1
|
Lagrange J, Van De Velde G, Lacolley P, Regnault V, Bascetin R. Underestimated role of macromolecular crowding in bioengineered in vitro models of health and diseases. Mater Today Bio 2025; 32:101772. [PMID: 40331149 PMCID: PMC12053638 DOI: 10.1016/j.mtbio.2025.101772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 04/15/2025] [Accepted: 04/16/2025] [Indexed: 05/08/2025] Open
Abstract
Macromolecular crowding (MMC) is a ubiquitous phenomenon in biological systems that is largely overlooked in bioengineered in vitro cellular models. This comprehensive review examines the significant impact of both intracellular and extracellular MMC on cellular and molecular processes under physiological and pathological conditions. By synthesizing current knowledge and identifying critical gaps in our understanding of MMC, this review highlights the need to incorporate crowding into the development of in vitro models for studying health and diseases, as well as for drug discovery platforms. The pervasive nature of MMC in biological systems underscores its potential importance in various physiological and pathological processes, including protein aggregation disorders, cancer, and vascular diseases. Recognizing the ubiquitous influence of MMC could open new avenues for therapeutic interventions and deepen our understanding of fundamental biological processes.
Collapse
Affiliation(s)
- Jérémy Lagrange
- Université de Lorraine, Inserm, DCAC, F-54000, Nancy, France
- Université de Lorraine, CHRU-Nancy, Inserm, IHU INFINY, F-54000, Nancy, France
| | | | | | | | | |
Collapse
|
2
|
Fidaleo AM, Bach MD, Orbeta S, Abdullaev IF, Martino N, Adam AP, Boulos MA, Dulin NO, Paul AR, Kuo YH, Mongin AA. LRRC8A-containing anion channels promote glioblastoma proliferation via a WNK1/mTORC2-dependent mechanism. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.02.636139. [PMID: 39975357 PMCID: PMC11838495 DOI: 10.1101/2025.02.02.636139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Leucine-rich repeat-containing protein 8A (LRRC8A) is the essential subunit of ubiquitous volume-regulated anion channels (VRACs). LRCC8A is overexpressed in several cancers and promotes negative survival outcomes via a poorly defined mechanism. Here, we explored the role of LRRC8A and VRACs in the progression of glioblastoma (GBM), the most common and deadly primary brain tumor. We found that, as compared to healthy controls, LRRC8A mRNA was strongly upregulated in surgical GBM specimens, patient-derived GBM cell lines, and GBM datasets from The Cancer Genome Atlas (TCGA). Our in-silico analysis indicated that patients belonging to the lowest LRRC8A expression quartile demonstrated a trend for extended life expectancy. In patient-derived GBM cultures, siRNA-driven LRRC8A knockdown reduced cell proliferation and additionally decreased intracellular chloride levels and inhibited activity of mTOR complex 2. The antiproliferative effect of LRRC8A downregulation was recapitulated with a pharmacological inhibitor of VRAC. Our ensuing biochemical and molecular biology analyses established that the LRRC8A-containing VRACs facilitate GBM proliferation via a new mechanism involving non-enzymatic actions of the chloride-sensitive protein kinase WNK1. Accordingly, the chloride-bound WNK1 stimulates mTORC2 and the mTORC2-dependent protein kinases AKT and SGK, which promote proliferation. These findings establish the new mTORC2-centric axis for VRAC dependent regulation of cellular functions and uncover potential targets for GBM intervention.
Collapse
Affiliation(s)
- Antonio M Fidaleo
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, NY, USA
| | - Martin D Bach
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, NY, USA
| | - Shaina Orbeta
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, NY, USA
| | - Iskandar F Abdullaev
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, NY, USA
| | - Nina Martino
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
| | - Alejandro P Adam
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
| | - Mateo A Boulos
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, NY, USA
| | - Nickolai O Dulin
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, The University of Chicago, Chicago, IL, USA
| | - Alexandra R Paul
- Department of Neurosurgery, Albany Medical College, Albany, NY, USA
| | - Yu-Hung Kuo
- Neurosurgery, Luminis Health Anne Arundel Medical Center, Annapolis, MD, USA
| | - Alexander A Mongin
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, NY, USA
| |
Collapse
|
3
|
Naik P, McLeod DS, Bhutto IA, Edwards MM. Regional Alterations in Müller Cell Protein Expression in Human and a Rat Model of Geographic Atrophy. Invest Ophthalmol Vis Sci 2025; 66:21. [PMID: 39918478 PMCID: PMC11809446 DOI: 10.1167/iovs.66.2.21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Accepted: 01/14/2025] [Indexed: 02/12/2025] Open
Abstract
Purpose Despite being crucial to neuronal survival, the role Müller cells play in geographic atrophy (GA) has only recently been considered. We investigated whether Müller cells retain their normal functional profile or form a fibrotic scar when remodeling in human GA eyes and our subretinal sodium iodate (NaIO3) model. Methods Sprague Dawley rats given subretinal injections of NaIO3 (5 mg/mL) were sacrificed at 3 and 12 weeks. Cryosections and retinal flatmounts from rats and cryosections from human GA eyes were stained with antibodies against the Müller cell proteins glutamine synthetase (GS), inwardly rectifying potassium channel 4.1 (Kir4.1), aquaporin 4 (AQP4), cellular retinaldehyde-binding protein 1 (CRALBP), and glial fibrillary acidic protein (GFAP), as well as alpha smooth muscle actin (α-SMA), fibronectin, and collagens I and IV. The immunofluorescence intensity of AQP4 and Kir4.1 was quantified using Image J, and Kir4.1 protein levels were verified by western blot. Results In both human GA eyes and NaIO3-injected rats, Müller cell processes at the external limiting membrane (ELM) descent and in the subretinal membrane exhibited increased GS expression. GFAP was elevated throughout the Müller cells. AQP4 staining at the ELM descent was particularly pronounced throughout the radial processes, including those extending into the subretinal space. In NaIO3-injected rats, perivascular Kir4.1 expression significantly decreased in the atrophic retina, but expression increased in the subretinal glial membrane. α-SMA and extracellular matrix proteins were not detected in the subretinal membrane. Conclusions Our findings underscore the persistence of homeostatic proteins, albeit altered, in Müller cells as they remodel and extend into the subretinal space.
Collapse
Affiliation(s)
- Poonam Naik
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - D. Scott McLeod
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Imran A. Bhutto
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Malia M. Edwards
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| |
Collapse
|
4
|
Feldman LER, Mohapatra S, Jones RT, Scholtes M, Tilton CB, Orman MV, Joshi M, Deiter CS, Broneske TP, Qu F, Gutierrez C, Ye H, Clambey ET, Parker S, Mahmoudi T, Zuiverloon T, Costello JC, Theodorescu D. Regulation of volume-regulated anion channels alters sensitivity to platinum chemotherapy. SCIENCE ADVANCES 2024; 10:eadr9364. [PMID: 39671496 PMCID: PMC11641020 DOI: 10.1126/sciadv.adr9364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 11/08/2024] [Indexed: 12/15/2024]
Abstract
Cisplatin-based chemotherapy is used across many common tumor types, but resistance reduces the likelihood of long-term survival. We previously found the puromycin-sensitive aminopeptidase, NPEPPS, as a druggable driver of cisplatin resistance in vitro and in vivo and in patient-derived organoids. Here, we present a general mechanism where NPEPPS interacts with the volume-regulated anion channels (VRACs) to control cisplatin import into cells and thus regulate cisplatin response across a range of cancer types. We also find the NPEPPS/VRAC gene expression ratio is a predictive measure of cisplatin response in multiple cancer cohorts, showing the broad applicability of this mechanism. Our work describes a specific mechanism of cisplatin resistance, which, given the characteristics of NPEPPS as a drug target, has the potential to improve cancer patient outcomes. In addition, we describe an intracellular mechanism regulating VRAC activity, which is critical for volume regulation in normal cells - a finding with functional implications beyond cancer.
Collapse
Affiliation(s)
| | - Saswat Mohapatra
- Cedars-Sinai Samuel Oschin Comprehensive Cancer Institute, Los Angeles, CA, USA
| | - Robert T. Jones
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Mathijs Scholtes
- Department of Urology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Charlene B. Tilton
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Michael V. Orman
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Molishree Joshi
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Functional Genomics Facility, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Cailin S. Deiter
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Travis P. Broneske
- Functional Genomics Facility, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Fangyuan Qu
- Cedars-Sinai Samuel Oschin Comprehensive Cancer Institute, Los Angeles, CA, USA
| | - Corazon Gutierrez
- Cedars-Sinai Samuel Oschin Comprehensive Cancer Institute, Los Angeles, CA, USA
| | - Huihui Ye
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Eric T. Clambey
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Sarah Parker
- Smidt Heart Institute & Advanced Clinical Biosystems Research Institute, Cedars Sinai Medical Center, Los Angeles, CA, USA
| | - Tokameh Mahmoudi
- Department of Urology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, Netherlands
- Department of Biochemistry, Erasmus University Medical Center, Rotterdam, Netherlands
- Department of Pathology, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Tahlita Zuiverloon
- Department of Urology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, Netherlands
| | - James C. Costello
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- University of Colorado Comprehensive Cancer Center, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Dan Theodorescu
- Cedars-Sinai Samuel Oschin Comprehensive Cancer Institute, Los Angeles, CA, USA
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Urology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| |
Collapse
|
5
|
Biswas P, Roy P, Jana S, Ray D, Das J, Chaudhuri B, Basunia RR, Sinha B, Sinha DK. Exploring the role of macromolecular crowding and TNFR1 in cell volume control. eLife 2024; 13:e92719. [PMID: 39297502 PMCID: PMC11581439 DOI: 10.7554/elife.92719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 09/18/2024] [Indexed: 11/22/2024] Open
Abstract
The excessive cosolute densities in the intracellular fluid create a physicochemical condition called macromolecular crowding (MMC). Intracellular MMC entropically maintains the biochemical thermodynamic equilibria by favoring associative reactions while hindering transport processes. Rapid cell volume shrinkage during extracellular hypertonicity elevates the MMC and disrupts the equilibria, potentially ushering cell death. Consequently, cells actively counter the hypertonic stress through regulatory volume increase (RVI) and restore the MMC homeostasis. Here, we establish fluorescence anisotropy of EGFP as a reliable tool for studying cellular MMC and explore the spatiotemporal dynamics of MMC during cell volume instabilities under multiple conditions. Our studies reveal that the actin cytoskeleton enforces spatially varying MMC levels inside adhered cells. Within cell populations, MMC is uncorrelated with nuclear DNA content but anti-correlated with the cell spread area. Although different cell lines have statistically similar MMC distributions, their responses to extracellular hypertonicity vary. The intensity of the extracellular hypertonicity determines a cell's ability for RVI, which correlates with nuclear factor kappa beta (NFkB) activation. Pharmacological inhibition and knockdown experiments reveal that tumor necrosis factor receptor 1 (TNFR1) initiates the hypertonicity-induced NFkB signaling and RVI. At severe hypertonicities, the elevated MMC amplifies cytoplasmic microviscosity and hinders receptor interacting protein kinase 1 (RIPK1) recruitment at the TNFR1 complex, incapacitating the TNFR1-NFkB signaling and consequently, RVI. Together, our studies unveil the involvement of TNFR1-NFkB signaling in modulating RVI and demonstrate the pivotal role of MMC in determining cellular osmoadaptability.
Collapse
Affiliation(s)
- Parijat Biswas
- School of Biological Sciences, Indian Association for the Cultivation of ScienceKolkataIndia
| | - Priyanka Roy
- School of Biological Sciences, Indian Association for the Cultivation of ScienceKolkataIndia
| | - Subhamoy Jana
- School of Biological Sciences, Indian Association for the Cultivation of ScienceKolkataIndia
| | - Dipanjan Ray
- School of Biological Sciences, Indian Association for the Cultivation of ScienceKolkataIndia
| | - Jibitesh Das
- Department of Biological Sciences, Indian Institute of Science Education and Research KolkataKolkataIndia
| | - Bipasa Chaudhuri
- School of Biological Sciences, Indian Association for the Cultivation of ScienceKolkataIndia
| | - Ridita Ray Basunia
- School of Biological Sciences, Indian Association for the Cultivation of ScienceKolkataIndia
| | - Bidisha Sinha
- Department of Biological Sciences, Indian Institute of Science Education and Research KolkataKolkataIndia
| | - Deepak Kumar Sinha
- School of Biological Sciences, Indian Association for the Cultivation of ScienceKolkataIndia
| |
Collapse
|
6
|
Wagh SB, Berthold D, Majeed I, Lewis LK, Schilter D, Mertens B, Evidente A, van Otterlo WAL, Mathieu V, Kornienko A. Sphaeropsidin A C15-C16 Cross-Metathesis Analogues with Potent Anticancer Activity. ChemMedChem 2024; 19:e202400288. [PMID: 38895989 PMCID: PMC11645891 DOI: 10.1002/cmdc.202400288] [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: 04/21/2024] [Revised: 06/10/2024] [Accepted: 06/10/2024] [Indexed: 06/21/2024]
Abstract
We recently discovered that sphaeropsidin A (SphA), a fungal metabolite from Diplodia cupressi, overcomes apoptosis resistance in cancer cells by inducing cellular shrinkage by impairing regulatory volume increase. Previously, we prepared a pyrene-conjugated derivative of SphA by a cross-metathesis reaction involving the phytotoxin's C15,C16-alkene. This derivative's evaluation in a cancer cell panel revealed a significant increase in potency, with the IC50 values 5-10× lower than those displayed by the original natural product. Herein, we describe the preparation and anticancer evaluation of fifteen novel C15,C16-alkene cross-metathesis analogues in which the pyrene moiety was replaced with other aromatic or non-aromatic hydrophobic groups. The idea for this replacement was to prepare a family of compounds that would not be predicted to be mutagenic compared with the original pyrene analogue. We predict several of our new compounds to be non-mutagenic, while retaining the high potency of the original pyrene-containing analogues. Examples of these potential lead compounds included those containing pentamethylphenyl and triphenylethylene pendant groups. As an additional feature of the current investigation, we prepared several deuterated pyrene-containing compounds to overcome intellectual property issues associated with non-patentability of the original pyrene derivative.
