1
|
Xu P, Zou W, Yin W, Chen G, Gao G, Zhong X. Ion channels research in hPSC-RPE cells: bridging benchwork to clinical applications. J Transl Med 2024; 22:1073. [PMID: 39604931 PMCID: PMC11600670 DOI: 10.1186/s12967-024-05769-5] [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: 08/17/2024] [Accepted: 10/15/2024] [Indexed: 11/29/2024] Open
Abstract
Ion channels in retinal pigment epithelial (RPE) cells are crucial for retinal health and vision functions. Defects in such channels are intricately associated with the development of various retinopathies that cause blindness. Human pluripotent stem cells (hPSC)-derived RPE cells, including those from human-induced pluripotent stem cells (hiPSC) and human embryonic stem cells (hESC), have been used as in vitro models for investigating pathogenic mechanisms and screening potential therapeutic strategies for retinopathies. Therefore, the cellular status of hPSC-RPE cells, including maturity and physiologic functions, have been widely explored. Particularly, research on ion channels in hPSC-RPE cells can lead to the development of more stable models upon which robust investigations and clinical safety assessments can be performed. Moreover, the use of patient-specific hiPSC-RPE cells has significantly accelerated the clinical translation of gene therapy for retinal channelopathies, such as bestrophinopathies. This review consolidates current research on ion channels in hPSC-RPE cells, specifically Kir7.1, Bestrophin-1, CLC-2, and CaV1.3, providing a foundation for future research.
Collapse
Affiliation(s)
- Ping Xu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-sen University, Guangzhou, 510060, China
| | - Weisheng Zou
- Sun Yat-sen University Zhongshan School of Medicine, Guangzhou, 510080, China
| | - Wenjing Yin
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-sen University, Guangzhou, 510060, China
| | - Guifu Chen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-sen University, Guangzhou, 510060, China
| | - Guanjie Gao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-sen University, Guangzhou, 510060, China
| | - Xiufeng Zhong
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-sen University, Guangzhou, 510060, China.
| |
Collapse
|
2
|
Wang S, Li W, Chen M, Cao Y, Lu W, Li X. The retinal pigment epithelium: Functions and roles in ocular diseases. FUNDAMENTAL RESEARCH 2024; 4:1710-1718. [PMID: 39734536 PMCID: PMC11670733 DOI: 10.1016/j.fmre.2023.08.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 08/08/2023] [Accepted: 08/14/2023] [Indexed: 12/31/2024] Open
Abstract
The retinal pigment epithelium (RPE) between retinal photoreceptors and choroidal capillaries is a single layer of cells that are of critical importance to the eye. RPE cells are derived from the anterior neural plate of neuroectodermal origin. Instructed by specific molecules and signaling pathways, the RPE undergoes formation and maturation to form a functional unit together with photoreceptors. The RPE plays crucial roles in maintaining normal retinal structure and functions, such as phagocytosis; barrier function; transportation of nutrients, ions, and water; resistance to oxidative damage; maintenance of visual cycle; and production of various important factors. RPE cells have an efficient metabolic machinery to provide sufficient energy to the retina. RPE dysfunction or atrophy can lead to many retinopathies, such as age-related macular degeneration and proliferative vitreoretinopathy. Here, we discuss RPE development, functions, and roles in various ocular diseases, and the mechanisms involved. A better understanding of the functions of the RPE and related regulatory pathways may help identify novel or better therapies for the treatment of many blinding diseases.
Collapse
Affiliation(s)
- Shasha Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Wanhong Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Min Chen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Yihai Cao
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm 17165, Sweden
| | - Weisi Lu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Xuri Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| |
Collapse
|
3
|
Leclercq B, Mejlachowicz D, Zhu L, Jonet L, Mehanna C, Berdugo M, Irinopoulou T, Jaisser F, Zhao M, Behar-Cohen F. Differential Effect of Aldosterone or Mineralocorticoid Receptor Overexpression on Retinal Inflammation. Invest Ophthalmol Vis Sci 2024; 65:39. [PMID: 39453673 PMCID: PMC11512573 DOI: 10.1167/iovs.65.12.39] [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: 06/28/2024] [Accepted: 09/25/2024] [Indexed: 10/26/2024] Open
Abstract
Purpose Overactivation of the mineralocorticoid receptor (MR) pathway is proinflammatory and contributes to the pathogenesis of diabetic retinopathy and of age-related macular degeneration. Excess of aldosterone, the specific MR ligand, is known to stimulate the production of proinflammatory cytokines and chemokines in extrarenal tissues and cells. In the RPE/choroid complex, aldosterone upregulated genes encoding proteins of the inflammatory response and downregulated genes encoding proteins involved in synaptic activity and neurotransmitters. Yet, cortisol, which is the main MR ligand in the eye, is a potent anti-inflammatory endogenous glucocorticoid. The aim of the present work was to better understand the role of MR activation in retinal inflammation either by acute injection of aldosterone or overexpression of the receptor. Methods We first analyzed the retinal transcriptomic regulation induced by acute intraocular injection of aldosterone in the rat. Then, we used a transgenic rat overexpressing human MR (hMR) to also conduct retinal transcriptomic analysis as well as histological evaluation of the retina, retinal pigment epithelium and choroid. Results Our results show that acute intravitreal injection of aldosterone is highly proinflammatory, upregulating pathways related to microglial activation, oxidative stress, cell death, and downregulating pathways related to glial/neuronal cells activity and proper neurotransmission. On the other hand, hMR overexpression mediates a low-grade inflammation in the retina, associated with notable choroidal inflammation and choroidal neuropathy. Conclusions Consequences of hMR overexpression or aldosterone-injection on retinal transcriptome reveal very distinct pathological mechanisms, with only a few common genes regulated, most of them not being regulated in the same way. Although aldosterone is highly proinflammatory in the retina, MR overactivation in its physiologic milieu mediates a low-grade inflammation in the neural retina.
Collapse
Affiliation(s)
- Bastien Leclercq
- Centre de Recherche des Cordeliers, Inserm UMRS1138, Université Paris Cité, Sorbonne Université, Paris, France
| | - Dan Mejlachowicz
- Centre de Recherche des Cordeliers, Inserm UMRS1138, Université Paris Cité, Sorbonne Université, Paris, France
| | - Linxin Zhu
- Centre de Recherche des Cordeliers, Inserm UMRS1138, Université Paris Cité, Sorbonne Université, Paris, France
| | - Laurent Jonet
- Centre de Recherche des Cordeliers, Inserm UMRS1138, Université Paris Cité, Sorbonne Université, Paris, France
| | - Chadi Mehanna
- Hôpital Américain de Paris, Neuilly-sur-Seine, Paris, France
| | - Marianne Berdugo
- Centre de Recherche des Cordeliers, Inserm UMRS1138, Université Paris Cité, Sorbonne Université, Paris, France
| | | | - Fréderic Jaisser
- Centre de Recherche des Cordeliers, Inserm UMRS1138, Université Paris Cité, Sorbonne Université, Paris, France
| | - Min Zhao
- Centre de Recherche des Cordeliers, Inserm UMRS1138, Université Paris Cité, Sorbonne Université, Paris, France
| | - Francine Behar-Cohen
- Centre de Recherche des Cordeliers, Inserm UMRS1138, Université Paris Cité, Sorbonne Université, Paris, France
- Ophthalmopole Cochin University Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| |
Collapse
|
4
|
Visual pigment-deficient cones survive and mediate visual signaling despite the lack of outer segments. Proc Natl Acad Sci U S A 2022; 119:2115138119. [PMID: 35197287 PMCID: PMC8892328 DOI: 10.1073/pnas.2115138119] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/30/2021] [Indexed: 11/18/2022] Open
Abstract
Rhodopsin and cone opsins are essential for light detection in vertebrate rods and cones, respectively. It is well established that rhodopsin is required for rod phototransduction, outer segment disk morphogenesis, and rod viability. However, the roles of cone opsins are less well understood. In this study, we adopted a loss-of-function approach to investigate the physiological roles of cone opsins in mice. We showed that cones lacking cone opsins do not form normal outer segments due to the lack of disk morphogenesis. Surprisingly, cone opsin-deficient cones survive for at least 12 mo, which is in stark contrast to the rapid rod degeneration observed in rhodopsin-deficient mice, suggesting that cone opsins are dispensable for cone viability. Although the mutant cones do not respond to light directly, they maintain a normal dark current and continue to mediate visual signaling by relaying the rod signal through rod-cone gap junctions. Our work reveals a striking difference between the role of rhodopsin and cone opsins in photoreceptor viability.
Collapse
|
5
|
Chuang JZ, Yang N, Nakajima N, Otsu W, Fu C, Yang HH, Lee MP, Akbar AF, Badea TC, Guo Z, Nuruzzaman A, Hsu KS, Dunaief JL, Sung CH. Retinal pigment epithelium-specific CLIC4 mutant is a mouse model of dry age-related macular degeneration. Nat Commun 2022; 13:374. [PMID: 35042858 PMCID: PMC8766482 DOI: 10.1038/s41467-021-27935-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 12/21/2021] [Indexed: 12/12/2022] Open
Abstract
Age-related macular degeneration (AMD) is the leading cause of blindness among the elderly. Dry AMD has unclear etiology and no treatment. Lipid-rich drusen are the hallmark of dry AMD. An AMD mouse model and insights into drusenogenesis are keys to better understanding of this disease. Chloride intracellular channel 4 (CLIC4) is a pleomorphic protein regulating diverse biological functions. Here we show that retinal pigment epithelium (RPE)-specific Clic4 knockout mice exhibit a full spectrum of functional and pathological hallmarks of dry AMD. Multidisciplinary longitudinal studies of disease progression in these mice support a mechanistic model that links RPE cell-autonomous aberrant lipid metabolism and transport to drusen formation.
