1
|
Zhang Y, Pan C, Wang S, Zhou Y, Chen J, Yu X, Peng R, Zhang N, Yang H. Distinctive function of Tetraspanins: Implication in viral infections. Virulence 2025; 16:2474188. [PMID: 40053412 PMCID: PMC11901453 DOI: 10.1080/21505594.2025.2474188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 02/03/2025] [Accepted: 02/25/2025] [Indexed: 03/09/2025] Open
Abstract
Harboring four transmembrane domains in their structural hallmark, Tetraspanins (Tspans) are a family of glycoproteins with pivotal functions in a variety of biological and cellular processes. Through interacting laterally with each other or specific membrane proteins, Tspans organize tetraspanin-enriched microdomains (TEMs), modulating cellular signaling, adhesion, fusion, and proliferation. An abundance of evidence has identified the multiple functions in the progression of cancer as well as the underlying molecular mechanisms. Recently, plenty of studies have focused on the utilities of Tspans by pathogens for infection, especially the infection of viruses. The expression of Tspans correlates with the phase of viral infection, the type of virus, and targeted therapies. In particular, perturbations of Tspans in host cells can affect viral attachment, intracellular trafficking, translation, virus assembly, and release. In this review, we summarize and provide a historical overview of the discovery and characterization of various kinds of virus infection and highlight their diversity and complexity, along with the virus life cycle. Furthermore, we examined the current understanding of how various Tspans are involved in the regulatory mechanisms underlying viral infection. This review aims to offer a comprehensive understanding of the targeting of Tspans for therapeutic intervention in infections caused by diverse pathogens.
Collapse
Affiliation(s)
- Yuzhi Zhang
- School of Life Sciences, Northwestern Polytechnical University, Xi’an, Shaanxi, China
- Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment, Xi’an, China
- Research Center of Special Environmental Biomechanics & Medical Engineering, Northwestern Polytechnical University, Xi’an, Shaanxi, China
| | - Chengwei Pan
- Department of Mechanical Engineering, University of Victoria, Victoria, BC, Canada
| | - Sijie Wang
- School of Life Sciences, Northwestern Polytechnical University, Xi’an, Shaanxi, China
- Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment, Xi’an, China
- Research Center of Special Environmental Biomechanics & Medical Engineering, Northwestern Polytechnical University, Xi’an, Shaanxi, China
| | - Yidan Zhou
- School of Life Sciences, Northwestern Polytechnical University, Xi’an, Shaanxi, China
- Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment, Xi’an, China
- Research Center of Special Environmental Biomechanics & Medical Engineering, Northwestern Polytechnical University, Xi’an, Shaanxi, China
| | - Jiawei Chen
- School of Life Sciences, Northwestern Polytechnical University, Xi’an, Shaanxi, China
- Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment, Xi’an, China
- Research Center of Special Environmental Biomechanics & Medical Engineering, Northwestern Polytechnical University, Xi’an, Shaanxi, China
| | - Xiaoyu Yu
- School of Life Sciences, Northwestern Polytechnical University, Xi’an, Shaanxi, China
- Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment, Xi’an, China
- Research Center of Special Environmental Biomechanics & Medical Engineering, Northwestern Polytechnical University, Xi’an, Shaanxi, China
| | - Ruining Peng
- School of Life Sciences, Northwestern Polytechnical University, Xi’an, Shaanxi, China
- Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment, Xi’an, China
- Research Center of Special Environmental Biomechanics & Medical Engineering, Northwestern Polytechnical University, Xi’an, Shaanxi, China
| | - Nu Zhang
- School of Life Sciences, Northwestern Polytechnical University, Xi’an, Shaanxi, China
- Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment, Xi’an, China
- Research Center of Special Environmental Biomechanics & Medical Engineering, Northwestern Polytechnical University, Xi’an, Shaanxi, China
| | - Hui Yang
- School of Life Sciences, Northwestern Polytechnical University, Xi’an, Shaanxi, China
- Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment, Xi’an, China
- Research Center of Special Environmental Biomechanics & Medical Engineering, Northwestern Polytechnical University, Xi’an, Shaanxi, China
| |
Collapse
|
2
|
Zheng S, Zhang Y, Cai R, Cai B, Luo S, He S, Peng T, Wang W, Cui H, Li H, Lu X. The untold story of CD82: Exploring its non-canonical roles in cancer. Pathol Res Pract 2025; 270:155979. [PMID: 40252385 DOI: 10.1016/j.prp.2025.155979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2025] [Revised: 04/06/2025] [Accepted: 04/13/2025] [Indexed: 04/21/2025]
Abstract
CD82, traditionally recognized as a metastasis suppressor within the tetraspanin family, has emerged as a key player in diverse cancer-related processes beyond its canonical functions. This review highlights recent research on the non-canonical roles of CD82 in cancer progression, with a particular focus on its regulation of immune cell interactions, its impact on tumor microenvironment modulation, and its potential as both a therapeutic target and a biomarker. By examining the novel functions of CD82 in immune modulation and its influence on key signaling pathways, we propose that CD82 offers promising avenues for therapeutic interventions in cancer. This paper provides a comprehensive synthesis of the current understanding of CD82's expanded roles, underscoring its potential in improving cancer diagnosis and therapy.
Collapse
Affiliation(s)
- Shutao Zheng
- State Key Laboratory of Pathogenesis, Prevention, Treatment of Central Asian High Incidence Diseases, Clinical Medical Research Institute, First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region 830011, PR China
| | - Yao Zhang
- Beijing Beanstalk International Bilingual School, Beijing 100016, PR China
| | - Ren Cai
- State Key Laboratory of Pathogenesis, Prevention, Treatment of Central Asian High Incidence Diseases, Clinical Medical Research Institute, First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region 830011, PR China
| | - Bangwu Cai
- State Key Laboratory of Pathogenesis, Prevention, Treatment of Central Asian High Incidence Diseases, Clinical Medical Research Institute, First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region 830011, PR China
| | - Shujuan Luo
- State Key Laboratory of Pathogenesis, Prevention, Treatment of Central Asian High Incidence Diseases, Clinical Medical Research Institute, First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region 830011, PR China
| | - Shuo He
- State Key Laboratory of Pathogenesis, Prevention, Treatment of Central Asian High Incidence Diseases, Clinical Medical Research Institute, First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region 830011, PR China
| | - Tianyuan Peng
- State Key Laboratory of Pathogenesis, Prevention, Treatment of Central Asian High Incidence Diseases, Clinical Medical Research Institute, First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region 830011, PR China
| | - Wei Wang
- Department of Digestive Internal Medicine, the Affiliated Tumor Hospital of Xinjiang Medical University, PR China
| | - Hong Cui
- State Key Laboratory of Pathogenesis, Prevention, Treatment of Central Asian High Incidence Diseases, Clinical Medical Research Institute, First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region 830011, PR China
| | - Huifang Li
- Department of Breast Surgery, the First Affiliated Hospital of Xinjiang Medical University, PR China
| | - Xiaomei Lu
- State Key Laboratory of Pathogenesis, Prevention, Treatment of Central Asian High Incidence Diseases, Clinical Medical Research Institute, First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region 830011, PR China.
