18 results on '"SWELL1"'
Search Results
2. Trends in volume-regulated anion channel (VRAC) research: visualization and bibliometric analysis from 2014 to 2022.
- Author
-
Tianbao Liu, Yin Li, Dawei Wang, Stauber, Tobias, and Jiajun Zhao
- Subjects
BIBLIOMETRICS ,DATA visualization ,CEREBRAL edema ,ANIONS ,STOMACH cancer ,ION channels - Abstract
Objective: In this study, we utilized bibliometric methods to assess the worldwide scientific output and identify hotspots related to the research on the volume-regulated anion channel (VRAC) from 2014 to 2022. Methods: From Web of Science, we obtained studies related to VRAC published from 2014 to 2022. To analyzed the data, we utilized VOSviewer, a tool for visualizing network, to create networks based on the collaboration between countries, institutions, and authors. Additionally, we performed an analysis of journal co-citation, document citation, and co-occurrence of keywords. Furthermore, we employed CiteSpace (6.1. R6 Advanced) to analyzed keywords and co-cited references with the strongest burst. Results: The final analysis included a total of 278 related articles and reviews, covering the period from 2014 to 2022. The United States emerged as the leading country contributing to this field, while the University of Copenhagen stood out as the most prominent institution. The author with most publications and most citations was Thomas J. Jentsch. Among the cited references, the article by Voss et al. published in Science (2014) gained significant attention for its identification of LRRC8 heteromers as a crucial component of the volume-regulated anion channel VRAC. Pflügers Archiv European Journal of Physiology and Journal of Physiology-London were the leading journals in terms of the quantity of associated articles and citations. Through the analysis of keyword co-occurrence, it was discovered that VRAC is involved in various physiological processes including cell growth, migration, apoptosis, swelling, and myogenesis, as well as anion and organic osmolyte transport including chloride, taurine, glutamate and ATP. VRAC is also associated with related ion channels such as TMEM16A, TMEM16F, pannexin, and CFTR, and associated with various diseases including epilepsy, leukodystrophy, atherosclerosis, hypertension, cerebral edema, stroke, and different types of cancer including gastric cancer, glioblastoma and hepatocellular carcinoma. Furthermore, VRAC is involved in anti-tumor drug resistance by regulating the uptake of platinum-based drugs and temozolomide. Additionally, VRAC has been studied in the context of pharmacology involving DCPIB and flavonoids. Conclusion: The aim of this bibliometric analysis is to provide an overall perspective for research on VRAC. VRAC has become a topic of increasing interest, and our analysis shows that it continues to be a prominent area. This study offers insights into the investigation of VRAC channel and may guide researchers in identifying new directions for future research. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
3. Glial Chloride Channels in the Function of the Nervous System Across Species
- Author
-
Fernandez-Abascal, Jesus, Graziano, Bianca, Encalada, Nicole, Bianchi, Laura, Crusio, Wim E., Series Editor, Dong, Haidong, Series Editor, Radeke, Heinfried H., Series Editor, Rezaei, Nima, Series Editor, Steinlein, Ortrud, Series Editor, Xiao, Junjie, Series Editor, and Zhou, Lei, editor
- Published
- 2021
- Full Text
- View/download PDF
4. Activation of Swell1 in microglia suppresses neuroinflammation and reduces brain damage in ischemic stroke
- Author
-
Baoyi Chen, Cong Xie, Tengrui Shi, Shiqin Yue, Weiping Li, Guodong Huang, Yuan Zhang, and Wenlan Liu
- Subjects
Inflammation ,Swell1 ,Chloride channel ,Ischemic stroke ,Microglia ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
Cl− movement and Cl−-sensitive signal pathways contributes to the survival and switch of inflammatory phenotype of microglia and are believed to play a key role in the inflammatory brain injury after ischemic stroke. Here, we demonstrated an important role of Cl− transmembrane transporter Swell1, in the survival and M2-like polarization of microglia in ischemic stroke. Knockdown or overexpression of Swell1 in cultured microglia inhibited or increased hypotonic-activated Cl− currents, respectively, and these changes were completely blocked by the volume-regulated anion channels (VRACs) inhibitor DCPIB. Swell1 conditional knock-in mice promoted microglia survival in ischemic brain region and resulted in significant reductions in neural cell death, infarction volume and neurological deficits following transient middle cerebral artery occlusion (tMCAO). Using gene manipulating technique and pharmacological inhibitors, we further revealed that Swell1 opening led to SGK1 (a Cl−-sensitive kinase)-mediated activation of FOXO3a/CREB as well as WNK1 (another Cl−-sensitive kinase)-mediated SPAK/OSR1-CCCs activation, which promoted microglia survival and M2-like polarization, thereby attenuating neuroinflammation and ischemic brain injury. Taken together, our results demonstrated that Swell1 is an essential component of microglia VRACs and its activation protects against ischemic brain injury through promoting microglia survival and M2-like polarization.