Collapse
Affiliation(s)
- Jen-Zen Chuang
- Department of Ophthalmology, Margaret M. Dyson Vision Research Institute, Weill Cornell Medicine, 1300 York Avenue, New York, NY, 10065, USA.
| | - Nan Yang
- Department of Ophthalmology, Margaret M. Dyson Vision Research Institute, Weill Cornell Medicine, 1300 York Avenue, New York, NY, 10065, USA
| | - Nobuyuki Nakajima
- Department of Ophthalmology, Margaret M. Dyson Vision Research Institute, Weill Cornell Medicine, 1300 York Avenue, New York, NY, 10065, USA
- Department of Urology, Tokai University, Kanagawa, Japan
| | - Wataru Otsu
- Department of Ophthalmology, Margaret M. Dyson Vision Research Institute, Weill Cornell Medicine, 1300 York Avenue, New York, NY, 10065, USA
- Department of Biomedical Research Laboratory, Gifu Pharmaceutical University, Gifu, Japan
| | - Cheng Fu
- Department of Ophthalmology, Margaret M. Dyson Vision Research Institute, Weill Cornell Medicine, 1300 York Avenue, New York, NY, 10065, USA
| | - Howard Hua Yang
- The Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Maxwell Ping Lee
- The Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | | | - Tudor Constantin Badea
- National Eye Institute, National institute of Health, Bethesda, MD, USA
- Research and Development Institute, Transilvania University of Brasov, School of Medicine, Brasov, Romania
| | - Ziqi Guo
- Department of Ophthalmology, Margaret M. Dyson Vision Research Institute, Weill Cornell Medicine, 1300 York Avenue, New York, NY, 10065, USA
| | - Afnan Nuruzzaman
- Department of Ophthalmology, Margaret M. Dyson Vision Research Institute, Weill Cornell Medicine, 1300 York Avenue, New York, NY, 10065, USA
| | - Kuo-Shun Hsu
- Department of Ophthalmology, Margaret M. Dyson Vision Research Institute, Weill Cornell Medicine, 1300 York Avenue, New York, NY, 10065, USA
- Sloan Kettering Cancer Institute, New York, NY, USA
| | - Joshua L Dunaief
- FM Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ching-Hwa Sung
- Department of Ophthalmology, Margaret M. Dyson Vision Research Institute, Weill Cornell Medicine, 1300 York Avenue, New York, NY, 10065, USA.
- Department of Cell and Developmental Biology, Weill Cornell Medicine, 1300 York Avenue, New York, NY, 10065, USA.
| |
Collapse
|
6
|
Liu YV, Teng D, Konar GJ, Agakishiev D, Biggs-Garcia A, Harris-Bookman S, McNally MM, Garzon C, Sastry S, Singh MS. Characterization and allogeneic transplantation of a novel transgenic cone-rich donor mouse line. Exp Eye Res 2021; 210:108715. [PMID: 34343570 PMCID: PMC8429259 DOI: 10.1016/j.exer.2021.108715] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Revised: 06/26/2021] [Accepted: 07/30/2021] [Indexed: 01/10/2023]
Abstract
OBJECTIVES Cone photoreceptor transplantation is a potential treatment for macular diseases. The optimal conditions for cone transplantation are poorly understood, partly because of the scarcity of cones in donor mice. To facilitate allogeneic cone photoreceptor transplantation studies in mice, we aimed to create and characterize a donor mouse model containing a cone-rich retina with a cone-specific enhanced green fluorescent protein (EGFP) reporter. METHODS We generated OPN1LW-EGFP/NRL-/- mice by crossing NRL-/- and OPN1LW-EGFP mice. We characterized the anatomical phenotype of OPN1LW-EGFP/NRL-/- mice using multimodal confocal scanning laser ophthalmoscopy (cSLO) imaging, immunohistology, and transmission electron microscopy. We evaluated retinal function using electroretinography (ERG), including 465 and 525 nm chromatic stimuli. Retinal sheets and cell suspensions from OPN1LW-EGFP/NRL-/- mice were transplanted subretinally into immunodeficient Rd1 mice. RESULTS OPN1LW-EGFP/NRL-/- retinas were enriched with OPN1LW-EGFP+ and S-opsin+ cone photoreceptors in a dorsal-ventral distribution gradient. Cone photoreceptors co-expressing OPNL1W-EGFP and S-opsin significantly increased in OPN1LW-EGFP/NRL-/- compared to OPN1LW-EGFP mice. Temporal dynamics of rosette formation in the OPN1LW-EGFP/NRL-/- were similar as the NRL-/- with peak formation at P15. Rosettes formed preferentially in the ventral retina. The outer retina in P35 OPN1LW-EGFP/NRL-/- was thinner than NRL-/- controls. The OPN1LW-EGFP/NRL-/- ERG response amplitudes to 465 nm stimulation were similar to, but to 535 nm stimulation were lower than, NRL-/- controls. Three months after transplantation, the suspension grafts showed greater macroscopic degradation than sheet grafts. Retinal sheet grafts from OPN1LW-EGFP/NRL-/- mice showed greater S-opsin + cone survival than suspension grafts from the same strain. CONCLUSIONS OPN1LW-EGFP/NRL-/- retinae were enriched with S-opsin+ photoreceptors. Sustained expression of EGFP facilitated the longitudinal tracking of transplanted donor cells. Transplantation of cone-rich retinal grafts harvested prior to peak rosette formation survived and differentiated into cone photoreceptor subtypes. Photoreceptor sheet transplantation may promote greater macroscopic graft integrity and S-opsin+ cone survival than cell suspension transplantation, although the mechanism underlying this observation is unclear at present. This novel cone-rich reporter mouse strain may be useful to study the influence of graft structure on cone survival.
Collapse
Affiliation(s)
- Ying V Liu
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Derek Teng
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Gregory J Konar
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Dzhalal Agakishiev
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Alexis Biggs-Garcia
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sarah Harris-Bookman
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Minda M McNally
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Catalina Garzon
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Saalini Sastry
- Zanvyl Krieger School of Arts and Sciences, Johns Hopkins University, Baltimore, MD, USA
| | - Mandeep S Singh
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
7
|
A Literature-Derived Knowledge Graph Augments the Interpretation of Single Cell RNA-seq Datasets. Genes (Basel) 2021; 12:genes12060898. [PMID: 34200671 PMCID: PMC8229796 DOI: 10.3390/genes12060898] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 06/04/2021] [Accepted: 06/04/2021] [Indexed: 01/05/2023] Open
Abstract
Technology to generate single cell RNA-sequencing (scRNA-seq) datasets and tools to annotate them have advanced rapidly in the past several years. Such tools generally rely on existing transcriptomic datasets or curated databases of cell type defining genes, while the application of scalable natural language processing (NLP) methods to enhance analysis workflows has not been adequately explored. Here we deployed an NLP framework to objectively quantify associations between a comprehensive set of over 20,000 human protein-coding genes and over 500 cell type terms across over 26 million biomedical documents. The resultant gene-cell type associations (GCAs) are significantly stronger between a curated set of matched cell type-marker pairs than the complementary set of mismatched pairs (Mann Whitney p = 6.15 × 10−76, r = 0.24; cohen’s D = 2.6). Building on this, we developed an augmented annotation algorithm (single cell Annotation via Literature Encoding, or scALE) that leverages GCAs to categorize cell clusters identified in scRNA-seq datasets, and we tested its ability to predict the cellular identity of 133 clusters from nine datasets of human breast, colon, heart, joint, ovary, prostate, skin, and small intestine tissues. With the optimized settings, the true cellular identity matched the top prediction in 59% of tested clusters and was present among the top five predictions for 91% of clusters. scALE slightly outperformed an existing method for reference data driven automated cluster annotation, and we demonstrate that integration of scALE can meaningfully improve the annotations derived from such methods. Further, contextualization of differential expression analyses with these GCAs highlights poorly characterized markers of well-studied cell types, such as CLIC6 and DNASE1L3 in retinal pigment epithelial cells and endothelial cells, respectively. Taken together, this study illustrates for the first time how the systematic application of a literature-derived knowledge graph can expedite and enhance the annotation and interpretation of scRNA-seq data.
Collapse
|
8
|
Yokoyama R, Kojima H, Takai R, Ohta T, Maeda H, Miyashita K, Mutoh M, Terasaki M. Effects of CLIC4 on Fucoxanthinol-Induced Apoptosis in Human Colorectal Cancer Cells. Nutr Cancer 2020; 73:889-898. [PMID: 33703973 DOI: 10.1080/01635581.2020.1779760] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Fucoxanthin is a marine xanthophyll found in edible brown algae, and a metabolite, fucoxanthinol (FxOH), possesses a potent apoptosis inducing effect in many cancer cells. Chloride intracellular channel 4 (CLIC4) is a member of the CLIC family that plays an important role in cancer development and apoptosis. However, the role of CLIC4 in FxOH-induced apoptosis is not well understood. In this study, we investigated whether CLIC4 affects the apoptotic properties of FxOH in human colorectal cancer (CRC) cells under FxOH treatment. Treating human CRC DLD-1 cells with 5.0 μmol/L FxOH significantly induced apoptosis. FxOH downregulated CLIC4, integrin β1, NHERF2 and pSmad2 (Ser465/467) by 0.6-, 0.7-, 0.7-, and 0.5-fold, respectively, compared with control cells without alteration of Rab35 expression. No colocalizing change was observed in CLIC4-related proteins in either control or FxOH-treated cells. CLIC4 knockdown suppressed cell growth and apoptosis. Interestingly, apoptosis induction by FxOH almost disappeared with CLIC4 knockdown. Our findings suggested that CLIC4 could be involved in FxOH-induced apoptosis in human CRC.