| |
Collapse
|
3
|
Arone C, Martial S, Burlaud-Gaillard J, Thoulouze MI, Roingeard P, Dutartre H, Muriaux D. HTLV-1 biofilm polarization maintained by tetraspanin CD82 is required for efficient viral transmission. mBio 2023; 14:e0132623. [PMID: 37889017 PMCID: PMC10746275 DOI: 10.1128/mbio.01326-23] [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: 05/24/2023] [Accepted: 09/11/2023] [Indexed: 10/28/2023] Open
Abstract
IMPORTANCE In the early stages of infection, human T-lymphotropic virus type 1 (HTLV-1) dissemination within its host is believed to rely mostly on cell-to-cell contacts. Past studies unveiled a novel mechanism of HTLV-1 intercellular transmission based on the remodeling of the host-cell extracellular matrix and the generation of cell-surface viral assemblies whose structure, composition, and function resemble bacterial biofilms. These polarized aggregates of infectious virions, identified as viral biofilms, allow the bulk delivery of viruses to target cells and may help to protect virions from immune attacks. However, viral biofilms' molecular and functional description is still in its infancy, although it is crucial to fully decipher retrovirus pathogenesis. Here, we explore the function of cellular tetraspanins (CD9, CD81, CD82) that we detect inside HTLV-1 particles within biofilms. Our results demonstrate specific roles for CD82 in the cell-surface distribution and intercellular transmission of HTLV-1 biofilms, which we document as two essential parameters for efficient viral transmission. At last, our findings indicate that N-glycosylation of cell-surface molecules, including CD82, is required for the polarization of HTLV-1 biofilms and for the efficient transmission of HTLV-1 between T-lymphocytes.
Collapse
Affiliation(s)
- Coline Arone
- Infectious Disease Research Institute of Montpellier (IRIM), UMR CNRS, Montpellier, France
| | - Samuel Martial
- Center for International Research on Infectiology (CIRI), UMR Inserm, Lyon, France
| | | | | | - Philippe Roingeard
- IBiSA Electron Microscopy Platform of Tours University, UMR Inserm, Tours, France
| | - Hélène Dutartre
- Center for International Research on Infectiology (CIRI), UMR Inserm, Lyon, France
| | - Delphine Muriaux
- Infectious Disease Research Institute of Montpellier (IRIM), UMR CNRS, Montpellier, France
| |
Collapse
|
4
|
Lee H, Han JH, Kang YJ, Hwangbo H, Yoon A, Kim HS, Lee D, Lee SY, Choi BH, Kim JJ, Kim SR, Choi YH, Hur J. CD82 attenuates TGF-β1-mediated epithelial-mesenchymal transition by blocking smad-dependent signaling in ARPE-19 cells. Front Pharmacol 2022; 13:991056. [PMID: 36386228 PMCID: PMC9640495 DOI: 10.3389/fphar.2022.991056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 10/17/2022] [Indexed: 11/27/2022] Open
Abstract
In retinal pigment epithelial (RPE) cells, transforming growth factor-beta (TGF-β) plays a critical role in epithelial-mesenchymal transition (EMT), which contributes to various fibrotic retinal disorders. In the present study, we investigated the effect of recombinant human cluster of differentiation 82 (rhCD82), a tumor metastasis suppressor, on TGF-β-induced EMT in the human RPE cell line APRE-19. The results show that TGF-β1 significantly enhanced cell migration, invasion and the expression of EMT-mediate factors in ARPE-19 cells. However, rhCD82 markedly inhibited cell mobility and the expression of epithelial marker, zonula occludens-1, as well as increased the expression of mesenchymal markers, such as vimentin and α-smooth muscle actin in TGF-β1-treated APRE-19 cells. In addition, TGF-β1 upregulated the phosphorylation of Smad, extracellular signal regulated kinase (ERK) and glycogen synthase kinase-3β (GSK-3β), but only phosphorylation of Smad was suppressed by rhCD82. Noteworthy, rhCD82 greatly suppressed the expression of TGF-β receptor I (TGFRI), TGFRII and integrins in TGF-β1-treated APRE-19 cells. In particular, the result of molecular docking analysis and structural modeling show that rhCD82 partially interacts with the TGF-β1 binding sites of TGFRI, TGFRII, integrin β1 and integrin αv. Taken together, this finding suggested that rhCD82 suppressed TGF-β1-induced EMT of RPE by blocking of Smad-dependent pathway, which is caused by rhCD82 interaction with TGFRs and integrins, suggesting new insight into CD82 as a potential therapeutic strategy in fibrotic retinal disorders.