Collapse
Affiliation(s)
- Sachin B. Wagh
- Department of Chemistry and Biochemistry, Texas State University, San Marcos, Texas 78666, USA
| | - Dino Berthold
- Department of Chemistry and Polymer Sciences, Stellenbosch University, Stellenbosch, Matieland 7602, Western Cape, South Africa
| | - Iram Majeed
- Department of Chemistry and Biochemistry, Texas State University, San Marcos, Texas 78666, USA
| | - L. Kevin Lewis
- Department of Chemistry and Biochemistry, Texas State University, San Marcos, Texas 78666, USA
| | - David Schilter
- Department of Chemistry and Biochemistry, Texas State University, San Marcos, Texas 78666, USA
| | - Birgit Mertens
- Sciensano, Rue Juliette Wytsmanstraat 14, 1050 Brussels, Belgium
| | - Antonio Evidente
- Institute of Sciences of Food Production, National Research Council, Via Amendola 122/O, 70125 Bari, Italy
| | - Willem A. L. van Otterlo
- Department of Chemistry and Polymer Sciences, Stellenbosch University, Stellenbosch, Matieland 7602, Western Cape, South Africa
| | - Véronique Mathieu
- Department of Pharmacotherapy and Pharmaceutics, Chemistry and Biochemistry, Faculté de Pharmacie, Université Libre de Bruxelles, Brussels, Belgium
- ULB Cancer Research Center, U-CRC, Université Libre de Bruxelles, Brussels, Belgium
| | - Alexander Kornienko
- Department of Chemistry and Biochemistry, Texas State University, San Marcos, Texas 78666, USA
| |
Collapse
|
7
|
Centrone M, Saltarella I, D'Agostino M, Ranieri M, Venneri M, Di Mise A, Simone L, Pisani F, Valenti G, Frassanito MA, Tamma G. RhoB plays a central role in hyperosmolarity-induced cell shrinkage in renal cells. J Cell Physiol 2024; 239:e31343. [PMID: 38946197 DOI: 10.1002/jcp.31343] [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: 11/28/2023] [Revised: 06/06/2024] [Accepted: 06/11/2024] [Indexed: 07/02/2024]
Abstract
The small Rho GTP-binding proteins are important cell morphology, function, and apoptosis regulators. Unlike other Rho proteins, RhoB can be subjected to either geranylgeranylation (RhoB-GG) or farnesylation (RhoB-F), making that the only target of the farnesyltransferase inhibitor (FTI). Fluorescence resonance energy transfer experiments revealed that RhoB is activated by hyperosmolarity. By contrast, hyposmolarity did not affect RhoB activity. Interestingly, treatment with farnesyltransferase inhibitor-277 (FTI-277) decreased the cell size. To evaluate whether RhoB plays a role in volume reduction, renal collecting duct MCD4 cells and Human Kidney, HK-2 were transiently transfected with RhoB-wildtype-Enhance Green Fluorescence Protein (RhoB-wt-EGFP) and RhoB-CLLL-EGFP which cannot undergo farnesylation. A calcein-based fluorescent assay revealed that hyperosmolarity caused a significant reduction of cell volume in mock and RhoB-wt-EGFP-expressing cells. By contrast, cells treated with FTI-277 or expressing the RhoB-CLLL-EGFP mutant did not properly respond to hyperosmolarity with respect to mock and RhoB-wt-EGFP expressing cells. These findings were further confirmed by 3D-LSCM showing that RhoB-CLLL-EGFP cells displayed a significant reduction in cell size compared to cells expressing RhoB-wt-EGFP. Moreover, flow cytometry analysis revealed that RhoB-CLLL-EGFP expressing cells as well as FTI-277-treated cells showed a significant increase in cell apoptosis. Together, these data suggested that: (i) RhoB is sensitive to hyperosmolarity and not to hyposmolarity; (ii) inhibition of RhoB farnesylation associates with an increase in cell apoptosis, likely suggesting that RhoB might be a paramount player controlling apoptosis by interfering with responses to cell volume change.
Collapse
Affiliation(s)
- Mariangela Centrone
- Department of Biosciences Biotechnologies and Environment, University of Bari Aldo Moro, Bari, Italy
| | - Ilaria Saltarella
- Department of Precision and Regenerative Medicine and Ionian Area, Section of Pharmacology, School of Medicine, University of Bari Aldo Moro, Bari, Italy
| | - Mariagrazia D'Agostino
- Department of Biosciences Biotechnologies and Environment, University of Bari Aldo Moro, Bari, Italy
| | - Marianna Ranieri
- Department of Biosciences Biotechnologies and Environment, University of Bari Aldo Moro, Bari, Italy
| | - Maria Venneri
- Istituti Clinici Scientifici Maugeri SPA SB IRCCS, Bari, Italy
| | - Annarita Di Mise
- Department of Biosciences Biotechnologies and Environment, University of Bari Aldo Moro, Bari, Italy
| | - Laura Simone
- Fondazione IRCCS Casa Sollievo della Sofferenza, Cancer Stem Cells Unit, San Giovanni Rotondo, Italy
| | - Francesco Pisani
- Department of Biosciences Biotechnologies and Environment, University of Bari Aldo Moro, Bari, Italy
| | - Giovanna Valenti
- Department of Biosciences Biotechnologies and Environment, University of Bari Aldo Moro, Bari, Italy
| | - Maria A Frassanito
- Department of Precision and Regenerative Medicine and Ionian Area, Section of Clinical Pathology, University of Bari Aldo Moro, Bari, Italy
| | - Grazia Tamma
- Department of Biosciences Biotechnologies and Environment, University of Bari Aldo Moro, Bari, Italy
| |
Collapse
|
8
|
Muhammed M, Tengku Azizan TRPB, Mohd Noor MH, Abu Hassim H, Che'Amat A, Saleh AB, Han MHW, Ab Latip MQ. In vitro evaluation of oral contraceptives on long-tailed macaque ( Macaca fascicularis) primary ovarian cells. Heliyon 2024; 10:e36185. [PMID: 39247268 PMCID: PMC11378886 DOI: 10.1016/j.heliyon.2024.e36185] [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: 07/28/2023] [Revised: 08/12/2024] [Accepted: 08/12/2024] [Indexed: 09/10/2024] Open
Abstract
Hormonal contraception has been advocated as an alternative population control method for the long-tailed macaque population, which has increased exponentially due to anthropogenic changes and incidental food subsidies from human food waste. Risks of increased zoonosis and conflict are imminent if the population growth of long-tailed macaques is unchecked. However, there's a gap in the literature about the effect of hormonal contraceptives on long-tailed macaque reproductive tissues cell line. The present study aims to investigate the effect of oral contraceptives (Nordette, Noriday, and Ella) on long-tailed macaque ovarian cells. We determine the cell viability and cytotoxicity as well as the morphological changes of the drugs on long-tailed macaque ovarian cells using the MTT assay, Acridine orange/propidium iodide double staining method, morphological examination, and the 4, 6-diamidino-2-phenylindole (DAPI) staining method. For the MTT assay, The drugs were dissolved in culture media before use to have a concentration ranging from 0.5 μg/mL, 2.5 μg/mL, 0.125 μg/mL, 0.0625 μg/mL, and 0.0315 μg/mL to have three replicates for each treatment. In contrast, the concentration of 0.0315 μg/mL was used for the morphological and histopathological analysis. The result of the study indicates that human oral contraceptives (Nordette, Noriday, and Ella) inhibit the growth of long-tailed macaque ovarian cells and induce apoptosis in a concentration- and time-dependent manner (at a concentration of 0.0315 μg/mL and an IC50 lower than 10 μg/mL), With a statistically significant value of ****P < 0.001 for each drug compared to the negative control. The result of the present study contributes toward addressing the gap in the literature on the effect of oral contraceptives in long-tailed macaque ovarian cells. Hence, we conclude that human oral contraceptives (Nordette, Noriday, and Ella) are safe and effective in long-tailed macaque ovarian cells as such could be used to develop non-invasive oral contraceptives for controlling the population of long-tailed macaques as an alternative population control method.
Collapse
Affiliation(s)
- Mikail Muhammed
- Department of Veterinary Preclinical Sciences, Faculty of Veterinary Medicine, Universiti Putra Malaysia, 43400, Serdang, Selangor Darul-Ehsan, Malaysia
- Department of Veterinary Clinical Studies, Faculty of Veterinary Medicine, Universiti Malaysia Kelantan, 1610, Pengkalan Chepa, Kelantan, Malaysia
| | - Tengku Rinalfi Putra Bin Tengku Azizan
- Department of Veterinary Preclinical Sciences, Faculty of Veterinary Medicine, Universiti Putra Malaysia, 43400, Serdang, Selangor Darul-Ehsan, Malaysia
| | - Mohd Hezmee Mohd Noor
- Department of Veterinary Preclinical Sciences, Faculty of Veterinary Medicine, Universiti Putra Malaysia, 43400, Serdang, Selangor Darul-Ehsan, Malaysia
| | - Hasliza Abu Hassim
- Department of Veterinary Preclinical Sciences, Faculty of Veterinary Medicine, Universiti Putra Malaysia, 43400, Serdang, Selangor Darul-Ehsan, Malaysia
- Institute of Tropical Agriculture and Food Security (ITAFoS), Universiti Putra Malaysia, 43400, Serdang, Selangor Darul-Ehsan, Malaysia
| | - Azlan Che'Amat
- Department of Veterinary Clinical Studies, Faculty of Veterinary Medicine, Universiti Putra Malaysia, 43400, Serdang, Selangor Darul-Ehsan, Malaysia
| | - Annas Bin Saleh
- Department of Veterinary Laboratory Diagnostic, Faculty of Veterinary Medicine, Universiti Putra Malaysia, 43400, Serdang, Selangor Darul-Ehsan, Malaysia
- Institute of Tropical Agriculture and Food Security (ITAFoS), Universiti Putra Malaysia, 43400, Serdang, Selangor Darul-Ehsan, Malaysia
| | - Mark Hiew Wen Han
- Department of Veterinary Clinical Studies, Faculty of Veterinary Medicine, Universiti Putra Malaysia, 43400, Serdang, Selangor Darul-Ehsan, Malaysia
| | - Mohd Qayyum Ab Latip
- Department of Veterinary Preclinical Sciences, Faculty of Veterinary Medicine, Universiti Putra Malaysia, 43400, Serdang, Selangor Darul-Ehsan, Malaysia
| |
Collapse
|
9
|
Caro L, Wei AD, Thomas CA, Posch G, Uremis A, Franzi MC, Abell SJ, Laszlo AH, Gundlach JH, Ramirez JM, Ailion M. Mechanism of an animal toxin-antidote system. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.11.598564. [PMID: 38915716 PMCID: PMC11195288 DOI: 10.1101/2024.06.11.598564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Toxin-antidote systems are selfish genetic elements composed of a linked toxin and antidote. The peel-1 zeel-1 toxin-antidote system in C. elegans consists of a transmembrane toxin protein PEEL-1 which acts cell autonomously to kill cells. Here we investigate the molecular mechanism of PEEL-1 toxicity. We find that PEEL-1 requires a small membrane protein, PMPL-1, for toxicity. Together, PEEL-1 and PMPL-1 are sufficient for toxicity in a heterologous system, HEK293T cells, and cause cell swelling and increased cell permeability to monovalent cations. Using purified proteins, we show that PEEL-1 and PMPL-1 allow ion flux through lipid bilayers and generate currents which resemble ion channel gating. Our work suggests that PEEL-1 kills cells by co-opting PMPL-1 and creating a cation channel.
Collapse
Affiliation(s)
- Lews Caro
- Molecular and Cellular Biology Ph.D. Program, University of Washington; Seattle, WA 98195, USA
- Department of Biochemistry, University of Washington; Seattle, WA 91895, USA
| | - Aguan D. Wei
- Norcliffe Foundation Center for Integrative Brain Research, Seattle Children’s Research Institute; Seattle, WA 98101, USA
| | | | - Galen Posch
- Department of Biochemistry, University of Washington; Seattle, WA 91895, USA
| | - Ahmet Uremis
- Department of Biochemistry, University of Washington; Seattle, WA 91895, USA
| | | | - Sarah J. Abell
- Department of Physics, University of Washington; Seattle, WA 91895, USA
| | - Andrew H. Laszlo
- Department of Physics, University of Washington; Seattle, WA 91895, USA
| | - Jens H. Gundlach
- Department of Physics, University of Washington; Seattle, WA 91895, USA
| | - Jan-Marino Ramirez
- Norcliffe Foundation Center for Integrative Brain Research, Seattle Children’s Research Institute; Seattle, WA 98101, USA
- Department of Neurological Surgery, University of Washington School of Medicine; Seattle, WA 98104, USA
| | - Michael Ailion
- Molecular and Cellular Biology Ph.D. Program, University of Washington; Seattle, WA 98195, USA
- Department of Biochemistry, University of Washington; Seattle, WA 91895, USA
| |
Collapse
|
10
|
Untiet V. Astrocytic chloride regulates brain function in health and disease. Cell Calcium 2024; 118:102855. [PMID: 38364706 DOI: 10.1016/j.ceca.2024.102855] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 02/02/2024] [Accepted: 02/06/2024] [Indexed: 02/18/2024]
Abstract
Chloride ions (Cl-) play a pivotal role in synaptic inhibition in the central nervous system, primarily mediated through ionotropic mechanisms. A recent breakthrough emphathizes the significant influence of astrocytic intracellular chloride concentration ([Cl-]i) regulation, a field still in its early stages of exploration. Typically, the [Cl-]i in most animal cells is maintained at lower levels than the extracellular chloride [Cl-]o, a critical balance to prevent cell swelling due to osmotic pressure. Various Cl- transporters are expressed differently across cell types, fine-tuning the [Cl-]i, while Cl- gradients are utilised by several families of Cl- channels. Although the passive distribution of ions within cells is governed by basic biophysical principles, astrocytes actively expend energy to sustain [Cl-]i at much higher levels than those achieved passively, and much higher than neuronal [Cl-]i. Beyond the role in volume regulation, astrocytic [Cl-]i is dynamically linked to brain states and influences neuronal signalling in actively behaving animals. As a vital component of brain function, astrocytic [Cl-]i also plays a role in the development of disorders where inhibitory transmission is disrupted. This review synthesises the latest insights into astrocytic [Cl-]i, elucidating its role in modulating brain function and its implications in various pathophysiological conditions.
Collapse
Affiliation(s)
- Verena Untiet
- Division of Glial Disease and Therapeutics, Center for Translational Neuromedicine, University of Copenhagen, 2200 Copenhagen, Denmark.
| |
Collapse
|
11
|
Okada Y. Physiology of the volume-sensitive/regulatory anion channel VSOR/VRAC. Part 1: from its discovery and phenotype characterization to the molecular entity identification. J Physiol Sci 2024; 74:3. [PMID: 38238667 PMCID: PMC10795261 DOI: 10.1186/s12576-023-00897-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 12/27/2023] [Indexed: 01/22/2024]
Abstract
The volume-sensitive outwardly rectifying or volume-regulated anion channel, VSOR/VRAC, which was discovered in 1988, is expressed in most vertebrate cell types and is essentially involved in cell volume regulation after swelling and in the induction of cell death. This series of review articles describes what is already known and what remains to be uncovered about the functional and molecular properties as well as the physiological and pathophysiological roles of VSOR/VRAC. This Part 1 review article describes, from the physiological standpoint, first its discovery and significance in cell volume regulation, second its phenotypical properties, and third its molecular identification. Although the pore-forming core molecules and the volume-sensing subcomponent of VSOR/VRAC were identified as LRRC8 members and TRPM7 in 2014 and 2021, respectively, it is stressed that the identification of the molecular entity of VSOR/VRAC is still not complete enough to explain the full set of phenotypical properties.
Collapse
Affiliation(s)
- Yasunobu Okada
- National Institute for Physiological Sciences, 5-1 Higashiyama, Myodaiji, Okazaki, Aichi, 444-8787, Japan.
- Department of Integrative Physiology, Graduate School of Medicine, Akita University, Akita, Japan.
- Department of Physiology, School of Medicine, Aichi Medical University, Nagakute, Japan.
- Department of Physiology, Kyoto Prefectural University of Medicine, Kyoto, Japan.
- Cardiovascular Research Institute, Yokohama City University, Yokohama, Japan.