Collapse
Affiliation(s)
- Reo Yokoyama
- School of Pharmaceutical Sciences, Health Science University of Hokkaido, Ishikari-Tobetsu, Japan
| | - Hiroyuki Kojima
- School of Pharmaceutical Sciences, Health Science University of Hokkaido, Ishikari-Tobetsu, Japan
| | - Rie Takai
- Research Institute of Health Sciences, Health Science University of Hokkaido, Ishikari-Tobetsu, Japan
| | - Tohru Ohta
- Research Institute of Health Sciences, Health Science University of Hokkaido, Ishikari-Tobetsu, Japan
| | - Hayato Maeda
- Faculty of Agriculture and Life Science, Hirosaki University, Hirosaki, Aomori, Japan
| | - Kazuo Miyashita
- Laboratory of Biofunctional Material Chemistry, Division of Marine Bioscience, Graduate School of Fisheries Sciences, Hokkaido University, Hakodate, Hokkaido, Japan
| | - Michihiro Mutoh
- Epidemiology and Preventions Group, Center for Public Health Sciences, National Cancer Center, Chuo-ku, Tokyo, Japan
| | - Masaru Terasaki
- School of Pharmaceutical Sciences, Health Science University of Hokkaido, Ishikari-Tobetsu, Japan.,Cancer Prevention Laboratories, Health Sciences University of Hokkaido, Ishikari-Tobetsu, Hokkaido, Japan
| |
Collapse
|
9
|
Altered photoreceptor metabolism in mouse causes late stage age-related macular degeneration-like pathologies. Proc Natl Acad Sci U S A 2020; 117:13094-13104. [PMID: 32434914 PMCID: PMC7293639 DOI: 10.1073/pnas.2000339117] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The main cause for blindness in the elderly worldwide is age-related macular degeneration (AMD). What causes AMD remains unknown. The high metabolic demands of photoreceptors are thought to contribute to disease pathogenesis, yet whether photoreceptor metabolism differs in individuals with AMD has not been determined. Here, we show that photoreceptor metabolism does differ between diseased and nondiseased individuals. Mimicking the metabolic profile of diseased individuals in mouse resulted in the similar advanced pathologies as those that cause blindness in humans. A disease model with photoreceptors as a contributing factor explains also why AMD affects preferentially the macula; it is the region of highest photoreceptor density. The data might open new avenues to study the role of PRs in disease pathogenesis. Age-related macular degeneration (AMD) is the leading cause of blindness in the elderly. While the histopathology of the different disease stages is well characterized, the cause underlying the progression, from the early drusen stage to the advanced macular degeneration stage that leads to blindness, remains unknown. Here, we show that photoreceptors (PRs) of diseased individuals display increased expression of two key glycolytic genes, suggestive of a glucose shortage during disease. Mimicking aspects of this metabolic profile in PRs of wild-type mice by activation of the mammalian target of rapamycin complex 1 (mTORC1) caused early drusen-like pathologies, as well as advanced AMD-like pathologies. Mice with activated mTORC1 in PRs also displayed other early disease features, such as a delay in photoreceptor outer segment (POS) clearance and accumulation of lipofuscin in the retinal-pigmented epithelium (RPE) and of lipoproteins at the Bruch’s membrane (BrM), as well as changes in complement accumulation. Interestingly, formation of drusen-like deposits was dependent on activation of mTORC1 in cones. Both major types of advanced AMD pathologies, including geographic atrophy (GA) and neovascular pathologies, were also seen. Finally, activated mTORC1 in PRs resulted in a threefold reduction in di-docosahexaenoic acid (DHA)–containing phospholipid species. Feeding mice a DHA-enriched diet alleviated most pathologies. The data recapitulate many aspects of the human disease, suggesting that metabolic adaptations in photoreceptors could contribute to disease progression in AMD. Identifying the changes downstream of mTORC1 that lead to advanced pathologies in mouse might present new opportunities to study the role of PRs in AMD pathogenesis.
Collapse
|
10
|
Kars ME, Toklu Y, Arıkan Yorgun M, Neşelioğlu S, Eren F. Electrolyte, Nitric Oxide and Oxidative Stress Levels of Aqueous Humor in Patients with Retinal Detachment and Silicone Oil Tamponade. Curr Eye Res 2020; 45:1443-1450. [PMID: 32228105 DOI: 10.1080/02713683.2020.1749668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Purpose: To enlighten the pathogenesis of silicone oil (SiO)-related complications via measuring aqueous humor levels of electrolytes, nitric oxide (NO), and oxidative stress in SiO, retinal detachment (RD), and control groups. Materials and Methods: In this prospective study, 56 patients were grouped as SiO (n = 29), RD (n = 12), and control (n = 15). The results of pre- and post-operative ophthalmological examinations, aqueous humor electrolyte and NO levels, total antioxidant and oxidant status (TAS, TOS) and oxidative stress index (OSI) were analyzed. Results: SiO group had a higher mean Na+ level compared to controls (144.77 ± 11.48 vs 137.56 ± 6.57 mmol/kg, p = .02). Also, the mean Na+ and Cl- levels of RD group were higher than controls (149.04 ± 12.05 vs. 137.56 ± 6.57 mmol/kg, p = .02, 115.2 ± 7.79 vs 106.23 ± 8.99 mmol/kg, p = .031 for Na+ and Cl-, respectively). The mean NO level of RD group was higher than that of SiO group (51.07 ± 19.56 vs. 34.07 ± 13.84 μM, p = .009). The mean TAS and TOS were lower in SiO group compared to controls (1.92 ± 0.64 vs. 2.49 ± 0.56 μmolTroloxEqv./L, p = .021, 34.98 ± 26.55 vs. 61.46 ± 22.69 μmolH2O2Eqv./L, p = .004 for TAS and TOS, respectively). Intraocular retention time of SiO demonstrated positive correlation with post-operative visual acuity (logMAR) and negative correlation with TOS. Conclusions: Elevated aqueous humor Na+ and Cl- in RD patients might reflect abolished function of ion channels on detached retina. Increased Na+ and lack of NO response to elevated intraocular pressure in SiO-filled eyes might contribute to secondary cataract and glaucoma formation. SiO is associated with low levels of oxidative stress in aqueous humor; however, increased intraocular retention time of SiO is related to a poor visual outcome.
Collapse
Affiliation(s)
- Meltem Ece Kars
- Department of Ophthalmology, Yıldırım Beyazıt University, Ankara Atatürk Training and Research Hospital , Ankara, Turkey
| | - Yasin Toklu
- Department of Ophthalmology, Yıldırım Beyazıt University, Ankara Atatürk Training and Research Hospital , Ankara, Turkey
| | - Mücella Arıkan Yorgun
- Department of Ophthalmology, Ankara Atatürk Training and Research Hospital , Ankara, Turkey
| | - Salim Neşelioğlu
- Department of Biochemistry, Yıldırım Beyazıt University, Ankara Atatürk Training and Research Hospital , Ankara, Turkey
| | - Funda Eren
- Department of Biochemistry, Ankara Atatürk Training and Research Hospital , Ankara, Turkey
| |
Collapse
|
11
|
Carofino BL, Dinshaw KM, Ho PY, Cataisson C, Michalowski AM, Ryscavage A, Alkhas A, Wong NW, Koparde V, Yuspa SH. Head and neck squamous cancer progression is marked by CLIC4 attenuation in tumor epithelium and reciprocal stromal upregulation of miR-142-3p, a novel post-transcriptional regulator of CLIC4. Oncotarget 2019; 10:7251-7275. [PMID: 31921386 PMCID: PMC6944452 DOI: 10.18632/oncotarget.27387] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 12/02/2019] [Indexed: 02/06/2023] Open
Abstract
Chloride intracellular channel 4 (CLIC4) is a tumor suppressor implicated in processes including growth arrest, differentiation, and apoptosis. CLIC4 protein expression is diminished in the tumor parenchyma during progression in squamous cell carcinoma (SCC) and other neoplasms, but the underlying mechanisms have not been identified. Data from The Cancer Genome Atlas suggest this is not driven by genomic alterations. However, screening and functional assays identified miR-142-3p as a regulator of CLIC4. CLIC4 and miR-142-3p expression are inversely correlated in head and neck (HN) SCC and cervical SCC, particularly in advanced stage cancers. In situ localization revealed that stromal immune cells, not tumor cells, are the predominant source of miR-142-3p in HNSCC. Furthermore, HNSCC single-cell expression data demonstrated that CLIC4 is lower in tumor epithelial cells than in stromal fibroblasts and endothelial cells. Tumor-specific downregulation of CLIC4 was confirmed in an SCC xenograft model concurrent with immune cell infiltration and miR-142-3p upregulation. These findings provide the first evidence of CLIC4 regulation by miRNA. Furthermore, the distinct localization of CLIC4 and miR-142-3p within the HNSCC tumor milieu highlight the limitations of bulk tumor analysis and provide critical considerations for both future mechanistic studies and use of miR-142-3p as a HNSCC biomarker.
Collapse
Affiliation(s)
- Brandi L. Carofino
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Kayla M. Dinshaw
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
- Department of Molecular and Cellular Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Pui Yan Ho
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
- Department of Pediatrics, Division of Stem Cell Transplantation and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Christophe Cataisson
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Aleksandra M. Michalowski
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Andrew Ryscavage
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | | | - Nathan W. Wong
- CCR Collaborative Bioinformatics Resource (CCBR), Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
- Advanced Biomedical Computational Science, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Vishal Koparde
- CCR Collaborative Bioinformatics Resource (CCBR), Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
- Advanced Biomedical Computational Science, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Stuart H. Yuspa
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| |
Collapse
|
12
|
Hsu KS, Otsu W, Li Y, Wang HC, Chen S, Tsang SH, Chuang JZ, Sung CH. CLIC4 regulates late endosomal trafficking and matrix degradation activity of MMP14 at focal adhesions in RPE cells. Sci Rep 2019; 9:12247. [PMID: 31439888 PMCID: PMC6706427 DOI: 10.1038/s41598-019-48438-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 08/05/2019] [Indexed: 12/17/2022] Open
Abstract
Dysregulation in the extracellular matrix (ECM) microenvironment surrounding the retinal pigment epithelium (RPE) has been implicated in the etiology of proliferative vitreoretinopathy and age-related macular degeneration. The regulation of ECM remodeling by RPE cells is not well understood. We show that membrane-type matrix metalloproteinase 14 (MMP14) is central to ECM degradation at the focal adhesions in human ARPE19 cells. The matrix degradative activity, but not the assembly, of the focal adhesion is regulated by chloride intracellular channel 4 (CLIC4). CLIC4 is co-localized with MMP14 in the late endosome. CLIC4 regulates the proper sorting of MMP14 into the lumen of the late endosome and its proteolytic activation in lipid rafts. CLIC4 has the newly-identified “late domain” motif that binds to MMP14 and to Tsg101, a component of the endosomal sorting complex required for transport (ESCRT) complex. Unlike the late domain mutant CLIC4, wild-type CLIC4 can rescue the late endosomal sorting defect of MMP14. Finally, CLIC4 knockdown inhibits the apical secretion of MMP2 in polarized human RPE monolayers. These results, taken together, demonstrate that CLIC4 is a novel matrix microenvironment modulator and a novel regulator for late endosomal cargo sorting. Moreover, the late endosomal sorting of MMP14 actively regulates its surface activation in RPE cells.
