1
|
Enderlin J, Rieu Q, Réty S, Vanoni EM, Roux S, Dégardin J, César Q, Augustin S, Nous C, Cai B, Fontaine V, Sennlaub F, Nandrot EF. Retinal atrophy, inflammation, phagocytic and metabolic disruptions develop in the MerTK-cleavage-resistant mouse model. Front Neurosci 2024; 18:1256522. [PMID: 38680449 PMCID: PMC11047123 DOI: 10.3389/fnins.2024.1256522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 03/11/2024] [Indexed: 05/01/2024] Open
Abstract
In the eye, cells from the retinal pigment epithelium (RPE) facing the neurosensory retina exert several functions that are all crucial for long-term survival of photoreceptors (PRs) and vision. Among those, RPE cells phagocytose under a circadian rhythm photoreceptor outer segment (POS) tips that are constantly subjected to light rays and oxidative attacks. The MerTK tyrosine kinase receptor is a key element of this phagocytic machinery required for POS internalization. Recently, we showed that MerTK is subjected to the cleavage of its extracellular domain to finely control its function. In addition, monocytes in retinal blood vessels can migrate inside the inner retina and differentiate into macrophages expressing MerTK, but their role in this context has not been studied yet. We thus investigated the ocular phenotype of MerTK cleavage-resistant (MerTKCR) mice to understand the relevance of this characteristic on retinal homeostasis at the RPE and macrophage levels. MerTKCR retinae appear to develop and function normally, as observed in retinal sections, by electroretinogram recordings and optokinetic behavioral tests. Monitoring of MerTKCR and control mice between the ages of 3 and 18 months showed the development of large degenerative areas in the central retina as early as 4 months when followed monthly by optical coherence tomography (OCT) plus fundus photography (FP)/autofluorescence (AF) detection but not by OCT alone. The degenerative areas were associated with AF, which seems to be due to infiltrated macrophages, as observed by OCT and histology. MerTKCR RPE primary cultures phagocytosed less POS in vitro, while in vivo, the circadian rhythm of POS phagocytosis was deregulated. Mitochondrial function and energy production were reduced in freshly dissected RPE/choroid tissues at all ages, thus showing a metabolic impairment not present in macrophages. RPE anomalies were detected by electron microscopy, including phagosomes retained in the apical area and vacuoles. Altogether, this new mouse model displays a novel phenotype that could prove useful to understanding the interplay between RPE and PRs in inflammatory retinal degenerations and highlights new roles for MerTK in the regulation of the energetic metabolism and the maintenance of the immune privilege in the retina.
Collapse
Affiliation(s)
- Julie Enderlin
- INSERM, CNRS, Institut de la Vision, Therapeutics Department, Sorbonne Université, Paris, France
| | - Quentin Rieu
- INSERM, CNRS, Institut de la Vision, Therapeutics Department, Sorbonne Université, Paris, France
| | - Salomé Réty
- INSERM, CNRS, Institut de la Vision, Therapeutics Department, Sorbonne Université, Paris, France
| | - Elora M. Vanoni
- INSERM, CNRS, Institut de la Vision, Therapeutics Department, Sorbonne Université, Paris, France
| | - Solène Roux
- INSERM, CNRS, Institut de la Vision, Therapeutics Department, Sorbonne Université, Paris, France
| | - Julie Dégardin
- INSERM, CNRS, Institut de la Vision, Therapeutics Department, Sorbonne Université, Paris, France
| | - Quénol César
- INSERM, CNRS, Institut de la Vision, Therapeutics Department, Sorbonne Université, Paris, France
| | - Sébastien Augustin
- INSERM, CNRS, Institut de la Vision, Therapeutics Department, Sorbonne Université, Paris, France
| | - Caroline Nous
- INSERM, CNRS, Institut de la Vision, Therapeutics Department, Sorbonne Université, Paris, France
| | - Bishuang Cai
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Valérie Fontaine
- INSERM, CNRS, Institut de la Vision, Therapeutics Department, Sorbonne Université, Paris, France
| | - Florian Sennlaub
- INSERM, CNRS, Institut de la Vision, Therapeutics Department, Sorbonne Université, Paris, France
| | - Emeline F. Nandrot
- INSERM, CNRS, Institut de la Vision, Therapeutics Department, Sorbonne Université, Paris, France
| |
Collapse
|
2
|
Ghosh M, McGurk F, Norris R, Dong A, Nair S, Jellison E, Murphy P, Verma R, Shapiro LH. The Implant-Induced Foreign Body Response Is Limited by CD13-Dependent Regulation of Ubiquitination of Fusogenic Proteins. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:663-676. [PMID: 38149920 PMCID: PMC10828181 DOI: 10.4049/jimmunol.2300688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 11/29/2023] [Indexed: 12/28/2023]
Abstract
Implanted medical devices, from artificial heart valves and arthroscopic joints to implantable sensors, often induce a foreign body response (FBR), a form of chronic inflammation resulting from the inflammatory reaction to a persistent foreign stimulus. The FBR is characterized by a subset of multinucleated giant cells (MGCs) formed by macrophage fusion, the foreign body giant cells (FBGCs), accompanied by inflammatory cytokines, matrix deposition, and eventually deleterious fibrotic implant encapsulation. Despite efforts to improve biocompatibility, implant-induced FBR persists, compromising the utility of devices and making efforts to control the FBR imperative for long-term function. Controlling macrophage fusion in FBGC formation presents a logical target to prevent implant failure, but the actual contribution of FBGCs to FBR-induced damage is controversial. CD13 is a molecular scaffold, and in vitro induction of CD13KO bone marrow progenitors generates many more MGCs than the wild type, suggesting that CD13 regulates macrophage fusion. In the mesh implant model of FBR, CD13KO mice produced significantly more peri-implant FBGCs with enhanced TGF-β expression and increased collagen deposition versus the wild type. Prior to fusion, increased protrusion and microprotrusion formation accompanies hyperfusion in the absence of CD13. Expression of fusogenic proteins driving cell-cell fusion was aberrantly sustained at high levels in CD13KO MGCs, which we show is due to a novel CD13 function, to our knowledge, regulating ubiquitin/proteasomal protein degradation. We propose CD13 as a physiologic brake limiting aberrant macrophage fusion and the FBR, and it may be a novel therapeutic target to improve the success of implanted medical devices. Furthermore, our data directly implicate FBGCs in the detrimental fibrosis that characterizes the FBR.
Collapse
Affiliation(s)
- Mallika Ghosh
- Centers for Vascular Biology, University of Connecticut Medical School, Farmington, CT
| | - Fraser McGurk
- Centers for Vascular Biology, University of Connecticut Medical School, Farmington, CT
| | - Rachael Norris
- Department of Cell Biology, University of Connecticut Medical School, Farmington, CT
| | - Andy Dong
- Centers for Vascular Biology, University of Connecticut Medical School, Farmington, CT
| | - Sreenidhi Nair
- Centers for Vascular Biology, University of Connecticut Medical School, Farmington, CT
| | - Evan Jellison
- Department of Immunology, University of Connecticut Medical School, Farmington, CT
| | - Patrick Murphy
- Centers for Vascular Biology, University of Connecticut Medical School, Farmington, CT
| | - Rajkumar Verma
- Department of Neuroscience, University of Connecticut Medical School, Farmington, CT
| | - Linda H. Shapiro
- Centers for Vascular Biology, University of Connecticut Medical School, Farmington, CT
| |
Collapse
|
3
|
Ma Y, Yao Y, Meng X, Fu H, Li J, Luan X, Liu M, Liu H, Gu W, Hou L, Meng Q. Hemolymph exosomes inhibit Spiroplasma eriocheiris infection by promoting Tetraspanin-mediated hemocyte phagocytosis in crab. FASEB J 2024; 38:e23433. [PMID: 38226893 DOI: 10.1096/fj.202302182r] [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: 10/25/2023] [Revised: 12/26/2023] [Accepted: 01/04/2024] [Indexed: 01/17/2024]
Abstract
Exosomes released from infected cells are thought to play an important role in the dissemination of pathogens, as well as in host-derived immune molecules during infection. As an intracellular pathogen, Spiroplasma eriocheiris is harmful to multiple crustaceans. However, the immune mechanism of exosomes during Spiroplasma infection has not been investigated. Here, we found exosomes derived from S. eriocheiris-infected crabs could facilitate phagocytosis and apoptosis of hemocytes, resulting in increased crab survival and suppression of Spiroplasma intracellular replication. Proteomic analysis revealed the altered abundance of EsTetraspanin may confer resistance to S. eriocheiris, possibly by mediating hemocyte phagocytosis in Eriocheir sinensis. Specifically, knockdown of EsTetraspanin in E. sinensis increased susceptibility to S. eriocheiris infection and displayed compromised phagocytic ability, whereas overexpression of EsTetraspanin in Drosophila S2 cells inhibited S. eriocheiris infection. Further, it was confirmed that intramuscular injection of recombinant LEL domain of EsTetraspanin reduced the mortality of S. eriocheiris-infected crabs. Blockade with anti-EsTetraspanin serum could exacerbate S. eriocheiris invasion of hemocytes and impair hemocyte phagocytic activity. Taken together, our findings prove for the first time that exosomes modulate phagocytosis to resist pathogenic infection in invertebrates, which is proposed to be mediated by exosomal Tetraspanin, supporting the development of preventative strategies against Spiroplasma infection.
Collapse
Affiliation(s)
- Yubo Ma
- Jiangsu Key Laboratory for Aquatic Crustacean Diseases, College of Marine Science and Engineering, Nanjing Normal University, Nanjing, China
| | - Yu Yao
- Jiangsu Key Laboratory for Aquatic Crustacean Diseases, College of Marine Science and Engineering, Nanjing Normal University, Nanjing, China
| | - Xiang Meng
- Jiangsu Key Laboratory for Aquatic Crustacean Diseases, College of Marine Science and Engineering, Nanjing Normal University, Nanjing, China
| | - Hui Fu
- Jiangsu Key Laboratory for Aquatic Crustacean Diseases, College of Marine Science and Engineering, Nanjing Normal University, Nanjing, China
| | - Jiaying Li
- Jiangsu Key Laboratory for Aquatic Crustacean Diseases, College of Marine Science and Engineering, Nanjing Normal University, Nanjing, China
| | - Xiaoqi Luan
- Jiangsu Key Laboratory for Aquatic Crustacean Diseases, College of Marine Science and Engineering, Nanjing Normal University, Nanjing, China
| | - Min Liu
- Jiangsu Key Laboratory for Aquatic Crustacean Diseases, College of Marine Science and Engineering, Nanjing Normal University, Nanjing, China
| | - Hongli Liu
- Jiangsu Key Laboratory for Aquatic Crustacean Diseases, College of Marine Science and Engineering, Nanjing Normal University, Nanjing, China
| | - Wei Gu
- Jiangsu Key Laboratory for Aquatic Crustacean Diseases, College of Marine Science and Engineering, Nanjing Normal University, Nanjing, China
- Engineering Lab of Henan Province for Aquatic Animal Disease Control, College of Fisheries, Henan Normal University, Xinxiang, China
- Co-Innovation Center for Marine Bio-Industry Technology of Jiangsu Province, Lianyungang, Jiangsu, PR China
| | - Libo Hou
- Jiangsu Key Laboratory for Aquatic Crustacean Diseases, College of Marine Science and Engineering, Nanjing Normal University, Nanjing, China
- Engineering Lab of Henan Province for Aquatic Animal Disease Control, College of Fisheries, Henan Normal University, Xinxiang, China
| | - Qingguo Meng
- Jiangsu Key Laboratory for Aquatic Crustacean Diseases, College of Marine Science and Engineering, Nanjing Normal University, Nanjing, China
- Engineering Lab of Henan Province for Aquatic Animal Disease Control, College of Fisheries, Henan Normal University, Xinxiang, China
- Co-Innovation Center for Marine Bio-Industry Technology of Jiangsu Province, Lianyungang, Jiangsu, PR China
| |
Collapse
|
4
|
Lieffrig SA, Gyimesi G, Mao Y, Finnemann SC. Clearance phagocytosis by the retinal pigment epithelial during photoreceptor outer segment renewal: Molecular mechanisms and relation to retinal inflammation. Immunol Rev 2023; 319:81-99. [PMID: 37555340 PMCID: PMC10615845 DOI: 10.1111/imr.13264] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 07/18/2023] [Indexed: 08/10/2023]
Abstract
Mammalian photoreceptor outer segment renewal is a highly coordinated process that hinges on timed cell signaling between photoreceptor neurons and the adjacent retinal pigment epithelial (RPE). It is a strictly rhythmic, synchronized process that underlies in part circadian regulation. We highlight findings from recently developed methods that quantify distinct phases of outer segment renewal in retinal tissue. At light onset, outer segments expose the conserved "eat-me" signal phosphatidylserine exclusively at their distal, most aged tip. A coordinated two-receptor efferocytosis process follows, in which ligands bridge outer segment phosphatidylserine with the RPE receptors αvβ5 integrin, inducing cytosolic signaling toward Rac1 and focal adhesion kinase/MERTK, and with MERTK directly, additionally inhibiting RhoA/ROCK and thus enabling F-actin dynamics favoring outer segment fragment engulfment. Photoreceptors and RPE persist for life with each RPE cell in the eye servicing dozens of overlying photoreceptors. Thus, RPE cells phagocytose more often and process more material than any other cell type. Mutant mice with impaired outer segment renewal largely retain functional photoreceptors and retinal integrity. However, when anti-inflammatory signaling in the RPE via MERTK or the related TYRO3 is lacking, catastrophic inflammation leads to immune cell infiltration that swiftly destroys the retina causing blindness.