- Graduate University for Advanced Studies (SOKENDAI), Hayama, Kanagawa, Japan.
| |
Collapse
|
12
|
Kostritskaia Y, Klüssendorf M, Pan YE, Hassani Nia F, Kostova S, Stauber T. Physiological Functions of the Volume-Regulated Anion Channel VRAC/LRRC8 and the Proton-Activated Chloride Channel ASOR/TMEM206. Handb Exp Pharmacol 2024; 283:181-218. [PMID: 37468723 DOI: 10.1007/164_2023_673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/21/2023]
Abstract
Volume-regulated anion channels (VRACs) and the acid-sensitive outwardly rectifying anion channel (ASOR) mediate flux of chloride and small organic anions. Although known for a long time, they were only recently identified at the molecular level. VRACs are heteromers consisting of LRRC8 proteins A to E. Combining the essential LRRC8A with different LRRC8 paralogues changes key properties of VRAC such as conductance or substrate selectivity, which is how VRACs are involved in multiple physiological functions including regulatory volume decrease, cell proliferation and migration, cell death, purinergic signalling, fat and glucose metabolism, insulin signalling, and spermiogenesis. VRACs are also involved in pathological conditions, such as the neurotoxic release of glutamate and aspartate. Certain VRACs are also permeable to larger, organic anions, including antibiotics and anti-cancer drugs, making them an interesting therapeutic target. ASOR, also named proton-activated chloride channel (PAC), is formed by TMEM206 homotrimers on the plasma membrane and on endosomal compartments where it mediates chloride flux in response to extracytosolic acidification and plays a role in the shrinking and maturation of macropinosomes. ASOR has been shown to underlie neuronal swelling which causes cell death after stroke as well as promoting the metastasis of certain cancers, making them intriguing therapeutic targets as well.
Collapse
Affiliation(s)
- Yulia Kostritskaia
- Institute for Molecular Medicine, MSH Medical School Hamburg, Hamburg, Germany
| | - Malte Klüssendorf
- Institute for Molecular Medicine, MSH Medical School Hamburg, Hamburg, Germany
| | - Yingzhou Edward Pan
- Institute for Molecular Medicine, MSH Medical School Hamburg, Hamburg, Germany
| | - Fatemeh Hassani Nia
- Institute for Molecular Medicine, MSH Medical School Hamburg, Hamburg, Germany
| | - Simona Kostova
- Institute for Molecular Medicine, MSH Medical School Hamburg, Hamburg, Germany
| | - Tobias Stauber
- Institute for Molecular Medicine, MSH Medical School Hamburg, Hamburg, Germany.
| |
Collapse
|
13
|
Toshtemirova G, Rustamova S, Tsiferova N, Maksimcheva G, Merzlyak P, Kurbannazarova R, Sabirov R. Effect of pinocembrin on thymocyte proliferation and death. JOURNAL OF HERBMED PHARMACOLOGY 2024; 13:137-143. [DOI: 10.34172/jhp.2024.48225] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 10/14/2023] [Indexed: 05/19/2025] Open
Abstract
Introduction: Cell volume regulation is critical for cellular proliferation and death. Pinocembrin effectively suppresses the volume regulation in thymocytes under hypoosmotic stress by blocking the volume-sensitive anion channel. This study aims to evaluate the effects of this flavonoid on thymocyte proliferation and death. Methods: Thymocytes were cultured in RPMI-1640 medium supplemented with 10% fetal bovine serum, and the cell number was determined by cloud-based automated cell counting (Corning). Necrotic and apoptotic cell death were evaluated by propidium iodide- and annexin V-staining, respectively. Results: Pinocembrin at 10–50 μM caused suppression of primary cultured thymocyte proliferation with a half-maximal effect of 28.4 ± 0.2 μM. The cell counts did not fall below the control level at the doses of 100–150 μM. The fraction of spontaneously necrotic cells was ~26% of the total population and increased to ~51% in the presence of dexamethasone. The fraction of spontaneously apoptotic cells increased by this glucocorticoid from 3.6% to 16.7%. Pinocembrin protected thymocytes from necrosis both in spontaneous and dexamethasone-induced death, reducing the fraction of necrotic cells by ~40–50% at 150 μM. Pinocembrin attenuated dexamethasone-induced apoptotic death, reducing the fraction of annexin-positive cells to the control (spontaneous) level. Conclusion: Our results suggest that pinocembrin arrests thymocyte proliferation without essential killing. Under conditions of massive death (e.g., during inflammation, when the level of glucocorticoids increases sharply both physiologically and as a result of pharmacotherapy), pinocembrin protects immuno-competent cells from necrotic and apoptotic death.
Collapse
Affiliation(s)
- Gulnoza Toshtemirova
- Institute of Biophysics and Biochemistry, National University of Uzbekistan, Tashkent, Uzbekistan
| | - Sarvinoz Rustamova
- Institute of Biophysics and Biochemistry, National University of Uzbekistan, Tashkent, Uzbekistan
| | - Nargiza Tsiferova
- Institute of Biophysics and Biochemistry, National University of Uzbekistan, Tashkent, Uzbekistan
- Center for Advanced Technologies, Ministry of Higher Education, Science and Innovation of the Republic of Uzbekistan
| | - Galina Maksimcheva
- Institute of Biophysics and Biochemistry, National University of Uzbekistan, Tashkent, Uzbekistan
- Center for Advanced Technologies, Ministry of Higher Education, Science and Innovation of the Republic of Uzbekistan
| | - Petr Merzlyak
- Institute of Biophysics and Biochemistry, National University of Uzbekistan, Tashkent, Uzbekistan
| | - Ranohon Kurbannazarova
- Institute of Biophysics and Biochemistry, National University of Uzbekistan, Tashkent, Uzbekistan
- Department of Biophysics, National University of Uzbekistan, Tashkent, Uzbekistan
| | - Ravshan Sabirov
- Institute of Biophysics and Biochemistry, National University of Uzbekistan, Tashkent, Uzbekistan
- Department of Biophysics, National University of Uzbekistan, Tashkent, Uzbekistan
| |
Collapse
|
14
|
Yoshie S, Murono S, Hazama A. Approach for Elucidating the Molecular Mechanism of Epithelial to Mesenchymal Transition in Fibrosis of Asthmatic Airway Remodeling Focusing on Cl - Channels. Int J Mol Sci 2023; 25:289. [PMID: 38203460 PMCID: PMC10779031 DOI: 10.3390/ijms25010289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 12/15/2023] [Accepted: 12/16/2023] [Indexed: 01/12/2024] Open
Abstract
Airway remodeling caused by asthma is characterized by structural changes of subepithelial fibrosis, goblet cell metaplasia, submucosal gland hyperplasia, smooth muscle cell hyperplasia, and angiogenesis, leading to symptoms such as dyspnea, which cause marked quality of life deterioration. In particular, fibrosis exacerbated by asthma progression is reportedly mediated by epithelial-mesenchymal transition (EMT). It is well known that the molecular mechanism of EMT in fibrosis of asthmatic airway remodeling is closely associated with several signaling pathways, including the TGF-β1/Smad, TGF-β1/non-Smad, and Wnt/β-catenin signaling pathways. However, the molecular mechanism of EMT in fibrosis of asthmatic airway remodeling has not yet been fully clarified. Given that Cl- transport through Cl- channels causes passive water flow and consequent changes in cell volume, these channels may be considered to play a key role in EMT, which is characterized by significant morphological changes. In the present article, we highlight how EMT, which causes fibrosis and carcinogenesis in various tissues, is strongly associated with activation or inactivation of Cl- channels and discuss whether Cl- channels can lead to elucidation of the molecular mechanism of EMT in fibrosis of asthmatic airway remodeling.
Collapse
Affiliation(s)
- Susumu Yoshie
- Department of Cellular and Integrative Physiology, Graduate School of Medicine, Fukushima Medical University, Fukushima 960-1295, Japan
| | - Shigeyuki Murono
- Department of Otolaryngology Head and Neck Surgery, Graduate School of Medicine, Fukushima Medical University, Fukushima 960-1295, Japan
| | - Akihiro Hazama
- Department of Cellular and Integrative Physiology, Graduate School of Medicine, Fukushima Medical University, Fukushima 960-1295, Japan
| |
Collapse
|
15
|
Mao W, Wang Z, Wen S, Lin Y, Gu J, Sun J, Wang H, Cao Q, Xu Y, Xu X, Cai X. LRRC8A promotes Glaesserella parasuis cytolethal distending toxin-induced p53-dependent apoptosis in NPTr cells. Virulence 2023; 14:2287339. [PMID: 38018865 PMCID: PMC10732598 DOI: 10.1080/21505594.2023.2287339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Accepted: 11/19/2023] [Indexed: 11/30/2023] Open
Abstract
Glaesserella parasuis is an early colonizer of the swine upper respiratory tract and can break through the respiratory barrier for further invasion. However, the mechanisms underlying G. parasuis increases epithelial barrier permeability remain unclear. This study demonstrates that G. parasuis cytolethal distending toxin (CDT) induces p53-dependent apoptosis in new-born piglet tracheal (NPTr) cells. Moreover, we report for the first time that leucine-rich repeat-containing protein 8A (LRRC8A), an essential subunit of the volume-regulated anion channel (VRAC), involves in apoptosis of NPTr cells mediated by G. parasuis CDT. Pharmacological inhibition of VRAC with either PPQ-102 or NS3728 largely attenuated CDT-induced apoptosis in NPTr cells. Additionally, experiments with cells knocked down for LRRC8A using small interfering ribonucleic acid (siRNA) or knocked out LRRC8A using CRISPR/Cas9 technology showed a significant reduction in CDT-induced apoptosis. Conversely, re-expression of Sus scrofa LRRC8A in LRRC8A-/- NPTr cells efficiently complemented the CDT-induced apoptosis. In summary, these findings suggest that LRRC8A is pivotal for G. parasuis CDT-induced apoptosis, providing novel insights into the mechanism of apoptosis caused by CDT.
Collapse
Affiliation(s)
- Weiting Mao
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Zhichao Wang
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Siting Wen
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
| | - Yan Lin
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
| | - Jiayun Gu
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
| | - Ju Sun
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
| | - Huan Wang
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
| | - Qi Cao
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
| | - Yindi Xu
- Institute of Animal Husbandry and Veterinary Research, Henan Academy of Agricultural Sciences, Zhengzhou, China
| | - Xiaojuan Xu
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
| | - Xuwang Cai
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
| |
Collapse
|
16
|
Wu X, Yi X, Zhao B, Zhi Y, Xu Z, Cao Y, Cao X, Pang J, Yung KKL, Zhang S, Liu S, Zhou P. The volume regulated anion channel VRAC regulates NLRP3 inflammasome by modulating itaconate efflux and mitochondria function. Pharmacol Res 2023; 198:107016. [PMID: 38006980 DOI: 10.1016/j.phrs.2023.107016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 11/19/2023] [Accepted: 11/22/2023] [Indexed: 11/27/2023]
Abstract
The NLRP3 inflammasome is a supramolecular complex that is linked to sterile and pathogen-dependent inflammation, and its excessive activation underlies many diseases. Ion flux disturbance and cell volume regulation are both reported to mediate NLRP3 inflammasome activation, but the underlying orchestrating signaling remains not fully elucidated. The volume-regulated anion channel (VRAC), formed by LRRC8 proteins, is an important constituent that controls cell volume by permeating chloride and organic osmolytes in response to cell swelling. We now demonstrate that Lrrc8a, the essential component of VRAC, plays a central and specific role in canonical NLRP3 inflammasome activation. Moreover, VRAC acts downstream of K+ efflux for NLRP3 stimuli that require K+ efflux. Mechanically, our data demonstrate that VRAC modulates itaconate efflux and damaged mitochondria production for NLRP3 inflammasome activation. Further in vivo experiments show mice with Lrrc8a deficiency in myeloid cells were protected from lipopolysaccharides (LPS)-induced endotoxic shock. Taken together, this work identifies VRAC as a key regulator of NLRP3 inflammasome and innate immunity by regulating mitochondrial adaption for macrophage activation and highlights VRAC as a prospective drug target for the treatment of NLRP3 inflammasome and itaconate related diseases.
Collapse
Affiliation(s)
- Xiaoyan Wu
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Xin Yi
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Boxin Zhao
- Department of Pharmacy, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yuanxing Zhi
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Ziwei Xu
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Ying Cao
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Xiong Cao
- Key Laboratory of Mental Health of the Ministry of Education, Key Laboratory of Psychiatric Disorders of Guangdong Province, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Jianxin Pang
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Ken Kin Lam Yung
- Department of Science and Environmental Studies, the Education University of Hong Kong, Hong Kong, China
| | - Shiqing Zhang
- JNU-HKUST Joint Laboratory for Neuroscience and Innovative Drug Research, College of Pharmacy, Jinan University, Guangzhou, China.
| | - Shuwen Liu
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China.
| | - Pingzheng Zhou
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China.
| |
Collapse
|
17
|
Marunaka Y. Physiological roles of chloride ions in bodily and cellular functions. J Physiol Sci 2023; 73:31. [PMID: 37968609 PMCID: PMC10717538 DOI: 10.1186/s12576-023-00889-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 10/31/2023] [Indexed: 11/17/2023]
Abstract
Physiological roles of Cl-, a major anion in the body, are not well known compared with those of cations. This review article introduces: (1) roles of Cl- in bodily and cellular functions; (2) the range of cytosolic Cl- concentration ([Cl-]c); (3) whether [Cl-]c could change with cell volume change under an isosmotic condition; (4) whether [Cl-]c could change under conditions where multiple Cl- transporters and channels contribute to Cl- influx and efflux in an isosmotic state; (5) whether the change in [Cl-]c could be large enough to act as signals; (6) effects of Cl- on cytoskeletal tubulin polymerization through inhibition of GTPase activity and tubulin polymerization-dependent biological activity; (7) roles of cytosolic Cl- in cell proliferation; (8) Cl--regulatory mechanisms of ciliary motility; (9) roles of Cl- in sweet/umami taste receptors; (10) Cl--regulatory mechanisms of with-no-lysine kinase (WNK); (11) roles of Cl- in regulation of epithelial Na+ transport; (12) relationship between roles of Cl- and H+ in body functions.
Collapse
Affiliation(s)
- Yoshinori Marunaka
- Medical Research Institute, Kyoto Industrial Health Association, General Incorporated Foundation, 67 Kitatsuboi-Cho, Nishinokyo, Nakagyo-Ku, Kyoto, 604-8472, Japan.
- Research Organization of Science and Technology, Ritsumeikan University, Kusatsu, 525-8577, Japan.
- Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-Ku, Kyoto, 602-8566, Japan.
| |
Collapse
|
18
|
Okada Y, Numata T, Sabirov RZ, Kashio M, Merzlyak PG, Sato-Numata K. Cell death induction and protection by activation of ubiquitously expressed anion/cation channels. Part 3: the roles and properties of TRPM2 and TRPM7. Front Cell Dev Biol 2023; 11:1246955. [PMID: 37842082 PMCID: PMC10576435 DOI: 10.3389/fcell.2023.1246955] [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: 06/25/2023] [Accepted: 09/15/2023] [Indexed: 10/17/2023] Open
Abstract
Cell volume regulation (CVR) is a prerequisite for animal cells to survive and fulfill their functions. CVR dysfunction is essentially involved in the induction of cell death. In fact, sustained normotonic cell swelling and shrinkage are associated with necrosis and apoptosis, and thus called the necrotic volume increase (NVI) and the apoptotic volume decrease (AVD), respectively. Since a number of ubiquitously expressed ion channels are involved in the CVR processes, these volume-regulatory ion channels are also implicated in the NVI and AVD events. In Part 1 and Part 2 of this series of review articles, we described the roles of swelling-activated anion channels called VSOR or VRAC and acid-activated anion channels called ASOR or PAC in CVR and cell death processes. Here, Part 3 focuses on therein roles of Ca2+-permeable non-selective TRPM2 and TRPM7 cation channels activated by stress. First, we summarize their phenotypic properties and molecular structure. Second, we describe their roles in CVR. Since cell death induction is tightly coupled to dysfunction of CVR, third, we focus on their participation in the induction of or protection against cell death under oxidative, acidotoxic, excitotoxic, and ischemic conditions. In this regard, we pay attention to the sensitivity of TRPM2 and TRPM7 to a variety of stress as well as to their capability to physicall and functionally interact with other volume-related channels and membrane enzymes. Also, we summarize a large number of reports hitherto published in which TRPM2 and TRPM7 channels are shown to be involved in cell death associated with a variety of diseases or disorders, in some cases as double-edged swords. Lastly, we attempt to describe how TRPM2 and TRPM7 are organized in the ionic mechanisms leading to cell death induction and protection.
