1
|
Juknevičienė M, Balnytė I, Valančiūtė A, Alonso MM, Preikšaitis A, Sužiedėlis K, Stakišaitis D. Differential Impact of Valproic Acid on SLC5A8, SLC12A2, SLC12A5, CDH1, and CDH2 Expression in Adult Glioblastoma Cells. Biomedicines 2024; 12:1416. [PMID: 39061990 PMCID: PMC11274075 DOI: 10.3390/biomedicines12071416] [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: 03/16/2024] [Revised: 06/13/2024] [Accepted: 06/22/2024] [Indexed: 07/28/2024] Open
Abstract
Valproic acid (VPA) has anticancer, anti-inflammatory, and epigenetic effects. The study aimed to determine the expression of carcinogenesis-related SLC5A8, SLC12A2, SLC12A5, CDH1, and CDH2 in adult glioblastoma U87 MG and T98G cells and the effects of 0.5 mM, 0.75 mM, and 1.5 mM doses of VPA. RNA gene expression was determined by RT-PCR. GAPDH was used as a control. U87 and T98G control cells do not express SLC5A8 or CDH1. SLC12A5 was expressed in U87 control but not in T98G control cells. The SLC12A2 expression in the U87 control was significantly lower than in the T98G control. T98G control cells showed significantly higher CDH2 expression than U87 control cells. VPA treatment did not affect SLC12A2 expression in U87 cells, whereas treatment dose-dependently increased SLC12A2 expression in T98G cells. Treatment with 1.5 mM VPA induced SLC5A8 expression in U87 cells, while treatment of T98G cells with VPA did not affect SLC5A8 expression. Treatment of U87 cells with VPA significantly increased SLC12A5 expression. VPA increases CDH1 expression depending on the VPA dose. CDH2 expression was significantly increased only in the U87 1.5 mM VPA group. Tested VPA doses significantly increased CDH2 expression in T98G cells. When approaching treatment tactics, assessing the cell's sensitivity to the agent is essential.
Collapse
Affiliation(s)
- Milda Juknevičienė
- Department of Histology and Embryology, Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania; (M.J.); (I.B.); (A.V.)
| | - Ingrida Balnytė
- Department of Histology and Embryology, Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania; (M.J.); (I.B.); (A.V.)
| | - Angelija Valančiūtė
- Department of Histology and Embryology, Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania; (M.J.); (I.B.); (A.V.)
| | - Marta Marija Alonso
- Department of Pediatrics, Clínica Universidad de Navarra, University of Navarra, 31008 Pamplona, Spain;
| | - Aidanas Preikšaitis
- Centre of Neurosurgery, Clinic of Neurology and Neurosurgery, Faculty of Medicine, Vilnius University, 03101 Vilnius, Lithuania;
| | - Kęstutis Sužiedėlis
- Laboratory of Molecular Oncology, National Cancer Institute, 08660 Vilnius, Lithuania;
| | - Donatas Stakišaitis
- Department of Histology and Embryology, Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania; (M.J.); (I.B.); (A.V.)
- Laboratory of Molecular Oncology, National Cancer Institute, 08660 Vilnius, Lithuania;
| |
Collapse
|
2
|
Zheng G, Chattopadhyay S, Sundquist J, Sundquist K, Ji J. Antihypertensive drug targets and breast cancer risk: a two-sample Mendelian randomization study. Eur J Epidemiol 2024; 39:535-548. [PMID: 38396187 PMCID: PMC11219410 DOI: 10.1007/s10654-024-01103-x] [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: 07/12/2023] [Accepted: 01/15/2024] [Indexed: 02/25/2024]
Abstract
Findings on the correlation between the use of antihypertensive medication and the risk of breast cancer (BC) have been inconsistent. We performed a two-sample Mendelian randomization (MR) using instrumental variables to proxy changes in gene expressions of antihypertensive medication targets to interrogate this. Genetic instruments for expression of antihypertensive drug target genes were identified with expression quantitative trait loci in blood, which should be associated with systolic blood pressure to proxy for the effect of antihypertensive drug. The association between genetic variants and BC risk were obtained from genome-wide association study summary statistics. The summary-based MR was employed to estimate the drug effects on BC risk. We further performed sensitivity analyses to confirm the discovered MR associations such as assessment of horizontal pleiotropy, colocalization, and multiple tissue enrichment analyses. The overall BC risk was only associated with SLC12A2 gene expression at a Bonferroni-corrected threshold. One standard deviation (SD) decrease of SLC12A2 gene expression in blood was associated with a decrease of 1.12 (95%CI, 0.80-1.58) mmHg of systolic blood pressure, but a 16% increased BC risk (odds ratio, 1.16, 95% confidential interval, 1.06-1.28). This signal was further observed for estrogen receptor positive (ER +) BC (1.17, 1.06-1.28). In addition, one SD decrease in expression of PDE1B in blood was associated with 7% decreased risk of ER + BC (0.93, 0.90-0.97). We detected no evidence of horizontal pleiotropy for these associations and the probability of the causal variants being shared between the gene expression and BC risk was 81.5, 40.5 and 66.8%, respectively. No significant association was observed between other target gene expressions and BC risk. Changes in expression of SLC12A2 and PDE1B mediated possibly via antihypertensive drugs may result in increased and decreased BC risk, respectively.
Collapse
Affiliation(s)
- Guoqiao Zheng
- Center for Primary Health Care Research, Lund University/Region Skåne, Jan Waldenströms Gata 35, 205 02, Malmö, Sweden.
| | - Subhayan Chattopadhyay
- Division of Clinical Genetics, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Jan Sundquist
- Center for Primary Health Care Research, Lund University/Region Skåne, Jan Waldenströms Gata 35, 205 02, Malmö, Sweden
- Department of Family Medicine and Community Health, Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Community-Based Healthcare Research and Education (CoHRE), Department of Functional Pathology, School of Medicine, Shimane University, Matsue, Japan
| | - Kristina Sundquist
- Center for Primary Health Care Research, Lund University/Region Skåne, Jan Waldenströms Gata 35, 205 02, Malmö, Sweden
- Department of Family Medicine and Community Health, Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Community-Based Healthcare Research and Education (CoHRE), Department of Functional Pathology, School of Medicine, Shimane University, Matsue, Japan
| | - Jianguang Ji
- Center for Primary Health Care Research, Lund University/Region Skåne, Jan Waldenströms Gata 35, 205 02, Malmö, Sweden.
| |
Collapse
|
3
|
Anagnostakis F, Kokkorakis M, Markouli M, Piperi C. Impact of Solute Carrier Transporters in Glioma Pathology: A Comprehensive Review. Int J Mol Sci 2023; 24:ijms24119393. [PMID: 37298344 DOI: 10.3390/ijms24119393] [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: 04/20/2023] [Revised: 05/25/2023] [Accepted: 05/26/2023] [Indexed: 06/12/2023] Open
Abstract
Solute carriers (SLCs) are essential for brain physiology and homeostasis due to their role in transporting necessary substances across cell membranes. There is an increasing need to further unravel their pathophysiological implications since they have been proposed to play a pivotal role in brain tumor development, progression, and the formation of the tumor microenvironment (TME) through the upregulation and downregulation of various amino acid transporters. Due to their implication in malignancy and tumor progression, SLCs are currently positioned at the center of novel pharmacological targeting strategies and drug development. In this review, we discuss the key structural and functional characteristics of the main SLC family members involved in glioma pathogenesis, along with their potential targeting options to provide new opportunities for CNS drug design and more effective glioma management.
Collapse
Affiliation(s)
- Filippos Anagnostakis
- Department of Medical and Surgical Sciences, University of Bologna, 40126 Bologna, Italy
| | - Michail Kokkorakis
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, 9700 RB Groningen, The Netherlands
| | - Mariam Markouli
- Department of Medicine, Boston Medical Center, Boston University School of Medicine, Boston, MA 02118, USA
| | - Christina Piperi
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| |
Collapse
|
4
|
Xiu M, Li L, Li Y, Gao Y. An update regarding the role of WNK kinases in cancer. Cell Death Dis 2022; 13:795. [PMID: 36123332 PMCID: PMC9485243 DOI: 10.1038/s41419-022-05249-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 09/07/2022] [Accepted: 09/08/2022] [Indexed: 01/23/2023]
Abstract
Mammalian WNK kinases (WNKs) are serine/threonine kinases that contain four members, WNK1-4. They function to maintain ion homeostasis and regulate blood pressure in mammals. Recent studies have revealed that the dysregulation of WNKs contributes to tumor growth, metastasis, and angiogenesis through complex mechanisms, especially through phosphorylating kinase substrates SPS1-related proline/alanine-rich kinase (SPAK) and oxidative stress-responsive kinase 1 (OSR1). Here, we review and discuss the relationships between WNKs and several key factors/biological processes in cancer, including ion channels, cation chloride cotransporters, sodium bicarbonate cotransporters, signaling pathways, angiogenesis, autophagy, and non-coding RNAs. In addition, the potential drugs for targeting WNK-SPAK/OSR1 signaling have also been discussed. This review summarizes and discusses knowledge of the roles of WNKs in cancer, which provides a comprehensive reference for future studies.
Collapse
Affiliation(s)
- Mengxi Xiu
- grid.24516.340000000123704535Department of Oncology, Shanghai East Hospital, Tongji University School of Medicine, 200120 Shanghai, China
| | - Li Li
- grid.24516.340000000123704535Department of Oncology, Shanghai East Hospital, Tongji University School of Medicine, 200120 Shanghai, China
| | - Yandong Li
- grid.24516.340000000123704535Department of Oncology, Shanghai East Hospital, Tongji University School of Medicine, 200120 Shanghai, China
| | - Yong Gao
- grid.24516.340000000123704535Department of Oncology, Shanghai East Hospital, Tongji University School of Medicine, 200120 Shanghai, China
| |
Collapse
|
5
|
Hou CY, Ma CY, Yuh CH. WNK1 kinase signaling in metastasis and angiogenesis. Cell Signal 2022; 96:110371. [DOI: 10.1016/j.cellsig.2022.110371] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Revised: 05/24/2022] [Accepted: 05/25/2022] [Indexed: 02/06/2023]
|
6
|
Damanskienė E, Balnytė I, Valančiūtė A, Alonso MM, Stakišaitis D. Different Effects of Valproic Acid on SLC12A2, SLC12A5 and SLC5A8 Gene Expression in Pediatric Glioblastoma Cells as an Approach to Personalised Therapy. Biomedicines 2022; 10:968. [PMID: 35625705 PMCID: PMC9138981 DOI: 10.3390/biomedicines10050968] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Revised: 04/19/2022] [Accepted: 04/20/2022] [Indexed: 02/04/2023] Open
Abstract
Valproic acid (VPA) is a histone deacetylase inhibitor with sex-specific immunomodulatory and anticancer effects. This study aimed to investigate the effect of 0.5 and 0.75 mM VPA on NKCC1 (SLC12A2), KCC2 (SLC12A5) and SLC5A8 (SLC5A8) co-transporter gene expressions in pediatric PBT24 (boy's) and SF8628 (girl's) glioblastoma cells. The SLC12A2, SLC12A5 and SLC5A8 RNA expressions were determined by the RT-PCR method. The SLC12A2 and SLC5A8 expressions did not differ between the PBT24 and SF8628 controls. The SLC12A5 expression in the PBT24 control was significantly higher than in the SF8628 control. VPA treatment significantly increased the expression of SLC12A2 in PBT24 but did not affect SF8628 cells. VPA increased the SLC12A5 expression in PBT24 and SF8628 cells. The SLC12A5 expression of the PBT24-treated cells was significantly higher than in corresponding SF8628 groups. Both VPA doses increased the SLC5A8 expression in PBT24 and SF8628 cells, but the expression was significantly higher in the PBT24-treated, compared to the respective SF8628 groups. The SLC5A8 expression in PBT24-treated cells was 10-fold higher than in SF8628 cells. The distinct effects of VPA on the expression of SLC12A2, SLC12A5 and SLC5A8 in PBT24 and SF8628 glioblastoma cells suggest differences in tumor cell biology that may be gender-related.