- Published
- 2023
- Full Text
- View/download PDF
5. Effect of Swell1 on regulating chondrocyte hypertrophy during the condylar osteochondral development process in mice.
- Author
-
Chen, Long, Chen, Yu, Xu, Yanting, Shen, Steve GF., and Dai, Jiewen
- Subjects
- *
MANDIBULAR condyle , *ENDOCHONDRAL ossification , *BONE density , *CHONDROGENESIS , *COMPACT bone , *HYPERTROPHY , *CELL size - Abstract
Chondrocyte hypertrophy is a significant factor in cartilage development, yet the molecular mechanism for cell volume expand during the process is remains unclear. In the present study, the relationship between Swell1, a cell volume regulated anion channel, and chondrocyte hypertrophy was explored. The results reveal that the spatiotemporal expression of Swell1 was similar with the development process of hypertrophic chondrocytes in condyles. Through Col10a1 mediated knock out of Swell1 in hypertrophy chondrocytes, we found that there are less obvious boundary between different condylar cartilage layers in which increased hypertrophic chondrocytes were scattered in all three cartilage layers. The cortical bone mass and bone mineral density in the subchondral bone significantly increased. Additionally, knock out of Swell1 could increase the expression of OCN in the femur condyle. Based on the aforementioned findings, a conclusion could be drawn that Swell1 is a significant factor in chondrocyte hypertrophy during the condylar osteochondral development process, and there was some difference between the mandibular and femur condyles, which will provide some new clues for understanding the development of cartilage and related diseases. • Chondrocyte hypertrophy is very important in the process of endochondral osteogenesis. •The spatiotemporal expression of Swell1 was similar with the development process of hypertrophic chondrocytes in condyles. •Swell1 affects bone and cartilage morphology during the development of mouse condyle. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
6. LRRC8/VRAC volume-regulated anion channels are crucial for hearing.
- Author
-
Knecht DA, Zeziulia M, Bhavsar MB, Puchkov D, Maier H, and Jentsch TJ
- Subjects
- Animals, Mice, Hearing, Cochlea metabolism, Cochlea pathology, Hair Cells, Auditory metabolism, Hair Cells, Auditory pathology, Stria Vascularis metabolism, Stria Vascularis pathology, Deafness metabolism, Deafness pathology, Deafness genetics, Voltage-Dependent Anion Channels metabolism, Voltage-Dependent Anion Channels genetics, Membrane Proteins metabolism, Membrane Proteins genetics, Mice, Knockout
- Abstract
Hearing crucially depends on cochlear ion homeostasis as evident from deafness elicited by mutations in various genes encoding cation or anion channels and transporters. Ablation of ClC‑K/barttin chloride channels causes deafness by interfering with the positive electrical potential of the endolymph, but roles of other anion channels in the inner ear have not been studied. Here we report the intracochlear distribution of all five LRRC8 subunits of VRAC, a volume-regulated anion channel that transports chloride, metabolites, and drugs such as the ototoxic anti-cancer drug cisplatin, and explore its physiological role by ablating its subunits. Sensory hair cells express all LRRC8 isoforms, whereas only LRRC8A, D and E were found in the potassium-secreting epithelium of the stria vascularis. Cochlear disruption of the essential LRRC8A subunit, or combined ablation of LRRC8D and E, resulted in cochlear degeneration and congenital deafness of Lrrc8a
-/- mice. It was associated with a progressive degeneration of the organ of Corti and its innervating spiral ganglion. Like disruption of ClC-K/barttin, loss of VRAC severely reduced the endocochlear potential. However, the mechanism underlying this reduction seems different. Disruption of VRAC, but not ClC-K/barttin, led to an almost complete loss of Kir4.1 (KCNJ10), a strial K+ channel crucial for the generation of the endocochlear potential. The strong downregulation of Kir4.1 might be secondary to a loss of VRAC-mediated transport of metabolites regulating inner ear redox potential such as glutathione. Our study extends the knowledge of the role of cochlear ion transport in hearing and ototoxicity., Competing Interests: Conflict of interest The authors declare that they have no conflicts of interest with the contents of this article., (Copyright © 2024 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2024