Collapse
Affiliation(s)
- Kuo-Shun Hsu
- Department of Ophthalmology, Weill Medical College of Cornell University, New York, NY, USA.,Department of Surgery, Colorectal Service and Laboratory of Signal Transduction, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Wataru Otsu
- Department of Ophthalmology, Weill Medical College of Cornell University, New York, NY, USA.,Department of Biomedical Research Laboratory, Gifu Pharmaceutical University, Gifu, Japan
| | - Yao Li
- Department of Ophthalmology, Columbia University, New York, NY, USA
| | - Heuy-Ching Wang
- Ocular Trauma Task Area, US Army Institute of Surgical Research, Joint Base San Antonio-Fort Sam Houston, TX, San Antonio, USA
| | - Shuibing Chen
- Department of Surgery and Department of Biochemistry, Weill Medical College of Cornell University, New York, NY, USA
| | - Stephen H Tsang
- Department of Ophthalmology, Columbia University, New York, NY, USA.,Institute of Human Nutrition, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA.,Jonas Children's Vision Care and Bernard & Shirlee Brown Glaucoma Laboratory, Edward S. Harkness Eye Institute, New York-Presbyterian Hospital, New York, NY, USA.,Department of Pathology & Cell Biology, and Columbia Stem Cell Initiative, Columbia University Medical Center, New York, NY, USA
| | - Jen-Zen Chuang
- Department of Ophthalmology, Weill Medical College of Cornell University, New York, NY, USA
| | - Ching-Hwa Sung
- Department of Ophthalmology, Weill Medical College of Cornell University, New York, NY, USA. .,Department of Cell and Developmental Biology, Weill Medical College of Cornell University, New York, NY, USA.
| |
Collapse
|
13
|
Kim YK, Refaeli I, Brooks CR, Jing P, Gulieva RE, Hughes MR, Cruz NM, Liu Y, Churchill AJ, Wang Y, Fu H, Pippin JW, Lin LY, Shankland SJ, Vogl AW, McNagny KM, Freedman BS. Gene-Edited Human Kidney Organoids Reveal Mechanisms of Disease in Podocyte Development. Stem Cells 2017; 35:2366-2378. [PMID: 28905451 DOI: 10.1002/stem.2707] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Accepted: 09/04/2017] [Indexed: 11/07/2022]
Abstract
A critical event during kidney organogenesis is the differentiation of podocytes, specialized epithelial cells that filter blood plasma to form urine. Podocytes derived from human pluripotent stem cells (hPSC-podocytes) have recently been generated in nephron-like kidney organoids, but the developmental stage of these cells and their capacity to reveal disease mechanisms remains unclear. Here, we show that hPSC-podocytes phenocopy mammalian podocytes at the capillary loop stage (CLS), recapitulating key features of ultrastructure, gene expression, and mutant phenotype. hPSC-podocytes in vitro progressively establish junction-rich basal membranes (nephrin+ podocin+ ZO-1+ ) and microvillus-rich apical membranes (podocalyxin+ ), similar to CLS podocytes in vivo. Ultrastructural, biophysical, and transcriptomic analysis of podocalyxin-knockout hPSCs and derived podocytes, generated using CRISPR/Cas9, reveals defects in the assembly of microvilli and lateral spaces between developing podocytes, resulting in failed junctional migration. These defects are phenocopied in CLS glomeruli of podocalyxin-deficient mice, which cannot produce urine, thereby demonstrating that podocalyxin has a conserved and essential role in mammalian podocyte maturation. Defining the maturity of hPSC-podocytes and their capacity to reveal and recapitulate pathophysiological mechanisms establishes a powerful framework for studying human kidney disease and regeneration. Stem Cells 2017;35:2366-2378.
Collapse
Affiliation(s)
- Yong Kyun Kim
- Division of Nephrology, Department of Medicine, University of Washington School of Medicine, Seattle, Washington, USA
- Kidney Research Institute, Department of Medicine, University of Washington School of Medicine, Seattle, Washington, USA
- Institute for Stem Cell and Regenerative Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, Washington, USA
- Department of Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, Washington, USA
| | - Ido Refaeli
- The Biomedical Research Centre, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Craig R Brooks
- Division of Nephrology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Peifeng Jing
- Department of Electrical Engineering, University of Washington, Seattle, Washington, USA
| | - Ramila E Gulieva
- Division of Nephrology, Department of Medicine, University of Washington School of Medicine, Seattle, Washington, USA
- Kidney Research Institute, Department of Medicine, University of Washington School of Medicine, Seattle, Washington, USA
- Institute for Stem Cell and Regenerative Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, Washington, USA
- Department of Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, Washington, USA
| | - Michael R Hughes
- The Biomedical Research Centre, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Nelly M Cruz
- Division of Nephrology, Department of Medicine, University of Washington School of Medicine, Seattle, Washington, USA
- Kidney Research Institute, Department of Medicine, University of Washington School of Medicine, Seattle, Washington, USA
- Institute for Stem Cell and Regenerative Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, Washington, USA
- Department of Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, Washington, USA
| | - Yannan Liu
- Department of Electrical Engineering, University of Washington, Seattle, Washington, USA
| | - Angela J Churchill
- Division of Nephrology, Department of Medicine, University of Washington School of Medicine, Seattle, Washington, USA
- Kidney Research Institute, Department of Medicine, University of Washington School of Medicine, Seattle, Washington, USA
- Institute for Stem Cell and Regenerative Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, Washington, USA
- Department of Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, Washington, USA
| | - Yuliang Wang
- Institute for Stem Cell and Regenerative Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, Washington, USA
- Paul G. Allen School of Computer Science and Engineering, University of Washington, Seattle, Washington, USA
| | - Hongxia Fu
- Institute for Stem Cell and Regenerative Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, Washington, USA
- Department of Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, Washington, USA
- Division of Hematology, Department of Medicine, University of Washington School of Medicine, Seattle, Washington, USA
- Department of Bioengineering, University of Washington School of Medicine, Seattle, Washington, USA
| | - Jeffrey W Pippin
- Division of Nephrology, Department of Medicine, University of Washington School of Medicine, Seattle, Washington, USA
- Department of Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, Washington, USA
| | - Lih Y Lin
- Department of Electrical Engineering, University of Washington, Seattle, Washington, USA
| | - Stuart J Shankland
- Division of Nephrology, Department of Medicine, University of Washington School of Medicine, Seattle, Washington, USA
- Department of Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, Washington, USA
| | - A Wayne Vogl
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Kelly M McNagny
- The Biomedical Research Centre, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Benjamin S Freedman
- Division of Nephrology, Department of Medicine, University of Washington School of Medicine, Seattle, Washington, USA
- Kidney Research Institute, Department of Medicine, University of Washington School of Medicine, Seattle, Washington, USA
- Institute for Stem Cell and Regenerative Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, Washington, USA
- Department of Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, Washington, USA
| |
Collapse
|
14
|
Argenzio E, Moolenaar WH. Emerging biological roles of Cl- intracellular channel proteins. J Cell Sci 2017; 129:4165-4174. [PMID: 27852828 DOI: 10.1242/jcs.189795] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Cl- intracellular channels (CLICs) are a family of six evolutionary conserved cytosolic proteins that exist in both soluble and membrane-associated forms; however, their functions have long been elusive. Soluble CLICs adopt a glutathione S-transferase (GST)-fold, can induce ion currents in artificial membranes and show oxidoreductase activity in vitro, but there is no convincing evidence of CLICs having such activities in vivo. Recent studies have revealed a role for CLIC proteins in Rho-regulated cortical actin dynamics as well as vesicular trafficking and integrin recycling, the latter of which are under the control of Rab GTPases. In this Commentary, we discuss the emerging roles of CLIC proteins in these processes and the lessons learned from gene-targeting studies. We also highlight outstanding questions regarding the molecular function(s) of these important but still poorly understood proteins.
Collapse
Affiliation(s)
- Elisabetta Argenzio
- Division of Cell Biology, The Netherlands Cancer Institute, Amsterdam 1066CX, The Netherlands
| | - Wouter H Moolenaar
- Division of Cell Biology, The Netherlands Cancer Institute, Amsterdam 1066CX, The Netherlands
| |
Collapse
|
15
|
Hsu KS, Chuang JZ, Sung CH. The Biology of Ciliary Dynamics. Cold Spring Harb Perspect Biol 2017; 9:cshperspect.a027904. [PMID: 28062565 DOI: 10.1101/cshperspect.a027904] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The cilium is an evolutionally conserved apical membrane protrusion that senses and transduces diverse signals to regulate a wide range of cellular activities. The cilium is dynamic in length, structure, and protein composition. Dysregulation of ciliary dynamics has been linked with ciliopathies and other human diseases. The cilium undergoes cell-cycle-dependent assembly and disassembly, with ciliary resorption linked with G1-S transition and cell-fate choice. In the resting cell, the cilium remains sensitive to environmental cues for remodeling during tissue homeostasis and repair. Recent findings further reveal an interplay between the cilium and extracellular vesicles and identify bioactive cilium-derived vesicles, posing a previously unrecognized role of cilia for sending signals. The photoreceptor outer segment is a notable dynamic cilium. A recently discovered protein transport mechanism in photoreceptors maintains light-regulated homeostasis of ciliary length.