Collapse
Affiliation(s)
- Eligija Damanskienė
- Department of Histology and Embryology, Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania; (E.D.); (I.B.); (A.V.)
| | - Ingrida Balnytė
- Department of Histology and Embryology, Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania; (E.D.); (I.B.); (A.V.)
| | - Angelija Valančiūtė
- Department of Histology and Embryology, Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania; (E.D.); (I.B.); (A.V.)
| | - Marta Marija Alonso
- Department of Pediatrics, Clínica Universidad de Navarra, University of Navarra, 31008 Pamplona, Spain;
| | - Donatas Stakišaitis
- Department of Histology and Embryology, Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania; (E.D.); (I.B.); (A.V.)
- Laboratory of Molecular Oncology, National Cancer Institute, 08660 Vilnius, Lithuania
| |
Collapse
|
7
|
Zhong L, Yang P, Zhang C, Wang Z, Jiang T, Chen B, Shan X, Qiu X. Long-term adjuvant administration of temozolomide impacts serum ions concentration in high-grade glioma. Chin Neurosurg J 2022; 8:6. [PMID: 35216639 PMCID: PMC8876447 DOI: 10.1186/s41016-022-00271-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Accepted: 01/27/2022] [Indexed: 11/10/2022] Open
Abstract
Background Adjuvant temozolomide (TMZ) chemotherapy with standard regimen remarkably improves survival in patients with high-grade glioma (HGG). However, the influence of long-term TMZ chemotherapy on serum ions concentration is unclear. Methods One hundred and thirty-eight patients with HGG were included. Their blood samples were collected for blood biochemistry and routine test. The alteration in serum ions concentration, total protein, albumin, globin, and blood cells counts were used to identify the impact of long-term TMZ chemotherapy. Results Through the comparation of quantitative value of diverse parameters among different chemotherapy cycles, we identified that serum potassium concentration had a downward trend after TMZ administration (1st vs. 6th, p < 0.001; 1st vs. 12th, p < 0.001). Additionally, the correlation analysis showed that platelets was negatively correlated with chemotherapy cycles (r = − 0.649, p = 0.023). The hematological adverse events mainly centered on grade 1 to 2. Conclusion Long-term administration of TMZ may lead to serum ions disturbance. Besides the myelosuppression, we should pay attention to the alteration in serum ions concentration, and give patients proper symptomatic treatment when necessary. Supplementary Information The online version contains supplementary material available at 10.1186/s41016-022-00271-7.
Collapse
Affiliation(s)
- Liyun Zhong
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Pei Yang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Chinese Glioma Genome Atlas Network (CGGA) and Asian Glioma Genome Atlas Network (AGGA), Beijing, China
| | - Chuanbao Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Chinese Glioma Genome Atlas Network (CGGA) and Asian Glioma Genome Atlas Network (AGGA), Beijing, China
| | - Zheng Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Chinese Glioma Genome Atlas Network (CGGA) and Asian Glioma Genome Atlas Network (AGGA), Beijing, China
| | - Tao Jiang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Chinese Glioma Genome Atlas Network (CGGA) and Asian Glioma Genome Atlas Network (AGGA), Beijing, China.,Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, China.,China National Clinical Research Center for Neurological Diseases, 119 South 4th Ring Road West, Beijing, 100070, China
| | - Baoshi Chen
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Xia Shan
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China. .,Chinese Glioma Genome Atlas Network (CGGA) and Asian Glioma Genome Atlas Network (AGGA), Beijing, China. .,Department of Radiotherapy, Beijing Tiantan Hospital, Capital Medical University, 119 South 4th Ring Road West, Beijing, 100070, China.
| | - Xiaoguang Qiu
- Department of Radiotherapy, Beijing Tiantan Hospital, Capital Medical University, 119 South 4th Ring Road West, Beijing, 100070, China.
| |
Collapse
|
8
|
Damanskienė E, Balnytė I, Valančiūtė A, Alonso MM, Preikšaitis A, Stakišaitis D. The Different Temozolomide Effects on Tumorigenesis Mechanisms of Pediatric Glioblastoma PBT24 and SF8628 Cell Tumor in CAM Model and on Cells In Vitro. Int J Mol Sci 2022; 23:ijms23042001. [PMID: 35216113 PMCID: PMC8877228 DOI: 10.3390/ijms23042001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 02/04/2022] [Accepted: 02/06/2022] [Indexed: 02/05/2023] Open
Abstract
It is necessary to elucidate the individual effects of temozolomide (TMZ) on carcinogenesis and tumor resistance to chemotherapy mechanisms. The study aimed to investigate the TMZ 50 and 100 μM dose effect difference between PBT24 and SF8628 cell line high-grade pediatric glioblastoma (phGBM) xenografts in a chicken chorioallantoic membrane (CAM) model, on PCNA and EZH2 immunohistochemical expression in the tumor and on the expression of NKCC1, KCC2, E- and N-cadherin genes in TMZ-treated and control cell groups in vitro. TMZ at a 100 μg dose reduced the incidence of PBT24 xenograft invasion into the CAM, CAM thickening and the number of blood vessels in the CAM (p < 0.05), but did not affect the SF8628 tumor in the CAM model. The TMZ impact on PBT24 and SF8628 tumor PCNA expression was similarly significantly effective but did not alter EZH2 expression in the studied tumors. The TMZ at 50 μM caused significantly increased RNA expression of the NKCC1 gene in both studied cell types compared with controls (p < 0.05). The expression of the KCC2 gene was increased in PBT24 TMZ-treated cells (p < 0.05), and no TMZ effect was found in SF8628-treated cells. The study supports the suggestion that individual sensitivity to TMZ should be assessed when starting treatment.
Collapse
Affiliation(s)
- Eligija Damanskienė
- Department of Histology and Embryology, Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania; (I.B.); (A.V.)
- Correspondence: (E.D.); (D.S.)
| | - Ingrida Balnytė
- Department of Histology and Embryology, Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania; (I.B.); (A.V.)
| | - Angelija Valančiūtė
- Department of Histology and Embryology, Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania; (I.B.); (A.V.)
| | - Marta Maria Alonso
- Department of Pediatrics, Clínica Universidad de Navarra, University of Navarra, 31008 Pamplona, Spain;
| | - Aidanas Preikšaitis
- Centre of Neurosurgery, Clinic of Neurology and Neurosurgery, Faculty of Medicine, Vilnius University, 03101 Vilnius, Lithuania;
| | - Donatas Stakišaitis
- Department of Histology and Embryology, Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania; (I.B.); (A.V.)
- Laboratory of Molecular Oncology, National Cancer Institute, 08660 Vilnius, Lithuania
- Correspondence: (E.D.); (D.S.)
| |
Collapse
|
9
|
Wang Y, Wu B, Long S, QiangLiu, Li G. WNK3 promotes the invasiveness of glioma cell lines under hypoxia by inducing the epithelial-to-mesenchymal transition. Transl Neurosci 2021; 12:320-329. [PMID: 34513083 PMCID: PMC8389507 DOI: 10.1515/tnsci-2020-0180] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Revised: 07/27/2021] [Accepted: 07/28/2021] [Indexed: 12/21/2022] Open
Abstract
Background The primary features of malignant glioma include high rates of mortality and recurrence, uncontrollable invasiveness, strong angiogenesis, and widespread hypoxia. The hypoxic microenvironment is an important factor affecting the malignant progression of glioma. However, the molecular mechanisms underlying glioma adaption in hypoxic microenvironments are poorly understood. Objective The work presented in this paper focuses on the role of WNK3 gene in glioma invasion under hypoxic conditions. Furthermore, we aim to explore its role in epithelial-to-mesenchymal transition (EMT). Methods ShRNA targeting WNK3 transfection was used to knockdown the WNK3 expression in U87 cells. We used western blot analysis to detect the relative expression of proteins in U87 cells. The effect of WNK3 on cell migration was explored using a transwell assay in the U87 cell line. We also evaluated WNK3 expression levels in glioma samples by immunohistochemistry analysis. Results WNK3 expression was significantly higher in high-grade (III and IV) gliomas than in low-grade (I and II) gliomas. WNK3 expression was up-regulated in U87 cells when cultured in a hypoxic environment in addition; WNK3 knockdown inhibited the invasion of U87 glioma cells by regulating the EMT, especially under hypoxic conditions. Conclusion These findings suggested that WNK3 plays an important role in the hypoxic microenvironment of glioma and might also be a candidate for therapeutic application in the treatment of glioma.
Collapse
Affiliation(s)
- Yue Wang
- Department of Neurosurgery, Weifang People's Hospital, Weifang, China
| | - Bingbing Wu
- Department of Neurosurgery, First Affiliated Hospital of China Medical University, Shenyang 110001, China
| | - Shengrong Long
- Department of Neurosurgery, First Affiliated Hospital of China Medical University, Shenyang 110001, China
| | - QiangLiu
- Department of Neurosurgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, China
| | - Guangyu Li
- Department of Neurosurgery, First Affiliated Hospital of China Medical University, Shenyang 110001, China
| |
Collapse
|
10
|
Chang KF, Chang JT, Huang XF, Huang YC, Li CY, Weng JC, Hsiao CY, Hsu HJ, Tsai NM. Cedrus atlantica Extract Suppress Glioblastoma Growth through Promotion of Genotoxicity and Apoptosis: In Vitro and In Vivo Studies. Int J Med Sci 2021; 18:2417-2430. [PMID: 33967620 PMCID: PMC8100640 DOI: 10.7150/ijms.54468] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 04/07/2021] [Indexed: 01/31/2023] Open
Abstract
Glioblastoma (GBM) is the most common malignant primary brain tumor in humans, exhibiting highly infiltrative growth and drug resistance to conventional chemotherapy. Cedrus atlantica (CAt) extract has been shown to decrease postoperative pain and inhibit the growth of K562 leukemia cells. The aim of this study was to assess the anti-GBM activity and molecular mechanism of CAt extract in vitro and in vivo. The results showed that CAt extract greatly suppressed GBM cells both in vitro and in vivo and enhanced the survival rate in subcutaneous and orthotopic animal models. Moreover, CAt extract increased the level of ROS and induced DNA damage, resulting in cell cycle arrest at the G0/G1 phase and cell apoptosis. Western blotting results indicated that CAt extract regulates p53/p21 and CDK4/cyclin D1 protein expression and activates extrinsic and intrinsic apoptosis. Furthermore, CAt extract enhanced the cytotoxicity of Temozolomide and decreased AKT/mTOR signaling by combination treatment. In toxicity assays, CAt extract exhibited low cytotoxicity toward normal cells or organs in vitro and in vivo. CAt extract suppresses the growth of GBM by induction of genotoxicity and activation of apoptosis. The results of this study suggest that CAt extract can be developed as a therapeutic agent or adjuvant for GBM treatment in the future.