- Full Text
- View/download PDF
7. More than just a pressure relief valve: physiological roles of volume-regulated LRRC8 anion channels.
- Author
-
Lingye Chen, König, Benjamin, Tianbao Liu, Pervaiz, Sumaira, Razzaque, Yasmin S., and Stauber, Tobias
- Subjects
- *
RELIEF valves , *CALCIUM channels , *CELL communication , *CELLULAR control mechanisms , *ANIONS , *GENOME editing - Abstract
The volume-regulated anion channel (VRAC) is a key player in the volume regulation of vertebrate cells. This ubiquitously expressed channel opens upon osmotic cell swelling and potentially other cues and releases chloride and organic osmolytes, which contributes to regulatory volume decrease (RVD). A plethora of studies have proposed a wide range of physiological roles for VRAC beyond volume regulation including cell proliferation, differentiation and migration, apoptosis, intercellular communication by direct release of signaling molecules and by supporting the exocytosis of insulin. VRAC was additionally implicated in pathological states such as cancer therapy resistance and excitotoxicity under ischemic conditions. Following extensive investigations, 5 years ago leucine-rich repeat-containing family 8 (LRRC8) heteromers containing LRRC8A were identified as the pore-forming components of VRAC. Since then, molecular biological approaches have allowed further insight into the biophysical properties and structure of VRAC. Heterologous expression, siRNA-mediated downregulation and genome editing in cells, as well as the use of animal models have enabled the assessment of the proposed physiological roles, together with the identification of new functions including spermatogenesis and the uptake of antibiotics and platinum-based cancer drugs. This review discusses the recent molecular biological insights into the physiology of VRAC in relation to its previously proposed roles. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
8. Structural insights into anion selectivity and activation mechanism of LRRC8 volume-regulated anion channels.
- Author
-
Liu, Heng, Polovitskaya, Maya M., Yang, Linlin, Li, Meiling, Li, Hongyue, Han, Zhen, Wu, Jianguo, Zhang, Qiansen, Jentsch, Thomas J., and Liao, Jun
- Abstract
Volume-regulated anion channels (VRACs) are hexamers of LRRC8 proteins that are crucial for cell volume regulation. N termini (NTs) of the obligatory LRRC8A subunit modulate VRACs activation and ion selectivity, but the underlying mechanisms remain poorly understood. Here, we report a 2.8-Å cryo-electron microscopy structure of human LRRC8A that displays well-resolved NTs. Amino-terminal halves of NTs fold back into the pore and constrict the permeation path, thereby determining ion selectivity together with an extracellular selectivity filter with which it works in series. They also interact with pore-surrounding helices and support their compact arrangement. The C-terminal halves of NTs interact with intracellular loops that are crucial for channel activation. Molecular dynamics simulations indicate that low ionic strength increases NT mobility and expands the radial distance between pore-surrounding helices. Our work suggests an unusual pore architecture with two selectivity filters in series and a mechanism for VRAC activation by cell swelling. [Display omitted] • Cryo-EM structure of LRRC8A volume-regulated anion channel reveals back-folded N termini • N termini form a selectivity filter in series with a second one at the external pore opening • Molecular dynamics calculations suggest that low ionic strength partially unwinds N termini • The unwound N termini may modulate channel activation by cell swelling Liu et al. show that the N terminus of LRRC8A, resolved in their structure of a volume-regulated VRAC anion channel, inserts into the pore. It forms a second selectivity filter in series with an external one. Its partial unfolding under low ionic strength may trigger activation of VRAC by cell swelling. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