Collapse
Affiliation(s)
- Hyesook Lee
- Department of Convergence Medicine, Pusan National University School of Medicine, Yangsan, South Korea
| | - Jung-Hwa Han
- Department of Convergence Medicine, Pusan National University School of Medicine, Yangsan, South Korea
- PNU GRAND Convergence Medical Science Education Research Center, Pusan National University School of Medicine, Yangsan, South Korea
| | - Yun Jeong Kang
- Department of Convergence Medicine, Pusan National University School of Medicine, Yangsan, South Korea
| | - Hyun Hwangbo
- Department of Biochemistry, Dong-eui University College of Korean Medicine, Busan, South Korea
| | - Aeseon Yoon
- Department of Convergence Medicine, Pusan National University School of Medicine, Yangsan, South Korea
| | - Hyung-Sik Kim
- Department of Oral Biochemistry, Dental and Life Science Institute, School of Dentistry, Pusan National University, Yangsan, South Korea
| | - Dongjun Lee
- Department of Convergence Medicine, Pusan National University School of Medicine, Yangsan, South Korea
| | - Soo Yong Lee
- Division of Cardiology, Department of Internal Medicine and Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, South Korea
| | - Byung Hyun Choi
- Division of Hepato-Biliary-Pancreatic Surgery and Transplantation, Department of Surgery, Pusan National University School of Medicine and Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, South Korea
| | - Jae-Joon Kim
- Medical Oncology and Hematology, Department of Internal Medicine, Pusan National University Yangsan Hospital, Yangsan, South Korea
| | - Seo Rin Kim
- Department of Nephrology and Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, South Korea
| | - Yung Hyun Choi
- Department of Biochemistry, Dong-eui University College of Korean Medicine, Busan, South Korea
- Anti-Aging Research Center and Core-Facility Center for Tissue Regeneration, Dong-eui University, Busan, South Korea
- *Correspondence: Yung Hyun Choi, ; Jin Hur,
| | - Jin Hur
- Department of Convergence Medicine, Pusan National University School of Medicine, Yangsan, South Korea
- PNU GRAND Convergence Medical Science Education Research Center, Pusan National University School of Medicine, Yangsan, South Korea
- *Correspondence: Yung Hyun Choi, ; Jin Hur,
| |
Collapse
|
5
|
Reppert N, Lang T. A conserved sequence in the small intracellular loop of tetraspanins forms an M-shaped inter-helix turn. Sci Rep 2022; 12:4494. [PMID: 35296690 PMCID: PMC8927573 DOI: 10.1038/s41598-022-07243-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 02/15/2022] [Indexed: 11/30/2022] Open
Abstract
Tetraspanins are a family of small proteins with four transmembrane segments (TMSs) playing multiple roles in human physiology. Nevertheless, we know little about the factors determining their structure. In the study at hand, we focus on the small intracellular loop (SIL) between TMS2 and TMS3. There we have identified a conserved five amino acid core region with three charged residues forming an M-shaped backbone, which we call M-motif. The M´s plane runs parallel to the membrane surface and the central amino acid constitutes the inter-helix turning point. At the second position of the M-motif, in tetraspanin crystal structures we identified a glutamate oriented towards a lysine in the juxtamembrane region of TMS1. Using Tspan17 as example, we find that by mutating either the glutamate or juxtamembrane-lysine, but not upon glutamate/lysine swapping, expression level, maturation and ER-exit are reduced. We conclude that the SIL is more than a short linking segment but propose it is involved in shaping the tertiary structure of tetraspanins.
Collapse
Affiliation(s)
- Nikolas Reppert
- Department of Membrane Biochemistry, Life and Medical Sciences (LIMES) Institute, University of Bonn, Carl-Troll-Straße 31, 53115, Bonn, Germany.
| | - Thorsten Lang
- Department of Membrane Biochemistry, Life and Medical Sciences (LIMES) Institute, University of Bonn, Carl-Troll-Straße 31, 53115, Bonn, Germany.
| |
Collapse
|
6
|
HIV-1 and HTLV-1 Transmission Modes: Mechanisms and Importance for Virus Spread. Viruses 2022; 14:v14010152. [PMID: 35062355 PMCID: PMC8779814 DOI: 10.3390/v14010152] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 01/07/2022] [Accepted: 01/11/2022] [Indexed: 12/13/2022] Open
Abstract
So far, only two retroviruses, human immunodeficiency virus (HIV) (type 1 and 2) and human T-cell lymphotropic virus type 1 (HTLV-1), have been recognized as pathogenic for humans. Both viruses mainly infect CD4+ T lymphocytes. HIV replication induces the apoptosis of CD4 lymphocytes, leading to the development of acquired immunodeficiency syndrome (AIDS). After a long clinical latency period, HTLV-1 can transform lymphocytes, with subsequent uncontrolled proliferation and the manifestation of a disease called adult T-cell leukemia (ATLL). Certain infected patients develop neurological autoimmune disorder called HTLV-1-associated myelopathy, also known as tropical spastic paraparesis (HAM/TSP). Both viruses are transmitted between individuals via blood transfusion, tissue/organ transplantation, breastfeeding, and sexual intercourse. Within the host, these viruses can spread utilizing either cell-free or cell-to-cell modes of transmission. In this review, we discuss the mechanisms and importance of each mode of transmission for the biology of HIV-1 and HTLV-1.
Collapse
|
7
|
Zhu T, Sun Y, Chen X. Arabidopsis Tetraspanins Facilitate Virus Infection via Membrane-Recognition GCCK/RP Motif and Cysteine Residues. FRONTIERS IN PLANT SCIENCE 2022; 13:805633. [PMID: 35310653 PMCID: PMC8927881 DOI: 10.3389/fpls.2022.805633] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Accepted: 02/04/2022] [Indexed: 05/07/2023]
Abstract
Tetraspanins (TETs) function as key molecular scaffolds for surface signal recognition and transduction via the assembly of tetraspanin-enriched microdomains. TETs' function in mammalian has been intensively investigated for the organization of multimolecular membrane complexes, regulation of cell migration and cellular adhesion, whereas plant TET studies lag far behind. Animal and plant TETs share similar topologies, despite the hallmark of "CCG" in the large extracellular loop of animal TETs, plant TETs contain a plant specific GCCK/RP motif and more conserved cysteine residues. Here, we showed that the GCCK/RP motif is responsible for TET protein association with the plasma membrane. Moreover, the conserved cysteine residues located within or neighboring the GCCK/RP motif are both crucial for TET anchoring to membrane. During virus infection, the intact TET3 protein enhanced but GCCK/RP motif or cysteine residues-deficient TET3 variants abolished the cell-to-cell movement capability of virus. This study provides cellular evidence that the GCCK/RP motif and the conserved cysteine residues are the primary determinants for the distribution and function of TET proteins in Arabidopsis.