Collapse
Affiliation(s)
- Kai-Fu Chang
- Institute of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan, ROC.,Department of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung 40201, Taiwan, ROC
| | - Jinghua Tsai Chang
- Institute of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan, ROC
| | - Xiao-Fan Huang
- Institute of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan, ROC.,Department of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung 40201, Taiwan, ROC
| | - Ya-Chih Huang
- Institute of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan, ROC.,Department of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung 40201, Taiwan, ROC
| | - Chia-Yu Li
- Department of Life and Death, Nanhua University, Chiayi 62249, Taiwan, ROC
| | - Jun-Cheng Weng
- Department of Medical Imaging and Radiological Sciences, Chang Gung University, Taoyuan 33302, Taiwan, ROC
| | - Chih-Yen Hsiao
- Division of Nephrology, Department of Internal Medicine, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chia-Yi, 60002, Taiwan, ROC.,Department of Hospital and Health Care Administration, Chia Nan University of Pharmacy and Science, Tainan, 71710, Taiwan, ROC
| | - Hui-Ju Hsu
- Department of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung 40201, Taiwan, ROC.,Clinical Laboratory, Chung Shan Medical University Hospital, Taichung 40201, Taiwan, ROC
| | - Nu-Man Tsai
- Department of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung 40201, Taiwan, ROC.,Clinical Laboratory, Chung Shan Medical University Hospital, Taichung 40201, Taiwan, ROC
| |
Collapse
|
11
|
Takayasu T, Kurisu K, Esquenazi Y, Ballester LY. Ion Channels and Their Role in the Pathophysiology of Gliomas. Mol Cancer Ther 2020; 19:1959-1969. [PMID: 33008831 PMCID: PMC7577395 DOI: 10.1158/1535-7163.mct-19-0929] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 03/24/2020] [Accepted: 08/06/2020] [Indexed: 01/10/2023]
Abstract
Malignant gliomas are the most common primary central nervous system tumors and their prognosis is very poor. In recent years, ion channels have been demonstrated to play important roles in tumor pathophysiology such as regulation of gene expression, cell migration, and cell proliferation. In this review, we summarize the current knowledge on the role of ion channels on the development and progression of gliomas. Cell volume changes through the regulation of ion flux, accompanied by water flux, are essential for migration and invasion. Signaling pathways affected by ion channel activity play roles in cell survival and cell proliferation. Moreover, ion channels are involved in glioma-related seizures, sensitivity to chemotherapy, and tumor metabolism. Ion channels are potential targets for the treatment of these lethal tumors. Despite our increased understanding of the contributions of ion channels to glioma biology, this field remains poorly studied. This review summarizes the current literature on this important topic.
Collapse
Affiliation(s)
- Takeshi Takayasu
- Department of Pathology and Laboratory Medicine, The University of Texas Health Science Center at Houston, Houston, Texas
- Department of Neurosurgery, Institute of Biomedical and Health Sciences, Hiroshima University, Higashihiroshima, Hiroshima, Japan
| | - Kaoru Kurisu
- Department of Neurosurgery, Institute of Biomedical and Health Sciences, Hiroshima University, Higashihiroshima, Hiroshima, Japan
| | - Yoshua Esquenazi
- Vivian L. Smith Department of Neurosurgery, The University of Texas Health Science Center at Houston, Medical School, Houston, Texas.
- Memorial Hermann Hospital-TMC, Houston, Texas
| | - Leomar Y Ballester
- Department of Pathology and Laboratory Medicine, The University of Texas Health Science Center at Houston, Houston, Texas.
- Vivian L. Smith Department of Neurosurgery, The University of Texas Health Science Center at Houston, Medical School, Houston, Texas
- Memorial Hermann Hospital-TMC, Houston, Texas
| |
Collapse
|
12
|
Luo L, Wang J, Ding D, Hasan MN, Yang SS, Lin SH, Schreppel P, Sun B, Yin Y, Erker T, Sun D. Role of NKCC1 Activity in Glioma K + Homeostasis and Cell Growth: New Insights With the Bumetanide-Derivative STS66. Front Physiol 2020; 11:911. [PMID: 32848856 PMCID: PMC7413028 DOI: 10.3389/fphys.2020.00911] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 07/08/2020] [Indexed: 12/01/2022] Open
Abstract
Introduction: Na+-K+-2Cl− cotransporter isoform 1 (NKCC1) is important in regulating intracellular K+ and Cl− homeostasis and cell volume. In this study, we investigated a role of NKCC1 in regulating glioma K+ influx and proliferation in response to apoptosis inducing chemotherapeutic drug temozolomide (TMZ). The efficacy of a new bumetanide (BMT)-derivative NKCC1 inhibitor STS66 [3-(butylamino)-2-phenoxy-5-[(2, 2, 2-trifluoroethylamino) methyl] benzenesulfonamide] in blocking NKCC1 activity was compared with well-established NKCC1 inhibitor BMT. Methods: NKCC1 activity in cultured mouse GL26 and SB28-GFP glioma cells was measured by Rb+ (K+) influx. The WNK1-SPAK/OSR1-NKCC1 signaling and AKT/ERK-mTOR signaling protein expression and activation were assessed by immunoblotting. Cell growth was determined by bromodeoxyuridine (BrdU) incorporation assay, MTT proliferation assay, and cell cycle analysis. Impact of STS66 and BMT on cell Rb+ influx and growth was measured in glioma cells treated with or without TMZ. Results: Rb+ influx assay showed that 10 μM BMT markedly decreased the total Rb+ influx and no additional inhibition detected at >10 μM BMT. In contrast, the maximum effects of STS66 on Rb+ influx inhibition were at 40–60 μM. Both BMT and STS66 reduced TMZ-mediated NKCC1 activation and protein upregulation. Glioma cell growth can be reduced by STS66. The most robust inhibition of glioma growth, cell cycle, and AKT/ERK signaling was achieved by the TMZ + STS66 treatment. Conclusion: The new BMT-derivative NKCC1 inhibitor STS66 is more effective than BMT in reducing glioma cell growth in part by inhibiting NKCC1-mediated K+ influx. TMZ + STS66 combination treatment reduces glioma cell growth via inhibiting cell cycle and AKT-ERK signaling.
Collapse
Affiliation(s)
- Lanxin Luo
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, United States.,Department of Neurosurgery, University of Minnesota, Minneapolis, MN, United States
| | - Jun Wang
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, United States.,Department of Neurology, The Second Hospital of Dalian Medical University, Dalian, China
| | - Dawei Ding
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, United States.,Department of Neurosurgery, University of Minnesota, Minneapolis, MN, United States
| | - Md Nabiul Hasan
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, United States
| | - Sung-Sen Yang
- Division of Nephrology, Department of Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Shih-Hua Lin
- Division of Nephrology, Department of Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Philipp Schreppel
- Department of Medicinal Chemistry, University of Vienna, Vienna, Austria
| | - Baoshan Sun
- Pólo Dois Portos, Instituto National de Investigação Agrária e Veterinária, I.P., Quinta da Almoinha, Dois Portos, Portugal
| | - Yan Yin
- Department of Neurology, The Second Hospital of Dalian Medical University, Dalian, China
| | - Thomas Erker
- Department of Medicinal Chemistry, University of Vienna, Vienna, Austria
| | - Dandan Sun
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, United States.,Veterans Affairs Pittsburgh Health Care System, Geriatric Research, Educational and Clinical Center, Pittsburgh, PA, United States
| |
Collapse
|
13
|
Luo L, Guan X, Begum G, Ding D, Gayden J, Hasan MN, Fiesler VM, Dodelson J, Kohanbash G, Hu B, Amankulor NM, Jia W, Castro MG, Sun B, Sun D. Blockade of Cell Volume Regulatory Protein NKCC1 Increases TMZ-Induced Glioma Apoptosis and Reduces Astrogliosis. Mol Cancer Ther 2020; 19:1550-1561. [PMID: 32393472 PMCID: PMC11792748 DOI: 10.1158/1535-7163.mct-19-0910] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 01/29/2020] [Accepted: 05/04/2020] [Indexed: 11/16/2022]
Abstract
Glioma is one of the most common primary malignant tumors of the central nervous system accounting for approximately 40% of all intracranial tumors. Temozolomide is a conventional chemotherapy drug for adjuvant treatment of patients with high-risk gliomas, including grade II to grade IV. Our bioinformatic analysis of The Cancer Genome Atlas and Chinese Glioma Genome Atlas datasets and immunoblotting assay show that SLC12A2 gene and its encoded Na+-K+-2Cl- cotransporter isoform 1 (NKCC1) protein are abundantly expressed in grade II-IV gliomas. NKCC1 regulates cell volume and intracellular Cl- concentration, which promotes glioma cell migration, resistance to temozolomide, and tumor-related epilepsy in experimental glioma models. Using mouse syngeneic glioma models with intracranial transplantation of two different glioma cell lines (GL26 and SB28), we show that NKCC1 protein in glioma tumor cells as well as in tumor-associated reactive astrocytes was significantly upregulated in response to temozolomide monotherapy. Combination therapy of temozolomide with the potent NKCC1 inhibitor bumetanide reduced tumor proliferation, potentiated the cytotoxic effects of temozolomide, decreased tumor-associated reactive astrogliosis, and restored astrocytic GLT-1 and GLAST glutamate transporter expression. The combinatorial therapy also led to suppressed tumor growth and prolonged survival of mice bearing GL26 glioma cells. Taken together, these results demonstrate that NKCC1 protein plays multifaceted roles in the pathogenesis of glioma tumors and presents as a therapeutic target for reducing temozolomide-mediated resistance and tumor-associated astrogliosis.
Collapse
Affiliation(s)
- Lanxin Luo
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, Liaoning, China
- School of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang, Liaoning, China
- Department of Neurology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Xiudong Guan
- Department of Neurology, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Chinese National Clinical Research Center for Neurological Diseases, Beijing, China
- Beijing Neurosurgical Institute, Beijing, China
- Chinese Glioma Genome Atlas Network, Beijing, China
| | - Gulnaz Begum
- Department of Neurology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Dawei Ding
- Department of Neurology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Jenesis Gayden
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Md Nabiul Hasan
- Department of Neurology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Victoria M Fiesler
- Department of Neurology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Jacob Dodelson
- Department of Neurology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Gary Kohanbash
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Baoli Hu
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Nduka M Amankulor
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Wang Jia
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Chinese National Clinical Research Center for Neurological Diseases, Beijing, China
- Beijing Neurosurgical Institute, Beijing, China
- Chinese Glioma Genome Atlas Network, Beijing, China
| | - Maria G Castro
- Department of Neurosurgery and Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Baoshan Sun
- School of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang, Liaoning, 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, Pittsburgh, Pennsylvania.
- Veterans Affairs Pittsburgh Health Care System, Geriatric Research, Educational and Clinical Center, Pittsburgh, Pennsylvania
| |
Collapse
|
14
|
Demian WL, Persaud A, Jiang C, Coyaud É, Liu S, Kapus A, Kafri R, Raught B, Rotin D. The Ion Transporter NKCC1 Links Cell Volume to Cell Mass Regulation by Suppressing mTORC1. Cell Rep 2020; 27:1886-1896.e6. [PMID: 31067471 DOI: 10.1016/j.celrep.2019.04.034] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 02/13/2019] [Accepted: 04/05/2019] [Indexed: 01/08/2023] Open
Abstract
mTORC1 regulates cellular growth and is activated by growth factors and by essential amino acids such as Leu. Leu enters cells via the Leu transporter LAT1-4F2hc (LAT1). Here we show that the Na+/K+/2Cl- cotransporter NKCC1 (SLC12A2), a known regulator of cell volume, is present in complex with LAT1. We further show that NKCC1 depletion or deletion enhances LAT1 activity, as well as activation of Akt and Erk, leading to activation of mTORC1 in cells, colonic organoids, and mouse colon. Moreover, NKCC1 depletion reduces intracellular Na+ concentration and cell volume (size) and mass and stimulates cell proliferation. NKCC1, therefore, suppresses mTORC1 by inhibiting its key activating signaling pathways. Importantly, by linking ion transport and cell volume regulation to mTORC1 function, NKCC1 provides a long-sought link connecting cell volume (size) to cell mass regulation.
