1
|
Shao Z, Hao Q, Chen J, Lu Y. TSPAN15 enhances EMT-mediated metastasis of HCC by promoting autophagy through BTRC-mediated PDCD4 degradation. Mol Immunol 2025; 183:203-212. [PMID: 40398082 DOI: 10.1016/j.molimm.2025.05.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2025] [Revised: 05/08/2025] [Accepted: 05/12/2025] [Indexed: 05/23/2025]
Abstract
BACKGROUND Cumulative evidence shows that Tetraspanin 15 (TSPAN15) shows a high degree of consistency in a variety of tumor characteristics, which has attracted extensive attention from researchers. We used TSPAN15 as a starting point to explore the role and mechanism of TSPAN15 in in hepatocellular carcinoma (HCC). METHODS Using database analysis, recombinant plasmid transfection technology, transwell, autophagic flux analysis and western blotting, the effects of TSPAN15 on autophagy, invasion, epithelial-mesenchymal transition (EMT) of HCC cells, and tumor growth and metastasis were elucidated after silencing TSPAN15 in HCC cells. The effect of TSPAN15 on tumor growth was detected by using xenograft model of nude mice. RESULTS Based on the online database and immunohistochemistry analysis, it was found that the mRNA and protein expression of TSPAN15 in HCC tissues was significantly higher than that in normal liver tissues or adjacent non-cancerous tissues. High expression of TSPAN15 was an independent risk factor for poor prognosis in TCGA-LIHC patients. TSPAN15 silencing inhibited HCC autophagy and autophagy-induced migration, invasion and EMT as well as tumor growth and metastasis. Mechanistically, TSPAN15 contributed to programmed cell death 4 (PDCD4) proteasomal degradation through physical interaction with beta-transducin repeat containing (BTRC), thus activing autophagy. Rescue experiments revealed that PDCD4 effectively inhibited TSPAN15-induced autophagy, migration, invasion and EMT. CONCLUSION Abnormally expressed TSPAN15 promotes the degradation of tumor suppressor gene PDCD4 through ubiquitination, thereby promoting autophagy and autophagy-mediated EMT and metastasis of HCC cells, demonstrating the importance of TSPAN15 in the molecular etiology of HCC and its potential therapeutic value.
Collapse
Affiliation(s)
- Zicheng Shao
- Suzhou Medical College of Soochow University, Suzhou, Jiangsu 215000, China; Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, China.
| | - Qingya Hao
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, China
| | - Jie Chen
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, China
| | - Yuhua Lu
- Suzhou Medical College of Soochow University, Suzhou, Jiangsu 215000, China; Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, China.
| |
Collapse
|
2
|
Rubinstein E, Théry C, Zimmermann P. Tetraspanins affect membrane structures and the trafficking of molecular partners: what impact on extracellular vesicles? Biochem Soc Trans 2025; 0:BST20240523. [PMID: 40135387 DOI: 10.1042/bst20240523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 03/03/2025] [Accepted: 03/07/2025] [Indexed: 03/27/2025]
Abstract
Tetraspanins are a family of 33 proteins in mammals believed to play a crucial role in the compartmentalization of various associated proteins within cells and membranes. Recent studies have elucidated the structure of several tetraspanin members, revealing that while the four transmembrane domains typically adopt a cone-shaped configuration in crystals, other conformations are also possible. This cone-shaped structure may explain why tetraspanins are often enriched in curved and tubular cellular structures, such as microvilli, tunneling nanotubes, retraction fibers, or at the site of virus budding, and may contribute to the formation or maintenance of these structures. Tetraspanins have also been detected on midbody remnants and migrasomes, as well as on extracellular vesicles (EVs), for which CD9, CD81, and CD63 are widely used as markers. Although their impact on certain membrane structures and their ability to regulate the function and trafficking of associated proteins would suggest a potential role of tetraspanins either in EV formation or in regulating their protein composition, or both, efforts to characterize these roles have been complicated by conflicting results. In line with the interaction of certain tetraspanins with cholesterol, two recent studies have suggested that the presence or organization of oxysterols and cholesterol in EVs may be regulated by Tspan6 and CD63, respectively, paving the way for further research on the influence of tetraspanins on the lipid composition of EVs.
Collapse
Affiliation(s)
- Eric Rubinstein
- Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Sorbonne Université, Inserm, CNRS, Paris, France
| | - Clotilde Théry
- Institut Curie Research Center, PSL Research University, INSERM U932, Paris, France
- Institut Curie Research Center, CurieCoreTech Extracellular Vesicles, Paris, France
| | - Pascale Zimmermann
- Equipe labellisée Ligue 2024, Centre de Recherche en Cancérologie de Marseille (CRCM), Aix-Marseille Université, Inserm, CNRS, Institut Paoli-Calmettes, Marseille, France
- Department of Human Genetics, KU Leuven, Leuven, Belgium
| |
Collapse
|
3
|
Xu K, Feng H, Zhao R, Huang Y. Targeting Tetraspanins at Cell Interfaces: Functional Modulation and Exosome-Based Drug Delivery for Precise Disease Treatment. ChemMedChem 2025; 20:e202400664. [PMID: 39415492 DOI: 10.1002/cmdc.202400664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 10/13/2024] [Accepted: 10/16/2024] [Indexed: 10/18/2024]
Abstract
Tetraspanins are key players in various physiological and pathological processes, including malignancy, immune response, fertilization, and infectious disease. Affinity ligands targeting the interactions between tetraspanins and partner proteins are promising for modulating downstream signaling pathways, thus emerging as attractive candidates for interfering related biological functions. Due to the involvement in vesicle biogenesis and cargo trafficking, tetraspanins are also regarded as exosome markers, and become molecular targets for drug loading and delivery. Given the rapid development in these areas, this minireview focuses on recent advances in design and engineering of affinity binders toward tetraspanins including CD63, CD81, and CD9. Their mechanism of actions in modulating protein interactions at cell interfaces and treatment of malignant diseases are discussed. Strategies for constructing exosome-based drug delivery platforms are also reviewed, with emphasis on the important roles of tetraspanins and the affinity ligands. Finally, challenges and future development of tetraspanin-targeting therapy and exosomal drug delivery platforms are also discussed.
Collapse
Affiliation(s)
- Kun Xu
- Beijing National Laboratory for Molecular Sciences, CAS Key Laboratory of Analytical Chemistry for Living Biosystems, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
- School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Huixia Feng
- Beijing National Laboratory for Molecular Sciences, CAS Key Laboratory of Analytical Chemistry for Living Biosystems, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
- School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Rui Zhao
- Beijing National Laboratory for Molecular Sciences, CAS Key Laboratory of Analytical Chemistry for Living Biosystems, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
- School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yanyan Huang
- Beijing National Laboratory for Molecular Sciences, CAS Key Laboratory of Analytical Chemistry for Living Biosystems, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
- School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| |
Collapse
|
4
|
Ramseier NT, Jing H, Anderson J, Hu YS. Superresolution Imaging Reveals the Spatial Organization of CD81 Microdomains in Regulating Membrane Signaling on Jurkat T Cell Microvilli. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.07.627345. [PMID: 39677771 PMCID: PMC11643289 DOI: 10.1101/2024.12.07.627345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Tetraspanin proteins are closely associated with high-curvature membrane structures and play key roles in organizing membrane domains and regulating membrane signaling in immune cells. However, their specific roles in regulating T cell membrane signaling, particularly within the microvilli often characteristic of these cells, remain poorly understood. Here, we used Jurkat T cells as a model system and investigated CD81 as a member of the tetraspanin family. Using total internal reflection fluorescence (TIRF) microscopy and structured illumination microscopy (SIM), we identified an enrichment of the tetraspanin CD81 microdomains along the actin-rich membrane microvilli. At the distal end of the microvilli, SIM images revealed the spatial colocalization of CD81 with T cell receptors (TCR) and CD63, implying a potential role for CD81 in regulating TCR signaling in conjunction with CD63. Spatial analysis of CD81 and CD63 microdomains from the dual-color SIM data revealed their preference for associating with each other. Cluster analysis of direct stochastic optical reconstruction microscopy (dSTORM) data revealed that in vitro T cell activation results in reduced domain sizes and increased domain separation of CD81. These findings provide visual evidence of the spatial organization and rearrangement of CD81 on the T cell microvilli, highlighting its potential role in signal regulation on specialized membrane protrusions.
Collapse
Affiliation(s)
- Neal T. Ramseier
- Department of Chemistry, College of Liberal Arts and Sciences, University of Illinois Chicago, Chicago, IL 60607, USA
| | - Haoran Jing
- Department of Chemistry, College of Liberal Arts and Sciences, University of Illinois Chicago, Chicago, IL 60607, USA
| | - Jesse Anderson
- Department of Chemical Engineering, College of Engineering, University of Illinois Chicago, Chicago, IL 60607, USA
| | - Ying S. Hu
- Department of Chemistry, College of Liberal Arts and Sciences, University of Illinois Chicago, Chicago, IL 60607, USA
| |
Collapse
|
5
|
Dharan R, Sorkin R. Tetraspanin proteins in membrane remodeling processes. J Cell Sci 2024; 137:jcs261532. [PMID: 39051897 DOI: 10.1242/jcs.261532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/27/2024] Open
Abstract
Membrane remodeling is a fundamental cellular process that is crucial for physiological functions such as signaling, membrane fusion and cell migration. Tetraspanins (TSPANs) are transmembrane proteins of central importance to membrane remodeling events. During these events, TSPANs are known to interact with themselves and other proteins and lipids; however, their mechanism of action in controlling membrane dynamics is not fully understood. Since these proteins span the membrane, membrane properties such as rigidity, curvature and tension can influence their behavior. In this Review, we summarize recent studies that explore the roles of TSPANs in membrane remodeling processes and highlight the unique structural features of TSPANs that mediate their interactions and localization. Further, we emphasize the influence of membrane curvature on TSPAN distribution and membrane domain formation and describe how these behaviors affect cellular functions. This Review provides a comprehensive perspective on the multifaceted function of TSPANs in membrane remodeling processes and can help readers to understand the intricate molecular mechanisms that govern cellular membrane dynamics.
Collapse
Affiliation(s)
- Raviv Dharan
- School of Chemistry , Raymond & Beverly Sackler Faculty of Exact Sciences , Tel Aviv University, 6997801, Tel Aviv, Israel
- Center for Physics and Chemistry of Living Systems , Tel Aviv University, 6997801, Tel Aviv, Israel
| | - Raya Sorkin
- School of Chemistry , Raymond & Beverly Sackler Faculty of Exact Sciences , Tel Aviv University, 6997801, Tel Aviv, Israel
- Center for Physics and Chemistry of Living Systems , Tel Aviv University, 6997801, Tel Aviv, Israel
| |
Collapse
|
6
|
Schmidt SC, Massenberg A, Homsi Y, Sons D, Lang T. Microscopic clusters feature the composition of biochemical tetraspanin-assemblies and constitute building-blocks of tetraspanin enriched domains. Sci Rep 2024; 14:2093. [PMID: 38267610 PMCID: PMC10808221 DOI: 10.1038/s41598-024-52615-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 01/21/2024] [Indexed: 01/26/2024] Open
Abstract
Biochemical approaches revealed that tetraspanins are multi-regulatory proteins forming a web, where they act in tetraspanin-enriched-microdomains (TEMs). A microscopic criterion differentiating between web and TEMs is lacking. Using super-resolution microcopy, we identify co-assemblies between the tetraspanins CD9 and CD81 and CD151 and CD81. CD9 assemblies contain as well the CD9/CD81-interaction partner EWI-2. Moreover, CD9 clusters are proximal to clusters of the CD81-interaction partner CD44 and CD81-/EWI-2-interacting ezrin-radixin-moesin proteins. Assemblies scatter unorganized across the cell membrane; yet, upon EWI-2 elevation, they agglomerate into densely packed arranged-crowds in a process independent from actin dynamics. In conclusion, microscopic clusters are equivalent to biochemical tetraspanin-assemblies, defining in their entirety the tetraspanin web. Cluster-agglomeration enriches tetraspanins, which makes agglomerations to a microscopic complement of TEMs. The microscopic classification of tetraspanin assemblies advances our understanding of this enigmatic protein family, whose members play roles in a plethora of cellular functions, diseases, and pathogen infections.