Collapse
Affiliation(s)
- Tingyu Zhu
- College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
- Horticultural Plant Biology and Metabolomics Center, Haixia Institute of Science and Technology, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Yanbiao Sun
- Horticultural Plant Biology and Metabolomics Center, Haixia Institute of Science and Technology, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Xu Chen
- Horticultural Plant Biology and Metabolomics Center, Haixia Institute of Science and Technology, Fujian Agriculture and Forestry University, Fuzhou, China
- *Correspondence: Xu Chen,
| |
Collapse
|
8
|
Kim Y, Mensah GA, Al Sharif S, Pinto DO, Branscome H, Yelamanchili SV, Cowen M, Erickson J, Khatkar P, Mahieux R, Kashanchi F. Extracellular Vesicles from Infected Cells Are Released Prior to Virion Release. Cells 2021; 10:781. [PMID: 33916140 PMCID: PMC8066806 DOI: 10.3390/cells10040781] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 03/24/2021] [Accepted: 03/29/2021] [Indexed: 12/29/2022] Open
Abstract
Here, we have attempted to address the timing of EV and virion release from virally infected cells. Uninfected (CEM), HIV-1-infected (J1.1), and human T cell leukemia virus-1 (HTLV-1)-infected (HUT102) cells were synchronized in G0. Viral latency was reversed by increasing gene expression with the addition of serum-rich media and inducers. Supernatants and cell pellets were collected post-induction at different timepoints and assayed for extracellular vesicle (EV) and autophagy markers; and for viral proteins and RNAs. Tetraspanins and autophagy-related proteins were found to be differentially secreted in HIV-1- and HTLV-1-infected cells when compared with uninfected controls. HIV-1 proteins were present at 6 h and their production increased up to 24 h. HTLV-1 proteins peaked at 6 h and plateaued. HIV-1 and HTLV-1 RNA production correlated with viral protein expression. Nanoparticle tracking analysis (NTA) showed increase of EV concentration over time in both uninfected and infected samples. Finally, the HIV-1 supernatant from the 6-h samples was found not to be infectious; however, the virus from the 24-h samples was successfully rescued and infectious. Overall, our data indicate that EV release may occur prior to viral release from infected cells, thereby implicating a potentially significant effect of EVs on uninfected recipient cells prior to subsequent viral infection and spread.
Collapse
Affiliation(s)
- Yuriy Kim
- Laboratory of Molecular Virology, School of Systems Biology, George Mason University, Manassas, VA 20110, USA; (Y.K.); (G.A.M.); (S.A.S.); (D.O.P.); (H.B.); (M.C.); (J.E.); (P.K.)
| | - Gifty A. Mensah
- Laboratory of Molecular Virology, School of Systems Biology, George Mason University, Manassas, VA 20110, USA; (Y.K.); (G.A.M.); (S.A.S.); (D.O.P.); (H.B.); (M.C.); (J.E.); (P.K.)
| | - Sarah Al Sharif
- Laboratory of Molecular Virology, School of Systems Biology, George Mason University, Manassas, VA 20110, USA; (Y.K.); (G.A.M.); (S.A.S.); (D.O.P.); (H.B.); (M.C.); (J.E.); (P.K.)
| | - Daniel O. Pinto
- Laboratory of Molecular Virology, School of Systems Biology, George Mason University, Manassas, VA 20110, USA; (Y.K.); (G.A.M.); (S.A.S.); (D.O.P.); (H.B.); (M.C.); (J.E.); (P.K.)
| | - Heather Branscome
- Laboratory of Molecular Virology, School of Systems Biology, George Mason University, Manassas, VA 20110, USA; (Y.K.); (G.A.M.); (S.A.S.); (D.O.P.); (H.B.); (M.C.); (J.E.); (P.K.)
| | - Sowmya V. Yelamanchili
- Department of Anesthesiology, University of Nebraska Medical Center, Omaha, NE 68198, USA;
| | - Maria Cowen
- Laboratory of Molecular Virology, School of Systems Biology, George Mason University, Manassas, VA 20110, USA; (Y.K.); (G.A.M.); (S.A.S.); (D.O.P.); (H.B.); (M.C.); (J.E.); (P.K.)
| | - James Erickson
- Laboratory of Molecular Virology, School of Systems Biology, George Mason University, Manassas, VA 20110, USA; (Y.K.); (G.A.M.); (S.A.S.); (D.O.P.); (H.B.); (M.C.); (J.E.); (P.K.)
| | - Pooja Khatkar
- Laboratory of Molecular Virology, School of Systems Biology, George Mason University, Manassas, VA 20110, USA; (Y.K.); (G.A.M.); (S.A.S.); (D.O.P.); (H.B.); (M.C.); (J.E.); (P.K.)
| | - Renaud Mahieux
- International Center for Research in Infectiology, Retroviral Oncogenesis Laboratory, INSERM U1111-Université Claude Bernard Lyon 1, Ecole Normale Superieure de Lyon, Université de Lyon, Fondation Pour La Recherche Médicale, Labex Ecofect, 69007 Lyon, France;
| | - Fatah Kashanchi
- Laboratory of Molecular Virology, School of Systems Biology, George Mason University, Manassas, VA 20110, USA; (Y.K.); (G.A.M.); (S.A.S.); (D.O.P.); (H.B.); (M.C.); (J.E.); (P.K.)
| |
Collapse
|
9
|
Dual Pathways of Human Immunodeficiency Virus Type 1 Envelope Glycoprotein Trafficking Modulate the Selective Exclusion of Uncleaved Oligomers from Virions. J Virol 2021; 95:JVI.01369-20. [PMID: 33148792 DOI: 10.1128/jvi.01369-20] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 10/24/2020] [Indexed: 12/14/2022] Open
Abstract
The human immunodeficiency virus type 1 (HIV-1) envelope glycoprotein (Env) trimer is transported through the secretory pathway to the infected cell surface and onto virion particles. In the Golgi, the gp160 Env precursor is modified by complex sugars and proteolytically cleaved to produce the mature functional Env trimer, which resists antibody neutralization. We observed mostly uncleaved gp160 and smaller amounts of cleaved gp120 and gp41 Envs on the surface of HIV-1-infected or Env-expressing cells; however, cleaved Envs were relatively enriched in virions and virus-like particles (VLPs). This relative enrichment of cleaved Env in VLPs was observed for wild-type Envs, for Envs lacking the cytoplasmic tail, and for CD4-independent, conformationally flexible Envs. On the cell surface, we identified three distinct populations of Envs: (i) the cleaved Env was transported through the Golgi, was modified by complex glycans, formed trimers that cross-linked efficiently, and was recognized by broadly neutralizing antibodies; (ii) a small fraction of Env modified by complex carbohydrates escaped cleavage in the Golgi; and (iii) the larger population of uncleaved Env lacked complex carbohydrates, cross-linked into diverse oligomeric forms, and was recognized by poorly neutralizing antibodies. This last group of more "open" Env oligomers reached the cell surface in the presence of brefeldin A, apparently bypassing the Golgi apparatus. Relative to Envs transported through the Golgi, these uncleaved Envs were counterselected for virion incorporation. By employing two pathways for Env transport to the surface of infected cells, HIV-1 can misdirect host antibody responses toward conformationally flexible, uncleaved Env without compromising virus infectivity.IMPORTANCE The envelope glycoprotein (Env) trimers on the surface of human immunodeficiency virus type 1 (HIV-1) mediate the entry of the virus into host cells and serve as targets for neutralizing antibodies. The cleaved, functional Env is incorporated into virus particles from the surface of the infected cell. We found that an uncleaved form of Env is transported to the cell surface by an unconventional route, but this nonfunctional Env is mostly excluded from the virus. Thus, only one of the pathways by which Env is transported to the surface of infected cells results in efficient incorporation into virus particles, potentially allowing the uncleaved Env to act as a decoy to the host immune system without compromising virus infectivity.
