201
|
Liu J, Zhu G, Jia N, Wang W, Wang Y, Yin M, Jiang X, Huang Y, Zhang J. CD9 regulates keratinocyte migration by negatively modulating the sheddase activity of ADAM17. Int J Biol Sci 2019; 15:493-506. [PMID: 30745837 PMCID: PMC6367546 DOI: 10.7150/ijbs.29404] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 12/13/2018] [Indexed: 12/17/2022] Open
Abstract
CD9 is a trans-membrane protein, and has recently been implicated in different physiological and cellular processes, such as cell migration and adhesion. According to previous study, down-regulation of CD9 contributes to keratinocyte migration, critical for wound re-epithelialization. Nevertheless, it is widely believed that tetraspanin CD9 does not have ligands or function as the cell surface receptor, rather it is thought to associate with other transmembrane molecules, thereby mediate keratinocyte migration. Little is known about how CD9 associates with transmembrane molecules in migratory keratinocytes. Here, using confocal microscopy, we observed that tetraspanin CD9 and ADAM17 co-localized on the surface of keratinocytes in the course of wound repair in vivo and in vitro. Co-immunoprecipitation experiments demonstrated a direct association between CD9 and ADAM17 in HaCaT cells and C57-MKs. Functional studies revealed that down-regulation or over-expression of CD9 exerted negative regulatory effects on ADAM17 sheddase activity. This activity is involved in CD9-regulated cell motility and migration. Further studies found that ADAM17 inhibitor-TAPI-2 or siADAM17 significantly abolished the enhanced effect of keratinocyte migration induced by CD9 down-regulation. Meanwhile, the sheddase activity of ADAM17 was inhibited by TAPI-2, which decreased this release of AREG and HB-EGF in CD9-silenced HaCat cells and C57-MKs. Importantly, neutralizing antibody against HB-EGF significant weakened keratinocyte migration and motility in CD9-silenced keratinocytes, and the inhibition of CD9-regulated keratinocyte migration by siADAM17 was rescued by addition of recombinant HB-EGF, activating EGFR/ERK pathway. Collectively, our results suggest that ADAM17 sheddase activity is activated by down-regulation of CD9, thereby mediating shedding of HB-EGF and activation of EGFR/ERK signaling, which crucially affects the keratinocyte migration and wound healing.
Collapse
Affiliation(s)
- Jie Liu
- Institute of Burn Research, State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Third Military Medical University(Army Medical University), Chongqing, China
| | - Guoqin Zhu
- Institute of Burn Research, State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Third Military Medical University(Army Medical University), Chongqing, China
| | - Naixin Jia
- Institute of Burn Research, State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Third Military Medical University(Army Medical University), Chongqing, China
| | - Weiyi Wang
- Institute of Burn Research, State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Third Military Medical University(Army Medical University), Chongqing, China
| | - Yuan Wang
- Institute of Burn Research, State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Third Military Medical University(Army Medical University), Chongqing, China
| | - Meifang Yin
- Institute of Burn Research, State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Third Military Medical University(Army Medical University), Chongqing, China
| | - Xuping Jiang
- Institute of Burn Research, State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Third Military Medical University(Army Medical University), Chongqing, China
| | - Yuesheng Huang
- Institute of Burn Research, State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Third Military Medical University(Army Medical University), Chongqing, China
| | - Jiaping Zhang
- Institute of Burn Research, State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Third Military Medical University(Army Medical University), Chongqing, China
- Department of plastic Surgery, Southwest Hospital, Third Military Medical University(Army Medical University), Chongqing, China
| |
Collapse
|
202
|
Mechanisms of receptor shedding in platelets. Blood 2018; 132:2535-2545. [DOI: 10.1182/blood-2018-03-742668] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 10/12/2018] [Indexed: 02/07/2023] Open
Abstract
Abstract
The ability to upregulate and downregulate surface-exposed proteins and receptors is a powerful process that allows a cell to instantly respond to its microenvironment. In particular, mobile cells in the bloodstream must rapidly react to conditions where infection or inflammation are detected, and become proadhesive, phagocytic, and/or procoagulant. Platelets are one such blood cell that must rapidly acquire and manage proadhesive and procoagulant properties in order to execute their primary function in hemostasis. The regulation of platelet membrane properties is achieved via several mechanisms, one of which involves the controlled metalloproteolytic release of adhesion receptors and other proteins from the platelet surface. Proteolysis effectively lowers receptor density and reduces the reactivity of platelets, and is a mechanism to control robust platelet activation. Recent research has also established clear links between levels of platelet receptors and platelet lifespan. In this review, we will discuss the current knowledge of metalloproteolytic receptor regulation in the vasculature with emphasis on the platelet receptor system to highlight how receptor density can influence both platelet function and platelet survival.
Collapse
|
203
|
Exosomes-the enigmatic regulators of bone homeostasis. Bone Res 2018; 6:36. [PMID: 30534458 PMCID: PMC6286319 DOI: 10.1038/s41413-018-0039-2] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Revised: 10/18/2018] [Accepted: 10/19/2018] [Indexed: 12/12/2022] Open
Abstract
Exosomes are a heterogeneous group of cell-derived membranous structures, which mediate crosstalk interaction between cells. Recent studies have revealed a close relationship between exosomes and bone homeostasis. It is suggested that bone cells can spontaneously secret exosomes containing proteins, lipids and nucleic acids, which then to regulate osteoclastogenesis and osteogenesis. However, the network of regulatory activities of exosomes in bone homeostasis as well as their therapeutic potential in bone injury remain largely unknown. This review will detail and discuss the characteristics of exosomes, the regulatory activities of exosomes in bone homeostasis as well as the clinical potential of exosomes in bone injury. Vesicles known as exosomes may prove to be valuable clinical tools once their function is clarified. Exosomes were discovered in the 1980s but not observed in bone tissue until 2003. Minghao Zheng of the University of Western Australia, together with colleagues elsewhere, has reviewed the biology of exosomes, their role in maintaining bones, and their potential clinical uses. Exosomes carry lipids, proteins, and nucleic acids between cells. They are released by every type of bone cell, with the role of each exosome determined by its specific contents. Exosome-mediated crosstalk is involved in regulating bone remodeling, and exosomes have also been implicated in myelomas. Recent work has shown that exosome treatment can improve fracture healing. The authors conclude that a better understanding of the role of exosomes in bone homeostasis will unlock their significant clinical potential.
Collapse
|
204
|
Vicente-Manzanares M, Sánchez-Madrid F. Targeting the integrin interactome in human disease. Curr Opin Cell Biol 2018; 55:17-23. [DOI: 10.1016/j.ceb.2018.05.010] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 05/07/2018] [Accepted: 05/14/2018] [Indexed: 12/21/2022]
|
205
|
Zuidema A, Wang W, Kreft M, Te Molder L, Hoekman L, Bleijerveld OB, Nahidiazar L, Janssen H, Sonnenberg A. Mechanisms of integrin αVβ5 clustering in flat clathrin lattices. J Cell Sci 2018; 131:jcs221317. [PMID: 30301780 DOI: 10.1242/jcs.221317] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 09/27/2018] [Indexed: 12/17/2023] Open
Abstract
The family of integrin transmembrane receptors is essential for the normal function of multicellular organisms by facilitating cell-extracellular matrix adhesion. The vitronectin-binding integrin αVβ5 localizes to focal adhesions (FAs) as well as poorly characterized flat clathrin lattices (FCLs). Here, we show that, in human keratinocytes, αVβ5 is predominantly found in FCLs, and formation of the αVβ5-containing FCLs requires the presence of vitronectin as ligand, Ca2+, and the clathrin adaptor proteins ARH (also known as LDLRAP1), Numb and EPS15/EPS15L1. Integrin chimeras, containing the extracellular and transmembrane domains of β5 and the cytoplasmic domains of β1 or β3, almost exclusively localize in FAs. Interestingly, lowering actomyosin-mediated contractility promotes integrin redistribution to FLCs in an integrin tail-dependent manner, while increasing cellular tension favors αVβ5 clustering in FAs. Our findings strongly indicate that clustering of integrin αVβ5 in FCLs is dictated by the β5 subunit cytoplasmic domain, cellular tension and recruitment of specific adaptor proteins to the β5 subunit cytoplasmic domains.
Collapse
Affiliation(s)
- Alba Zuidema
- Division of Cell Biology I, The Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam 1066 CX, The Netherlands
| | - Wei Wang
- Division of Cell Biology I, The Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam 1066 CX, The Netherlands
| | - Maaike Kreft
- Division of Cell Biology I, The Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam 1066 CX, The Netherlands
| | - Lisa Te Molder
- Division of Cell Biology I, The Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam 1066 CX, The Netherlands
| | - Liesbeth Hoekman
- Mass spectrometry/Proteomics Facility, The Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam 1066 CX, The Netherlands
| | - Onno B Bleijerveld
- Mass spectrometry/Proteomics Facility, The Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam 1066 CX, The Netherlands
| | - Leila Nahidiazar
- Division of Cell Biology I, The Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam 1066 CX, The Netherlands
| | - Hans Janssen
- Electron Microscopy Facility, The Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam 1066 CX, The Netherlands
| | - Arnoud Sonnenberg
- Division of Cell Biology I, The Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam 1066 CX, The Netherlands
| |
Collapse
|
206
|
Cao J, Tan X. Comparative analysis of the tetraspanin gene family in six teleost fishes. FISH & SHELLFISH IMMUNOLOGY 2018; 82:432-441. [PMID: 30145201 DOI: 10.1016/j.fsi.2018.08.048] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Revised: 07/02/2018] [Accepted: 08/22/2018] [Indexed: 06/08/2023]
Abstract
Tetraspanins are a family of membrane proteins, which play important roles in many aspects of cell biology and physiology via binding other tetraspanins or proteins. In this study, we identified 251 putative tetraspanin genes in 6 teleost fishes. Conserved gene organization and motif distribution suggested their functional relevance existing in each group. Synteny analyses implied conserved and dynamic evolution characteristics of this gene family in several vertebrates. We also found that some recombination events have accelerated the evolution of this gene family. Moreover, a few positive selection sites were identified. Expression patterns of some tetraspanins were further studied under organophosphorus stress using transcriptome sequencing. Functional network analyses identified some interacting genes that exhibited 174 interactions, which reflected the diversity of tetraspanin binding proteins. The results will provide a foundation for the further functional investigation of the tetraspanin genes in fishes.
Collapse
Affiliation(s)
- Jun Cao
- Institute of Life Sciences, Jiangsu University, Zhenjiang, 212013, China.
| | - Xiaona Tan
- Institute of Life Sciences, Jiangsu University, Zhenjiang, 212013, China
| |
Collapse
|
207
|
Liao Y, Chang HC, Liang FX, Chung PJ, Wei Y, Nguyen TP, Zhou G, Talebian S, Krey LC, Deng FM, Wong TW, Chicote JU, Grifo JA, Keefe DL, Shapiro E, Lepor H, Wu XR, DeSalle R, Garcia-España A, Kim SY, Sun TT. Uroplakins play conserved roles in egg fertilization and acquired additional urothelial functions during mammalian divergence. Mol Biol Cell 2018; 29:3128-3143. [PMID: 30303751 PMCID: PMC6340209 DOI: 10.1091/mbc.e18-08-0496] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Uroplakin (UP) tetraspanins and their associated proteins are major mammalian urothelial differentiation products that form unique two-dimensional crystals of 16-nm particles (“urothelial plaques”) covering the apical urothelial surface. Although uroplakins are highly expressed only in mammalian urothelium and are often referred to as being urothelium specific, they are also expressed in several mouse nonurothelial cell types in stomach, kidney, prostate, epididymis, testis/sperms, and ovary/oocytes. In oocytes, uroplakins colocalize with CD9 on cell-surface and multivesicular body-derived exosomes, and the cytoplasmic tail of UPIIIa undergoes a conserved fertilization-dependent, Fyn-mediated tyrosine phosphorylation that also occurs in Xenopus laevis eggs. Uroplakin knockout and antibody blocking reduce mouse eggs’ fertilization rate in in vitro fertilization assays, and UPII/IIIa double-knockout mice have a smaller litter size. Phylogenetic analyses showed that uroplakin sequences underwent significant mammal-specific changes. These results suggest that, by mediating signal transduction and modulating membrane stability that do not require two-dimensional-crystal formation, uroplakins can perform conserved and more ancestral fertilization functions in mouse and frog eggs. Uroplakins acquired the ability to form two-dimensional-crystalline plaques during mammalian divergence, enabling them to perform additional functions, including umbrella cell enlargement and the formation of permeability and mechanical barriers, to protect/modify the apical surface of the modern-day mammalian urothelium.