Collapse
Affiliation(s)
- Stephanie A. Lieffrig
- Center for Cancer, Genetic Diseases and Gene Regulation, Department of Biological Sciences, Fordham University, Bronx, NY
| | - Gavin Gyimesi
- Center for Cancer, Genetic Diseases and Gene Regulation, Department of Biological Sciences, Fordham University, Bronx, NY
| | | | - Silvia C. Finnemann
- Center for Cancer, Genetic Diseases and Gene Regulation, Department of Biological Sciences, Fordham University, Bronx, NY
| |
Collapse
|
5
|
Muir V, Sagadiev S, Liu S, Holder U, Armendariz AM, Suchland E, Meitlis I, Camp N, Giltiay N, Tam JM, Garner EC, Wivagg CN, Shows D, James RG, Lacy-Hulbert A, Acharya M. Transcriptomic analysis of pathways associated with ITGAV/alpha(v) integrin-dependent autophagy in human B cells. Autophagy 2023; 19:926-942. [PMID: 36016494 PMCID: PMC9980515 DOI: 10.1080/15548627.2022.2113296] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Macroautophagy/autophagy proteins have been linked with the development of immune-mediated diseases including lupus, but the mechanisms for this are unclear due to the complex roles of these proteins in multiple immune cell types. We have previously shown that a form of noncanonical autophagy induced by ITGAV/alpha(v) integrins regulates B cell activation by viral and self-antigens, in mice. Here, we investigate the involvement of this pathway in B cells from human tissues. Our data reveal that autophagy is specifically induced in the germinal center and memory B cell subpopulations of human tonsils and spleens. Transcriptomic analysis show that the induction of autophagy is related to unique aspects of activated B cells such as mitochondrial metabolism. To understand the function of ITGAV/alpha(v) integrin-dependent autophagy in human B cells, we used CRISPR-mediated knockdown of autophagy genes. Integrating data from primary B cells and knockout cells, we found that ITGAV/alpha(v)-dependent autophagy limits activation of specific pathways related to B cell responses, while promoting others. These data provide new mechanistic links for autophagy and B-cell-mediated immune dysregulation in diseases such as lupus.
Collapse
Affiliation(s)
- Virginia Muir
- Immunology Program, Benaroya Research Institute at Virginia Mason, Seattle, WA, USA
| | - Sara Sagadiev
- Immunology Program, Benaroya Research Institute at Virginia Mason, Seattle, WA, USA.,Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA, USA
| | - Shuozhi Liu
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA, USA
| | - Ursula Holder
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA, USA
| | - Andrea M Armendariz
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA, USA
| | - Emmaline Suchland
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA, USA
| | - Iana Meitlis
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA, USA
| | - Nathan Camp
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA, USA
| | - Natalia Giltiay
- Departments of Rheumatology, University of Washington, Seattle, WA, USA
| | - Jenny M Tam
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA.,Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Ethan C Garner
- Department of Molecular and Cell Biology, Harvard University, Cambridge, MA, USA
| | - Carl N Wivagg
- Department of Molecular and Cell Biology, Harvard University, Cambridge, MA, USA
| | - Donna Shows
- Immunology Program, Benaroya Research Institute at Virginia Mason, Seattle, WA, USA
| | - Richard G James
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA, USA.,Department of Pediatric, University of Washington, Seattle, WA, USA.,Department of Pharmacology, University of Washington, Seattle, WA, USA
| | - Adam Lacy-Hulbert
- Immunology Program, Benaroya Research Institute at Virginia Mason, Seattle, WA, USA.,Department of Immunology, University of Washington, Seattle, WA, USA
| | - Mridu Acharya
- Immunology Program, Benaroya Research Institute at Virginia Mason, Seattle, WA, USA.,Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA, USA.,Department of Pediatric, University of Washington, Seattle, WA, USA
| |
Collapse
|
6
|
Mercau ME, Akalu YT, Mazzoni F, Gyimesi G, Alberto EJ, Kong Y, Hafler BP, Finnemann SC, Rothlin CV, Ghosh S. Inflammation of the retinal pigment epithelium drives early-onset photoreceptor degeneration in Mertk-associated retinitis pigmentosa. SCIENCE ADVANCES 2023; 9:eade9459. [PMID: 36662852 PMCID: PMC9858494 DOI: 10.1126/sciadv.ade9459] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 12/19/2022] [Indexed: 05/17/2023]
Abstract
Severe, early-onset photoreceptor (PR) degeneration associated with MERTK mutations is thought to result from failed phagocytosis by retinal pigment epithelium (RPE). Notwithstanding, the severity and onset of PR degeneration in mouse models of Mertk ablation are determined by the hypomorphic expression or the loss of the Mertk paralog Tyro3. Here, we find that loss of Mertk and reduced expression/loss of Tyro3 led to RPE inflammation even before eye-opening. Incipient RPE inflammation cascaded to involve microglia activation and PR degeneration with monocyte infiltration. Inhibition of RPE inflammation with the JAK1/2 inhibitor ruxolitinib mitigated PR degeneration in Mertk-/- mice. Neither inflammation nor severe, early-onset PR degeneration was observed in mice with defective phagocytosis alone. Thus, inflammation drives severe, early-onset PR degeneration-associated with Mertk loss of function.
Collapse
Affiliation(s)
- Maria E. Mercau
- Department of Immunobiology, School of Medicine, Yale University, New Haven, CT 06520, USA
| | - Yemsratch T. Akalu
- Department of Immunobiology, School of Medicine, Yale University, New Haven, CT 06520, USA
| | - Francesca Mazzoni
- Center for Cancer, Genetic Diseases and Gene Regulation, Department of Biological Sciences, Fordham University, Bronx, NY 10458, USA
| | - Gavin Gyimesi
- Center for Cancer, Genetic Diseases and Gene Regulation, Department of Biological Sciences, Fordham University, Bronx, NY 10458, USA
| | - Emily J. Alberto
- Department of Immunobiology, School of Medicine, Yale University, New Haven, CT 06520, USA
| | - Yong Kong
- Department of Molecular Biophysics and Biochemistry, W. M. Keck Foundation Biotechnology Resource Laboratory, School of Medicine, Yale University, New Haven, CT 06520, USA
| | - Brian P. Hafler
- Department of Ophthalmology, School of Medicine, Yale University, New Haven, CT 06520, USA
- Department of Pathology, School of Medicine, Yale University, New Haven, CT 06520, USA
| | - Silvia C. Finnemann
- Center for Cancer, Genetic Diseases and Gene Regulation, Department of Biological Sciences, Fordham University, Bronx, NY 10458, USA
| | - Carla V. Rothlin
- Department of Immunobiology, School of Medicine, Yale University, New Haven, CT 06520, USA
- Department of Pharmacology, School of Medicine, Yale University, New Haven, CT 06520, USA
| | - Sourav Ghosh
- Department of Pharmacology, School of Medicine, Yale University, New Haven, CT 06520, USA
- Department of Neurology, School of Medicine, Yale University, New Haven, CT 06520, USA
| |
Collapse
|
7
|
Yang R, Liu Q, Zhang M. The Past and Present Lives of the Intraocular Transmembrane Protein CD36. Cells 2022; 12:cells12010171. [PMID: 36611964 PMCID: PMC9818597 DOI: 10.3390/cells12010171] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 12/19/2022] [Accepted: 12/27/2022] [Indexed: 01/03/2023] Open
Abstract
Cluster of differentiation 36 (CD36) belongs to the B2 receptors of the scavenger receptor class B family, which is comprised of single-chain secondary transmembrane glycoproteins. It is present in a variety of cell types, including monocytes, macrophages, microvascular endothelial cells, adipocytes, hepatocytes, platelets, skeletal muscle cells, kidney cells, cardiomyocytes, taste bud cells, and a variety of other cell types. CD36 can be localized on the cell surface, mitochondria, endoplasmic reticulum, and endosomes, playing a role in lipid accumulation, oxidative stress injury, apoptosis, and inflammatory signaling. Recent studies have found that CD36 is expressed in a variety of ocular cells, including retinal pigment epithelium (RPE), retinal microvascular endothelial cells, retinal ganglion cells (RGC), Müller cells, and photoreceptor cells, playing an important role in eye diseases, such as age-related macular degeneration (AMD), diabetic retinopathy (DR), and glaucoma. Therefore, a comprehensive understanding of CD36 function and downstream signaling pathways is of great significance for the prevention and treatment of eye diseases. This article reviews the molecular characteristics, distribution, and function of scavenger receptor CD36 and its role in ophthalmology in order to deepen the understanding of CD36 in eye diseases and provide new ideas for treatment strategies.
Collapse
Affiliation(s)
- Rucui Yang
- Joint Shantou International Eye Center of Shantou University and The Chinese University of Hong Kong, Shantou 515041, China
- Department of Ophthalmology, Shantou University Medical College, Shantou University, Shantou 515041, China
| | - Qingping Liu
- Joint Shantou International Eye Center of Shantou University and The Chinese University of Hong Kong, Shantou 515041, China
| | - Mingzhi Zhang
- Joint Shantou International Eye Center of Shantou University and The Chinese University of Hong Kong, Shantou 515041, China
| |
Collapse
|
8
|
Lewandowski D, Sander CL, Tworak A, Gao F, Xu Q, Skowronska-Krawczyk D. Dynamic lipid turnover in photoreceptors and retinal pigment epithelium throughout life. Prog Retin Eye Res 2022; 89:101037. [PMID: 34971765 PMCID: PMC10361839 DOI: 10.1016/j.preteyeres.2021.101037] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 12/13/2021] [Accepted: 12/15/2021] [Indexed: 12/13/2022]
Abstract
The retinal pigment epithelium-photoreceptor interphase is renewed each day in a stunning display of cellular interdependence. While photoreceptors use photosensitive pigments to convert light into electrical signals, the RPE supports photoreceptors in their function by phagocytizing shed photoreceptor tips, regulating the blood retina barrier, and modulating inflammatory responses, as well as regenerating the 11-cis-retinal chromophore via the classical visual cycle. These processes involve multiple protein complexes, tightly regulated ligand-receptors interactions, and a plethora of lipids and protein-lipids interactions. The role of lipids in maintaining a healthy interplay between the RPE and photoreceptors has not been fully delineated. In recent years, novel technologies have resulted in major advancements in understanding several facets of this interplay, including the involvement of lipids in phagocytosis and phagolysosome function, nutrient recycling, and the metabolic dependence between the two cell types. In this review, we aim to integrate the complex role of lipids in photoreceptor and RPE function, emphasizing the dynamic exchange between the cells as well as discuss how these processes are affected in aging and retinal diseases.
Collapse
Affiliation(s)
- Dominik Lewandowski
- Department of Ophthalmology, Center for Translational Vision Research, School of Medicine, UC Irvine, Irvine, CA, USA
| | - Christopher L Sander
- Department of Ophthalmology, Center for Translational Vision Research, School of Medicine, UC Irvine, Irvine, CA, USA; Department of Pharmacology, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Aleksander Tworak
- Department of Ophthalmology, Center for Translational Vision Research, School of Medicine, UC Irvine, Irvine, CA, USA
| | - Fangyuan Gao
- Department of Ophthalmology, Center for Translational Vision Research, School of Medicine, UC Irvine, Irvine, CA, USA
| | - Qianlan Xu
- Department of Physiology and Biophysics, Center for Translational Vision Research, School of Medicine, UC Irvine, Irvine, CA, USA
| | - Dorota Skowronska-Krawczyk
- Department of Ophthalmology, Center for Translational Vision Research, School of Medicine, UC Irvine, Irvine, CA, USA; Department of Physiology and Biophysics, Center for Translational Vision Research, School of Medicine, UC Irvine, Irvine, CA, USA.
| |
Collapse
|
9
|
Rieu Q, Bougoüin A, Zagar Y, Chatagnon J, Hamieh A, Enderlin J, Huby T, Nandrot EF. Pleiotropic Roles of Scavenger Receptors in Circadian Retinal Phagocytosis: A New Function for Lysosomal SR-B2/LIMP-2 at the RPE Cell Surface. Int J Mol Sci 2022; 23:ijms23073445. [PMID: 35408805 PMCID: PMC8998831 DOI: 10.3390/ijms23073445] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 03/08/2022] [Accepted: 03/17/2022] [Indexed: 11/29/2022] Open
Abstract
The retinal phagocytic machinery resembles the one used by macrophages to clear apoptotic cells. However, in the retina, the permanent contact between photoreceptor outer segments (POS) and retinal pigment epithelial (RPE) cells requires a tight control of this circadian machinery. In addition to the known receptors synchronizing POS internalization, several others are expressed by RPE cells. Notably, scavenger receptor CD36 has been shown to intervene in the internalization speed. We thus investigated members of the scavenger receptor family class A SR-AI and MARCO and class B CD36, SR-BI and SR-B2/LIMP-2 using immunoblotting, immunohisto- and immunocytochemistry, lipid raft flotation gradients, phagocytosis assays after siRNA/antibody inhibition, RT-qPCR and western blot analysis along the light:dark cycle. All receptors were expressed by RPE cell lines and tissues and colocalized with POS, except SR-BI. All receptors were associated with lipid rafts, and even more upon POS challenge. SR-B2/LIMP-2 inhibition suggested a role in the control of the internalization speed similar to CD36. In vivo, MARCO and CD36 displayed rhythmic gene and protein expression patterns concomitant with the phagocytic peak. Taken together, our results indicate that CD36 and SR-B2/LIMP-2 play a direct regulatory role in POS phagocytosis dynamics, while the others such as MARCO might participate in POS clearance by RPE cells either as co-receptors or via an indirect process.
Collapse
Affiliation(s)
- Quentin Rieu
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012 Paris, France; (Q.R.); (A.B.); (Y.Z.); (J.C.); (A.H.); (J.E.)
| | - Antoine Bougoüin
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012 Paris, France; (Q.R.); (A.B.); (Y.Z.); (J.C.); (A.H.); (J.E.)
| | - Yvrick Zagar
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012 Paris, France; (Q.R.); (A.B.); (Y.Z.); (J.C.); (A.H.); (J.E.)
| | - Jonathan Chatagnon
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012 Paris, France; (Q.R.); (A.B.); (Y.Z.); (J.C.); (A.H.); (J.E.)
| | - Abdallah Hamieh
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012 Paris, France; (Q.R.); (A.B.); (Y.Z.); (J.C.); (A.H.); (J.E.)
| | - Julie Enderlin
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012 Paris, France; (Q.R.); (A.B.); (Y.Z.); (J.C.); (A.H.); (J.E.)
| | - Thierry Huby
- Sorbonne Université, INSERM, UMR-S 1166, F-75013 Paris, France;
| | - Emeline F. Nandrot
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012 Paris, France; (Q.R.); (A.B.); (Y.Z.); (J.C.); (A.H.); (J.E.)