Collapse
Affiliation(s)
- Sara C Schmidt
- Faculty of Mathematics and Natural Sciences, Life & Medical Sciences (LIMES) Institute, University of Bonn, Carl-Troll-Straße 31, 53115, Bonn, Germany
| | - Annika Massenberg
- Faculty of Mathematics and Natural Sciences, Life & Medical Sciences (LIMES) Institute, University of Bonn, Carl-Troll-Straße 31, 53115, Bonn, Germany
| | - Yahya Homsi
- Faculty of Mathematics and Natural Sciences, Life & Medical Sciences (LIMES) Institute, University of Bonn, Carl-Troll-Straße 31, 53115, Bonn, Germany
| | - Dominik Sons
- Faculty of Mathematics and Natural Sciences, Life & Medical Sciences (LIMES) Institute, University of Bonn, Carl-Troll-Straße 31, 53115, Bonn, Germany
| | - Thorsten Lang
- Faculty of Mathematics and Natural Sciences, Life & Medical Sciences (LIMES) Institute, University of Bonn, Carl-Troll-Straße 31, 53115, Bonn, Germany.
| |
Collapse
|
7
|
Carey TR, Kozminsky M, Hall J, Vargas-Zapata V, Geiger K, Coscoy L, Sohn LL. Detecting Intact Virus Using Exogenous Oligonucleotide Labels. Anal Chem 2022; 94:7619-7627. [PMID: 35584293 DOI: 10.1021/acs.analchem.2c00835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The COVID-19 pandemic has revealed how an emerging pathogen can cause a sudden and dramatic increase in demand for viral testing. Testing pooled samples could meet this demand; however, the sensitivity of reverse transcription quantitative polymerase chain reaction (RT-qPCR), the gold standard, significantly decreases with an increasing number of samples pooled. Here, we introduce detection of intact virus by exogenous-nucleotide reaction (DIVER), a method that quantifies intact virus and is robust to sample dilution. As demonstrated using two models of severe acute respiratory syndrome coronavirus 2, DIVER first tags membraned particles with exogenous oligonucleotides, then captures the tagged particles on beads functionalized with a virus-specific capture agent (in this instance, angiotensin-converting enzyme 2), and finally quantifies the oligonucleotide tags using qPCR. Using spike-presenting liposomes and spike-pseudotyped lentivirus, we show that DIVER can detect 1 × 105 liposomes and 100 plaque-forming units of lentivirus and can successfully identify positive samples in pooling experiments. Overall, DIVER is well positioned for efficient sample pooling and clinical validation.
Collapse
Affiliation(s)
- Thomas R Carey
- UC Berkeley-UC San Francisco Graduate Program in Bioengineering, University of California, Berkeley, 306 Stanley Hall, Berkeley, California 94720, United States
| | - Molly Kozminsky
- California Institute for Quantitative Biosciences, University of California, Berkeley, 174 Stanley Hall, Berkeley, California 94720, United States
| | - Jennifer Hall
- Department of Molecular and Cellular Biology, University of California, Berkeley, 3200 Weill Hall, Berkeley, California 94720, United States
| | - Valerie Vargas-Zapata
- Department of Molecular and Cellular Biology, University of California, Berkeley, 3200 Weill Hall, Berkeley, California 94720, United States
| | - Kristina Geiger
- Department of Molecular and Cellular Biology, University of California, Berkeley, 3200 Weill Hall, Berkeley, California 94720, United States
| | - Laurent Coscoy
- Department of Molecular and Cellular Biology, University of California, Berkeley, 3200 Weill Hall, Berkeley, California 94720, United States
| | - Lydia L Sohn
- UC Berkeley-UC San Francisco Graduate Program in Bioengineering, University of California, Berkeley, 306 Stanley Hall, Berkeley, California 94720, United States.,Department of Mechanical Engineering, University of California, Berkeley, 5118 Etcheverry Hall, Berkeley, California 94720, United States
| |
Collapse
|
8
|
Siu KK, Serrão VHB, Ziyyat A, Lee JE. The cell biology of fertilization: Gamete attachment and fusion. J Cell Biol 2021; 220:e202102146. [PMID: 34459848 PMCID: PMC8406655 DOI: 10.1083/jcb.202102146] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 08/13/2021] [Accepted: 08/16/2021] [Indexed: 02/06/2023] Open
Abstract
Fertilization is defined as the union of two gametes. During fertilization, sperm and egg fuse to form a diploid zygote to initiate prenatal development. In mammals, fertilization involves multiple ordered steps, including the acrosome reaction, zona pellucida penetration, sperm-egg attachment, and membrane fusion. Given the success of in vitro fertilization, one would think that the mechanisms of fertilization are understood; however, the precise details for many of the steps in fertilization remain a mystery. Recent studies using genetic knockout mouse models and structural biology are providing valuable insight into the molecular basis of sperm-egg attachment and fusion. Here, we review the cell biology of fertilization, specifically summarizing data from recent structural and functional studies that provide insights into the interactions involved in human gamete attachment and fusion.
Collapse
Affiliation(s)
- Karen K. Siu
- Department of Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Vitor Hugo B. Serrão
- Department of Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Ahmed Ziyyat
- Université de Paris, Institut Cochin, Institut National de la Santé et de la Recherche Médicale, Centre National de la Recherche Scientifique, Paris, France
- Service d’Histologie, d’Embryologie, Biologie de la Reproduction, Assistance Publique - Hôpitaux de Paris, Hôpital Cochin, Paris, France
| | - Jeffrey E. Lee
- Department of Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
9
|
Ren K, Blosser MC, Malmstadt N. Light-Triggered Unique Shape Transformation of Giant Polymersomes with Tubular Protrusions. Macromol Rapid Commun 2021; 42:e2100474. [PMID: 34553805 DOI: 10.1002/marc.202100474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 09/04/2021] [Indexed: 11/10/2022]
Abstract
Light-triggered unique shape transformation of calcein-loaded giant polymersomes with tubular protrusions, which serve as a reservoir membrane area during the shape transformation, is reported here. Under irradiation at the excitation wavelength of calcein, the tubular protrusions form strings of budded vesicles and then reintegrate into the mother vesicle. The initial giant polymersomes transform to two connected spherical vesicles via two pathways to alleviate the osmotic pressure imbalance across the vesicle membrane. The two connected spherical vesicles further transform to a mother vesicle with an inner daughter vesicle after switching off the light to relieve the bending energy. The finding provides a promising platform to mimic cell morphology changes.
Collapse
Affiliation(s)
- Kaixuan Ren
- Department of Polymer Materials, School of Materials Science and Engineering, Shanghai University, Shanghai, 200444, P. R. China.,Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, 925 Bloom Walk, Los Angeles, CA, 90089-1211, USA
| | - Matthew C Blosser
- Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, 925 Bloom Walk, Los Angeles, CA, 90089-1211, USA
| | - Noah Malmstadt
- Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, 925 Bloom Walk, Los Angeles, CA, 90089-1211, USA.,Department of Chemistry, University of Southern California, 840 Downey Way, Los Angeles, CA, 90089-0744, USA.,Department of Biomedical Engineering, University of Southern California, 3650 McClintock Avenue, Los Angeles, CA, 90089-1111, USA
| |
Collapse
|
10
|
Bray ER, Oropallo AR, Grande DA, Kirsner RS, Badiavas EV. Extracellular Vesicles as Therapeutic Tools for the Treatment of Chronic Wounds. Pharmaceutics 2021; 13:1543. [PMID: 34683836 PMCID: PMC8541217 DOI: 10.3390/pharmaceutics13101543] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 09/16/2021] [Accepted: 09/17/2021] [Indexed: 12/17/2022] Open
Abstract
Chronic wounds develop when the orderly process of cutaneous wound healing is delayed or disrupted. Development of a chronic wound is associated with significant morbidity and financial burden to the individual and health-care system. Therefore, new therapeutic modalities are needed to address this serious condition. Mesenchymal stem cells (MSCs) promote skin repair, but their clinical use has been limited due to technical challenges. Extracellular vesicles (EVs) are particles released by cells that carry bioactive molecules (lipids, proteins, and nucleic acids) and regulate intercellular communication. EVs (exosomes, microvesicles, and apoptotic bodies) mediate key therapeutic effects of MSCs. In this review we examine the experimental data establishing a role for EVs in wound healing. Then, we explore techniques for designing EVs to function as a targeted drug delivery system and how EVs can be incorporated into biomaterials to produce a personalized wound dressing. Finally, we discuss the status of clinically deploying EVs as a therapeutic agent in wound care.
Collapse
Affiliation(s)
- Eric R. Bray
- Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (E.R.B.); (R.S.K.)
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Alisha R. Oropallo
- Comprehensive Wound Healing Center and Hyperbarics, Department of Vascular Surgery, Donald and Barbara Zucker School of Medicine, Hofstra/Northwell Health, Hempstead, NY 11549, USA; (A.R.O.); (D.A.G.)
- Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY 11030, USA
| | - Daniel A. Grande
- Comprehensive Wound Healing Center and Hyperbarics, Department of Vascular Surgery, Donald and Barbara Zucker School of Medicine, Hofstra/Northwell Health, Hempstead, NY 11549, USA; (A.R.O.); (D.A.G.)
- Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY 11030, USA
- Department of Orthopedic Surgery, Long Island Jewish Medical Center, Northwell Health, New Hyde Park, NY 11040, USA
| | - Robert S. Kirsner
- Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (E.R.B.); (R.S.K.)
| | - Evangelos V. Badiavas
- Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (E.R.B.); (R.S.K.)
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
11
|
Cholesterol plays a decisive role in tetraspanin assemblies during bilayer deformations. Biosystems 2021; 209:104505. [PMID: 34403719 DOI: 10.1016/j.biosystems.2021.104505] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Revised: 07/30/2021] [Accepted: 08/07/2021] [Indexed: 11/22/2022]
Abstract
The tetraspanin family plays key roles in many physiological processes, such as, tumour invasion, cell motility, virus infection, cell attachment and entry. Tetraspanins function as molecular scaffolds organized in microdomains with interesting downstream cellular consequences. However, despite their relevance in human physiology, the precise mechanisms of their various functions remain elusive. In particular, the full-length CD81 tetraspanin has interesting cholesterol-related properties that modulate its activity in cells. In this work, we study the opening transition of CD81 under different conditions. We propose that such conformational change is a collaborative process enhanced by simultaneous interactions between multiple identical CD81 tetraspanins. With molecular dynamics simulations we describe the crucial role of a ternary lipid bilayer with cholesterol in CD81 conformational dynamics, observing two emergent properties: first, clusters of CD81 collectively segregate one tetraspanin while favouring one opening transition, second, cumulative cholesterol sequestering by CD81 tetraspanins inhibits large membrane deformations due to local density variations.
Collapse
|
12
|
Carey TR, Kozminsky M, Hall J, Vargas-Zapata V, Geiger K, Coscoy L, Sohn LL. Toward Community Surveillance: Detecting Intact SARS-CoV-2 Using Exogeneous Oligonucleotide Labels. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2021:2021.03.23.21254201. [PMID: 33791715 PMCID: PMC8010747 DOI: 10.1101/2021.03.23.21254201] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
The persistence of the COVID-19 pandemic demands a dramatic increase in testing efficiency. Testing pooled samples for SARS-CoV-2 could meet this need; however, the sensitivity of RT-qPCR, the gold standard, significantly decreases with an increasing number of samples pooled. Here, we introduce DIVER, a method that quantifies intact virus and is robust to sample dilution. DIVER first tags viral particles with exogeneous oligonucleotides, then captures the tagged particles on ACE2-functionalized beads, and finally quantifies the oligonucleotide tags using qPCR. Using spike-presenting liposomes and Spike-pseudotyped lentivirus as SARS-CoV-2 models, we show that DIVER can detect 1×10 5 liposomes and 100 pfu lentivirus and can successfully identify positive samples in pooling experiments. Overall, DIVER is well-positioned for efficient sample pooling and expanded community surveillance.
Collapse
|
13
|
Lorico A, Lorico-Rappa M, Karbanová J, Corbeil D, Pizzorno G. CD9, a tetraspanin target for cancer therapy? Exp Biol Med (Maywood) 2021; 246:1121-1138. [PMID: 33601913 DOI: 10.1177/1535370220981855] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
In the present minireview, we intend to provide a brief history of the field of CD9 involvement in oncogenesis and in the metastatic process of cancer, considering its potential value as a tumor-associated antigenic target. Over the years, CD9 has been identified as a favorable prognostic marker or predictor of metastatic potential depending on the cancer type. To understand its implications in cancer beside its use as an antigenic biomarker, it is essential to know its physiological functions, including its molecular partners in a given cell system. Moreover, the discovery that CD9 is one of the most specific and broadly expressed markers of extracellular membrane vesicles, nanometer-sized entities that are released into extracellular space and various physiological body fluids and play a role in intercellular communication under physiological and pathological conditions, notably the establishment of cancer metastases, has added a new dimension to our knowledge of CD9 function in cancer. Here, we will discuss these issues as well as the possible cancer therapeutic implications of CD9, their limitations, and pitfalls.
