1
|
Kaida A, Igarashi Y, Nojima H, Nakayama M, Okada R, Takahashi R, Kobayashi H, Miura M. Uncovering Cell Cycle-Dependent Effects on Cell Survival in Near-Infrared Photoimmunotherapy. Exp Cell Res 2025; 448:114570. [PMID: 40273966 DOI: 10.1016/j.yexcr.2025.114570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 04/21/2025] [Accepted: 04/21/2025] [Indexed: 04/26/2025]
Abstract
Near-infrared photoimmunotherapy (NIR-PIT) is an innovative cancer treatment that selectively induces cell death in cancer cells. Cetuximab-IRdye700DX (Cmab-IR700) conjugate is commonly used in NIR-PIT for head and neck squamous cell carcinoma (HNSCC) because of the frequent overexpression of epidermal growth factor receptor (EGFR) in HNSCC cells. This study examined the influence of cell cycle phases on the response and sensitivity to NIR-PIT in cell lines expressing a fluorescent ubiquitination-based cell cycle indicator (Fucci). The timing of cell death was quantified using time-lapse imaging and a clonogenic assay was used to assess cell survival. The results indicated that the timing of cell death varied among cell lines, with G1-phase cells in HSC3 and CAL33 lines showing slower cell death than those in the S/G2/M phases, whereas HeLa cells exhibited no cell cycle phase-dependent correlation. Cell rupture was predominant in HSC3 and CAL33 cells, whereas HeLa cells exhibited a combination of cell rupture and swelling. Clonogenic survival differed among the cell lines, mirroring variations in the timing of cell death. Among CAL33 and HeLa cells, G1-phase cells demonstrated greater resistance to NIR-PIT. EGFR expression levels, which varied according to cell line and cell cycle phase, were associated with sensitivity to NIR-PIT. Additionally, L-ascorbic acid-treated HeLa cells exhibited increased time to cell death and reduced NIR-PIT sensitivity, which may be due to reactive oxygen species. These findings provide information for the development of NIR-PIT strategies based on cell cycle kinetics to enhance therapeutic outcomes.
Collapse
Affiliation(s)
- Atsushi Kaida
- Department of Dental Radiology and Radiation Oncology, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Japan.
| | - Yuriko Igarashi
- Department of Dental Radiology and Radiation Oncology, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Japan
| | - Hitomi Nojima
- Department of Dental Radiology and Radiation Oncology, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Japan
| | - Mio Nakayama
- Department of Dental Radiology and Radiation Oncology, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Japan; Department of Oral and Maxillofacial Surgical Oncology, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Japan
| | - Ryuhei Okada
- Department of Head and Neck Surgery, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Japan
| | - Ryosuke Takahashi
- Department of Head and Neck Surgery, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Japan
| | - Hisataka Kobayashi
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892-1088, USA
| | - Masahiko Miura
- Department of Dental Radiology and Radiation Oncology, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Japan.
| |
Collapse
|
2
|
Szabo M, Cs. Szabo B, Kurtan K, Varga Z, Panyi G, Nagy P, Zakany F, Kovacs T. Look Beyond Plasma Membrane Biophysics: Revealing Considerable Variability of the Dipole Potential Between Plasma and Organelle Membranes of Living Cells. Int J Mol Sci 2025; 26:889. [PMID: 39940660 PMCID: PMC11816637 DOI: 10.3390/ijms26030889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Revised: 01/14/2025] [Accepted: 01/20/2025] [Indexed: 02/16/2025] Open
Abstract
Due to the lack of measurement techniques suitable for examining compartments of intact, living cells, membrane biophysics is almost exclusively investigated in the plasma membrane despite the fact that its alterations in intracellular organelles may also contribute to disease pathogenesis. Here, we employ a novel, easy-to-use, confocal microscopy-based approach utilizing F66, an environment-sensitive fluorophore in combination with fluorescent organelle markers and quantitative image analysis to determine the magnitude of the molecular order-related dipole potential in the plasma membrane and intracellular organelles of various tumor and neural cell lines. Our comparative analysis demonstrates considerable intracellular variations of the dipole potential that may be large enough to modulate protein functions, with an inward decreasing gradient on the route of the secretory/endocytic pathway (plasma membrane >> lysosome > Golgi > endoplasmic reticulum), whereas mitochondrial membranes are characterized by a dipole potential slightly larger than that of lysosomes. Our approach is suitable and sensitive enough to quantify membrane biophysical properties selectively in intracellular compartments and their comparative analysis in intact, living cells, and, therefore, to identify the affected organelles and potential therapeutic targets in diseases associated with alterations in membrane lipid composition and thus biophysics such as tumors, metabolic, neurodegenerative, or lysosomal storage disorders.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Florina Zakany
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (M.S.); (B.C.S.); (K.K.); (Z.V.); (G.P.); (P.N.)