Collapse
Affiliation(s)
- Kuo-Shun Hsu
- The Margaret M. Dyson Vision Research Institute, Weill Medical College of Cornell University, New York, New York 10065
| | - Jen-Zen Chuang
- The Margaret M. Dyson Vision Research Institute, Weill Medical College of Cornell University, New York, New York 10065
| | - Ching-Hwa Sung
- Departments of Cell and Developmental Biology, Weill Cornell Medical College, New York, New York 10065
| |
Collapse
|
16
|
Tavasoli M, Al-Momany A, Wang X, Li L, Edwards JC, Ballermann BJ. Both CLIC4 and CLIC5A activate ERM proteins in glomerular endothelium. Am J Physiol Renal Physiol 2016; 311:F945-F957. [PMID: 27582103 DOI: 10.1152/ajprenal.00353.2016] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 08/25/2016] [Indexed: 01/17/2023] Open
Abstract
The chloride intracellular channel (CLIC) 5A is expressed at very high levels in renal glomeruli, in both endothelial cells (EC) and podocytes. CLIC5A stimulates Rac1- and phosphatidylinositol (4,5)-bisphosphate-dependent ERM (ezrin, radixin, moesin) activation. ERM proteins, in turn, function in lumen formation and in the development of actin-based cellular projections. In mice lacking CLIC5A, ERM phosphorylation is profoundly reduced in podocytes, but preserved in glomerular EC. Since glomerular EC also express CLIC4, we reasoned that, if CLIC4 activates ERM proteins like CLIC5A, then CLIC4 could compensate for the CLIC5A loss in glomerular EC. In glomeruli of CLIC5-deficient mice, CLIC4 expression was upregulated and colocalized with moesin and ezrin in glomerular EC, but not in podocytes. In cultured glomerular EC, CLIC4 silencing reduced ERM phosphorylation and cytoskeletal association, and expression of exogenous CLIC4 or CLIC5A rescued ERM de-phosphorylation due to CLIC4 silencing. In mice lacking either CLIC4 or CLIC5, ERM phosphorylation was retained in glomerular EC, but, in mice lacking both CLIC4 and CLIC5, glomerular EC ERM phosphorylation was profoundly reduced. Although glomerular EC fenestrae developed normally in dual CLIC4/CLIC5-deficient mice, the density of fenestrae declined substantially by 8 mo of age, along with the deposition of subendothelial electron-lucent material. The dual CLIC4/CLIC5-deficient mice developed spontaneous proteinuria, glomerular cell proliferation, and matrix deposition. Thus CLIC4 stimulates ERM activation and can compensate for CLIC5A in glomerular EC. The findings indicate that CLIC4/CLIC5A-mediated ERM activation is required for maintenance of the glomerular capillary architecture.
Collapse
Affiliation(s)
- Mahtab Tavasoli
- Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Abass Al-Momany
- Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada; and
| | - Xin Wang
- Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Laiji Li
- Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - John C Edwards
- Department of Internal Medicine, St. Louis University, St. Louis, Missouri
| | - Barbara J Ballermann
- Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada; .,Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada; and
| |
Collapse
|
17
|
Cashman SM, Gracias J, Adhi M, Kumar-Singh R. Adenovirus-mediated delivery of Factor H attenuates complement C3 induced pathology in the murine retina: a potential gene therapy for age-related macular degeneration. J Gene Med 2016; 17:229-43. [PMID: 26369397 DOI: 10.1002/jgm.2865] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Revised: 08/19/2015] [Accepted: 09/11/2015] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Age-related macular degeneration (AMD) is the most common cause of blindness in the elderly, with no therapy available for 90% of patients. Recent genetic evidence implicates activation of complement in the pathogenesis of AMD. We have recently discovered that adenovirus (Ad)-mediated expression of complement component C3 (AdCMVC3) in the murine retina recapitulates many of the pathological features found in human AMD. In the present study, utilizing a gene therapy approach, we examine whether Ad-mediated expression of complement Factor H (AdCAGfH) attenuates AdCMVC3-mediated retinal pathology. METHODS AdCMVC3 was co-injected with either AdCAGfH or a negative control virus expressing green fluorescent protein (AdCMVGFP) into the subretinal space of adult mice. The resulting retinal pathology was analyzed by histology and immunocytochemistry and retinal function was quantified by electroretinography. RESULTS Morphological and functional analyses indicated that AdCMVC3-mediated retinal pathology could be attenuated by AdCAGfH. Specifically, endothelial cell proliferation was reduced by 91% and atrophy of retinal pigment epithelium (RPE) could be attenuated by 69%. AdCAGfH injected eyes exhibited 90-150% greater A-wave and 120-180% greater B-wave amplitudes relative to control eyes. Immunocytochemical analysis of rhodopsin and RPE65 was consistent with the rescue of photoreceptors and RPE in AdCAGfH injected eyes. CONCLUSIONS C3-induced pathology in murine retina can be attenuated by Ad-mediated expression of Factor H. Expression of Factor H is worthy of further study as a potential gene therapy for AMD.
Collapse
Affiliation(s)
- Siobhan M Cashman
- Department of Developmental, Molecular and Chemical Biology and the Department of Ophthalmology, Tufts University School of Medicine, Boston, MA, USA
| | - Jessica Gracias
- Department of Developmental, Molecular and Chemical Biology and the Department of Ophthalmology, Tufts University School of Medicine, Boston, MA, USA
| | - Mehreen Adhi
- Department of Developmental, Molecular and Chemical Biology and the Department of Ophthalmology, Tufts University School of Medicine, Boston, MA, USA
| | - Rajendra Kumar-Singh
- Department of Developmental, Molecular and Chemical Biology and the Department of Ophthalmology, Tufts University School of Medicine, Boston, MA, USA
| |
Collapse
|
18
|
Salao K, Jiang L, Li H, Tsai VWW, Husaini Y, Curmi PMG, Brown LJ, Brown DA, Breit SN. CLIC1 regulates dendritic cell antigen processing and presentation by modulating phagosome acidification and proteolysis. Biol Open 2016; 5:620-30. [PMID: 27113959 PMCID: PMC4874360 DOI: 10.1242/bio.018119] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Intracellular chloride channel protein 1 (CLIC1) participates in inflammatory processes by regulating macrophage phagosomal functions such as pH and proteolysis. Here, we sought to determine if CLIC1 can regulate adaptive immunity by actions on dendritic cells (DCs), the key professional antigen presenting cells. To do this, we first generated bone marrow-derived DCs (BMDCs) from germline CLIC1 gene-deleted (CLIC1−/−) and wild-type (CLIC1+/+) mice, then studied them in vitro and in vivo. We found phagocytosis triggered cytoplasmic CLIC1 translocation to the phagosomal membrane where it regulated phagosomal pH and proteolysis. Phagosomes from CLIC1−/− BMDCs displayed impaired acidification and proteolysis, which could be reproduced if CLIC1+/+, but not CLIC1−/− cells, were treated with IAA94, a CLIC family ion channel blocker. CLIC1−/− BMDC displayed reduced in vitro antigen processing and presentation of full-length myelin oligodendrocyte glycoprotein (MOG) and reduced MOG-induced experimental autoimmune encephalomyelitis. These data suggest that CLIC1 regulates DC phagosomal pH to ensure optimal processing of antigen for presentation to antigen-specific T-cells. Further, they indicate that CLIC1 is a novel therapeutic target to help reduce the adaptive immune response in autoimmune diseases. Summary: DC phagosomes from CLIC1−/− mice display impaired acidification and in vivo and in vitro antigen processing and presentation, revealing CLIC1−/− as a potential therapeutic target in reducing the adaptive immune response in autoimmune diseases.
Collapse
Affiliation(s)
- Kanin Salao
- St Vincent's Centre for Applied Medical Research, St. Vincent's Hospital and University of New South Wales, Sydney, New South Wales 2010, Australia
| | - Lele Jiang
- St Vincent's Centre for Applied Medical Research, St. Vincent's Hospital and University of New South Wales, Sydney, New South Wales 2010, Australia
| | - Hui Li
- St Vincent's Centre for Applied Medical Research, St. Vincent's Hospital and University of New South Wales, Sydney, New South Wales 2010, Australia
| | - Vicky W-W Tsai
- St Vincent's Centre for Applied Medical Research, St. Vincent's Hospital and University of New South Wales, Sydney, New South Wales 2010, Australia
| | - Yasmin Husaini
- St Vincent's Centre for Applied Medical Research, St. Vincent's Hospital and University of New South Wales, Sydney, New South Wales 2010, Australia
| | - Paul M G Curmi
- School of Physics, University of New South Wales, Sydney, New South Wales 2052, Australia
| | - Louise J Brown
- Department of Chemistry and Biomolecular Sciences, Macquarie University, Sydney, New South Wales 2109, Australia
| | - David A Brown
- St Vincent's Centre for Applied Medical Research, St. Vincent's Hospital and University of New South Wales, Sydney, New South Wales 2010, Australia
| | - Samuel N Breit
- St Vincent's Centre for Applied Medical Research, St. Vincent's Hospital and University of New South Wales, Sydney, New South Wales 2010, Australia
| |
Collapse
|
19
|
Chou SY, Hsu KS, Otsu W, Hsu YC, Luo YC, Yeh C, Shehab SS, Chen J, Shieh V, He GA, Marean MB, Felsen D, Ding A, Poppas DP, Chuang JZ, Sung CH. CLIC4 regulates apical exocytosis and renal tube luminogenesis through retromer- and actin-mediated endocytic trafficking. Nat Commun 2016; 7:10412. [PMID: 26786190 PMCID: PMC4736046 DOI: 10.1038/ncomms10412] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Accepted: 12/04/2015] [Indexed: 02/07/2023] Open
Abstract
Chloride intracellular channel 4 (CLIC4) is a mammalian homologue of EXC-4 whose mutation is associated with cystic excretory canals in nematodes. Here we show that CLIC4-null mouse embryos exhibit impaired renal tubulogenesis. In both developing and developed kidneys, CLIC4 is specifically enriched in the proximal tubule epithelial cells, in which CLIC4 is important for luminal delivery, microvillus morphogenesis, and endolysosomal biogenesis. Adult CLIC4-null proximal tubules display aberrant dilation. In MDCK 3D cultures, CLIC4 is expressed on early endosome, recycling endosome and apical transport carriers before reaching its steady-state apical membrane localization in mature lumen. CLIC4 suppression causes impaired apical vesicle coalescence and central lumen formation, a phenotype that can be rescued by Rab8 and Cdc42. Furthermore, we show that retromer- and branched actin-mediated trafficking on early endosome regulates apical delivery during early luminogenesis. CLIC4 selectively modulates retromer-mediated apical transport by negatively regulating the formation of branched actin on early endosomes.