Collapse
Affiliation(s)
- Aurelio Lorico
- Touro University College of Medicine, Henderson, NV 89014, USA.,Mediterranean Institute of Oncology, Viagrande 95029, Italy
| | | | - Jana Karbanová
- Biotechnology Center and Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Dresden 01307, Germany
| | - Denis Corbeil
- Biotechnology Center and Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Dresden 01307, Germany
| | - Giuseppe Pizzorno
- University of Tennessee Health Science Center, Memphis, TN 38163, USA.,Erlanger Health System, Chattanooga, TN 37403 , USA
| |
Collapse
|
14
|
Zhang Y, Wang J, Ding Y, Zhang J, Xu Y, Xu J, Zheng S, Yang H. Migrasome and Tetraspanins in Vascular Homeostasis: Concept, Present, and Future. Front Cell Dev Biol 2020; 8:438. [PMID: 32612990 PMCID: PMC7308473 DOI: 10.3389/fcell.2020.00438] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 05/11/2020] [Indexed: 12/18/2022] Open
Abstract
Cell migration plays a critical role in vascular homeostasis. Under noxious stimuli, endothelial cells (ECs) migration always contributes to vascular repair, while enhanced migration of vascular smooth muscle cells (VSMCs) will lead to pathological vascular remodeling. Moreover, vascular activities are involved in communication between ECs and VSMCs, between ECs and immune cells, et al. Recently, Ma et al. (2015) discovered a novel migration-dependent organelle “migrasome,” which mediated release of cytoplasmic contents, and this process was defined as “migracytosis.” The formation of migrasome is precisely regulated by tetraspanins (TSPANs), cholesterol and integrins. Migrasomes can be taken up by neighboring cells, and migrasomes are distributed in many kinds of cells and tissues, such as in blood vessel, human serum, and in ischemic brain of human and mouse. In addition, the migrasome elements TSPANs are wildly expressed in cardiovascular system. Therefore, TSPANs, migrasomes and migracytosis might play essential roles in regulating vascular homeostasis. In this review, we will discuss the discoveries of migration-dependent migrasome and migracytosis, migrasome formation, the basic differences between migrasomes and exosomes, the distributions and functions of migrasome, the functions of migrasome elements TSPANs in vascular biology, and discuss the possible roles of migrasomes and migracytosis in vascular homeostasis.
Collapse
Affiliation(s)
- Yaxing Zhang
- Department of Traditional Chinese Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jing Wang
- Department of Ophthalmology, Qingdao Fubai Eye Hospital, Qingdao, China
| | - Yungang Ding
- Department of Ophthalmology, Qingdao Ludong Eye Hospital, Qingdao, China
| | - Jiongshan Zhang
- Department of Traditional Chinese Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yan Xu
- Department of Gastrointestinal Endoscopy, Guangzhou Cadre Health Management Center/Guangzhou Eleventh People's Hospital, Guangzhou, China
| | - Jingting Xu
- Biofeedback Laboratory, Xinhua College of Sun Yat-sen University, Guangzhou, China
| | - Shuhui Zheng
- Research Center for Translational Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Hongzhi Yang
- Department of Traditional Chinese Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
15
|
Beyenbach KW, Schöne F, Breitsprecher LF, Tiburcy F, Furuse M, Izumi Y, Meyer H, Jonusaite S, Rodan AR, Paululat A. The septate junction protein Tetraspanin 2A is critical to the structure and function of Malpighian tubules in Drosophila melanogaster. Am J Physiol Cell Physiol 2020; 318:C1107-C1122. [PMID: 32267718 DOI: 10.1152/ajpcell.00061.2020] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Tetraspanin-2A (Tsp2A) is an integral membrane protein of smooth septate junctions in Drosophila melanogaster. To elucidate its structural and functional roles in Malpighian tubules, we used the c42-GAL4/UAS system to selectively knock down Tsp2A in principal cells of the tubule. Tsp2A localizes to smooth septate junctions (sSJ) in Malpighian tubules in a complex shared with partner proteins Snakeskin (Ssk), Mesh, and Discs large (Dlg). Knockdown of Tsp2A led to the intracellular retention of Tsp2A, Ssk, Mesh, and Dlg, gaps and widening spaces in remaining sSJ, and tumorous and cystic tubules. Elevated protein levels together with diminished V-type H+-ATPase activity in Tsp2A knockdown tubules are consistent with cell proliferation and reduced transport activity. Indeed, Malpighian tubules isolated from Tsp2A knockdown flies failed to secrete fluid in vitro. The absence of significant transepithelial voltages and resistances manifests an extremely leaky epithelium that allows secreted solutes and water to leak back to the peritubular side. The tubular failure to excrete fluid leads to extracellular volume expansion in the fly and to death within the first week of adult life. Expression of the c42-GAL4 driver begins in Malpighian tubules in the late embryo and progresses upstream to distal tubules in third instar larvae, which can explain why larvae survive Tsp2A knockdown and adults do not. Uncontrolled cell proliferation upon Tsp2A knockdown confirms the role of Tsp2A as tumor suppressor in addition to its role in sSJ structure and transepithelial transport.
Collapse
Affiliation(s)
- Klaus W Beyenbach
- Department of Zoology/Developmental Biology, University of Osnabrück, Osnabrück, Germany.,Department of Animal Physiology, University of Osnabrück, Osnabrück, Germany
| | - Frederike Schöne
- Department of Zoology/Developmental Biology, University of Osnabrück, Osnabrück, Germany
| | | | - Felix Tiburcy
- Department of Animal Physiology, University of Osnabrück, Osnabrück, Germany
| | - Mikio Furuse
- Division of Cell Structure, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Japan.,Department of Physiological Sciences, School of Life Science, Sokendai, The Graduate University for Advanced Studies, Okazaki, Japan
| | - Yasushi Izumi
- Division of Cell Structure, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Japan.,Department of Physiological Sciences, School of Life Science, Sokendai, The Graduate University for Advanced Studies, Okazaki, Japan
| | - Heiko Meyer
- Department of Zoology/Developmental Biology, University of Osnabrück, Osnabrück, Germany
| | - Sima Jonusaite
- Division of Nephrology and Hypertension, Department of Internal Medicine, Molecular Medicine Program, University of Utah, Salt Lake City, Utah.,Department of Integrative Biology, University of Guelph, Guelph, Ontario, Canada
| | - Aylin R Rodan
- Division of Nephrology and Hypertension, Department of Internal Medicine, Molecular Medicine Program, University of Utah, Salt Lake City, Utah
| | - Achim Paululat
- Department of Zoology/Developmental Biology, University of Osnabrück, Osnabrück, Germany
| |
Collapse
|
16
|
Umeda R, Satouh Y, Takemoto M, Nakada-Nakura Y, Liu K, Yokoyama T, Shirouzu M, Iwata S, Nomura N, Sato K, Ikawa M, Nishizawa T, Nureki O. Structural insights into tetraspanin CD9 function. Nat Commun 2020; 11:1606. [PMID: 32231207 PMCID: PMC7105497 DOI: 10.1038/s41467-020-15459-7] [Citation(s) in RCA: 126] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 03/06/2020] [Indexed: 01/16/2023] Open
Abstract
Tetraspanins play critical roles in various physiological processes, ranging from cell adhesion to virus infection. The members of the tetraspanin family have four membrane-spanning domains and short and large extracellular loops, and associate with a broad range of other functional proteins to exert cellular functions. Here we report the crystal structure of CD9 and the cryo-electron microscopic structure of CD9 in complex with its single membrane-spanning partner protein, EWI-2. The reversed cone-like molecular shape of CD9 generates membrane curvature in the crystalline lipid layers, which explains the CD9 localization in regions with high membrane curvature and its implications in membrane remodeling. The molecular interaction between CD9 and EWI-2 is mainly mediated through the small residues in the transmembrane region and protein/lipid interactions, whereas the fertilization assay revealed the critical involvement of the LEL region in the sperm-egg fusion, indicating the different dependency of each binding domain for other partner proteins.
Collapse
Affiliation(s)
- Rie Umeda
- Department of Biological Sciences Graduate School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, Japan
| | - Yuhkoh Satouh
- Laboratory of Molecular Traffic, Institute for Molecular and Cellular Regulation (IMCR), Gunma University, Maebashi, 371-8512, Japan
- Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka, Japan
| | - Mizuki Takemoto
- Department of Biological Sciences Graduate School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, Japan
- Preferred Networks, Inc., Bunkyo-ku, Tokyo, Japan
| | - Yoshiko Nakada-Nakura
- Department of Cell Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kehong Liu
- Department of Cell Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Takeshi Yokoyama
- Laboratory for Protein Functional and Structural Biology, RIKEN Center for Biosystems Dynamics Research, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa, 230-0045, Japan
| | - Mikako Shirouzu
- Laboratory for Protein Functional and Structural Biology, RIKEN Center for Biosystems Dynamics Research, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa, 230-0045, Japan
| | - So Iwata
- Department of Cell Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- RIKEN SPring-8 Center, 1-1-1 Kouto, Sayo-cho, Sayo-gun, Hyogo, Japan
| | - Norimichi Nomura
- Department of Cell Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Ken Sato
- Laboratory of Molecular Traffic, Institute for Molecular and Cellular Regulation (IMCR), Gunma University, Maebashi, 371-8512, Japan
- Gunma University Initiative for Advanced Research (GIAR), Gunma University, Maebashi, 371-8512, Japan
| | - Masahito Ikawa
- Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka, Japan
| | - Tomohiro Nishizawa
- Department of Biological Sciences Graduate School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, Japan.
- Precursory Research for Embryonic Science and Technology (PRESTO), Japan Science and Technology, Bunkyo-ku, Tokyo, Japan.
| | - Osamu Nureki
- Department of Biological Sciences Graduate School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, Japan.
| |
Collapse
|
17
|
Cao L, Wu X, Bai Y, Wu X, Gu S. Anti-inflammatory and antioxidant activities of probiotic powder containing Lactobacillus plantarum 1.2567 in necrotic enteritis model of broiler chickens. Livest Sci 2019. [DOI: 10.1016/j.livsci.2019.03.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
18
|
Dahmane S, Doucet C, Le Gall A, Chamontin C, Dosset P, Murcy F, Fernandez L, Salas D, Rubinstein E, Mougel M, Nollmann M, Milhiet PE. Nanoscale organization of tetraspanins during HIV-1 budding by correlative dSTORM/AFM. NANOSCALE 2019; 11:6036-6044. [PMID: 30869094 DOI: 10.1039/c8nr07269h] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
Membrane partition and remodeling play a key role in numerous cell mechanisms, especially in viral replication cycles where viruses subvert the plasma membrane to enter and escape from the host cell. Specifically assembly and release of HIV-1 particles require specific cellular components, which are recruited to the egress site by the viral protein Gag. We previously demonstrated that HIV-1 assembly alters both partitioning and dynamics of the tetraspanins CD9 and CD81, which are key players in many infectious processes, forming enriched areas where the virus buds. In this study we correlated super resolution microscopy mapping of tetraspanins with membrane topography delineated by atomic force microscopy (AFM) in Gag-expressing cells. We revealed that CD9 is specifically trapped within the nascent viral particles, especially at buds tips, suggesting that Gag mediates CD9 and CD81 depletion from the plasma membrane. In addition, we showed that CD9 is organized as small membrane assemblies of few tens of nanometers that can coalesce upon Gag expression.
Collapse
Affiliation(s)
- Selma Dahmane
- Centre de Biochimie Structurale (CBS), INSERM, CNRS, Univ Montpellier, France.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Jankovicova J, Secova P, Manaskova-Postlerova P, Simonik O, Frolikova M, Chmelikova E, Horovska L, Michalkova K, Dvorakova-Hortova K, Antalikova J. Detection of CD9 and CD81 tetraspanins in bovine and porcine oocytes and embryos. Int J Biol Macromol 2018; 123:931-938. [PMID: 30452988 DOI: 10.1016/j.ijbiomac.2018.11.161] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Revised: 11/15/2018] [Accepted: 11/16/2018] [Indexed: 12/11/2022]
Abstract
Tetraspanins are multifunctional molecules located in specific microdomains on the plasma membrane. Thanks to their ability to form networks with other proteins they can participate in many cellular functions. Tetraspanins are part of the interactive network in gametes; however, their precise role in fertilization is not yet clear. The aim of this study was to compare the localization of CD9 and CD81 tetraspanins during oocyte maturation and early development of the embryos in bovine and porcine model. CD9 was detected on the oocyte plasma membrane and vesicles in the perivitelline space of bovine oocytes and embryos. We suggest that CD9 could be a component involved in transzonal projections. Based on the results of in vitro fertilization assay, CD9 and CD81 seem to be part of a more complex fusion network on the plasma membrane of bovine oocytes. On the other hand, both tetraspanins showed a clustered expression pattern on the plasma membrane and inner margin of zona pellucida (ZP) in porcine oocytes and embryos. We found a new species-specific pattern of CD9 and CD81 distribution in ZP which could reflect their specialized role in processes associated with cell adhesion and intercellular communication upon fertilization.