Collapse
Affiliation(s)
- Yasunobu Okada
- National Institute for Physiological Sciences (NIPS), Okazaki, Japan
- Department of Integrative Physiology, Graduate School of Medicine, AkitaUniversity, Akita, Japan
- Department of Physiology, School of Medicine, Aichi Medical Uniersity, Nagakute, Japan
- Department of Physiology, Kyoto Prefectural University of Medicine, Kyoto, Japan
- Cardiovascular Research Institute, Yokohama City University, Yokohama, Japan
| | - Tomohiro Numata
- Department of Integrative Physiology, Graduate School of Medicine, AkitaUniversity, Akita, Japan
| | - Ravshan Z. Sabirov
- Institute of Biophysics and Biochemistry, National University of Uzbekistan, Tashkent, Uzbekistan
| | - Makiko Kashio
- National Institute for Physiological Sciences (NIPS), Okazaki, Japan
- Department of Physiology, School of Medicine, Aichi Medical Uniersity, Nagakute, Japan
| | - Peter G. Merzlyak
- Institute of Biophysics and Biochemistry, National University of Uzbekistan, Tashkent, Uzbekistan
| | - Kaori Sato-Numata
- Department of Integrative Physiology, Graduate School of Medicine, AkitaUniversity, Akita, Japan
| |
Collapse
|
19
|
Giordano ME, Udayan G, Guascito MR, De Bartolomeo AR, Carlino A, Conte M, Contini D, Lionetto MG. Apoptotic volume decrease (AVD) in A 549 cells exposed to water-soluble fraction of particulate matter (PM 10). Front Physiol 2023; 14:1218687. [PMID: 37492639 PMCID: PMC10364053 DOI: 10.3389/fphys.2023.1218687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Accepted: 06/21/2023] [Indexed: 07/27/2023] Open
Abstract
Exposure to atmospheric particulate matter (PM) is recognized as a human health risk factor of great concern. The present work aimed to study the cellular mechanisms underlying cytotoxic effects of airborne particulate matter <10 µm in size (PM10), sampled in an urban background site from January to May 2020, on A549 cells. In particular, the study addressed if PM10 exposure can be a main factor in the induction of the Apoptotic Volume Decrease (AVD), which is one of the first events of apoptosis, and if the generation of intracellular oxidative stress can be involved in the PM10 induction of apoptosis in A549 cells. The cytotoxicity of PM10 samples was measured by MTT test on cells exposed for 24 h to the PM10 aqueous extracts, cell volume changes were monitored by morphometric analysis of the cells, apoptosis appearance was detected by annexin V and the induction of intracellular oxidative stress was evaluated by the ROS sensitive CM-H2DCFDA fluorescent probe. The results showed cytotoxic effects ascribable to apoptotic death in A549 cells exposed for 24 h to aqueous extracts of airborne winter PM10 samples characterized by high PM10 value and organic carbon content. The detected reduced cell viability in winter samples ranged from 55% to 100%. Normotonic cell volume reduction (ranging from about 60% to 30% cell volume decrease) after PM10 exposure was already detectable after the first 30 min clearly indicating the ability of PM10, mainly arising from biomass burning, to induce Apoptotic Volume Decrease (AVD) in A549 cells. AVD was prevented by the pre-treatment with 0.5 mM SITS indicating the activation of Cl- efflux presumably through the activation of VRAC channels. The exposure of A549 cells to PM10 aqueous extracts was able to induce intracellular oxidative stress detected by using the ROS-sensitive probe CM-H2DCFDA. The PM10-induced oxidative stress was statistically significantly correlated with cell viability inhibition and with apoptotic cell shrinkage. It was already evident after 15 min exposure representing one of the first cellular effects caused by PM exposure. This result suggests the role of oxidative stress in the PM10 induction of AVD as one of the first steps in cytotoxicity.
Collapse
Affiliation(s)
- M E Giordano
- Department Biological and Environmental Sciences and Technologies (DiSTeBA), Salento University, Lecce, Italy
| | - G Udayan
- Department Biological and Environmental Sciences and Technologies (DiSTeBA), Salento University, Lecce, Italy
| | - M R Guascito
- Department Biological and Environmental Sciences and Technologies (DiSTeBA), Salento University, Lecce, Italy
| | - A R De Bartolomeo
- Department Biological and Environmental Sciences and Technologies (DiSTeBA), Salento University, Lecce, Italy
| | - A Carlino
- Department Biological and Environmental Sciences and Technologies (DiSTeBA), Salento University, Lecce, Italy
| | - M Conte
- Institute of Atmospheric Sciences and Climate, ISAC-CNR, Rome, Italy
| | - D Contini
- Institute of Atmospheric Sciences and Climate, ISAC-CNR, Lecce, Italy
| | - M G Lionetto
- Department Biological and Environmental Sciences and Technologies (DiSTeBA), Salento University, Lecce, Italy
- NBFC, National Biodiversity Future Center, Palermo, Italy
| |
Collapse
|
20
|
Empitu MA, Kikyo M, Shirata N, Yamada H, Makino SI, Kadariswantiningsih IN, Aizawa M, Patrakka J, Nishimori K, Asanuma K. Inhibition of Importin- α -Mediated Nuclear Localization of Dendrin Attenuates Podocyte Loss and Glomerulosclerosis. J Am Soc Nephrol 2023; 34:1222-1239. [PMID: 37134307 PMCID: PMC10356163 DOI: 10.1681/asn.0000000000000150] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 03/27/2023] [Indexed: 05/05/2023] Open
Abstract
SIGNIFICANCE STATEMENT Nuclear translocation of dendrin is observed in injured podocytes, but the mechanism and its consequence are unknown. In nephropathy mouse models, dendrin ablation attenuates proteinuria, podocyte loss, and glomerulosclerosis. The nuclear translocation of dendrin promotes c-Jun N -terminal kinase phosphorylation in podocytes, altering focal adhesion and enhancing cell detachment-induced apoptosis. We identified mediation of dendrin nuclear translocation by nuclear localization signal 1 (NLS1) sequence and adaptor protein importin- α . Inhibition of importin- α prevents nuclear translocation of dendrin, decreases podocyte loss, and attenuates glomerulosclerosis in nephropathy models. Thus, inhibiting importin- α -mediated nuclear translocation of dendrin is a potential strategy to halt podocyte loss and glomerulosclerosis. BACKGROUND Nuclear translocation of dendrin is observed in the glomeruli in numerous human renal diseases, but the mechanism remains unknown. This study investigated that mechanism and its consequence in podocytes. METHODS The effect of dendrin deficiency was studied in adriamycin (ADR) nephropathy model and membrane-associated guanylate kinase inverted 2 ( MAGI2 ) podocyte-specific knockout ( MAGI2 podKO) mice. The mechanism and the effect of nuclear translocation of dendrin were studied in podocytes overexpressing full-length dendrin and nuclear localization signal 1-deleted dendrin. Ivermectin was used to inhibit importin- α . RESULTS Dendrin ablation reduced albuminuria, podocyte loss, and glomerulosclerosis in ADR-induced nephropathy and MAGI2 podKO mice. Dendrin deficiency also prolonged the lifespan of MAGI2 podKO mice. Nuclear dendrin promoted c-Jun N -terminal kinase phosphorylation that subsequently altered focal adhesion, reducing cell attachment and enhancing apoptosis in cultured podocytes. Classical bipartite nuclear localization signal sequence and importin- α mediate nuclear translocation of dendrin. The inhibition of importin- α / β reduced dendrin nuclear translocation and apoptosis in vitro as well as albuminuria, podocyte loss, and glomerulosclerosis in ADR-induced nephropathy and MAGI2 podKO mice. Importin- α 3 colocalized with nuclear dendrin in the glomeruli of FSGS and IgA nephropathy patients. CONCLUSIONS Nuclear translocation of dendrin promotes cell detachment-induced apoptosis in podocytes. Therefore, inhibiting importin- α -mediated dendrin nuclear translocation is a potential strategy to prevent podocyte loss and glomerulosclerosis.
Collapse
Affiliation(s)
- Maulana A. Empitu
- Department of Nephrology, Graduate School of Medicine, Chiba University, Chiba, Japan
- Faculty of Medicine, Airlangga University, Surabaya, Indonesia
| | - Mitsuhiro Kikyo
- Sohyaku, Innovative Research Division, Mitsubishi Tanabe Pharmaceutical Corporation, Kanagawa, Japan
- Medical Innovation Center, TMK Project, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Naritoshi Shirata
- Sohyaku, Innovative Research Division, Mitsubishi Tanabe Pharmaceutical Corporation, Kanagawa, Japan
- Medical Innovation Center, TMK Project, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hiroyuki Yamada
- Department of Nephrology, Graduate School of Medicine, Chiba University, Chiba, Japan
- Medical Innovation Center, TMK Project, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Department of Nephrology, Kyoto University Hospital, Kyoto, Japan
| | - Shin-ichi Makino
- Department of Nephrology, Graduate School of Medicine, Chiba University, Chiba, Japan
- Medical Innovation Center, TMK Project, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Department of Nephrology, Kyoto University Hospital, Kyoto, Japan
| | - Ika N. Kadariswantiningsih
- Department of Nephrology, Graduate School of Medicine, Chiba University, Chiba, Japan
- Faculty of Medicine, Airlangga University, Surabaya, Indonesia
| | - Masashi Aizawa
- Department of Nephrology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Jaakko Patrakka
- Karolinska Institute/AstraZeneca Integrated Cardio Metabolic Center (ICMC), Huddinge, Sweden
- Division of Pathology, Department of Laboratory Medicine, Karolinska University Hospital Huddinge, Huddinge, Sweden
| | - Katsuhiko Nishimori
- Department of Bioregulation and Pharmacological Medicine and Department of Obesity and Internal Inflammation, Fukushima Medical University, Fukushima, Japan
| | - Katsuhiko Asanuma
- Department of Nephrology, Graduate School of Medicine, Chiba University, Chiba, Japan
- Medical Innovation Center, TMK Project, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
21
|
Balistrieri A, Makino A, Yuan JXJ. Pathophysiology and pathogenic mechanisms of pulmonary hypertension: role of membrane receptors, ion channels, and Ca 2+ signaling. Physiol Rev 2023; 103:1827-1897. [PMID: 36422993 PMCID: PMC10110735 DOI: 10.1152/physrev.00030.2021] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 11/11/2022] [Accepted: 11/19/2022] [Indexed: 11/25/2022] Open
Abstract
The pulmonary circulation is a low-resistance, low-pressure, and high-compliance system that allows the lungs to receive the entire cardiac output. Pulmonary arterial pressure is a function of cardiac output and pulmonary vascular resistance, and pulmonary vascular resistance is inversely proportional to the fourth power of the intraluminal radius of the pulmonary artery. Therefore, a very small decrease of the pulmonary vascular lumen diameter results in a significant increase in pulmonary vascular resistance and pulmonary arterial pressure. Pulmonary arterial hypertension is a fatal and progressive disease with poor prognosis. Regardless of the initial pathogenic triggers, sustained pulmonary vasoconstriction, concentric vascular remodeling, occlusive intimal lesions, in situ thrombosis, and vascular wall stiffening are the major and direct causes for elevated pulmonary vascular resistance in patients with pulmonary arterial hypertension and other forms of precapillary pulmonary hypertension. In this review, we aim to discuss the basic principles and physiological mechanisms involved in the regulation of lung vascular hemodynamics and pulmonary vascular function, the changes in the pulmonary vasculature that contribute to the increased vascular resistance and arterial pressure, and the pathogenic mechanisms involved in the development and progression of pulmonary hypertension. We focus on reviewing the pathogenic roles of membrane receptors, ion channels, and intracellular Ca2+ signaling in pulmonary vascular smooth muscle cells in the development and progression of pulmonary hypertension.
Collapse
Affiliation(s)
- Angela Balistrieri
- Section of Physiology, Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, University of California, San Diego, La Jolla, California
- Harvard University, Cambridge, Massachusetts
| | - Ayako Makino
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Diego, La Jolla, California
| | - Jason X-J Yuan
- Section of Physiology, Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, University of California, San Diego, La Jolla, California
| |
Collapse
|
22
|
Cancer as a Channelopathy—Appreciation of Complimentary Pathways Provides a Different Perspective for Developing Treatments. Cancers (Basel) 2022; 14:cancers14194627. [PMID: 36230549 PMCID: PMC9562872 DOI: 10.3390/cancers14194627] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 09/17/2022] [Accepted: 09/21/2022] [Indexed: 12/15/2022] Open
Abstract
Simple Summary While improvements in technology have improved our ability to treat many forms of cancer when diagnosed at an early stage of the disease, the ability to improve survival and quality of life for patients with late stage disease has been limited, largely due to the ability of cancer cells to evade destruction when treatments block preferred paths for survival. Here, we review the role that ions and ion channels play in normal cell function, the development of disease and their role in the life and death of a cell. It is hoped that viewing cancer from the perspective of altered ion channel expression and ion balance may provide a novel approach for developing more effective treatments for this devastating disease. Abstract Life depends upon the ability of cells to evaluate and adapt to a constantly changing environment and to maintain internal stability to allow essential biochemical reactions to occur. Ions and ion channels play a crucial role in this process and are essential for survival. Alterations in the expression of the transmembrane proteins responsible for maintaining ion balance that occur as a result of mutations in the genetic code or in response to iatrogenically induced changes in the extracellular environment is a characteristic feature of oncogenesis and identifies cancer as one of a constellation of diseases known as channelopathies. The classification of cancer as a channelopathy provides a different perspective for viewing the disease. Potentially, it may expand opportunities for developing novel ways to affect or reverse the deleterious changes that underlie establishing and sustaining disease and developing tolerance to therapeutic attempts at treatment. The role of ions and ion channels and their interactions in the cell’s ability to maintain ionic balance, homeostasis, and survival are reviewed and possible approaches that mitigate gain or loss of ion channel function to contribute to new or enhance existing cancer therapies are discussed.