Collapse
Affiliation(s)
- Szu-Yi Chou
- Department of Ophthalmology, Weill Cornell Medical College, 1300 York Avenue, New York City, New York 10065, USA
| | - Kuo-Shun Hsu
- Department of Ophthalmology, Weill Cornell Medical College, 1300 York Avenue, New York City, New York 10065, USA
| | - Wataru Otsu
- Department of Ophthalmology, Weill Cornell Medical College, 1300 York Avenue, New York City, New York 10065, USA
| | - Ya-Chu Hsu
- Department of Ophthalmology, Weill Cornell Medical College, 1300 York Avenue, New York City, New York 10065, USA
| | - Yun-Cin Luo
- Department of Ophthalmology, Weill Cornell Medical College, 1300 York Avenue, New York City, New York 10065, USA
| | - Celine Yeh
- Department of Ophthalmology, Weill Cornell Medical College, 1300 York Avenue, New York City, New York 10065, USA
| | - Syed S. Shehab
- Institute for Pediatric Urology, Department of Urology, Weill Cornell Medical College, 1300 York Avenue, New York City, New York 10065, USA
| | - Jie Chen
- Institute for Pediatric Urology, Department of Urology, Weill Cornell Medical College, 1300 York Avenue, New York City, New York 10065, USA
| | - Vincent Shieh
- Department of Ophthalmology, Weill Cornell Medical College, 1300 York Avenue, New York City, New York 10065, USA
| | - Guo-an He
- Department of Microbiology and Immunology, Weill Cornell Medical College, 1300 York Avenue, New York City, New York 10065, USA
| | - Michael B. Marean
- Institute for Pediatric Urology, Department of Urology, Weill Cornell Medical College, 1300 York Avenue, New York City, New York 10065, USA
| | - Diane Felsen
- Institute for Pediatric Urology, Department of Urology, Weill Cornell Medical College, 1300 York Avenue, New York City, New York 10065, USA
| | - Aihao Ding
- Department of Microbiology and Immunology, Weill Cornell Medical College, 1300 York Avenue, New York City, New York 10065, USA
| | - Dix P. Poppas
- Institute for Pediatric Urology, Department of Urology, Weill Cornell Medical College, 1300 York Avenue, New York City, New York 10065, USA
| | - Jen-Zen Chuang
- Department of Ophthalmology, Weill Cornell Medical College, 1300 York Avenue, New York City, New York 10065, USA
| | - Ching-Hwa Sung
- Department of Ophthalmology, Weill Cornell Medical College, 1300 York Avenue, New York City, New York 10065, USA
- Department of Microbiology and Immunology, Weill Cornell Medical College, 1300 York Avenue, New York City, New York 10065, USA
| |
Collapse
|
20
|
Sauvanet C, Wayt J, Pelaseyed T, Bretscher A. Structure, Regulation, and Functional Diversity of Microvilli on the Apical Domain of Epithelial Cells. Annu Rev Cell Dev Biol 2015; 31:593-621. [DOI: 10.1146/annurev-cellbio-100814-125234] [Citation(s) in RCA: 110] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Cécile Sauvanet
- Department of Molecular Biology and Genetics and Weill Institute for Molecular and Cell Biology, Cornell University, Ithaca, New York 14853;
| | - Jessica Wayt
- Department of Molecular Biology and Genetics and Weill Institute for Molecular and Cell Biology, Cornell University, Ithaca, New York 14853;
| | - Thaher Pelaseyed
- Department of Molecular Biology and Genetics and Weill Institute for Molecular and Cell Biology, Cornell University, Ithaca, New York 14853;
| | - Anthony Bretscher
- Department of Molecular Biology and Genetics and Weill Institute for Molecular and Cell Biology, Cornell University, Ithaca, New York 14853;
| |
Collapse
|
21
|
Systems biology of ion channels and transporters in tumor angiogenesis: An omics view. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2015; 1848:2647-56. [DOI: 10.1016/j.bbamem.2014.10.031] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Revised: 10/09/2014] [Accepted: 10/20/2014] [Indexed: 01/19/2023]
|
22
|
Pfister S, Weber T, Härtig W, Schwerdel C, Elsaesser R, Knuesel I, Fritschy JM. Novel role of cystic fibrosis transmembrane conductance regulator in maintaining adult mouse olfactory neuronal homeostasis. J Comp Neurol 2014; 523:406-30. [PMID: 25271146 DOI: 10.1002/cne.23686] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Revised: 09/25/2014] [Accepted: 09/26/2014] [Indexed: 02/03/2023]
Abstract
The olfactory epithelium (OE) of mice deficient in cystic fibrosis transmembrane conductance regulator (CFTR) exhibits ion transport deficiencies reported in human CF airways, as well as progressive neuronal loss, suggesting defects in olfactory neuron homeostasis. Microvillar cells, a specialized OE cell-subtype, have been implicated in maintaining tissue homeostasis. These cells are endowed with a PLCβ2/IP3 R3/TRPC6 signal transduction pathway modulating release of neuropeptide Y (NPY), which stimulates OE stem cell activity. It is unknown, however, whether microvillar cells also mediate the deficits observed in CFTR-null mice. Here we show that Cftr mRNA in mouse OE is exclusively localized in microvillar cells and CFTR immunofluorescence is coassociated with the scaffolding protein NHERF-1 and PLCβ2 in microvilli. In CFTR-null mice, PLCβ2 was undetectable, NHERF-1 mislocalized, and IP3 R3 more intensely stained, along with increased levels of NPY, suggesting profound alteration of the PLCβ2/IP3 R3 signaling pathway. In addition, basal olfactory neuron homeostasis was altered, shown by increased progenitor cell proliferation, differentiation, and apoptosis and by reduced regenerative capacity following methimazole-induced neurodegeneration. The importance of CFTR in microvillar cells was further underscored by decreased thickness of the OE mucus layer and increased numbers of immune cells within this tissue in CFTR-KO mice. Finally, we observed enhanced immune responses to an acute viral-like infection, as well as hyper-responsiveness to chemical and physical stimuli applied intranasally. Taken together, these data strengthen the notion that microvillar cells in the OE play a key role in maintaining tissue homeostasis and identify several mechanisms underlying this regulation through the multiple functions of CFTR.
Collapse
Affiliation(s)
- Sandra Pfister
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland; Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| | | | | | | | | | | | | |
Collapse
|
23
|
Dynamics of ezrin and EBP50 in regulating microvilli on the apical aspect of epithelial cells. Biochem Soc Trans 2014; 42:189-94. [PMID: 24450650 DOI: 10.1042/bst20130263] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Microvilli are found on the apical surface of epithelial cells. Recent studies on the microvillar proteins ezrin and EBP50 (ezrin/radixin/moesin-binding phosphoprotein of 50 kDa) have revealed both the dynamics and the regulation of microvillar components, and how a dynamic ezrin phosphocycle is necessary to confine microvilli to the apical membrane. In the present review, we first summarize the background to allow us to place these advances in context.
Collapse
|
24
|
Phelps DS, Umstead TM, Floros J. Sex differences in the acute in vivo effects of different human SP-A variants on the mouse alveolar macrophage proteome. J Proteomics 2014; 108:427-44. [PMID: 24954098 DOI: 10.1016/j.jprot.2014.06.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Revised: 05/28/2014] [Accepted: 06/10/2014] [Indexed: 01/06/2023]
Abstract
UNLABELLED Surfactant protein A (SP-A) is involved in lung innate immunity. Humans have two SP-A genes, SFTPA1 and SFTPA2, each with several variants. We examined the in vivo effects of treatment with specific SP-A variants on the alveolar macrophage (AM) proteome from SP-A knockout (KO) mice. KO mice received either SP-A1, SP-A2, or both. AM were collected and their proteomes examined with 2D-DIGE. We identified 90 proteins and categorized them as related to actin/cytoskeleton, oxidative stress, protease balance/chaperones, regulation of inflammation, and regulatory/developmental processes. SP-A1 and SP-A2 had different effects on the AM proteome and these effects differed between sexes. In males more changes occurred in the oxidative stress, protease/chaperones, and inflammation groups with SP-A2 treatment than with SP-A1. In females most SP-A1-induced changes were in the actin/cytoskeletal and oxidative stress groups. We conclude that after acute SP-A1 and SP-A2 treatment, sex-specific differences were observed in the AM proteomes from KO mice, and that these sex differences differ in response to SP-A1 and SP-A2. Females are more responsive to SP-A1, whereas the gene-specific differences in males were minimal. These observations not only demonstrate the therapeutic potential of exogenous SP-A, but also illustrate sex- and gene-specific differences in the response to it. BIOLOGICAL SIGNIFICANCE This study shows that changes occur in the alveolar macrophage proteome in response to a single in vivo treatment with exogenous SP-A1 and/or SP-A2. We demonstrate that SP-A1 and SP-A2 have different effects on the AM proteome and that sex differences exist in the response to each SP-A1 and SP-A2 gene product. This study illustrates the potential of exogenous SP-A1 and SP-A2 treatment for the manipulation of macrophage function and indicates that the specific SP-A variant used for treatment may vary with sex and with the cellular functions being modified. The observed changes may contribute to sex differences in the incidence of some lung diseases.