| | - Tamas Kovacs
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (M.S.); (B.C.S.); (K.K.); (Z.V.); (G.P.); (P.N.)
| |
Collapse
|
3
|
Cs Szabo B, Szabo M, Nagy P, Varga Z, Panyi G, Kovacs T, Zakany F. Novel insights into the modulation of the voltage-gated potassium channel K V1.3 activation gating by membrane ceramides. J Lipid Res 2024; 65:100596. [PMID: 39019344 PMCID: PMC11367112 DOI: 10.1016/j.jlr.2024.100596] [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: 04/26/2024] [Revised: 07/08/2024] [Accepted: 07/10/2024] [Indexed: 07/19/2024] Open
Abstract
Membrane lipids extensively modulate the activation gating of voltage-gated potassium channels (KV), however, much less is known about the mechanisms of ceramide and glucosylceramide actions including which structural element is the main intramolecular target and whether there is any contribution of indirect, membrane biophysics-related mechanisms to their actions. We used two-electrode voltage-clamp fluorometry capable of recording currents and fluorescence signals to simultaneously monitor movements of the pore domain (PD) and the voltage sensor domain (VSD) of the KV1.3 ion channel after attaching an MTS-TAMRA fluorophore to a cysteine introduced into the extracellular S3-S4 loop of the VSD. We observed rightward shifts in the conductance-voltage (G-V) relationship, slower current activation kinetics, and reduced current amplitudes in response to loading the membrane with C16-ceramide (Cer) or C16-glucosylceramide (GlcCer). When analyzing VSD movements, only Cer induced a rightward shift in the fluorescence signal-voltage (F-V) relationship and slowed fluorescence activation kinetics, whereas GlcCer exerted no such effects. These results point at a distinctive mechanism of action with Cer primarily targeting the VSD, while GlcCer only the PD of KV1.3. Using environment-sensitive probes and fluorescence-based approaches, we show that Cer and GlcCer similarly increase molecular order in the inner, hydrophobic regions of bilayers, however, Cer induces a robust molecular reorganization at the membrane-water interface. We propose that this unique ordering effect in the outermost membrane layer in which the main VSD rearrangement involving an outward sliding of the top of S4 occurs can explain the VSD targeting mechanism of Cer, which is unavailable for GlcCer.
Collapse
Affiliation(s)
- Bence Cs Szabo
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Mate Szabo
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Peter Nagy
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Zoltan Varga
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Gyorgy Panyi
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Tamas Kovacs
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.
| | - Florina Zakany
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.