Collapse
|
23
|
Figueroa B, Xu FX, Hu R, Men S, Fu D. Quantitative Imaging of Intracellular Density with Ratiometric Stimulated Raman Scattering Microscopy. J Phys Chem B 2022; 126:7595-7603. [PMID: 36135097 DOI: 10.1021/acs.jpcb.2c04355] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Cell size and density are tightly controlled in mammalian cells. They impact a wide range of physiological functions, including osmoregulation, tissue homeostasis, and growth regulation. Compared to size, density variation for a given cell type is typically much smaller, implying that cell-type-specific density plays an important role in cell function. However, little is known about how cell density affects cell function or how it is regulated. Current tools for intracellular cell density measurements are limited to either suspended cells or cells grown on 2D substrates, neither of which recapitulate the physiology of single cells in intact tissue. While optical measurements have the potential to noninvasively measure cell density in situ, light scattering in multicellular systems prevents direct quantification. Here, we introduce an intracellular density imaging technique based on ratiometric stimulated Raman scattering microscopy (rSRS). It uses intrinsic vibrational information from intracellular macromolecules to quantify dry mass density. Moreover, water is used as an internal standard to correct for aberration and light scattering effects. We demonstrate real-time measurement of intracellular density and show that density is tightly regulated across different cell types and can be used to differentiate cell types as well as cell states. We further demonstrate dynamic imaging of density change in response to osmotic challenge as well as intracellular density imaging of a 3D tumor spheroid. Our technique has the potential for imaging intracellular density in intact tissue and understanding density regulation and its role in tissue homeostasis.
Collapse
Affiliation(s)
- Benjamin Figueroa
- Department of Chemistry, University of Washington, Seattle, Washington 98195, United States
| | - Fiona Xi Xu
- Department of Chemistry, University of Washington, Seattle, Washington 98195, United States
| | - Ruoqian Hu
- Department of Chemistry, University of Washington, Seattle, Washington 98195, United States
| | - Shuaiqian Men
- Department of Chemistry, University of Washington, Seattle, Washington 98195, United States
| | - Dan Fu
- Department of Chemistry, University of Washington, Seattle, Washington 98195, United States
| |
Collapse
|
24
|
The Comparative Experimental Study of Sodium and Magnesium Dichloroacetate Effects on Pediatric PBT24 and SF8628 Cell Glioblastoma Tumors Using a Chicken Embryo Chorioallantoic Membrane Model and on Cells In Vitro. Int J Mol Sci 2022; 23:ijms231810455. [PMID: 36142368 PMCID: PMC9499689 DOI: 10.3390/ijms231810455] [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: 08/16/2022] [Revised: 08/31/2022] [Accepted: 09/07/2022] [Indexed: 11/16/2022] Open
Abstract
In this study, pyruvate dehydrogenase kinase-1 inhibition with dichloroacetate (DCA) was explored as an alternative cancer therapy. The study’s aim was to compare the effectiveness of NaDCA and MgDCA on pediatric glioblastoma PBT24 and SF8628 tumors and cells. The treatment effects were evaluated on xenografts growth on a chicken embryo chorioallantoic membrane. The PCNA, EZH2, p53, survivin expression in tumor, and the SLC12A2, SLC12A5, SLC5A8, CDH1, and CDH2 expression in cells were studied. The tumor groups were: control, cells treated with 10 mM and 5 mM of NaDCA, and 5 mM and 2.5 mM of MgDCA. The cells were also treated with 3 mM DCA. Both the 10 mM DCA preparations significantly reduced PBT24 and SF8624 tumor invasion rates, while 5 mM NaDCA reduced it only in the SF8628 tumors. The 5 mM MgDCA inhibited tumor-associated neoangiogenesis in PBT24; both doses of NaDCA inhibited tumor-associated neoangiogenesis in SF8628. The 10 mM DCA inhibited the expression of markers tested in PBT24 and SF8628 tumors, but the 5 mM DCA affect on their expression depended on the cation. The DCA treatment did not affect the SLC12A2, SLC12A5, and SLC5A8 expression in cells but increased CDH1 expression in SF8628. The tumor response to DCA at different doses indicated that a contrast between NaDCA and MgDCA effectiveness reflects the differences in the tested cells’ biologies.
Collapse
|
25
|
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
|
26
|
Koumangoye R. The role of Cl - and K + efflux in NLRP3 inflammasome and innate immune response activation. Am J Physiol Cell Physiol 2022; 322:C645-C652. [PMID: 35171697 DOI: 10.1152/ajpcell.00421.2021] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Inflammation is part of innate immunity and is a natural response of the body to bacteria, virus, any other pathogen infections, or to damaged tissues. However, too much inflammation or chronic inflammation contributes to a wide variety of diseases such as inflammatory bowel disease, cancer, type 2 diabetes, heart disease, or autoimmune disease such as rheumatoid arthritis. Recent studies underscored the critical role of K+ and Cl- efflux in the activation of the inflammasome. The NLRP3 inflammasome is a multiprotein complex that mediates the production of the proinflammatory cytokines IL-1β and IL-18 and initiates the inflammatory cell death or pyroptosis. The NLRP3 inflammasome can be activated by multiple stimuli such as extracellular ATP, microbial toxins, ROS, mitochondria DNA or particulate matter. Although the precise mechanisms of NLRP3 activation and regulation by these diverse agonists remain unclear, multiple reports indicate that all NLRP3 agonists ultimately lead to a drop in intracellular concentration of potassium (K+ efflux) and chloride (Cl- efflux). The WNK-SPAK/OSR1-[N]KCC pathway plays a critical role maintaining K+ and Cl- ions concentration in the cell. Recent advances indicate that the WNK-SPAK-[N]KCC pathway play a role in the activation of the innate immune response. This review highlights recent discoveries detailing how ion transport regulates innate immune cell response to inflammatory stimuli.
Collapse
Affiliation(s)
- Rainelli Koumangoye
- Department of Anesthesiology, Vanderbilt University School of Medicine, Nashville, TN, United States
| |
Collapse
|
27
|
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
|
28
|
Ion Channel Involvement in Tumor Drug Resistance. J Pers Med 2022; 12:jpm12020210. [PMID: 35207698 PMCID: PMC8878471 DOI: 10.3390/jpm12020210] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 01/28/2022] [Accepted: 02/02/2022] [Indexed: 11/30/2022] Open
Abstract
Over 90% of deaths in cancer patients are attributed to tumor drug resistance. Resistance to therapeutic agents can be due to an innate property of cancer cells or can be acquired during chemotherapy. In recent years, it has become increasingly clear that regulation of membrane ion channels is an important mechanism in the development of chemoresistance. Here, we review the contribution of ion channels in drug resistance of various types of cancers, evaluating their potential in clinical management. Several molecular mechanisms have been proposed, including evasion of apoptosis, cell cycle arrest, decreased drug accumulation in cancer cells, and activation of alternative escape pathways such as autophagy. Each of these mechanisms leads to a reduction of the therapeutic efficacy of administered drugs, causing more difficulty in cancer treatment. Thus, targeting ion channels might represent a good option for adjuvant therapies in order to counteract chemoresistance development.
Collapse
|
29
|
Kittl M, Winklmayr M, Preishuber-Pflügl J, Strobl V, Gaisberger M, Ritter M, Jakab M. Low pH Attenuates Apoptosis by Suppressing the Volume-Sensitive Outwardly Rectifying (VSOR) Chloride Current in Chondrocytes. Front Cell Dev Biol 2022; 9:804105. [PMID: 35186954 PMCID: PMC8847443 DOI: 10.3389/fcell.2021.804105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 12/31/2021] [Indexed: 11/25/2022] Open
Abstract
In a variety of physiological and pathophysiological conditions, cells are exposed to acidic environments. Severe synovial fluid acidification also occurs in a progressive state of osteoarthritis (OA) affecting articular chondrocytes. In prior studies extracellular acidification has been shown to protect cells from apoptosis but the underlying mechanisms remain elusive. In the present study, we demonstrate that the inhibition of Cl− currents plays a significant role in the antiapoptotic effect of acidification in human articular chondrocytes. Drug-induced apoptosis was analyzed after exposure to staurosporine by caspase 3/7 activity and by annexin-V/7-actinomycin D (7-AAD) staining, followed by flow cytometry. Cell viability was assessed by resazurin, CellTiter-Glo and CellTiter-Fluor assays. Cl− currents and the mean cell volume were determined using the whole cell patch clamp technique and the Coulter method, respectively. The results reveal that in C28/I2 cells extracellular acidification decreases caspase 3/7 activity, enhances cell viability following staurosporine treatment and gradually deactivates the volume-sensitive outwardly rectifying (VSOR) Cl− current. Furthermore, the regulatory volume decrease (RVD) as well as the apoptotic volume decrease (ADV), which represents an early event during apoptosis, were absent under acidic conditions after hypotonicity-induced cell swelling and staurosporine-induced apoptosis, respectively. Like acidosis, the VSOR Cl− current inhibitor DIDS rescued chondrocytes from apoptotic cell death and suppressed AVD after induction of apoptosis with staurosporine. Similar to acidosis and DIDS, the VSOR channel blockers NPPB, niflumic acid (NFA) and DCPIB attenuated the staurosporine-induced AVD. NPPB and NFA also suppressed staurosporine-induced caspase 3/7 activation, while DCPIB and Tamoxifen showed cytotoxic effects per se. From these data, we conclude that the deactivation of VSOR Cl− currents impairs cell volume regulation under acidic conditions, which is likely to play an important role in the survivability of human articular chondrocytes.
Collapse
Affiliation(s)
- Michael Kittl
- Center for Physiology, Pathophysiology and Biophysics, Institute for Physiology and Pathophysiology—Salzburg, Paracelsus Medical University, Salzburg, Austria
- Ludwig Boltzmann Institute for Arthritis and Rehabilitation, Salzburg, Austria
- *Correspondence: Michael Kittl,
| | - Martina Winklmayr
- Center for Physiology, Pathophysiology and Biophysics, Institute for Physiology and Pathophysiology—Salzburg, Paracelsus Medical University, Salzburg, Austria
- Ludwig Boltzmann Institute for Arthritis and Rehabilitation, Salzburg, Austria
| | - Julia Preishuber-Pflügl
- Center for Physiology, Pathophysiology and Biophysics, Institute for Physiology and Pathophysiology—Salzburg, Paracelsus Medical University, Salzburg, Austria
- Gastein Research Institute, Paracelsus Medical University, Salzburg, Austria
| | - Victoria Strobl
- Center for Physiology, Pathophysiology and Biophysics, Institute for Physiology and Pathophysiology—Salzburg, Paracelsus Medical University, Salzburg, Austria
- Gastein Research Institute, Paracelsus Medical University, Salzburg, Austria
| | - Martin Gaisberger
- Center for Physiology, Pathophysiology and Biophysics, Institute for Physiology and Pathophysiology—Salzburg, Paracelsus Medical University, Salzburg, Austria
- Ludwig Boltzmann Institute for Arthritis and Rehabilitation, Salzburg, Austria
- Gastein Research Institute, Paracelsus Medical University, Salzburg, Austria
| | - Markus Ritter
- Center for Physiology, Pathophysiology and Biophysics, Institute for Physiology and Pathophysiology—Salzburg, Paracelsus Medical University, Salzburg, Austria
- Ludwig Boltzmann Institute for Arthritis and Rehabilitation, Salzburg, Austria
- Gastein Research Institute, Paracelsus Medical University, Salzburg, Austria
- Center for Physiology, Pathophysiology and Biophysics, Institute for Physiology, Pathophysiology and Biophysics—Nuremberg, Paracelsus Medical University, Nuremberg, Germany
| | - Martin Jakab
- Center for Physiology, Pathophysiology and Biophysics, Institute for Physiology and Pathophysiology—Salzburg, Paracelsus Medical University, Salzburg, Austria
- Ludwig Boltzmann Institute for Arthritis and Rehabilitation, Salzburg, Austria
| |
Collapse
|
30
|
Okada Y, Sabirov RZ, Merzlyak PG, Numata T, Sato-Numata K. Properties, Structures, and Physiological Roles of Three Types of Anion Channels Molecularly Identified in the 2010's. Front Physiol 2022; 12:805148. [PMID: 35002778 PMCID: PMC8733619 DOI: 10.3389/fphys.2021.805148] [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: 10/29/2021] [Accepted: 12/06/2021] [Indexed: 11/24/2022] Open
Abstract
Molecular identification was, at last, successfully accomplished for three types of anion channels that are all implicated in cell volume regulation/dysregulation. LRRC8A plus LRRC8C/D/E, SLCO2A1, and TMEM206 were shown to be the core or pore-forming molecules of the volume-sensitive outwardly rectifying anion channel (VSOR) also called the volume-regulated anion channel (VRAC), the large-conductance maxi-anion channel (Maxi-Cl), and the acid-sensitive outwardly rectifying anion channel (ASOR) also called the proton-activated anion channel (PAC) in 2014, 2017, and 2019, respectively. More recently in 2020 and 2021, we have identified the S100A10-annexin A2 complex and TRPM7 as the regulatory proteins for Maxi-Cl and VSOR/VRAC, respectively. In this review article, we summarize their biophysical and structural properties as well as their physiological roles by comparing with each other on the basis of their molecular insights. We also point out unsolved important issues to be elucidated soon in the future.
Collapse
Affiliation(s)
- Yasunobu Okada
- National Institute for Physiological Sciences (NIPS), Okazaki, Japan.,Department of Physiology, School of Medicine, Aichi Medical University, Nagakute, Japan.,Department of Physiology, Kyoto Prefectural University of Medicine, Kyoto, Japan.,Cardiovascular Research Institute, Yokohama City University, Yokohama, Japan
| | - Ravshan Z Sabirov
- Laboratory of Molecular Physiology, Institute of Biophysics and Biochemistry, National University of Uzbekistan, Tashkent, Uzbekistan
| | - Petr G Merzlyak
- Laboratory of Molecular Physiology, Institute of Biophysics and Biochemistry, National University of Uzbekistan, Tashkent, Uzbekistan
| | - Tomohiro Numata
- Department of Integrative Physiology, Graduate School of Medicine, Akita University, Akita, Japan
| | - Kaori Sato-Numata
- Department of Integrative Physiology, Graduate School of Medicine, Akita University, Akita, Japan.,Japan Society for the Promotion of Science, Tokyo, Japan
| |
Collapse
|
31
|
Wilczyński B, Dąbrowska A, Saczko J, Kulbacka J. The Role of Chloride Channels in the Multidrug Resistance. MEMBRANES 2021; 12:38. [PMID: 35054564 PMCID: PMC8781147 DOI: 10.3390/membranes12010038] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 12/23/2021] [Indexed: 12/19/2022]
Abstract
Nowadays, one of medicine's main and most challenging aims is finding effective ways to treat cancer. Unfortunately, although there are numerous anti-cancerous drugs, such as cisplatin, more and more cancerous cells create drug resistance. Thus, it is equally important to find new medicines and research the drug resistance phenomenon and possibilities to avoid this mechanism. Ion channels, including chloride channels, play an important role in the drug resistance phenomenon. Our article focuses on the chloride channels, especially the volume-regulated channels (VRAC) and CLC chloride channels family. VRAC induces multidrug resistance (MDR) by causing apoptosis connected with apoptotic volume decrease (AVD) and VRAC are responsible for the transport of anti-cancerous drugs such as cisplatin. VRACs are a group of heterogenic complexes made from leucine-rich repetition with 8A (LRRC8A) and a subunit LRRC8B-E responsible for the properties. There are probably other subunits, which can create those channels, for example, TTYH1 and TTYH2. It is also known that the ClC family is involved in creating MDR in mainly two mechanisms-by changing the cell metabolism or acidification of the cell. The most researched chloride channel from this family is the CLC-3 channel. However, other channels are playing an important role in inducing MDR as well. In this paper, we review the role of chloride channels in MDR and establish the role of the channels in the MDR phenomenon.
