi: 10.3389/fphys.2013.00316

PubMed Abstract | CrossRef Full Text | Google Scholar

Huang, Y., Jiang, N., Ji, Y., Xiong, Z., and Zha, X. (2015). Two aspects of ASIC function: synaptic plasticity and neuronal injury. Neuropharmacology 94, 42–48. doi: 10.1016/j.neuropharm.2014.12.010

PubMed Abstract | CrossRef Full Text | Google Scholar

Jiang, Q., Li, M. H., Papasian, C. J., Branigan, D., Xiong, Z. G., Wang, J. Q., et al. (2009). Characterization of acid-sensing ion channels in medium spiny neurons of mouse striatum. Neuroscience 162, 55–66. doi: 10.1016/j.neuroscience.2009.04.029

PubMed Abstract | CrossRef Full Text | Google Scholar

Kapoor, N., Bartoszewski, R., Qadri, Y. J., Bebok, Z., Bubien, J. K., Fuller, C. M., et al. (2009). Knockdown of ASIC1 and epithelial sodium channel subunits inhibits glioblastoma whole cell current and cell migration. J. Biol. Chem. 284, 24526–24541. doi: 10.1074/jbc.M109.037390

PubMed Abstract | CrossRef Full Text | Google Scholar

Kapoor, N., Lee, W., Clark, E., Bartoszewski, R., McNicholas, C. M., Latham, C. B., et al. (2011). Interaction of ASIC1 and ENaC subunits in human glioma cells and rat astrocytes. Am. J. Physiol,. Cell Physiol. 300, C1246–1259. doi: 10.1152/ajpcell.00199.2010

PubMed Abstract | CrossRef Full Text | Google Scholar

Kellenberger, S., and Schild, L. (2015). International union of basic and clinical pharmacology. XCI. structure, function, and pharmacology of acid-sensing ion channels and the epithelial Na+ channel. Pharmacol. Rev. 67, 1–35. doi: 10.1124/pr.114.009225

PubMed Abstract | CrossRef Full Text | Google Scholar

Khaddour, K., Johanns, T. M., and Ansstas, G. (2020). The landscape of novel therapeutics and challenges in glioblastoma multiforme: contemporary state and future directions. Pharmaceuticals 13:389. doi: 10.3390/ph13110389

PubMed Abstract | CrossRef Full Text | Google Scholar

Krishtal, O. (2015). Receptor for protons: First observations on acid sensing ion channels. Neuropharmacology 94, 4–8. doi: 10.1016/j.neuropharm.2014.12.014

PubMed Abstract | CrossRef Full Text | Google Scholar

Lin, Y. C., Liu, Y. C., Huang, Y. Y., and Lien, C. C. (2010). High-density expression of Ca2+–permeable ASIC1a channels in NG2 glia of rat hippocampus. PLoS ONE 5:e12665. doi: 10.1371/journal.pone.0012665

PubMed Abstract | CrossRef Full Text | Google Scholar

MacLean, D. M., and Jayaraman, V. (2017). Deactivation kinetics of acid-sensing ion channel 1a are strongly pH-sensitive. Proc. Natl. Acad. Sci. USA. 114, E2504–E2513. doi: 10.1073/pnas.1620508114

PubMed Abstract | CrossRef Full Text | Google Scholar

Mandel, J. J., Youssef, M., Ludmir, E., Yust-Katz, S., Patel, A. J., and De Groot, J. F. (2018). Highlighting the need for reliable clinical trials in glioblastoma. Expert Rev. Anticancer Ther. 18, 1031–1040. doi: 10.1080/14737140.2018.1496824

PubMed Abstract | CrossRef Full Text | Google Scholar

Medikonda, R., Dunn, G., Rahman, M., Fecci, P., and Lim, M. (2021). A review of glioblastoma immunotherapy. J. Neurooncol. 151, 41–53. doi: 10.1007/s11060-020-03448-1

PubMed Abstract | CrossRef Full Text | Google Scholar

Ortega-Ramírez, A., Vega, R., and Soto, E. (2017). Acid-sensing ion channels as potential therapeutic targets in neurodegeneration and neuroinflammation. Mediators Inflamm. 2017:3728096. doi: 10.1155/2017/3728096

PubMed Abstract | CrossRef Full Text | Google Scholar

Ostrom, Q. T., Patil, N., Cioffi, G., Waite, K., Kruchko, C., and Barnholtz-Sloan, J. S. (2020). CBTRUS statistical report: primary brain and other central nervous system tumors diagnosed in the United States in 2013-2017. Neuro Oncol. 22, iv1–iv96. doi: 10.1093/neuonc/noaa200

PubMed Abstract | CrossRef Full Text | Google Scholar

Ou, A., Yung, W. K. A., and Majd, N. (2020). Molecular mechanisms of treatment resistance in glioblastoma. Int. J. Mol. Sci. 22:351. doi: 10.3390/ijms22010351

PubMed Abstract | CrossRef Full Text | Google Scholar

Rehncrona, S. (2005). Brain acidosis. Ann. Emerg. Med. 14, 770–776. doi: 10.1016/S0196-0644(85)80055-X

PubMed Abstract | CrossRef Full Text | Google Scholar

Rooj, A. K., McNicholas, C. M., Bartoszewski, R., Bebok, Z., Benos, D. J., and Fuller, C. M. (2012). Glioma-specific cation conductance regulates migration and cell cycle progression. J. Biol. Chem. 287, 4053–4065. doi: 10.1074/jbc.M111.311688

PubMed Abstract | CrossRef Full Text | Google Scholar

Sheng, Y., Wu, B., Leng, T., Zhu, L., and Xiong, Z. (2021). Acid-sensing ion channel 1 (ASIC1) mediates weak acid-induced migration of human malignant glioma cells. Am. J. Cancer Res. 11, 997–1008.