Collapse
Affiliation(s)
- Yi Liao
- Department of Cell Biology, New York University School of Medicine, New York, NY 10016
| | - Hung-Chi Chang
- Department of Obstetrics and Gynecology, New York University School of Medicine, New York, NY 10016.,Department of Obstetrics and Gynecology, National Taiwan University, Taipei 10617, Taiwan
| | - Feng-Xia Liang
- Department of Cell Biology, New York University School of Medicine, New York, NY 10016
| | | | - Yuan Wei
- Department of Cell Biology, New York University School of Medicine, New York, NY 10016
| | - Tuan-Phi Nguyen
- Department of Cell Biology, New York University School of Medicine, New York, NY 10016
| | - Ge Zhou
- Regeneron, Tarrytown, NY 10591
| | - Sheeva Talebian
- Department of Obstetrics and Gynecology, New York University School of Medicine, New York, NY 10016
| | - Lewis C Krey
- Department of Obstetrics and Gynecology, New York University School of Medicine, New York, NY 10016
| | - Fang-Ming Deng
- Department of Pathology, New York University School of Medicine, New York, NY 10016.,Department of Urology, New York University School of Medicine, New York, NY 10016
| | - Tak-Wah Wong
- Department of Dermatology, National Cheng Kung University, Tainan 701, Taiwan
| | - Javier U Chicote
- Unitat De Recerca, Hospital Joan XXIII, Institut de Investigacio Sanitaria Pere Virgili (IISPV), Universitat Rovira i Virgili, Tarragona 43007, Spain
| | - James A Grifo
- Department of Obstetrics and Gynecology, New York University School of Medicine, New York, NY 10016
| | - David L Keefe
- Department of Obstetrics and Gynecology, New York University School of Medicine, New York, NY 10016
| | - Ellen Shapiro
- Department of Urology, New York University School of Medicine, New York, NY 10016
| | - Herbert Lepor
- Department of Urology, New York University School of Medicine, New York, NY 10016.,Sackler Institute of Comparative Genomics, American Museum of Natural History, New York, NY 10024
| | - Xue-Ru Wu
- Department of Pathology, New York University School of Medicine, New York, NY 10016.,Department of Urology, New York University School of Medicine, New York, NY 10016.,Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, NY 10016
| | - Robert DeSalle
- Veterans Affairs New York Harbor Healthcare System, New York, NY 10010
| | - Antonio Garcia-España
- Unitat De Recerca, Hospital Joan XXIII, Institut de Investigacio Sanitaria Pere Virgili (IISPV), Universitat Rovira i Virgili, Tarragona 43007, Spain
| | - Sang Yong Kim
- Department of Pathology, New York University School of Medicine, New York, NY 10016
| | - Tung-Tien Sun
- Department of Cell Biology, New York University School of Medicine, New York, NY 10016.,Department of Urology, New York University School of Medicine, New York, NY 10016.,The Ronald O. Perelman Department of Dermatology, New York University School of Medicine, New York, NY 10016.,Sackler Institute of Comparative Genomics, American Museum of Natural History, New York, NY 10024
| |
Collapse
|
208
|
Genome-wide association studies for corneal and refractive astigmatism in UK Biobank demonstrate a shared role for myopia susceptibility loci. Hum Genet 2018; 137:881-896. [PMID: 30306274 PMCID: PMC6267700 DOI: 10.1007/s00439-018-1942-8] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Accepted: 09/25/2018] [Indexed: 01/08/2023]
Abstract
Previous studies have suggested that naturally occurring genetic variation contributes to the risk of astigmatism. The purpose of this investigation was to identify genetic markers associated with corneal and refractive astigmatism in a large-scale European ancestry cohort (UK Biobank) who underwent keratometry and autorefraction at an assessment centre. Genome-wide association studies for corneal and refractive astigmatism were performed in individuals of European ancestry (N = 86,335 and 88,005 respectively), with the mean corneal astigmatism or refractive astigmatism in fellow eyes analysed as a quantitative trait (dependent variable). Genetic correlation between the two traits was calculated using LD Score regression. Gene-based and gene-set tests were carried out using MAGMA. Single marker-based association tests for corneal astigmatism identified four genome-wide significant loci (P < 5 × 10-8) near the genes ZC3H11B (1q41), LINC00340 (6p22.3), HERC2/OCA2 (15q13.1) and NPLOC4/TSPAN10 (17q25.3). Three of these loci also demonstrated genome-wide significant association with refractive astigmatism: LINC00340, HERC2/OCA2 and NPLOC4/TSPAN10. The genetic correlation between corneal and refractive astigmatism was 0.85 (standard error = 0.068, P = 1.37 × 10-35). Here, we have undertaken the largest genome-wide association studies for corneal and refractive astigmatism to date and identified four novel loci for corneal astigmatism, two of which were also novel loci for refractive astigmatism. These loci have previously demonstrated association with axial length (ZC3H11B), myopia (NPLOC4), spherical equivalent refractive error (LINC00340) and eye colour (HERC2). The shared role of these novel candidate genes for astigmatism lends further support to the shared genetic susceptibility of myopia and astigmatism.
Collapse
|
209
|
Dias MVS, Costa CS, daSilva LLP. The Ambiguous Roles of Extracellular Vesicles in HIV Replication and Pathogenesis. Front Microbiol 2018; 9:2411. [PMID: 30364166 PMCID: PMC6191503 DOI: 10.3389/fmicb.2018.02411] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 09/20/2018] [Indexed: 12/14/2022] Open
Abstract
Cells from all kingdoms of life can release membrane-enclosed vesicles to the extracellular milieu. These extracellular vesicles (EVs) may function as mediators of intercellular communication, allowing the transfer of biologically active molecules between cells and organisms. It has become clear that HIV particles and certain types of EVs, such as exosomes, share many similarities regarding morphology, composition, and biogenesis. This review presents a summary of the literature describing the intricate relationship between HIV and EVs biogenesis. Also, we discuss the latest progress toward understanding the mechanisms by which EVs influence HIV pathogenesis, as well as, how HIV modulates EVs composition in infected cells to facilitate viral spread.
Collapse
Affiliation(s)
- Marcos V S Dias
- Center for Virus Research, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil.,Department of Cell and Molecular Biology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Cristina S Costa
- Center for Virus Research, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil.,Department of Cell and Molecular Biology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Luis L P daSilva
- Center for Virus Research, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil.,Department of Cell and Molecular Biology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| |
Collapse
|
210
|
Inhibition of Tetraspanin Functions Impairs Human Papillomavirus and Cytomegalovirus Infections. Int J Mol Sci 2018; 19:ijms19103007. [PMID: 30279342 PMCID: PMC6212908 DOI: 10.3390/ijms19103007] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 09/23/2018] [Accepted: 09/27/2018] [Indexed: 12/14/2022] Open
Abstract
Tetraspanins are suggested to regulate the composition of cell membrane components and control intracellular transport, which leaves them vulnerable to utilization by pathogens such as human papillomaviruses (HPV) and cytomegaloviruses (HCMV) to facilitate host cell entry and subsequent infection. In this study, by means of cellular depletion, the cluster of differentiation (CD) tetraspanins CD9, CD63, and CD151 were found to reduce HPV16 infection in HeLa cells by 50 to 80%. Moreover, we tested recombinant proteins or peptides of specific tetraspanin domains on their effect on the most oncogenic HPV type, HPV16, and HCMV. We found that the C-terminal tails of CD63 and CD151 significantly inhibited infections of both HPV16 and HCMV. Although CD9 was newly identified as a key cellular factor for HPV16 infection, the recombinant CD9 C-terminal peptide had no effect on infection. Based on the determined half-maximal inhibitory concentration (IC50), we classified CD63 and CD151 C-terminal peptides as moderate to potent inhibitors of HPV16 infection in HeLa and HaCaT cells, and in EA.hy926, HFF (human foreskin fibroblast) cells, and HEC-LTT (human endothelial cell-large T antigen and telomerase) cells for HCMV, respectively. These results indicate that HPV16 and HCMV share similar cellular requirements for their entry into host cells and reveal the necessity of the cytoplasmic CD151 and CD63 C-termini in virus infections. Furthermore, this highlights the suitability of these peptides for functional investigation of tetraspanin domains and as inhibitors of pathogen infections.
Collapse
|
211
|
Romancino DP, Buffa V, Caruso S, Ferrara I, Raccosta S, Notaro A, Campos Y, Noto R, Martorana V, Cupane A, Giallongo A, d'Azzo A, Manno M, Bongiovanni A. Palmitoylation is a post-translational modification of Alix regulating the membrane organization of exosome-like small extracellular vesicles. Biochim Biophys Acta Gen Subj 2018; 1862:2879-2887. [PMID: 30251702 DOI: 10.1016/j.bbagen.2018.09.004] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 09/04/2018] [Accepted: 09/06/2018] [Indexed: 12/11/2022]
Abstract
BACKGROUND Virtually all cell types have the capacity to secrete nanometer-sized extracellular vesicles, which have emerged in recent years as potent signal transducers and cell-cell communicators. The multifunctional protein Alix is a bona fide exosomal regulator and skeletal muscle cells can release Alix-positive nano-sized extracellular vesicles, offering a new paradigm for understanding how myofibers communicate within skeletal muscle and with other organs. S-palmitoylation is a reversible lipid post-translational modification, involved in different biological processes, such as the trafficking of membrane proteins, achievement of stable protein conformations, and stabilization of protein interactions. METHODS Here, we have used an integrated biochemical-biophysical approach to determine whether S-palmitoylation contributes to the regulation of extracellular vesicle production in skeletal muscle cells. RESULTS We ascertained that Alix is S-palmitoylated and that this post-translational modification influences its protein-protein interaction with CD9, a member of the tetraspanin protein family. Furthermore, we showed that the structural organization of the lipid bilayer of the small (nano-sized) extracellular vesicle membrane with altered palmitoylation is qualitatively different compared to mock control vesicles. CONCLUSIONS We propose that S-palmitoylation regulates the function of Alix in facilitating the interactions among extracellular vesicle-specific regulators and maintains the proper structural organization of exosome-like extracellular vesicle membranes. GENERAL SIGNIFICANCE Beyond its biological relevance, our study also provides the means for a comprehensive structural characterization of EVs.
Collapse
Affiliation(s)
- Daniele P Romancino
- Institute of Biomedicine and Molecular Immunology (IBIM), National Research Council (CNR) of Italy, Palermo, Italy
| | - Valentina Buffa
- Institute of Biomedicine and Molecular Immunology (IBIM), National Research Council (CNR) of Italy, Palermo, Italy
| | - Stefano Caruso
- UMR-1162, Functional Genomics of Solid Tumors, Inserm, Paris 1162, France
| | - Ines Ferrara
- Institute of Biomedicine and Molecular Immunology (IBIM), National Research Council (CNR) of Italy, Palermo, Italy
| | - Samuele Raccosta
- Institute of Biophysics (IBF), National Research Council (CNR) of Italy, Palermo, Italy
| | - Antonietta Notaro
- Institute of Biomedicine and Molecular Immunology (IBIM), National Research Council (CNR) of Italy, Palermo, Italy
| | - Yvan Campos
- Department of Genetics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Rosina Noto
- Institute of Biophysics (IBF), National Research Council (CNR) of Italy, Palermo, Italy
| | - Vincenzo Martorana
- Institute of Biophysics (IBF), National Research Council (CNR) of Italy, Palermo, Italy
| | - Antonio Cupane
- Department of Physics and Chemistry, University of Palermo, Italy
| | - Agata Giallongo
- Institute of Biomedicine and Molecular Immunology (IBIM), National Research Council (CNR) of Italy, Palermo, Italy
| | - Alessandra d'Azzo
- Department of Genetics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Mauro Manno
- Institute of Biophysics (IBF), National Research Council (CNR) of Italy, Palermo, Italy
| | - Antonella Bongiovanni
- Institute of Biomedicine and Molecular Immunology (IBIM), National Research Council (CNR) of Italy, Palermo, Italy.
| |
Collapse
|
212
|
Wang L, Kempton JB, Brigande JV. Gene Therapy in Mouse Models of Deafness and Balance Dysfunction. Front Mol Neurosci 2018; 11:300. [PMID: 30210291 PMCID: PMC6123355 DOI: 10.3389/fnmol.2018.00300] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 08/06/2018] [Indexed: 12/16/2022] Open
Abstract
Therapeutic strategies to restore hearing and balance in mouse models of inner ear disease aim to rescue sensory function by gene replacement, augmentation, knock down or knock out. Modalities to achieve therapeutic effects have utilized virus-mediated transfer of wild type genes and small interfering ribonucleic acids; systemic and focal administration of antisense oligonucleotides (ASO) and designer small molecules; and lipid-mediated transfer of Cas 9 ribonucleoprotein (RNP) complexes. This work has established that gene or drug administration to the structurally and functionally immature, early neonatal mouse inner ear prior to hearing onset is a prerequisite for the most robust therapeutic responses. These observations may have significant implications for translating mouse inner ear gene therapies to patients. The human fetus hears by gestational week 19, suggesting that a corollary window of therapeutic efficacy closes early in the second trimester of pregnancy. We hypothesize that fetal therapeutics deployed prior to hearing onset may be the most effective approach to preemptively manage genetic mutations that cause deafness and vestibular dysfunction. We assert that gene therapy studies in higher vertebrate model systems with fetal hearing onset and a comparable acoustic range and sensitivity to that of humans are an essential step to safely and effectively translate murine gene therapies to the clinic.