Collapse
Affiliation(s)
- Jana Jankovicova
- Laboratory of Reproductive Physiology, Institute of Animal Biochemistry and Genetics, Centre of Biosciences, Slovak Academy of Sciences, Bratislava, Slovak Republic
| | - Petra Secova
- Laboratory of Reproductive Physiology, Institute of Animal Biochemistry and Genetics, Centre of Biosciences, Slovak Academy of Sciences, Bratislava, Slovak Republic
| | - Pavla Manaskova-Postlerova
- Laboratory of Reproductive Biology, Institute of Biotechnology CAS, v.v.i., BIOCEV, Vestec, Czech Republic; Department of Veterinary Sciences, Faculty of Agrobiology, Food and Natural Resources, Czech University of Life Sciences Prague, Czech Republic
| | - Ondrej Simonik
- Laboratory of Reproductive Biology, Institute of Biotechnology CAS, v.v.i., BIOCEV, Vestec, Czech Republic; Department of Veterinary Sciences, Faculty of Agrobiology, Food and Natural Resources, Czech University of Life Sciences Prague, Czech Republic
| | - Michaela Frolikova
- Laboratory of Reproductive Biology, Institute of Biotechnology CAS, v.v.i., BIOCEV, Vestec, Czech Republic
| | - Eva Chmelikova
- Department of Veterinary Sciences, Faculty of Agrobiology, Food and Natural Resources, Czech University of Life Sciences Prague, Czech Republic
| | - Lubica Horovska
- Laboratory of Reproductive Physiology, Institute of Animal Biochemistry and Genetics, Centre of Biosciences, Slovak Academy of Sciences, Bratislava, Slovak Republic
| | - Katarina Michalkova
- Laboratory of Reproductive Physiology, Institute of Animal Biochemistry and Genetics, Centre of Biosciences, Slovak Academy of Sciences, Bratislava, Slovak Republic
| | - Katerina Dvorakova-Hortova
- Laboratory of Reproductive Biology, Institute of Biotechnology CAS, v.v.i., BIOCEV, Vestec, Czech Republic; Department of Zoology, Faculty of Science, Charles University, Prague, Czech Republic.
| | - Jana Antalikova
- Laboratory of Reproductive Physiology, Institute of Animal Biochemistry and Genetics, Centre of Biosciences, Slovak Academy of Sciences, Bratislava, Slovak Republic.
| |
Collapse
|
20
|
Huang C, Fu C, Wren JD, Wang X, Zhang F, Zhang YH, Connel SA, Chen T, Zhang XA. Tetraspanin-enriched microdomains regulate digitation junctions. Cell Mol Life Sci 2018; 75:3423-3439. [PMID: 29589089 PMCID: PMC6615572 DOI: 10.1007/s00018-018-2803-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 02/18/2018] [Accepted: 03/21/2018] [Indexed: 12/22/2022]
Abstract
Tetraspanins co-emerged with multi-cellular organisms during evolution and are typically localized at the cell–cell interface, [corrected] and form tetraspanin-enriched microdomains (TEMs) by associating with each other and other membrane molecules. Tetraspanins affect various biological functions, but how tetraspanins engage in multi-faceted functions at the cellular level is largely unknown. When cells interact, the membrane microextrusions at the cell-cell interfaces form dynamic, digit-like structures between cells, which we term digitation junctions (DJs). We found that (1) tetraspanins CD9, CD81, and CD82 and (2) TEM-associated molecules integrin α3β1, CD44, EWI2/PGRL, and PI-4P are present in DJs of epithelial, endothelial, and cancer cells. Tetraspanins and their associated molecules also regulate the formation and development of DJs. Moreover, (1) actin cytoskeleton, RhoA, and actomyosin activities and (2) growth factor receptor-Src-MAP kinase signaling, but not PI-3 kinase, regulate DJs. Finally, we showed that DJs consist of various forms in different cells. Thus, DJs are common, interactive structures between cells, and likely affect cell adhesion, migration, and communication. TEMs probably modulate various cell functions through DJs. Our findings highlight that DJ morphogenesis reflects the transition between cell-matrix adhesion and cell-cell adhesion and involves both cell-cell and cell-matrix adhesion molecules.
Collapse
Affiliation(s)
- Chao Huang
- Stephenson Cancer Center and Department of Physiology, University of Oklahoma Health Sciences Center, BRC Building West Room 1474, 975 N.E. 10th Street, Oklahoma City, OK, 73104, USA
| | - Chenying Fu
- Stephenson Cancer Center and Department of Physiology, University of Oklahoma Health Sciences Center, BRC Building West Room 1474, 975 N.E. 10th Street, Oklahoma City, OK, 73104, USA
| | - Jonathan D Wren
- Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Xuejun Wang
- Stephenson Cancer Center and Department of Physiology, University of Oklahoma Health Sciences Center, BRC Building West Room 1474, 975 N.E. 10th Street, Oklahoma City, OK, 73104, USA
| | - Feng Zhang
- Stephenson Cancer Center and Department of Physiology, University of Oklahoma Health Sciences Center, BRC Building West Room 1474, 975 N.E. 10th Street, Oklahoma City, OK, 73104, USA
| | - Yanhui H Zhang
- University of Tennessee Health Science Center, Memphis, TN, USA
| | | | - Taosheng Chen
- St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Xin A Zhang
- Stephenson Cancer Center and Department of Physiology, University of Oklahoma Health Sciences Center, BRC Building West Room 1474, 975 N.E. 10th Street, Oklahoma City, OK, 73104, USA.
| |
Collapse
|
21
|
Jin X, Dimitriadis EK, Liu Y, Combs CA, Chang J, Varsano N, Stempinski E, Flores R, Jackson SN, Muller L, Woods AS, Addadi L, Kruth HS. Macrophages Shed Excess Cholesterol in Unique Extracellular Structures Containing Cholesterol Microdomains. Arterioscler Thromb Vasc Biol 2018; 38:1504-1518. [PMID: 29853567 PMCID: PMC6023747 DOI: 10.1161/atvbaha.118.311269] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 05/16/2018] [Indexed: 12/20/2022]
Abstract
Supplemental Digital Content is available in the text. Objective— Cells use various mechanisms to maintain cellular cholesterol homeostasis including efflux of cholesterol from the cellular plasma membrane to cholesterol acceptors such as HDLs (high-density lipoproteins). Little is known about the transfer of cholesterol from cells into the extracellular matrix. Using a unique monoclonal antibody that detects ordered cholesterol arrays (ie, cholesterol micro[or nano]-domains), we previously identified that particles containing these cholesterol domains accumulate in the extracellular matrix during cholesterol enrichment of human monocyte-derived macrophages and are found in atherosclerotic lesions. In this study, we further investigate these deposited particles containing cholesterol microdomains and discover their unexpected morphology. Approach and Results— Although appearing spherical at the resolution of the conventional fluorescence microscope, super-resolution immunofluorescence and atomic force microscopy of in situ cholesterol microdomains, and immunoelectron microscopy of isolated cholesterol microdomains revealed that the microdomains are not vesicles or 3-dimensional crystals but rather appear as branching irregularly shaped deposits of varying size. These cholesterol microdomain-containing deposits are shed from the plasma membrane into the extracellular matrix. Conclusions— To date, research on cellular excretion of excess cholesterol has demonstrated cellular cholesterol efflux in the form of membranous vesicles and discoidal HDL particles released into the fluid-phase medium. Shedding of plasma membrane cholesterol microdomains provides an additional mechanism for cells such as macrophages to maintain plasma membrane cholesterol homeostasis. Furthermore, recognition that macrophages shed cholesterol microdomains into the extracellular matrix is important to our understanding of extracellular buildup of cholesterol in atherosclerosis.
Collapse
Affiliation(s)
- Xueting Jin
- From the Experimental Atherosclerosis Section, National Heart, Lung, and Blood Institute (X.J., Y.L., J.C., R.F., H.S.K.)
| | - Emilios K Dimitriadis
- Scanning Probe Microscopy Unit, National Institute of Biomedical Imaging and Bioengineering (E.K.D.)
| | - Ying Liu
- From the Experimental Atherosclerosis Section, National Heart, Lung, and Blood Institute (X.J., Y.L., J.C., R.F., H.S.K.)
| | - Christian A Combs
- Light Microscopy Core, National Heart, Lung, and Blood Institute (C.A.C.)
| | - Janet Chang
- From the Experimental Atherosclerosis Section, National Heart, Lung, and Blood Institute (X.J., Y.L., J.C., R.F., H.S.K.)
| | - Neta Varsano
- Department of Structural Biology, Weizmann Institute of Science, Rehovot, Israel (N.V., L.A.)
| | - Erin Stempinski
- Electron Microscopy Core, National Heart, Lung, and Blood Institute (E.S.)
| | - Rhonda Flores
- From the Experimental Atherosclerosis Section, National Heart, Lung, and Blood Institute (X.J., Y.L., J.C., R.F., H.S.K.)
| | - Shelley N Jackson
- Structural Biology Core, National Institute of Drug Abuse (S.N.J., L.M., A.S.W.), National Institutes of Health, Baltimore, MD
| | - Ludovic Muller
- Structural Biology Core, National Institute of Drug Abuse (S.N.J., L.M., A.S.W.), National Institutes of Health, Baltimore, MD
| | - Amina S Woods
- Structural Biology Core, National Institute of Drug Abuse (S.N.J., L.M., A.S.W.), National Institutes of Health, Baltimore, MD
| | - Lia Addadi
- Department of Structural Biology, Weizmann Institute of Science, Rehovot, Israel (N.V., L.A.)
| | - Howard S Kruth
- From the Experimental Atherosclerosis Section, National Heart, Lung, and Blood Institute (X.J., Y.L., J.C., R.F., H.S.K.)
| |
Collapse
|
22
|
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
|
23
|
Genome-Wide Screen for New Components of the Drosophila melanogaster Torso Receptor Tyrosine Kinase Pathway. G3-GENES GENOMES GENETICS 2018; 8:761-769. [PMID: 29363515 PMCID: PMC5844297 DOI: 10.1534/g3.117.300491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Patterning of the Drosophila embryonic termini by the Torso (Tor) receptor pathway has long served as a valuable paradigm for understanding how receptor tyrosine kinase signaling is controlled. However, the mechanisms that underpin the control of Tor signaling remain to be fully understood. In particular, it is unclear how the Perforin-like protein Torso-like (Tsl) localizes Tor activity to the embryonic termini. To shed light on this, together with other aspects of Tor pathway function, we conducted a genome-wide screen to identify new pathway components that operate downstream of Tsl. Using a set of molecularly defined chromosomal deficiencies, we screened for suppressors of ligand-dependent Tor signaling induced by unrestricted Tsl expression. This approach yielded 59 genomic suppressor regions, 11 of which we mapped to the causative gene, and a further 29 that were mapped to <15 genes. Of the identified genes, six represent previously unknown regulators of embryonic Tor signaling. These include twins (tws), which encodes an integral subunit of the protein phosphatase 2A complex, and α-tubulin at 84B (αTub84B), a major constituent of the microtubule network, suggesting that these may play an important part in terminal patterning. Together, these data comprise a valuable resource for the discovery of new Tor pathway components. Many of these may also be required for other roles of Tor in development, such as in the larval prothoracic gland where Tor signaling controls the initiation of metamorphosis.
Collapse
|
24
|
How tetraspanins shape endothelial and leukocyte nano-architecture during inflammation. Biochem Soc Trans 2017; 45:999-1006. [PMID: 28710286 DOI: 10.1042/bst20170163] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2017] [Revised: 06/07/2017] [Accepted: 06/09/2017] [Indexed: 01/13/2023]
Abstract
Tetraspanins are ubiquitous membrane proteins that induce local membrane curvature and hence co-ordinate cell-to-cell contacts. This review highlights their role in inflammation, which requires control of the nano-architecture of attachment sites between endothelial cells and leukocytes. The active role of endothelial cells in preparing for transmigration of leukocytes and determining the severity of an inflammation is often underscored. A clear hint to endothelial pre-activation is their ability to protrude clustered adhesion proteins upward prior to leukocyte contact. The elevation of molecular adhesive platforms toward the blood stream is crucially dependent on tetraspanins. In addition, leukocytes require tetraspanins for their activation. The example of the B-cell receptor is referenced in some detail here, since it provides deeper insights into the receptor-coreceptor interplay. To lift the role of tetraspanins from an abstract model of inflammation toward a player of clinical significance, two pathologies are analyzed for the known contributions of tetraspanins. The recent publication of the first crystal structure of a full-length tetraspanin revealed a cholesterol-binding site, which provides a strong link to the pathophysiological condition of atherosclerosis. Dysregulation of the inflammatory cascade in autoimmune diseases by endothelial cells is exemplified by the involvement of tetraspanins in multiple sclerosis.
Collapse
|
25
|
Oligomerization of the Tetraspanin CD81 via the Flexibility of Its δ-Loop. Biophys J 2017; 110:2463-2474. [PMID: 27276264 DOI: 10.1016/j.bpj.2016.05.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Revised: 05/03/2016] [Accepted: 05/04/2016] [Indexed: 01/28/2023] Open
Abstract
Tetraspanins are master organizers in the plasma membrane, forming tetraspanin-enriched microdomains with one another and other surface molecules. Their rod-shaped structure includes a large extracellular loop (LEL) that plays a pivotal role in tetraspanin network formation. We performed comparative atomistic and coarse-grain molecular-dynamics simulations of the LEL in isolation and full-length CD81, and reproduced LEL flexibility patterns known from wet-lab experiments in which the LEL δ-loop region showed a pronounced flexibility. In a 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphocholine lipid bilayer and a plasma membrane environment, the conformational flexibility of the δ-loop initiates CD81-CD81 contacts for oligomerization. Furthermore, in the plasma membrane, CD81-ganglioside bridges arising from preformed glycolipid patches cross-link the complexes. The data suggest that exposing a flexible domain enables binding to interaction partners by circumventing the restriction of orientation and conformational freedom of membrane proteins.