PubMed Abstract | Google Scholar

Shergalis, A., Bankhead, A. I. I. I., Luesakul, U., Muangsin, N., and Neamati, N. (2018). Current challenges and opportunities in treating glioblastoma. Pharmacol. Rev. 70, 412–445. doi: 10.1124/pr.117.014944

PubMed Abstract | CrossRef Full Text | Google Scholar

Siesj,ö, B. K., Katsura, K., Mellergård, P., Ekholm, A., Lundgren, J., and Smith, M. L. (1993). Acidosis-related brain damage. Prog. Brain Res. 96, 23–48. doi: 10.1016/S0079-6123(08)63257-4

CrossRef Full Text | Google Scholar

Siesjö, B. K. (1982). Lactic acidosis in the brain: occurrence, triggering mechanisms and pathophysiological importance. Ciba Found. Symp.87, 77–100. doi: 10.1002/9780470720691.ch5

PubMed Abstract | CrossRef Full Text | Google Scholar

Storozhuka, M., Cherninskyia, A., Maximyuka, O., Isaeva, D., and Krishtala, O. (2021). Acid-sensing ion channels: focus on physiological and some pathological roles in the brain. Curr. Neuropharmacol. doi: 10.2174/1570159X19666210125151824. [Epub ahead of print].

PubMed Abstract | CrossRef Full Text | Google Scholar

Sun, X., Zhao, D., Li, Y., Sun, Y., Lei, X., Zhang, J., et al. (2013). Regulation of ASIC1 by Ca2+/calmodulin-dependent protein kinase II in human glioblastoma multiforme. Oncol. Rep. 30, 2852–2858. doi: 10.3892/or.2013.2777

PubMed Abstract | CrossRef Full Text | Google Scholar

Suter, R. K., Rodriguez-Blanco, J., and Ayad, N. G. (2020). Epigenetic pathways and plasticity in brain tumors. Neurobiol. Dis.145:105060. doi: 10.1016/j.nbd.2020.105060

PubMed Abstract | CrossRef Full Text | Google Scholar

Tian, Y., Bresenitz, P., Reska, A., Moussaoui, L. E., Beier, C. P., and Gründer, S. (2017). Glioblastoma cancer stem cell lines express functional acid sensing ion channels ASIC1a and ASIC3. Sci. Rep. 7:13674. doi: 10.1038/s41598-017-13666-9

PubMed Abstract | CrossRef Full Text | Google Scholar

Uchitel, O. D., González Inchauspe, C., and Weissmann, C. (2019). Synaptic signals mediated by protons and acid-sensing ion channels. Synapse 73:e22120. doi: 10.1002/syn.22120

PubMed Abstract | CrossRef Full Text | Google Scholar

Vullo, S., and Kellenberger, S. (2020). A molecular view of the function and pharmacology of acid-sensing ion channels. Pharmacol. Res. 154:104166. doi: 10.1016/j.phrs.2019.02.005

PubMed Abstract | CrossRef Full Text | Google Scholar

Waldmann, R., Champigny, G., Bassilana, F., Heurteaux, C., and Lazdunski, M. (1997). A proton-gated cation channel involved in acid-sensing. Nature 386, 173–177. doi: 10.1038/386173a0

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, R., Gurguis, C., Gu, W., Ko, E. A., Lim, I., Bang, H., et al. (2015). Ion channel gene expression predicts survival in glioma patients. Sci. Rep. 5:11593. doi: 10.1038/srep11593

PubMed Abstract | CrossRef Full Text | Google Scholar

Wemmie, J. A., Taugher, R. J., and Kreple, C. J. (2013). Acid-sensing ion channels in pain and disease. Nat. Rev. Neurosci. 14, 461–471. doi: 10.1038/nrn3529

PubMed Abstract | CrossRef Full Text | Google Scholar

Wen, P. Y., Weller, M., Lee, E. Q., Alexander, B. M., Barnholtz-Sloan, J. S., Barthel, F. P., et al. (2020). Glioblastoma in adults: a Society for Neuro-Oncology (SNO) and European Society of Neuro-Oncology (EANO) consensus review on current management and future directions. Neuro-oncology. 22, 1073–1113. doi: 10.1093/neuonc/noaa106

PubMed Abstract | CrossRef Full Text | Google Scholar

Xiong, Z. G., Zhu, X. M., Chu, X. P., Minami, M., Hey, J., Wei, W. L., et al. (2004). Neuroprotection in ischemia: blocking calcium-permeable acid-sensing ion channels. Cell 118, 687–698. doi: 10.1016/j.cell.2004.08.026

PubMed Abstract | CrossRef Full Text | Google Scholar

Yu, L., Zhang, G., and Qi, S. (2021). Aggressive treatment in glioblastoma: what determines the survival of patients? J. Neurol. Surg. A Cent. Eur. Neurosurg. 82, 112–117. doi: 10.1055/s-0040-1713172

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: acid-sensing ion channel 1, human malignant glioma cells, migration, acidosis, patch-clamp recording

Citation: Shah S, Chu Y, Cegielski V and Chu X-P (2021) Acid-Sensing Ion Channel 1 Contributes to Weak Acid-Induced Migration of Human Malignant Glioma Cells. Front. Physiol. 12:734418. doi: 10.3389/fphys.2021.734418

Received: 01 July 2021; Accepted: 16 August 2021;
Published: 07 September 2021.

Edited by:

Osvaldo D. Uchitel, University of Buenos Aires, Argentina

Reviewed by:

Mingli Liu, Morehouse School of Medicine, United States

Copyright © 2021 Shah, Chu, Cegielski and Chu. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Xiang-Ping Chu, chux@umkc.edu

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.