Collapse
|
10
|
Neviani V, van Deventer S, Wörner TP, Xenaki KT, van de Waterbeemd M, Rodenburg RNP, Wortel IMN, Kuiper JK, Huisman S, Granneman J, van Bergen En Henegouwen PMP, Heck AJR, van Spriel AB, Gros P. Site-specific functionality and tryptophan mimicry of lipidation in tetraspanin CD9. FEBS J 2020; 287:5323-5344. [PMID: 32181977 PMCID: PMC7818406 DOI: 10.1111/febs.15295] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 01/19/2020] [Accepted: 03/13/2020] [Indexed: 12/18/2022]
Abstract
Lipidation of transmembrane proteins regulates many cellular activities, including signal transduction, cell–cell communication, and membrane trafficking. However, how lipidation at different sites in a membrane protein affects structure and function remains elusive. Here, using native mass spectrometry we determined that wild‐type human tetraspanins CD9 and CD81 exhibit nonstochastic distributions of bound acyl chains. We revealed CD9 lipidation at its three most frequent lipidated sites suffices for EWI‐F binding, while cysteine‐to‐alanine CD9 mutations markedly reduced binding of EWI‐F. EWI‐F binding by CD9 was rescued by mutating all or, albeit to a lesser extent, only the three most frequently lipidated sites into tryptophans. These mutations did not affect the nanoscale distribution of CD9 in cell membranes, as shown by super‐resolution microscopy using a CD9‐specific nanobody. Thus, these data demonstrate site‐specific, possibly conformation‐dependent, functionality of lipidation in tetraspanin CD9 and identify tryptophan mimicry as a possible biochemical approach to study site‐specific transmembrane‐protein lipidation.
Collapse
Affiliation(s)
- Viviana Neviani
- Crystal and Structural Chemistry, Department of Chemistry, Bijvoet Center for Biomolecular Research, Utrecht University, The Netherlands
| | - Sjoerd van Deventer
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud UMC, Nijmegen, The Netherlands
| | - Tobias P Wörner
- Biomolecular Mass Spectrometry and Proteomics, Department of Chemistry, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, The Netherlands
| | - Katerina T Xenaki
- Cell Biology, Department of Biology, Utrecht University, The Netherlands
| | - Michiel van de Waterbeemd
- Biomolecular Mass Spectrometry and Proteomics, Department of Chemistry, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, The Netherlands
| | - Remco N P Rodenburg
- Crystal and Structural Chemistry, Department of Chemistry, Bijvoet Center for Biomolecular Research, Utrecht University, The Netherlands
| | - Inge M N Wortel
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud UMC, Nijmegen, The Netherlands
| | - Jeroen K Kuiper
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud UMC, Nijmegen, The Netherlands
| | - Sofie Huisman
- Crystal and Structural Chemistry, Department of Chemistry, Bijvoet Center for Biomolecular Research, Utrecht University, The Netherlands
| | - Joke Granneman
- Crystal and Structural Chemistry, Department of Chemistry, Bijvoet Center for Biomolecular Research, Utrecht University, The Netherlands
| | | | - Albert J R Heck
- Biomolecular Mass Spectrometry and Proteomics, Department of Chemistry, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, The Netherlands
| | - Annemiek B van Spriel
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud UMC, Nijmegen, The Netherlands
| | - Piet Gros
- Crystal and Structural Chemistry, Department of Chemistry, Bijvoet Center for Biomolecular Research, Utrecht University, The Netherlands
| |
Collapse
|
11
|
Cao J, Tan X. Comparative analysis of the tetraspanin gene family in six teleost fishes. FISH & SHELLFISH IMMUNOLOGY 2018; 82:432-441. [PMID: 30145201 DOI: 10.1016/j.fsi.2018.08.048] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Revised: 07/02/2018] [Accepted: 08/22/2018] [Indexed: 06/08/2023]
Abstract
Tetraspanins are a family of membrane proteins, which play important roles in many aspects of cell biology and physiology via binding other tetraspanins or proteins. In this study, we identified 251 putative tetraspanin genes in 6 teleost fishes. Conserved gene organization and motif distribution suggested their functional relevance existing in each group. Synteny analyses implied conserved and dynamic evolution characteristics of this gene family in several vertebrates. We also found that some recombination events have accelerated the evolution of this gene family. Moreover, a few positive selection sites were identified. Expression patterns of some tetraspanins were further studied under organophosphorus stress using transcriptome sequencing. Functional network analyses identified some interacting genes that exhibited 174 interactions, which reflected the diversity of tetraspanin binding proteins. The results will provide a foundation for the further functional investigation of the tetraspanin genes in fishes.