Collapse
Affiliation(s)
- Wael L Demian
- Program in Cell Biology, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Biochemistry Department, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Avinash Persaud
- Program in Cell Biology, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Biochemistry Department, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Chong Jiang
- Program in Cell Biology, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Étienne Coyaud
- Princess Margaret Cancer Centre, University Health Network, Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 2C1, Canada
| | - Shixuan Liu
- Biochemistry Department, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Andras Kapus
- Biochemistry Department, University of Toronto, Toronto, ON M5S 1A8, Canada; St. Michael Hospital Research Institute, Toronto, ON M5B 1W8, Canada
| | - Ran Kafri
- Biochemistry Department, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Brian Raught
- Princess Margaret Cancer Centre, University Health Network, Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 2C1, Canada
| | - Daniela Rotin
- Program in Cell Biology, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Biochemistry Department, University of Toronto, Toronto, ON M5S 1A8, Canada.
| |
Collapse
|
15
|
Sun H, Long S, Wu B, Liu J, Li G. NKCC1 involvement in the epithelial-to-mesenchymal transition is a prognostic biomarker in gliomas. PeerJ 2020; 8:e8787. [PMID: 32211242 PMCID: PMC7081783 DOI: 10.7717/peerj.8787] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 02/23/2020] [Indexed: 12/16/2022] Open
Abstract
Background Gliomas are the most prevalent type of intracranial tumors. NKCC1 is an important regulator in tumor cell volume. We noticed that abnormally high NKCC1 expression resulted in changes in the shape and adhesion of glioma cells. However, little is known about the role of NKCC1 in the epithelial-mesenchymal transition (EMT) of gliomas. This study aims to clarify the biological function of NKCC1 in glioblastoma multiforme (GBM) progression. Methods Using data from The Cancer Genome Atlas (TCGA), we performed a Kaplan–Meier analysis on NKCC1 expression levels to estimate the rate of survival of mesenchymal GBM patients. The correlation between NKCC1 and EMT-related proteins was analyzed from the Gene Expression Profiling Interactive Analysis (GEPIA) server. We conducted Gene Set Enrichment Analysis (GSEA) to verify molecular signatures and pathways. We then studied the expression of NKCC1 in grade I–IV glioma tissue samples collected from patients using immunohistochemistry (IHC). Finally, we evaluated the effects of NKCC1 migration and invasion on the cellular behaviors of U251 cells using the transwell assay and western blots. Results High NKCC1 expression was associated with poor prognoses in mesenchymal GBM. Our results suggest a correlation between NKCC1 and EMT-protein markers: CDH2 and VIM. GSEA showed that gliomas, TGF-beta signaling and EMT were enriched in the NKCC1 high expression phenotype. Higher expression levels of NKCC1 in gliomas correlate with higher glioma grades. Transwell assay and western blot results demonstrated that the knockdown of NKCC1 led to a reduction in migration and invasion, while also inhibiting MMP-2 and MMP-9 expression in U251. Conclusion These results suggest that high expression of NKCC1 regulates EMT in gliomas, providing a new therapeutic strategy for addressing the spread of gliomas by inhibiting the spread of intracranial tumors.
Collapse
Affiliation(s)
- Huaiyu Sun
- Department of Neurosurgery, First Hospital of China Medical University, Shenyang, China
| | - Shengrong Long
- Department of Neurosurgery, First Hospital of China Medical University, Shenyang, China
| | - Bingbing Wu
- Department of Neurosurgery, First Hospital of China Medical University, Shenyang, China
| | - Jia Liu
- Department of Neurosurgery, First Hospital of China Medical University, Shenyang, China
| | - Guangyu Li
- Department of Neurosurgery, First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
16
|
Pu W, Qiu J, Nassar ZD, Shaw PN, McMahon KA, Ferguson C, Parton RG, Riggins GJ, Harris JM, Parat MO. A role for caveola-forming proteins caveolin-1 and CAVIN1 in the pro-invasive response of glioblastoma to osmotic and hydrostatic pressure. J Cell Mol Med 2020; 24:3724-3738. [PMID: 32065471 PMCID: PMC7131935 DOI: 10.1111/jcmm.15076] [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: 11/14/2019] [Revised: 01/28/2020] [Accepted: 01/29/2020] [Indexed: 12/27/2022] Open
Abstract
In solid tumours, elevated interstitial fluid pressure (osmotic and hydrostatic pressure) is a barrier to drug delivery and correlates with poor prognosis. Glioblastoma (GBM) further experience compressive force when growing within a space limited by the skull. Caveolae are proposed to play mechanosensing roles, and caveola‐forming proteins are overexpressed in GBM. We asked whether caveolae mediate the GBM response to osmotic pressure. We evaluated in vitro the influence of spontaneous or experimental down‐regulation of caveola‐forming proteins (caveolin‐1, CAVIN1) on the proteolytic profile and invasiveness of GBM cells in response to osmotic pressure. In response to osmotic pressure, GBM cell lines expressing caveola‐forming proteins up‐regulated plasminogen activator (uPA) and/or matrix metalloproteinases (MMPs), some EMT markers and increased their in vitro invasion potential. Down‐regulation of caveola‐forming proteins impaired this response and prevented hyperosmolarity‐induced mRNA expression of the water channel aquaporin 1. CRISPR ablation of caveola‐forming proteins further lowered expression of matrix proteases and EMT markers in response to hydrostatic pressure, as a model of mechanical force. GBM respond to pressure by increasing matrix‐degrading enzyme production, mesenchymal phenotype and invasion. Caveola‐forming proteins mediate, at least in part, the pro‐invasive response of GBM to pressure. This may represent a novel target in GBM treatment.
Collapse
Affiliation(s)
- Wenjun Pu
- School of Pharmacy, The University of Queensland, Brisbane, Queensland, Australia
| | - Jiawen Qiu
- School of Pharmacy, The University of Queensland, Brisbane, Queensland, Australia
| | - Zeyad D Nassar
- School of Medicine and Freemasons Foundation Centre for Men's Health, South Australian Health and Medical Research Institute, University of Adelaide, Adelaide, South Australia, Australia
| | - Paul N Shaw
- School of Pharmacy, The University of Queensland, Brisbane, Queensland, Australia
| | - Kerrie-Ann McMahon
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Charles Ferguson
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Robert G Parton
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia.,Centre for Microscopy and Microanalysis, The University of Queensland, Brisbane, Queensland, Australia
| | - Gregory J Riggins
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jonathan M Harris
- Institute of Health Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Marie-Odile Parat
- School of Pharmacy, The University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|
17
|
Guan X, Hasan MN, Begum G, Kohanbash G, Carney KE, Pigott VM, Persson AI, Castro MG, Jia W, Sun D. Blockade of Na/H exchanger stimulates glioma tumor immunogenicity and enhances combinatorial TMZ and anti-PD-1 therapy. Cell Death Dis 2018; 9:1010. [PMID: 30262908 PMCID: PMC6160445 DOI: 10.1038/s41419-018-1062-3] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 08/21/2018] [Accepted: 09/10/2018] [Indexed: 12/28/2022]
Abstract
The weak immunogenicity of gliomas presents a barrier for effective immunotherapy. Na/H exchanger isoform 1 (NHE1) maintains alkaline intracellular pH (pHi) of glioma cells and acidic microenvironment. In addition, NHE1 is expressed in tumor-associated microglia and tumor-associated macrophages (TAMs) and involved in protumoral communications between glioma and TAMs. Therefore, we hypothesize that NHE1 plays a role in developing tumor resistance and immunosuppressive tumor microenvironment. In this study, we investigated the efficacy of pharmacological inhibition of NHE1 on combinatorial therapies. Here we show that temozolomide (TMZ) treatment stimulates NHE1 protein expression in two intracranial syngeneic mouse glioma models (SB28, GL26). Pharmacological inhibition of NHE1 potentiated the cytotoxic effects of TMZ, leading to reduced tumor growth and increased median survival of mice. Blockade of NHE1 stimulated proinflammatory activation of TAM and increased cytotoxic T cell infiltration into tumors. Combining TMZ, anti-PD-1 antibody treatment with NHE1 blockade significantly prolonged the median survival in the mouse glioma model. These results demonstrate that pharmacological inhibition of NHE1 protein presents a new strategy for potentiating TMZ-induced cytotoxicity and increasing tumor immunogenicity for immunotherapy to improve glioma therapy.
Collapse
Affiliation(s)
- Xiudong Guan
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
- Chinese National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Md Nabiul Hasan
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Gulnaz Begum
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Gary Kohanbash
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Karen E Carney
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Victoria M Pigott
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Anders I Persson
- Department of Neurology, University of California, San Francisco, CA, USA
- Department of Neurological Surgery, University of California, San Francisco, CA, USA
| | - Maria G Castro
- Department of Neurological Surgery, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Wang Jia
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
- Chinese National Clinical Research Center for Neurological Diseases, Beijing, China.
- Beijing Neurosurgical Institute, Beijing, China.
| | - Dandan Sun
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
18
|
Chemo-resistance of A172 glioblastoma cells is controlled by miR-1271-regulated Bcl-2. Biomed Pharmacother 2018; 108:734-740. [PMID: 30248541 DOI: 10.1016/j.biopha.2018.08.102] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 07/21/2018] [Accepted: 08/17/2018] [Indexed: 11/21/2022] Open
Abstract
MicroRNAs (miRNAs) have been reported to exert important effects on the initiation, progression and metastasis of glioblastoma multiforme (GBM). In this study, we aimed to explore the regulation role of miR-1271 on the development of GBM. We found that miR-1271 was a Bcl-2-targeting miRNA, and the levels of miR-1271was decreased in samples from patients with GBM, compared with those from corresponding normal tissue samples. On the other hand, the levels of miR-1271 were inversely related to the levels of Bcl-2, which have been significantly increased in GBM samples. The overall survival was poorer in patients with low levels of miR-1271, compared to those with high levels of miR-1271. In vitro, the chemo-resistant cell survival mediated with Bcl-2 was inhibited by overexpression of miR-1271 and was enhanced by depletion of miR-1271. Thus, the chemo-resistance of GBM cells may be promoted after suppressing miR-1271 through cell survival mediated with Bcl-2. The prognosis of patients with GBM receiving chemotherapy may be improved by overexpressing miR-1271 in cancerous cells.
Collapse
|
19
|
Ma H, Li T, Tao Z, Hai L, Tong L, Yi L, Abeysekera IR, Liu P, Xie Y, Li J, Yuan F, Zhang C, Yang Y, Ming H, Yu S, Yang X. NKCC1 promotes EMT-like process in GBM via RhoA and Rac1 signaling pathways. J Cell Physiol 2018; 234:1630-1642. [PMID: 30159893 PMCID: PMC6282979 DOI: 10.1002/jcp.27033] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 06/22/2018] [Indexed: 12/30/2022]
Abstract
Glioblastoma is the most common and lethal primary intracranial tumor. As the key regulator of tumor cell volume, sodium‐potassium‐chloride cotransporter 1 (NKCC1) expression increases along with the malignancy of the glioma, and NKCC1 has been implicated in glioblastoma invasion. However, little is known about the role of NKCC1 in the epithelial‐mesenchymal transition‐like process in gliomas. We noticed that aberrantly elevated expression of NKCC1 leads to changes in the shape, polarity, and adhesion of cells in glioma. Here, we investigated whether NKCC1 promotes an epithelial–mesenchymal transition (EMT)‐like process in gliomas via the RhoA and Rac1 signaling pathways. Pharmacological inhibition and knockdown of NKCC1 both decrease the expressions of mesenchymal markers, such as N‐cadherin, vimentin, and snail, whereas these treatments increase the expression of the epithelial marker E‐cadherin. These findings indicate that NKCC1 promotes an EMT‐like process in gliomas. The underlying mechanism is the facilitation of the binding of Rac1 and RhoA to GTP by NKCC1, which results in a significant enhancement of the EMT‐like process. Specific inhibition or knockdown of NKCC1 both attenuate activated Rac1 and RhoA, and the pharmacological inhibitions of Rac1 and RhoA both impair the invasion and migration abilities of gliomas. Furthermore, we illustrated that NKCC1 knockdown abolished the dissemination and spread of glioma cells in a nude mouse intracranial model. These findings suggest that elevated NKCC1 activity acts in the regulation of an EMT‐like process in gliomas, and thus provides a novel therapeutic strategy for targeting the invasiveness of gliomas, which might help to inhibit the spread of malignant intracranial tumors.