9. Trends in volume-regulated anion channel (VRAC) research: visualization and bibliometric analysis from 2014 to 2022.
- Author
-
Liu T, Li Y, Wang D, Stauber T, and Zhao J
- Abstract
Objective: In this study, we utilized bibliometric methods to assess the worldwide scientific output and identify hotspots related to the research on the volume-regulated anion channel (VRAC) from 2014 to 2022. Methods: From Web of Science, we obtained studies related to VRAC published from 2014 to 2022. To analyzed the data, we utilized VOSviewer, a tool for visualizing network, to create networks based on the collaboration between countries, institutions, and authors. Additionally, we performed an analysis of journal co-citation, document citation, and co-occurrence of keywords. Furthermore, we employed CiteSpace (6.1. R6 Advanced) to analyzed keywords and co-cited references with the strongest burst. Results: The final analysis included a total of 278 related articles and reviews, covering the period from 2014 to 2022. The United States emerged as the leading country contributing to this field, while the University of Copenhagen stood out as the most prominent institution. The author with most publications and most citations was Thomas J. Jentsch. Among the cited references, the article by Voss et al. published in Science (2014) gained significant attention for its identification of LRRC8 heteromers as a crucial component of the volume-regulated anion channel VRAC. Pflügers Archiv European Journal of Physiology and Journal of Physiology-London were the leading journals in terms of the quantity of associated articles and citations. Through the analysis of keyword co-occurrence, it was discovered that VRAC is involved in various physiological processes including cell growth, migration, apoptosis, swelling, and myogenesis, as well as anion and organic osmolyte transport including chloride, taurine, glutamate and ATP. VRAC is also associated with related ion channels such as TMEM16A, TMEM16F, pannexin, and CFTR, and associated with various diseases including epilepsy, leukodystrophy, atherosclerosis, hypertension, cerebral edema, stroke, and different types of cancer including gastric cancer, glioblastoma and hepatocellular carcinoma. Furthermore, VRAC is involved in anti-tumor drug resistance by regulating the uptake of platinum-based drugs and temozolomide. Additionally, VRAC has been studied in the context of pharmacology involving DCPIB and flavonoids. Conclusion: The aim of this bibliometric analysis is to provide an overall perspective for research on VRAC. VRAC has become a topic of increasing interest, and our analysis shows that it continues to be a prominent area. This study offers insights into the investigation of VRAC channel and may guide researchers in identifying new directions for future research., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2023 Liu, Li, Wang, Stauber and Zhao.)
- Published
- 2023
- Full Text
- View/download PDF
10. Activation of Swell1 in microglia suppresses neuroinflammation and reduces brain damage in ischemic stroke.
- Author
-
Chen, Baoyi, Xie, Cong, Shi, Tengrui, Yue, Shiqin, Li, Weiping, Huang, Guodong, Zhang, Yuan, and Liu, Wenlan
- Subjects
- *
ISCHEMIC stroke , *BRAIN damage , *MICROGLIA , *NEUROINFLAMMATION , *CELL death - Abstract
Cl− movement and Cl−-sensitive signal pathways contributes to the survival and switch of inflammatory phenotype of microglia and are believed to play a key role in the inflammatory brain injury after ischemic stroke. Here, we demonstrated an important role of Cl− transmembrane transporter Swell1, in the survival and M2-like polarization of microglia in ischemic stroke. Knockdown or overexpression of Swell1 in cultured microglia inhibited or increased hypotonic-activated Cl− currents, respectively, and these changes were completely blocked by the volume-regulated anion channels (VRACs) inhibitor DCPIB. Swell1 conditional knock-in mice promoted microglia survival in ischemic brain region and resulted in significant reductions in neural cell death, infarction volume and neurological deficits following transient middle cerebral artery occlusion (tMCAO). Using gene manipulating technique and pharmacological inhibitors, we further revealed that Swell1 opening led to SGK1 (a Cl−-sensitive kinase)-mediated activation of FOXO3a/CREB as well as WNK1 (another Cl−-sensitive