Collapse
|
26
|
Joint features and complementarities of Tspan8 and CD151 revealed in knockdown and knockout models. Biochem Soc Trans 2017; 45:437-447. [PMID: 28408484 DOI: 10.1042/bst20160298] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Revised: 02/04/2017] [Accepted: 02/13/2017] [Indexed: 02/06/2023]
Abstract
Tetraspanins are highly conserved 4-transmembrane proteins which form molecular clusters with a large variety of transmembrane and cytosolic proteins. By these associations tetraspanins are engaged in a multitude of biological processes. Furthermore, tetraspanin complexes are located in specialized microdomains, called tetraspanin-enriched microdomains (TEMs). TEMs provide a signaling platform and are poised for invagination and vesicle formation. These vesicles can be released as exosomes (Exo) and are important in cell contact-independent intercellular communication. Here, we summarize emphasizing knockdown and knockout models' pathophysiological joint and selective activities of CD151 and Tspan8, and discuss the TEM-related engagement of CD151 and Tspan8 in Exo activities.
Collapse
|
27
|
Gaibelet G, Tercé F, Allart S, Lebrun C, Collet X, Jamin N, Orlowski S. Fluorescent probes for detecting cholesterol-rich ordered membrane microdomains: entangled relationships between structural analogies in the membrane and functional homologies in the cell. AIMS BIOPHYSICS 2017. [DOI: 10.3934/biophy.2017.1.121] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
|
28
|
Abstract
Stem cells are critical to maintaining steady-state organ homeostasis and regenerating injured tissues. Recent intriguing reports implicate extracellular vesicles (EVs) as carriers for the distribution of morphogens and growth and differentiation factors from tissue parenchymal cells to stem cells, and conversely, stem cell-derived EVs carrying certain proteins and nucleic acids can support healing of injured tissues. We describe approaches to make use of engineered EVs as technology platforms in therapeutics and diagnostics in the context of stem cells. For some regenerative therapies, natural and engineered EVs from stem cells may be superior to single-molecule drugs, biologics, whole cells, and synthetic liposome or nanoparticle formulations because of the ease of bioengineering with multiple factors while retaining superior biocompatibility and biostability and posing fewer risks for abnormal differentiation or neoplastic transformation. Finally, we provide an overview of current challenges and future directions of EVs as potential therapeutic alternatives to cells for clinical applications.
Collapse
Affiliation(s)
- Milad Riazifar
- Department of Pharmaceutical Sciences, University of California, Irvine, California 92697; .,Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, California 92697.,Chao Family Comprehensive Cancer Center, University of California, Irvine, Orange, California 92868.,Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California, Irvine, California 92697.,Department of Biomedical Engineering, University of California, Irvine, California 92697.,Department of Biological Chemistry, University of California, Irvine, California 92697
| | - Egest J Pone
- Department of Pharmaceutical Sciences, University of California, Irvine, California 92697; .,Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, California 92697.,Chao Family Comprehensive Cancer Center, University of California, Irvine, Orange, California 92868.,Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California, Irvine, California 92697.,Department of Biomedical Engineering, University of California, Irvine, California 92697.,Department of Biological Chemistry, University of California, Irvine, California 92697
| | - Jan Lötvall
- Krefting Research Centre, Institute of Medicine, The Sahlgrenska Academy, Göteborg University, SE-405 30 Göteborg, Sweden.,Codiak BioSciences Inc., Woburn, Massachusetts 01801
| | - Weian Zhao
- Department of Pharmaceutical Sciences, University of California, Irvine, California 92697; .,Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, California 92697.,Chao Family Comprehensive Cancer Center, University of California, Irvine, Orange, California 92868.,Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California, Irvine, California 92697.,Department of Biomedical Engineering, University of California, Irvine, California 92697.,Department of Biological Chemistry, University of California, Irvine, California 92697
| |
Collapse
|
29
|
Feng J, Huang C, Wren JD, Wang DW, Yan J, Zhang J, Sun Y, Han X, Zhang XA. Tetraspanin CD82: a suppressor of solid tumors and a modulator of membrane heterogeneity. Cancer Metastasis Rev 2016; 34:619-33. [PMID: 26335499 DOI: 10.1007/s10555-015-9585-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Tetraspanin CD82 suppresses the progression and metastasis of a wide range of solid malignant tumors. However, its roles in tumorigenesis and hematopoietic malignancy remain unclear. Ubiquitously expressed CD82 restrains cell migration and cell invasion by modulating both cell-matrix and cell-cell adhesiveness and confining outside-in pro-motility signaling. This restraint at least contributes to, if not determines, the metastasis-suppressive activity and, also likely, the physiological functions of CD82. As a modulator of cell membrane heterogeneity, CD82 alters microdomains, trafficking, and topography of the membrane by changing the membrane molecular landscape. The functional activities of membrane molecules and the cytoskeletal interaction of the cell membrane are subsequently altered, followed by changes in cellular functions. Given its pathological and physiological importance, CD82 is a promising candidate for clinically predicting and blocking tumor progression and metastasis and also an emerging model protein for mechanistically understanding cell membrane organization and heterogeneity.
Collapse
Affiliation(s)
- Jin Feng
- Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Chao Huang
- Stephenson Cancer Center and Department of Physiology, University of Oklahoma Health Sciences Center, BRC 1474, 975 NE 10th Street, Oklahoma City, OK, 73104, USA
| | - Jonathan D Wren
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Dao-Wen Wang
- Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Jizhou Yan
- Institute for Marine Biosystem and Neurosciences, Shanghai Ocean University, Shanghai, China
| | - Jiexin Zhang
- Department of Biochemistry, Nanjing Medical University, Nanjing, China
| | - Yujie Sun
- Department of Biochemistry, Nanjing Medical University, Nanjing, China
| | - Xiao Han
- Department of Biochemistry, Nanjing Medical University, Nanjing, China
| | - Xin A Zhang
- Stephenson Cancer Center and Department of Physiology, University of Oklahoma Health Sciences Center, BRC 1474, 975 NE 10th Street, Oklahoma City, OK, 73104, USA.
| |
Collapse
|
30
|
Heiler S, Wang Z, Zöller M. Pancreatic cancer stem cell markers and exosomes - the incentive push. World J Gastroenterol 2016; 22:5971-6007. [PMID: 27468191 PMCID: PMC4948278 DOI: 10.3748/wjg.v22.i26.5971] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2016] [Revised: 06/03/2016] [Accepted: 06/28/2016] [Indexed: 02/06/2023] Open
Abstract
Pancreatic cancer (PaCa) has the highest death rate and incidence is increasing. Poor prognosis is due to late diagnosis and early metastatic spread, which is ascribed to a minor population of so called cancer stem cells (CSC) within the mass of the primary tumor. CSC are defined by biological features, which they share with adult stem cells like longevity, rare cell division, the capacity for self renewal, differentiation, drug resistance and the requirement for a niche. CSC can also be identified by sets of markers, which for pancreatic CSC (Pa-CSC) include CD44v6, c-Met, Tspan8, alpha6beta4, CXCR4, CD133, EpCAM and claudin7. The functional relevance of CSC markers is still disputed. We hypothesize that Pa-CSC markers play a decisive role in tumor progression. This is fostered by the location in glycolipid-enriched membrane domains, which function as signaling platform and support connectivity of the individual Pa-CSC markers. Outside-in signaling supports apoptosis resistance, stem cell gene expression and tumor suppressor gene repression as well as miRNA transcription and silencing. Pa-CSC markers also contribute to motility and invasiveness. By ligand binding host cells are triggered towards creating a milieu supporting Pa-CSC maintenance. Furthermore, CSC markers contribute to the generation, loading and delivery of exosomes, whereby CSC gain the capacity for a cell-cell contact independent crosstalk with the host and neighboring non-CSC. This allows Pa-CSC exosomes (TEX) to reprogram neighboring non-CSC towards epithelial mesenchymal transition and to stimulate host cells towards preparing a niche for metastasizing tumor cells. Finally, TEX communicate with the matrix to support tumor cell motility, invasion and homing. We will discuss the possibility that CSC markers are the initial trigger for these processes and what is the special contribution of CSC-TEX.
Collapse
|
31
|
Yoon JC, Yang CM, Song Y, Lee JM. Natural killer cells in hepatitis C: Current progress. World J Gastroenterol 2016; 22:1449-1460. [PMID: 26819513 PMCID: PMC4721979 DOI: 10.3748/wjg.v22.i4.1449] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Revised: 09/30/2015] [Accepted: 12/01/2015] [Indexed: 02/06/2023] Open
Abstract
Patients infected with the hepatitis C virus (HCV) are characterized by a high incidence of chronic infection, which results in chronic hepatitis, liver cirrhosis, and hepatocellular carcinoma. The functional impairment of HCV-specific T cells is associated with the evolution of an acute infection to chronic hepatitis. While T cells are the important effector cells in adaptive immunity, natural killer (NK) cells are the critical effector cells in innate immunity to virus infections. The findings of recent studies on NK cells in hepatitis C suggest that NK cell responses are indeed important in each phase of HCV infection. In the early phase, NK cells are involved in protective immunity to HCV. The immune evasion strategies used by HCV may target NK cells and might contribute to the progression to chronic hepatitis C. NK cells may control HCV replication and modulate hepatic fibrosis in the chronic phase. Further investigations are, however, needed, because a considerable number of studies observed functional impairment of NK cells in chronic HCV infection. Interestingly, the enhanced NK cell responses during interferon-α-based therapy of chronic hepatitis C indicate successful treatment. In spite of the advances in research on NK cells in hepatitis C, establishment of more physiological HCV infection model systems is needed to settle unsolved controversies over the role and functional status of NK cells in HCV infection.
Collapse
|
32
|
Carquin M, D'Auria L, Pollet H, Bongarzone ER, Tyteca D. Recent progress on lipid lateral heterogeneity in plasma membranes: From rafts to submicrometric domains. Prog Lipid Res 2015; 62:1-24. [PMID: 26738447 DOI: 10.1016/j.plipres.2015.12.004] [Citation(s) in RCA: 103] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Revised: 12/22/2015] [Accepted: 12/22/2015] [Indexed: 12/29/2022]
Abstract
The concept of transient nanometric domains known as lipid rafts has brought interest to reassess the validity of the Singer-Nicolson model of a fluid bilayer for cell membranes. However, this new view is still insufficient to explain the cellular control of surface lipid diversity or membrane deformability. During the past decades, the hypothesis that some lipids form large (submicrometric/mesoscale vs nanometric rafts) and stable (>min vs s) membrane domains has emerged, largely based on indirect methods. Morphological evidence for stable submicrometric lipid domains, well-accepted for artificial and highly specialized biological membranes, was further reported for a variety of living cells from prokaryot es to yeast and mammalian cells. However, results remained questioned based on limitations of available fluorescent tools, use of poor lipid fixatives, and imaging artifacts due to non-resolved membrane projections. In this review, we will discuss recent evidence generated using powerful and innovative approaches such as lipid-specific toxin fragments that support the existence of submicrometric domains. We will integrate documented mechanisms involved in the formation and maintenance of these domains, and provide a perspective on their relevance on membrane deformability and regulation of membrane protein distribution.
Collapse
Affiliation(s)
- Mélanie Carquin
- CELL Unit, de Duve Institute & Université Catholique de Louvain, UCL B1.75.05, Avenue Hippocrate, 75, B-1200 Brussels, Belgium
| | - Ludovic D'Auria
- The Myelin Regeneration Group at the Dept. Anatomy & Cell Biology, College of Medicine, University of Illinois, 808 S. Wood St. MC512, Chicago, IL. 60612. USA
| | - Hélène Pollet
- CELL Unit, de Duve Institute & Université Catholique de Louvain, UCL B1.75.05, Avenue Hippocrate, 75, B-1200 Brussels, Belgium
| | - Ernesto R Bongarzone
- The Myelin Regeneration Group at the Dept. Anatomy & Cell Biology, College of Medicine, University of Illinois, 808 S. Wood St. MC512, Chicago, IL. 60612. USA
| | - Donatienne Tyteca
- CELL Unit, de Duve Institute & Université Catholique de Louvain, UCL B1.75.05, Avenue Hippocrate, 75, B-1200 Brussels, Belgium.
| |
Collapse
|
33
|
Homsi Y, Schloetel JG, Scheffer KD, Schmidt TH, Destainville N, Florin L, Lang T. The extracellular δ-domain is essential for the formation of CD81 tetraspanin webs. Biophys J 2015; 107:100-13. [PMID: 24988345 DOI: 10.1016/j.bpj.2014.05.028] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2013] [Revised: 05/14/2014] [Accepted: 05/20/2014] [Indexed: 12/15/2022] Open
Abstract
CD81 is a ubiquitously expressed member of the tetraspanin family. It forms large molecular platforms, so-called tetraspanin webs that play physiological roles in a variety of cellular functions and are involved in viral and parasite infections. We have investigated which part of the CD81 molecule is required for the formation of domains in the cell membranes of T-cells and hepatocytes. Surprisingly, we find that large CD81 platforms assemble via the short extracellular δ-domain, independent from a strong primary partner binding and from weak interactions mediated by palmitoylation. The δ-domain is also essential for the platforms to function during viral entry. We propose that, instead of stable binary interactions, CD81 interactions via the small δ-domain, possibly involving a dimerization step, play the key role in organizing CD81 into large tetraspanin webs and controlling its function.