Collapse
Affiliation(s)
- Lingyan Wang
- Oregon Hearing Research Center, Department of Otolaryngology, Oregon Health & Science University, Portland, OR, United States
| | - J Beth Kempton
- Oregon Hearing Research Center, Department of Otolaryngology, Oregon Health & Science University, Portland, OR, United States
| | - John V Brigande
- Oregon Hearing Research Center, Department of Otolaryngology, Oregon Health & Science University, Portland, OR, United States
| |
Collapse
|
213
|
Fish EJ, Irizarry KJ, DeInnocentes P, Ellis CJ, Prasad N, Moss AG, Curt Bird R. Malignant canine mammary epithelial cells shed exosomes containing differentially expressed microRNA that regulate oncogenic networks. BMC Cancer 2018; 18:832. [PMID: 30126376 PMCID: PMC6102898 DOI: 10.1186/s12885-018-4750-6] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Accepted: 08/14/2018] [Indexed: 12/18/2022] Open
Abstract
Background Breast (mammary) cancers in human (BC) and canine (CMT) patients share clinical, pathological, and molecular similarities that suggest dogs may be a useful translational model. Many cancers, including BC, shed exosomes that contain microRNAs (miRs) into the microenvironment and circulation, and these may represent biomarkers of metastasis and tumor phenotype. Methods Three normal canine mammary epithelial cell (CMEC) cultures and 5 CMT cell lines were grown in serum-free media. Exosomes were isolated from culture media by ultracentrifugation then profiled by transmission electron microscopy, dynamic light scattering, and Western blot. Exosomal small RNA was deep-sequenced on an Illumina HiSeq2500 sequencer and validated by qRT-PCR. In silico bioinformatic analysis was carried out to determine microRNA gene and pathway targets. Results CMEC and CMT cell lines shed round, “cup-shaped” exosomes approximately 150–200 nm, and were immunopositive for exosomal marker CD9. Deep-sequencing averaged ~ 15 million reads/sample. Three hundred thirty-eight unique miRs were detected, with 145 having > ±1.5-fold difference between one or more CMT and CMEC samples. Gene ontology analysis revealed that the upregulated miRs in this exosomal population regulate a number of relevant oncogenic networks. Several miRNAs including miR-18a, miR-19a and miR-181a were predicted in silico to target the canine estrogen receptor (ESR1α). Conclusions CMEC and CMT cells shed exosomes in vitro that contain differentially expressed miRs. CMT exosomal RNA expresses a limited number of miRs that are up-regulated relative to CMEC, and these are predicted to target biologically relevant hormone receptors and oncogenic pathways. These results may inform future studies of circulating exosomes and the utility of miRs as biomarkers of breast cancer in women and dogs. Electronic supplementary material The online version of this article (10.1186/s12885-018-4750-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Eric J Fish
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, 166 Greene Hall, Auburn, AL, 36849, USA.
| | - Kristopher J Irizarry
- College of Veterinary Medicine, Western University of Health Sciences, Pomona, CA, 91766, USA
| | - Patricia DeInnocentes
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, 166 Greene Hall, Auburn, AL, 36849, USA
| | - Connor J Ellis
- College of Veterinary Medicine, Western University of Health Sciences, Pomona, CA, 91766, USA
| | - Nripesh Prasad
- Genomic Services Laboratory, Hudson Alpha Institute for Biotechnology, Huntsville, AL, 35806, USA
| | - Anthony G Moss
- Department of Biology, College of Science and Mathematics, Auburn University, Auburn, AL, 36849, USA
| | - R Curt Bird
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, 166 Greene Hall, Auburn, AL, 36849, USA
| |
Collapse
|
214
|
Pathogens and Their Effect on Exosome Biogenesis and Composition. Biomedicines 2018; 6:biomedicines6030079. [PMID: 30041409 PMCID: PMC6164629 DOI: 10.3390/biomedicines6030079] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 07/17/2018] [Accepted: 07/17/2018] [Indexed: 12/12/2022] Open
Abstract
Exosomes are nanosized membrane microvesicles (30⁻100 nm) that have the capability to communicate intercellularly and transport cell components (i.e., miRNA, mRNA, proteins and DNA). Exosomes are found in nearly every cell type (i.e., mast cells, dendritic, tumor, and macrophages). There have been many studies that have shown the importance of exosome function as well as their unique packaging and targeting abilities. These characteristics make exosomes ideal candidates to act as biomarkers and therapeutics for disease. We will discuss the biogenesis, composition, and relationship of exosomes with non-viral microbial infections including gram-negative bacteria, gram-positive bacteria, Leishmania and Trypanosoma cruzi.
Collapse
|
215
|
Thomson-Luque R, Wang C, Ntumngia FB, Xu S, Szekeres K, Conway A, Adapa SR, Barnes SJ, Adams JH, Jiang RHY. In-depth phenotypic characterization of reticulocyte maturation using mass cytometry. Blood Cells Mol Dis 2018; 72:22-33. [PMID: 30007855 PMCID: PMC6097872 DOI: 10.1016/j.bcmd.2018.06.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 06/24/2018] [Accepted: 06/24/2018] [Indexed: 12/17/2022]
Abstract
Progress towards an in-depth understanding of the final steps of the erythroid lineage development is paramount for many hematological diseases. We have characterized the final stages of reticulocyte maturation from bone marrow to peripheral blood using for the first time single-cell Mass Cytometry (CyTOF). We were able to measure the expression of 31 surface markers within a single red blood cell (RBC). We demonstrate the validity of CyTOF for RBC phenotyping by confirming the progressive reduction of transferrin receptor 1 (CD71) during reticulocyte maturation to mature RBC. We highlight the high-dimensional nature of mass cytometry data by correlating the expression of multiple proteins on individual RBCs. We further describe a more drastic reduction pattern for a component of the alpha4/beta1 integrin CD49d at the very early steps of reticulocyte maturation in bone marrow and directly linked with the mitochondria remnants clearance pattern. The enhanced and accurate RBC phenotyping potential of CyTOF described herein could be beneficial to decipher RBC preferences, as well as still not well understood receptor-ligand interaction of some hemotropic parasites such as the malaria causing agent Plasmodium vivax.
Collapse
Affiliation(s)
- Richard Thomson-Luque
- Center for Global Health and Infectious Diseases Research, College of Public Health, University of South Florida, Tampa, USA
| | - Chengqi Wang
- Center for Global Health and Infectious Diseases Research, College of Public Health, University of South Florida, Tampa, USA
| | - Francis B Ntumngia
- Center for Global Health and Infectious Diseases Research, College of Public Health, University of South Florida, Tampa, USA
| | - Shulin Xu
- Center for Global Health and Infectious Diseases Research, College of Public Health, University of South Florida, Tampa, USA
| | - Karoly Szekeres
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, USA
| | - Amy Conway
- Center for Global Health and Infectious Diseases Research, College of Public Health, University of South Florida, Tampa, USA
| | - Swamy Rakesh Adapa
- Center for Global Health and Infectious Diseases Research, College of Public Health, University of South Florida, Tampa, USA
| | - Samantha J Barnes
- Center for Global Health and Infectious Diseases Research, College of Public Health, University of South Florida, Tampa, USA
| | - John H Adams
- Center for Global Health and Infectious Diseases Research, College of Public Health, University of South Florida, Tampa, USA.
| | - Rays H Y Jiang
- Center for Global Health and Infectious Diseases Research, College of Public Health, University of South Florida, Tampa, USA.
| |
Collapse
|
216
|
Matthews AL, Koo CZ, Szyroka J, Harrison N, Kanhere A, Tomlinson MG. Regulation of Leukocytes by TspanC8 Tetraspanins and the "Molecular Scissor" ADAM10. Front Immunol 2018; 9:1451. [PMID: 30013551 PMCID: PMC6036176 DOI: 10.3389/fimmu.2018.01451] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Accepted: 06/12/2018] [Indexed: 01/16/2023] Open
Abstract
A disintegrin and metalloproteinase 10 (ADAM10) is a ubiquitous transmembrane protein that functions as a "molecular scissor" to cleave the extracellular regions from its transmembrane target proteins. ADAM10 is well characterized as the ligand-dependent activator of Notch proteins, which control cell fate decisions. Indeed, conditional knockouts of ADAM10 in mice reveal impaired B-, T-, and myeloid cell development and/or function. ADAM10 cleaves many other leukocyte-expressed substrates. On B-cells, ADAM10 cleavage of the low-affinity IgE receptor CD23 promotes allergy and asthma, cleavage of ICOS ligand impairs antibody responses, and cleavage of the BAFF-APRIL receptor transmembrane activator and CAML interactor, and BAFF receptor, reduce B-cell survival. On microglia, increased ADAM10 cleavage of a rare variant of the scavenger receptor triggering receptor expressed on myeloid cells 2 may increase susceptibility to Alzheimer's disease. We and others recently showed that ADAM10 interacts with one of six different regulatory tetraspanin membrane proteins, which we termed the TspanC8 subgroup, comprising Tspan5, Tspan10, Tspan14, Tspan15, Tspan17, and Tspan33. The TspanC8s are required for ADAM10 exit from the endoplasmic reticulum, and emerging evidence suggests that they dictate ADAM10 subcellular localization and substrate specificity. Therefore, we propose that ADAM10 should not be regarded as a single scissor, but as six different scissors with distinct substrate specificities, depending on the associated TspanC8. In this review, we collate recent transcriptomic data to present the TspanC8 repertoires of leukocytes, and we discuss the potential role of the six TspanC8/ADAM10 scissors in leukocyte development and function.
Collapse
Affiliation(s)
- Alexandra L Matthews
- School of Biosciences, College of Life and Environmental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Chek Ziu Koo
- School of Biosciences, College of Life and Environmental Sciences, University of Birmingham, Birmingham, United Kingdom.,Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham, Birmingham, United Kingdom
| | - Justyna Szyroka
- School of Biosciences, College of Life and Environmental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Neale Harrison
- School of Biosciences, College of Life and Environmental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Aditi Kanhere
- School of Biosciences, College of Life and Environmental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Michael G Tomlinson
- School of Biosciences, College of Life and Environmental Sciences, University of Birmingham, Birmingham, United Kingdom.,Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
217
|
Moharir A, Gay L, Appadurai D, Keener J, Babst M. Eisosomes are metabolically regulated storage compartments for APC-type nutrient transporters. Mol Biol Cell 2018; 29:2113-2127. [PMID: 29927345 PMCID: PMC6232963 DOI: 10.1091/mbc.e17-11-0691] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Eisosomes are lipid domains of the yeast plasma membrane that share similarities to caveolae of higher eukaryotes. Eisosomes harbor APC-type nutrient transporters for reasons that are poorly understood. Our analyses support the model that eisosomes function as storage compartments, keeping APC transporters in a stable, inactive state. By regulating eisosomes, yeast is able to balance the number of proton-driven APC transporters with the proton-pumping activity of Pma1, thereby maintaining the plasma membrane proton gradient. Environmental or metabolic changes that disrupt the proton gradient cause the rapid restructuring of eisosomes and results in the removal of the APC transporters from the cell surface. Furthermore, we show evidence that eisosomes require the presence of APC transporters, suggesting that regulating activity of nutrient transporters is a major function of eisosomes.
Collapse
Affiliation(s)
- Akshay Moharir
- Henry Eyring Center for Cell and Genome Science, University of Utah, Salt Lake City, UT 84112
| | - Lincoln Gay
- Henry Eyring Center for Cell and Genome Science, University of Utah, Salt Lake City, UT 84112
| | - Daniel Appadurai
- Henry Eyring Center for Cell and Genome Science, University of Utah, Salt Lake City, UT 84112
| | - James Keener
- Department of Mathematics, University of Utah, Salt Lake City, UT 84112
| | - Markus Babst
- Henry Eyring Center for Cell and Genome Science, University of Utah, Salt Lake City, UT 84112
| |
Collapse
|
218
|
Murru L, Vezzoli E, Longatti A, Ponzoni L, Falqui A, Folci A, Moretto E, Bianchi V, Braida D, Sala M, D'Adamo P, Bassani S, Francolini M, Passafaro M. Pharmacological Modulation of AMPAR Rescues Intellectual Disability-Like Phenotype in Tm4sf2-/y Mice. Cereb Cortex 2018; 27:5369-5384. [PMID: 28968657 PMCID: PMC5939231 DOI: 10.1093/cercor/bhx221] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Accepted: 08/02/2017] [Indexed: 01/02/2023] Open
Abstract
Intellectual disability affects 2–3% of the world's population and typically begins during childhood, causing impairments in social skills and cognitive abilities. Mutations in the TM4SF2 gene, which encodes the TSPAN7 protein, cause a severe form of intellectual disability, and currently, no therapy is able to ameliorate this cognitive impairment. We previously reported that, in cultured neurons, shRNA-mediated down-regulation of TSPAN7 affects AMPAR trafficking by enhancing PICK1–GluA2 interaction, thereby increasing the intracellular retention of AMPAR. Here, we found that loss of TSPAN7 function in mice causes alterations in hippocampal excitatory synapse structure and functionality as well as cognitive impairment. These changes occurred along with alterations in AMPAR expression levels. We also found that interfering with PICK1–GluA2 binding restored synaptic function in Tm4sf2−/y mice. Moreover, potentiation of AMPAR activity via the administration of the ampakine CX516 reverted the neurological phenotype observed in Tm4sf2−/y mice, suggesting that pharmacological modulation of AMPAR may represent a new approach for treating patients affected by TM4SF2 mutations and intellectual disability.