Collapse
Affiliation(s)
- Haiwen Ma
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China.,Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Tianjin, China.,Key Laboratory of Post-trauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin, China.,Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, China
| | - Tao Li
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China.,Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Tianjin, China.,Key Laboratory of Post-trauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin, China.,Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, China
| | - Zhennan Tao
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China.,Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Tianjin, China.,Key Laboratory of Post-trauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin, China.,Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, China
| | - Long Hai
- Department of Radiation Oncology, Henan Cancer Hospital, The Affiliated Cancer Hospital of Zhengzhou University, Henan, China
| | - Luqing Tong
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China.,Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Tianjin, China.,Key Laboratory of Post-trauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin, China.,Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, China
| | - Li Yi
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China.,Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Tianjin, China.,Key Laboratory of Post-trauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin, China.,Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, China
| | - Iruni R Abeysekera
- Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Peidong Liu
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China.,Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Tianjin, China
| | - Yang Xie
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China.,Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Tianjin, China
| | - Jiabo Li
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China.,Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Tianjin, China
| | - Feng Yuan
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China.,Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Tianjin, China
| | - Chen Zhang
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Yihan Yang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China.,Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Tianjin, China
| | - Haolang Ming
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China.,Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Tianjin, China
| | - Shengping Yu
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China.,Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Tianjin, China
| | - Xuejun Yang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China.,Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Tianjin, China.,Key Laboratory of Post-trauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin, China.,Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, China
| |
Collapse
|
20
|
Ilkhanizadeh S, Sabelström H, Miroshnikova YA, Frantz A, Zhu W, Idilli A, Lakins JN, Schmidt C, Quigley DA, Fenster T, Yuan E, Trzeciak JR, Saxena S, Lindberg OR, Mouw JK, Burdick JA, Magnitsky S, Berger MS, Phillips JJ, Arosio D, Sun D, Weaver VM, Weiss WA, Persson AI. Antisecretory Factor-Mediated Inhibition of Cell Volume Dynamics Produces Antitumor Activity in Glioblastoma. Mol Cancer Res 2018; 16:777-790. [PMID: 29431617 PMCID: PMC5932284 DOI: 10.1158/1541-7786.mcr-17-0413] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 12/13/2017] [Accepted: 01/24/2018] [Indexed: 12/31/2022]
Abstract
Interstitial fluid pressure (IFP) presents a barrier to drug uptake in solid tumors, including the aggressive primary brain tumor glioblastoma (GBM). It remains unclear how fluid dynamics impacts tumor progression and can be targeted therapeutically. To address this issue, a novel telemetry-based approach was developed to measure changes in IFP during progression of GBM xenografts. Antisecretory factor (AF) is an endogenous protein that displays antisecretory effects in animals and patients. Here, endogenous induction of AF protein or exogenous administration of AF peptide reduced IFP and increased drug uptake in GBM xenografts. AF inhibited cell volume regulation of GBM cells, an effect that was phenocopied in vitro by the sodium-potassium-chloride cotransporter 1 (SLC12A2/NKCC1) inhibitor bumetanide. As a result, AF induced apoptosis and increased survival in GBM models. In vitro, the ability of AF to reduce GBM cell proliferation was phenocopied by bumetanide and NKCC1 knockdown. Next, AF's ability to sensitize GBM cells to the alkylating agent temozolomide, standard of care in GBM patients, was evaluated. Importantly, combination of AF induction and temozolomide treatment blocked regrowth in GBM xenografts. Thus, AF-mediated inhibition of cell volume regulation represents a novel strategy to increase drug uptake and improve outcome in GBM. Mol Cancer Res; 16(5); 777-90. ©2018 AACR.
Collapse
Affiliation(s)
- Shirin Ilkhanizadeh
- Department of Neurology, University of California, San Francisco, San Francisco, California
- Brain Tumor Research Center (BTRC) at the Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California
| | - Hanna Sabelström
- Department of Neurology, University of California, San Francisco, San Francisco, California
- Sandler Neurosciences Center, University of California, San Francisco, San Francisco, California
| | | | - Aaron Frantz
- Department of Neurology, University of California, San Francisco, San Francisco, California
- Sandler Neurosciences Center, University of California, San Francisco, San Francisco, California
| | - Wen Zhu
- Department of Neurology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Aurora Idilli
- Institute of Biophysics, CNR and FBK, Trento, Italy
- CIBIO, University of Trento, Trento, Italy
| | - Jon N Lakins
- Department of Surgery, University of California, San Francisco, San Francisco, California
| | - Christin Schmidt
- Department of Neurology, University of California, San Francisco, San Francisco, California
- Sandler Neurosciences Center, University of California, San Francisco, San Francisco, California
| | - David A Quigley
- Brain Tumor Research Center (BTRC) at the Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, California
| | - Trenten Fenster
- Department of Neurology, University of California, San Francisco, San Francisco, California
- Sandler Neurosciences Center, University of California, San Francisco, San Francisco, California
| | - Edith Yuan
- Department of Neurology, University of California, San Francisco, San Francisco, California
- Sandler Neurosciences Center, University of California, San Francisco, San Francisco, California
| | - Jacqueline R Trzeciak
- Department of Neurology, University of California, San Francisco, San Francisco, California
- Sandler Neurosciences Center, University of California, San Francisco, San Francisco, California
| | - Supna Saxena
- Department of Neurology, University of California, San Francisco, San Francisco, California
- Sandler Neurosciences Center, University of California, San Francisco, San Francisco, California
| | - Olle R Lindberg
- Brain Tumor Research Center (BTRC) at the Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, California
- Department of Pathology, University of California, San Francisco, San Francisco, California
| | - Janna K Mouw
- Department of Surgery, University of California, San Francisco, San Francisco, California
| | - Jason A Burdick
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Sergey Magnitsky
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California
| | - Mitchel S Berger
- Brain Tumor Research Center (BTRC) at the Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, California
| | - Joanna J Phillips
- Brain Tumor Research Center (BTRC) at the Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, California
- Department of Pathology, University of California, San Francisco, San Francisco, California
| | - Daniele Arosio
- Institute of Biophysics, CNR and FBK, Trento, Italy
- CIBIO, University of Trento, Trento, Italy
| | - Dandan Sun
- Department of Neurology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Valerie M Weaver
- Department of Surgery, University of California, San Francisco, San Francisco, California
- Brain Tumor Research Center (BTRC) at the Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California
| | - William A Weiss
- Department of Neurology, University of California, San Francisco, San Francisco, California
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, California
- Brain Tumor Research Center (BTRC) at the Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California
| | - Anders I Persson
- Department of Neurology, University of California, San Francisco, San Francisco, California.
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, California
- Sandler Neurosciences Center, University of California, San Francisco, San Francisco, California
- Brain Tumor Research Center (BTRC) at the Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California
| |
Collapse
|
21
|
Mrowczynski OD, Bourcier AJ, Liao J, Langan ST, Specht CS, Rizk EB. The predictive potential of hyponatremia for glioblastoma patient survival. J Neurooncol 2018; 138:99-104. [DOI: 10.1007/s11060-018-2774-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 01/20/2018] [Indexed: 11/24/2022]
|
22
|
Reactive Astrocytes in Glioblastoma Multiforme. Mol Neurobiol 2018; 55:6927-6938. [PMID: 29363044 DOI: 10.1007/s12035-018-0880-8] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 01/07/2018] [Indexed: 12/17/2022]
Abstract
Despite the multidisciplinary integration in the therapeutic management of glioblastoma multiforme (GBM), the prognosis of GBM patients is poor. There is growing recognition that the cells in the tumor microenvironment play a vital role in regulating the progression of glioma. Astrocytes are an important component of the blood-brain barrier (BBB) as well as the tripartite synapse neural network to promote bidirectional communication with neurons under physiological conditions. Emerging evidence shows that tumor-associated reactive astrocytes interact with glioma cells and facilitate the progression, aggression, and survival of tumors by releasing different cytokines. Communication between reactive astrocytes and glioma cells is further promoted through ion channels and ion transporters, which augment the migratory capacity and invasiveness of tumor cells by modifying H+ and Ca2+ concentrations and stimulating volume changes in the cell. This in part contributes to the loss of epithelial polarization, initiating epithelial-mesenchymal transition. Therefore, this review will summarize the recent findings on the role of reactive astrocytes in the progression of GBM and in the development of treatment-resistant glioma. In addition, the involvement of ion channels and transporters in bridging the interactions between tumor cells and astrocytes and their potential as new therapeutic anti-tumor targets will be discussed.
Collapse
|
23
|
Li Y, Scott R, Hooper AR, Bartholomeusz GA, Kornienko A, Bills GF. Aspergillus candidus is a newly recognized source of sphaeropsidin A: Isolation, semi-synthetic derivatization and anticancer evaluation. Bioorg Med Chem Lett 2017; 27:5436-5440. [DOI: 10.1016/j.bmcl.2017.11.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Revised: 10/30/2017] [Accepted: 11/01/2017] [Indexed: 10/18/2022]
|
24
|
Molecular features and physiological roles of K +-Cl - cotransporter 4 (KCC4). Biochim Biophys Acta Gen Subj 2017; 1861:3154-3166. [PMID: 28935604 DOI: 10.1016/j.bbagen.2017.09.007] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 09/15/2017] [Indexed: 12/27/2022]
Abstract
A K+-Cl- cotransport system was documented for the first time during the mid-seventies in sheep and goat red blood cells. It was then described as a Na+-independent and ouabain-insensitive ion carrier that could be stimulated by cell swelling and N-ethylmaleimide (NEM), a thiol-reacting agent. Twenty years later, this system was found to be dispensed by four different isoforms in animal cells. The first one was identified in the expressed sequence tag (EST) database by Gillen et al. based on the assumption that it would be homologous to the Na+-dependent K+-Cl- cotransport system for which the molecular identity had already been uncovered. Not long after, the three other isoforms were once again identified in the EST databank. Among those, KCC4 has generated much interest a few years ago when it was shown to sustain distal renal acidification and hearing development in mouse. As will be seen in this review, many additional roles were ascribed to this isoform, in keeping with its wide distribution in animal species. However, some of them have still not been confirmed through animal models of gene inactivation or overexpression. Along the same line, considerable knowledge has been acquired on the mechanisms by which KCC4 is regulated and the environmental cues to which it is sensitive. Yet, it is inferred to some extent from historical views and extrapolations.
Collapse
|
25
|
Zhou Y, Sun W, Chen N, Xu C, Wang X, Dong K, Zhang B, Zhang J, Hao N, Sun A, Wei H, He F, Jiang Y. Discovery of NKCC1 as a potential therapeutic target to inhibit hepatocellular carcinoma cell growth and metastasis. Oncotarget 2017; 8:66328-66342. [PMID: 29029515 PMCID: PMC5630415 DOI: 10.18632/oncotarget.20240] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 07/11/2017] [Indexed: 01/20/2023] Open
Abstract
Metastasis is the essential cause for the high mortality of hepatocellular carcinoma (HCC). In order to investigate the mechanism of metastasis, and to discover therapeutic targets for HCC, the quantitative proteomic technique was applied to characterize the plasma membrane proteins of two HCC cell lines with low (MHCC97L) or high (MHCC97H) metastatic potentials. One of the plasma membrane proteins, sodium-potassium-chloride cotransporter 1 (NKCC1), was upregulated in MHCC97H cell line. Immunohistochemistry result in HCC patients showed that NKCC1 expression was associated with poor differentiation and microvascular invasion. Knockdown of NKCC1 via RNA interference reduced HCC cell proliferation and invasion abilities in vitro and in vivo, whereas over-expression of NKCC1 significantly increased HCC cell proliferation and invasion abilities in vitro and in vivo. Additionally, blocking NKCC1 activity with bumetanide attenuated the proliferation and invasion abilities of HCC cells in vitro and limited the HCC growth in vivo. Further results suggested that NKCC1 promotes the invasion ability via MMP-2 activity, and that the WNK1/OSR1/NKCC1 signal pathway might play roles in HCC metastasis. For the first time, our study demonstrated that NKCC1 plays a role in HCC metastasis, and could be served as a potential target to inhibit HCC cell growth and metastasis.