Collapse
Affiliation(s)
- Bartosz Wilczyński
- Faculty of Medicine, Wroclaw Medical University, L. Pasteura 1, 50-367 Wroclaw, Poland; (B.W.); (A.D.)
| | - Alicja Dąbrowska
- Faculty of Medicine, Wroclaw Medical University, L. Pasteura 1, 50-367 Wroclaw, Poland; (B.W.); (A.D.)
| | - Jolanta Saczko
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland;
| | - Julita Kulbacka
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland;
| |
Collapse
|
32
|
Hollembeak JE, Model MA. Stability of Intracellular Protein Concentration under Extreme Osmotic Challenge. Cells 2021; 10:cells10123532. [PMID: 34944039 PMCID: PMC8700764 DOI: 10.3390/cells10123532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 12/06/2021] [Accepted: 12/10/2021] [Indexed: 11/16/2022] Open
Abstract
Cell volume (CV) regulation is typically studied in short-term experiments to avoid complications resulting from cell growth and division. By combining quantitative phase imaging (by transport-of-intensity equation) with CV measurements (by the exclusion of an external absorbing dye), we were able to monitor the intracellular protein concentration (PC) in HeLa and 3T3 cells for up to 48 h. Long-term PC remained stable in solutions with osmolarities ranging from one-third to almost twice the normal. When cells were subjected to extreme hypoosmolarity (one-quarter of normal), their PC did not decrease as one might expect, but increased; a similar dehydration response was observed at high concentrations of ionophore gramicidin. Highly dilute media, or even moderately dilute in the presence of cytochalasin, caused segregation of water into large protein-free vacuoles, while the surrounding cytoplasm remained at normal density. These results suggest that: (1) dehydration is a standard cellular response to severe stress; (2) the cytoplasm resists prolonged dilution. In an attempt to investigate the mechanism behind the homeostasis of PC, we tested the inhibitors of the protein kinase complex mTOR and the volume-regulated anion channels (VRAC). The initial results did not fully elucidate whether these elements are directly involved in PC maintenance.
Collapse
|
33
|
Kolobkova Y, Pervaiz S, Stauber T. The expanding toolbox to study the LRRC8-formed volume-regulated anion channel VRAC. CURRENT TOPICS IN MEMBRANES 2021; 88:119-163. [PMID: 34862024 DOI: 10.1016/bs.ctm.2021.10.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
The volume-regulated anion channel (VRAC) is activated upon cell swelling and facilitates the passive movement of anions across the plasma membrane in cells. VRAC function underlies many critical homeostatic processes in vertebrate cells. Among them are the regulation of cell volume and membrane potential, glutamate release and apoptosis. VRAC is also permeable for organic osmolytes and metabolites including some anti-cancer drugs and antibiotics. Therefore, a fundamental understanding of VRAC's structure-function relationships, its physiological roles, its utility for therapy of diseases, and the development of compounds modulating its activity are important research frontiers. Here, we describe approaches that have been applied to study VRAC since it was first described more than 30 years ago, providing an overview of the recent methodological progress. The diverse applications reflecting a compromise between the physiological situation, biochemical definition, and biophysical resolution range from the study of VRAC activity using a classic electrophysiology approach, to the measurement of osmolytes transport by various means and the investigation of its activation using a novel biophysical approach based on fluorescence resonance energy transfer.
Collapse
Affiliation(s)
- Yulia Kolobkova
- Department of Human Medicine and Institute for Molecular Medicine, MSH Medical School Hamburg, Germany
| | - Sumaira Pervaiz
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Germany
| | - Tobias Stauber
- Department of Human Medicine and Institute for Molecular Medicine, MSH Medical School Hamburg, Germany; Institute of Chemistry and Biochemistry, Freie Universität Berlin, Germany.
| |
Collapse
|
34
|
Ritter M, Mongin AA, Valenti G, Okada Y. Editorial: Ion and Water Transport in Cell Death. Front Cell Dev Biol 2021; 9:757033. [PMID: 34568348 PMCID: PMC8458750 DOI: 10.3389/fcell.2021.757033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 08/13/2021] [Indexed: 11/17/2022] Open
Affiliation(s)
- Markus Ritter
- Center for Physiology, Pathophysiology and Biophysics, Institute for Physiology and Pathophysiology, Paracelsus Medical University, Salzburg, Austria.,Kathmandu University, Dhulikhel, Nepal.,Ludwig Boltzmann Institute for Arthritis and Rehabilitation, Salzburg, Austria.,Gastein Research Institute, Paracelsus Medical University, Salzburg, Austria
| | - Alexander A Mongin
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, NY, United States
| | - Giovanna Valenti
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari Aldo Moro, Bari, Italy
| | - Yasunobu Okada
- Division of Cell Signaling, National Institute for Physiological Sciences (NIPS), Okazaki, Japan.,Department of Physiology, School of Medicine, Aichi Medical University, Nagakute, Japan.,Department of Physiology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
35
|
Hamed M, Osman AGM, Badrey AEA, Soliman HAM, Sayed AEDH. Microplastics-Induced Eryptosis and Poikilocytosis in Early-Juvenile Nile Tilapia ( Oreochromis niloticus). Front Physiol 2021; 12:742922. [PMID: 34650449 PMCID: PMC8507840 DOI: 10.3389/fphys.2021.742922] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 08/30/2021] [Indexed: 02/05/2023] Open
Abstract
This study aims to assess the impact of microplastics (MPs) on erythrocytes using eryptosis (apoptosis) and an erythron profile (poikilocytosis and nuclear abnormalities), considered to be novel biomarkers in Nile tilapia (Oreochromis niloticus). In this study, four groups of fish were used: The first was the control group. In the second group, 1 mg/L of MPs was introduced to the samples. The third group was exposed to 10 mg/L of MPs. Finally, the fourth group was exposed to 100 mg/L of MPs for 15 days, following 15 days of recovery. The fish treated with MPs experienced an immense rise in the eryptosis percentage, poikilocytosis, and nuclear abnormalities of red blood cells (RBCs) compared with the control group in a concentration-dependent manner. Poikilocytosis of MP-exposed groups included sickle cell shape, schistocyte, elliptocyte, acanthocyte, and other shapes. Nuclear abnormalities of the MPs-exposed groups included micronuclei, binucleated erythrocytes, notched, lobed, blebbed, and hemolyzed nuclei. After the recovery period, a greater percentage of eryptosis, poikilocytotic cells, and nuclear abnormalities in RBCs were still evident in the groups exposed to MPs when crosschecked with the control group. The results show concerning facts regarding the toxicity of MPs in tilapia.
Collapse
Affiliation(s)
- Mohamed Hamed
- Department of Zoology, Faculty of Science, Al Azhar University (Assiut Branch), Cairo, Egypt
| | - Alaa G. M. Osman
- Department of Zoology, Faculty of Science, Al Azhar University (Assiut Branch), Cairo, Egypt
| | - Ahmed E. A. Badrey
- Department of Zoology, Faculty of Science, Al Azhar University (Assiut Branch), Cairo, Egypt
| | | | | |
Collapse
|
36
|
Toki S, Yoshimaru T, Matsushita Y, Aihara H, Ono M, Tsuneyama K, Sairyo K, Katagiri T. The survival and proliferation of osteosarcoma cells are dependent on the mitochondrial BIG3-PHB2 complex formation. Cancer Sci 2021; 112:4208-4219. [PMID: 34363714 PMCID: PMC8486206 DOI: 10.1111/cas.15099] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 07/28/2021] [Accepted: 08/04/2021] [Indexed: 11/29/2022] Open
Abstract
Previous studies reported the critical role of the brefeldin A-inhibited guanine nucleotide exchange protein 3-prohibitin 2 (BIG3-PHB2) complex in modulating estrogen signaling activation in breast cancer cells, yet its pathophysiological roles in osteosarcoma (OS) cells remain elusive. Here, we report a novel function of BIG3-PHB2 in OS malignancy. BIG3-PHB2 complexes were localized mainly in mitochondria in OS cells, unlike in estrogen-dependent breast cancer cells. Depletion of endogenous BIG3 expression by small interfering RNA (siRNA) treatment led to significant inhibition of OS cell growth. Disruption of BIG3-PHB2 complex formation by treatment with specific peptide inhibitor also resulted in significant dose-dependent suppression of OS cell growth, migration, and invasion resulting from G2/M-phase arrest and in PARP cleavage, ultimately leading to PARP-1/apoptosis-inducing factor (AIF) pathway activation-dependent apoptosis in OS cells. Subsequent proteomic and bioinformatic pathway analyses revealed that disruption of the BIG3-PHB2 complex might lead to downregulation of inner mitochondrial membrane protein complex activity. Our findings indicate that the mitochondrial BIG3-PHB2 complex might regulate PARP-1/AIF pathway-dependent apoptosis during OS cell proliferation and progression and that disruption of this complex may be a promising therapeutic strategy for OS.
Collapse
Affiliation(s)
- Shunichi Toki
- Division of Genome Medicine, Advanced Institute of Medical Sciences, Tokushima University, Tokushima, Japan.,Department of Orthopedics, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Tetsuro Yoshimaru
- Division of Genome Medicine, Advanced Institute of Medical Sciences, Tokushima University, Tokushima, Japan
| | - Yosuke Matsushita
- Division of Genome Medicine, Advanced Institute of Medical Sciences, Tokushima University, Tokushima, Japan
| | - Hitoshi Aihara
- Division of Genome Medicine, Advanced Institute of Medical Sciences, Tokushima University, Tokushima, Japan
| | - Masaya Ono
- Department of Proteomics, National Cancer Center Research Institute, Tokyo, Japan
| | - Koichi Tsuneyama
- Department of Pathology and Laboratory Medicine, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Koichi Sairyo
- Department of Orthopedics, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Toyomasa Katagiri
- Division of Genome Medicine, Advanced Institute of Medical Sciences, Tokushima University, Tokushima, Japan
| |
Collapse
|
37
|
Okada Y, Sato-Numata K, Sabirov RZ, Numata T. Cell Death Induction and Protection by Activation of Ubiquitously Expressed Anion/Cation Channels. Part 2: Functional and Molecular Properties of ASOR/PAC Channels and Their Roles in Cell Volume Dysregulation and Acidotoxic Cell Death. Front Cell Dev Biol 2021; 9:702317. [PMID: 34307382 PMCID: PMC8299559 DOI: 10.3389/fcell.2021.702317] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 06/18/2021] [Indexed: 12/18/2022] Open
Abstract
For survival and functions of animal cells, cell volume regulation (CVR) is essential. Major hallmarks of necrotic and apoptotic cell death are persistent cell swelling and shrinkage, and thus they are termed the necrotic volume increase (NVI) and the apoptotic volume decrease (AVD), respectively. A number of ubiquitously expressed anion and cation channels play essential roles not only in CVR but also in cell death induction. This series of review articles address the question how cell death is induced or protected with using ubiquitously expressed ion channels such as swelling-activated anion channels, acid-activated anion channels, and several types of TRP cation channels including TRPM2 and TRPM7. In the Part 1, we described the roles of swelling-activated VSOR/VRAC anion channels. Here, the Part 2 focuses on the roles of the acid-sensitive outwardly rectifying (ASOR) anion channel, also called the proton-activated chloride (PAC) anion channel, which is activated by extracellular protons in a manner sharply dependent on ambient temperature. First, we summarize phenotypical properties, the molecular identity, and the three-dimensional structure of ASOR/PAC. Second, we highlight the unique roles of ASOR/PAC in CVR dysfunction and in the induction of or protection from acidotoxic cell death under acidosis and ischemic conditions.
Collapse
Affiliation(s)
- Yasunobu Okada
- National Institute for Physiological Sciences (NIPS), Okazaki, Japan.,Department of Physiology, School of Medicine, Aichi Medical University, Nagakute, Japan.,Department of Physiology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kaori Sato-Numata
- Department of Physiology, School of Medicine, Fukuoka University, Fukuoka, Japan.,Japan Society for the Promotion of Science, Tokyo, Japan
| | - Ravshan Z Sabirov
- Laboratory of Molecular Physiology, Institute of Biophysics and Biochemistry, National University of Uzbekistan, Tashkent, Uzbekistan
| | - Tomohiro Numata
- Department of Physiology, School of Medicine, Fukuoka University, Fukuoka, Japan
| |
Collapse
|
38
|
Kim GN, Hah YS, Seong H, Yoo WS, Choi MY, Cho HY, Yun SP, Kim SJ. The Role of Nuclear Factor of Activated T Cells 5 in Hyperosmotic Stress-Exposed Human Lens Epithelial Cells. Int J Mol Sci 2021; 22:ijms22126296. [PMID: 34208226 PMCID: PMC8230750 DOI: 10.3390/ijms22126296] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 06/08/2021] [Accepted: 06/09/2021] [Indexed: 11/16/2022] Open
Abstract
We investigated the role of nuclear factor of activated T cells 5 (NFAT5) under hyperosmotic conditions in human lens epithelial cells (HLECs). Hyperosmotic stress decreased the viability of human lens epithelial B-3 cells and significantly increased NFAT5 expression. Hyperosmotic stress-induced cell death occurred to a greater extent in NFAT5-knockout (KO) cells than in NFAT5 wild-type (NFAT5 WT) cells. Bcl-2 and Bcl-xl expression was down-regulated in NFAT5 WT cells and NFAT5 KO cells under hyperosmotic stress. Pre-treatment with a necroptosis inhibitor (necrostatin-1) significantly blocked hyperosmotic stress-induced death of NFAT5 KO cells, but not of NFAT5 WT cells. The phosphorylation levels of receptor-interacting protein kinase 1 (RIP1) and RIP3, which indicate the occurrence of necroptosis, were up-regulated in NFAT5 KO cells, suggesting that death of these cells is predominantly related to the necroptosis pathway. This finding is the first to report that necroptosis occurs when lens epithelial cells are exposed to hyperosmolar conditions, and that NFAT5 is involved in this process.
Collapse
Affiliation(s)
- Gyu-Nam Kim
- Department of Ophthalmology, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Gyeongsang National University Hospital, Jinju 52727, Korea; (G.-N.K.); (H.S.); (W.-S.Y.); (M.-Y.C.)
| | - Young-Sool Hah
- Biomedical Research Institute, Gyeongsang National University Hospital, Jinju 52727, Korea; (Y.-S.H.); (H.-Y.C.)
| | - Hyemin Seong
- Department of Ophthalmology, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Gyeongsang National University Hospital, Jinju 52727, Korea; (G.-N.K.); (H.S.); (W.-S.Y.); (M.-Y.C.)
- Department of Pharmacology and Convergence Medical Science, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Korea
| | - Woong-Sun Yoo
- Department of Ophthalmology, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Gyeongsang National University Hospital, Jinju 52727, Korea; (G.-N.K.); (H.S.); (W.-S.Y.); (M.-Y.C.)
| | - Mee-Young Choi
- Department of Ophthalmology, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Gyeongsang National University Hospital, Jinju 52727, Korea; (G.-N.K.); (H.S.); (W.-S.Y.); (M.-Y.C.)
| | - Hee-Young Cho
- Biomedical Research Institute, Gyeongsang National University Hospital, Jinju 52727, Korea; (Y.-S.H.); (H.-Y.C.)
| | - Seung Pil Yun
- Department of Pharmacology and Convergence Medical Science, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Korea
- Correspondence: (S.P.Y.); (S.-J.K.); Tel.: +82-55-772-8071 (S.P.Y.); +82-55-750-8468 (S.-J.K.)
| | - Seong-Jae Kim
- Department of Ophthalmology, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Gyeongsang National University Hospital, Jinju 52727, Korea; (G.-N.K.); (H.S.); (W.-S.Y.); (M.-Y.C.)