Collapse
Affiliation(s)
- Luca Murru
- CNR Institute of Neuroscience, 20129 Milano, Italy
| | - Elena Vezzoli
- Department of Medical Biotechnology and Translational Medicine, Università degli studi di Milano, Via Vanvitelli 32, 20129 Milano, Italy.,Department of Pharmacological and Biomolecular Sciences (DiSFeB), Università di Milano, Via Balzaretti 9, 20133 Milano, Italy.,Department of Biosciences and Centre for Stem Cell Research, University of Milan and Istituto Nazionale di Genetica Molecolare "Romeo ed Enrica Invernizzi" Milan, Italy
| | - Anna Longatti
- CNR Institute of Neuroscience, 20129 Milano, Italy.,Department of Pharmacological and Biomolecular Sciences (DiSFeB), Università di Milano, Via Balzaretti 9, 20133 Milano, Italy
| | - Luisa Ponzoni
- Department of Medical Biotechnology and Translational Medicine, Università degli studi di Milano, Via Vanvitelli 32, 20129 Milano, Italy.,Fondazione Umberto Veronesi, Piazza Velasca 5, 20122 Milan, Italy
| | - Andrea Falqui
- Biological and Environmental Sciences and Engineering Division, King Abdullah University for Science and Technology (KAUST), Thuwal 23955-6900, Kingdom of Saudi Arabia
| | | | | | - Veronica Bianchi
- Division of Neuroscience, IRCSS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Daniela Braida
- Department of Medical Biotechnology and Translational Medicine, Università degli studi di Milano, Via Vanvitelli 32, 20129 Milano,Italy
| | | | - Patrizia D'Adamo
- Division of Neuroscience, IRCSS San Raffaele Scientific Institute, 20132 Milan, Italy
| | | | - Maura Francolini
- Department of Medical Biotechnology and Translational Medicine, Università degli studi di Milano, Via Vanvitelli 32, 20129 Milano,Italy
| | | |
Collapse
|
219
|
Schaper F, van Spriel AB. Antitumor Immunity Is Controlled by Tetraspanin Proteins. Front Immunol 2018; 9:1185. [PMID: 29896201 PMCID: PMC5986925 DOI: 10.3389/fimmu.2018.01185] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Accepted: 05/14/2018] [Indexed: 12/27/2022] Open
Abstract
Antitumor immunity is shaped by the different types of immune cells that are present in the tumor microenvironment (TME). In particular, environmental signals (for instance, soluble factors or cell–cell contact) transmitted through the plasma membrane determine whether immune cells are activated or inhibited. Tetraspanin proteins are emerging as central building blocks of the plasma membrane by their capacity to cluster immune receptors, enzymes, and signaling molecules into the tetraspanin web. Whereas some tetraspanins (CD81, CD151, CD9) are widely and broadly expressed, others (CD53, CD37, Tssc6) have an expression pattern restricted to hematopoietic cells. Studies using genetic mouse models have identified important immunological functions of these tetraspanins on different leukocyte subsets, and as such, may be involved in the immune response against tumors. While multiple studies have been performed with regards to deciphering the function of tetraspanins on cancer cells, the effect of tetraspanins on immune cells in the antitumor response remains understudied. In this review, we will focus on tetraspanins expressed by immune cells and discuss their potential role in antitumor immunity. New insights in tetraspanin function in the TME and possible prognostic and therapeutic roles of tetraspanins will be discussed.
Collapse
Affiliation(s)
- Fleur Schaper
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Annemiek B van Spriel
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| |
Collapse
|
220
|
Florin L, Lang T. Tetraspanin Assemblies in Virus Infection. Front Immunol 2018; 9:1140. [PMID: 29887866 PMCID: PMC5981178 DOI: 10.3389/fimmu.2018.01140] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Accepted: 05/07/2018] [Indexed: 12/23/2022] Open
Abstract
Tetraspanins (Tspans) are a family of four-span transmembrane proteins, known as plasma membrane “master organizers.” They form Tspan-enriched microdomains (TEMs or TERMs) through lateral association with one another and other membrane proteins. If multiple microdomains associate with each other, larger platforms can form. For infection, viruses interact with multiple cell surface components, including receptors, activating proteases, and signaling molecules. It appears that Tspans, such as CD151, CD82, CD81, CD63, CD9, Tspan9, and Tspan7, coordinate these associations by concentrating the interacting partners into Tspan platforms. In addition to mediating viral attachment and entry, these platforms may also be involved in intracellular trafficking of internalized viruses and assist in defining virus assembly and exit sites. In conclusion, Tspans play a role in viral infection at different stages of the virus replication cycle. The present review highlights recently published data on this topic, with a focus on events at the plasma membrane. In light of these findings, we propose a model for how Tspan interactions may organize cofactors for viral infection into distinct molecular platforms.
Collapse
Affiliation(s)
- Luise Florin
- Department of Medical Microbiology and Hygiene, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Thorsten Lang
- Department of Membrane Biochemistry, Life & Medical Sciences Institute, University of Bonn, Bonn, Germany
| |
Collapse
|
221
|
Saiz ML, Rocha-Perugini V, Sánchez-Madrid F. Tetraspanins as Organizers of Antigen-Presenting Cell Function. Front Immunol 2018; 9:1074. [PMID: 29875769 PMCID: PMC5974036 DOI: 10.3389/fimmu.2018.01074] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Accepted: 04/30/2018] [Indexed: 12/19/2022] Open
Abstract
Professional antigen-presenting cells (APCs) include dendritic cells, monocytes, and B cells. APCs internalize and process antigens, producing immunogenic peptides that enable antigen presentation to T lymphocytes, which provide the signals that trigger T-cell activation, proliferation, and differentiation, and lead to adaptive immune responses. After detection of microbial antigens through pattern recognition receptors (PRRs), APCs migrate to secondary lymphoid organs where antigen presentation to T lymphocytes takes place. Tetraspanins are membrane proteins that organize specialized membrane platforms, called tetraspanin-enriched microdomains, which integrate membrane receptors, like PRR and major histocompatibility complex class II (MHC-II), adhesion proteins, and signaling molecules. Importantly, through the modulation of the function of their associated membrane partners, tetraspanins regulate different steps of the immune response. Several tetraspanins can positively or negatively regulate the activation threshold of immune receptors. They also play a role during migration of APCs by controlling the surface levels and spatial arrangement of adhesion molecules and their subsequent intracellular signaling. Finally, tetraspanins participate in antigen processing and are important for priming of naïve T cells through the control of T-cell co-stimulation and MHC-II-dependent antigen presentation. In this review, we discuss the role of tetraspanins in APC biology and their involvement in effective immune responses.
Collapse
Affiliation(s)
- Maria Laura Saiz
- Servicio de Inmunología, Hospital de la Princesa, Instituto de Investigación Sanitaria La Princesa, Madrid, Spain.,Vascular Pathophysiology Research Area, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - Vera Rocha-Perugini
- Servicio de Inmunología, Hospital de la Princesa, Instituto de Investigación Sanitaria La Princesa, Madrid, Spain.,Vascular Pathophysiology Research Area, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - Francisco Sánchez-Madrid
- Servicio de Inmunología, Hospital de la Princesa, Instituto de Investigación Sanitaria La Princesa, Madrid, Spain.,Vascular Pathophysiology Research Area, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain.,CIBER Cardiovascular, Madrid, Spain
| |
Collapse
|
222
|
Whole Exome Sequencing Identifies New Host Genomic Susceptibility Factors in Empyema Caused by Streptococcus pneumoniae in Children: A Pilot Study. Genes (Basel) 2018; 9:genes9050240. [PMID: 29751582 PMCID: PMC5977180 DOI: 10.3390/genes9050240] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 04/25/2018] [Accepted: 04/26/2018] [Indexed: 11/17/2022] Open
Abstract
Pneumonia is the leading cause of death amongst infectious diseases. Streptococcus pneumoniae is responsible for about 25% of pneumonia cases worldwide, and it is a major cause of childhood mortality. We carried out a whole exome sequencing (WES) study in eight patients with complicated cases of pneumococcal pneumonia (empyema). An initial assessment of statistical association of WES variation with pneumonia was carried out using data from the 1000 Genomes Project (1000G) for the Iberian Peninsula (IBS) as reference controls. Pseudo-replication statistical analyses were carried out using different European control groups. Association tests pointed to single nucleotide polymorphism (SNP) rs201967957 (gene MEIS1; chromosome 2; p-valueIBS = 3.71 × 10-13) and rs576099063 (gene TSPAN15; chromosome 10; p-valueIBS = 2.36 × 10-8) as the best candidate variants associated to pneumococcal pneumonia. A burden gene test of pathogenicity signaled four genes, namely, OR9G9, MUC6, MUC3A and APOB, which carry significantly increased pathogenic variation when compared to controls. By analyzing various transcriptomic data repositories, we found strong supportive evidence for the role of MEIS1, TSPAN15 and APOBR (encoding the receptor of the APOB protein) in pneumonia in mouse and human models. Furthermore, the association of the olfactory receptor gene OR9G9 has recently been related to some viral infectious diseases, while the role of mucin genes (MUC6 and MUC3A), encoding mucin glycoproteins, are well-known factors related to chronic obstructive airway disease. WES emerges as a promising technique to disentangle the genetic basis of host genome susceptibility to infectious respiratory diseases.
Collapse
|
223
|
Katikaneni DS, Jin L. B cell MHC class II signaling: A story of life and death. Hum Immunol 2018; 80:37-43. [PMID: 29715484 DOI: 10.1016/j.humimm.2018.04.013] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 04/08/2018] [Accepted: 04/25/2018] [Indexed: 01/17/2023]
Abstract
MHC class II regulates B cell activation, proliferation, and differentiation during cognate B cell-T cell interaction. This is, in part, due to the MHC class II signaling in B cells. Activation of MHC Class II in human B cells or "primed" murine B cells leads to tyrosine phosphorylation, calcium mobilization, AKT, ERK, JNK activation. In addition, crosslinking MHC class II with monoclonal Abs kill malignant human B cells. Several humanized anti-HLA-DR/MHC class II monoclonal Abs entered clinical trials for lymphoma/leukemia and MHC class II-expressing melanomas. Mechanistically, MHC class II is associated with a wealth of transmembrane proteins including the B cell-specific signaling proteins CD79a/b, CD19 and a group of four-transmembrane proteins including tetraspanins and the apoptotic protein MPYS/STING. Furthermore, MHC class II signals are compartmentalized in the tetraspanin-enriched microdomains. In this review, we discuss our current understanding of MHC class II signaling in B cells focusing on its physiological significance and the therapeutic potential.
Collapse
Affiliation(s)
- Divya Sai Katikaneni
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Florida, Gainesville, FL 32610, United States
| | - Lei Jin
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Florida, Gainesville, FL 32610, United States.
| |
Collapse
|
224
|
Helenius A. Virus Entry: Looking Back and Moving Forward. J Mol Biol 2018; 430:1853-1862. [PMID: 29709571 PMCID: PMC7094621 DOI: 10.1016/j.jmb.2018.03.034] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 03/15/2018] [Accepted: 03/16/2018] [Indexed: 12/29/2022]
Abstract
Research over a period of more than half a century has provided a reasonably accurate picture of mechanisms involved in animal virus entry into their host cells. Successive steps in entry include binding to receptors, endocytosis, passage through one or more membranes, targeting to specific sites within the cell, and uncoating of the genome. For some viruses, the molecular interactions are known in great detail. However, as more viruses are analyzed, and as the focus shifts from tissue culture to in vivo experiments, it is evident that viruses display considerable redundancy and flexibility in receptor usage, endocytic mechanism, location of penetration, and uncoating mechanism. For many viruses, the picture is still elusive because the interactions that they engage in rely on sophisticated adaptation to complex cellular functions and defense mechanisms. Studies using a broad combination of technologies have provided detailed information on the entry and uncoating of many animal viruses. Not only the identity of cell surface receptors but their distribution in plasma membrane and in microdomains defines cell tropism and infection efficiency. The majority of viruses enter by endocytic mechanisms and penetrate into the cytosol intracellularly from a variety of different organelles. The picture is often elusive because many viruses display redundancy in receptor choice and entry strategy.
Collapse
Affiliation(s)
- Ari Helenius
- ETH Zurich, Institute of Biochemistry, Otto-Stern-Weg 3, Zurich 8093, Switzerland.
| |
Collapse
|
225
|
TSPAN12 Is a Norrin Co-receptor that Amplifies Frizzled4 Ligand Selectivity and Signaling. Cell Rep 2018; 19:2809-2822. [PMID: 28658627 DOI: 10.1016/j.celrep.2017.06.004] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Revised: 03/29/2017] [Accepted: 05/27/2017] [Indexed: 11/23/2022] Open
Abstract
Accessory proteins in Frizzled (FZD) receptor complexes are thought to determine ligand selectivity and signaling amplitude. Genetic evidence indicates that specific combinations of accessory proteins and ligands mediate vascular β-catenin signaling in different CNS structures. In the retina, the tetraspanin TSPAN12 and the ligand norrin (NDP) mediate angiogenesis, and both genes are linked to familial exudative vitreoretinopathy (FEVR), yet the molecular function of TSPAN12 remains poorly understood. Here, we report that TSPAN12 is an essential component of the NDP receptor complex and interacts with FZD4 and NDP via its extracellular loops, consistent with an action as co-receptor that enhances FZD4 ligand selectivity for NDP. FEVR-linked mutations in TSPAN12 prevent the incorporation of TSPAN12 into the NDP receptor complex. In vitro and in Xenopus embryos, TSPAN12 alleviates defects of FZD4 M105V, a mutation that destabilizes the NDP/FZD4 interaction. This study sheds light on the poorly understood function of accessory proteins in FZD signaling.
Collapse
|
226
|
Frolikova M, Manaskova-Postlerova P, Cerny J, Jankovicova J, Simonik O, Pohlova A, Secova P, Antalikova J, Dvorakova-Hortova K. CD9 and CD81 Interactions and Their Structural Modelling in Sperm Prior to Fertilization. Int J Mol Sci 2018; 19:ijms19041236. [PMID: 29671763 PMCID: PMC5979608 DOI: 10.3390/ijms19041236] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 04/10/2018] [Accepted: 04/13/2018] [Indexed: 11/16/2022] Open
Abstract
Proteins CD9 and CD81 are members of the tetraspanin superfamily and were detected in mammalian sperm, where they are suspected to form an active tetraspanin web and to participate in sperm–egg membrane fusion. The importance of these two proteins during the early stages of fertilization is supported by the complete sterility of CD9/CD81 double null female mice. In this study, the putative mechanism of CD9/CD81 involvement in tetraspanin web formation in sperm and its activity prior to fertilization was addressed. Confocal microscopy and colocalization assay was used to determine a mutual CD9/CD81 localization visualised in detail by super-resolution microscopy, and their interaction was address by co-immunoprecipitation. The species-specific traits in CD9 and CD81 distribution during sperm maturation were compared between mice and humans. A mutual position of CD9/CD81 is shown in human spermatozoa in the acrosomal cap, however in mice, CD9 and CD81 occupy a distinct area. During the acrosome reaction in human sperm, only CD9 is relocated, compared to the relocation of both proteins in mice. The structural modelling of CD9 and CD81 homologous and possibly heterologous network formation was used to propose their lateral Cis as well as Trans interactions within the sperm membrane and during sperm–egg membrane fusion.