Collapse
Affiliation(s)
- Yaya Zhou
- State Key Laboratory of Proteomics, National Center for Protein Sciences Beijing, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing 102206, P. R. China
| | - Wei Sun
- State Key Laboratory of Proteomics, National Center for Protein Sciences Beijing, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing 102206, P. R. China
| | - Ning Chen
- State Key Laboratory of Proteomics, National Center for Protein Sciences Beijing, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing 102206, P. R. China
| | - Chen Xu
- State Key Laboratory of Proteomics, National Center for Protein Sciences Beijing, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing 102206, P. R. China
| | - Xinxin Wang
- Department of Pathology, Beijing Youan Hospital of Capital Medical University, Beijing 100069, P. R. China
| | - Kun Dong
- Department of Pathology, Beijing Youan Hospital of Capital Medical University, Beijing 100069, P. R. China
| | - Binxue Zhang
- State Key Laboratory of Proteomics, National Center for Protein Sciences Beijing, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing 102206, P. R. China
| | - Jian Zhang
- State Key Laboratory of Proteomics, National Center for Protein Sciences Beijing, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing 102206, P. R. China
| | - Ning Hao
- State Key Laboratory of Proteomics, National Center for Protein Sciences Beijing, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing 102206, P. R. China
| | - Aihua Sun
- State Key Laboratory of Proteomics, National Center for Protein Sciences Beijing, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing 102206, P. R. China
| | - Handong Wei
- State Key Laboratory of Proteomics, National Center for Protein Sciences Beijing, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing 102206, P. R. China
| | - Fuchu He
- State Key Laboratory of Proteomics, National Center for Protein Sciences Beijing, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing 102206, P. R. China.,Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, P. R. China
| | - Ying Jiang
- State Key Laboratory of Proteomics, National Center for Protein Sciences Beijing, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing 102206, P. R. China
| |
Collapse
|
26
|
Abstract
WNK kinases, along with their upstream regulators (CUL3/KLHL3) and downstream targets (the SPAK/OSR1 kinases and the cation-Cl- cotransporters [CCCs]), comprise a signaling cascade essential for ion homeostasis in the kidney and nervous system. Recent work has furthered our understanding of the WNKs in epithelial transport, cell volume homeostasis, and GABA signaling, and uncovered novel roles for this pathway in immune cell function and cell proliferation.
Collapse
Affiliation(s)
- Masoud Shekarabi
- Department of Neuroscience, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Jinwei Zhang
- Departments of Neurosurgery, Centers for Mendelian Genomics, Yale School of Medicine, New Haven, CT 06477, USA; MRC Protein Phosphorylation and Ubiquitylation Unit, College of Life Sciences, University of Dundee, Dundee DD1 5EH, Scotland, UK
| | - Arjun R Khanna
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Neurosurgery, Harvard Medical School, Boston, MA 02115, USA
| | - David H Ellison
- Division of Nephrology & Hypertension, Department of Medicine, Oregon Health & Science University, Portland, Oregon 97239, USA; VA Portland Health Care System, Portland, OR 97239, USA
| | - Eric Delpire
- Department of Anesthesiology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Kristopher T Kahle
- Departments of Neurosurgery, Pediatrics, and Cellular & Molecular Physiology, Centers for Mendelian Genomics, Yale School of Medicine, New Haven, CT 06477, USA.
| |
Collapse
|
27
|
Kim KH, Yeo SG, Yoo BC, Myung JK. Identification of calgranulin B interacting proteins and network analysis in gastrointestinal cancer cells. PLoS One 2017; 12:e0171232. [PMID: 28152021 PMCID: PMC5289589 DOI: 10.1371/journal.pone.0171232] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Accepted: 01/17/2017] [Indexed: 01/14/2023] Open
Abstract
Calgranulin B is known to be involved in tumor development, but the underlying molecular mechanism is not clear. To gain insight into possible roles of calgranulin B, we screened for calgranulin B-interacting molecules in the SNU-484 gastric cancer and the SNU-81 colon cancer cells. Calgranulin B-interacting partners were identified by yeast two-hybrid and functional information was obtained by computational analysis. Most of the calgranulin B-interacting partners were involved in metabolic and cellular processes, and found to have molecular function of binding and catalytic activities. Interestingly, 46 molecules in the network of the calgranulin B-interacting proteins are known to be associated with cancer and FKBP2 was found to interact with calgranulin B in both SNU-484 and SNU-81 cells. Polyubiquitin-C encoded by UBC, which exhibited an interaction with calgranulin B, has been associated with various molecules of the extracellular space and plasma membrane identified in our screening, including Na-K-Cl cotransporter 1 and dystonin in SNU-484 cells, and ATPase subunit beta-1 in SNU-81 cells. Our data provide novel insight into the roles of calgranulin B of gastrointestinal cancer cells, and offer new clues suggesting calgranulin B acts as an effector molecule through which the cell can communicate with the tumor microenvironment via polyubiquitin-C.
Collapse
Affiliation(s)
- Kyung-Hee Kim
- Omics Core Laboratory, Research Institute, National Cancer Center, Goyang-si, Gyeonggi-do, Republic of Korea
- Colorectal Cancer Branch, Research Institute, National Cancer Center, Goyang-si, Gyeonggi-do, Republic of Korea
| | - Seung-Gu Yeo
- Department of Radiation Oncology, Soonchunhyang University College of Medicine, Cheonan, Chungnam, Republic of Korea
| | - Byong Chul Yoo
- Colorectal Cancer Branch, Research Institute, National Cancer Center, Goyang-si, Gyeonggi-do, Republic of Korea
| | - Jae Kyung Myung
- Department of System Cancer Science, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang-si, Gyeonggi-do, Republic of Korea
| |
Collapse
|
28
|
Zhang Z, Zhou Q, Miao Y, Tian H, Li Y, Feng X, Song X. MiR-429 induces apoptosis of glioblastoma cell through Bcl-2. Tumour Biol 2016; 37:15607–15613. [PMID: 26511969 DOI: 10.1007/s13277-015-4291-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Accepted: 10/19/2015] [Indexed: 01/04/2023] Open
Abstract
An essential role of microRNAs (miRNAs) has been acknowledged in the tumorigenesis of glioblastoma multiforme (GBM). Very recently, miR-429 was reported to have a potential of suppressing cancer growth. However, whether miR-429 may similarly regulate growth of GBM remains unknown. Here, we analyzed the levels of miR-429 and anti-apoptotic protein Bcl-2 in GBM specimens. We combined bioinformatics analyses and luciferase reporter assay to determine the relationship between miR-429 and Bcl-2 in GBM cells. Cell survival upon temozolomide treatment was analyzed in a CCK assay. Cell apoptosis was measured by fluorescein isothiocyanate (FITC) Annexin V apoptosis detection assay. We found that miR-429 levels were significantly decreased and Bcl-2 levels were significantly increased in GBM specimens, compared to the paired adjacent non-tumor brain tissue. Moreover, the levels of miR-429 and Bcl-2 inversely correlated. Low-miR-429 subjects had an overall inferior survival, compared to high-miR-429 subjects. MiR-429 targeted the 3'-UTR of Bcl-2 mRNA to inhibit its translation. Overexpression of miR-429 inhibited Bcl-2-mediated cell survival against temozolomide-induced apoptosis, while depletion of miR-429 augmented it. Together, our data suggest that miR-429 suppression in GBM promotes Bcl-2-mediated cancer cell survival against chemotherapy-induced cell death. Re-expression of miR-429 levels in GBM cells may improve the outcome of chemotherapy.
Collapse
Affiliation(s)
- Zhenxing Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Liaoning Medical University, Jinzhou, 121001, China
| | - Qingqing Zhou
- Department of Neurosurgery, The First Affiliated Hospital of Liaoning Medical University, Jinzhou, 121001, China
| | - Ye Miao
- Department of Neurosurgery, The First Affiliated Hospital of Liaoning Medical University, Jinzhou, 121001, China
| | - He Tian
- Department of Histology and Embryology, Liaoning Medical University, 3-40 Songpo Road, Jinzhou, 121001, China
| | - Yang Li
- Department of Neurosurgery, The First Affiliated Hospital of Liaoning Medical University, Jinzhou, 121001, China
| | - Xu Feng
- Department of Neurosurgery, The First Affiliated Hospital of Liaoning Medical University, Jinzhou, 121001, China
| | - Xiaofeng Song
- Department of Histology and Embryology, Liaoning Medical University, 3-40 Songpo Road, Jinzhou, 121001, China.
| |
Collapse
|
29
|
Cong D, Zhu W, Kuo JS, Hu S, Sun D. Ion transporters in brain tumors. Curr Med Chem 2016; 22:1171-81. [PMID: 25620102 DOI: 10.2174/0929867322666150114151946] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Revised: 01/05/2015] [Accepted: 01/08/2015] [Indexed: 11/22/2022]
Abstract
Ion transporters are important in regulation of ionic homeostasis, cell volume, and cellular signal transduction under physiological conditions. They have recently emerged as important players in cancer progression. In this review, we discussed two important ion transporter proteins, sodium-potassium-chloride cotransporter isoform 1 (NKCC-1) and sodium-hydrogen exchanger isoform 1 (NHE-1) in Glioblastoma multiforme (GBM) and other malignant tumors. NKCC-1 is a Na(+)- dependent Cl(-) transporter that mediates the movement of Na(+), K(+), and Cl(-) ions across the plasma membrane and maintains cell volume and intracellular K(+) and Cl(-) homeostasis. NHE-1 is a ubiquitously expressed cell membrane protein which regulates intracellular pH (pH(i)) and extracellular pH (pH(e)) homeostasis and cell volume. Here, we summarized recent pre-clinical experimental studies on NKCC-1 and NHE-1 in GBM and other malignant tumors, such as breast cancer, hepatocellular carcinoma, and lung cancer cells. These studies illustrated that pharmacological inhibition or down-regulation of these ion transporter proteins reduces proliferation, increases apoptosis, and suppresses migration and invasion of cancer cells. These new findings reveal the potentials of these ion transporters as new targets for cancer diagnosis and/or treatment.
Collapse
Affiliation(s)
| | | | | | | | - Dandan Sun
- Department of Neurology, University of Pittsburgh Medical School, S-598 South Biomedical Science Tower (BST), 3500 Terrace St., Pittsburgh, PA 15213, USA.
| |
Collapse
|
30
|
Sun PL, Jin Y, Park SY, Kim H, Park E, Jheon S, Kim K, Lee CT, Chung JH. Expression of Na+-K+-2Cl- cotransporter isoform 1 (NKCC1) predicts poor prognosis in lung adenocarcinoma and EGFR-mutated adenocarcinoma patients. QJM 2016; 109:237-44. [PMID: 26559081 DOI: 10.1093/qjmed/hcv207] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Accepted: 10/29/2015] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Sodium-potassium-chloride cotransporter isoform 1 (NKCC1) is an active ions cotransporter and modulates cellular volume and migration. NKCC1 blockers can inhibit cancer cell growth. AIM We aimed to elucidate the expression and prognostic significance of NKCC1 in non-small cell lung cancer (NSCLC). METHODS We retrospectively analyzed 788 NSCLC patients with either adenocarcinoma (n = 503) or squamous cell carcinoma (n = 285) by immunohistochemistry to correlate NKCC1 expression with clinicopathologic and survival outcomes. RESULTS In adenocarcinoma, high NKCC1 expression was associated with larger tumor size (P = 0.013), vascular invasion (P < 0.001), lymphatic invasion (P < 0.001), perineural invasion (P = 0.019) and advanced pathologic stage (P < 0.001), but there are no significant correlations between NKCC1 expression and clinicopathological parameters in squamous cell carcinoma. Patients with high NKCC1 expression had significantly shorter disease-free survival (DFS;P < 0.001) and shorter overall survival (OS;P < 0.001) than those with low NKCC1 expression in adenocarcinoma. In squamous cell carcinoma, NKCC1 expression was not associated with prognosis. Multivariate analysis revealed that high NKCC1 expression was an independent prognostic factor for DFS in lung adenocarcinomas (HR, 1.709; 95% CI 1.029-2.130;P = 0.033) and for OS inEGFR-mutated adenocarcinoma patients (HR, 3.165; 95% CI 1.424-7.035;P = 0.005). CONCLUSION NKCC1 high expression predicted a bad clinical outcome for lung adenocarcinoma patients andEGFR-mutated subgroup. Therefore, NKCC1 may play a role in lung adenocarcinoma and novel therapeutic tactics could be developed by targeting NKCC1 protein.