| |
Collapse
|
4
|
Orlikowska-Rzeznik H, Versluis J, Bakker HJ, Piatkowski L. Cholesterol Changes Interfacial Water Alignment in Model Cell Membranes. J Am Chem Soc 2024; 146:13151-13162. [PMID: 38687869 PMCID: PMC11099968 DOI: 10.1021/jacs.4c00474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 04/20/2024] [Accepted: 04/22/2024] [Indexed: 05/02/2024]
Abstract
The nanoscopic layer of water that directly hydrates biological membranes plays a critical role in maintaining the cell structure, regulating biochemical processes, and managing intermolecular interactions at the membrane interface. Therefore, comprehending the membrane structure, including its hydration, is essential for understanding the chemistry of life. While cholesterol is a fundamental lipid molecule in mammalian cells, influencing both the structure and dynamics of cell membranes, its impact on the structure of interfacial water has remained unknown. We used surface-specific vibrational sum-frequency generation spectroscopy to study the effect of cholesterol on the structure and hydration of monolayers of the lipids 1,2-dipalmitoyl-sn-glycero-3-phosphocholine (DPPC), 1,2-dioleoyl-sn-glycero-3-phosphocholine (DOPC), and egg sphingomyelin (SM). We found that for the unsaturated lipid DOPC, cholesterol intercalates in the membrane without significantly changing the orientation of the lipid tails and the orientation of the water molecules hydrating the headgroups of DOPC. In contrast, for the saturated lipids DPPC and SM, the addition of cholesterol leads to clearly enhanced packing and ordering of the hydrophobic tails. It is also observed that the orientation of the water hydrating the lipid headgroups is enhanced upon the addition of cholesterol. These results are important because the orientation of interfacial water molecules influences the cell membranes' dipole potential and the strength and specificity of interactions between cell membranes and peripheral proteins and other biomolecules. The lipid nature-dependent role of cholesterol in altering the arrangement of interfacial water molecules offers a fresh perspective on domain-selective cellular processes, such as protein binding.
Collapse
Affiliation(s)
- Hanna Orlikowska-Rzeznik
- Faculty
of Materials Engineering and Technical Physics, Poznan University of Technology, 60-965 Poznan, Poland
| | - Jan Versluis
- AMOLF,
Ultrafast Spectroscopy, 1098 XG Amsterdam, The Netherlands
| | - Huib J. Bakker
- AMOLF,
Ultrafast Spectroscopy, 1098 XG Amsterdam, The Netherlands
| | - Lukasz Piatkowski
- Faculty
of Materials Engineering and Technical Physics, Poznan University of Technology, 60-965 Poznan, Poland
| |
Collapse
|
5
|
Sarkar P, Chattopadhyay A. Interplay of Cholesterol and Actin in Neurotransmitter GPCR Signaling: Insights from Chronic Cholesterol Depletion Using Statin. ACS Chem Neurosci 2023; 14:3855-3868. [PMID: 37804226 DOI: 10.1021/acschemneuro.3c00472] [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: 10/09/2023] Open
Abstract
Serotonin1A receptors are important neurotransmitter receptors in the G protein-coupled receptor (GPCR) family and modulate a variety of neurological, behavioral, and cognitive functions. We recently showed that chronic cholesterol depletion by statins, potent inhibitors of HMG-CoA reductase (the rate-limiting enzyme in cholesterol biosynthesis), leads to polymerization of the actin cytoskeleton that alters lateral diffusion of serotonin1A receptors. However, cellular signaling by the serotonin1A receptor under chronic cholesterol depletion remains unexplored. In this work, we explored signaling by the serotonin1A receptor under statin-treated condition. We show that cAMP signaling by the receptor is reduced upon lovastatin treatment due to reduction in cholesterol as well as polymerization of the actin cytoskeleton. To the best of our knowledge, these results constitute the first report describing the effect of chronic cholesterol depletion on the signaling of a G protein-coupled neuronal receptor. An important message arising from these results is that it is prudent to include the contribution of actin polymerization while analyzing changes in membrane protein function due to chronic cholesterol depletion by statins. Notably, our results show that whereas actin polymerization acts as a negative regulator of cAMP signaling, cholesterol could act as a positive modulator. These results assume significance in view of reports highlighting symptoms of anxiety and depression in humans upon statin administration and the role of serotonin1A receptors in anxiety and depression. Overall, these results reveal a novel role of actin polymerization induced by chronic cholesterol depletion in modulating GPCR signaling, which could act as a potential therapeutic target.