Collapse
Affiliation(s)
- Michaela Frolikova
- Group of Reproductive Biology, Institute of Biotechnology, Czech Academy of Sciences, v.v.i., BIOCEV, Prumyslova 595, 252 50 Vestec, Czech Republic.
| | - Pavla Manaskova-Postlerova
- Group of Reproductive Biology, Institute of Biotechnology, Czech Academy of Sciences, v.v.i., BIOCEV, Prumyslova 595, 252 50 Vestec, Czech Republic.
- Department of Veterinary Sciences, Faculty of Agrobiology, Food and Natural Resources, University of Life Sciences Prague, Kamycka 129, 165 00 Prague, Czech Republic.
| | - Jiri Cerny
- Laboratory of Structural Bioinformatics of Proteins, Institute of Biotechnology Czech Academy of Sciences, v.v.i., BIOCEV, Prumyslova 595, 252 50 Vestec, Czech Republic.
| | - Jana Jankovicova
- Laboratory of Reproductive Physiology, Institute of Animal Biochemistry and Genetics Centre of Biosciences Slovak Academy of Sciences, Dubravska Cesta 9, 845 05 Bratislava, Slovakia.
| | - Ondrej Simonik
- Group of Reproductive Biology, Institute of Biotechnology, Czech Academy of Sciences, v.v.i., BIOCEV, Prumyslova 595, 252 50 Vestec, Czech Republic.
- Department of Veterinary Sciences, Faculty of Agrobiology, Food and Natural Resources, University of Life Sciences Prague, Kamycka 129, 165 00 Prague, Czech Republic.
| | - Alzbeta Pohlova
- Group of Reproductive Biology, Institute of Biotechnology, Czech Academy of Sciences, v.v.i., BIOCEV, Prumyslova 595, 252 50 Vestec, Czech Republic.
- Department of Biochemistry, Faculty of Science, Charles University, Hlavova 2030/8, 128 43 Prague, Czech Republic.
| | - Petra Secova
- Laboratory of Reproductive Physiology, Institute of Animal Biochemistry and Genetics Centre of Biosciences Slovak Academy of Sciences, Dubravska Cesta 9, 845 05 Bratislava, Slovakia.
| | - Jana Antalikova
- Laboratory of Reproductive Physiology, Institute of Animal Biochemistry and Genetics Centre of Biosciences Slovak Academy of Sciences, Dubravska Cesta 9, 845 05 Bratislava, Slovakia.
| | - Katerina Dvorakova-Hortova
- Group of Reproductive Biology, Institute of Biotechnology, Czech Academy of Sciences, v.v.i., BIOCEV, Prumyslova 595, 252 50 Vestec, Czech Republic.
- Department of Zoology, Faculty of Science, Charles University, Vinicna 7, 128 44 Prague, Czech Republic.
| |
Collapse
|
227
|
Steinbacher T, Kummer D, Ebnet K. Junctional adhesion molecule-A: functional diversity through molecular promiscuity. Cell Mol Life Sci 2018; 75:1393-1409. [PMID: 29238845 PMCID: PMC11105642 DOI: 10.1007/s00018-017-2729-0] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 12/04/2017] [Accepted: 12/11/2017] [Indexed: 12/27/2022]
Abstract
Cell adhesion molecules (CAMs) of the immunoglobulin superfamily (IgSF) regulate important processes such as cell proliferation, differentiation and morphogenesis. This activity is primarily due to their ability to initiate intracellular signaling cascades at cell-cell contact sites. Junctional adhesion molecule-A (JAM-A) is an IgSF-CAM with a short cytoplasmic tail that has no catalytic activity. Nevertheless, JAM-A is involved in a variety of biological processes. The functional diversity of JAM-A resides to a large part in a C-terminal PDZ domain binding motif which directly interacts with nine different PDZ domain-containing proteins. The molecular promiscuity of its PDZ domain motif allows JAM-A to recruit protein scaffolds to specific sites of cell-cell adhesion and to assemble signaling complexes at those sites. Here, we review the molecular characteristics of JAM-A, including its dimerization, its interaction with scaffolding proteins, and the phosphorylation of its cytoplasmic domain, and we describe how these characteristics translate into diverse biological activities.
Collapse
Affiliation(s)
- Tim Steinbacher
- Institute-Associated Research Group: Cell Adhesion and Cell Polarity, Institute of Medical Biochemistry, ZMBE, University of Münster, Von-Esmarch-Str. 56, 48149, Münster, Germany
- Cells-In-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, Münster, Germany
| | - Daniel Kummer
- Institute-Associated Research Group: Cell Adhesion and Cell Polarity, Institute of Medical Biochemistry, ZMBE, University of Münster, Von-Esmarch-Str. 56, 48149, Münster, Germany
- Interdisciplinary Clinical Research Center (IZKF), University of Münster, Münster, Germany
| | - Klaus Ebnet
- Institute-Associated Research Group: Cell Adhesion and Cell Polarity, Institute of Medical Biochemistry, ZMBE, University of Münster, Von-Esmarch-Str. 56, 48149, Münster, Germany.
- Cells-In-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, Münster, Germany.
- Interdisciplinary Clinical Research Center (IZKF), University of Münster, Münster, Germany.
| |
Collapse
|
228
|
Jin Y, Takeda Y, Kondo Y, Tripathi LP, Kang S, Takeshita H, Kuhara H, Maeda Y, Higashiguchi M, Miyake K, Morimura O, Koba T, Hayama Y, Koyama S, Nakanishi K, Iwasaki T, Tetsumoto S, Tsujino K, Kuroyama M, Iwahori K, Hirata H, Takimoto T, Suzuki M, Nagatomo I, Sugimoto K, Fujii Y, Kida H, Mizuguchi K, Ito M, Kijima T, Rakugi H, Mekada E, Tachibana I, Kumanogoh A. Double deletion of tetraspanins CD9 and CD81 in mice leads to a syndrome resembling accelerated aging. Sci Rep 2018; 8:5145. [PMID: 29572511 PMCID: PMC5865149 DOI: 10.1038/s41598-018-23338-x] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Accepted: 03/09/2018] [Indexed: 01/29/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) has been recently characterized as a disease of accelerated lung aging, but the mechanism remains unclear. Tetraspanins have emerged as key players in malignancy and inflammatory diseases. Here, we found that CD9/CD81 double knockout (DKO) mice with a COPD-like phenotype progressively developed a syndrome resembling human aging, including cataracts, hair loss, and atrophy of various organs, including thymus, muscle, and testis, resulting in shorter survival than wild-type (WT) mice. Consistent with this, DNA microarray analysis of DKO mouse lungs revealed differential expression of genes involved in cell death, inflammation, and the sirtuin-1 (SIRT1) pathway. Accordingly, expression of SIRT1 was reduced in DKO mouse lungs. Importantly, siRNA knockdown of CD9 and CD81 in lung epithelial cells additively decreased SIRT1 and Foxo3a expression, but reciprocally upregulated the expression of p21 and p53, leading to reduced cell proliferation and elevated apoptosis. Furthermore, deletion of these tetraspanins increased the expression of pro-inflammatory genes and IL-8. Hence, CD9 and CD81 might coordinately prevent senescence and inflammation, partly by maintaining SIRT1 expression. Altogether, CD9/CD81 DKO mice represent a novel model for both COPD and accelerated senescence.
Collapse
Affiliation(s)
- Yingji Jin
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Yoshito Takeda
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan.
| | | | - Lokesh P Tripathi
- National Institute of Biomedical Innovation, Health and Nutrition, Ibaraki, Osaka, Japan
| | - Sujin Kang
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Hikari Takeshita
- Department of Geriatric Medicine &, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Hanako Kuhara
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Yohei Maeda
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Masayoshi Higashiguchi
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Kotaro Miyake
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Osamu Morimura
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Taro Koba
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Yoshitomo Hayama
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Shohei Koyama
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Kaori Nakanishi
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Takeo Iwasaki
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Satoshi Tetsumoto
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Kazuyuki Tsujino
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Muneyoshi Kuroyama
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Kota Iwahori
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Haruhiko Hirata
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Takayuki Takimoto
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Mayumi Suzuki
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Izumi Nagatomo
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Ken Sugimoto
- Department of Geriatric Medicine &, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Yuta Fujii
- Sumitomo Dainippon Pharma Co., Ltd, Osaka, Japan
| | - Hiroshi Kida
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Kenji Mizuguchi
- National Institute of Biomedical Innovation, Health and Nutrition, Ibaraki, Osaka, Japan
| | - Mari Ito
- National Institute of Biomedical Innovation, Health and Nutrition, Ibaraki, Osaka, Japan
| | - Takashi Kijima
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Hiromi Rakugi
- Department of Geriatric Medicine &, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Eisuke Mekada
- Department of Cell Biology, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Isao Tachibana
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Atsushi Kumanogoh
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| |
Collapse
|
229
|
Wendt GR, Collins JNR, Pei J, Pearson MS, Bennett HM, Loukas A, Berriman M, Grishin NV, Collins JJ. Flatworm-specific transcriptional regulators promote the specification of tegumental progenitors in Schistosoma mansoni. eLife 2018; 7:e33221. [PMID: 29557781 PMCID: PMC5927768 DOI: 10.7554/elife.33221] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 03/19/2018] [Indexed: 01/13/2023] Open
Abstract
Schistosomes infect more than 200 million people. These parasitic flatworms rely on a syncytial outer coat called the tegument to survive within the vasculature of their host. Although the tegument is pivotal for their survival, little is known about maintenance of this tissue during the decades schistosomes survive in the bloodstream. Here, we demonstrate that the tegument relies on stem cells (neoblasts) to specify fusogenic progenitors that replace tegumental cells lost to turnover. Molecular characterization of neoblasts and tegumental progenitors led to the discovery of two flatworm-specific zinc finger proteins that are essential for tegumental cell specification. These proteins are homologous to a protein essential for neoblast-driven epidermal maintenance in free-living flatworms. Therefore, we speculate that related parasites (i.e., tapeworms and flukes) employ similar strategies to control tegumental maintenance. Since parasitic flatworms infect every vertebrate species, understanding neoblast-driven tegumental maintenance could identify broad-spectrum therapeutics to fight diseases caused by these parasites.
Collapse
Affiliation(s)
- George R Wendt
- Department of PharmacologyUT Southwestern Medical CenterDallasTexas
| | - Julie NR Collins
- Department of PharmacologyUT Southwestern Medical CenterDallasTexas
| | - Jimin Pei
- Department of BiophysicsUT Southwestern Medical CenterDallasTexas
- Howard Hughes Medical InstituteUT Southwestern Medical CenterDallasTexas
| | - Mark S Pearson
- Center for Biodiscovery and Molecular Development of TherapeuticsAustralian Institute of Tropical Health and Medicine, James Cook UniversityCairnsAustralia
| | - Hayley M Bennett
- Wellcome Sanger InstituteWellcome Genome CampusHinxtonUnited Kingdom
| | - Alex Loukas
- Center for Biodiscovery and Molecular Development of TherapeuticsAustralian Institute of Tropical Health and Medicine, James Cook UniversityCairnsAustralia
| | - Matthew Berriman
- Wellcome Sanger InstituteWellcome Genome CampusHinxtonUnited Kingdom
| | - Nick V Grishin
- Department of BiophysicsUT Southwestern Medical CenterDallasTexas
- Howard Hughes Medical InstituteUT Southwestern Medical CenterDallasTexas
| | - James J Collins
- Department of PharmacologyUT Southwestern Medical CenterDallasTexas
| |
Collapse
|
230
|
Zhu Y, Ailane N, Sala-Valdés M, Haghighi-Rad F, Billard M, Nguyen V, Saffroy R, Lemoine A, Rubinstein E, Boucheix C, Greco C. Multi-factorial modulation of colorectal carcinoma cells motility - partial coordination by the tetraspanin Co-029/tspan8. Oncotarget 2018; 8:27454-27470. [PMID: 28418857 PMCID: PMC5432348 DOI: 10.18632/oncotarget.16247] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 02/20/2017] [Indexed: 12/30/2022] Open
Abstract
Colorectal carcinoma cells Isreco1 display an ability to migrate controlled by a complex set of signals issued from the membrane. By comparing cells infected by mycoplasmas and mycoplasmas free cells, we have established that basal 2D migration is dependent on a double signal mediated by the collagen receptors integrins alpha1/2 and the Toll-Like receptor TLR2. The signal issued from mycoplasmas can be replaced by a TLR2 ligand and the functional effect is neutralized by silencing of MyD88. Following previous observation that downregulation of E-cadherin/p120 catenin increases cell motility, we now report that EGFR or CD44 inhibition have a similar effect on cell motility that is restricted to tetraspanin Co-029/tspan8 transduced IsrecoI cells (Is1-Co029). The modulation of cell migration linked to EGFR or CD44 can be neutralized by antagonizing Co-029 with the mAb Ts29.1 or by RNA interference. Altogether these data point to a crucial role of Co-029 in the modulation of colon cancer cell motility which could be related to the protumoral effect reported for this tetraspanin. Among surface molecules able to mediate Co-029 function, E-cadherin, EGFR and CD44 appear as likely candidates.