Collapse
Affiliation(s)
- Ping-Li Sun
- From the Department of Pathology, Second Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Yan Jin
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Soo Young Park
- Department of Pathology, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Republic of Korea
| | - Hyojin Kim
- Department of Pathology, Seoul National University Hospital, Seoul National University College of Medicine, Jongno-Gu, Seoul, Republic of Korea
| | - Eunhyang Park
- Department of Pathology, Seoul National University Hospital, Seoul National University College of Medicine, Jongno-Gu, Seoul, Republic of Korea
| | - Sanghoon Jheon
- Department of Thoracic and Cardiovascular Surgery, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Republic of Korea
| | - Kwhanmien Kim
- Department of Thoracic and Cardiovascular Surgery, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Republic of Korea
| | - Choon-Taek Lee
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Republic of Korea
| | - Jin-Haeng Chung
- Department of Pathology, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Republic of Korea
| |
Collapse
|
31
|
Huberfeld G, Vecht CJ. Seizures and gliomas — towards a single therapeutic approach. Nat Rev Neurol 2016; 12:204-16. [DOI: 10.1038/nrneurol.2016.26] [Citation(s) in RCA: 118] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
32
|
Simon OJ, Müntefering T, Grauer OM, Meuth SG. The role of ion channels in malignant brain tumors. J Neurooncol 2015; 125:225-35. [PMID: 26334315 DOI: 10.1007/s11060-015-1896-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2015] [Accepted: 08/14/2015] [Indexed: 12/15/2022]
Abstract
Malignant gliomas are the most common primary brain tumors and have poor clinical prognosis, despite multimodal therapeutic strategies. In recent years, ion channels have emerged as major players in tumor pathophysiology regarding all hallmarks of cancer. Since ion channels are easily accessible structures, they may prove to be effective targets for canner therapy, although their broad expression pattern and role in physiological processes should be taken into consideration. This review summarizes the current knowledge on the role of ion channels in the pathophysiology of malignant gliomas, especially glioblastoma, and evaluates their potential role in targeted antiglioma therapy.
Collapse
Affiliation(s)
- Ole J Simon
- Department of Neurology, University of Münster, Albert-Schweitzer-Campus 1, 48149, Münster, Germany.
| | - Thomas Müntefering
- Department of Neurology, University of Münster, Albert-Schweitzer-Campus 1, 48149, Münster, Germany
| | - Oliver M Grauer
- Department of Neurology, University of Münster, Albert-Schweitzer-Campus 1, 48149, Münster, Germany
| | - Sven G Meuth
- Department of Neurology, University of Münster, Albert-Schweitzer-Campus 1, 48149, Münster, Germany
| |
Collapse
|
33
|
Fu P, Tang R, Yu Z, Huang S, Xie M, Luo X, Wang W. Bumetanide-induced NKCC1 inhibition attenuates oxygen-glucose deprivation-induced decrease in proliferative activity and cell cycle progression arrest in cultured OPCs via p-38 MAPKs. Brain Res 2015; 1613:110-9. [PMID: 25881895 DOI: 10.1016/j.brainres.2015.04.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Revised: 03/23/2015] [Accepted: 04/04/2015] [Indexed: 11/28/2022]
Abstract
The Na-K-Cl co-transporter 1 (NKCC1; a member of the cation-chloride co-transporter family) mediates the coupled movement of Na(+) and/or K(+) with Cl(-) across the plasma membrane of cells (Haas and Forbush, 2000, Annu. Rev. Physiol., 62, 515-534; Russell, 2000, Physiol. Rev., 80, 211-276). Although it acts as an important regulator of cell volume, secretion, and modulator of cell apoptosis and proliferation (Chen et al., 2005, J. Cereb. Blood Flow Metab., 25, 54-66; Kahle et al., 2008, Nat. Clin. Pract. Neurol., 4, 490-503; Kidokoro et al., 2014, Am. J. Physiol. Ren. Physiol., 306, F1155-F1160; Wang et al., 2011, Cell. Physiol. Biochem., 28, 703-714), NKCC1׳s effects on oligodendrocyte precursor cells (OPCs) have not been characterized. The aim of this study was to investigate whether and to what extent inhibition of NKCC1 alters oxygen glucose deprivation (OGD)-induced cell cycle progression. In the present study, we demonstrated that inhibition of NKCC1 with bumetanide attenuates the decrease in OGD-induced DNA synthesis in cultured OPCs. Western blots showed that NKCC1 inhibition led to an increased expression of cyclin D1, CDK 4, and cyclin E in OGD-treated cells. Furthermore, our results showed bumetanide attenuated the decrease in OGD-induced proliferation and arrest of cell cycle progression via the P-38 MAPK signaling cascade. Thus, NKCC1 plays important roles in the proliferation of OPCs under OGD-induced stress.
Collapse
Affiliation(s)
- Peicai Fu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China
| | - Ronghua Tang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China
| | - Zhiyuan Yu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China; Key Laboratory of Neurological Diseases (HUST), Ministry of Education of China, Wuhan 430030, PR China
| | - Shanshan Huang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China
| | - Minjie Xie
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China; Key Laboratory of Neurological Diseases (HUST), Ministry of Education of China, Wuhan 430030, PR China
| | - Xiang Luo
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China
| | - Wei Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China; Key Laboratory of Neurological Diseases (HUST), Ministry of Education of China, Wuhan 430030, PR China.
| |
Collapse
|
34
|
Mathieu V, Chantôme A, Lefranc F, Cimmino A, Miklos W, Paulitschke V, Mohr T, Maddau L, Kornienko A, Berger W, Vandier C, Evidente A, Delpire E, Kiss R. Sphaeropsidin A shows promising activity against drug-resistant cancer cells by targeting regulatory volume increase. Cell Mol Life Sci 2015; 72:3731-46. [PMID: 25868554 DOI: 10.1007/s00018-015-1902-6] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Revised: 03/10/2015] [Accepted: 04/02/2015] [Indexed: 12/19/2022]
Abstract
Despite the recent advances in the treatment of tumors with intrinsic chemotherapy resistance, such as melanoma and renal cancers, their prognosis remains poor and new chemical agents with promising activity against these cancers are urgently needed. Sphaeropsidin A, a fungal metabolite whose anticancer potential had previously received little attention, was isolated from Diplodia cupressi and found to display specific anticancer activity in vitro against melanoma and kidney cancer subpanels in the National Cancer Institute (NCI) 60-cell line screen. The NCI data revealed a mean LC50 of ca. 10 µM and a cellular sensitivity profile that did not match that of any other agent in the 765,000 compound database. Subsequent mechanistic studies in melanoma and other multidrug-resistant in vitro cancer models showed that sphaeropsidin A can overcome apoptosis as well as multidrug resistance by inducing a marked and rapid cellular shrinkage related to the loss of intracellular Cl(-) and the decreased HCO3 (-) concentration in the culture supernatant. These changes in ion homeostasis and the absence of effects on the plasma membrane potential were attributed to the sphaeropsidin A-induced impairment of regulatory volume increase (RVI). Preliminary results also indicate that depending on the type of cancer, the sphaeropsidin A effects on RVI could be related to Na-K-2Cl electroneutral cotransporter or Cl(-)/HCO3 (-) anion exchanger(s) targeting. This study underscores the modulation of ion-transporter activity as a promising therapeutic strategy to combat drug-resistant cancers and identifies the fungal metabolite, sphaeropsidin A, as a lead to develop anticancer agents targeting RVI in cancer cells.
Collapse
Affiliation(s)
- Véronique Mathieu
- Laboratoire de Cancérologie et de Toxicologie Expérimentale, Faculté de Pharmacie, Université Libre de Bruxelles (ULB), Campus de la Plaine, Boulevard du Triomphe, 1050, Brussels, Belgium.
| | - Aurélie Chantôme
- Inserm UMR 1069, Université François Rabelais and network "Ion channels and cancer - Canceropole Grand Ouest", Tours, France
| | - Florence Lefranc
- Service de Neurochirurgie, Hôpital Erasme, Université Libre de Bruxelles, Brussels, Belgium
| | - Alessio Cimmino
- Dipartimento di Scienze Chimiche, Università di Napoli Federico II, Complesso Universitario Monte Sant'Angelo, Via Cintia 4, 80126, Naples, Italy
| | - Walter Miklos
- Department of Medicine I, Institute of Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Verena Paulitschke
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Thomas Mohr
- Department of Medicine I, Institute of Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Lucia Maddau
- Dipartimento di Agraria, Sezione di Patologia vegetale ed Entomologia, Università degli Studi di Sassari, Viale Italia 39, 07100, Sassari, Italy
| | - Alexander Kornienko
- Department of Chemistry and Biochemistry, Texas State University, San Marcos, TX, 78666, USA
| | - Walter Berger
- Department of Medicine I, Institute of Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Christophe Vandier
- Inserm UMR 1069, Université François Rabelais and network "Ion channels and cancer - Canceropole Grand Ouest", Tours, France
| | - Antonio Evidente
- Dipartimento di Scienze Chimiche, Università di Napoli Federico II, Complesso Universitario Monte Sant'Angelo, Via Cintia 4, 80126, Naples, Italy
| | - Eric Delpire
- Department of Anesthesiology, Vanderbilt University Medical School, Nashville, TN, USA
| | - Robert Kiss
- Laboratoire de Cancérologie et de Toxicologie Expérimentale, Faculté de Pharmacie, Université Libre de Bruxelles (ULB), Campus de la Plaine, Boulevard du Triomphe, 1050, Brussels, Belgium
| |
Collapse
|
35
|
Anti-angiogenic Effects of Bumetanide Revealed by DCE-MRI with a Biodegradable Macromolecular Contrast Agent in a Colon Cancer Model. Pharm Res 2015; 32:3029-43. [PMID: 25840948 DOI: 10.1007/s11095-015-1684-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Accepted: 03/19/2015] [Indexed: 10/23/2022]
Abstract
PURPOSE To assess the antiangiogenic effect of bumetanide with dynamic contrast enhanced (DCE)-MRI and a biodegradable macromolecular MRI contrast agent. METHODS A new polydisulfide containing macrocyclic gadolinium (Gd(III)) chelates, poly([(Gd-DOTA)-DETA]-co-DTBP) (GODP), was synthesized as a safe biodegradable macromolecular MRI contrast agent for DCE-MRI. Nude mice bearing flank HT29 colon cancer xenografts were then treated daily with either bumetanide or saline for a total of 3 weeks. DCE-MRI was performed before and after the treatment weekly. The DCE-MRI data were analyzed using the adiabiatic approximation to the tissue homogeneity (AATH) model to assess the change of tumor vascularity in response to the treatment. Immunohistochemistry (IHC) and western blot were performed to study tumor angiogenic biomarkers and hypoxia. RESULTS DCE-MRI with GODP revealed that bumetanide reduced vascular permeability and plasma volume fraction by a significantly greater extent than the saline control therapy after 3 weeks of therapy. These changes were verified by the significant decline of CD31 and VEGF expression in the bumetanide treatment group. Despite a significant regression in vascularity, the tumors remained highly proliferative. Overexpression of the transcription factor HIF-1α in response to elevated hypoxia is thought to be the driving force behind the uninterrupted tumor expansion. CONCLUSION This study demonstrated the effectiveness of DCE-MRI with GODP in detecting vascular changes following the administration of bumetanide. Bumetanide has the potential to curtail growth of the tumor vasculature and can be employed in future therapeutic strategies.