Collapse
Affiliation(s)
- David S Phelps
- The Center for Host Defense, Inflammation, and Lung Disease (CHILD) Research, Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Todd M Umstead
- The Center for Host Defense, Inflammation, and Lung Disease (CHILD) Research, Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Joanna Floros
- The Center for Host Defense, Inflammation, and Lung Disease (CHILD) Research, Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, PA, USA; Department of Obstetrics and Gynecology, The Pennsylvania State University College of Medicine, Hershey, PA, USA.
| |
Collapse
|
25
|
Padmakumar V, Masiuk KE, Luger D, Lee C, Coppola V, Tessarollo L, Hoover SB, Karavanova I, Buonanno A, Simpson RM, Yuspa SH. Detection of differential fetal and adult expression of chloride intracellular channel 4 (CLIC4) protein by analysis of a green fluorescent protein knock-in mouse line. BMC DEVELOPMENTAL BIOLOGY 2014; 14:24. [PMID: 24886590 PMCID: PMC4073518 DOI: 10.1186/1471-213x-14-24] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Accepted: 05/13/2014] [Indexed: 01/17/2023]
Abstract
Background Chloride Intracellular Channel 4 (CLIC4) is one of seven members in the closely related CLIC protein family. CLIC4 is involved in multiple cellular processes including apoptosis, cellular differentiation, inflammation and endothelial tubulogenesis. Despite over a decade of research, no comprehensive in situ expression analysis of CLIC4 in a living organism has been reported. In order to fulfill this goal, we generated a knock-in mouse to express Green Fluorescent Protein (GFP) from the CLIC4 locus, thus substituting the GFP coding region for CLIC4. We used GFP protein expression to eliminate cross reaction with other CLIC family members. Results We analyzed CLIC4 expression during embryonic development and adult organs. During mid and late gestation, CLIC4 expression is modulated particularly in fetal brain, heart, thymus, liver and kidney as well as in developing brown adipose tissue and stratifying epidermis. In the adult mouse, CLIC4 is highly expressed globally in vascular endothelial cells as well as in liver, lung alveolar septae, pancreatic acini, spermatogonia, renal proximal tubules, cardiomyocytes and thymic epithelial cells. Neural expression included axonal tracks, olfactory bulb, Purkinje cell layer and dentate gyrus. Renal CLIC4 expression was most pronounced in proximal tubules, although altered renal function was not detected in the absence of CLIC4. Myeloid cells and B cells of the spleen are rich in CLIC4 expression as are CD4 and CD8 positive T cells. Conclusions In a comprehensive study detailing CLIC4 expression in situ in a mouse model that excludes cross reaction with other family members, we were able to document previously unreported expression for CLIC4 in developing fetus, particularly the brain. In addition, compartmentalized expression of CLIC4 in specific adult tissues and cells provides a focus to explore potential functions of this protein not addressed previously.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Stuart H Yuspa
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, Bethesda, MD, USA.
| |
Collapse
|
26
|
Argenzio E, Margadant C, Leyton-Puig D, Janssen H, Jalink K, Sonnenberg A, Moolenaar WH. CLIC4 regulates cell adhesion and β1 integrin trafficking. J Cell Sci 2014; 127:5189-203. [DOI: 10.1242/jcs.150623] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Chloride intracellular channel (CLIC) protein CLIC4 exists in both soluble and membrane-associated forms, and is implicated in diverse cellular processes, ranging from ion channel formation to intracellular membrane remodeling. CLIC4 is rapidly recruited to the plasma membrane by lysophosphatidic acid (LPA) and serum, suggesting a possible role for CLIC4 in exocytic-endocytic trafficking. However, the function and subcellular target(s) of CLIC4 remain elusive. Here we show that in HeLa and MDA-MB-231 cells, CLIC4 knockdown decreases cell-matrix adhesion, cell spreading and integrin signalling, while increasing cell motility. LPA stimulates the recruitment of CLIC4 to β1 integrins at the plasma membrane and in Rab35-positive endosomes. CLIC4 is required for both the internalization and the serum/LPA-induced recycling of β1 integrins, but not for EGF receptor trafficking. Furthermore, we show that CLIC4 suppresses Rab35 activity and antagonizes Rab35-dependent regulation of β1-integrin trafficking. Our results define CLIC4 as a regulator of Rab35 activity and serum/LPA-dependent integrin trafficking.
Collapse
|
27
|
Maurya DK, Menini A. Developmental expression of the calcium-activated chloride channels TMEM16A and TMEM16B in the mouse olfactory epithelium. Dev Neurobiol 2013; 74:657-75. [PMID: 24318978 DOI: 10.1002/dneu.22159] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2013] [Revised: 12/02/2013] [Accepted: 12/03/2013] [Indexed: 01/21/2023]
Abstract
Calcium-activated chloride channels are involved in several physiological processes including olfactory perception. TMEM16A and TMEM16B, members of the transmembrane protein 16 family (TMEM16), are responsible for calcium-activated chloride currents in several cells. Both are present in the olfactory epithelium of adult mice, but little is known about their expression during embryonic development. Using immunohistochemistry we studied their expression in the mouse olfactory epithelium at various stages of prenatal development from embryonic day (E) 12.5 to E18.5 as well as in postnatal mice. At E12.5, TMEM16A immunoreactivity was present at the apical surface of the entire olfactory epithelium, but from E16.5 became restricted to a region near the transition zone with the respiratory epithelium, where localized at the apical part of supporting cells and in their microvilli. In contrast, TMEM16B immunoreactivity was present at E14.5 at the apical surface of the entire olfactory epithelium, increased in subsequent days, and localized to the cilia of mature olfactory sensory neurons. These data suggest different functional roles for TMEM16A and TMEM16B in the developing as well as in the postnatal olfactory epithelium. The presence of TMEM16A at the apical part and in microvilli of supporting cells is consistent with a role in the regulation of the chloride ionic composition of the mucus covering the apical surface of the olfactory epithelium, whereas the localization of TMEM16B to the cilia of mature olfactory sensory neurons is consistent with a role in olfactory signal transduction.
Collapse
Affiliation(s)
- Devendra Kumar Maurya
- Laboratory of Olfactory Transduction, SISSA, International School for Advanced Studies, Via Bonomea 265, Trieste, 34136, Italy
| | | |
Collapse
|
28
|
Viswanatha R, Wayt J, Ohouo PY, Smolka MB, Bretscher A. Interactome analysis reveals ezrin can adopt multiple conformational states. J Biol Chem 2013; 288:35437-51. [PMID: 24151071 DOI: 10.1074/jbc.m113.505669] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Ezrin, a member of the ezrin-radixin-moesin family (ERM), is an essential regulator of the structure of microvilli on the apical aspect of epithelial cells. Ezrin provides a linkage between membrane-associated proteins and F-actin, oscillating between active/open and inactive/closed states, and is regulated in part by phosphorylation of a C-terminal threonine. In the open state, ezrin can bind a number of ligands, but in the closed state the ligand-binding sites are inaccessible. In vitro analysis has proposed that there may be a third hyperactivated form of ezrin. To gain a better understanding of ezrin, we conducted an unbiased proteomic analysis of ezrin-binding proteins in an epithelial cell line, Jeg-3. We refined our list of interactors by comparing the interactomes using quantitative mass spectrometry between wild-type ezrin, closed ezrin, open ezrin, and hyperactivated ezrin. The analysis reveals several novel interactors confirmed by their localization to microvilli, as well as a significant class of proteins that bind closed ezrin. Taken together, the data indicate that ezrin can exist in three different conformational states, and different ligands "perceive" ezrin conformational states differently.
Collapse
Affiliation(s)
- Raghuvir Viswanatha
- From the Department of Molecular Biology and Genetics and Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, New York 14853
| | | | | | | | | |
Collapse
|
29
|
Jiang L, Phang JM, Yu J, Harrop SJ, Sokolova AV, Duff AP, Wilk KE, Alkhamici H, Breit SN, Valenzuela SM, Brown LJ, Curmi PMG. CLIC proteins, ezrin, radixin, moesin and the coupling of membranes to the actin cytoskeleton: a smoking gun? BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2013; 1838:643-57. [PMID: 23732235 DOI: 10.1016/j.bbamem.2013.05.025] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2013] [Revised: 05/20/2013] [Accepted: 05/21/2013] [Indexed: 12/20/2022]
Abstract
The CLIC proteins are a highly conserved family of metazoan proteins with the unusual ability to adopt both soluble and integral membrane forms. The physiological functions of CLIC proteins may include enzymatic activity in the soluble form and anion channel activity in the integral membrane form. CLIC proteins are associated with the ERM proteins: ezrin, radixin and moesin. ERM proteins act as cross-linkers between membranes and the cortical actin cytoskeleton. Both CLIC and ERM proteins are controlled by Rho family small GTPases. CLIC proteins, ERM and Rho GTPases act in a concerted manner to control active membrane processes including the maintenance of microvillar structures, phagocytosis and vesicle trafficking. All of these processes involve the interaction of membranes with the underlying cortical actin cytoskeleton. The relationships between Rho GTPases, CLIC proteins, ERM proteins and the membrane:actin cytoskeleton interface are reviewed. Speculative models are proposed involving the formation of localised multi-protein complexes on the membrane surface that assemble via multiple weak interactions. This article is part of a Special Issue entitled: Reciprocal influences between cell cytoskeleton and membrane channels, receptors and transporters. Guest Editor: Jean Claude Hervé.