Collapse
Affiliation(s)
- Yingying Zhu
- Inserm, UMR-S 935, SFR André Lwoff, Villejuif, France.,Université Paris-Sud 11, Paris, France.,Department of Oncology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Naouel Ailane
- Inserm, UMR-S 935, SFR André Lwoff, Villejuif, France.,Université Paris-Sud 11, Paris, France
| | - Monica Sala-Valdés
- Inserm, UMR-S 935, SFR André Lwoff, Villejuif, France.,Université Paris-Sud 11, Paris, France
| | - Farhad Haghighi-Rad
- Inserm, UMR-S 935, SFR André Lwoff, Villejuif, France.,Université Paris-Sud 11, Paris, France
| | - Martine Billard
- Inserm, UMR-S 935, SFR André Lwoff, Villejuif, France.,Université Paris-Sud 11, Paris, France
| | - Viet Nguyen
- Université Paris-Sud 11, Paris, France.,Inserm, UMS-33, SFR André Lwoff, Villejuif, France
| | - Raphael Saffroy
- Université Paris-Sud 11, Paris, France.,Inserm UMR-S 1193, SFR André Lwoff, Villejuif, France.,AP HP, Hôpital Paul-Brousse, Department of Biochemistry, Villejuif, France
| | - Antoinette Lemoine
- Université Paris-Sud 11, Paris, France.,Inserm UMR-S 1193, SFR André Lwoff, Villejuif, France.,AP HP, Hôpital Paul-Brousse, Department of Biochemistry, Villejuif, France
| | - Eric Rubinstein
- Inserm, UMR-S 935, SFR André Lwoff, Villejuif, France.,Université Paris-Sud 11, Paris, France
| | - Claude Boucheix
- Inserm, UMR-S 935, SFR André Lwoff, Villejuif, France.,Université Paris-Sud 11, Paris, France
| | - Céline Greco
- Inserm, UMR-S 935, SFR André Lwoff, Villejuif, France.,Université Paris-Sud 11, Paris, France.,AP HP, Hôpital Necker, Department of Pain and Palliative Medicine, Paris, France
| |
Collapse
|
231
|
Bucher F, Friedlander M, Yea K. Antibody targeting TSPAN12/β-catenin signaling in vasoproliferative retinopathy. Oncotarget 2018; 9:12538-12539. [PMID: 29560082 PMCID: PMC5849146 DOI: 10.18632/oncotarget.23401] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Accepted: 12/16/2017] [Indexed: 11/25/2022] Open
Affiliation(s)
- Felicitas Bucher
- Department of New Biology, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Republic of Korea
| | - Martin Friedlander
- Department of New Biology, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Republic of Korea
| | - Kyungmoo Yea
- Department of New Biology, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Republic of Korea
| |
Collapse
|
232
|
Liang D. A Salutary Role of Reactive Oxygen Species in Intercellular Tunnel-Mediated Communication. Front Cell Dev Biol 2018; 6:2. [PMID: 29503816 PMCID: PMC5821100 DOI: 10.3389/fcell.2018.00002] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 01/18/2018] [Indexed: 12/17/2022] Open
Abstract
The reactive oxygen species, generally labeled toxic due to high reactivity without target specificity, are gradually uncovered as signaling molecules involved in a myriad of biological processes. But one important feature of ROS roles in macromolecule movement has not caught attention until recent studies with technique advance and design elegance have shed lights on ROS signaling for intercellular and interorganelle communication. This review begins with the discussions of genetic and chemical studies on the regulation of symplastic dye movement through intercellular tunnels in plants (plasmodesmata), and focuses on the ROS regulatory mechanisms concerning macromolecule movement including small RNA-mediated gene silencing movement and protein shuttling between cells. Given the premise that intercellular tunnels (bridges) in mammalian cells are the key physical structures to sustain intercellular communication, movement of macromolecules and signals is efficiently facilitated by ROS-induced membrane protrusions formation, which is analogously applied to the interorganelle communication in plant cells. Although ROS regulatory differences between plant and mammalian cells exist, the basis for ROS-triggered conduit formation underlies a unifying conservative theme in multicellular organisms. These mechanisms may represent the evolutionary advances that have enabled multicellularity to gain the ability to generate and utilize ROS to govern material exchanges between individual cells in oxygenated environment.
Collapse
Affiliation(s)
- Dacheng Liang
- Hubei Collaborative Innovation Center for Grain Industry, School of Agriculture, Yangtze University, Jingzhou, China.,Engineering Research Center of Ecology and Agricultural Use of Wetland, Ministry of Education, Yangtze University, Jingzhou, China
| |
Collapse
|
233
|
Zhao K, Erb U, Hackert T, Zöller M, Yue S. Distorted leukocyte migration, angiogenesis, wound repair and metastasis in Tspan8 and Tspan8/CD151 double knockout mice indicate complementary activities of Tspan8 and CD51. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2018; 1865:379-391. [DOI: 10.1016/j.bbamcr.2017.11.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 10/21/2017] [Accepted: 11/10/2017] [Indexed: 02/07/2023]
|
234
|
Alpha-Secretase ADAM10 Regulation: Insights into Alzheimer's Disease Treatment. Pharmaceuticals (Basel) 2018; 11:ph11010012. [PMID: 29382156 PMCID: PMC5874708 DOI: 10.3390/ph11010012] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Revised: 01/23/2018] [Accepted: 01/23/2018] [Indexed: 02/07/2023] Open
Abstract
ADAM (a disintegrin and metalloproteinase) is a family of widely expressed, transmembrane and secreted proteins of approximately 750 amino acids in length with functions in cell adhesion and proteolytic processing of the ectodomains of diverse cell-surface receptors and signaling molecules. ADAM10 is the main α-secretase that cleaves APP (amyloid precursor protein) in the non-amyloidogenic pathway inhibiting the formation of β-amyloid peptide, whose accumulation and aggregation leads to neuronal degeneration in Alzheimer’s disease (AD). ADAM10 is a membrane-anchored metalloprotease that sheds, besides APP, the ectodomain of a large variety of cell-surface proteins including cytokines, adhesion molecules and notch. APP cleavage by ADAM10 results in the production of an APP-derived fragment, sAPPα, which is neuroprotective. As increased ADAM10 activity protects the brain from β-amyloid deposition in AD, this strategy has been proved to be effective in treating neurodegenerative diseases, including AD. Here, we describe the physiological mechanisms regulating ADAM10 expression at different levels, aiming to propose strategies for AD treatment. We report in this review on the physiological regulation of ADAM10 at the transcriptional level, by epigenetic factors, miRNAs and/or translational and post-translational levels. In addition, we describe the conditions that can change ADAM10 expression in vitro and in vivo, and discuss how this knowledge may help in AD treatment. Regulation of ADAM10 is achieved by multiple mechanisms that include transcriptional, translational and post-translational strategies, which we will summarize in this review.
Collapse
|
235
|
Oh J, Perry JSA, Pua H, Irgens-Möller N, Ishido S, Hsieh CS, Shin JS. MARCH1 protects the lipid raft and tetraspanin web from MHCII proteotoxicity in dendritic cells. J Cell Biol 2018; 217:1395-1410. [PMID: 29371232 PMCID: PMC5881489 DOI: 10.1083/jcb.201611141] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 10/25/2017] [Accepted: 01/08/2018] [Indexed: 12/12/2022] Open
Abstract
Dendritic cells (DCs) produce major histocompatibility complex II (MHCII) in large amounts to function as professional antigen presenting cells. Paradoxically, DCs also ubiquitinate and degrade MHCII in a constitutive manner. Mice deficient in the MHCII-ubiquitinating enzyme membrane-anchored RING-CH1, or the ubiquitin-acceptor lysine of MHCII, exhibit a substantial reduction in the number of regulatory T (Treg) cells, but the underlying mechanism was unclear. Here we report that ubiquitin-dependent MHCII turnover is critical to maintain homeostasis of lipid rafts and the tetraspanin web in DCs. Lack of MHCII ubiquitination results in the accumulation of excessive quantities of MHCII in the plasma membrane, and the resulting disruption to lipid rafts and the tetraspanin web leads to significant impairment in the ability of DCs to engage and activate thymocytes for Treg cell differentiation. Thus, ubiquitin-dependent MHCII turnover represents a novel quality-control mechanism by which DCs maintain homeostasis of membrane domains that support DC's Treg cell-selecting function.
Collapse
Affiliation(s)
- Jaehak Oh
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA.,Sandler Asthma Basic Research Center, University of California, San Francisco, San Francisco, CA
| | - Justin S A Perry
- Department of Pathology, University of California, San Francisco, San Francisco, CA
| | - Heather Pua
- Sandler Asthma Basic Research Center, University of California, San Francisco, San Francisco, CA.,Department of Pathology, University of California, San Francisco, San Francisco, CA
| | - Nicole Irgens-Möller
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA.,Sandler Asthma Basic Research Center, University of California, San Francisco, San Francisco, CA
| | - Satoshi Ishido
- Department of Microbiology, Hyogo College of Medicine 1-1, Mukogawa-cho, Nishinomiya, Japan
| | - Chyi-Song Hsieh
- Department of Internal Medicine, Division of Rheumatology, Washington University School of Medicine, St. Louis, MO
| | - Jeoung-Sook Shin
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA .,Sandler Asthma Basic Research Center, University of California, San Francisco, San Francisco, CA
| |
Collapse
|
236
|
He P, Wang S, Zhang X, Gao Y, Niu W, Dong N, Shi X, Geng Y, Ma Q, Li M, Jiang B, Li JL. Tspan5 is an independent favourable prognostic factor and suppresses tumour growth in gastric cancer. Oncotarget 2018; 7:40160-40173. [PMID: 27223087 PMCID: PMC5130000 DOI: 10.18632/oncotarget.9514] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2016] [Accepted: 04/26/2016] [Indexed: 12/16/2022] Open
Abstract
Tetraspanins are believed to interact with specific partner proteins forming tetraspanin-enriched microdomains and regulate some aspects of partner protein functions. However, the role of Tspan5 during pathological processes, particularly in cancer biology, remains unknown. Here we report that Tspan5 is significantly downregulated in gastric cancer (GC) and closely associated with clinicopathological features including tumour size and TNM stage. The expression of Tspan5 is inversely correlated with patient overall survival and is an independent prognostic factor in GC. Upregulation of Tspan5 in tumour cells results in inhibition of cell proliferation and colony formation in vitro and suppression of xenograft growth of GC by reducing tumour cell proliferation in vivo. Thus, Tspan5 functions as a tumour suppressor in stomach to control the tumour growth. Mechanistically, Tspan5 inhibits the cell cycle transition from G1-S phase by increasing the expression of p27 and p15 and decreasing the expression of cyclin D1, CDK4, pRB and E2F1. The correlation of Tspan5 expression with the expression of p27, p15, cyclin D1, CDK4, pRB and E2F1 in vivo are also revealed in xenografted tumours. Reconstitution of either cyclin D1 or CDK4 in Tspan5-overexpressing GC cells rescues the inhibitory phenotype produced by Tspan5, suggesting that cyclin D1/CDK4 play a dominant role in mediating the suppression of tumour growth by Tspan5 in GC. Our results suggest that Tspan5 may serve as a prognostic biomarker for predicting outcome of GC patients and provide new insights into the pathogenesis of GC and rational for the development of clinical intervention strategies against GC.
Collapse
Affiliation(s)
- Peirong He
- School of Biotechnology, Southern Medical University, Guangzhou 510515, China.,Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Suihai Wang
- School of Biotechnology, Southern Medical University, Guangzhou 510515, China
| | - Xuefeng Zhang
- School of Biotechnology, Southern Medical University, Guangzhou 510515, China
| | - Yanjun Gao
- School of Biotechnology, Southern Medical University, Guangzhou 510515, China
| | - Wenbo Niu
- School of Biotechnology, Southern Medical University, Guangzhou 510515, China
| | - Ningning Dong
- School of Biotechnology, Southern Medical University, Guangzhou 510515, China
| | - Xiangyi Shi
- School of Biotechnology, Southern Medical University, Guangzhou 510515, China
| | - Yan Geng
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Qiang Ma
- School of Biotechnology, Southern Medical University, Guangzhou 510515, China
| | - Ming Li
- School of Biotechnology, Southern Medical University, Guangzhou 510515, China
| | - Bo Jiang
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Ji-Liang Li
- School of Biotechnology, Southern Medical University, Guangzhou 510515, China.,Institute of Translational and Stratified Medicine, Plymouth University Peninsula Schools of Medicine and Dentistry, Plymouth PL6 8BU, U.K
| |
Collapse
|
237
|
van Niel G, D'Angelo G, Raposo G. Shedding light on the cell biology of extracellular vesicles. Nat Rev Mol Cell Biol 2018; 19:213-228. [PMID: 29339798 DOI: 10.1038/nrm.2017.125] [Citation(s) in RCA: 5479] [Impact Index Per Article: 782.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Extracellular vesicles are a heterogeneous group of cell-derived membranous structures comprising exosomes and microvesicles, which originate from the endosomal system or which are shed from the plasma membrane, respectively. They are present in biological fluids and are involved in multiple physiological and pathological processes. Extracellular vesicles are now considered as an additional mechanism for intercellular communication, allowing cells to exchange proteins, lipids and genetic material. Knowledge of the cellular processes that govern extracellular vesicle biology is essential to shed light on the physiological and pathological functions of these vesicles as well as on clinical applications involving their use and/or analysis. However, in this expanding field, much remains unknown regarding the origin, biogenesis, secretion, targeting and fate of these vesicles.