Collapse
Affiliation(s)
- Jun Cao
- Institute of Life Sciences, Jiangsu University, Zhenjiang, 212013, China.
| | - Xiaona Tan
- Institute of Life Sciences, Jiangsu University, Zhenjiang, 212013, China
| |
Collapse
|
12
|
Florin L, Lang T. Tetraspanin Assemblies in Virus Infection. Front Immunol 2018; 9:1140. [PMID: 29887866 PMCID: PMC5981178 DOI: 10.3389/fimmu.2018.01140] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Accepted: 05/07/2018] [Indexed: 12/23/2022] Open
Abstract
Tetraspanins (Tspans) are a family of four-span transmembrane proteins, known as plasma membrane “master organizers.” They form Tspan-enriched microdomains (TEMs or TERMs) through lateral association with one another and other membrane proteins. If multiple microdomains associate with each other, larger platforms can form. For infection, viruses interact with multiple cell surface components, including receptors, activating proteases, and signaling molecules. It appears that Tspans, such as CD151, CD82, CD81, CD63, CD9, Tspan9, and Tspan7, coordinate these associations by concentrating the interacting partners into Tspan platforms. In addition to mediating viral attachment and entry, these platforms may also be involved in intracellular trafficking of internalized viruses and assist in defining virus assembly and exit sites. In conclusion, Tspans play a role in viral infection at different stages of the virus replication cycle. The present review highlights recently published data on this topic, with a focus on events at the plasma membrane. In light of these findings, we propose a model for how Tspan interactions may organize cofactors for viral infection into distinct molecular platforms.
Collapse
Affiliation(s)
- Luise Florin
- Department of Medical Microbiology and Hygiene, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Thorsten Lang
- Department of Membrane Biochemistry, Life & Medical Sciences Institute, University of Bonn, Bonn, Germany
| |
Collapse
|
13
|
Termini CM, Gillette JM. Tetraspanins Function as Regulators of Cellular Signaling. Front Cell Dev Biol 2017; 5:34. [PMID: 28428953 PMCID: PMC5382171 DOI: 10.3389/fcell.2017.00034] [Citation(s) in RCA: 194] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Accepted: 03/22/2017] [Indexed: 01/10/2023] Open
Abstract
Tetraspanins are molecular scaffolds that distribute proteins into highly organized microdomains consisting of adhesion, signaling, and adaptor proteins. Many reports have identified interactions between tetraspanins and signaling molecules, finding unique downstream cellular consequences. In this review, we will explore these interactions as well as the specific cellular responses to signal activation, focusing on tetraspanin regulation of adhesion-mediated (integrins/FAK), receptor-mediated (EGFR, TNF-α, c-Met, c-Kit), and intracellular signaling (PKC, PI4K, β-catenin). Additionally, we will summarize our current understanding for how tetraspanin post-translational modifications (palmitoylation, N-linked glycosylation, and ubiquitination) can regulate signal propagation. Many of the studies outlined in this review suggest that tetraspanins offer a potential therapeutic target to modulate aberrant signal transduction pathways that directly impact a host of cellular behaviors and disease states.
Collapse
Affiliation(s)
- Christina M Termini
- Department of Pathology, University of New Mexico Health Sciences CenterAlbuquerque, NM, USA
| | - Jennifer M Gillette
- Department of Pathology, University of New Mexico Health Sciences CenterAlbuquerque, NM, USA
| |
Collapse
|
14
|
Determining antigen specificity of a monoclonal antibody using genome-scale CRISPR-Cas9 knockout library. J Immunol Methods 2016; 439:8-14. [DOI: 10.1016/j.jim.2016.09.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2016] [Revised: 09/16/2016] [Accepted: 09/20/2016] [Indexed: 12/26/2022]
|
15
|
Termini CM, Lidke KA, Gillette JM. Tetraspanin CD82 Regulates the Spatiotemporal Dynamics of PKCα in Acute Myeloid Leukemia. Sci Rep 2016; 6:29859. [PMID: 27417454 PMCID: PMC4945921 DOI: 10.1038/srep29859] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 06/22/2016] [Indexed: 02/08/2023] Open
Abstract
Patients with acute myeloid leukemia (AML) have increased myeloid cells within their bone marrow that exhibit aberrant signaling. Therefore, therapeutic targets that modulate disrupted signaling cascades are of significant interest. In this study, we demonstrate that the tetraspanin membrane scaffold, CD82, regulates protein kinase c alpha (PKCα)-mediated signaling critical for AML progression. Utilizing a palmitoylation mutant form of CD82 with disrupted membrane organization, we find that the CD82 scaffold controls PKCα expression and activation. Combining single molecule and ensemble imaging measurements, we determine that CD82 stabilizes PKCα activation at the membrane and regulates the size of PKCα membrane clusters. Further evaluation of downstream effector signaling identified robust and sustained activation of ERK1/2 upon CD82 overexpression that results in enhanced AML colony formation. Together, these data propose a mechanism where CD82 membrane organization regulates sustained PKCα signaling that results in an aggressive leukemia phenotype. These observations suggest that the CD82 scaffold may be a potential therapeutic target for attenuating aberrant signal transduction in AML.
Collapse
Affiliation(s)
- Christina M Termini
- Department of Pathology, University of New Mexico Health Sciences Center, University of New Mexico, MSC 08-4640, Albuquerque, NM 87131, USA
| | - Keith A Lidke
- Department of Physics and Astronomy, University of New Mexico, MSC 07-4220, Albuquerque, NM 87131, USA
| | - Jennifer M Gillette
- Department of Pathology, University of New Mexico Health Sciences Center, University of New Mexico, MSC 08-4640, Albuquerque, NM 87131, USA
| |
Collapse
|
16
|
Hou CY, Lin JHY, Lin SJ, Kuo WC, Lin HT. Down-regulation of CD53 expression in Epinephelus coioides under LPS, poly (I:C), and cytokine stimulation. FISH & SHELLFISH IMMUNOLOGY 2016; 51:143-152. [PMID: 26631805 DOI: 10.1016/j.fsi.2015.11.032] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Revised: 11/22/2015] [Accepted: 11/23/2015] [Indexed: 06/05/2023]
Abstract
Tetraspanins are a group of cell surface molecules involved in cell adhesion, motility, metastasis, signal transduction, and immune cell activation. Members of the tetraspanin family include CD9, CD37, CD63, CD53, and others. However, few tetraspanins have been investigated in teleosts. In this study, we obtained the open reading frame of CD53 cDNA from orange spotted grouper (Epinephelus coioices), an economically important fish. The predicted amino acid structure contains four membrane-spanning domains and a conserved CCG motif. The amino acid identity between human and grouper CD53 was only 38%; however, both CD53 proteins share the same structure. Quantitative real-time PCR revealed that mRNA is abundant in immune organs, including the head and trunk kidneys, spleen, thymus, gill, and blood. Immunochemistry and immunofluorescence analyses further revealed that CD53 was majorly expressed in the leukocytes of various organs. Finally, mRNA and protein expression for CD53 was down-regulated in fish treated with immune stimulators, including LPS, Poly (I:C), Vibrio, recombinant grouper IL-6, and CCL4. Our results indicate that the expression of CD53 may play important roles in pathogen invasion and inflammation reaction.