- Correspondence: ; Tel.: +33-1-5346-2541; Fax: +33-1-5346-2602
| |
Collapse
|
10
|
Kummer D, Steinbacher T, Thölmann S, Schwietzer MF, Hartmann C, Horenkamp S, Demuth S, Peddibhotla SS, Brinkmann F, Kemper B, Schnekenburger J, Brandt M, Betz T, Liashkovich I, Kouzel IU, Shahin V, Corvaia N, Rottner K, Tarbashevich K, Raz E, Greune L, Schmidt MA, Gerke V, Ebnet K. A JAM-A-tetraspanin-αvβ5 integrin complex regulates contact inhibition of locomotion. J Biophys Biochem Cytol 2022; 221:213070. [PMID: 35293964 PMCID: PMC8931538 DOI: 10.1083/jcb.202105147] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 12/16/2021] [Accepted: 01/21/2022] [Indexed: 12/30/2022] Open
Abstract
Contact inhibition of locomotion (CIL) is a process that regulates cell motility upon collision with other cells. Improper regulation of CIL has been implicated in cancer cell dissemination. Here, we identify the cell adhesion molecule JAM-A as a central regulator of CIL in tumor cells. JAM-A is part of a multimolecular signaling complex in which tetraspanins CD9 and CD81 link JAM-A to αvβ5 integrin. JAM-A binds Csk and inhibits the activity of αvβ5 integrin-associated Src. Loss of JAM-A results in increased activities of downstream effectors of Src, including Erk1/2, Abi1, and paxillin, as well as increased activity of Rac1 at cell-cell contact sites. As a consequence, JAM-A-depleted cells show increased motility, have a higher cell-matrix turnover, and fail to halt migration when colliding with other cells. We also find that proper regulation of CIL depends on αvβ5 integrin engagement. Our findings identify a molecular mechanism that regulates CIL in tumor cells and have implications on tumor cell dissemination.
Collapse
Affiliation(s)
- Daniel Kummer
- Institute-associated Research Group “Cell Adhesion and Cell Polarity”, Münster, Germany,Institute of Medical Biochemistry, ZMBE, University of Münster, Münster, Germany,Interdisciplinary Clinical Research Center (IZKF), University of Münster, Münster, Germany
| | - Tim Steinbacher
- Institute-associated Research Group “Cell Adhesion and Cell Polarity”, Münster, Germany,Institute of Medical Biochemistry, ZMBE, University of Münster, Münster, Germany
| | - Sonja Thölmann
- Institute-associated Research Group “Cell Adhesion and Cell Polarity”, Münster, Germany,Institute of Medical Biochemistry, ZMBE, University of Münster, Münster, Germany
| | - Mariel Flavia Schwietzer
- Institute-associated Research Group “Cell Adhesion and Cell Polarity”, Münster, Germany,Institute of Medical Biochemistry, ZMBE, University of Münster, Münster, Germany
| | - Christian Hartmann
- Institute-associated Research Group “Cell Adhesion and Cell Polarity”, Münster, Germany,Institute of Medical Biochemistry, ZMBE, University of Münster, Münster, Germany
| | - Simone Horenkamp
- Institute-associated Research Group “Cell Adhesion and Cell Polarity”, Münster, Germany,Institute of Medical Biochemistry, ZMBE, University of Münster, Münster, Germany
| | - Sabrina Demuth
- Institute-associated Research Group “Cell Adhesion and Cell Polarity”, Münster, Germany,Institute of Medical Biochemistry, ZMBE, University of Münster, Münster, Germany
| | - Swetha S.D. Peddibhotla
- Institute-associated Research Group “Cell Adhesion and Cell Polarity”, Münster, Germany,Institute of Medical Biochemistry, ZMBE, University of Münster, Münster, Germany
| | - Frauke Brinkmann
- Institute-associated Research Group “Cell Adhesion and Cell Polarity”, Münster, Germany,Institute of Medical Biochemistry, ZMBE, University of Münster, Münster, Germany
| | - Björn Kemper
- Biomedical Technology Center, Medical Faculty, University of Münster, Münster, Germany
| | - Jürgen Schnekenburger
- Biomedical Technology Center, Medical Faculty, University of Münster, Münster, Germany
| | - Matthias Brandt
- Institute-associated Research Group “Mechanics of Cellular Systems”, Institute of Cell Biology, ZMBE, University of Münster, Münster, Germany
| | - Timo Betz
- Institute-associated Research Group “Mechanics of Cellular Systems”, Institute of Cell Biology, ZMBE, University of Münster, Münster, Germany
| | - Ivan Liashkovich
- Institute of Physiology II, University of Münster, Münster, Germany
| | - Ivan U. Kouzel
- Sars International Centre for Marine Molecular Biology University of Bergen Thormøhlensgt, Bergen, Norway
| | - Victor Shahin
- Institute of Physiology II, University of Münster, Münster, Germany
| | - Nathalie Corvaia
- Centre d’Immunologie Pierre Fabre (CIPF), Saint-Julien-en-Genevois, France
| | - Klemens Rottner
- Divison of Molecular Cell Biology, Zoological Institute, Technical University Braunschweig, Braunschweig, Germany,Molecular Cell Biology Group, Department of Cell Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | | | - Erez Raz
- Institute of Cell Biology, ZMBE, University of Münster, Münster, Germany,Cells-in-Motion Cluster of Excellence (EXC 1003—CiM), University of Münster, 48419 Münster, Germany
| | - Lilo Greune
- Institute of Infectiology, ZMBE, University of Münster, Münster, Germany
| | | | - Volker Gerke
- Institute of Medical Biochemistry, ZMBE, University of Münster, Münster, Germany,Cells-in-Motion Cluster of Excellence (EXC 1003—CiM), University of Münster, 48419 Münster, Germany
| | - Klaus Ebnet
- Institute-associated Research Group “Cell Adhesion and Cell Polarity”, Münster, Germany,Institute of Medical Biochemistry, ZMBE, University of Münster, Münster, Germany,Interdisciplinary Clinical Research Center (IZKF), University of Münster, Münster, Germany,Cells-in-Motion Cluster of Excellence (EXC 1003—CiM), University of Münster, 48419 Münster, Germany
| |
Collapse
|
11
|
Scavenger receptor MARCO contributes to macrophage phagocytosis and clearance of tumor cells. Exp Cell Res 2021; 408:112862. [PMID: 34626585 DOI: 10.1016/j.yexcr.2021.112862] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Revised: 09/04/2021] [Accepted: 10/04/2021] [Indexed: 11/22/2022]
Abstract
Macrophage receptor with collagenous structure (MARCO) is a member of the class A scavenger receptor family which is expressed on the cell surface of macrophages. It is well known that MARCO avidly binds to unopsonized pathogens, leading to its ingestion by macrophages. However, the role of MARCO in the recognition and phagocytosis of tumor cells by macrophages remains poorly understood. In this study, we used lentiviral technology to knockdown and overexpress MARCO and investigated the ability of macrophages to phagocytose tumor cells. Our results showed that MARCO expression was correlated with the ability of macrophages to carry on phagocytosis. MARCO knockdown led to significant decreases in the number of engulfment pseudopodia of macrophages and inhibition of the phagocytosis of tumor cells. On the other hand, MARCO overexpression elevated activity of SYK, PI3K and Rac1 in macrophages, which led to changes in macrophage morphology and enhanced phagocytosis by promoting formation of stress fibers and pseudopodia. By Co-IP analysis we showed that MARCO directly binds to the β5 integrin of SL4 tumor cells. In summary, these results demonstrated the important role for MARCO in demonstrated tumor cells uptake and clearance by macrophages.
Collapse
|
12
|
Flores‐Bellver M, Mighty J, Aparicio‐Domingo S, Li KV, Shi C, Zhou J, Cobb H, McGrath P, Michelis G, Lenhart P, Bilousova G, Heissel S, Rudy MJ, Coughlan C, Goodspeed AE, Becerra SP, Redenti S, Canto‐Soler MV. Extracellular vesicles released by human retinal pigment epithelium mediate increased polarised secretion of drusen proteins in response to AMD stressors. J Extracell Vesicles 2021; 10:e12165. [PMID: 34750957 PMCID: PMC8575963 DOI: 10.1002/jev2.12165] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 10/06/2021] [Accepted: 10/25/2021] [Indexed: 12/22/2022] Open
Abstract
Age-related macular degeneration (AMD) is a leading cause of blindness worldwide. Drusen are key contributors to the etiology of AMD and the ability to modulate drusen biogenesis could lead to therapeutic strategies to slow or halt AMD progression. The mechanisms underlying drusen biogenesis, however, remain mostly unknown. Here we demonstrate that under homeostatic conditions extracellular vesicles (EVs) secreted by retinal pigment epithelium (RPE) cells are enriched in proteins associated with mechanisms involved in AMD pathophysiology, including oxidative stress, immune response, inflammation, complement system and drusen composition. Furthermore, we provide first evidence that drusen-associated proteins are released as cargo of extracellular vesicles secreted by RPE cells in a polarised apical:basal mode. Notably, drusen-associated proteins exhibited distinctive directional secretion modes in homeostatic conditions and, differential modulation of this directional secretion in response to AMD stressors. These observations underpin the existence of a finely-tuned mechanism regulating directional apical:basal sorting and secretion of drusen-associated proteins via EVs, and its modulation in response to mechanisms involved in AMD pathophysiology. Collectively, our results strongly support an active role of RPE-derived EVs as a key source of drusen proteins and important contributors to drusen development and growth.
Collapse
Affiliation(s)
- Miguel Flores‐Bellver
- CellSight Ocular Stem Cell and Regeneration ProgramDepartment of OphthalmologySue Anschutz‐Rodgers Eye CenterUniversity of Colorado, School of MedicineAuroraColoradoUSA
| | - Jason Mighty
- Lehman CollegeBronxNew YorkUSA
- Biology Doctoral ProgramThe Graduate School and University CenterCity University of New YorkNew YorkNew YorkUSA
| | - Silvia Aparicio‐Domingo
- CellSight Ocular Stem Cell and Regeneration ProgramDepartment of OphthalmologySue Anschutz‐Rodgers Eye CenterUniversity of Colorado, School of MedicineAuroraColoradoUSA
| | - Kang V. Li
- CellSight Ocular Stem Cell and Regeneration ProgramDepartment of OphthalmologySue Anschutz‐Rodgers Eye CenterUniversity of Colorado, School of MedicineAuroraColoradoUSA
| | - Cui Shi
- Lehman CollegeBronxNew YorkUSA
- Biology Doctoral ProgramThe Graduate School and University CenterCity University of New YorkNew YorkNew YorkUSA
| | | | - Hannah Cobb
- CellSight Ocular Stem Cell and Regeneration ProgramDepartment of OphthalmologySue Anschutz‐Rodgers Eye CenterUniversity of Colorado, School of MedicineAuroraColoradoUSA
| | - Patrick McGrath
- Department of DermatologyUniversity of Colorado School of MedicineAuroraColoradoUSA
| | - German Michelis
- Section of Protein Structure and FunctionNEINIHBethesdaMarylandUSA
| | - Patricia Lenhart
- CellSight Ocular Stem Cell and Regeneration ProgramDepartment of OphthalmologySue Anschutz‐Rodgers Eye CenterUniversity of Colorado, School of MedicineAuroraColoradoUSA
| | - Ganna Bilousova
- Department of DermatologyUniversity of Colorado School of MedicineAuroraColoradoUSA
- Charles C. Gates Center for Regenerative MedicineUniversity of Colorado School of MedicineAuroraColoradoUSA
- Linda Crnic Institute for Down SyndromeUniversity of Colorado School of MedicineAuroraColoradoUSA
| | - Søren Heissel
- Proteomics Resource CenterThe Rockefeller UniversityNew YorkNew YorkUSA
| | - Michael J. Rudy
- Department of NeurologyUniversity of Colorado School of MedicineAuroraColoradoUSA
| | - Christina Coughlan
- University of Colorado Alzheimer's and Cognition CenterDepartment of NeurologyLinda Crnic Institute for Down SyndromeUniversity of Colorado Anschutz Medical CampusAuroraColoradoUSA
| | - Andrew E. Goodspeed
- Department of PharmacologyUniversity of Colorado Anschutz Medical CampusAuroraColoradoUSA
- University of Colorado Cancer CenterUniversity of Colorado Anschutz Medical CampusAuroraColoradoUSA
| | | | - Stephen Redenti
- Lehman CollegeBronxNew YorkUSA
- Biology Doctoral ProgramThe Graduate School and University CenterCity University of New YorkNew YorkNew YorkUSA
- Biochemistry Doctoral ProgramThe Graduate SchoolCity University of New YorkNew YorkNew YorkUSA
| | - M. Valeria Canto‐Soler
- CellSight Ocular Stem Cell and Regeneration ProgramDepartment of OphthalmologySue Anschutz‐Rodgers Eye CenterUniversity of Colorado, School of MedicineAuroraColoradoUSA
- Charles C. Gates Center for Regenerative MedicineUniversity of Colorado School of MedicineAuroraColoradoUSA
| |
Collapse
|
13
|
Liu C, Yang C, Wang M, Jiang S, Yi Q, Wang W, Wang L, Song L. A CD63 Homolog Specially Recruited to the Fungi-Contained Phagosomes Is Involved in the Cellular Immune Response of Oyster Crassostrea gigas. Front Immunol 2020; 11:1379. [PMID: 32793193 PMCID: PMC7387653 DOI: 10.3389/fimmu.2020.01379] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Accepted: 05/29/2020] [Indexed: 02/02/2023] Open
Abstract
Cluster of differentiation 63 (CD63), a four-transmembrane glycoprotein in the subfamily of tetraspanin, has been widely recognized as a gateway from the infection of foreign invaders to the immune defense of hosts. Its role in Pacific oyster Crassostrea gigas is, however, yet to be discovered. This work makes contributions by identifying CgCD63H, a CD63 homolog with four transmembrane domains and one conservative CCG motif, and establishing its role as a receptor that participates in immune recognition and hemocyte phagocytosis. The presence of CgCD63H messenger RNA (mRNA) in hepatopancreas, labial palps, gill, and hemocytes is confirmed. The expression level of mRNA in hemocytes is found significantly (p < 0.01) upregulated after the injection of Vibrio splendidus. CgCD63H protein, typically distributed over the plasma membrane of oyster hemocytes, is recruited to the Yarrowia lipolytica-containing phagosomes after the stimulation of Y. lipolytica. The recombinant CgCD63H protein expresses binding capacity to glucan (GLU), peptidoglycan (PGN), and lipopolysaccharide (LPS) in the presence of lyophilized hemolymph. The phagocytic rate of hemocytes toward V. splendidus and Y. lipolytica is significantly inhibited (p < 0.01) after incubation with anti-CgCD63H antibody. Our work further suggests that CgCD63H functions as a receptor involved in the immune recognition and hemocyte phagocytosis against invading pathogen, which can be a marker candidate for the hemocyte typing in C. gigas.