Collapse
Affiliation(s)
- Guillaume van Niel
- Center of Psychiatry and Neurosciences, INSERM U895, Paris 75014, France
| | - Gisela D'Angelo
- Institut Curie, Paris Sciences et Lettres Research University, Centre National de la Recherche Scientifique UMR144, Structure and Membrane Compartments, Paris F-75005, France
| | - Graça Raposo
- Institut Curie, Paris Sciences et Lettres Research University, Centre National de la Recherche Scientifique UMR144, Structure and Membrane Compartments, Paris F-75005, France
| |
Collapse
|
238
|
Witkowska M, Smolewski P, Robak T. Investigational therapies targeting CD37 for the treatment of B-cell lymphoid malignancies. Expert Opin Investig Drugs 2018; 27:171-177. [PMID: 29323537 DOI: 10.1080/13543784.2018.1427730] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
INTRODUCTION While chemotherapy still remains a cornerstone of oncologic therapy, immunotherapy with monoclonal antibodies has steadily improved the treatment strategy for several hematologic malignancies. New treatment options need to be developed for relapsed and refractory non-Hodgkin lymphoma (NHL) patients. Currently, novel agents targeting specific molecules on the surface of lymphoma cells, such as anti-CD37 antibodies, are under considerable investigation. Here we report on anti-CD37 targeting for the treatment of patients with B-cell NHL. AREAS COVERED CD37 seems to be the perfect therapeutic target in patients with NHL. The CD37 antigen is abundantly expressed in B-cells, but is absent on normal stem cells and plasma cells. It is hoped that anti-CD37 monoclonal antibodies will increase the efficacy and reduce toxicity in patients with both newly diagnosed and relapsed and refractory disease. Recent clinical trials have shown promising outcomes for these agents, administered both as monotherapy and in combination with standard chemotherapeutics. EXPERT OPINION The development of new therapeutic options might help to avoid cytotoxic chemotherapy entirely in some clinical settings. This article presents the latest state of the art on the new treatment strategies in NHL patients. It also discusses recently approved agents and available clinical trial data.
Collapse
Affiliation(s)
- Magdalena Witkowska
- a Department of Experimental Hematology , Medical University of Lodz , Lodz , Poland
| | - Piotr Smolewski
- a Department of Experimental Hematology , Medical University of Lodz , Lodz , Poland
| | - Tadeusz Robak
- b Department of Hematology , Medical University of Lodz , Lodz , Poland
| |
Collapse
|
239
|
Human Neural Stem Cell Extracellular Vesicles Improve Tissue and Functional Recovery in the Murine Thromboembolic Stroke Model. Transl Stroke Res 2017; 9:530-539. [PMID: 29285679 PMCID: PMC6132936 DOI: 10.1007/s12975-017-0599-2] [Citation(s) in RCA: 204] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Revised: 12/12/2017] [Accepted: 12/14/2017] [Indexed: 02/08/2023]
Abstract
Over 700 drugs have failed in stroke clinical trials, an unprecedented rate thought to be attributed in part to limited and isolated testing often solely in “young” rodent models and focusing on a single secondary injury mechanism. Here, extracellular vesicles (EVs), nanometer-sized cell signaling particles, were tested in a mouse thromboembolic (TE) stroke model. Neural stem cell (NSC) and mesenchymal stem cell (MSC) EVs derived from the same pluripotent stem cell (PSC) line were evaluated for changes in infarct volume as well as sensorimotor function. NSC EVs improved cellular, tissue, and functional outcomes in middle-aged rodents, whereas MSC EVs were less effective. Acute differences in lesion volume following NSC EV treatment were corroborated by MRI in 18-month-old aged rodents. NSC EV treatment has a positive effect on motor function in the aged rodent as indicated by beam walk, instances of foot faults, and strength evaluated by hanging wire test. Increased time with a novel object also indicated that NSC EVs improved episodic memory formation in the rodent. The therapeutic effect of NSC EVs appears to be mediated by altering the systemic immune response. These data strongly support further preclinical development of a NSC EV-based stroke therapy and warrant their testing in combination with FDA-approved stroke therapies.
Collapse
|
240
|
Böker KO, Lemus-Diaz N, Rinaldi Ferreira R, Schiller L, Schneider S, Gruber J. The Impact of the CD9 Tetraspanin on Lentivirus Infectivity and Exosome Secretion. Mol Ther 2017; 26:634-647. [PMID: 29221804 DOI: 10.1016/j.ymthe.2017.11.008] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Revised: 10/24/2017] [Accepted: 11/11/2017] [Indexed: 12/18/2022] Open
Abstract
Efficient transduction tools are a hallmark for both research and therapy development. Here, we introduce new insights into the generation of lentiviral vectors with improved performance by utilizing producer cells with increased production rates of extracellular vesicles through CD9 overexpression. Most human cells secrete small vesicles from their surface (microvesicles) or intraluminal endosome-derived membranes (exosomes). In particular, enhanced levels of the tetraspanin CD9 result in significantly increased numbers of extracellular vesicles with exosome-like features that were secreted from four different human cell lines. Intriguingly, exosomes and their biogenesis route display similarities to lentivirus and we examined the impact of CD9 expression on release and infectivity of recombinant lentiviral vectors. Although the titers of released viral particles were not increased upon production in high CD9 cells, we observed improved performance in terms of both speed and efficiency of lentiviral gene delivery into numerous human cell lines, including HEK293, HeLa, SH-SY5Y, as well as B and T lymphocytes. Here, we demonstrate that enhanced CD9 enables lentiviral transduction in the absence of any pseudotyping viral glycoprotein or fusogenic molecule. Our findings indicate an important role of CD9 for lentiviral vector and exosome biogenesis and point out a remarkable function of this tetraspanin in membrane fusion, viral infectivity, and exosome-mediated horizontal information transfer.
Collapse
Affiliation(s)
- Kai O Böker
- Junior Research Group Medical RNA Biology, German Primate Center, Leibniz Institute for Primate Research, Kellnerweg 4, 37077 Göttingen, Germany; Department for Trauma Surgery, Orthopaedics and Plastic Surgery, University Medical Center, Robert-Koch-Straße 40, 37075 Göttingen, Germany
| | - Nicolas Lemus-Diaz
- Junior Research Group Medical RNA Biology, German Primate Center, Leibniz Institute for Primate Research, Kellnerweg 4, 37077 Göttingen, Germany
| | - Rafael Rinaldi Ferreira
- Junior Research Group Medical RNA Biology, German Primate Center, Leibniz Institute for Primate Research, Kellnerweg 4, 37077 Göttingen, Germany
| | - Lara Schiller
- Junior Research Group Medical RNA Biology, German Primate Center, Leibniz Institute for Primate Research, Kellnerweg 4, 37077 Göttingen, Germany
| | - Stefan Schneider
- Junior Research Group Medical RNA Biology, German Primate Center, Leibniz Institute for Primate Research, Kellnerweg 4, 37077 Göttingen, Germany
| | - Jens Gruber
- Junior Research Group Medical RNA Biology, German Primate Center, Leibniz Institute for Primate Research, Kellnerweg 4, 37077 Göttingen, Germany.
| |
Collapse
|
241
|
Haining EJ, Matthews AL, Noy PJ, Romanska HM, Harris HJ, Pike J, Morowski M, Gavin RL, Yang J, Milhiet PE, Berditchevski F, Nieswandt B, Poulter NS, Watson SP, Tomlinson MG. Tetraspanin Tspan9 regulates platelet collagen receptor GPVI lateral diffusion and activation. Platelets 2017; 28:629-642. [PMID: 28032533 PMCID: PMC5706974 DOI: 10.1080/09537104.2016.1254175] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Revised: 10/06/2016] [Accepted: 10/20/2016] [Indexed: 12/14/2022]
Abstract
The tetraspanins are a superfamily of four-transmembrane proteins, which regulate the trafficking, lateral diffusion and clustering of the transmembrane proteins with which they interact. We have previously shown that tetraspanin Tspan9 is expressed on platelets. Here we have characterised gene-trap mice lacking Tspan9. The mice were viable with normal platelet numbers and size. Tspan9-deficient platelets were specifically defective in aggregation and secretion induced by the platelet collagen receptor GPVI, despite normal surface GPVI expression levels. A GPVI activation defect was suggested by partially impaired GPVI-induced protein tyrosine phosphorylation. In mechanistic experiments, Tspan9 and GPVI co-immunoprecipitated and co-localised, but super-resolution imaging revealed no defects in collagen-induced GPVI clustering on Tspan9-deficient platelets. However, single particle tracking using total internal reflection fluorescence microscopy showed that GPVI lateral diffusion was reduced by approximately 50% in the absence of Tspan9. Therefore, Tspan9 plays a fine-tuning role in platelet activation by regulating GPVI membrane dynamics.
Collapse
Affiliation(s)
- Elizabeth J. Haining
- School of Biosciences, College of Life and Environmental Sciences, University of Birmingham, Birmingham, UK
| | - Alexandra L. Matthews
- School of Biosciences, College of Life and Environmental Sciences, University of Birmingham, Birmingham, UK
| | - Peter J. Noy
- School of Biosciences, College of Life and Environmental Sciences, University of Birmingham, Birmingham, UK
| | | | - Helen J. Harris
- School of Biosciences, College of Life and Environmental Sciences, University of Birmingham, Birmingham, UK
| | - Jeremy Pike
- School of Biosciences, College of Life and Environmental Sciences, University of Birmingham, Birmingham, UK
- PSIBS Doctoral Training Centre, School of Chemistry, University of Birmingham, Birmingham, UK
| | - Martina Morowski
- Department of Experimental Biomedicine, University Hospital, University of Würzburg, Würzburg, Germany
| | - Rebecca L. Gavin
- School of Biosciences, College of Life and Environmental Sciences, University of Birmingham, Birmingham, UK
| | - Jing Yang
- School of Biosciences, College of Life and Environmental Sciences, University of Birmingham, Birmingham, UK
| | - Pierre-Emmanuel Milhiet
- INSERM U1054, CNRS, UMR 5048, Centre de Biochimie Structurale, Montpellier University, France
| | - Fedor Berditchevski
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Bernhard Nieswandt
- Department of Experimental Biomedicine, University Hospital, University of Würzburg, Würzburg, Germany
| | - Natalie S. Poulter
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Steve P. Watson
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Michael G. Tomlinson
- School of Biosciences, College of Life and Environmental Sciences, University of Birmingham, Birmingham, UK
| |
Collapse
|
242
|
Choi SI, Kim SY, Lee JH, Kim JY, Cho EW, Kim IG. Osteopontin production by TM4SF4 signaling drives a positive feedback autocrine loop with the STAT3 pathway to maintain cancer stem cell-like properties in lung cancer cells. Oncotarget 2017; 8:101284-101297. [PMID: 29254164 PMCID: PMC5731874 DOI: 10.18632/oncotarget.21021] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Accepted: 08/26/2017] [Indexed: 01/16/2023] Open
Abstract
Transmembrane 4 L6 family proteins have been known to promote cancer. In this study, we demonstrated that transmembrane 4 L6 family member 4 (TM4SF4), which is induced by γ-radiation in non-small cell lung cancer (NSCLC) cells, is involved in epithelial-to-mesenchymal transition (EMT) and cancer stem cell (CSC) properties of NSCLC through the regulation of osteopontin (OPN). Forced TM4SF4 overexpression in A549 cells increased the secretion of OPN, which activates CD44 or integrin signaling and thus maintains EMT-associated CSC-like properties. OPN, known as a downstream target of β-catenin/T-cell factor 4 (TCF-4), was induced by up-regulated β-catenin via TM4SF4-driven phosphorylation of glycogen synthase kinase 3b (GSK3β). TCF4 complexed to promoter regions of OPN in TM4SF4-overexpressing A549 cells was also confirmed by chromatin immunoprecipitation. Knockout of either β-catenin or TCF4-suppressed OPN expression, demonstrating that both factors are essential for OPN expression in NSCLC cells. OPN secreted by TM4SF4/GSK3β/β-catenin signaling activated the JAK2/STAT3 or FAK/STAT3 pathway, which also up-regulates OPN expression in an autocrine manner and consequently maintains the self-renewal and metastatic capacity of cancer cells. Neutralizing antibody to OPN blocked the autocrine activation of OPN expression, consequently weakened the metastatic and self-renewal capacity of cancer cells. Collectively, our findings indicate that TM4SF4-triggered OPN expression is involved in the persistent reinforcement of EMT or cancer stemness by creating a positive feedback autocrine loop with JAK2/STAT3 or FAK/STAT3 pathways.