Collapse
Affiliation(s)
- Yahya Homsi
- Department of Membrane Biochemistry, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Jan-Gero Schloetel
- Department of Membrane Biochemistry, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Konstanze D Scheffer
- Department of Medical Microbiology and Hygiene, University Medical Centre of the Johannes Gutenberg University, Mainz, Germany
| | - Thomas H Schmidt
- Department of Membrane Biochemistry, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Nicolas Destainville
- Université Toulouse 3-Paul Sabatier, UPS, Laboratoire de Physique Théorique (IRSAMC), Toulouse, France
| | - Luise Florin
- Department of Medical Microbiology and Hygiene, University Medical Centre of the Johannes Gutenberg University, Mainz, Germany
| | - Thorsten Lang
- Department of Membrane Biochemistry, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany.
| |
Collapse
|
34
|
Yamada M, Sekiguchi K. Molecular Basis of Laminin-Integrin Interactions. CURRENT TOPICS IN MEMBRANES 2015; 76:197-229. [PMID: 26610915 DOI: 10.1016/bs.ctm.2015.07.002] [Citation(s) in RCA: 96] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Laminins are composed of three polypeptide chains, designated as α, β, and γ. The C-terminal region of laminin heterotrimers, containing coiled-coil regions, short tails, and laminin globular (LG) domains, is necessary and sufficient for binding to integrins, which are the major laminin receptor class. Laminin recognition by integrins critically requires the α chain LG domains and a glutamic acid residue of the γ chain at the third position from the C-terminus. Furthermore, the C-terminal region of the β chain contains a short amino acid sequence that modulates laminin affinity for integrins. Thus, all three of the laminin chains act cooperatively to facilitate integrin binding. Mammals possess 5 α (α1-5), 3 β (β1-3), and 3 γ (γ1-3) chains, combinations of which give rise to 16 distinct laminin isoforms. Each isoform is expressed in a tissue-specific and developmental stage-specific manner, exerting its functions through binding of integrins. In this review, we detail the current knowledge surrounding the molecular basis and physiological relevance of specific interactions between laminins and integrins, and describe the mechanisms underlying laminin action through integrins.
Collapse
Affiliation(s)
- Masashi Yamada
- Laboratory of Extracellular Matrix Biochemistry, Institute for Protein Research, Osaka University, Suita, Osaka, Japan
| | - Kiyotoshi Sekiguchi
- Laboratory of Extracellular Matrix Biochemistry, Institute for Protein Research, Osaka University, Suita, Osaka, Japan
| |
Collapse
|
35
|
Fox RM, Andrew DJ. Changes in organelle position and epithelial architecture associated with loss of CrebA. Biol Open 2015; 4:317-30. [PMID: 25681391 PMCID: PMC4359738 DOI: 10.1242/bio.201411205] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Drosophila CrebA facilitates high-level secretion by transcriptional upregulation of the protein components of the core secretory machinery. In CrebA mutant embryos, both salivary gland (SG) morphology and epidermal cuticle secretion are abnormal, phenotypes similar to those observed with mutations in core secretory pathway component genes. Here, we examine the cellular defects associated with CrebA loss in the SG epithelium. Apically localized secretory vesicles are smaller and less abundant, consistent with overall reductions in secretion. Unexpectedly, global mislocalization of cellular organelles and excess membrane accumulation in the septate junctions (SJs) are also observed. Whereas mutations in core secretory pathway genes lead to organelle localization defects similar to those of CrebA mutants, they have no effect on SJ-associated membrane. Mutations in tetraspanin genes, which are normally repressed by CrebA, have mild defects in SJ morphology that are rescued by simultaneous CrebA loss. Correspondingly, removal of several tetraspanins gives partial rescue of the CrebA SJ phenotype, supporting a role for tetraspanins in SJ organization.
Collapse
Affiliation(s)
- Rebecca M Fox
- The Department of Cell Biology, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Deborah J Andrew
- The Department of Cell Biology, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
36
|
HIV-1 Nef and Vpu are functionally redundant broad-spectrum modulators of cell surface receptors, including tetraspanins. J Virol 2014; 88:14241-57. [PMID: 25275127 DOI: 10.1128/jvi.02333-14] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
UNLABELLED HIV-1 Nef and Vpu are thought to optimize virus replication in the infected host, at least in part via their ability to interfere with vesicular host cell trafficking. Despite the use of distinct molecular mechanisms, Nef and Vpu share specificity for some molecules such as CD4 and major histocompatibility complex class I (MHC-I), while disruption of intracellular transport of the host cell restriction factor CD317/tetherin represents a specialized activity of Vpu not exerted by HIV-1 Nef. To establish a profile of host cell receptors whose intracellular transport is affected by Nef, Vpu, or both, we comprehensively analyzed the effect of these accessory viral proteins on cell surface receptor levels on A3.01 T lymphocytes. Thirty-six out of 105 detectable receptors were significantly downregulated by HIV-1 Nef, revealing a previously unappreciated scope with which HIV-1 Nef remodels the cell surface of infected cells. Remarkably, the effects of HIV-1 Vpu on host cell receptor exposure largely matched those of HIV-1 Nef in breadth and specificity (32 of 105, all also targeted by Nef), even though the magnitude was generally less pronounced. Of particular note, cell surface exposure of all members of the tetraspanin (TSPAN) protein family analyzed was reduced by both Nef and Vpu, and the viral proteins triggered the enrichment of TSPANs in a perinuclear area of the cell. While Vpu displayed significant colocalization and physical association with TSPANs, interactions of Nef with TSPANs were less robust. TSPANs thus emerge as a major target of deregulation in host cell vesicular transport by HIV-1 Nef and Vpu. The conservation of this activity in two independent accessory proteins suggests its importance for the spread of HIV-1 in the infected host. IMPORTANCE In this paper, we define that HIV-1 Nef and Vpu display a surprising functional overlap and affect the cell surface exposure of a previously unexpected breadth of cellular receptors. Our analyses furthermore identify the tetraspanin protein family as a previously unrecognized target of Nef and Vpu activity. These findings have implications for the interpretation of effects detected for these accessory gene products on individual host cell receptors and illustrate the coevolution of Nef and Vpu function.
Collapse
|
37
|
Wei Q, Zhang F, Richardson MM, Roy NH, Rodgers W, Liu Y, Zhao W, Fu C, Ding Y, Huang C, Chen Y, Sun Y, Ding L, Hu Y, Ma JX, Boulton ME, Pasula S, Wren JD, Tanaka S, Huang X, Thali M, Hämmerling GJ, Zhang XA. CD82 restrains pathological angiogenesis by altering lipid raft clustering and CD44 trafficking in endothelial cells. Circulation 2014; 130:1493-504. [PMID: 25149363 DOI: 10.1161/circulationaha.114.011096] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND Angiogenesis is crucial for many pathological processes and becomes a therapeutic strategy against diseases ranging from inflammation to cancer. The regulatory mechanism of angiogenesis remains unclear. Although tetraspanin CD82 is widely expressed in various endothelial cells (ECs), its vascular function is unknown. METHODS AND RESULTS Angiogenesis was examined in Cd82-null mice with in vivo and ex vivo morphogenesis assays. Cellular functions, molecular interactions, and signaling were analyzed in Cd82-null ECs. Angiogenic responses to various stimuli became markedly increased upon Cd82 ablation. Major changes in Cd82-null ECs were enhanced migration and invasion, likely resulting from the upregulated expression of cell adhesion molecules such as CD44 and integrins at the cell surface and subsequently elevated outside-in signaling. Gangliosides, lipid raft clustering, and CD44-membrane microdomain interactions were increased in the plasma membrane of Cd82-null ECs, leading to less clathrin-independent endocytosis and then more surface presence of CD44. CONCLUSIONS Our study reveals that CD82 restrains pathological angiogenesis by inhibiting EC movement, that lipid raft clustering and cell adhesion molecule trafficking modulate angiogenic potential, that transmembrane protein modulates lipid rafts, and that the perturbation of CD82-ganglioside-CD44 signaling attenuates pathological angiogenesis.
Collapse
Affiliation(s)
- Quan Wei
- From the West China Hospital, Sichuan University, Chengdu, China (Q.W.); University of Oklahoma Health Science Center, Oklahoma City (Q.W., F.Z., M.M.R., W.R., Y.L., C.F., Y.D., C.H., L.D., Y.H., J.M., X.A.Z.); University of Vermont, Burlington (N.H.R., M.T.); University of Tennessee, Memphis (W.Z., Y.C., Y.S.); Tongji Hospital, Wuhan, China (Y.D., X.H.); Indiana University, Indianapolis (M.E.B.); Oklahoma Medical Research Foundation, Oklahoma City (S.P., J.D.W.); and German Cancer Research Center, Heidelberg, Germany (S.T., G.J.H.)
| | - Feng Zhang
- From the West China Hospital, Sichuan University, Chengdu, China (Q.W.); University of Oklahoma Health Science Center, Oklahoma City (Q.W., F.Z., M.M.R., W.R., Y.L., C.F., Y.D., C.H., L.D., Y.H., J.M., X.A.Z.); University of Vermont, Burlington (N.H.R., M.T.); University of Tennessee, Memphis (W.Z., Y.C., Y.S.); Tongji Hospital, Wuhan, China (Y.D., X.H.); Indiana University, Indianapolis (M.E.B.); Oklahoma Medical Research Foundation, Oklahoma City (S.P., J.D.W.); and German Cancer Research Center, Heidelberg, Germany (S.T., G.J.H.)
| | - Mekel M Richardson
- From the West China Hospital, Sichuan University, Chengdu, China (Q.W.); University of Oklahoma Health Science Center, Oklahoma City (Q.W., F.Z., M.M.R., W.R., Y.L., C.F., Y.D., C.H., L.D., Y.H., J.M., X.A.Z.); University of Vermont, Burlington (N.H.R., M.T.); University of Tennessee, Memphis (W.Z., Y.C., Y.S.); Tongji Hospital, Wuhan, China (Y.D., X.H.); Indiana University, Indianapolis (M.E.B.); Oklahoma Medical Research Foundation, Oklahoma City (S.P., J.D.W.); and German Cancer Research Center, Heidelberg, Germany (S.T., G.J.H.)
| | - Nathan H Roy
- From the West China Hospital, Sichuan University, Chengdu, China (Q.W.); University of Oklahoma Health Science Center, Oklahoma City (Q.W., F.Z., M.M.R., W.R., Y.L., C.F., Y.D., C.H., L.D., Y.H., J.M., X.A.Z.); University of Vermont, Burlington (N.H.R., M.T.); University of Tennessee, Memphis (W.Z., Y.C., Y.S.); Tongji Hospital, Wuhan, China (Y.D., X.H.); Indiana University, Indianapolis (M.E.B.); Oklahoma Medical Research Foundation, Oklahoma City (S.P., J.D.W.); and German Cancer Research Center, Heidelberg, Germany (S.T., G.J.H.)
| | - William Rodgers
- From the West China Hospital, Sichuan University, Chengdu, China (Q.W.); University of Oklahoma Health Science Center, Oklahoma City (Q.W., F.Z., M.M.R., W.R., Y.L., C.F., Y.D., C.H., L.D., Y.H., J.M., X.A.Z.); University of Vermont, Burlington (N.H.R., M.T.); University of Tennessee, Memphis (W.Z., Y.C., Y.S.); Tongji Hospital, Wuhan, China (Y.D., X.H.); Indiana University, Indianapolis (M.E.B.); Oklahoma Medical Research Foundation, Oklahoma City (S.P., J.D.W.); and German Cancer Research Center, Heidelberg, Germany (S.T., G.J.H.)
| | - Yuechueng Liu
- From the West China Hospital, Sichuan University, Chengdu, China (Q.W.); University of Oklahoma Health Science Center, Oklahoma City (Q.W., F.Z., M.M.R., W.R., Y.L., C.F., Y.D., C.H., L.D., Y.H., J.M., X.A.Z.); University of Vermont, Burlington (N.H.R., M.T.); University of Tennessee, Memphis (W.Z., Y.C., Y.S.); Tongji Hospital, Wuhan, China (Y.D., X.H.); Indiana University, Indianapolis (M.E.B.); Oklahoma Medical Research Foundation, Oklahoma City (S.P., J.D.W.); and German Cancer Research Center, Heidelberg, Germany (S.T., G.J.H.)