Collapse
Affiliation(s)
- Conghui Liu
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, China.,Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China.,Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, China
| | - Chuanyan Yang
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, China.,Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, China.,Dalian Key Laboratory of Aquatic Animal Disease Prevention and Control, Dalian Ocean University, Dalian, China
| | - Mengqiang Wang
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
| | - Shuai Jiang
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
| | - Qilin Yi
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, China.,Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, China.,Dalian Key Laboratory of Aquatic Animal Disease Prevention and Control, Dalian Ocean University, Dalian, China
| | - Weilin Wang
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China.,Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, China.,Dalian Key Laboratory of Aquatic Animal Disease Prevention and Control, Dalian Ocean University, Dalian, China
| | - Lingling Wang
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, China.,Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, China.,Dalian Key Laboratory of Aquatic Animal Disease Prevention and Control, Dalian Ocean University, Dalian, China
| | - Linsheng Song
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, China.,Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, China
| |
Collapse
|
14
|
Oguri Y, Shinoda K, Kim H, Alba DL, Bolus WR, Wang Q, Brown Z, Pradhan RN, Tajima K, Yoneshiro T, Ikeda K, Chen Y, Cheang RT, Tsujino K, Kim CR, Greiner VJ, Datta R, Yang CD, Atabai K, McManus MT, Koliwad SK, Spiegelman BM, Kajimura S. CD81 Controls Beige Fat Progenitor Cell Growth and Energy Balance via FAK Signaling. Cell 2020; 182:563-577.e20. [PMID: 32615086 DOI: 10.1016/j.cell.2020.06.021] [Citation(s) in RCA: 184] [Impact Index Per Article: 36.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 03/30/2020] [Accepted: 06/09/2020] [Indexed: 01/03/2023]
Abstract
Adipose tissues dynamically remodel their cellular composition in response to external cues by stimulating beige adipocyte biogenesis; however, the developmental origin and pathways regulating this process remain insufficiently understood owing to adipose tissue heterogeneity. Here, we employed single-cell RNA-seq and identified a unique subset of adipocyte progenitor cells (APCs) that possessed the cell-intrinsic plasticity to give rise to beige fat. This beige APC population is proliferative and marked by cell-surface proteins, including PDGFRα, Sca1, and CD81. Notably, CD81 is not only a beige APC marker but also required for de novo beige fat biogenesis following cold exposure. CD81 forms a complex with αV/β1 and αV/β5 integrins and mediates the activation of integrin-FAK signaling in response to irisin. Importantly, CD81 loss causes diet-induced obesity, insulin resistance, and adipose tissue inflammation. These results suggest that CD81 functions as a key sensor of external inputs and controls beige APC proliferation and whole-body energy homeostasis.
Collapse
Affiliation(s)
- Yasuo Oguri
- UCSF Diabetes Center, University of California, San Francisco, San Francisco, CA, USA; Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, USA; Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA, USA; Beth Israel Deaconess Medical Center, Division of Endocrinology, Diabetes & Metabolism, Harvard Medical School, Boston, MA, USA
| | - Kosaku Shinoda
- UCSF Diabetes Center, University of California, San Francisco, San Francisco, CA, USA; Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, USA; Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA, USA; Department of Medicine and Molecular Pharmacology, Albert Einstein College of Medicine, New York, NY, USA
| | - Hyeonwoo Kim
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Diana L Alba
- UCSF Diabetes Center, University of California, San Francisco, San Francisco, CA, USA; Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - W Reid Bolus
- UCSF Diabetes Center, University of California, San Francisco, San Francisco, CA, USA; Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Qiang Wang
- UCSF Diabetes Center, University of California, San Francisco, San Francisco, CA, USA; Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, USA; Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA, USA; Beth Israel Deaconess Medical Center, Division of Endocrinology, Diabetes & Metabolism, Harvard Medical School, Boston, MA, USA
| | - Zachary Brown
- UCSF Diabetes Center, University of California, San Francisco, San Francisco, CA, USA; Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, USA; Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA, USA
| | - Rachana N Pradhan
- UCSF Diabetes Center, University of California, San Francisco, San Francisco, CA, USA; Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, USA; Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA, USA
| | - Kazuki Tajima
- UCSF Diabetes Center, University of California, San Francisco, San Francisco, CA, USA; Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, USA; Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA, USA
| | - Takeshi Yoneshiro
- UCSF Diabetes Center, University of California, San Francisco, San Francisco, CA, USA; Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, USA; Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA, USA
| | - Kenji Ikeda
- UCSF Diabetes Center, University of California, San Francisco, San Francisco, CA, USA; Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, USA; Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA, USA; Department of Molecular Endocrinology and Metabolism, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yong Chen
- UCSF Diabetes Center, University of California, San Francisco, San Francisco, CA, USA; Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, USA; Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA, USA; Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Rachel T Cheang
- UCSF Diabetes Center, University of California, San Francisco, San Francisco, CA, USA; Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Kazuyuki Tsujino
- Department of Respiratory Medicine, National Hospital Organization Osaka Toneyama Medical Center, Osaka, Japan
| | - Caroline R Kim
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Vanille Juliette Greiner
- UCSF Diabetes Center, University of California, San Francisco, San Francisco, CA, USA; Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA, USA
| | - Ritwik Datta
- Department of Medicine, Lung Biology Center, Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, USA
| | - Christopher D Yang
- Department of Medicine, Lung Biology Center, Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, USA
| | - Kamran Atabai
- Department of Medicine, Lung Biology Center, Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, USA
| | - Michael T McManus
- UCSF Diabetes Center, University of California, San Francisco, San Francisco, CA, USA; Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA, USA
| | - Suneil K Koliwad
- UCSF Diabetes Center, University of California, San Francisco, San Francisco, CA, USA; Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | | | - Shingo Kajimura
- UCSF Diabetes Center, University of California, San Francisco, San Francisco, CA, USA; Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, USA; Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA, USA; Beth Israel Deaconess Medical Center, Division of Endocrinology, Diabetes & Metabolism, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
15
|
Iwagawa T, Aihara Y, Umutoni D, Baba Y, Murakami A, Miyado K, Watanabe S. Cd9 Protects Photoreceptors from Injury and Potentiates Edn2 Expression. Invest Ophthalmol Vis Sci 2020; 61:7. [PMID: 32150249 PMCID: PMC7401443 DOI: 10.1167/iovs.61.3.7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Purpose Cd9 is a tetraspanin membrane protein that plays various roles in tissue development and disease pathogenesis, especially in cancer, but the expression patterns and function of Cd9 in retinal development and disease are not well understood. We asked its roles during retinal photoreceptor degeneration by using CD9-knockout mice. Methods Cd9 knockout mice and rd1 mice were used to examine roles of Cd9 for progression of photoreceptor degeneration. Reverse transcription-polymerase chain reaction and immunohistochemistry were mainly used as analytical methods. Results Cd9 transcripts were only weakly expressed in retina at embryonic day 14, but its expression level subsequently increased and peaked at around postnatal day 12. In 6-week-old female mice derived retina, mRNA expression decreased slightly but was maintained at a significant level. Published RNA-sequencing data and immunohistochemistry indicated that Cd9 was expressed abundantly in Müller glia and weakly in other retinal neurons. Notably, when photoreceptors were damaged, Cd9 expression was increased in rod photoreceptors and decreased in Müller glia. Cd9 knockout mice retinas developed normally; however, once the retina suffered damage, degeneration of photoreceptors was more severe in Cd9 knockout retinas than control retinas. Induction of Edn2, which is known to protect against photoreceptor damage, was severely hampered. In addition, induction of Socs3, which is downstream of gp130 (Il6st), was weaker in Cd9 knockout retinas. Conclusions Taken together, these findings indicate that, although Cd9 was dispensable for normal gross morphological development, it protected rod photoreceptors and enhanced Edn2 expression, possibly through modulation of gp130 signaling.
Collapse
|
16
|
Lakkaraju A, Umapathy A, Tan LX, Daniele L, Philp NJ, Boesze-Battaglia K, Williams DS. The cell biology of the retinal pigment epithelium. Prog Retin Eye Res 2020; 78:100846. [PMID: 32105772 PMCID: PMC8941496 DOI: 10.1016/j.preteyeres.2020.100846] [Citation(s) in RCA: 245] [Impact Index Per Article: 49.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 02/19/2020] [Accepted: 02/23/2020] [Indexed: 02/07/2023]
Abstract
The retinal pigment epithelium (RPE), a monolayer of post-mitotic polarized epithelial cells, strategically situated between the photoreceptors and the choroid, is the primary caretaker of photoreceptor health and function. Dysfunction of the RPE underlies many inherited and acquired diseases that cause permanent blindness. Decades of research have yielded valuable insight into the cell biology of the RPE. In recent years, new technologies such as live-cell imaging have resulted in major advancement in our understanding of areas such as the daily phagocytosis and clearance of photoreceptor outer segment tips, autophagy, endolysosome function, and the metabolic interplay between the RPE and photoreceptors. In this review, we aim to integrate these studies with an emphasis on appropriate models and techniques to investigate RPE cell biology and metabolism, and discuss how RPE cell biology informs our understanding of retinal disease.
Collapse
Affiliation(s)
- Aparna Lakkaraju
- Department of Ophthalmology, University of California, San Francisco, San Francisco, CA, USA
| | - Ankita Umapathy
- Department of Ophthalmology and Stein Eye Institute, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA; Department of Neurobiology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Li Xuan Tan
- Department of Ophthalmology, University of California, San Francisco, San Francisco, CA, USA
| | - Lauren Daniele
- Department of Biochemistry, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Nancy J Philp
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Kathleen Boesze-Battaglia
- Department of Biochemistry, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - David S Williams
- Department of Ophthalmology and Stein Eye Institute, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA; Department of Neurobiology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA.
| |
Collapse
|
17
|
Sørensen NB. Subretinal surgery: functional and histological consequences of entry into the subretinal space. Acta Ophthalmol 2019; 97 Suppl A114:1-23. [PMID: 31709751 DOI: 10.1111/aos.14249] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND AND OBJECTIVES Gene-therapy, stem-cell transplantation and surgical robots hold the potential for treatment of currently untreatable retinal degenerative diseases. All of the techniques require entry into the subretinal space, which is a potential space located between the retina and the retinal pigment epithelium (RPE). Knowledge about obstacles and critical steps in relation to subretinal procedures is therefore needed. This thesis explores the functional and histological consequences of separation of the retina from the RPE, extensive RPE damage, a large cut in the retina (retinotomy) and RPE phagocytosis in a porcine model. METHODS Experiments were performed in 106 female domestic pigs of Danish landrace distributed over five studies. Under general anesthesia, different procedures for expansion of the subretinal space were conducted. Outcomes were visual function measured electrophysiologically with multifocal electroretinogram (mfERG) and retinal morphology examined histologically. Study I: The effect of anesthesia on mfERG was examined by repeated recordings for 3 hr in isoflurane or propofol anesthesia. Outcome was mfERG amplitude. Study II: Consequences of a large separation of the photoreceptors from the RPE were examined by injecting a perfluorocarbon-liquid (decalin) into the subretinal space. Two weeks after, in a second surgery, decalin was withdrawn. Outcomes were mfERG and histology 4 weeks after decalin injection. Study III: Extensive RPE damage was examined by expanding the subretinal space with saline and removing large sheets of RPE-cells through a retinotomy. Outcomes were mfERG and histology 2, 4 and 6 weeks after the procedure. Study IV: Consequences of a large retinotomy were examined by similar procedures as in Study III, but in study IV only a few RPE cells were removed. Outcomes were mfERG and histology 2 and 6 weeks after surgery. Study V: Clearance of the subretinal space was examined by injecting fluorescent latex beads of various sizes into the subretinal space. Outcome was histologic location of the beads at different time intervals after the procedure. RESULTS Study I: MfERG amplitudes decreased linearly as a function of time in propofol or isoflurane anesthesia. Duration of mfERG recording could be decreased without compromising quality, and thereby could time in anesthesia be reduced. Study II: MfERG and histology remained normal after reattachment of a large and 2-week long separation of the photoreceptors and RPE. Repeated entry into the subretinal space was well tolerated. Fluid injection into the subretinal space constitutes a risk of RPE-damage. Study III: Removal of large sheets of retinal pigment epithelial cells triggered a widespread rhegmatogenous-like retinal detachment resulting in visual loss. Study IV: A large retinotomy with limited damage of the RPE was well tolerated, and visual function was preserved. Study V: Subretinal latex beads up to 4 μm were phagocytosed by the RPE and passed into the sub-RPE space. Beads up to 2 μm travelled further through the Bruch's membrane and were found in the choroid, sclera and inside blood vessels. CONCLUSION A large expansion of the subretinal space, repeated entry, a large retinotomy and limited RPE damage is well tolerated and retinal function is preserved. Subretinal injection of fluid can damage the RPE and extensive RPE damage can induce a rhegmatogenous-like retinal detachment with loss of visual function. Foreign substances exit the subretinal space and can reach the systemic circulation.