Collapse
Affiliation(s)
- Soo Im Choi
- Department of Radiation Biology, Environmental Radiation Research Group, Korea Atomic Energy Research Institute, Yuseong-Gu, Daejeon 34057, Korea
| | - Seo Yoen Kim
- Department of Radiation Biology, Environmental Radiation Research Group, Korea Atomic Energy Research Institute, Yuseong-Gu, Daejeon 34057, Korea
| | - Jei Ha Lee
- Department of Radiation Biology, Environmental Radiation Research Group, Korea Atomic Energy Research Institute, Yuseong-Gu, Daejeon 34057, Korea.,Department of Radiation Biotechnology and Applied Radioisotope, Korea University of Science and Technology (UST), Yuseong-Gu, Daejeon 34057, Korea
| | - Jung Yul Kim
- Department of Radiation Biology, Environmental Radiation Research Group, Korea Atomic Energy Research Institute, Yuseong-Gu, Daejeon 34057, Korea.,Department of Radiation Biotechnology and Applied Radioisotope, Korea University of Science and Technology (UST), Yuseong-Gu, Daejeon 34057, Korea
| | - Eun Wie Cho
- Rare Disease Research Center, Korea Research Institute of Bioscience and Biotechnology, Yuseong-Gu, Daejeon 34141, Korea
| | - In-Gyu Kim
- Department of Radiation Biology, Environmental Radiation Research Group, Korea Atomic Energy Research Institute, Yuseong-Gu, Daejeon 34057, Korea.,Department of Radiation Biotechnology and Applied Radioisotope, Korea University of Science and Technology (UST), Yuseong-Gu, Daejeon 34057, Korea
| |
Collapse
|
243
|
Zuidscherwoude M, Worah K, van der Schaaf A, Buschow SI, van Spriel AB. Differential expression of tetraspanin superfamily members in dendritic cell subsets. PLoS One 2017; 12:e0184317. [PMID: 28880937 PMCID: PMC5589240 DOI: 10.1371/journal.pone.0184317] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2017] [Accepted: 08/22/2017] [Indexed: 11/18/2022] Open
Abstract
Dendritic cells (DCs), which are essential for initiating immune responses, are comprised of different subsets. Tetraspanins organize dendritic cell membranes by facilitating protein-protein interactions within the so called tetraspanin web. In this study we analyzed expression of the complete tetraspanin superfamily in primary murine (CD4+, CD8+, pDC) and human DC subsets (CD1c+, CD141+, pDC) at the transcriptome and proteome level. Different RNA and protein expression profiles for the tetraspanin genes across human and murine DC subsets were identified. Although RNA expression levels of CD37 and CD82 were not significantly different between human DC subsets, CD9 RNA was highly expressed in pDCs, while CD9 protein expression was lower. This indicates that relative RNA and protein expression levels are not always in agreement. Both murine CD8α+ DCs and its regarded human counterpart, CD141+ DCs, displayed relatively high protein levels of CD81. CD53 protein was highly expressed on human pDCs in contrast to the relatively low protein expression of most other tetraspanins. This study demonstrates that tetraspanins are differentially expressed by human and murine DC subsets which provides a valuable resource that will aid the understanding of tetraspanin function in DC biology.
Collapse
Affiliation(s)
- Malou Zuidscherwoude
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Kuntal Worah
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
- Centre for Molecular and Biomolecular Informatics (CMBI), Radboud University Medical Center, Nijmegen, the Netherlands
| | - Alie van der Schaaf
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Sonja I. Buschow
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, the Netherlands
| | - Annemiek B. van Spriel
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
- * E-mail:
| |
Collapse
|
244
|
Grove J, Hu K, Farquhar MJ, Goodall M, Walker L, Jamshad M, Drummer HE, Bill RM, Balfe P, McKeating JA. A new panel of epitope mapped monoclonal antibodies recognising the prototypical tetraspanin CD81. Wellcome Open Res 2017; 2:82. [PMID: 29090272 PMCID: PMC5657224 DOI: 10.12688/wellcomeopenres.12058.1] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/16/2017] [Indexed: 12/26/2022] Open
Abstract
Background: Tetraspanins are small transmembrane proteins, found in all higher eukaryotes, that compartmentalize cellular membranes through interactions with partner proteins. CD81 is a prototypical tetraspanin and contributes to numerous physiological and pathological processes, including acting as a critical entry receptor for hepatitis C virus (HCV). Antibody engagement of tetraspanins can induce a variety of effects, including actin cytoskeletal rearrangements, activation of MAPK-ERK signaling and cell migration. However, the epitope specificity of most anti-tetraspanin antibodies is not known, limiting mechanistic interpretation of these studies. Methods: We generated a panel of monoclonal antibodies (mAbs) specific for CD81 second extracellular domain (EC2) and performed detailed epitope mapping with a panel of CD81 mutants. All mAbs were screened for their ability to inhibit HCV infection and E2-CD81 association. Nanoscale distribution of cell surface CD81 was investigated by scanning electron microscopy. Results: The antibodies were classified in two epitope groups targeting opposing sides of EC2. We observed a wide range of anti-HCV potencies that were independent of their epitope grouping, but associated with their relative affinity for cell-surface expressed CD81. Scanning electron microscopy identified at least two populations of CD81; monodisperse and higher-order assemblies, consistent with tetraspanin-enriched microdomains. Conclusions: These novel antibodies provide well-characterised tools to investigate CD81 function, including HCV entry, and have the potential to provide insights into tetraspanin biology in general.
Collapse
Affiliation(s)
- Joe Grove
- Institute of Immunity and Transplantation, Division of Infection and Immunity, , University College London, London, NW3 2PF, UK
| | - Ke Hu
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, B15 2TT, UK
| | - Michelle J. Farquhar
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, B15 2TT, UK
| | - Margaret Goodall
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, B15 2TT, UK
| | - Lucas Walker
- Institute of Immunity and Transplantation, Division of Infection and Immunity, , University College London, London, NW3 2PF, UK
| | - Mohammed Jamshad
- Institute for Microbiology and Infection, School of Biosciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - Heidi E. Drummer
- Centre for Biomedical Resear, Burnet Institute, Melbourne, VIC, 3004, Australia
| | - Roslyn M. Bill
- School of Life and Health Sciences, Aston University, Birmingham, B4 7ET, UK
| | - Peter Balfe
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, B15 2TT, UK
| | - Jane A. McKeating
- Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7BN, UK
| |
Collapse
|
245
|
Wang J, Zhou Y, Li D, Sun X, Deng Y, Zhao Q. TSPAN
31 is a critical regulator on transduction of survival and apoptotic signals in hepatocellular carcinoma cells. FEBS Lett 2017; 591:2905-2918. [DOI: 10.1002/1873-3468.12737] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Revised: 06/20/2017] [Accepted: 06/21/2017] [Indexed: 12/27/2022]
Affiliation(s)
- Jianglin Wang
- Department of Biochemistry and Molecular Biology GMU‐GIBH Joint School of Life Sciences Guangzhou Medical University China
| | - Yuting Zhou
- Department of Molecular & Cellular Biochemistry College of Medicine University of Kentucky Lexington KY USA
| | - Dan Li
- Department of Biochemistry and Molecular Biology GMU‐GIBH Joint School of Life Sciences Guangzhou Medical University China
| | - Xuemeng Sun
- Department of Biochemistry and Molecular Biology GMU‐GIBH Joint School of Life Sciences Guangzhou Medical University China
| | - Yuanfei Deng
- Department of Biochemistry and Molecular Biology GMU‐GIBH Joint School of Life Sciences Guangzhou Medical University China
| | - Qing Zhao
- Department of Biochemistry and Molecular Biology GMU‐GIBH Joint School of Life Sciences Guangzhou Medical University China
- Sino‐French Hoffmann Institute Guangzhou Medical University China
| |
Collapse
|
246
|
Differential regulation of cellular functions by the C-termini of transmembrane 4 L six family proteins in 2- or 3-dimensional environment. Oncotarget 2017; 8:13277-13292. [PMID: 28129652 PMCID: PMC5355095 DOI: 10.18632/oncotarget.14809] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2016] [Accepted: 12/27/2016] [Indexed: 01/04/2023] Open
Abstract
The transmembrane 4 L six family proteins TM4SF1, TM4SF4, and TM4SF5 share 40-50% overall sequence identity, but their C-terminus identity is limited. It may be likely that the C-termini of the members are important and unique for own regulatory functions. We thus examined how the TM4SF5 C-terminus affected cellular functions differentially from other family members. Using colon cancer cells expressing wildtype (WT), C-terminus-deleted, or chimeric mutants, diverse cellular functions were explored in 2-dimensional (2D) and 3-dimensional (3D) condition. The C-termini of the proteins were relatively comparable with respect to 2D cell proliferation, although each C-terminal-deletion mutant exhibited increased proliferation relative to the WT. Using chimeric constructs, we found that the TM4SF5 C-terminus was critical for regulating the diverse metastatic functions of TM4SF5, and could positively replace the C-termini of other family members. Replacement of the TM4SF1 or TM4SF4 C-terminus with that of TM4SF5 increased spheroids growth, transwell migration, and invasive dissemination from spheroids in 3D collagen gels. TM4SF5-mediated effects required its extracellular loop 2 linked to the C-terminus via the transmembrane domain 4, with causing c-Src activation. Altogether, the C-terminus of TM4SF5 appears to mediate pro-migratory roles, depending on a structural relay from the second extracellular loop to the C-terminus.
Collapse
|
247
|
TSPAN7, effector of actin nucleation required for dendritic cell-mediated transfer of HIV-1 to T cells. Biochem Soc Trans 2017; 45:703-708. [PMID: 28620031 DOI: 10.1042/bst20160439] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Revised: 03/15/2017] [Accepted: 03/21/2017] [Indexed: 01/18/2023]
Abstract
Dendritic cells (DCs) have essential roles in early detection of pathogens and activation of both innate and adaptive immune responses. Whereas human DCs are resistant to productive HIV-1 replication, they have a unique ability to take up virus and transmit it efficiently to T lymphocytes. By doing that, HIV-1 may evade, at least in part, the first line of defense of the immune system, exploiting DCs instead to facilitate rapid infection of a large pool of immune cells. While performing an shRNA screen in human primary monocyte-derived DCs, to gain insights into this cell biological process, we discovered the role played by tetraspanin-7 (TSPAN7). This member of the tetraspanin family appears to be a positive regulator of actin nucleation and stabilization, through the ARP2/3 complex. By doing so, TSPAN7 limits HIV-1 endocytosis and maintains viral particles on actin-rich dendrites for an efficient transfer toward T lymphocytes. While studying the function of TSPAN7 in the control of actin nucleation, we also discovered the existence in DCs of two opposing forces at the plasma membrane: actin nucleation, a protrusive force which seems to counterbalance actomyosin contraction.
Collapse
|
248
|
Molecular interactions shaping the tetraspanin web. Biochem Soc Trans 2017; 45:741-750. [PMID: 28620035 DOI: 10.1042/bst20160284] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 03/14/2017] [Accepted: 03/15/2017] [Indexed: 12/31/2022]
Abstract
To facilitate the myriad of different (signaling) processes that take place at the plasma membrane, cells depend on a high degree of membrane protein organization. Important mediators of this organization are tetraspanin proteins. Tetraspanins interact laterally among themselves and with partner proteins to control the spatial organization of membrane proteins in large networks called the tetraspanin web. The molecular interactions underlying the formation of the tetraspanin web were hitherto mainly described based on their resistance to different detergents, a classification which does not necessarily correlate with functionality in the living cell. To look at these interactions from a more physiological point of view, this review discusses tetraspanin interactions based on their function in the tetraspanin web: (1) intramolecular interactions supporting tetraspanin structure, (2) tetraspanin-tetraspanin interactions supporting web formation, (3) tetraspanin-partner interactions adding functional partners to the web and (4) cytosolic tetraspanin interactions regulating intracellular signaling. The recent publication of the first full-length tetraspanin crystal structure sheds new light on both the intra- and intermolecular tetraspanin interactions that shape the tetraspanin web. Furthermore, recent molecular dynamic modeling studies indicate that the binding strength between tetraspanins and between tetraspanins and their partners is the complex sum of both promiscuous and specific interactions. A deeper insight into this complex mixture of interactions is essential to our fundamental understanding of the tetraspanin web and its dynamics which constitute a basic building block of the cell surface.
Collapse
|
249
|
Scissor sisters: regulation of ADAM10 by the TspanC8 tetraspanins. Biochem Soc Trans 2017; 45:719-730. [PMID: 28620033 PMCID: PMC5473022 DOI: 10.1042/bst20160290] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Revised: 02/26/2017] [Accepted: 02/28/2017] [Indexed: 12/20/2022]
Abstract
A disintegrin and metalloprotease 10 (ADAM10) is a ubiquitously expressed transmembrane protein which is essential for embryonic development through activation of Notch proteins. ADAM10 regulates over 40 other transmembrane proteins and acts as a ‘molecular scissor’ by removing their extracellular regions. ADAM10 is also a receptor for α-toxin, a major virulence factor of Staphylococcus aureus. Owing to the importance of its substrates, ADAM10 is a potential therapeutic target for cancer, neurodegenerative diseases such as Alzheimer's and prion diseases, bacterial infection and inflammatory diseases such as heart attack, stroke and asthma. However, targetting ADAM10 is likely to result in toxic side effects. The tetraspanins are a superfamily of 33 four-transmembrane proteins in mammals which interact with and regulate specific partner proteins within membrane nanodomains. Tetraspanins appear to have a cone-shaped structure with a cholesterol-binding cavity, which may enable tetraspanins to undergo cholesterol-regulated conformational change. An emerging paradigm for tetraspanin function is the regulation of ADAM10 by the TspanC8 subgroup of tetraspanins, namely Tspan5, 10, 14, 15, 17 and 33. This review will describe how TspanC8s are required for ADAM10 trafficking from the endoplasmic reticulum and its enzymatic maturation. Moreover, different TspanC8s localise ADAM10 to different subcellular localisations and may cause ADAM10 to adopt distinct conformations and cleavage of distinct substrates. We propose that ADAM10 should now be regarded as six different scissor proteins depending on the interacting TspanC8. Therapeutic targetting of specific TspanC8/ADAM10 complexes could allow ADAM10 targetting in a cell type- or substrate-specific manner, to treat certain diseases while minimising toxicity.
Collapse
|
250
|
Hassuna NA, Monk PN, Ali F, Read RC, Partridge LJ. A role for the tetraspanin proteins in Salmonella infection of human macrophages. J Infect 2017; 75:115-124. [DOI: 10.1016/j.jinf.2017.06.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2017] [Revised: 05/31/2017] [Accepted: 06/02/2017] [Indexed: 10/19/2022]
|