| | - Wenyuan Zhao
- From the West China Hospital, Sichuan University, Chengdu, China (Q.W.); University of Oklahoma Health Science Center, Oklahoma City (Q.W., F.Z., M.M.R., W.R., Y.L., C.F., Y.D., C.H., L.D., Y.H., J.M., X.A.Z.); University of Vermont, Burlington (N.H.R., M.T.); University of Tennessee, Memphis (W.Z., Y.C., Y.S.); Tongji Hospital, Wuhan, China (Y.D., X.H.); Indiana University, Indianapolis (M.E.B.); Oklahoma Medical Research Foundation, Oklahoma City (S.P., J.D.W.); and German Cancer Research Center, Heidelberg, Germany (S.T., G.J.H.)
| | - Chenying Fu
- From the West China Hospital, Sichuan University, Chengdu, China (Q.W.); University of Oklahoma Health Science Center, Oklahoma City (Q.W., F.Z., M.M.R., W.R., Y.L., C.F., Y.D., C.H., L.D., Y.H., J.M., X.A.Z.); University of Vermont, Burlington (N.H.R., M.T.); University of Tennessee, Memphis (W.Z., Y.C., Y.S.); Tongji Hospital, Wuhan, China (Y.D., X.H.); Indiana University, Indianapolis (M.E.B.); Oklahoma Medical Research Foundation, Oklahoma City (S.P., J.D.W.); and German Cancer Research Center, Heidelberg, Germany (S.T., G.J.H.)
| | - Yingjun Ding
- From the West China Hospital, Sichuan University, Chengdu, China (Q.W.); University of Oklahoma Health Science Center, Oklahoma City (Q.W., F.Z., M.M.R., W.R., Y.L., C.F., Y.D., C.H., L.D., Y.H., J.M., X.A.Z.); University of Vermont, Burlington (N.H.R., M.T.); University of Tennessee, Memphis (W.Z., Y.C., Y.S.); Tongji Hospital, Wuhan, China (Y.D., X.H.); Indiana University, Indianapolis (M.E.B.); Oklahoma Medical Research Foundation, Oklahoma City (S.P., J.D.W.); and German Cancer Research Center, Heidelberg, Germany (S.T., G.J.H.)
| | - Chao Huang
- From the West China Hospital, Sichuan University, Chengdu, China (Q.W.); University of Oklahoma Health Science Center, Oklahoma City (Q.W., F.Z., M.M.R., W.R., Y.L., C.F., Y.D., C.H., L.D., Y.H., J.M., X.A.Z.); University of Vermont, Burlington (N.H.R., M.T.); University of Tennessee, Memphis (W.Z., Y.C., Y.S.); Tongji Hospital, Wuhan, China (Y.D., X.H.); Indiana University, Indianapolis (M.E.B.); Oklahoma Medical Research Foundation, Oklahoma City (S.P., J.D.W.); and German Cancer Research Center, Heidelberg, Germany (S.T., G.J.H.)
| | - Yuanjian Chen
- From the West China Hospital, Sichuan University, Chengdu, China (Q.W.); University of Oklahoma Health Science Center, Oklahoma City (Q.W., F.Z., M.M.R., W.R., Y.L., C.F., Y.D., C.H., L.D., Y.H., J.M., X.A.Z.); University of Vermont, Burlington (N.H.R., M.T.); University of Tennessee, Memphis (W.Z., Y.C., Y.S.); Tongji Hospital, Wuhan, China (Y.D., X.H.); Indiana University, Indianapolis (M.E.B.); Oklahoma Medical Research Foundation, Oklahoma City (S.P., J.D.W.); and German Cancer Research Center, Heidelberg, Germany (S.T., G.J.H.)
| | - Yao Sun
- From the West China Hospital, Sichuan University, Chengdu, China (Q.W.); University of Oklahoma Health Science Center, Oklahoma City (Q.W., F.Z., M.M.R., W.R., Y.L., C.F., Y.D., C.H., L.D., Y.H., J.M., X.A.Z.); University of Vermont, Burlington (N.H.R., M.T.); University of Tennessee, Memphis (W.Z., Y.C., Y.S.); Tongji Hospital, Wuhan, China (Y.D., X.H.); Indiana University, Indianapolis (M.E.B.); Oklahoma Medical Research Foundation, Oklahoma City (S.P., J.D.W.); and German Cancer Research Center, Heidelberg, Germany (S.T., G.J.H.)
| | - Lexi Ding
- From the West China Hospital, Sichuan University, Chengdu, China (Q.W.); University of Oklahoma Health Science Center, Oklahoma City (Q.W., F.Z., M.M.R., W.R., Y.L., C.F., Y.D., C.H., L.D., Y.H., J.M., X.A.Z.); University of Vermont, Burlington (N.H.R., M.T.); University of Tennessee, Memphis (W.Z., Y.C., Y.S.); Tongji Hospital, Wuhan, China (Y.D., X.H.); Indiana University, Indianapolis (M.E.B.); Oklahoma Medical Research Foundation, Oklahoma City (S.P., J.D.W.); and German Cancer Research Center, Heidelberg, Germany (S.T., G.J.H.)
| | - Yang Hu
- From the West China Hospital, Sichuan University, Chengdu, China (Q.W.); University of Oklahoma Health Science Center, Oklahoma City (Q.W., F.Z., M.M.R., W.R., Y.L., C.F., Y.D., C.H., L.D., Y.H., J.M., X.A.Z.); University of Vermont, Burlington (N.H.R., M.T.); University of Tennessee, Memphis (W.Z., Y.C., Y.S.); Tongji Hospital, Wuhan, China (Y.D., X.H.); Indiana University, Indianapolis (M.E.B.); Oklahoma Medical Research Foundation, Oklahoma City (S.P., J.D.W.); and German Cancer Research Center, Heidelberg, Germany (S.T., G.J.H.)
| | - Jian-Xing Ma
- From the West China Hospital, Sichuan University, Chengdu, China (Q.W.); University of Oklahoma Health Science Center, Oklahoma City (Q.W., F.Z., M.M.R., W.R., Y.L., C.F., Y.D., C.H., L.D., Y.H., J.M., X.A.Z.); University of Vermont, Burlington (N.H.R., M.T.); University of Tennessee, Memphis (W.Z., Y.C., Y.S.); Tongji Hospital, Wuhan, China (Y.D., X.H.); Indiana University, Indianapolis (M.E.B.); Oklahoma Medical Research Foundation, Oklahoma City (S.P., J.D.W.); and German Cancer Research Center, Heidelberg, Germany (S.T., G.J.H.)
| | - Michael E Boulton
- From the West China Hospital, Sichuan University, Chengdu, China (Q.W.); University of Oklahoma Health Science Center, Oklahoma City (Q.W., F.Z., M.M.R., W.R., Y.L., C.F., Y.D., C.H., L.D., Y.H., J.M., X.A.Z.); University of Vermont, Burlington (N.H.R., M.T.); University of Tennessee, Memphis (W.Z., Y.C., Y.S.); Tongji Hospital, Wuhan, China (Y.D., X.H.); Indiana University, Indianapolis (M.E.B.); Oklahoma Medical Research Foundation, Oklahoma City (S.P., J.D.W.); and German Cancer Research Center, Heidelberg, Germany (S.T., G.J.H.)
| | - Satish Pasula
- From the West China Hospital, Sichuan University, Chengdu, China (Q.W.); University of Oklahoma Health Science Center, Oklahoma City (Q.W., F.Z., M.M.R., W.R., Y.L., C.F., Y.D., C.H., L.D., Y.H., J.M., X.A.Z.); University of Vermont, Burlington (N.H.R., M.T.); University of Tennessee, Memphis (W.Z., Y.C., Y.S.); Tongji Hospital, Wuhan, China (Y.D., X.H.); Indiana University, Indianapolis (M.E.B.); Oklahoma Medical Research Foundation, Oklahoma City (S.P., J.D.W.); and German Cancer Research Center, Heidelberg, Germany (S.T., G.J.H.)
| | - Jonathan D Wren
- From the West China Hospital, Sichuan University, Chengdu, China (Q.W.); University of Oklahoma Health Science Center, Oklahoma City (Q.W., F.Z., M.M.R., W.R., Y.L., C.F., Y.D., C.H., L.D., Y.H., J.M., X.A.Z.); University of Vermont, Burlington (N.H.R., M.T.); University of Tennessee, Memphis (W.Z., Y.C., Y.S.); Tongji Hospital, Wuhan, China (Y.D., X.H.); Indiana University, Indianapolis (M.E.B.); Oklahoma Medical Research Foundation, Oklahoma City (S.P., J.D.W.); and German Cancer Research Center, Heidelberg, Germany (S.T., G.J.H.)
| | - Satoshi Tanaka
- From the West China Hospital, Sichuan University, Chengdu, China (Q.W.); University of Oklahoma Health Science Center, Oklahoma City (Q.W., F.Z., M.M.R., W.R., Y.L., C.F., Y.D., C.H., L.D., Y.H., J.M., X.A.Z.); University of Vermont, Burlington (N.H.R., M.T.); University of Tennessee, Memphis (W.Z., Y.C., Y.S.); Tongji Hospital, Wuhan, China (Y.D., X.H.); Indiana University, Indianapolis (M.E.B.); Oklahoma Medical Research Foundation, Oklahoma City (S.P., J.D.W.); and German Cancer Research Center, Heidelberg, Germany (S.T., G.J.H.)
| | - Xiaolin Huang
- From the West China Hospital, Sichuan University, Chengdu, China (Q.W.); University of Oklahoma Health Science Center, Oklahoma City (Q.W., F.Z., M.M.R., W.R., Y.L., C.F., Y.D., C.H., L.D., Y.H., J.M., X.A.Z.); University of Vermont, Burlington (N.H.R., M.T.); University of Tennessee, Memphis (W.Z., Y.C., Y.S.); Tongji Hospital, Wuhan, China (Y.D., X.H.); Indiana University, Indianapolis (M.E.B.); Oklahoma Medical Research Foundation, Oklahoma City (S.P., J.D.W.); and German Cancer Research Center, Heidelberg, Germany (S.T., G.J.H.)
| | - Markus Thali
- From the West China Hospital, Sichuan University, Chengdu, China (Q.W.); University of Oklahoma Health Science Center, Oklahoma City (Q.W., F.Z., M.M.R., W.R., Y.L., C.F., Y.D., C.H., L.D., Y.H., J.M., X.A.Z.); University of Vermont, Burlington (N.H.R., M.T.); University of Tennessee, Memphis (W.Z., Y.C., Y.S.); Tongji Hospital, Wuhan, China (Y.D., X.H.); Indiana University, Indianapolis (M.E.B.); Oklahoma Medical Research Foundation, Oklahoma City (S.P., J.D.W.); and German Cancer Research Center, Heidelberg, Germany (S.T., G.J.H.)
| | - Günter J Hämmerling
- From the West China Hospital, Sichuan University, Chengdu, China (Q.W.); University of Oklahoma Health Science Center, Oklahoma City (Q.W., F.Z., M.M.R., W.R., Y.L., C.F., Y.D., C.H., L.D., Y.H., J.M., X.A.Z.); University of Vermont, Burlington (N.H.R., M.T.); University of Tennessee, Memphis (W.Z., Y.C., Y.S.); Tongji Hospital, Wuhan, China (Y.D., X.H.); Indiana University, Indianapolis (M.E.B.); Oklahoma Medical Research Foundation, Oklahoma City (S.P., J.D.W.); and German Cancer Research Center, Heidelberg, Germany (S.T., G.J.H.)
| | - Xin A Zhang
- From the West China Hospital, Sichuan University, Chengdu, China (Q.W.); University of Oklahoma Health Science Center, Oklahoma City (Q.W., F.Z., M.M.R., W.R., Y.L., C.F., Y.D., C.H., L.D., Y.H., J.M., X.A.Z.); University of Vermont, Burlington (N.H.R., M.T.); University of Tennessee, Memphis (W.Z., Y.C., Y.S.); Tongji Hospital, Wuhan, China (Y.D., X.H.); Indiana University, Indianapolis (M.E.B.); Oklahoma Medical Research Foundation, Oklahoma City (S.P., J.D.W.); and German Cancer Research Center, Heidelberg, Germany (S.T., G.J.H.).
| |
Collapse
|
38
|
Abstract
Tetraspanins are a family of proteins with four transmembrane domains that play a role in many aspects of cell biology and physiology; they are also used by several pathogens for infection and regulate cancer progression. Many tetraspanins associate specifically and directly with a limited number of proteins, and also with other tetraspanins, thereby generating a hierarchical network of interactions. Through these interactions, tetraspanins are believed to have a role in cell and membrane compartmentalization. In this Cell Science at a Glance article and the accompanying poster, we describe the basic principles underlying tetraspanin-based assemblies and highlight examples of how tetraspanins regulate the trafficking and function of their partner proteins that are required for the normal development and function of several organs, including, in humans, the eye, the kidney and the immune system.