Collapse
Affiliation(s)
- Nina Buus Sørensen
- Department of Ophthalmology Copenhagen University Hospital Rigshospitalet København Denmark
- Department of Neurology Zealand University Hospital Køge Denmark
| |
Collapse
|
18
|
Yu C, Muñoz LE, Mallavarapu M, Herrmann M, Finnemann SC. Annexin A5 regulates surface αvβ5 integrin for retinal clearance phagocytosis. J Cell Sci 2019; 132:jcs.232439. [PMID: 31515275 DOI: 10.1242/jcs.232439] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 09/01/2019] [Indexed: 11/20/2022] Open
Abstract
Diurnal clearance phagocytosis by the retinal pigment epithelium (RPE) is a conserved efferocytosis process whose binding step is mediated by αvβ5 integrin receptors. Two related annexins, A5 (ANXA5) and A6 (ANXA6), share an αvβ5 integrin-binding motif. Here, we report that ANXA5, but not ANXA6, regulates the binding capacity for spent photoreceptor outer segment fragments or apoptotic cells by fibroblasts and RPE. Similar to αvβ5-deficient RPE, ANXA5-/- RPE in vivo lacks the diurnal burst of phagocytosis that follows photoreceptor shedding in wild-type retina. Increasing ANXA5 in cells lacking αvβ5 or increasing αvβ5 in cells lacking ANXA5 does not affect particle binding. Association of cytosolic ANXA5 and αvβ5 integrin in RPE in culture and in vivo further supports their functional interdependence. Silencing ANXA5 is sufficient to reduce levels of αvβ5 receptors at the apical phagocytic surface of RPE cells. The effect of ANXA5 on surface αvβ5 and on particle binding requires the C-terminal ANXA5 annexin repeat but not its unique N-terminus. These results identify a novel role for ANXA5 specifically in the recognition and binding step of clearance phagocytosis, which is essential to retinal physiology.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Chen Yu
- Department of Biological Sciences, Center for Cancer, Genetic Disease, and Gene Regulation, Fordham University, Bronx, NY 10458, USA
| | - Luis E Muñoz
- Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Department of Internal Medicine 3 - Rheumatology and Immunology, Universitätsklinikum Erlangen, 90154 Erlangen, Germany
| | - Mallika Mallavarapu
- Department of Biological Sciences, Center for Cancer, Genetic Disease, and Gene Regulation, Fordham University, Bronx, NY 10458, USA
| | - Martin Herrmann
- Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Department of Internal Medicine 3 - Rheumatology and Immunology, Universitätsklinikum Erlangen, 90154 Erlangen, Germany
| | - Silvia C Finnemann
- Department of Biological Sciences, Center for Cancer, Genetic Disease, and Gene Regulation, Fordham University, Bronx, NY 10458, USA
| |
Collapse
|
19
|
Reyes-Ruiz JM, Osuna-Ramos JF, De Jesús-González LA, Hurtado-Monzón AM, Farfan-Morales CN, Cervantes-Salazar M, Bolaños J, Cigarroa-Mayorga OE, Martín-Martínez ES, Medina F, Fragoso-Soriano RJ, Chávez-Munguía B, Salas-Benito JS, Del Angel RM. Isolation and characterization of exosomes released from mosquito cells infected with dengue virus. Virus Res 2019; 266:1-14. [PMID: 30930201 DOI: 10.1016/j.virusres.2019.03.015] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 03/13/2019] [Accepted: 03/20/2019] [Indexed: 12/21/2022]
Abstract
Exosomes are endocytic origin small-membrane vesicles secreted to the extracellular space by most cell types. Exosomes released from virus infected-cells can mediate the cell-to-cell communication to promote or modulate viral transmission. Dengue virus (DENV) is an arbovirus transmitted by Aedes mosquitoes bite to humans. Interestingly, the role of exosomes during the DENV infection in mammalian cells has already been described. However, little is known about exosomes derived from infected mosquito cells. Thus, the exosomes released from DENV-infected C6/36 cells were isolated, purified and analyzed using an antibody against the tetraspanin CD9 from human that showed cross-reactivity with the homologs to human CD9 found in Aedes albopictus (AalCD9). The exosomes from DENV infected cells were larger than the exosomes secreted from uninfected cells, contained virus-like particles, and they were able to infect naïve C6/36 cells, suggesting that exosomes are playing a role in virus dissemination.
Collapse
Affiliation(s)
- José Manuel Reyes-Ruiz
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (CINVESTAV-IPN), Mexico City, Mexico
| | - Juan Fidel Osuna-Ramos
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (CINVESTAV-IPN), Mexico City, Mexico
| | - Luis Adrián De Jesús-González
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (CINVESTAV-IPN), Mexico City, Mexico
| | - Arianna Mahely Hurtado-Monzón
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (CINVESTAV-IPN), Mexico City, Mexico
| | - Carlos Noe Farfan-Morales
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (CINVESTAV-IPN), Mexico City, Mexico
| | - Margot Cervantes-Salazar
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (CINVESTAV-IPN), Mexico City, Mexico
| | - Jeni Bolaños
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (CINVESTAV-IPN), Mexico City, Mexico
| | - Oscar E Cigarroa-Mayorga
- Departamento de Tecnologías Avanzadas, Unidad Profesional Interdisciplinaria en Ingeniería y Tecnologías Avanzadas del Instituto Politécnico Nacional, Mexico City, Mexico
| | - Eduardo San Martín-Martínez
- Centro de Investigación en Ciencias Aplicada y Tecnología Avanzada del Instituto Politécnico Nacional (CICATA-IPN), Mexico City, Mexico
| | - Fernando Medina
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (CINVESTAV-IPN), Mexico City, Mexico
| | | | - Bibiana Chávez-Munguía
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (CINVESTAV-IPN), Mexico City, Mexico
| | - Juan Santiago Salas-Benito
- Maestría en Ciencias en Biomedicina Molecular, Escuela Nacional de Medicina y Homeopatía, Instituto Politécnico Nacional, Mexico City, Mexico; Doctorado en Ciencias en Biotecnología, Escuela Nacional de Medicina y Homeopatía, Instituto Politécnico Nacional, Mexico City, Mexico
| | - Rosa M Del Angel
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (CINVESTAV-IPN), Mexico City, Mexico.
| |
Collapse
|
20
|
Sørensen NB, Christiansen AT, Kjær TW, Klemp K, la Cour M, Heegaard S, Kiilgaard JF. Bruch's membrane allows unhindered passage of up to 2 μm latex beads in an in vivo porcine model. Exp Eye Res 2018; 180:1-7. [PMID: 30468719 DOI: 10.1016/j.exer.2018.11.019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Revised: 11/06/2018] [Accepted: 11/19/2018] [Indexed: 11/26/2022]
Abstract
PURPOSE It has been proposed that changes in the permeability of Bruch's membrane play a role in the pathogenesis of age-related macular degeneration (AMD). This paper investigates, in an in vivo porcine model, the migration of fluorescent latex beads across the Bruch's membrane after subretinal injection. METHODS Forty-one healthy eyes of 33 three-month-old domestic pigs received a subretinal injection of 0.5, 1.0, 2.0, or 4.0 μm fluorescent latex beads. Between three hours and five weeks after injection evaluations were performed with fundus photographs and histology. Fluorescent beads were identified in unstained histologic sections using the rhodamine filter with the light microscope. RESULTS The fluorescent latex beads relocated from the subretinal space. Intact beads up to 2.0 μm were found in the choroid, sclera, and extrascleral space. The smaller beads were also found inside choroidal and extrascleral blood vessels. In contrast, the larger beads of 4.0 μm did not pass the Bruch's membrane. CONCLUSION Subretinally implanted beads up to 2.0 μm pass the Bruch's membrane intact and cross the blood-ocular barrier. The intact beads are found in the choroid, sclera and inside blood vessels. The results give reason to consider the role of subretinal clearance and passage of Bruch's membrane in the development of AMD.
Collapse
Affiliation(s)
- Nina Buus Sørensen
- Department of Ophthalmology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark.
| | | | | | - Kristian Klemp
- Department of Ophthalmology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Morten la Cour
- Department of Ophthalmology, Copenhagen University Hospital, Rigshospitalet, Glostrup, Denmark
| | - Steffen Heegaard
- Department of Ophthalmology, Copenhagen University Hospital, Rigshospitalet, Glostrup, Denmark; Department of Pathology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Jens Folke Kiilgaard
- Department of Ophthalmology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
21
|
Kumawat AK, Yu C, Mann EA, Schridde A, Finnemann SC, Mowat AM. Expression and characterization of αvβ5 integrin on intestinal macrophages. Eur J Immunol 2018; 48:1181-1187. [PMID: 29676784 DOI: 10.1002/eji.201747318] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 02/23/2018] [Accepted: 04/17/2018] [Indexed: 12/27/2022]
Abstract
Macrophages play a crucial role in maintaining homeostasis in the intestine, but the underlying mechanisms have not yet been elucidated fully. Here, we show for the first time that mature intestinal macrophages in mouse intestine express high levels of αvβ5 integrin, which acts as a receptor for the uptake of apoptotic cells and can activate molecules involved in several aspects of tissue homeostasis such as angiogenesis and remodeling of the ECM. αvβ5 is not expressed by other immune cells in the intestine, is already present on intestinal macrophages soon after birth, and its expression is not dependent on the microbiota. In adults, αvβ5 is induced during the differentiation of monocytes in response to the local environment and it confers intestinal macrophages with the ability to promote engulfment of apoptotic cells via engagement of the bridging molecule milk fat globule EGF-like molecule 8. In the absence of αvβ5, there are fewer monocytes in the mucosa and mature intestinal macrophages have decreased expression of metalloproteases and IL 10. Mice lacking αvβ5 on haematopoietic cells show increased susceptibility to chemical colitis and we conclude that αvβ5 contributes to the tissue repair by regulating the homeostatic properties of intestinal macrophages.
Collapse
Affiliation(s)
- Ashok K Kumawat
- Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, Scotland.,School of Medical Sciences, Örebro University, Örebro, Sweden
| | - Chen Yu
- Department of Biological Sciences, Center for Cancer, Genetic Diseases and Gene Regulation, Fordham University, Bronx, NY, USA
| | - Elizabeth A Mann
- Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, Scotland
| | - Anika Schridde
- Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, Scotland
| | - Silvia C Finnemann
- Department of Biological Sciences, Center for Cancer, Genetic Diseases and Gene Regulation, Fordham University, Bronx, NY, USA
| | - Allan McI Mowat
- Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, Scotland
| |
Collapse
|
22
|
Ao J, Wood JP, Chidlow G, Gillies MC, Casson RJ. Retinal pigment epithelium in the pathogenesis of age-related macular degeneration and photobiomodulation as a potential therapy? Clin Exp Ophthalmol 2018; 46:670-686. [PMID: 29205705 DOI: 10.1111/ceo.13121] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Revised: 11/27/2017] [Accepted: 11/28/2017] [Indexed: 12/11/2022]
Abstract
The retinal pigment epithelium (RPE) comprises a monolayer of cells located between the neuroretina and the choriocapillaries. The RPE serves several important functions in the eye: formation of the blood-retinal barrier, protection of the retina from oxidative stress, nutrient delivery and waste disposal, ionic homeostasis, phagocytosis of photoreceptor outer segments, synthesis and release of growth factors, reisomerization of all-trans-retinal during the visual cycle, and establishment of ocular immune privilege. Age-related macular degeneration (AMD) is the leading cause of blindness in developed countries. Dysfunction of the RPE has been associated with the pathogenesis of AMD in relation to increased oxidative stress, mitochondrial destabilization and complement dysregulation. Photobiomodulation or near infrared light therapy which refers to non-invasive irradiation of tissue with light in the far-red to near-infrared light spectrum (630-1000 nm), is an intervention that specifically targets key mechanisms of RPE dysfunction that are implicated in AMD pathogenesis. The current evidence for the efficacy of photobiomodulation in AMD is poor but its safety profile and proposed mechanisms of action motivate further research as a novel therapy for AMD.
Collapse
Affiliation(s)
- Jack Ao
- South Australian Institute of Ophthalmology, University of Adelaide, Adelaide, South Australia, Australia
| | - John Pm Wood
- South Australian Institute of Ophthalmology, University of Adelaide, Adelaide, South Australia, Australia
| | - Glyn Chidlow
- South Australian Institute of Ophthalmology, University of Adelaide, Adelaide, South Australia, Australia
| | - Mark C Gillies
- The Save Sight Institute, Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Robert J Casson
- South Australian Institute of Ophthalmology, University of Adelaide, Adelaide, South Australia, Australia
| |
Collapse
|
23
|
Vance DT, Dufresne J, Florentinus-Mefailoski A, Tucholska M, Trimble W, Grinstein S, Marshall JG. A phagocytosis assay for oxidized low-density lipoprotein versus immunoglobulin G-coated microbeads in human U937 macrophages. Anal Biochem 2016; 500:24-34. [DOI: 10.1016/j.ab.2016.01.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Revised: 12/24/2015] [Accepted: 01/11/2016] [Indexed: 10/22/2022]
|
24
|
Sethna S, Chamakkala T, Gu X, Thompson TC, Cao G, Elliott MH, Finnemann SC. Regulation of Phagolysosomal Digestion by Caveolin-1 of the Retinal Pigment Epithelium Is Essential for Vision. J Biol Chem 2016; 291:6494-506. [PMID: 26814131 DOI: 10.1074/jbc.m115.687004] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Indexed: 01/09/2023] Open
Abstract
Caveolin-1 associates with the endo/lysosomal machinery of cells in culture, suggesting that it functions at these organelles independently of its contribution to cell surface caveolae. Here we explored mice lacking caveolin-1 specifically in the retinal pigment epithelium (RPE). The RPE supports neighboring photoreceptors via diurnal phagocytosis of spent photoreceptor outer segment fragments. Like mice lacking caveolin-1 globally, (RPE)CAV1(-/-) mice developed a normal RPE and neural retina but showed reduced rod photoreceptor light responses, indicating that lack of caveolin-1 affects photoreceptor function in a non-cell-autonomous manner. (RPE)CAV1(-/-) RPE in situ showed normal particle engulfment but delayed phagosome clearance and reversed diurnal profiles of levels and activities of lysosomal enzymes. Therefore, eliminating caveolin-1 specifically impairs phagolysosomal degradation by the RPE in vivo. Endogenous caveolin-1 was recruited to maturing phagolysosomes in RPE cells in culture. Consistent with these in vivo data, a moderate increase (to ∼ 2.5-fold) or decrease (by half) of caveolin-1 protein levels in RPE cells in culture was sufficient to accelerate or impair phagolysosomal digestion, respectively. A mutant form of caveolin-1 that fails to reach the cell surface augmented degradation like wild-type caveolin-1. Acidic lysosomal pH and increased protease activity are essential for digestion. We show that halving caveolin-1 protein levels significantly alkalinized lysosomal pH and decreased lysosomal enzyme activities. Taken together, our results reveal a novel role for intracellular caveolin-1 in modulating phagolysosomal function. Moreover, they show, for the first time, that organellar caveolin-1 significantly affects tissue functionality in vivo.