Collapse
|
36
|
TRPM7 channels regulate glioma stem cell through STAT3 and Notch signaling pathways. Cell Signal 2014; 26:2773-81. [PMID: 25192910 DOI: 10.1016/j.cellsig.2014.08.020] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Accepted: 08/17/2014] [Indexed: 12/11/2022]
Abstract
Glioblastoma multiforme (GBM) is the most common and aggressive primary brain tumor in adults with median survival time of 14.6 months. A small fraction of cancer stem cells (CSC) initiate and maintain tumors thus driving glioma tumorigenesis and being responsible for resistance to classical chemo- and radio-therapies. It is desirable to identify signaling pathways related to CSC to develop novel therapies to selectively target them. Transient receptor potential cation channel, subfamily M, member 7, also known as TRPM7 is a ubiquitous, Ca(2+) and Mg(2+) permeable ion channels that are special in being both an ion channel and a serine/threonine kinase. In studies of glioma cells silenced for TRPM7, we demonstrated that Notch (Notch1, JAG1, Hey2, and Survivin) and STAT3 pathways are down regulated in glioma cells grown in monolayer. Furthermore, phospho-STAT3, Notch target genes and CSC markers (ALDH1 and CD133) were significantly higher in spheroid glioma CSCs when compared with monolayer cultures. The results further show that tyrosine-phosphorylated STAT3 binds and activates the ALDH1 promoters in glioma cells. We found that TRMP7-induced upregulation of ALDH1 expression is associated with increases in ALDH1 activity and is detectable in stem-like cells when expanded as spheroid CSCs. Finally, TRPM7 promotes proliferation, migration and invasion of glioma cells. These demonstrate that TRPM7 activates JAK2/STAT3 and/or Notch signaling pathways and leads to increased cell proliferation and migration. These findings for the first time demonstrates that TRPM7 (1) activates a previously unrecognized STAT3→ALDH1 pathway, and (2) promotes the induction of ALDH1 activity in glioma cells.
Collapse
|
37
|
Cong D, Zhu W, Shi Y, Pointer KB, Clark PA, Shen H, Kuo JS, Hu S, Sun D. Upregulation of NHE1 protein expression enables glioblastoma cells to escape TMZ-mediated toxicity via increased H⁺ extrusion, cell migration and survival. Carcinogenesis 2014; 35:2014-24. [PMID: 24717311 DOI: 10.1093/carcin/bgu089] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Sodium-hydrogen exchanger isoform 1 (NHE1) plays a role in survival and migration/invasion of several cancers and is an emerging new therapeutic target. However, the role of NHE1 in glioblastoma and the interaction of NHE1 expression and function in glioblastoma cells with cytotoxic temozolomide (TMZ) therapy remain unknown. In this study, we detected high levels of NHE1 protein only in primary human glioma cells (GC), glioma xenografts and glioblastoma, but not in human neural stem cells or astrocytes. GC exhibited an alkaline resting pHi (7.46±0.04) maintained by robust NHE1-mediated H(+) extrusion. GC treatment with TMZ for 2-24h triggered a transient decrease in pHi, which recovered by 48h and correlated with concurrent upregulation of NHE1 protein expression. NHE1 protein was colocalized with ezrin at lamellipodia and probably involved in GC migration. The TMZ-treated GC exhibited increased migration and invasion, which was attenuated by addition of NHE1 inhibitor HOE-642. Most importantly, NHE1 inhibition prevented prosurvival extracellular signal-regulated kinase activation and accelerated TMZ-induced apoptosis. Taken together, our study provides the first evidence that GC upregulate NHE1 protein to maintain alkaline pHi. Combining TMZ therapy with NHE1 inhibition suppresses GC migration and invasion, and also augments TMZ-induced apoptosis. These findings strongly suggest that NHE1 is an important cytoprotective mechanism in GC and presents a new therapeutic strategy of combining NHE1 inhibition and TMZ chemotherapy.
Collapse
Affiliation(s)
- Damin Cong
- Department of Neurological Surgery, The Second Affiliated Hospital of the Harbin Medical University, Harbin 150086, China, Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Wen Zhu
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Yejie Shi
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Kelli B Pointer
- Department of Neurological Surgery, University of Wisconsin, Madison, WI 53705, USA
| | - Paul A Clark
- Department of Neurological Surgery, University of Wisconsin, Madison, WI 53705, USA
| | - Hongmei Shen
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - John S Kuo
- Department of Neurological Surgery, University of Wisconsin, Madison, WI 53705, USA, Carbone Cancer Center, University of Wisconsin, Madison, WI 53705, USA and, Pittsburgh, PA 15213, USA and
| | - Shaoshan Hu
- Department of Neurological Surgery, The Second Affiliated Hospital of the Harbin Medical University, Harbin 150086, China
| | - Dandan Sun
- Department of Neurological Surgery, The Second Affiliated Hospital of the Harbin Medical University, Harbin 150086, China, Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA, Veterans Affairs Pittsburgh Health Care System, Geriatric Research, Educational and Clinical Center, Pittsburgh, PA, 15213, USA
| |
Collapse
|
38
|
Zhu W, Begum G, Pointer K, Clark PA, Yang SS, Lin SH, Kahle KT, Kuo JS, Sun D. WNK1-OSR1 kinase-mediated phospho-activation of Na+-K+-2Cl- cotransporter facilitates glioma migration. Mol Cancer 2014; 13:31. [PMID: 24555568 PMCID: PMC3936893 DOI: 10.1186/1476-4598-13-31] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2013] [Accepted: 02/13/2014] [Indexed: 12/22/2022] Open
Abstract
Background The bumetanide (BMT)-sensitive Na+-K+-2Cl- cotransporter isoform 1 (NKCC1) maintains cell volume homeostasis by increasing intracellular K+ and Cl- content via regulatory volume increase (RVI). Expression levels of NKCC1 positively correlate with the histological grade and severity of gliomas, the most common primary adult brain tumors, and up-regulated NKCC1 activity facilitates glioma cell migration and apoptotic resistance to the chemotherapeutic drug temozolomide (TMZ). However, the cellular mechanisms underlying NKCC1 functional up-regulation in glioma and in response to TMZ administration remain unknown. Methods Expression of NKCC1 and its upstream kinases With-No-K (Lysine) kinase 1 (WNK1) and oxidative stress-responsive kinase-1 (OSR1) in different human glioma cell lines and glioma specimens were detected by western blotting and immunostaining. Live cell imaging and microchemotaxis assay were applied to record glioma cell movements under different treatment conditions. Fluorescence indicators were utilized to measure cell volume, intracellular K+ and Cl- content to reflect the activity of NKCC1 on ion transportation. Small interfering RNA (siRNA)-mediated knockdown of WNK1 or OSR1 was used to explore their roles in regulation of NKCC1 activity in glioma cells. Results of different treatment groups were compared by one-way ANOVA using the Bonferroni post-hoc test in the case of multiple comparisons. Results We show that compared to human neural stem cells and astrocytes, human glioma cells exhibit robust increases in the activation and phosphorylation of NKCC1 and its two upstream regulatory kinases, WNK1 and OSR1. siRNA-mediated knockdown of WNK1 or OSR1 reduces intracellular K+ and Cl- content and RVI in glioma cells by abolishing NKCC1 regulatory phospho-activation. Unexpectedly, TMZ activates the WNK1/OSR1/NKCC1 signaling pathway and enhances glioma migration. Pharmacological inhibition of NKCC1 with its potent inhibitor BMT or siRNA knockdown of WNK1 or OSR1 significantly decreases glioma cell migration after TMZ treatment. Conclusion Together, our data show a novel role for the WNK1/OSR1/NKCC1 pathway in basal and TMZ-induced glioma migration, and suggest that glioma treatment with TMZ might be improved by drugs that inhibit elements of the WNK1/OSR1/NKCC1 signaling pathway.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Dandan Sun
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
39
|
Abstract
Between 30% and 50% of patients with brain tumors first present with a seizure, and up to 30% more will develop seizures later. Therefore, optimal management of these patients requires a rational approach to the use of antiseizure medications. Based on current evidence, prophylactic prescription of long-term antiepileptic drugs (AEDs) in patients with brain tumors in patients who did not present with seizures is not justified. Because of the high risk of recurrence, however, AED treatment should be strongly considered after a single seizure considered to be due to a tumor. Because of the lack of well-controlled randomized trials, the decision on which AED provides the best risk-benefit ratio in the individual patient is based mostly on physician's judgment rather than sound scientific evidence. In patients who may require chemotherapy, a non-enzyme-inducing AED is preferred for initial treatment to minimize the risk of drug interactions that impact adversely on the outcome of anticancer chemotherapy. Several retrospective studies in seizure patients with glioblastoma treated with chemotherapy have provided evidence for a moderately improved survival with the use of valproic acid, possibly due to inhibition of histone deacetylase. However, valproic acid may also increase the hematologic toxicity of antineoplastic drugs, presumably by inhibiting their metabolism, and may independently impair hemostasis, which is of some concern for patients who require surgical intervention. Among newer generation AEDs, levetiracetam has a number of advantageous features, including availability of a parenteral formulation, but other agents such as gabapentin, lamotrigine, oxcarbazepine, topiramate, and zonisamide may also be considered. Potentially more effective treatments targeting specific mechanisms of epileptogenesis and ictogenesis are being investigated. Resection of the tumor, radiation therapy, or chemotherapy can bring refractory seizures under control or prolong the duration of seizure freedom, an effect that does not appear to be necessarily related to removal or shrinkage of the tumor mass. In patients with a successfully treated tumor and an overall good prognosis for long-term survival, gradual discontinuation of AEDs may be considered.
Collapse
Affiliation(s)
- Emilio Perucca
- Department of Internal Medicine and Therapeutics, University of Pavia and C Mondino National Neurological Institute, Pavia, Italy
| |
Collapse
|
40
|
Hoffmann EK, Lambert IH. Ion channels and transporters in the development of drug resistance in cancer cells. Philos Trans R Soc Lond B Biol Sci 2014; 369:20130109. [PMID: 24493757 PMCID: PMC3917363 DOI: 10.1098/rstb.2013.0109] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Multi-drug resistance (MDR) to chemotherapy is the major challenge in the treatment of cancer. MDR can develop by numerous mechanisms including decreased drug uptake, increased drug efflux and the failure to undergo drug-induced apoptosis. Evasion of drug-induced apoptosis through modulation of ion transporters is the main focus of this paper and we demonstrate how pro-apoptotic ion channels are downregulated, while anti-apoptotic ion transporters are upregulated in MDR. We also discuss whether upregulation of ion transport proteins that are important for proliferation contribute to MDR. Finally, we discuss the possibility that the development of MDR involves sequential and localized upregulation of ion channels involved in proliferation and migration and a concomitant global and persistent downregulation of ion channels involved in apoptosis.
Collapse
Affiliation(s)
- Else K Hoffmann
- Department of Biology, University of Copenhagen, , 13 Universitetsparken, Copenhagen Ø 2100, Denmark
| | | |
Collapse
|
41
|
Shiizaki S, Naguro I, Ichijo H. Activation mechanisms of ASK1 in response to various stresses and its significance in intracellular signaling. Adv Biol Regul 2013; 53:135-44. [PMID: 23031789 DOI: 10.1016/j.jbior.2012.09.006] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2012] [Accepted: 09/06/2012] [Indexed: 05/25/2023]
Abstract
Apoptosis signal-regulating kinase 1 (ASK1) is a member of the mitogen-activated protein kinase kinase kinase family. ASK1 activates c-jun N-terminal kinase (JNK) and p38 in response to various stimuli such as oxidative stress, endoplasmic reticulum stress, infection and calcium influx. Under these stress conditions, ASK1 plays important roles in intracellular signaling pathways and biological functions. Diverse proteins are known to interact with ASK1 and regulate the activity of ASK1. However, activation mechanisms of ASK1 and ASK1-binding proteins which regulate the activity of ASK1 have not been completely understood. In this review, we focus on the recent findings on ASK1 and update the regulatory mechanisms of ASK1 activity.
Collapse
Affiliation(s)
- Shigeru Shiizaki
- Laboratory of Cell signaling, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | | | | |
Collapse
|