kinase)-mediated SPAK/OSR1-CCCs activation, which promoted microglia survival and M2-like polarization, thereby attenuating neuroinflammation and ischemic brain injury. Taken together, our results demonstrated that Swell1 is an essential component of microglia VRACs and its activation protects against ischemic brain injury through promoting microglia survival and M2-like polarization. • Swell1 is an essential component of microglia VRAC and is significantly upregulated induced by ischemic stroke. • Swell1 conditional knock-in mice ameliorates ischemic injury and neuroinflammatory after ischemic stroke. • Swell1 opening promotes microglial survival and M2-type polarization via activating the Cl−-sensitive kinase signaling pathway. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
11. SWELL1 promotes cell growth and metastasis of hepatocellular carcinoma in vitro and in vivo
- Author
-
Mei Liu, Panpan Lu, Yujia Xia, Yuhui Fan, Xiaoyu Ji, Qiang Ding, Dean Tian, Lili Li, and Xin Li
- Subjects
0301 basic medicine ,Research paper ,MMP, mitochondrial membrane potential ,Hepatocellular carcinoma ,Proliferation ,PKCa, protein kinase C alpha ,Apoptosis ,Metastasis ,PVDF, polyvinylidene difluoride ,chemistry.chemical_compound ,0302 clinical medicine ,ROS, cellular reactive oxygen species ,Cell Movement ,Neoplasm Metastasis ,RNA, Small Interfering ,SPHK1, sphingosine kinase 1 ,ANOVA, analysis of variance ,Liver Neoplasms ,EdU, 5-ethynyl-2′-deoxyuridine ,Cell migration ,General Medicine ,LCK, lymphocyte-specific protein tyrosine kinase ,PL, phospholipase ,ddc ,Gene Expression Regulation, Neoplastic ,030220 oncology & carcinogenesis ,RNA Interference ,Disease Susceptibility ,RVD, regulatory volume decrease ,IHC, immunohistochemistry ,PI3K, phosphoinositide 3-kinase ,Signal Transduction ,VRAC, volume-regulated anion channel ,Carcinoma, Hepatocellular ,Epithelial-Mesenchymal Transition ,Protein Kinase C-alpha ,MAP Kinase Signaling System ,Biology ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,In vivo ,CCK8, Cell Counting Kit-8 ,Cell Line, Tumor ,medicine ,Humans ,DAPI ,Epithelial–mesenchymal transition ,SWELL1 ,PI3K/AKT/mTOR pathway ,S1P, sphingosine-1-phosphate ,Cell Proliferation ,Neoplasm Staging ,GAPDH, Glyceraldehyde 3-phosphate dehydrogenase ,Cell growth ,qRT-PCR, quantitative real-time-PCR ,DCFH-DA, dichloro-dihydro-fluorescein diacetate ,Membrane Proteins ,medicine.disease ,digestive system diseases ,030104 developmental biology ,chemistry ,Cancer research ,BSA, bovine serum albumin ,HCC, hepatocellular carcinoma ,EMT, epithelial-to-mesenchymal transition ,DAPI, 4′, 6-diamidino-2-phenylindole - Abstract
Background SWELL1 was recently demonstrated to be an indispensable part of the volume-regulated anion channel (VRAC). VRAC is reported to participate in cell proliferation, survival, and migration. However, the correlation between SWELL1 and hepatocellular carcinoma (HCC) remains poorly-understood. In this study, we tried to explore the role of SWELL1 in HCC. Methods Immunohistochemistry and quantitative real-time-PCR (qRT-PCR) was used to measure SWELL1 expression in HCC samples obtained from patients with HCC. The effects of SWELL1 on HCC cell proliferation, apoptosis, and metastasis were analysed by corresponding cytological experiments including Cell Counting Kit-8 (CCK8), colony-forming, 5-ethynyl-2′-deoxyuridine (EdU), cell cycle analysis, TUNEL, Annexin V and PI staining, wound healing, transwell, and so on. BALB/c nude mice were used for the in vivo assays. qRT-PCR and western blotting was performed for molecular mechanisms. Findings SWELL1 was highly expressed in HCC tissues, and related to the poor prognosis. In vitro, the over-expression of SWELL1 significantly induced cell proliferation and migration, and inhibited apoptosis, whereas suppressing SWELL1 had the opposite effects. Moreover, knockdown of SWELL1 suppressed the growth and metastasis of HCC in vivo. Further experiments revealed that SWELL1 induced cell growth by activating the cyclinD1/CDK2 pathway via the connection with PKCa at the signalling level, and regulated cell migration through the JNK pathway in HCC. Interpretation SWELL1 acts as a promoter in the growth and metastasis of HCC cells and may be a potential intervention target for HCC. Fund This work is supported by the National Natural Science Foundation of China (No. 81572422 , 81700515 ).