Collapse
Affiliation(s)
- Stéphanie Charrin
- Inserm, U1004, F-94807, Villejuif, France Université Paris-Sud, Institut André Lwoff, F-94807 Villejuif, France
| | - Stéphanie Jouannet
- Inserm, U1004, F-94807, Villejuif, France Université Paris-Sud, Institut André Lwoff, F-94807 Villejuif, France
| | - Claude Boucheix
- Inserm, U1004, F-94807, Villejuif, France Université Paris-Sud, Institut André Lwoff, F-94807 Villejuif, France
| | - Eric Rubinstein
- Inserm, U1004, F-94807, Villejuif, France Université Paris-Sud, Institut André Lwoff, F-94807 Villejuif, France
| |
Collapse
|
39
|
Rossy J, Ma Y, Gaus K. The organisation of the cell membrane: do proteins rule lipids? Curr Opin Chem Biol 2014; 20:54-9. [PMID: 24815858 DOI: 10.1016/j.cbpa.2014.04.009] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Revised: 04/17/2014] [Accepted: 04/17/2014] [Indexed: 11/16/2022]
Abstract
Cell membranes are a complex adaptive system: they are constantly re-organised in response to extra- and intracellular inputs and their local and global structure ultimately determines how, where and when these inputs are processed. This requires a tight coupling of signalling and membranes in localised and specialised compartments. While lipids are essential components of cell membranes, they mostly lack a direct link to the input signals. Here we review how proteins can deform locally membranes, modify and reorganise lipids to form membrane domains and regulate properties like membrane charges and diffusion. From this point-of-view, it appears that proteins play a central role in regulating membrane organisation.
Collapse
Affiliation(s)
- Jérémie Rossy
- Centre for Vascular Research and Australian Centre for Nanomedicine, University of New South Wales, Sydney, Australia.
| | - Yuanqing Ma
- Centre for Vascular Research and Australian Centre for Nanomedicine, University of New South Wales, Sydney, Australia
| | - Katharina Gaus
- Centre for Vascular Research and Australian Centre for Nanomedicine, University of New South Wales, Sydney, Australia.
| |
Collapse
|
40
|
Dahmane S, Rubinstein E, Milhiet PE. Viruses and tetraspanins: lessons from single molecule approaches. Viruses 2014; 6:1992-2011. [PMID: 24800676 PMCID: PMC4036545 DOI: 10.3390/v6051992] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Revised: 03/24/2014] [Accepted: 04/10/2014] [Indexed: 12/15/2022] Open
Abstract
Tetraspanins are four-span membrane proteins that are widely distributed in multi-cellular organisms and involved in several infectious diseases. They have the unique property to form a network of protein-protein interaction within the plasma membrane, due to the lateral associations with one another and with other membrane proteins. Tracking tetraspanins at the single molecule level using fluorescence microscopy has revealed the membrane behavior of the tetraspanins CD9 and CD81 in epithelial cell lines, providing a first dynamic view of this network. Single molecule tracking highlighted that these 2 proteins can freely diffuse within the plasma membrane but can also be trapped, permanently or transiently, in tetraspanin-enriched areas. More recently, a similar strategy has been used to investigate tetraspanin membrane behavior in the context of human immunodeficiency virus type 1 (HIV-1) and hepatitis C virus (HCV) infection. In this review we summarize the main results emphasizing the relationship in terms of membrane partitioning between tetraspanins, some of their partners such as Claudin-1 and EWI-2, and viral proteins during infection. These results will be analyzed in the context of other membrane microdomains, stressing the difference between raft and tetraspanin-enriched microdomains, but also in comparison with virus diffusion at the cell surface. New advanced single molecule techniques that could help to further explore tetraspanin assemblies will be also discussed.
Collapse
Affiliation(s)
- Selma Dahmane
- Inserm, Unité 1054, Single Molecule Biophysics Department, Centre de Biochimie Structurale, 34090, Montpellier, France.
| | | | - Pierre-Emmanuel Milhiet
- Inserm, Unité 1054, Single Molecule Biophysics Department, Centre de Biochimie Structurale, 34090, Montpellier, France.
| |
Collapse
|
41
|
He J, Sun E, Bujny MV, Kim D, Davidson MW, Zhuang X. Dual function of CD81 in influenza virus uncoating and budding. PLoS Pathog 2013; 9:e1003701. [PMID: 24130495 PMCID: PMC3795033 DOI: 10.1371/journal.ppat.1003701] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2012] [Accepted: 08/29/2013] [Indexed: 12/26/2022] Open
Abstract
As an obligatory pathogen, influenza virus co-opts host cell machinery to harbor infection and to produce progeny viruses. In order to characterize the virus-host cell interactions, several genome-wide siRNA screens and proteomic analyses have been performed recently to identify host factors involved in influenza virus infection. CD81 has emerged as one of the top candidates in two siRNA screens and one proteomic study. The exact role played by CD81 in influenza infection, however, has not been elucidated thus far. In this work, we examined the effect of CD81 depletion on the major steps of the influenza infection. We found that CD81 primarily affected virus infection at two stages: viral uncoating during entry and virus budding. CD81 marked a specific endosomal population and about half of the fused influenza virus particles underwent fusion within the CD81-positive endosomes. Depletion of CD81 resulted in a substantial defect in viral fusion and infection. During virus assembly, CD81 was recruited to virus budding site on the plasma membrane, and in particular, to specific sub-viral locations. For spherical and slightly elongated influenza virus, CD81 was localized at both the growing tip and the budding neck of the progeny viruses. CD81 knockdown led to a budding defect and resulted in elongated budding virions with a higher propensity to remain attached to the plasma membrane. Progeny virus production was markedly reduced in CD81-knockdown cells even when the uncoating defect was compensated. In filamentous virus, CD81 was distributed at multiple sites along the viral filament. Taken together, these results demonstrate important roles of CD81 in both entry and budding stages of the influenza infection cycle. As a “Trojan Horse” that only encodes 13 viral proteins, influenza hijacks host cell machinery for productive infection. In this work, we studied the role of the host protein CD81 in influenza infection. We found that CD81 was important for influenza infection at two distinct stages: virus uncoating and virus budding. Specifically, during virus entry, more than half of internalized virus particles were trafficked into a specific CD81-positive endosomal population for virus uncoating. Depleting CD81 led to a significant defect in viral uncoating and infection. During virus egress, CD81 was recruited to virus assembly site, and incorporated into individual virions at specific sub-viral locations. CD81 depletion resulted in virions that failed to detach from the plasma membrane and a marked decrease in progeny virus production.
Collapse
Affiliation(s)
- Jiang He
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, Massachusetts, United States of America
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts, United States of America
| | - Eileen Sun
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts, United States of America
- Program in Virology, Harvard Medical School, Harvard University, Boston, Massachusetts, United States of America
| | - Miriam V. Bujny
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts, United States of America
| | - Doory Kim
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts, United States of America
| | - Michael W. Davidson
- National High Magnetic Field Laboratory and Department of Biological Science, The Florida State University, Tallahassee, Florida, United States of America
| | - Xiaowei Zhuang
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts, United States of America
- Department of Physics, Harvard University, Cambridge, Massachusetts, United States of America
- Howard Hughes Medical Institute, Cambridge, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
42
|
Benito-Martin A, Ucero AC, Zubiri I, Posada-Ayala M, Fernandez-Fernandez B, Cannata-Ortiz P, Sanchez-Nino MD, Ruiz-Ortega M, Egido J, Alvarez-Llamas G, Ortiz A. Osteoprotegerin in exosome-like vesicles from human cultured tubular cells and urine. PLoS One 2013; 8:e72387. [PMID: 24058411 PMCID: PMC3751949 DOI: 10.1371/journal.pone.0072387] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2013] [Accepted: 07/09/2013] [Indexed: 12/12/2022] Open
Abstract
Urinary exosomes have been proposed as potential diagnostic tools. TNF superfamily cytokines and receptors may be present in exosomes and are expressed by proximal tubular cells. We have now studied the expression of selected TNF superfamily proteins in exosome-like vesicles from cultured human proximal tubular cells and human urine and have identified additional proteins in these vesicles by LC-MS/MS proteomics. Human proximal tubular cells constitutively released exosome-like vesicles that did not contain the TNF superfamily cytokines TRAIL or TWEAK. However, exosome-like vesicles contained osteoprotegerin (OPG), a TNF receptor superfamily protein, as assessed by Western blot, ELISA or selected reaction monitoring by nLC-(QQQ)MS/MS. Twenty-one additional proteins were identified in tubular cell exosome-like vesicles, including one (vitamin D binding protein) that had not been previously reported in exosome-like vesicles. Twelve were extracellular matrix proteins, including the basement membrane proteins type IV collagen, nidogen-1, agrin and fibulin-1. Urine from chronic kidney disease patients contained a higher amount of exosomal protein and exosomal OPG than urine from healthy volunteers. Specifically OPG was increased in autosomal dominant polycystic kidney disease urinary exosome-like vesicles and expressed by cystic epithelium in vivo. In conclusion, OPG is present in exosome-like vesicles secreted by proximal tubular epithelial cells and isolated from Chronic Kidney Disease urine.
Collapse
Affiliation(s)
- Alberto Benito-Martin
- Department of Nephrology, Instituto de Investigaciones Sanitarias-Fundación Jiménez Díaz - Universidad Autonoma de Madrid, Madrid, Spain
| | - Alvaro Conrado Ucero
- Department of Nephrology, Instituto de Investigaciones Sanitarias-Fundación Jiménez Díaz - Universidad Autonoma de Madrid, Madrid, Spain
| | - Irene Zubiri
- Department of Immunology, Instituto de Investigaciones Sanitarias-Fundación Jiménez Díaz - Universidad Autonoma de Madrid, Madrid, Spain
| | - Maria Posada-Ayala
- Department of Immunology, Instituto de Investigaciones Sanitarias-Fundación Jiménez Díaz - Universidad Autonoma de Madrid, Madrid, Spain
| | - Beatriz Fernandez-Fernandez
- Department of Nephrology, Instituto de Investigaciones Sanitarias-Fundación Jiménez Díaz - Universidad Autonoma de Madrid, Madrid, Spain
| | - Pablo Cannata-Ortiz
- Department of Pathology, Instituto de Investigaciones Sanitarias-Fundación Jiménez Díaz - Universidad Autonoma de Madrid, Madrid, Spain
| | | | - Marta Ruiz-Ortega
- Department of Nephrology, Instituto de Investigaciones Sanitarias-Fundación Jiménez Díaz - Universidad Autonoma de Madrid, Madrid, Spain
| | - Jesus Egido
- Department of Nephrology, Instituto de Investigaciones Sanitarias-Fundación Jiménez Díaz - Universidad Autonoma de Madrid, Madrid, Spain
- Instituto Reina Sofia de Investigacion Nefrologica, Madrid, Spain
| | - Gloria Alvarez-Llamas
- Department of Immunology, Instituto de Investigaciones Sanitarias-Fundación Jiménez Díaz - Universidad Autonoma de Madrid, Madrid, Spain
| | - Alberto Ortiz
- Department of Nephrology, Instituto de Investigaciones Sanitarias-Fundación Jiménez Díaz - Universidad Autonoma de Madrid, Madrid, Spain
- Instituto Reina Sofia de Investigacion Nefrologica, Madrid, Spain
- * E-mail:
| |
Collapse
|
43
|
Yamada M, Mugnai G, Serada S, Yagi Y, Naka T, Sekiguchi K. Substrate-attached materials are enriched with tetraspanins and are analogous to the structures associated with rear-end retraction in migrating cells. Cell Adh Migr 2013; 7:304-14. [PMID: 23676281 PMCID: PMC3711998 DOI: 10.4161/cam.25041] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Substrate-attached materials (SAMs) are cellular feet that remain on substrates after the treatment of adherent cells with EGTA. SAMs are thought to contain cell adhesion machineries, but their biochemical properties have not been addressed in detail. To gain insight into the molecular mechanisms operating in cell adhesions, we comprehensively identified the protein components of SAMs by liquid chromatography coupled with tandem mass spectrometry, followed by immunoblot analysis. We found that the tetraspanins CD9, CD81, and CD151 were enriched in SAMs along with other transmembrane proteins that are known to associate with tetraspanins. Notably, integrins were detected in SAMs, but the components of focal adhesions were scarcely detected. These observations are reminiscent of the “footprints” that remain on substrates when the retraction fibers at the rear of migrating cells are released, because such footprints have been reported to contain tetraspanins and integrins but not focal adhesion proteins. In support of this hypothesis, the formation of SAMs was attenuated by inhibitors of ROCK, myosin II and dynamin, all of which are known to participate in rear-end retraction in migrating cells. Furthermore, SAMs left on collagen-coated substrates were found by electron microscopy to be fewer and thinner than those on laminin-coated substrates, reflecting the thin and fragile retraction fibers of cells migrating on collagen. Collectively, these results indicate that SAMs closely resemble the footprints and retraction fibers of migrating cells in their protein components, and that they are yielded by similar mechanisms.
Collapse
Affiliation(s)
- Masashi Yamada
- Laboratory of Extracellular Matrix Biochemistry, Institute for Protein Research, Osaka University, Suita, Japan
| | | | | | | | | | | |
Collapse
|