- Correspondence: (S.P.Y.); (S.-J.K.); Tel.: +82-55-772-8071 (S.P.Y.); +82-55-750-8468 (S.-J.K.)
| |
Collapse
|
39
|
Mitchell SJ, Rosenblatt J. Early mechanical selection of cell extrusion and extrusion signaling in cancer. Curr Opin Cell Biol 2021; 72:36-40. [PMID: 34034216 DOI: 10.1016/j.ceb.2021.04.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 04/19/2021] [Accepted: 04/20/2021] [Indexed: 12/28/2022]
Abstract
Epithelial cells use the process of extrusion to promote cell death while preserving a tight barrier. To extrude, a cell and its neighbors contract actin and myosin circumferentially and basolaterally to seamlessly squeeze it out of the epithelium. Recent research highlights how early apical pulsatile contractions within the extruding cell might orchestrate contraction in three dimensions so that a cell extrudes out apically. Along with apical constrictions, studies of ion channels and mathematical modeling reveal how differential contraction between cells helps select specific cells to extrude. In addition, several studies have offered new insights into pathways that use extrusion to eliminate transformed cells or cause an aberrant form of extrusion that promotes cell invasion.
Collapse
Affiliation(s)
- Saranne J Mitchell
- Biomedical Engineering Department, The University of Utah, Salt Lake City, UT, USA; The Randall Centre for Cell & Molecular Biophysics, Faculty of Life Sciences & Medicine, Schools of Basic & Medical Biosciences and Cancer & Pharmaceutical Sciences, UK
| | - Jody Rosenblatt
- Biomedical Engineering Department, The University of Utah, Salt Lake City, UT, USA; The Randall Centre for Cell & Molecular Biophysics, Faculty of Life Sciences & Medicine, Schools of Basic & Medical Biosciences and Cancer & Pharmaceutical Sciences, UK.
| |
Collapse
|
40
|
Prolonged maintenance of hematopoietic stem cells that escape from thrombopoietin deprivation. Blood 2021; 137:2609-2620. [PMID: 33657206 DOI: 10.1182/blood.2020005517] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 01/14/2021] [Indexed: 11/20/2022] Open
Abstract
Hematopoietic stem cells (HSC) rarely divide, rest in quiescence, and proliferate only upon stress hematopoiesis. The cytokine thrombopoietin (Thpo) has been perplexingly described to induce quiescence and promote self-renewal divisions in HSCs. To clarify the contradictory effect of Thpo, we conducted a detailed analysis on conventional (Thpo-/-) and liver-specific (Thpofl/fl;AlbCre+/-) Thpo-deletion models. Thpo-/- HSCs exhibited profound loss of quiescence, impaired cell cycle progression, and increased apoptosis. Thpo-/- HSCs also exhibited diminished mitochondrial mass and impaired mitochondrial bioenergetics. Abnormal HSC phenotypes in Thpo-/- mice were reversible after HSC transplantation into wild-type recipients. Moreover, Thpo-/- HSCs acquired quiescence with extended administration of a Thpo receptor agonist, romiplostim, and were prone to subsequent stem cell exhaustion during competitive bone marrow transplantation. Thpofl/fl;AlbCre+/- HSCs exhibited similar stem cell phenotypes but to a lesser degree compared with Thpo-/- HSCs. HSCs that survive Thpo deficiency acquire quiescence in a dose-dependent manner through the modification of their metabolic state.
Collapse
|
41
|
Ülger M, Sezer G, Özyazgan İ, Özocak H, Yay A, Balcıoğlu E, Yalçın B, Göç R, Ülger B, Özyazgan TM, Yakan B. The effect of erythropoietin and umbilical cord-derived mesenchymal stem cells on nerve regeneration in rats with sciatic nerve injury. J Chem Neuroanat 2021; 114:101958. [PMID: 33864937 DOI: 10.1016/j.jchemneu.2021.101958] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 04/05/2021] [Accepted: 04/06/2021] [Indexed: 02/06/2023]
Abstract
OBJECTIVE We aimed to investigate the effects of umbilical cord-derived mesenchymal stem cells and erythropoietin on nerve regeneration in the sciatic nerve 'crush injury' in a rat model. METHODS Experimental animals were randomly divided into 5 groups: Crush Injury, Sham, Crush Injury + Erythropoietin, Crush Injury + Mesenchymal Stem Cell, Crush Injury + Erythropoietin + Mesenchymal Stem Cell groups. Crush injury made with bulldog clamp. Mesencyhmal stem cells delivered by enjection locally. Erythropoietin administered by intraperitoneally. On the 0th, 14th and 28th days, all groups underwent a sciatic functional index test. On 28th day, sciatic nerves were harvested and histopathological appearance, axon number and axon diameter of the sciatic nerves were evaluated with Oil Red O staining. Immunoreactivity of nerve growth factor, neurofilament-H and caspase-3 were determined by immunofluorescence staining in nerve tissue. RESULTS In histopathological examination, axons and nerve bundles exhibiting normal nerve architecture in the Sham group. Crush Injury + Mesenchymal Stem Cell group has similar histological appearance to the Sham group. The number of axons were higher in the Mesenchymal Stem Cell groups compared to the Crush Injury group. Nerve growth factor immunoreactivity intensity was significantly lower in Crush Injury + Mesenchymal Stem Cell group compared to Crush Injury group. Neurofilament-H density was higher in the treatment groups when compared to the Crush Injury group. CONCLUSIONS In this study, it was found that umbilical cord-derived mesenchymal stem cells and erythropoietin treatments effects positively regeneration of crush injury caused by bulldog clamp in the sciatic nerve of rats.
Collapse
Affiliation(s)
- Menekşe Ülger
- Department of Histology and Embryology, Erciyes University, Faculty of Medicine, 38039, Kayseri, Turkey.
| | - Gülay Sezer
- Department of Pharmacology, Erciyes University, Faculty of Medicine, 38039, Kayseri, Turkey.
| | - İrfan Özyazgan
- Department of Plastic Reconstructive and Aesthetic Surgery, Erciyes University, Faculty of Medicine, 38039, Kayseri, Turkey.
| | - Hakan Özocak
- Department of Plastic Reconstructive and Aesthetic Surgery, Erciyes University, Faculty of Medicine, 38039, Kayseri, Turkey.
| | - Arzu Yay
- Department of Histology and Embryology, Erciyes University, Faculty of Medicine, 38039, Kayseri, Turkey.
| | - Esra Balcıoğlu
- Department of Histology and Embryology, Erciyes University, Faculty of Medicine, 38039, Kayseri, Turkey.
| | - Betül Yalçın
- Department of Histology and Embryology, Erciyes University, Faculty of Medicine, 38039, Kayseri, Turkey.
| | - Rümeysa Göç
- Department of Histology and Embryology, Cumhuriyet University, Faculty of Medicine, 058140, Sivas, Turkey.
| | - Birkan Ülger
- Department of Anesthesiology and Reanimation, Erciyes University, Faculty of Medicine, 38039, Kayseri, Turkey.
| | - Tuğçe Merve Özyazgan
- Department of Histology and Embryology, Erciyes University, Faculty of Medicine, 38039, Kayseri, Turkey.
| | - Birkan Yakan
- Department of Histology and Embryology, Erciyes University, Faculty of Medicine, 38039, Kayseri, Turkey.
| |
Collapse
|
42
|
Zhao T, Arbelet-Bonnin D, Tran D, Monetti E, Lehner A, Meimoun P, Kadono T, Dauphin A, Errakhi R, Reboutier D, Cangémi S, Kawano T, Mancuso S, El-Maarouf-Bouteau H, Laurenti P, Bouteau F. Biphasic activation of survival and death pathways in Arabidopsis thaliana cultured cells by sorbitol-induced hyperosmotic stress. PLANT SCIENCE : AN INTERNATIONAL JOURNAL OF EXPERIMENTAL PLANT BIOLOGY 2021; 305:110844. [PMID: 33691971 DOI: 10.1016/j.plantsci.2021.110844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 02/01/2021] [Accepted: 02/03/2021] [Indexed: 06/12/2023]
Abstract
Hyperosmotic stresses represent some of the most serious abiotic factors that adversely affect plants growth, development and fitness. Despite their central role, the early cellular events that lead to plant adaptive responses remain largely unknown. In this study, using Arabidopsis thaliana cultured cells we analyzed early cellular responses to sorbitol-induced hyperosmotic stress. We observed biphasic and dual responses of A. thaliana cultured cells to sorbitol-induced hyperosmotic stress. A first set of events, namely singlet oxygen (1O2) production and cell hyperpolarization due to a decrease in anion channel activity could participate to signaling and osmotic adjustment allowing cell adaptation and survival. A second set of events, namely superoxide anion (O2-) production by RBOHD-NADPH-oxidases and SLAC1 anion channel activation could participate in programmed cell death (PCD) of a part of the cell population. This set of events raises the question of how a survival pathway and a death pathway could be induced by the same hyperosmotic condition and what could be the meaning of the induction of two different behaviors in response to hyperosmotic stress.
Collapse
Affiliation(s)
- Tingting Zhao
- Université de Paris, Laboratoire des Energies de Demain, Paris, France
| | | | - Daniel Tran
- former EA3514, Université Paris Diderot, Paris, France
| | - Emanuela Monetti
- former EA3514, Université Paris Diderot, Paris, France; LINV-DiSPAA, Department of Agri-Food and Environmental Science, University of Florence, Viale delle Idee 30, 50019, Sesto Fiorentino (FI), Italy
| | - Arnaud Lehner
- former EA3514, Université Paris Diderot, Paris, France
| | - Patrice Meimoun
- Université de Paris, Laboratoire des Energies de Demain, Paris, France; former EA3514, Université Paris Diderot, Paris, France; Université de Paris, Paris Interdisciplinary Energy Research Institute (PIERI), Paris, France
| | - Takashi Kadono
- former EA3514, Université Paris Diderot, Paris, France; Graduate School of Environmental Engineering, University of Kitakyushu, 1-1, Hibikino, Wakamatsu-ku, Kitakyushu 808-0135, Japan
| | | | - Rafik Errakhi
- former EA3514, Université Paris Diderot, Paris, France
| | | | - Sylvie Cangémi
- Université de Paris, Laboratoire des Energies de Demain, Paris, France
| | - Tomonori Kawano
- LINV-DiSPAA, Department of Agri-Food and Environmental Science, University of Florence, Viale delle Idee 30, 50019, Sesto Fiorentino (FI), Italy; Graduate School of Environmental Engineering, University of Kitakyushu, 1-1, Hibikino, Wakamatsu-ku, Kitakyushu 808-0135, Japan; University of Florence LINV Kitakyushu Research Center (LINV@Kitakyushu), Kitakyushu, Japan; Université de Paris, Paris Interdisciplinary Energy Research Institute (PIERI), Paris, France
| | - Stefano Mancuso
- LINV-DiSPAA, Department of Agri-Food and Environmental Science, University of Florence, Viale delle Idee 30, 50019, Sesto Fiorentino (FI), Italy; University of Florence LINV Kitakyushu Research Center (LINV@Kitakyushu), Kitakyushu, Japan; Université de Paris, Paris Interdisciplinary Energy Research Institute (PIERI), Paris, France
| | | | - Patrick Laurenti
- Université de Paris, Laboratoire des Energies de Demain, Paris, France
| | - François Bouteau
- Université de Paris, Laboratoire des Energies de Demain, Paris, France; former EA3514, Université Paris Diderot, Paris, France; LINV-DiSPAA, Department of Agri-Food and Environmental Science, University of Florence, Viale delle Idee 30, 50019, Sesto Fiorentino (FI), Italy; University of Florence LINV Kitakyushu Research Center (LINV@Kitakyushu), Kitakyushu, Japan.
| |
Collapse
|
43
|
Mondéjar-Parreño G, Cogolludo A, Perez-Vizcaino F. Potassium (K +) channels in the pulmonary vasculature: Implications in pulmonary hypertension Physiological, pathophysiological and pharmacological regulation. Pharmacol Ther 2021; 225:107835. [PMID: 33744261 DOI: 10.1016/j.pharmthera.2021.107835] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 02/26/2021] [Accepted: 03/02/2021] [Indexed: 02/06/2023]
Abstract
The large K+ channel functional diversity in the pulmonary vasculature results from the multitude of genes expressed encoding K+ channels, alternative RNA splicing, the post-transcriptional modifications, the presence of homomeric or heteromeric assemblies of the pore-forming α-subunits and the existence of accessory β-subunits modulating the functional properties of the channel. K+ channels can also be regulated at multiple levels by different factors controlling channel activity, trafficking, recycling and degradation. The activity of these channels is the primary determinant of membrane potential (Em) in pulmonary artery smooth muscle cells (PASMC), providing an essential regulatory mechanism to dilate or contract pulmonary arteries (PA). K+ channels are also expressed in pulmonary artery endothelial cells (PAEC) where they control resting Em, Ca2+ entry and the production of different vasoactive factors. The activity of K+ channels is also important in regulating the population and phenotype of PASMC in the pulmonary vasculature, since they are involved in cell apoptosis, survival and proliferation. Notably, K+ channels play a major role in the development of pulmonary hypertension (PH). Impaired K+ channel activity in PH results from: 1) loss of function mutations, 2) downregulation of its expression, which involves transcription factors and microRNAs, or 3) decreased channel current as a result of increased vasoactive factors (e.g., hypoxia, 5-HT, endothelin-1 or thromboxane), exposure to drugs with channel-blocking properties, or by a reduction in factors that positively regulate K+ channel activity (e.g., NO and prostacyclin). Restoring K+ channel expression, its intracellular trafficking and the channel activity is an attractive therapeutic strategy in PH.
Collapse
Affiliation(s)
- Gema Mondéjar-Parreño
- Department of Pharmacology and Toxicology, School of Medicine, University Complutense of Madrid, Spain; Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain; Ciber Enfermedades Respiratorias (CIBERES), Spain
| | - Angel Cogolludo
- Department of Pharmacology and Toxicology, School of Medicine, University Complutense of Madrid, Spain; Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain; Ciber Enfermedades Respiratorias (CIBERES), Spain
| | - Francisco Perez-Vizcaino
- Department of Pharmacology and Toxicology, School of Medicine, University Complutense of Madrid, Spain; Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain; Ciber Enfermedades Respiratorias (CIBERES), Spain.
| |
Collapse
|
44
|
Cao L, Li S, Huang S, Shi D, Li X. AQP8 participates in oestrogen-mediated buffalo follicular development by regulating apoptosis of granulosa cells. Reprod Domest Anim 2021; 56:812-820. [PMID: 33639021 DOI: 10.1111/rda.13921] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 02/25/2021] [Indexed: 11/29/2022]
Abstract
Aquaporins (AQPs), a family of small membrane-spanning proteins, are involved in fluid transport, cell signalling and reproduction. Regulating AQP8 expression influences apoptosis of granulosa cells (GCs), ovarian folliculogenesis, oogenesis and early embryonic development in mice, but its role has never been investigated in other species. The aim of the present study was to characterize the AQP8 function in buffalo follicular development. The expression pattern of AQP8 in buffalo follicle was analysed by immunohistochemistry method. 17β-Estradiol (E2) or oestrogen receptor antagonist ICI182780 was used to treat GCs cultured in vitro, and the expression of AQP8 was detected using qRT-PCR. Its roles in apoptosis of buffalo GCs were investigated by shRNA technology. AQP8 was found to be expressed higher in secondary follicles (p < .05), and its mRNA level in GCs was upregulated by E2 via receptor-mediated mechanism in a dose-dependent manner. A 732-bp buffalo AQP8 coding region was obtained, which was highly conserved at the amino acid level among different species. AQP8-shRNA2 had more effective inhibition on target gene than AQP8-shRNA1 (66.49% vs. 58.31%) (p < .05). Knockdown of AQP8 induced GCs arrested at G2/M stage and occurred apoptosis. Compared with the control group, higher Caspase9 expression were observed in AQP8-shRNA2 lentivirus infected GCs (p < .05), while Bcl-2 and Bax expression levels had no obvious change (p > .05). Altogether, the above results indicate that AQP8 is involved in oestrogen-mediated regulation of buffalo follicular development by regulating cell cycle progression and apoptosis of GCs.