- Published
- 2019
12. The identification of a volume-regulated anion channel: an amazing Odyssey.
- Author
-
Pedersen, S. F., Klausen, T. K., and Nilius, B.
- Subjects
- *
CELLULAR control mechanisms , *LEUCINE , *CHLORIDE channels , *OSMOREGULATION , *PHYSIOLOGICAL effects of anions , *VOLTAGE-gated ion channels , *PATHOLOGICAL physiology , *PANNEXINS - Abstract
The volume-regulated anion channel ( VRAC) plays a pivotal role in cell volume regulation in essentially all cell types studied. Additionally, VRAC appears to contribute importantly to a wide range of other cellular functions and pathological events, including cell motility, cell proliferation, apoptosis and excitotoxic glutamate release in stroke. Although biophysically, pharmacologically and functionally thoroughly described, VRAC has until very recently remained a genetic orphan. The search for the molecular identity of VRAC has been long and has yielded multiple potential candidates, all of which eventually turned out to have properties not fully compatible with those of VRAC. Recently, two groups have independently identified the protein leucine-rich repeats containing 8A ( LRRC8A), belonging to family of proteins ( LRRC8A-E) distantly related to pannexins, as the likely pore-forming subunit of VRAC. In this brief review, we summarize the history of the discovery of VRAC, outline its basic biophysical and pharmacological properties, link these to several cellular functions in which VRAC appears to play important roles, and sketch the amazing search for the molecular identity of this channel. Finally, we describe properties of the LRRC8 proteins, highlight some features of the LRRC8A knockout mouse and discuss the impact of the discovery of LRRC8 as VRAC on future research. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
13. Unfulfilled Expectations Open New Horizons: What Have We Learned about Volume-Regulated Anion Channels in the Kidney?
- Author
-
Pochynyuk O and Palygin O
- Subjects
- Anions, Cell Size, Kidney, Membrane Proteins, Motivation
- Published
- 2022
- Full Text
- View/download PDF
14. Chronic ethanol consumption and HBV induce abnormal lipid metabolism through HBx/SWELL1/arachidonic acid signaling and activate Tregs in HBV-Tg mice.
- Author
-
Liu Z, Wang J, Liu L, Yuan H, Bu Y, Feng J, Liu Y, Yang G, Zhao M, Yuan Y, Zhang H, Yun H, and Zhang X
- Subjects
- Alcohol Drinking genetics, Animals, Cell Line, Tumor, Dinoprostone genetics, Disease Models, Animal, Ethanol adverse effects, Hep G2 Cells, Hepatitis B virology, Hepatitis B virus pathogenicity, Humans, Liver drug effects, Liver virology, Male, Mice, Mice, Inbred BALB C, Mice, Transgenic, RNA, Small Interfering genetics, Signal Transduction drug effects, Signal Transduction genetics, Sp1 Transcription Factor genetics, Spleen drug effects, Spleen virology, Trans-Activators genetics, Alcohol Drinking adverse effects, Arachidonic Acid genetics, Hepatitis B genetics, Lipid Metabolism drug effects, Lipid Metabolism genetics, Membrane Proteins genetics, T-Lymphocytes, Regulatory drug effects
- Abstract