Collapse
Affiliation(s)
- Parijat Sarkar
- CSIR-Centre for Cellular and Molecular Biology, Uppal Road, Hyderabad 500 007, India
| | - Amitabha Chattopadhyay
- CSIR-Centre for Cellular and Molecular Biology, Uppal Road, Hyderabad 500 007, India
- Academy of Scientific and Innovative Research, Ghaziabad 201 002, India
| |
Collapse
|
6
|
Sarkar P, Chattopadhyay A. Membrane Dipole Potential: An Emerging Approach to Explore Membrane Organization and Function. J Phys Chem B 2022; 126:4415-4430. [PMID: 35696090 DOI: 10.1021/acs.jpcb.2c02476] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Biological membranes are complex organized molecular assemblies of lipids and proteins that provide cells and membrane-bound intracellular organelles their individual identities by morphological compartmentalization. Membrane dipole potential originates from the electrostatic potential difference within the membrane due to the nonrandom arrangement (orientation) of amphiphile and solvent (water) dipoles at the membrane interface. In this Feature Article, we will focus on the measurement of dipole potential using electrochromic fluorescent probes and highlight interesting applications. In addition, we will focus on ratiometric fluorescence microscopic imaging technique to measure dipole potential in cellular membranes, a technique that can be used to address novel problems in cell biology which are otherwise difficult to address using available approaches. We envision that membrane dipole potential could turn out to be a convenient tool in exploring the complex interplay between membrane lipids and proteins and could provide novel insights in membrane organization and function.
Collapse
Affiliation(s)
- Parijat Sarkar
- CSIR-Centre for Cellular and Molecular Biology, Uppal Road, Hyderabad 500 007, India
| | | |
Collapse
|
7
|
Dutta A, Sarkar P, Shrivastava S, Chattopadhyay A. Effect of Hypoxia on the Function of the Human Serotonin 1A Receptor. ACS Chem Neurosci 2022; 13:1456-1466. [PMID: 35467841 DOI: 10.1021/acschemneuro.2c00181] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Cellular hypoxia causes numerous pathophysiological conditions associated with the disruption of oxygen homeostasis. Under oxygen-deficient conditions, cells adapt by controlling the cellular functions to facilitate the judicious use of available oxygen, such as cessation of cell growth and proliferation. In higher eukaryotes, the process of cholesterol biosynthesis is intimately coupled to the availability of oxygen, where the synthesis of one molecule of cholesterol requires 11 molecules of O2. Cholesterol is an essential component of higher eukaryotic membranes and is crucial for the physiological functions of several membrane proteins and receptors. The serotonin1A receptor, an important neurotransmitter G protein-coupled receptor associated with cognition and memory, has previously been shown to depend on cholesterol for its signaling and function. In this work, in order to explore the interdependence of oxygen levels, cholesterol biosynthesis, and the function of the serotonin1A receptor, we developed a cellular hypoxia model to explore the function of the human serotonin1A receptor heterologously expressed in Chinese hamster ovary cells. We observed cell cycle arrest at G1/S phase and the accumulation of lanosterol in cell membranes under hypoxic conditions, thereby validating our cellular model. Interestingly, we observed a significant reduction in ligand binding and disruption of downstream cAMP signaling of the serotonin1A receptor under hypoxic conditions. To the best of our knowledge, our results represent the first report linking the function of the serotonin1A receptor with hypoxia. From a broader perspective, these results contribute to our overall understanding of the molecular basis underlying neurological conditions often associated with hypoxia-induced brain dysfunction.