Collapse
Affiliation(s)
- Saumil Sethna
- From the Department of Biological Sciences, Center for Cancer Genetic Diseases and Gene Regulation, Fordham University, Bronx, New York 10458
| | - Tess Chamakkala
- From the Department of Biological Sciences, Center for Cancer Genetic Diseases and Gene Regulation, Fordham University, Bronx, New York 10458
| | - Xiaowu Gu
- the Department of Ophthalmology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, and
| | - Timothy C Thompson
- the Department of Genitourinary Medical Oncology, University of Texas M.D. Anderson Cancer Center, Houston, Texas 77030
| | - Guangwen Cao
- the Department of Genitourinary Medical Oncology, University of Texas M.D. Anderson Cancer Center, Houston, Texas 77030
| | - Michael H Elliott
- the Department of Ophthalmology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, and
| | - Silvia C Finnemann
- From the Department of Biological Sciences, Center for Cancer Genetic Diseases and Gene Regulation, Fordham University, Bronx, New York 10458,
| |
Collapse
|
25
|
Chen Y, Kennedy DJ, Ramakrishnan DP, Yang M, Huang W, Li Z, Xie Z, Chadwick AC, Sahoo D, Silverstein RL. Oxidized LDL-bound CD36 recruits an Na⁺/K⁺-ATPase-Lyn complex in macrophages that promotes atherosclerosis. Sci Signal 2015; 8:ra91. [PMID: 26350901 DOI: 10.1126/scisignal.aaa9623] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
One characteristic of atherosclerosis is the accumulation of lipid-laden macrophage foam cells in the arterial wall. We have previously shown that the binding of oxidized low-density lipoprotein (oxLDL) to the scavenger receptor CD36 activates the kinase Lyn, initiating a cascade that inhibits macrophage migration and is necessary for foam cell generation. We identified the plasma membrane ion transporter Na(+)/K(+)-ATPase as a key component in the macrophage oxLDL-CD36 signaling axis. Using peritoneal macrophages isolated from Atp1a1 heterozygous or Cd36-null mice, we demonstrated that CD36 recruited an Na(+)/K(+)-ATPase-Lyn complex for Lyn activation in response to oxLDL. Macrophages deficient in the α1 Na(+)/K(+)-ATPase catalytic subunit did not respond to activation of CD36, showing attenuated oxLDL uptake and foam cell formation, and oxLDL failed to inhibit migration of these macrophages. Furthermore, Apoe-null mice, which are a model of atherosclerosis, were protected from diet-induced atherosclerosis by global deletion of a single allele encoding the α1 Na(+)/K(+)-ATPase subunit or reconstitution with macrophages that lacked an allele encoding the α1 Na(+)/K(+)-ATPase subunit. These findings identify Na(+)/K(+)-ATPase as a potential target for preventing or treating atherosclerosis.
Collapse
Affiliation(s)
- Yiliang Chen
- Blood Research Institute, Blood Center of Wisconsin, Milwaukee, WI 53226, USA
| | - David J Kennedy
- Department of Medicine, University of Toledo, Health Science Campus, Toledo, OH 43614, USA
| | - Devi Prasadh Ramakrishnan
- Blood Research Institute, Blood Center of Wisconsin, Milwaukee, WI 53226, USA. Department of Molecular Medicine, Lerner College of Medicine, Case Western Reserve University, Cleveland, OH 44195, USA
| | - Moua Yang
- Blood Research Institute, Blood Center of Wisconsin, Milwaukee, WI 53226, USA. Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Wenxin Huang
- Blood Research Institute, Blood Center of Wisconsin, Milwaukee, WI 53226, USA
| | - Zhichuan Li
- Department of Physiology and Pharmacology, University of Toledo, Health Science Campus, Toledo, OH 43614, USA
| | - Zijian Xie
- Department of Physiology and Pharmacology, University of Toledo, Health Science Campus, Toledo, OH 43614, USA
| | - Alexandra C Chadwick
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Daisy Sahoo
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI 53226, USA. Department of Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA. Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Roy L Silverstein
- Blood Research Institute, Blood Center of Wisconsin, Milwaukee, WI 53226, USA. Department of Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA.
| |
Collapse
|
26
|
Hennig R, Goepferich A. Nanoparticles for the treatment of ocular neovascularizations. Eur J Pharm Biopharm 2015; 95:294-306. [DOI: 10.1016/j.ejpb.2015.02.027] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Revised: 02/13/2015] [Accepted: 02/27/2015] [Indexed: 12/27/2022]
|
27
|
Araínga M, Guo D, Wiederin J, Ciborowski P, McMillan J, Gendelman HE. Opposing regulation of endolysosomal pathways by long-acting nanoformulated antiretroviral therapy and HIV-1 in human macrophages. Retrovirology 2015; 12:5. [PMID: 25608975 PMCID: PMC4307176 DOI: 10.1186/s12977-014-0133-5] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Accepted: 12/19/2014] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND Long-acting nanoformulated antiretroviral therapy (nanoART) is designed to improve patient regimen adherence, reduce systemic drug toxicities, and facilitate clearance of human immunodeficiency virus type one (HIV-1) infection. While nanoART establishes drug depots within recycling and late monocyte-macrophage endosomes, whether or not this provides a strategic advantage towards viral elimination has not been elucidated. RESULTS We applied quantitative SWATH-MS proteomics and cell profiling to nanoparticle atazanavir (nanoATV)-treated and HIV-1 infected human monocyte-derived macrophages (MDM). Native ATV and uninfected cells served as controls. Both HIV-1 and nanoATV engaged endolysosomal trafficking for assembly and depot formation, respectively. Notably, the pathways were deregulated in opposing manners by the virus and the nanoATV, likely by viral clearance. Paired-sample z-scores, of the proteomic data sets, showed up- and down- regulation of Rab-linked endolysosomal proteins. NanoART and native ATV treated uninfected cells showed limited effects. The data was confirmed by Western blot. DAVID and KEGG bioinformatics analyses of proteomic data showed relationships between secretory, mobility and phagocytic cell functions and virus and particle trafficking. CONCLUSIONS We posit that modulation of endolysosomal pathways by antiretroviral nanoparticles provides a strategic path to combat HIV infection.
Collapse
Affiliation(s)
- Mariluz Araínga
- />Departments of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, 985880 Nebraska Medical Center, Omaha, NE 68198-5880 USA
| | - Dongwei Guo
- />Departments of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, 985880 Nebraska Medical Center, Omaha, NE 68198-5880 USA
- />Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198-5880 USA
| | - Jayme Wiederin
- />Departments of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, 985880 Nebraska Medical Center, Omaha, NE 68198-5880 USA
| | - Pawel Ciborowski
- />Departments of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, 985880 Nebraska Medical Center, Omaha, NE 68198-5880 USA
| | - JoEllyn McMillan
- />Departments of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, 985880 Nebraska Medical Center, Omaha, NE 68198-5880 USA
| | - Howard E Gendelman
- />Departments of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, 985880 Nebraska Medical Center, Omaha, NE 68198-5880 USA
- />Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198-5880 USA
| |
Collapse
|
28
|
In-depth characterisation of Retinal Pigment Epithelium (RPE) cells derived from human induced pluripotent stem cells (hiPSC). Neuromolecular Med 2014; 16:551-64. [PMID: 24801942 PMCID: PMC4119585 DOI: 10.1007/s12017-014-8308-8] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Accepted: 04/22/2014] [Indexed: 12/24/2022]
Abstract
Induced pluripotent stem cell (iPSC)-derived retinal pigment epithelium (RPE) has widely been appreciated as a promising tool to model human ocular disease emanating from primary RPE pathology. Here, we describe the successful reprogramming of adult human dermal fibroblasts to iPSCs and their differentiation to pure expandable RPE cells with structural and functional features characteristic for native RPE. Fibroblast cultures were established from skin biopsy material and subsequently reprogrammed following polycistronic lentiviral transduction with OCT4, SOX2, KLF4 and L-Myc. Fibroblast-derived iPSCs showed typical morphology, chromosomal integrity and a distinctive stem cell marker profile. Subsequent differentiation resulted in expandable pigmented hexagonal RPE cells. The cells revealed stable RNA expression of mature RPE markers RPE65, RLBP and BEST1. Immunolabelling verified localisation of BEST1 at the basolateral plasma membrane, and scanning electron microscopy showed typical microvilli at the apical side of iPSC-derived RPE cells. Transepithelial resistance was maintained at high levels during cell culture indicating functional formation of tight junctions. Secretion capacity was demonstrated for VEGF-A. Feeding of porcine photoreceptor outer segments revealed the proper ability of these cells for phagocytosis. IPSC-derived RPE cells largely maintained these properties after cryopreservation. Together, our study underlines that adult dermal fibroblasts can serve as a valuable resource for iPSC-derived RPE with characteristics highly reminiscent of true RPE cells. This will allow its broad application to establish cellular models for RPE-related human diseases.
Collapse
|
29
|
Mazzoni F, Safa H, Finnemann SC. Understanding photoreceptor outer segment phagocytosis: use and utility of RPE cells in culture. Exp Eye Res 2014; 126:51-60. [PMID: 24780752 DOI: 10.1016/j.exer.2014.01.010] [Citation(s) in RCA: 166] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2013] [Revised: 01/10/2014] [Accepted: 01/13/2014] [Indexed: 12/21/2022]
Abstract
RPE cells are the most actively phagocytic cells in the human body. In the eye, RPE cells face rod and cone photoreceptor outer segments at all times but contribute to shedding and clearance phagocytosis of distal outer segment tips only once a day. Analysis of RPE phagocytosis in situ has succeeded in identifying key players of the RPE phagocytic mechanism. Phagocytic processes comprise three distinct phases, recognition/binding, internalization, and digestion, each of which is regulated separately by phagocytes. Studies of phagocytosis by RPE cells in culture allow specifically analyzing and manipulating these distinct phases to identify their molecular mechanisms. Here, we compare similarities and differences of primary, immortalized, and stem cell-derived RPE cells in culture to RPE cells in situ with respect to phagocytic function. We discuss in particular potential pitfalls of RPE cell culture phagocytosis assays. Finally, we point out considerations for phagocytosis assay development for future studies.
Collapse
Affiliation(s)
- Francesca Mazzoni
- Department of Biological Sciences, Center for Cancer, Genetic Diseases, and Gene Regulation, Fordham University, Bronx, NY 10458, USA
| | - Hussein Safa
- Department of Biological Sciences, Center for Cancer, Genetic Diseases, and Gene Regulation, Fordham University, Bronx, NY 10458, USA
| | - Silvia C Finnemann
- Department of Biological Sciences, Center for Cancer, Genetic Diseases, and Gene Regulation, Fordham University, Bronx, NY 10458, USA.
| |
Collapse
|
30
|
Canton J, Neculai D, Grinstein S. Scavenger receptors in homeostasis and immunity. Nat Rev Immunol 2013; 13:621-34. [PMID: 23928573 DOI: 10.1038/nri3515] [Citation(s) in RCA: 593] [Impact Index Per Article: 49.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Scavenger receptors were originally identified by their ability to recognize and to remove modified lipoproteins; however, it is now appreciated that they carry out a striking range of functions, including pathogen clearance, lipid transport, the transport of cargo within the cell and even functioning as taste receptors. The large repertoire of ligands recognized by scavenger receptors and their broad range of functions are not only due to the wide range of receptors that constitute this family but also to their ability to partner with various co-receptors. The ability of individual scavenger receptors to associate with different co-receptors makes their responsiveness extremely versatile. This Review highlights recent insights into the structural features that determine the function of scavenger receptors and the emerging role that these receptors have in immune responses, notably in macrophage polarization and in the pathogenesis of diseases such as atherosclerosis and Alzheimer's disease.
Collapse
Affiliation(s)
- Johnathan Canton
- Cell Biology Program, Hospital for Sick Children, Toronto, Ontario M5G 1X8, Canada
| | | | | |
Collapse
|
31
|
Gustafson-Wagner E, Stipp CS. The CD9/CD81 tetraspanin complex and tetraspanin CD151 regulate α3β1 integrin-dependent tumor cell behaviors by overlapping but distinct mechanisms. PLoS One 2013; 8:e61834. [PMID: 23613949 PMCID: PMC3629153 DOI: 10.1371/journal.pone.0061834] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2013] [Accepted: 03/15/2013] [Indexed: 01/16/2023] Open
Abstract
Integrin α3β1 potently promotes cell motility on its ligands, laminin-332 and laminin-511, and this may help to explain why α3β1 has repeatedly been linked to breast carcinoma progression and metastasis. The pro-migratory functions of α3β1 depend strongly on lateral interactions with cell surface tetraspanin proteins. Tetraspanin CD151 interacts directly with the α3 integrin subunit and links α3β1 integrin to other tetraspanins, including CD9 and CD81. Loss of CD151 disrupts α3β1 association with other tetraspanins and impairs α3β1-dependent motility. However, the extent to which tetraspanins other than CD151 are required for specific α3β1 functions is unclear. To begin to clarify which aspects of α3β1 function require which tetraspanins, we created breast carcinoma cells depleted of both CD9 and CD81 by RNA interference. Silencing both of these closely related tetraspanins was required to uncover their contributions to α3β1 function. We then directly compared our CD9/CD81-silenced cells to CD151-silenced cells. Both CD9/CD81-silenced cells and CD151-silenced cells showed delayed α3β1-dependent cell spreading on laminin-332. Surprisingly, however, once fully spread, CD9/CD81-silenced cells, but not CD151-silenced cells, displayed impaired α3β1-dependent directed motility and altered front-rear cell morphology. Also unexpectedly, the CD9/CD81 complex, but not CD151, was required to promote α3β1 association with PKCα in breast carcinoma cells, and a PKC inhibitor mimicked aspects of the CD9/CD81-silenced cell motility defect. Our data reveal overlapping, but surprisingly distinct contributions of specific tetraspanins to α3β1 integrin function. Importantly, some of CD9/CD81's α3β1 regulatory functions may not require CD9/CD81 to be physically linked to α3β1 by CD151.