Rationale: Chronic ethanol consumption as a public health problem worldwide boosts the development of chronic liver diseases in hepatitis B virus (HBV)-infected patients. Arachidonic acid metabolite prostaglandin E2 (PGE2) activates regulatory T cells (Tregs) function. Here, we aim to investigate the underlying mechanism by which chronic ethanol consumption enriches the HBV-induced abnormal lipid metabolism and Tregs. Methods: The si-RNAs were used to weaken the expression of SWELL1 in HepG2, HepG2.2.15 and K180 cancer cell lines, followed by RNA sequencing from HepG2 cells. Arachidonic acid metabolite PGE2 and LTD4 were measured by ELISA assay in vivo and in vitro. Western blot analysis and RT-qPCR were used to examine HBx and SWELL1 and transcriptional factor Sp1 in clinical HCC samples and cell lines. The effect of chronic ethanol consumption on Tregs was tested by flow cytometry in HBV-Tg mice. The splenic Tregs were collected and analyzed by RNA sequencing. Results: The cooperative effect of ethanol and HBV in abnormal lipid metabolism was observed in vivo and in vitro . The depression of SWELL1 (or HBx) resulted in the reduction of lipid content and arachidonic acid metabolite, correlating with suppression of relative gene atlas. Ethanol and SWELL1 elevated the levels of PGE2 or LTD4 in the liver of mice and cell lines. Interestingly, the ethanol modulated abnormal lipid metabolism through activating HBx/Sp1/SWELL1/arachidonic acid signaling. Chronic ethanol consumption remarkably increased the population of PBL Tregs and splenic Tregs in HBV-Tg mice, consistently with the enhanced expression of PD-L1 in vivo and in vitro . Mechanically, RNA-seq data showed that multiple genes were altered in the transcriptomic atlas of Tregs sorting from ethanol-fed mice or HBV-Tg mice. Conclusion: The chronic ethanol intake enriches the HBV-enhanced abnormal lipid metabolism through HBx/SWELL1/arachidonic acid signaling and activates Tregs in mice., Competing Interests: Competing Interests: The authors have declared that no competing interest exists., (© The author(s).)
- Published
- 2020
- Full Text
- View/download PDF
15. The identification of a volume-regulated anion channel:an amazing Odyssey
- Author
-
Pedersen, Stine Helene Falsig, Klausen, Thomas Kjær, Nilius, B., Pedersen, Stine Helene Falsig, Klausen, Thomas Kjær, and Nilius, B.
- Published
- 2015
16. More than just a pressure relief valve: physiological roles of volume-regulated LRRC8 anion channels.
- Author
-
Chen L, König B, Liu T, Pervaiz S, Razzaque YS, and Stauber T
- Subjects
- Humans, Ion Channels metabolism, Membrane Proteins metabolism
- Abstract
The volume-regulated anion channel (VRAC) is a key player in the volume regulation of vertebrate cells. This ubiquitously expressed channel opens upon osmotic cell swelling and potentially other cues and releases chloride and organic osmolytes, which contributes to regulatory volume decrease (RVD). A plethora of studies have proposed a wide range of physiological roles for VRAC beyond volume regulation including cell proliferation, differentiation and migration, apoptosis, intercellular communication by direct release of signaling molecules and by supporting the exocytosis of insulin. VRAC was additionally implicated in pathological states such as cancer therapy resistance and excitotoxicity under ischemic conditions. Following extensive investigations, 5 years ago leucine-rich repeat-containing family 8 (LRRC8) heteromers containing LRRC8A were identified as the pore-forming components of VRAC. Since then, molecular biological approaches have allowed further insight into the biophysical properties and structure of VRAC. Heterologous expression, siRNA-mediated downregulation and genome editing in cells, as well as the use of animal models have enabled the assessment of the proposed physiological roles, together with the identification of new functions including spermatogenesis and the uptake of antibiotics and platinum-based cancer drugs. This review discusses the recent molecular biological insights into the physiology of VRAC in relation to its previously proposed roles.