Collapse
Affiliation(s)
- Aritri Dutta
- CSIR-Centre for Cellular and Molecular Biology, Uppal Road, Hyderabad 500 007, India
| | - Parijat Sarkar
- CSIR-Centre for Cellular and Molecular Biology, Uppal Road, Hyderabad 500 007, India
| | - Sandeep Shrivastava
- CSIR-Centre for Cellular and Molecular Biology, Uppal Road, Hyderabad 500 007, India
| | | |
Collapse
|
8
|
Huang X, Wei X, Qiao S, Zhang X, Li R, Hu S, Mao H, Liu P. Low Density Lipoprotein Receptor (LDLR) and 3-Hydroxy-3-Methylglutaryl Coenzyme a Reductase (HMGCR) Expression are Associated with Platinum-Resistance and Prognosis in Ovarian Carcinoma Patients. Cancer Manag Res 2021; 13:9015-9024. [PMID: 34908877 PMCID: PMC8664653 DOI: 10.2147/cmar.s337873] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 11/22/2021] [Indexed: 12/27/2022] Open
Abstract
Purpose The efficacy of post-surgery platinum-based chemotherapy, the primary choice for the treatment of ovarian cancer (OC), is greatly reduced by the development of drug-resistance. In this study, we investigated the association of expression low-density lipoprotein receptor (LDLR) and 3-hydroxy-3-methylglutaryl-CoA reductase (HMGCR), two cholesterol metabolism-related proteins, in OC tissues and chemoresistance and patient prognosis. Methods Survival analysis using LDLR and HMGCR expression in the ovarian cancer patients using the dataset of Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) was carried out online. A retrospective study was performed on 65 patients who had undergone surgery for ovarian cancer. In addition, patients were divided into 2 groups: platinum resistance group and platinum sensitivity group. Serum lipid metabolism data were collected and analyzed. Protein expressions of LDLR and HMGCR in ovarian cancer tissue were detected by immunohistochemistry. Results Online survival analysis showed that patients with higher LDLR expression had poorer prognosis than those with lower LDLR expression in ovarian cancer cells, while a higher HMGCR expression was associated with better OC prognosis. Overall survival (OS) and disease-free survival (DFS) were lower in patients with higher LDLR levels (OS: P=0.046, DFS: P=0.009). Platinum-resistant patients had higher levels of low-density lipoprotein (LDL) and cholesterol in serum as compared with platinum-sensitive patients (P<0.001). Immunohistochemistry showed that LDLR expression was high and HMGCR was low in platinum-resistant patients. Conclusion The expression of LDLR and HMGCR proteins, involved in the regulation of cholesterol metabolism and the plasma LDL and cholesterol levels were significantly different in platinum-resistant and platinum-sensitive ovarian cancer patients. We postulate that cholesterol metabolic reprogramming might play a role in platinum resistance in ovarian cancer.
Collapse
Affiliation(s)
- Xueyao Huang
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, People's Republic of China.,Shandong University, Jinan, Shandong, 250012, People's Republic of China
| | - Xuan Wei
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, People's Republic of China
| | - Sijing Qiao
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, People's Republic of China.,Shandong University, Jinan, Shandong, 250012, People's Republic of China
| | - Xue Zhang
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, People's Republic of China.,Shandong University, Jinan, Shandong, 250012, People's Republic of China
| | - Rui Li
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, People's Republic of China
| | - Shunxue Hu
- Department of Pathology, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, People's Republic of China
| | - Hongluan Mao
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, People's Republic of China.,Shandong University, Jinan, Shandong, 250012, People's Republic of China
| | - Peishu Liu
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, People's Republic of China.,Shandong University, Jinan, Shandong, 250012, People's Republic of China
| |
Collapse
|
9
|
Metabolic Depletion of Sphingolipids Does Not Alter Cell Cycle Progression in Chinese Hamster Ovary Cells. J Membr Biol 2021; 255:1-12. [PMID: 34392379 DOI: 10.1007/s00232-021-00198-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 08/02/2021] [Indexed: 10/20/2022]
Abstract
The cell cycle is a sequential multi-step process essential for growth and proliferation of cells comprising multicellular organisms. Although a number of proteins are known to modulate the cell cycle, the role of lipids in regulation of cell cycle is still emerging. In our previous work, we monitored the role of cholesterol in cell cycle progression in CHO-K1 cells. Since sphingolipids enjoy a functionally synergistic relationship with membrane cholesterol, in this work, we explored whether sphingolipids could modulate the eukaryotic cell cycle using CHO-K1 cells. Sphingolipids are essential components of eukaryotic cell membranes and are involved in a number of important cellular functions. To comprehensively monitor the role of sphingolipids on cell cycle progression, we carried out metabolic depletion of sphingolipids in CHO-K1 cells using inhibitors (fumonisin B1, myriocin, and PDMP) that block specific steps of the sphingolipid biosynthetic pathway and examined their effect on individual cell cycle phases. Our results show that metabolic inhibitors led to significant reduction in specific sphingolipids, yet such inhibition in sphingolipid biosynthesis did not show any effect on cell cycle progression in CHO-K1 cells. We speculate that any role of sphingolipids on cell cycle progression could be context and cell-type dependent, and cancer cells could be a better choice for monitoring such regulation, since sphingolipids are differentially modulated in these cells.
Collapse
|