Collapse
Affiliation(s)
- Chia-Yi Hou
- Institute of Biotechnology, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan 701, Taiwan; Chi Mei Medical Center, Liouying, No.201, Taikang, Liuying Dist., Tainan City 736, Taiwan
| | - John Han-You Lin
- Institute of Biotechnology, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan 701, Taiwan; Department of Biotechnology and Bioindustry Sciences, National Cheng Kung University, Tainan 701, Taiwan; Research Center of Agricultural Biotechnology, National Cheng Kung University, Tainan 701, Taiwan; Center of Biosciences, National Cheng Kung University, Tainan 701, Taiwan
| | - Shih-Jie Lin
- Institute of Biotechnology, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan 701, Taiwan
| | - Wan-Ching Kuo
- Institute of Biotechnology, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan 701, Taiwan
| | - Han-Tso Lin
- Department of Biotechnology, Ming Chuan University, Taoyuan County 333, Taiwan.
| |
Collapse
|
17
|
Molecular Mechanisms of HTLV-1 Cell-to-Cell Transmission. Viruses 2016; 8:74. [PMID: 27005656 PMCID: PMC4810264 DOI: 10.3390/v8030074] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 02/20/2016] [Accepted: 03/04/2016] [Indexed: 12/14/2022] Open
Abstract
The tumorvirus human T-cell lymphotropic virus type 1 (HTLV-1), a member of the delta-retrovirus family, is transmitted via cell-containing body fluids such as blood products, semen, and breast milk. In vivo, HTLV-1 preferentially infects CD4⁺ T-cells, and to a lesser extent, CD8⁺ T-cells, dendritic cells, and monocytes. Efficient infection of CD4⁺ T-cells requires cell-cell contacts while cell-free virus transmission is inefficient. Two types of cell-cell contacts have been described to be critical for HTLV-1 transmission, tight junctions and cellular conduits. Further, two non-exclusive mechanisms of virus transmission at cell-cell contacts have been proposed: (1) polarized budding of HTLV-1 into synaptic clefts; and (2) cell surface transfer of viral biofilms at virological synapses. In contrast to CD4⁺ T-cells, dendritic cells can be infected cell-free and, to a greater extent, via viral biofilms in vitro. Cell-to-cell transmission of HTLV-1 requires a coordinated action of steps in the virus infectious cycle with events in the cell-cell adhesion process; therefore, virus propagation from cell-to-cell depends on specific interactions between cellular and viral proteins. Here, we review the molecular mechanisms of HTLV-1 transmission with a focus on the HTLV-1-encoded proteins Tax and p8, their impact on host cell factors mediating cell-cell contacts, cytoskeletal remodeling, and thus, virus propagation.
Collapse
|
18
|
Marjon KD, Termini CM, Karlen KL, Saito-Reis C, Soria CE, Lidke KA, Gillette JM. Tetraspanin CD82 regulates bone marrow homing of acute myeloid leukemia by modulating the molecular organization of N-cadherin. Oncogene 2015; 35:4132-40. [PMID: 26592446 PMCID: PMC4877306 DOI: 10.1038/onc.2015.449] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Revised: 09/18/2015] [Accepted: 10/22/2015] [Indexed: 11/16/2022]
Abstract
Communication between acute myeloid leukemia (AML) and the bone marrow microenvironment is known to control disease progression. Therefore, regulation of AML cell trafficking and adhesion to the bone marrow is of significant interest. In this study, we demonstrate that differential expression of the membrane scaffold CD82 modulates the bone marrow homing of AML cells. By combining mutational analysis and super-resolution imaging, we identify membrane protein clustering by CD82 as a regulator of AML cell adhesion and bone marrow homing. Cluster analysis of super-resolution data indicates that N-linked glycosylation and palmitoylation of CD82 are both critical modifications that control the microdomain organization of CD82 as well as the nanoscale clustering of associated adhesion protein, N-cadherin. We demonstrate that inhibition of CD82 glycosylation increases the molecular packing of N-cadherin and promotes the bone marrow homing of AML cells. In contrast, we find that inhibition of CD82 palmitoylation disrupts the formation and organization of N-cadherin clusters and significantly diminishes bone marrow trafficking of AML. Taken together, these data establish a mechanism where the membrane organization of CD82, through specific post-translational modifications, regulates N-cadherin clustering and membrane density, which impacts the in vivo trafficking of AML cells. As such, these observations provide an alternative model for targeting AML where modulation of protein organization within the membrane may be an effective treatment therapy to disrupt the bone marrow homing potential of AML cells.
Collapse
Affiliation(s)
- K D Marjon
- Department of Pathology, University of New Mexico Health Sciences Center, University of New Mexico, Albuquerque, NM, USA
| | - C M Termini
- Department of Pathology, University of New Mexico Health Sciences Center, University of New Mexico, Albuquerque, NM, USA
| | - K L Karlen
- Department of Pathology, University of New Mexico Health Sciences Center, University of New Mexico, Albuquerque, NM, USA
| | - C Saito-Reis
- Department of Pathology, University of New Mexico Health Sciences Center, University of New Mexico, Albuquerque, NM, USA
| | - C E Soria
- Department of Pathology, University of New Mexico Health Sciences Center, University of New Mexico, Albuquerque, NM, USA
| | - K A Lidke
- Department of Physics and Astronomy, University of New Mexico, Albuquerque, NM, USA
| | - J M Gillette
- Department of Pathology, University of New Mexico Health Sciences Center, University of New Mexico, Albuquerque, NM, USA
| |
Collapse
|
19
|
Rizkallah G, Mahieux R, Dutartre H. [Intercellular transmission of HTLV-1: not all mechanisms have been revealed]. Med Sci (Paris) 2015; 31:629-37. [PMID: 26152167 DOI: 10.1051/medsci/20153106016] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
HTLV-1 is the etiological agent of Adult T cell Leukemia/Lymphoma (ATLL) and of HTLV-1-Associated Myelopathy/tropical spastic paraparesis (HAM/TSP). It is mainly detected in CD4+ lymphocytes in vivo, but proviral genomes have also been detected although less frequently, in CD8+ T lymphocytes, B lymphocytes, monocytes, macrophages, dendritic cells and other non-lymphoid cells. Virus spread is highly dependent on cell-cell contact. This mode of transmission is correlated with an increased ability of infected cells to migrate, a property linked to cytoskeleton reorganization induced by the viral Tax protein. Cell-to-cell transmission relies on at least three non-exclusive molecular pathways. First, a specialized area, the "virological synapse'' (VS) promotes direct transmission of budding HTLV-1 particles into a synaptic cleft formed between infected and uninfected cells. Second, HTLV-1 particles accumulate at the plasma membrane of infected cells in a biofilm-like extracellular viral assembly that resembles a bacterial biofilm. Viral biofilm is rapidly transmitted to uninfected cells when infected cells contact target cells. Finally, membrane extensions called inter-cellular conduits facilitate HTLV-1 proteins transfer from infected to uninfected target cells, and may stabilize cell-cell contacts. The aim of this review is to summarize the molecular mechanisms of these HTLV-1 transmission pathways.