- Published
- 2019
- Full Text
- View/download PDF
17. SWELL1 promotes cell growth and metastasis of hepatocellular carcinoma in vitro and in vivo.
- Author
-
Lu P, Ding Q, Li X, Ji X, Li L, Fan Y, Xia Y, Tian D, and Liu M
- Subjects
- Apoptosis genetics, Carcinoma, Hepatocellular metabolism, Cell Line, Tumor, Cell Movement, Cell Proliferation, Disease Susceptibility, Epithelial-Mesenchymal Transition genetics, Gene Expression Regulation, Neoplastic, Humans, Liver Neoplasms metabolism, MAP Kinase Signaling System, Membrane Proteins metabolism, Neoplasm Metastasis, Neoplasm Staging, Protein Kinase C-alpha metabolism, RNA Interference, RNA, Small Interfering, Signal Transduction, Carcinoma, Hepatocellular etiology, Carcinoma, Hepatocellular pathology, Liver Neoplasms etiology, Liver Neoplasms pathology, Membrane Proteins genetics
- Abstract
Background: SWELL1 was recently demonstrated to be an indispensable part of the volume-regulated anion channel (VRAC). VRAC is reported to participate in cell proliferation, survival, and migration. However, the correlation between SWELL1 and hepatocellular carcinoma (HCC) remains poorly-understood. In this study, we tried to explore the role of SWELL1 in HCC., Methods: Immunohistochemistry and quantitative real-time-PCR (qRT-PCR) was used to measure SWELL1 expression in HCC samples obtained from patients with HCC. The effects of SWELL1 on HCC cell proliferation, apoptosis, and metastasis were analysed by corresponding cytological experiments including Cell Counting Kit-8 (CCK8), colony-forming, 5-ethynyl-2'-deoxyuridine (EdU), cell cycle analysis, TUNEL, Annexin V and PI staining, wound healing, transwell, and so on. BALB/c nude mice were used for the in vivo assays. qRT-PCR and western blotting was performed for molecular mechanisms., Findings: SWELL1 was highly expressed in HCC tissues, and related to the poor prognosis. In vitro, the over-expression of SWELL1 significantly induced cell proliferation and migration, and inhibited apoptosis, whereas suppressing SWELL1 had the opposite effects. Moreover, knockdown of SWELL1 suppressed the growth and metastasis of HCC in vivo. Further experiments revealed that SWELL1 induced cell growth by activating the cyclinD1/CDK2 pathway via the connection with PKCa at the signalling level, and regulated cell migration through the JNK pathway in HCC., Interpretation: SWELL1 acts as a promoter in the growth and metastasis of HCC cells and may be a potential intervention target for HCC. FUND: This work is supported by the National Natural Science Foundation of China (No. 81572422, 81700515)., (Copyright © 2018. Published by Elsevier B.V.)
- Published
- 2019
- Full Text
- View/download PDF
18. Molecular Biology and Physiology of Volume-Regulated Anion Channel (VRAC).
- Author
-
Osei-Owusu J, Yang J, Vitery MDC, and Qiu Z
- Subjects
- Animals, Biological Transport, Humans, Signal Transduction, Anions metabolism, Cell Size, Ion Channels metabolism
- Abstract
The Volume-Regulated Anion Channel (VRAC) is activated by cell swelling and plays a key role in cell volume regulation. VRAC is ubiquitously expressed in vertebrate cells and also implicated in many other physiological and cellular processes including fluid secretion, glutamate release, membrane potential regulation, cell proliferation, migration, and apoptosis. Although its biophysical properties have been well characterized, the molecular identity of VRAC remained a mystery for almost three decades. The field was transformed by recent discoveries showing that the leucine-rich repeat-containing protein 8A (LRRC8A, also named SWELL1) and its four other homologs form heteromeric VRAC channels. The composition of LRRC8 subunits determines channel properties and substrate selectivity of a large variety of different VRACs. Incorporating purified SWELL1-containing protein complexes into lipid bilayers is sufficient to reconstitute channel activities, a finding that supports the decrease in intracellular ionic strength as the mechanism of VRAC activation during cell swelling. Characterization of Swell1 knockout mice uncovers the important role of VRAC in T cell development, pancreatic β-cell glucose-stimulated insulin secretion, and adipocyte metabolic function. The ability to permeate organic osmolytes and metabolites is a major feature of VRAC. The list of VRAC substrates is expected to grow, now also including some cancer drugs and antibiotics even under non-cell swelling conditions. Therefore, a critical role of VRAC in drug resistance and cell-cell communication is emerging. This review summarizes the exciting recent progress on the structure-function relationship and physiology of VRAC and discusses key future questions to be solved., (Copyright © 2018 Elsevier Inc. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.