Collapse
Affiliation(s)
- Lele Jiang
- St Vincent's Centre for Applied Medical Research, St Vincent's Hospital, Sydney, NSW 2010, Australia
| | - Juanita M Phang
- School of Physics, The University of New South Wales, Sydney, NSW 2052, Australia
| | - Jiang Yu
- School of Physics, The University of New South Wales, Sydney, NSW 2052, Australia
| | - Stephen J Harrop
- School of Physics, The University of New South Wales, Sydney, NSW 2052, Australia
| | - Anna V Sokolova
- Australian Nuclear Science and Technology Organisation, Lucas Heights, NSW, Australia
| | - Anthony P Duff
- Australian Nuclear Science and Technology Organisation, Lucas Heights, NSW, Australia
| | - Krystyna E Wilk
- School of Physics, The University of New South Wales, Sydney, NSW 2052, Australia
| | - Heba Alkhamici
- School of Medical and Molecular Biosciences, The University of Technology Sydney, Sydney, NSW 2007, Australia
| | - Samuel N Breit
- St Vincent's Centre for Applied Medical Research, St Vincent's Hospital, Sydney, NSW 2010, Australia
| | - Stella M Valenzuela
- School of Medical and Molecular Biosciences, The University of Technology Sydney, Sydney, NSW 2007, Australia
| | - Louise J Brown
- Department of Chemistry and Biomolecular Sciences, Macquarie University, Sydney, NSW 2109, Australia
| | - Paul M G Curmi
- St Vincent's Centre for Applied Medical Research, St Vincent's Hospital, Sydney, NSW 2010, Australia; School of Physics, The University of New South Wales, Sydney, NSW 2052, Australia.
| |
Collapse
|
30
|
de Smet MD, Julian K. Management of Combined Inflammatory and Rhegmatogenous Retinal Detachment. Retina 2013. [DOI: 10.1016/b978-1-4557-0737-9.00112-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
31
|
|
32
|
Phelps DS, Umstead TM, Floros J. Sex differences in the response of the alveolar macrophage proteome to treatment with exogenous surfactant protein-A. Proteome Sci 2012; 10:44. [PMID: 22824420 PMCID: PMC3570446 DOI: 10.1186/1477-5956-10-44] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2012] [Accepted: 06/29/2012] [Indexed: 01/12/2023] Open
Abstract
Background Male wild type (WT) C57BL/6 mice are less capable of clearing bacteria and surviving from bacterial pneumonia than females. However, if an oxidative stress (acute ozone exposure) occurs before infection, the advantage shifts to males who then survive at higher rates than females. We have previously demonstrated that survival in surfactant protein-A (SP-A) knockout (KO) mice compared to WT was significantly reduced. Because the alveolar macrophage (AM) is pivotal in host defense we hypothesized that SP-A and circulating sex hormones are responsible for these sex differences. We used 2D-DIGE to examine the relationship of sex and SP-A on the AM proteome. The role of SP-A was investigated by treating SP-A KO mice with exogenous SP-A for 6 and 18 hr and studying its effects on the AM proteome. Results We found: 1) less variance between KO males and females than between the WT counterparts by principal component analysis, indicating that SP-A plays a role in sex differences; 2) fewer changes in females when the total numbers of significantly changing protein spots or identified whole proteins in WT or 18 hr SP-A-treated males or females were compared to their respective KO groups; 3) more proteins with functions related to chaperones or protease balance and Nrf2-regulated proteins changed in response to SP-A in females than in males; and 4) the overall pattern of SP-A induced changes in actin-related proteins were similar in both sexes, although males had more significant changes. Conclusions Although there seems to be an interaction between sex and the effect of SP-A, it is unclear what the responsible mechanisms are. However, we found that several of the proteins that were expressed at significantly higher levels in females than in males in WT and/or in KO mice are known to interact with the estrogen receptor and may thus play a role in the SP-A/sex interaction. These include major vault protein, chaperonin subunit 2 (beta) (CCT2), and Rho GDP alpha dissociation inhibitor. We conclude that sex differences exist in the proteome of AM derived from male and female mice and that SP-A contributes to these sex differences.
Collapse
Affiliation(s)
- David S Phelps
- Center for Host defense, Inflammation, and Lung Disease(CHILD) Research and Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA.
| | | | | |
Collapse
|
33
|
Zhong J, Kong X, Zhang H, Yu C, Xu Y, Kang J, Yu H, Yi H, Yang X, Sun L. Inhibition of CLIC4 enhances autophagy and triggers mitochondrial and ER stress-induced apoptosis in human glioma U251 cells under starvation. PLoS One 2012; 7:e39378. [PMID: 22761775 PMCID: PMC3382619 DOI: 10.1371/journal.pone.0039378] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2012] [Accepted: 05/21/2012] [Indexed: 12/19/2022] Open
Abstract
CLIC4/mtCLIC, a chloride intracellular channel protein, localizes to mitochondria, endoplasmic reticulum (ER), nucleus and cytoplasm, and participates in the apoptotic response to stress. Apoptosis and autophagy, the main types of the programmed cell death, seem interconnected under certain stress conditions. However, the role of CLIC4 in autophagy regulation has yet to be determined. In this study, we demonstrate upregulation and nuclear translocation of the CLIC4 protein following starvation in U251 cells. CLIC4 siRNA transfection enhanced autophagy with increased LC3-II protein and puncta accumulation in U251 cells under starvation conditions. In that condition, the interaction of the 14-3-3 epsilon isoform with CLIC4 was abolished and resulted in Beclin 1 overactivation, which further activated autophagy. Moreover, inhibiting the expression of CLIC4 triggered both mitochondrial apoptosis involved in Bax/Bcl-2 and cytochrome c release under starvation and endoplasmic reticulum stress-induced apoptosis with CHOP and caspase-4 upregulation. These results demonstrate that CLIC4 nuclear translocation is an integral part of the cellular response to starvation. Inhibiting the expression of CLIC4 enhances autophagy and contributes to mitochondrial and ER stress-induced apoptosis under starvation.
Collapse
Affiliation(s)
- Jiateng Zhong
- Department of Pathophysiology, Norman Bethune College of Medicine, Jilin University, Changchun, Jilin, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Stuck MW, Conley SM, Naash MI. Defects in the outer limiting membrane are associated with rosette development in the Nrl-/- retina. PLoS One 2012; 7:e32484. [PMID: 22427845 PMCID: PMC3299663 DOI: 10.1371/journal.pone.0032484] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2011] [Accepted: 01/30/2012] [Indexed: 01/23/2023] Open
Abstract
The neural retinal leucine zipper (Nrl) knockout mouse is a widely used model to study cone photoreceptor development, physiology, and molecular biology in the absence of rods. In the Nrl−/− retina, rods are converted into functional cone-like cells. The Nrl−/− retina is characterized by large undulations of the outer nuclear layer (ONL) commonly known as rosettes. Here we explore the mechanism of rosette development in the Nrl−/− retina. We report that rosettes first appear at postnatal day (P)8, and that the structure of nascent rosettes is morphologically distinct from what is seen in the adult retina. The lumen of these nascent rosettes contains a population of aberrant cells protruding into the subretinal space that induce infolding of the ONL. Morphologically adult rosettes do not contain any cell bodies and are first detected at P15. The cells found in nascent rosettes are photoreceptors in origin but lack inner and outer segments. We show that the adherens junctions between photoreceptors and Müller glia which comprise the retinal outer limiting membrane (OLM) are not uniformly formed in the Nrl−/− retina and thus allow protrusion of a population of developing photoreceptors into the subretinal space where their maturation becomes delayed. These data suggest that the rosettes of the Nrl−/− retina arise due to defects in the OLM and delayed maturation of a subset of photoreceptors, and that rods may play an important role in the proper formation of the OLM.
Collapse
Affiliation(s)
| | | | - Muna I. Naash
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
- * E-mail:
| |
Collapse
|
35
|
Phelps DS, Umstead TM, Quintero OA, Yengo CM, Floros J. In vivo rescue of alveolar macrophages from SP-A knockout mice with exogenous SP-A nearly restores a wild type intracellular proteome; actin involvement. Proteome Sci 2011; 9:67. [PMID: 22035134 PMCID: PMC3219558 DOI: 10.1186/1477-5956-9-67] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2011] [Accepted: 10/28/2011] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Mice lacking surfactant protein-A (SP-A-/-; knockout; KO) exhibit increased vulnerability to infection and injury. Although many bronchoalveolar lavage (BAL) protein differences between KO and wild-type (WT) are rapidly reversed in KO after infection, their clinical course is still compromised. We studied the impact of SP-A on the alveolar macrophage (AM) proteome under basal conditions. Male SP-A KO mice were SP-A-treated (5 micrograms/mouse) and sacrificed in 6 or 18 hr. The AM proteomes of KO, SP-A-treated KO, and WT mice were studied by 2D-DIGE coupled with MALDI-ToF/ToF and AM actin distribution was examined by phalloidon staining. RESULTS We observed: a) significant differences from KO in WT or exogenous SP-A-treated in 45 of 76 identified proteins (both increases and decreases). These included actin-related/cytoskeletal proteins (involved in motility, phagocytosis, endocytosis), proteins of intracellular signaling, cell differentiation/regulation, regulation of inflammation, protease/chaperone function, and proteins related to Nrf2-mediated oxidative stress response pathway; b) SP-A-induced changes causing the AM proteome of the KO to resemble that of WT; and c) that SP-A treatment altered cell size and F-actin distribution. CONCLUSIONS These differences are likely to enhance AM function. The observations show for the first time that acute in vivo SP-A treatment of KO mice, under basal or unstimulated conditions, affects the expression of multiple AM proteins, alters F-actin distribution, and can restore much of the WT phenotype. We postulate that the SP-A-mediated expression profile of the AM places it in a state of "readiness" to successfully conduct its innate immune functions and ensure lung health.
Collapse
Affiliation(s)
- David S Phelps
- Center for Host defense, Inflammation, and Lung Disease (CHILD) Research and Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA.
| | | | | | | | | |
Collapse
|
36
|
Abstract
Humans possess the remarkable ability to perceive color, shape, and motion, and to differentiate between light intensities varied by over nine orders of magnitude. Phototransduction—the process in which absorbed photons are converted into electrical responses—is the first stage of visual processing, and occurs in the outer segment, the light-sensing organelle of the photoreceptor cell. Studies of genes linked to human inherited blindness have been crucial to understanding the biogenesis of the outer segment and membrane-trafficking of photoreceptors.
Collapse
Affiliation(s)
- Ching-Hwa Sung
- Dyson Vision Research Institute, Department of Ophthalmology, Weill Cornell Medical College, New York, NY 10065, USA.
| | | |
Collapse
|