Collapse
Affiliation(s)
| | - Christopher S. Stipp
- Department of Biology, University of Iowa, Iowa City, Iowa, United States of America
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, Iowa, United States of America
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa, United States of America
| |
Collapse
|
32
|
Morales SA, Telander DG, Leon D, Forward K, Braun J, Wadehra M, Gordon LK. Epithelial membrane protein 2 controls VEGF expression in ARPE-19 cells. Invest Ophthalmol Vis Sci 2013; 54:2367-72. [PMID: 23439602 DOI: 10.1167/iovs.12-11013] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
PURPOSE VEGF production by RPE cells has been shown to be important in regulating aberrant angiogenesis in the retina, which is responsible for multiple types of ocular pathology. EMP2 is highly expressed in the RPE and has been shown to regulate FAK activation, which is implicated in VEGF expression in other cell lines. The purpose of this study was to determine whether EMP2 regulates VEGF expression in the RPE cell line, ARPE-19. METHODS ARPE-19 cells were engineered to overexpress EMP2. EMP2 siRNA was used to decrease EMP2 expression. The small molecule inhibitor PP2 was used to inhibit FAK activation. VEGF levels were measured by Western blot and ELISA. Functional differences in secreted VEGF were assayed using HUVEC migration. RESULTS VEGF expression levels correlated with levels of EMP2. An increase of VEGF by 150% was observed in EMP2 overexpressing cells as compared with ARPE-19 cells. Concordantly, EMP2 knockdown resulted in a 57% decrease in VEGF expression. HUVEC migration (P = 0.01) and vessel tube formation (P < 0.01) were significantly increased when exposed to cell culture supernatants from EMP2 overexpressing cells. CONCLUSIONS This study establishes a novel connection between EMP2 and VEGF and may reflect either a direct effect through the tetraspan web or an indirect change through FAK activation. This connection is functionally significant. In addition to the direct use of anti-VEGF antibodies, modulation of EMP2 with impact on VEGF is potentially a distinct therapeutic target for the treatment of neovascularization associated with retinal diseases that involve pathologic angiogenesis.
Collapse
Affiliation(s)
- Shawn A Morales
- Department of Ophthalmology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, California 90095, USA
| | | | | | | | | | | | | |
Collapse
|
33
|
Harris HJ, Clerte C, Farquhar MJ, Goodall M, Hu K, Rassam P, Dosset P, Wilson GK, Balfe P, IJzendoorn SC, Milhiet PE, McKeating JA. Hepatoma polarization limits CD81 and hepatitis C virus dynamics. Cell Microbiol 2013; 15:430-45. [PMID: 23126643 PMCID: PMC3599488 DOI: 10.1111/cmi.12047] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2012] [Revised: 09/08/2012] [Accepted: 09/27/2012] [Indexed: 12/12/2022]
Abstract
Many viruses target the polarized epithelial apex during host invasion. In contrast, hepatitis C virus (HCV) engages receptors at the basal surface of hepatocytes in the polarized liver parenchyma. Hepatocyte polarization limits HCV entry by undefined mechanism(s). Given the recent reports highlighting a role for receptor mobility in pathogen entry, we studied the effect(s) of hepatocyte polarization on viral receptor and HCV pseudoparticle (HCVpp) dynamics using real-time fluorescence recovery after photobleaching and single particle tracking. Hepatoma polarization reduced CD81 and HCVpp dynamics at the basal membrane. Since cell polarization is accompanied by changes in the actin cytoskeleton and CD81 links to actin via its C-terminus, we studied the dynamics of a mutant CD81 lacking a C-terminal tail (CD81(ΔC)) and its effect(s) on HCVpp mobility and infection. CD81(ΔC) showed an increased frequency of confined trajectories and a reduction of Brownian diffusing molecules compared to wild-type protein in non-polarized cells. However, these changes were notobserved in polarized cells. HCVpp showed a significant reduction in Brownian diffusion and infection of CD81(ΔC) expressing non-polarized cells. In summary, these data highlight the dynamic nature of CD81 and demonstrate a role for CD81 lateral diffusion to regulate HCV infection in a polarization-dependent manner.
Collapse
Affiliation(s)
- H J Harris
- School of Immunity and Infection, University of BirminghamBirmingham, UK
- NIHR Centre for Liver Disease, University of BirminghamBirmingham, UK
| | - C Clerte
- Unité 1054, InsermMontpellier, France
- Centre de Biochimie Structurale, Université de Montpellier, CNRS, UMR 5048Montpellier, France
| | - M J Farquhar
- School of Immunity and Infection, University of BirminghamBirmingham, UK
- NIHR Centre for Liver Disease, University of BirminghamBirmingham, UK
| | - M Goodall
- School of Immunity and Infection, University of BirminghamBirmingham, UK
- NIHR Centre for Liver Disease, University of BirminghamBirmingham, UK
| | - K Hu
- School of Immunity and Infection, University of BirminghamBirmingham, UK
- NIHR Centre for Liver Disease, University of BirminghamBirmingham, UK
| | - P Rassam
- Unité 1054, InsermMontpellier, France
- Centre de Biochimie Structurale, Université de Montpellier, CNRS, UMR 5048Montpellier, France
| | - P Dosset
- Unité 1054, InsermMontpellier, France
- Centre de Biochimie Structurale, Université de Montpellier, CNRS, UMR 5048Montpellier, France
| | - G K Wilson
- School of Immunity and Infection, University of BirminghamBirmingham, UK
- NIHR Centre for Liver Disease, University of BirminghamBirmingham, UK
| | - P Balfe
- School of Immunity and Infection, University of BirminghamBirmingham, UK
- NIHR Centre for Liver Disease, University of BirminghamBirmingham, UK
| | - S C IJzendoorn
- Department of Cell Biology, University Medical Center Groningen, University of GroningenGroningen, The Netherlands
| | - P E Milhiet
- Unité 1054, InsermMontpellier, France
- Centre de Biochimie Structurale, Université de Montpellier, CNRS, UMR 5048Montpellier, France
| | - J A McKeating
- School of Immunity and Infection, University of BirminghamBirmingham, UK
- NIHR Centre for Liver Disease, University of BirminghamBirmingham, UK
- NIHR Liver Biomedical Research Unit, University of BirminghamBirmingham, UK
| |
Collapse
|
34
|
Multimolecular signaling complexes enable Syk-mediated signaling of CD36 internalization. Dev Cell 2013; 24:372-83. [PMID: 23395392 DOI: 10.1016/j.devcel.2013.01.007] [Citation(s) in RCA: 89] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2011] [Revised: 12/20/2012] [Accepted: 01/10/2013] [Indexed: 12/16/2022]
Abstract
CD36 is a versatile receptor known to play a central role in the development of atherosclerosis, the pathogenesis of malaria, and the removal of apoptotic cells. Remarkably, the short cytosolically exposed regions of CD36 lack identifiable motifs, which has hampered elucidation of its mode of signaling. Using a combination of phosphoprotein isolation, mass spectrometry, superresolution imaging, and gene silencing, we have determined that the receptor induces ligand internalization through a heteromeric complex consisting of CD36, β1 and/or β2 integrins, and the tetraspanins CD9 and/or CD81. This receptor complex serves to link CD36 to the adaptor FcRγ, which bears an immunoreceptor tyrosine activation motif. By coupling to FcRγ, CD36 is able to engage Src-family kinases and Syk, which in turn drives the internalization of CD36 and its bound ligands.
Collapse
|
35
|
Integrins and small GTPases as modulators of phagocytosis. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2013; 302:321-54. [PMID: 23351714 DOI: 10.1016/b978-0-12-407699-0.00006-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Phagocytosis is the mechanism whereby cells engulf large particles. This process has long been recognized as a critical component of the innate immune response, which constitutes the organism's defense against microorganisms. In addition, phagocytic internalization of apoptotic cells or cell fragments plays important roles in tissue homeostasis and remodeling. Phagocytosis requires target interactions with receptors on the plasma membrane of the phagocytic cell. Integrins have been identified as important mediators of particle clearance, in addition to their well-established roles in cell adhesion, migration and mechanotransduction. Indeed, these ubiquitously expressed proteins impart phagocytic capacity to epithelial, endothelial and mesenchymal cell types. The importance of integrins in particle internalization is emphasized by the ability of microbial and viral pathogens to exploit their signaling pathways to invade host cells, and by the wide variety of disorders that arise from abnormalities in integrin-dependent phagocytic uptake.
Collapse
|
36
|
Gropp M, Shilo V, Vainer G, Gov M, Gil Y, Khaner H, Matzrafi L, Idelson M, Kopolovic J, Zak NB, Reubinoff BE. Standardization of the teratoma assay for analysis of pluripotency of human ES cells and biosafety of their differentiated progeny. PLoS One 2012; 7:e45532. [PMID: 23049812 PMCID: PMC3458078 DOI: 10.1371/journal.pone.0045532] [Citation(s) in RCA: 86] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2012] [Accepted: 08/20/2012] [Indexed: 11/19/2022] Open
Abstract
Teratoma tumor formation is an essential criterion in determining the pluripotency of human pluripotent stem cells. However, currently there is no consistent protocol for assessment of teratoma forming ability. Here we present detailed characterization of a teratoma assay that is based on subcutaneous co-transplantation of defined numbers of undifferentiated human embryonic stem cells (hESCs) with mitotically inactivated feeder cells and Matrigel into immunodeficient mice. The assay was highly reproducible and 100% efficient when 100,000 hESCs were transplanted. It was sensitive, promoting teratoma formation after transplantation of 100 hESCs, though larger numbers of animals and longer follow-up were required. The assay could detect residual teratoma forming cells within differentiated hESC populations however its sensitivity was decreased in the presence of differentiated cells. Our data lay the foundation, for standardization of a teratoma assay for pluripotency analysis. The assay can also be used for bio-safety analysis of pluripotent stem cell-derived differentiated progeny.
Collapse
Affiliation(s)
- Michal Gropp
- The Hadassah Human Embryonic Stem Cell Research Center, The Goldyne Savad Institute of Gene Therapy and Department of Gynecology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | | | - Gilad Vainer
- The Department of Pathology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Miri Gov
- CellCure NeuroSciences Ltd., Jerusalem, Israel
| | - Yaniv Gil
- The Hadassah Human Embryonic Stem Cell Research Center, The Goldyne Savad Institute of Gene Therapy and Department of Gynecology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Hanita Khaner
- The Hadassah Human Embryonic Stem Cell Research Center, The Goldyne Savad Institute of Gene Therapy and Department of Gynecology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | | | - Maria Idelson
- The Hadassah Human Embryonic Stem Cell Research Center, The Goldyne Savad Institute of Gene Therapy and Department of Gynecology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Juri Kopolovic
- The Department of Pathology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | | | - Benjamin E. Reubinoff
- The Hadassah Human Embryonic Stem Cell Research Center, The Goldyne Savad Institute of Gene Therapy and Department of Gynecology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
- * E-mail:
| |
Collapse
|
37
|
Westenskow PD, Moreno SK, Krohne TU, Kurihara T, Zhu S, Zhang ZN, Zhao T, Xu Y, Ding S, Friedlander M. Using flow cytometry to compare the dynamics of photoreceptor outer segment phagocytosis in iPS-derived RPE cells. Invest Ophthalmol Vis Sci 2012; 53:6282-90. [PMID: 22871841 DOI: 10.1167/iovs.12-9721] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022] Open
Abstract
PURPOSE Retinal pigment epithelium (RPE) autologous grafts can be readily derived from induced pluripotent stem (iPS) cells. It is critical to stringently characterize iPS-RPE using standardized and quantifiable methods to be confident that they are safe and adequate replacements for diseased RPE before utilizing them in clinical settings. One important and required function is that the iPS-RPE phagocytose photoreceptor outer segments (POS). METHODS We developed a flow cytometry-based assay to monitor binding and internalization of FITC labeled POS by ARPE-19, human fetal RPE (hfRPE), and two types of iPS-RPE. Expression and density of α(v)β₅ integrin, CD36, and MerTK receptors, which are required for phagocytosis, were compared. RESULTS Trypsinization of treated RPE cells results in the release of bound POS. The number of freed POS, the percentage of cells that internalized POS, the brightness of the FITC signal from the cells, and the surface density of the phagocytosis receptors on single RPE cells were measured using flow cytometry. These assays reveal that receptor density is dynamic during differentiation and this can affect the binding and internalization dynamics of the RPE cells. Highly differentiated iPS-RPE phagocytose POS more efficiently than hfRPE. CONCLUSIONS Caution should be exercised to not use RPE grafts until demonstrating that they are fully functional. The density of the phagocytosis receptors is dynamic and may be used as a predictor for how well the iPS-RPE cells will function in vivo. The phagocytosis dynamics observed between iPS-RPE and primary RPE is very encouraging and adds to mounting evidence that iPS-RPE may be a viable replacement for dysfunctional or dying RPE in human patients.
Collapse
Affiliation(s)
- Peter D Westenskow
- Department of Cell Biology, The Scripps Research Institute, La Jolla, California, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Nandrot EF, Silva KE, Scelfo C, Finnemann SC. Retinal pigment epithelial cells use a MerTK-dependent mechanism to limit the phagocytic particle binding activity of αvβ5 integrin. Biol Cell 2012; 104:326-41. [PMID: 22289110 DOI: 10.1111/boc.201100076] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2011] [Accepted: 01/24/2012] [Indexed: 02/06/2023]
Abstract
BACKGROUND INFORMATION αvβ5 integrin and Mer tyrosine kinase (MerTK) receptors reside at the apical surface of the retinal pigment epithelium (RPE) in the eye to promote the diurnal, synchronised phagocytosis of shed photoreceptor outer segment fragments (POS) that is critical for vision. Phagocytosis assays studying RPE cells in culture have defined roles for αvβ5 in POS surface binding and for MerTK in engulfment of bound POS. Both receptors have thus far only been studied separately. It is therefore unknown if αvβ5 integrin activity in POS binding is independent of the engulfment function of RPE cells. This study investigates how increasing αvβ5 receptor levels affect POS binding and internalisation by wild-type (wt), αvβ5- or MerTK-deficient RPE. RESULTS β5 integrin-green fluorescent protein (β5-GFP) fusion proteins formed heterodimeric receptors with endogenous αv integrin subunits at the apical surface of mouse or rat RPE cells that co-immunoprecipitated focal adhesion kinase and redistributed with bound POS such as endogenous αvβ5 receptors. In β5(-/-) RPE cells, de novo formation of αvβ5-GFP receptors restored POS binding and internalisation up to, but not, above wt POS uptake levels. In wt RPE cells, increasing levels of αvβ5 surface receptors by over-expressing β5-GFP only moderately stimulated POS binding, even if POS internalisation was inhibited pharmacologically or by lowering incubation temperatures. In contrast, the same increase in αvβ5 receptor levels dramatically enhanced POS binding of RPE cells lacking MerTK. Furthermore, decreasing MerTK expression by RNA interference increased POS binding to endogenous αvβ5 receptors of wt RPE cells. CONCLUSIONS Expressing β5-GFP is sufficient to reverse phagocytic deficiencies of RPE cells derived from β5(-/-) mice, indicating that these cells do not irreversibly lose other components of the phagocytic machinery. RPE cells expressing the engulfment receptor MerTK control POS binding by limiting activity of endogenous αvβ5 and αvβ5-GFP integrins, although they reside at the apical, phagocytic surface. In contrast, RPE cells permanently or transiently losing MerTK expression lack this regulatory mechanism and bind excess POS via surface αvβ5 receptors. Taken together, these data reveal a novel feedback mechanism that restricts binding of POS to surface αvβ5 integrin receptors in RPE cells.