Collapse
Affiliation(s)
- Lihua Cao
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, Guangxi University, Nanning, China
| | - Sheng Li
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, Guangxi University, Nanning, China.,Huangshi Maternity and Children's Health Hospital of Edong Healthcare Group, Huangshi, China
| | - Shihai Huang
- College of Life Science and Technology, Guangxi University, Nanning, China
| | - Deshun Shi
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, Guangxi University, Nanning, China
| | - Xiangping Li
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, Guangxi University, Nanning, China
| |
Collapse
|
45
|
A novel and distinctive mode of cell death revealed by using non-thermal atmospheric pressure plasma: The involvements of reactive oxygen species and the translation inhibitor Pdcd4. Chem Biol Interact 2021; 338:109403. [PMID: 33582111 DOI: 10.1016/j.cbi.2021.109403] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 11/22/2020] [Accepted: 02/04/2021] [Indexed: 12/11/2022]
Abstract
Cells death is indispensable for embryonic development, tissue homeostasis, and the elimination of cancer, virally infected, or degenerated cells in multicellular organisms. It occurs not only via existing modes but also via unidentified modes, whose elucidation requires. Exposure to non-thermal atmospheric pressure plasma (NTAPP) has been demonstrated to induce cell death, probably because of its ability to generate reactive oxygen species (ROS). However, the mode of this cell death and its underlying mechanism remained elusive. Here we show cell death occurring in a novel and distinctive mode different from apoptosis and necrosis/necroptosis through a mechanism that ROS mediate the loss of the translation inhibitor Programmed cell death 4 (Pdcd4) when cells are cultured in solutions activated by NTAPP irradiation. Thus, our study performed with NTAPP-activated solutions may provide insight into the existence of the atypical cell death in cells and some features of its distinguishing mode and underlying mechanism.
Collapse
|
46
|
Okada Y, Sabirov RZ, Sato-Numata K, Numata T. Cell Death Induction and Protection by Activation of Ubiquitously Expressed Anion/Cation Channels. Part 1: Roles of VSOR/VRAC in Cell Volume Regulation, Release of Double-Edged Signals and Apoptotic/Necrotic Cell Death. Front Cell Dev Biol 2021; 8:614040. [PMID: 33511120 PMCID: PMC7835517 DOI: 10.3389/fcell.2020.614040] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 12/15/2020] [Indexed: 12/18/2022] Open
Abstract
Cell volume regulation (CVR) is essential for survival and functions of animal cells. Actually, normotonic cell shrinkage and swelling are coupled to apoptotic and necrotic cell death and thus called the apoptotic volume decrease (AVD) and the necrotic volume increase (NVI), respectively. A number of ubiquitously expressed anion and cation channels are involved not only in CVD but also in cell death induction. This series of review articles address the question how cell death is induced or protected with using ubiquitously expressed ion channels such as swelling-activated anion channels, acid-activated anion channels and several types of TRP cation channels including TRPM2 and TRPM7. The Part 1 focuses on the roles of the volume-sensitive outwardly rectifying anion channels (VSOR), also called the volume-regulated anion channel (VRAC), which is activated by cell swelling or reactive oxygen species (ROS) in a manner dependent on intracellular ATP. First we describe phenotypical properties, the molecular identity, and physical pore dimensions of VSOR/VRAC. Second, we highlight the roles of VSOR/VRAC in the release of organic signaling molecules, such as glutamate, glutathione, ATP and cGAMP, that play roles as double-edged swords in cell survival. Third, we discuss how VSOR/VRAC is involved in CVR and cell volume dysregulation as well as in the induction of or protection from apoptosis, necrosis and regulated necrosis under pathophysiological conditions.
Collapse
Affiliation(s)
- Yasunobu Okada
- National Institute for Physiological Sciences, Okazaki, Japan
- Department of Physiology, School of Medicine, Aichi Medical University, Nagakute, Japan
- Department of Physiology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Ravshan Z. Sabirov
- Laboratory of Molecular Physiology, Institute of Biophysics and Biochemistry, National University of Uzbekistan, Tashkent, Uzbekistan
| | - Kaori Sato-Numata
- Japan Society for the Promotion of Science, Tokyo, Japan
- Department of Physiology, School of Medicine, Fukuoka University, Fukuoka, Japan
| | - Tomohiro Numata
- Department of Physiology, School of Medicine, Fukuoka University, Fukuoka, Japan
| |
Collapse
|
47
|
Schnabel B, Kuhrt H, Wiedemann P, Bringmann A, Hollborn M. Osmotic regulation of aquaporin-8 expression in retinal pigment epithelial cells in vitro: Dependence on K ATP channel activation. Mol Vis 2020; 26:797-817. [PMID: 33456300 PMCID: PMC7803296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 12/28/2020] [Indexed: 12/02/2022] Open
Abstract
PURPOSE The expression of aquaporin-8 (AQP8), which plays a crucial role in the maintenance of the cellular fluid and electrolyte balance, was shown to be increased in RPE cells under hyperosmotic conditions. The aim of the present study was to investigate the mechanisms of hyperosmotic AQP8 gene expression and the localization of AQP8 in cultured human RPE cells. METHODS Hyperosmolarity was produced with the addition of 100 mM NaCl or 200 mM sucrose. Hypoxia was induced by cell culture in a 0.2% O2 atmosphere or the addition of the hypoxia mimetic CoCl2. Oxidative stress was induced by the addition of H2O2. Gene expression was determined with real-time RT-PCR analysis. AQP8 protein localization and secretion of VEGF were evaluated with immunocytochemistry, western blotting, and enzyme-linked immunosorbent assay (ELISA). RESULTS Immunocytochemical and western blot data suggest that the AQP8 protein is mainly located in the mitochondria. Extracellular hyperosmolarity, hypoxia, and oxidative stress induced increases in AQP8 gene expression. Hyperosmotic AQP8 gene expression was reduced by inhibitors of the p38 MAPK and PI3K signal transduction pathways, and by JAK2 and PLA2 inhibitors, and was in part mediated by the transcriptional activity of CREB. Hyperosmotic AQP8 gene expression was also reduced by autocrine/paracrine interleukin-1 signaling, the sulfonylureas glibenclamide and glipizide, which are known inhibitors of KATP channel activation, and a pannexin-blocking peptide. The KATP channel opener pinacidil increased the expression of AQP8 under control conditions. The cells contained Kir6.1 and SUR2B gene transcripts and displayed Kir6.1 immunoreactivity. siRNA-mediated knockdown of AQP8 caused increases in hypoxic VEGF gene expression and secretion and decreased cell viability under control, hyperosmotic, and hypoxic conditions. CONCLUSIONS The data indicate that hyperosmotic expression of AQP8 in RPE cells is dependent on the activation of KATP channels. The data suggest that AQP8 activity decreases the hypoxic VEGF expression and improves the viability of RPE cells which may have impact for ischemic retinal diseases like diabetic retinopathy and age-related macular degeneration.
Collapse
Affiliation(s)
- Benjamin Schnabel
- Department of Ophthalmology and Eye Hospital, University of Leipzig, Leipzig, Germany
| | - Heidrun Kuhrt
- Institute of Anatomy, Medical Faculty, University of Leipzig, Germany
| | - Peter Wiedemann
- Department of Ophthalmology and Eye Hospital, University of Leipzig, Leipzig, Germany
| | - Andreas Bringmann
- Department of Ophthalmology and Eye Hospital, University of Leipzig, Leipzig, Germany
| | - Margrit Hollborn
- Department of Ophthalmology and Eye Hospital, University of Leipzig, Leipzig, Germany
| |
Collapse
|
48
|
Bortner CD, Cidlowski JA. Ions, the Movement of Water and the Apoptotic Volume Decrease. Front Cell Dev Biol 2020; 8:611211. [PMID: 33324655 PMCID: PMC7723978 DOI: 10.3389/fcell.2020.611211] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 11/04/2020] [Indexed: 12/20/2022] Open
Abstract
The movement of water across the cell membrane is a natural biological process that occurs during growth, cell division, and cell death. Many cells are known to regulate changes in their cell volume through inherent compensatory regulatory mechanisms. Cells can sense an increase or decrease in their cell volume, and compensate through mechanisms known as a regulatory volume increase (RVI) or decrease (RVD) response, respectively. The transport of sodium, potassium along with other ions and osmolytes allows the movement of water in and out of the cell. These compensatory volume regulatory mechanisms maintain a cell at near constant volume. A hallmark of the physiological cell death process known as apoptosis is the loss of cell volume or cell shrinkage. This loss of cell volume is in stark contrast to what occurs during the accidental cell death process known as necrosis. During necrosis, cells swell or gain water, eventually resulting in cell lysis. Thus, whether a cell gains or loses water after injury is a defining feature of the specific mode of cell death. Cell shrinkage or the loss of cell volume during apoptosis has been termed apoptotic volume decrease or AVD. Over the years, this distinguishing feature of apoptosis has been largely ignored and thought to be a passive occurrence or simply a consequence of the cell death process. However, studies on AVD have defined an underlying movement of ions that result in not only the loss of cell volume, but also the activation and execution of the apoptotic process. This review explores the role ions play in controlling not only the movement of water, but the regulation of apoptosis. We will focus on what is known about specific ion channels and transporters identified to be involved in AVD, and how the movement of ions and water change the intracellular environment leading to stages of cell shrinkage and associated apoptotic characteristics. Finally, we will discuss these concepts as they apply to different cell types such as neurons, cardiomyocytes, and corneal epithelial cells.
Collapse
Affiliation(s)
- Carl D. Bortner
- Signal Transduction Laboratory, Department of Health and Human Services, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, United States
| | - John A. Cidlowski
- Signal Transduction Laboratory, Department of Health and Human Services, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, United States
| |
Collapse
|
49
|
Yurinskaya VE, Vereninov IA, Vereninov AA. Balance of Na +, K +, and Cl - Unidirectional Fluxes in Normal and Apoptotic U937 Cells Computed With All Main Types of Cotransporters. Front Cell Dev Biol 2020; 8:591872. [PMID: 33240889 PMCID: PMC7677585 DOI: 10.3389/fcell.2020.591872] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 09/30/2020] [Indexed: 12/30/2022] Open
Abstract
Fluxes of monovalent ions through the multiple pathways of the plasma membrane are highly interdependent, and their assessment by direct measurement is difficult or even impossible. Computation of the entire flux balance helps to identify partial flows and study the functional expression of individual transporters. Our previous computation of unidirectional fluxes in real cells ignored the ubiquitous cotransporters NKCC and KCC. Here, we present an analysis of the entire balance of unidirectional Na+, K+, and Cl- fluxes through the plasma membrane in human lymphoid U937 cells, taking into account not only the Na/K pump and electroconductive channels but all major types of cotransporters NC, NKCC, and KCC. Our calculations use flux equations based on the fundamental principles of macroscopic electroneutrality of the system, water balance, and the generally accepted thermodynamic dependence of ion fluxes on the driving force, and they do not depend on hypotheses about the molecular structure of the channel and transporters. A complete list of the major inward and outward Na+, K+, and Cl- fluxes is obtained for human lymphoid U937 cells at rest and during changes in the ion and water balance for the first 4 h of staurosporine-induced apoptosis. It is shown how the problem of the inevitable multiplicity of solutions to the flux equations, which arises with an increase in the number of ion pathways, can be solved in real cases by analyzing the ratio of ouabain-sensitive and ouabain-resistant parts of K+ (Rb+) influx (OSOR) and using additional experimental data on the effects of specific inhibitors. It is found that dynamics of changes in the membrane channels and transporters underlying apoptotic changes in the content of ions and water in cells, calculated without taking into account the KCC and NKCC cotransporters, differs only in details from that calculated for cells with KCC and NKCC. The developed approach to the assessment of unidirectional fluxes may be useful for understanding functional expression of ion channels and transporters in other cells under various conditions. Attached software allows reproduction of all calculated data under presented conditions and to study the effects of the condition variation.
Collapse
Affiliation(s)
- Valentina E Yurinskaya
- Laboratory of Cell Physiology, Institute of Cytology, Russian Academy of Sciences, St-Petersburg, Russia
| | - Igor A Vereninov
- Peter the Great St-Petersburg Polytechnic University, St-Petersburg, Russia
| | - Alexey A Vereninov
- Laboratory of Cell Physiology, Institute of Cytology, Russian Academy of Sciences, St-Petersburg, Russia
| |
Collapse
|
50
|
Giordano ME, Caricato R, Lionetto MG. Concentration Dependence of the Antioxidant and Prooxidant Activity of Trolox in HeLa Cells: Involvement in the Induction of Apoptotic Volume Decrease. Antioxidants (Basel) 2020; 9:antiox9111058. [PMID: 33137938 PMCID: PMC7693461 DOI: 10.3390/antiox9111058] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Revised: 10/22/2020] [Accepted: 10/26/2020] [Indexed: 12/14/2022] Open
Abstract
Trolox (6-hydroxy-2,5,7,8-tetramethylchroman-2-carboxylic acid), a hydrophilic analog of vitamin E, is known for its strong antioxidant activity, being a high radical scavenger of peroxyl and alkoxyl radicals. Under particular conditions, Trolox may also exhibit prooxidant properties. The present work aimed at studying the dual antioxidant/prooxidant behavior of Trolox over a wide range of concentrations (from 2.5 to 160 µM) in HeLa cells. In particular, the study addressed the dose-dependent effects of Trolox on the oxidative cell status and vitality of HeLa cells, focusing on the potential role of the vitamin E analog in the induction of one of the first steps of the apoptotic process, Apoptotic Volume Decrease (AVD). In HeLa cells, Trolox showed significant antioxidant activity, expressed as the ability to reduce the endogenous ROS production detected by the ROS-sensitive probe 5-(and-6)-chloromethyl-2′,7′-dichlorodihydrofluorescein diacetate (CM-H2DCFDA), at low concentrations (range: 2.5–15 µM), but exerted a dose-dependent prooxidant effect at higher concentrations after 24 h exposure. The prooxidant effect was paralleled by the reduction in cell viability due to the induction of the apoptotic process. The dual behavior, antioxidant at lower concentrations and prooxidant at higher concentrations, was evident also earlier after 2 h incubation, and it was paralleled by the isotonic shrinkage of the cells, ascribed to AVD. The use of SITS, known Cl− channel blocker, was able to completely inhibit the Trolox-induced isotonic cell shrinkage, demonstrating the involvement of the vitamin E analog in the alteration of cell volume homeostasis and, in turn, in the AVD induction. In conclusion, the study shed light on the concentration dependence of the Trolox antioxidant/prooxidant activity in HeLa cells and revealed its role in the induction of one of the first events of apoptosis, AVD, at high concentrations.
Collapse
|