Collapse
Affiliation(s)
- Emeline F Nandrot
- Department of Ophthalmology, Margaret M. Dyson Vision Research Institute, Weill Medical College of Cornell University, New York, NY 10065, USA
| | | | | | | |
Collapse
|
39
|
Bassani S, Cingolani LA. Tetraspanins: Interactions and interplay with integrins. Int J Biochem Cell Biol 2012; 44:703-8. [PMID: 22326999 DOI: 10.1016/j.biocel.2012.01.020] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2011] [Revised: 01/20/2012] [Accepted: 01/27/2012] [Indexed: 12/14/2022]
Abstract
Tetraspanins are small transmembrane proteins present on the cell surface of almost every eukaryotic cell. Through binding with other transmembrane and intracellular proteins, they regulate diverse cellular processes ranging from cell adhesion and motility to synapse formation and tumor progression. Here, we provide a brief overview of molecular, cellular and clinical studies to illustrate how the multiple functions of this fascinating family of molecules stem from their interplay with multiple molecular partners. In particular, we emphasize the special relationship between tetraspanins and the cell adhesion molecules integrins in regulating cell physiology in health and disease.
Collapse
Affiliation(s)
- Silvia Bassani
- CNR Institute of Neuroscience, Cellular and Molecular Pharmacology, Department of Pharmacology, University of Milan, Italy
| | | |
Collapse
|
40
|
Ruggiero L, Sarang Z, Szondy Z, Finnemann SC. αvβ5 integrin-dependent diurnal phagocytosis of shed photoreceptor outer segments by RPE cells is independent of the integrin coreceptor transglutaminase-2. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 723:731-7. [PMID: 22183400 PMCID: PMC3588855 DOI: 10.1007/978-1-4614-0631-0_93] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Affiliation(s)
- Linda Ruggiero
- Department of Biological Sciences, Fordham University, Bronx, NY 10458, USA
| | - Zsolt Sarang
- Department of Biochemistry and Molecular Biology, Apoptosis and Genomics Research Group, Hungarian Academy of Sciences, University of Debrecen, Debrecen, Hungary
| | - Zsuzsa Szondy
- Department of Biochemistry and Molecular Biology, Apoptosis and Genomics Research Group, Hungarian Academy of Sciences, University of Debrecen, Debrecen, Hungary
| | | |
Collapse
|
41
|
|
42
|
Conley SM, Stuck MW, Naash MI. Structural and functional relationships between photoreceptor tetraspanins and other superfamily members. Cell Mol Life Sci 2011; 69:1035-47. [PMID: 21655915 DOI: 10.1007/s00018-011-0736-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2011] [Revised: 05/12/2011] [Accepted: 05/16/2011] [Indexed: 12/14/2022]
Abstract
The two primary photoreceptor-specific tetraspanins are retinal degeneration slow (RDS) and rod outer segment membrane protein-1 (ROM-1). These proteins associate together to form different complexes necessary for the proper structure of the photoreceptor outer segment rim region. Mutations in RDS cause blinding retinal degenerative disease in both rods and cones by mechanisms that remain unknown. Tetraspanins are implicated in a variety of cellular processes and exert their function via the formation of tetraspanin-enriched microdomains. This review focuses on correlations between RDS and other members of the tetraspanin superfamily, particularly emphasizing protein structure, complex assembly, and post-translational modifications, with the goal of furthering our understanding of the structural and functional role of RDS and ROM-1 in outer segment morphogenesis and maintenance, and our understanding of the pathogenesis associated with RDS and ROM-1 mutations.
Collapse
Affiliation(s)
- Shannon M Conley
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, 73104, USA
| | | | | |
Collapse
|
43
|
Sparrow JR, Hicks D, Hamel CP. The retinal pigment epithelium in health and disease. Curr Mol Med 2011; 10:802-23. [PMID: 21091424 DOI: 10.2174/156652410793937813] [Citation(s) in RCA: 436] [Impact Index Per Article: 31.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2010] [Accepted: 09/13/2010] [Indexed: 12/15/2022]
Abstract
Retinal pigment epithelial cells (RPE) constitute a simple layer of cuboidal cells that are strategically situated behind the photoreceptor (PR) cells. The inconspicuousness of this monolayer contrasts sharply with its importance [1]. The relationship between the RPE and PR cells is crucial to sight; this is evident from basic and clinical studies demonstrating that primary dysfunctioning of the RPE can result in visual cell death and blindness. RPE cells carry out many functions including the conversion and storage of retinoid, the phagocytosis of shed PR outer segment membrane, the absorption of scattered light, ion and fluid transport and RPE-PR apposition. The magnitude of the demands imposed on this single layer of cells in order to execute these tasks, will become apparent to the reader of this review as will the number of clinical disorders that take origin from these cells.
Collapse
Affiliation(s)
- J R Sparrow
- Department of Ophthalmology, Columbia University, New York, NY 10032, USA.
| | | | | |
Collapse
|
44
|
Reisewitz S, Schroeder H, Tort N, Edwards KA, Baeumner AJ, Niemeyer CM. Capture and culturing of living cells on microstructured DNA substrates. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2010; 6:2162-2168. [PMID: 20730825 DOI: 10.1002/smll.201000776] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
A modular system for the DNA-directed immobilization of antibodies was applied to capture living cells on microstructured DNA surfaces. It is demonstrated in two different set-ups, static incubation and hydrodynamic flow, that this approach is well suited for specific capture and selection of cells from culture medium. The adhered cells show intact morphology and they can be cultivated to grow to dense monolayers, restricted to the lateral dimensions of DNA spots on the surface. Owing to the modularity of surface biofunctionalization, the system can readily be configured to serve as a matrix for adhesion and growth of different cells, as demonstrated by specific binding of human embryonic kidney cells (HEK293) and Hodgkin lymphoma L540cy cells onto patches bearing appropriate recognition moieties inside a microfluidic channel. We therefore anticipate that the systems described here should be useful for fundamental research in cell biology or applications in biomedical diagnostics, drug screening, and nanobiotechnology.
Collapse
Affiliation(s)
- Stephanie Reisewitz
- Technische Universität Dortmund, Fakultät Chemie, Biologisch-Chemische Mikrostrukturtechnik, Otto Hahn Str. 6, 44227 Dortmund, Germany
| | | | | | | | | | | |
Collapse
|
45
|
The effects of a CD81 null mutation on retinal pigment epithelium in mice. Neurochem Res 2010; 36:569-73. [PMID: 20882409 DOI: 10.1007/s11064-010-0268-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/08/2010] [Indexed: 10/19/2022]
Abstract
The present study examines the effects of Cd81-null mutation on the development of the retinal pigment epithelium (RPE), specifically cell size and number of cells with multiple nuclei. The outlines of RPE in retinal flat mounts were stained with rhodamine-labeled phalloidin and RPE nuclei with Hoechst stain. The RPE layer was sampled to define the number of cells, the size of the RPE cells and the number of nuclei within the cells. The Cd81-null mutation caused an increase in the number of cells within the RPE layer. The cells were smaller than those in the wild type mice. Furthermore there was an increase in the number of mono-nucleated cells. In the posterior portion of the eye there was a significant increase in the number of multi-nucleated cells. The data indicate that CD81 plays a significant role in the final stages of RPE development, controlling cell number and overall developmental pattern.
Collapse
|
46
|
Kevany BM, Palczewski K. Phagocytosis of retinal rod and cone photoreceptors. Physiology (Bethesda) 2010; 25:8-15. [PMID: 20134024 DOI: 10.1152/physiol.00038.2009] [Citation(s) in RCA: 309] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Photoreceptor cells maintain a roughly constant length by continuously generating new outer segments from their base while simultaneously releasing mature outer segments engulfed by the retinal pigment epithelium (RPE). Thus postmitotic RPE cells phagocytose an immense amount of material over a lifetime, disposing of photoreceptor cell waste while retaining useful content. This review focuses on current knowledge of outer segment phagocytosis, discussing the steps involved along with their critical participants as well as how various perturbations in outer segment (OS) disposal can lead to retinopathies.
Collapse
Affiliation(s)
- Brian M Kevany
- Department of Pharmacology, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA.
| | | |
Collapse
|
47
|
Abstract
Within the integrin family of cell adhesion receptors, integrins alpha3beta1, alpha6beta1, alpha6beta4 and alpha7beta1 make up a laminin-binding subfamily. The literature is divided on the role of these laminin-binding integrins in metastasis, with different studies indicating either pro- or antimetastatic functions. The opposing roles of the laminin-binding integrins in different settings might derive in part from their unusually robust associations with tetraspanin proteins. Tetraspanins organise integrins into multiprotein complexes within discrete plasma membrane domains termed tetraspanin-enriched microdomains (TEMs). TEM association is crucial to the strikingly rapid cell migration mediated by some of the laminin-binding integrins. However, emerging data suggest that laminin-binding integrins also promote the stability of E-cadherin-based cell-cell junctions, and that tetraspanins are essential for this function as well. Thus, TEM association endows the laminin-binding integrins with both pro-invasive functions (rapid migration) and anti-invasive functions (stable cell junctions), and the composition of TEMs in different cell types might help determine the balance between these opposing activities. Unravelling the tetraspanin control mechanisms that regulate laminin-binding integrins will help to define the settings where inhibiting the function of these integrins would be helpful rather than harmful, and may create opportunities to modulate integrin activity in more sophisticated ways than simple functional blockade.
Collapse
|
48
|
Yáñez-Mó M, Barreiro O, Gordon-Alonso M, Sala-Valdés M, Sánchez-Madrid F. Tetraspanin-enriched microdomains: a functional unit in cell plasma membranes. Trends Cell Biol 2009; 19:434-46. [DOI: 10.1016/j.tcb.2009.06.004] [Citation(s) in RCA: 439] [Impact Index Per Article: 27.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2009] [Revised: 06/16/2009] [Accepted: 06/17/2009] [Indexed: 12/14/2022]
|
49
|
Coffey GP, Rajapaksa R, Liu R, Sharpe O, Kuo CC, Krauss SW, Sagi Y, Davis RE, Staudt LM, Sharman JP, Robinson WH, Levy S. Engagement of CD81 induces ezrin tyrosine phosphorylation and its cellular redistribution with filamentous actin. J Cell Sci 2009; 122:3137-44. [PMID: 19654214 DOI: 10.1242/jcs.045658] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
CD81 is a tetraspanin family member involved in diverse cellular interactions in the immune and nervous systems and in cell fusion events. However, the mechanism of action of CD81 and of other tetraspanins has not been defined. We reasoned that identifying signaling molecules downstream of CD81 would provide mechanistic clues. We engaged CD81 on the surface of B-lymphocytes and identified the induced tyrosine-phosphorylated proteins by mass spectrometry. This analysis showed that the most prominent tyrosine phosphorylated protein was ezrin, an actin-binding protein and a member of the ezrin-radixin-moesin family. We also found that CD81 engagement induces spleen tyrosine kinase (Syk) and that Syk was involved in tyrosine phosphorylation of ezrin. After engagement of CD81, it colocalized with ezrin and F-actin, and this association was disrupted when Syk activation was blocked. Taken together, these studies suggest a model in which CD81 interfaces between the plasma membrane and the cytoskeleton by activating Syk, mobilizing ezrin, and recruiting F-actin to facilitate cytoskeletal reorganization and cell signaling. This mechanism might explain the pleiotropic effects induced in response to stimulation of cells by anti-CD81 antibodies or by the hepatitis C virus, which uses this molecule as its key receptor.
Collapse
Affiliation(s)
- Greg P Coffey
- Stanford University, School of Medicine, Division of Oncology, Stanford, CA 94305, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Wang AL, Lukas TJ, Yuan M, Du N, Handa JT, Neufeld AH. Changes in retinal pigment epithelium related to cigarette smoke: possible relevance to smoking as a risk factor for age-related macular degeneration. PLoS One 2009; 4:e5304. [PMID: 19390692 PMCID: PMC2669185 DOI: 10.1371/journal.pone.0005304] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2009] [Accepted: 03/30/2009] [Indexed: 11/19/2022] Open
Abstract
Age-related Macular Degeneration (AMD) is a major cause of central vision loss in the elderly and smoking is a primary risk factor associated with the prevalence and incidence of AMD. To better understand the cellular and molecular bases for the association between smoking and AMD, we determined the effects of Benzo(a)Pyrene (B(a)P), a toxic element in cigarette smoke, on cultured retinal pigment epithelia (RPE) and we examined the RPE/choroid from mice exposed to chronic cigarette smoke. We measured: mitochondrial DNA (mtDNA) damage, phagocytic activity, lysosomal enzymes, exosome markers and selected complement pathway components. In the presence of a non-cytotoxic dose of B(a)P, there was extensive mtDNA damage but no nuclear DNA damage. RPE phagocytic activity was not altered but there were increased lysosomal activity, exocytotic activity and complement pathway components. Retinas from mice exposed to cigarette smoke contained markers for mtDNA damage, exosomes and complement pathway components surrounding Bruch's membrane. Markers for these processes are found in drusen from AMD patients. Thus, smoking may cause damage to mtDNA and increased degradative processes in the RPE. These altered cell biological processes in the RPE may contribute to the formation of drusen in individuals who are cigarette smokers and underlie susceptibility to genetic mutations associated with AMD.
Collapse
Affiliation(s)
- Ai Ling Wang
- Forsythe Laboratory for the Investigation of the Aging Retina, Department of Ophthalmology, Northwestern University School of Medicine, Chicago, Illinois, United States of America.
| | | | | | | | | | | |
Collapse
|