Collapse
Affiliation(s)
- Gerges Rizkallah
- Équipe oncogenèse rétrovirale ; équipe labellisée Ligue nationale contre le cancer ; centre international de recherche en infectiologie, Inserm U1111-CNRS UMR5308, Lyon, France - École normale supérieure de Lyon, 46, allée d'Italie, 69007 Lyon, France - Université Lyon 1, Lyon Cedex 07, 69364, France
| | - Renaud Mahieux
- Équipe oncogenèse rétrovirale ; équipe labellisée Ligue nationale contre le cancer ; centre international de recherche en infectiologie, Inserm U1111-CNRS UMR5308, Lyon, France - École normale supérieure de Lyon, 46, allée d'Italie, 69007 Lyon, France - Université Lyon 1, Lyon Cedex 07, 69364, France
| | - Hélène Dutartre
- Équipe oncogenèse rétrovirale ; équipe labellisée Ligue nationale contre le cancer ; centre international de recherche en infectiologie, Inserm U1111-CNRS UMR5308, Lyon, France - École normale supérieure de Lyon, 46, allée d'Italie, 69007 Lyon, France - Université Lyon 1, Lyon Cedex 07, 69364, France
| |
Collapse
|
20
|
Termini CM, Cotter ML, Marjon KD, Buranda T, Lidke KA, Gillette JM. The membrane scaffold CD82 regulates cell adhesion by altering α4 integrin stability and molecular density. Mol Biol Cell 2014; 25:1560-73. [PMID: 24623721 PMCID: PMC4019488 DOI: 10.1091/mbc.e13-11-0660] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Hematopoietic stem/progenitor cell (HSPC) interactions with the bone marrow microenvironment are important for maintaining HSPC self-renewal and differentiation. In recent work, we identified the tetraspanin protein, CD82, as a regulator of HPSC adhesion and homing to the bone marrow, although the mechanism by which CD82 mediated adhesion was unclear. In the present study, we determine that CD82 expression alters cell-matrix adhesion, as well as integrin surface expression. By combining the superresolution microscopy imaging technique, direct stochastic optical reconstruction microscopy, with protein clustering algorithms, we identify a critical role for CD82 in regulating the membrane organization of α4 integrin subunits. Our data demonstrate that CD82 overexpression increases the molecular density of α4 within membrane clusters, thereby increasing cellular adhesion. Furthermore, we find that the tight packing of α4 into membrane clusters depend on CD82 palmitoylation and the presence of α4 integrin ligands. In combination, these results provide unique quantifiable evidence of CD82's contribution to the spatial arrangement of integrins within the plasma membrane and suggest that regulation of integrin density by tetraspanins is a critical component of cell adhesion.
Collapse
Affiliation(s)
- Christina M Termini
- Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131
| | - Maura L Cotter
- Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131
| | - Kristopher D Marjon
- Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131
| | - Tione Buranda
- Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131
| | - Keith A Lidke
- Department of Physics and Astronomy, University of New Mexico, Albuquerque, NM 87131
| | - Jennifer M Gillette
- Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131
| |
Collapse
|
21
|
Fogarty KH, Berk S, Grigsby IF, Chen Y, Mansky LM, Mueller JD. Interrelationship between cytoplasmic retroviral Gag concentration and Gag-membrane association. J Mol Biol 2013; 426:1611-24. [PMID: 24316368 DOI: 10.1016/j.jmb.2013.11.025] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Revised: 11/12/2013] [Accepted: 11/23/2013] [Indexed: 12/20/2022]
Abstract
The early events in the retrovirus assembly pathway, particularly the timing and nature of Gag translocation from the site of protein translation to the inner leaflet of the plasma membrane, are poorly understood. We have investigated the interrelationship between cytoplasmic Gag concentration and plasma membrane association using complementary live-cell biophysical fluorescence techniques in real time with both human T-cell leukemia virus type 1 (HTLV-1) and human immunodeficiency virus type 1 (HIV-1) Gag proteins. In particular, dual-color, z-scan fluorescence fluctuation spectroscopy in conjunction with total internal reflection fluorescence and conventional, epi-illumination imaging were utilized. Our results demonstrate that HTLV-1 Gag is capable of membrane targeting and particle assembly at low (i.e., nanomolar) cytoplasmic concentrations and that there is a critical threshold concentration (approaching micromolar) prior to the observation of HIV-1 Gag associated with the plasma membrane. These observations imply fundamental differences between HIV-1 and HTLV-1 Gag trafficking and membrane association.
Collapse
Affiliation(s)
- Keir H Fogarty
- Institute for Molecular Virology, University of Minnesota, Minneapolis, MN 55455, USA; Department of Diagnostic and Biological Sciences, School of Dentistry, University of Minnesota, Minneapolis, MN 55455, USA; School of Physics and Astronomy, University of Minnesota, Minneapolis, MN 55455, USA
| | - Serkan Berk
- Institute for Molecular Virology, University of Minnesota, Minneapolis, MN 55455, USA; Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | - Iwen F Grigsby
- Institute for Molecular Virology, University of Minnesota, Minneapolis, MN 55455, USA; Department of Diagnostic and Biological Sciences, School of Dentistry, University of Minnesota, Minneapolis, MN 55455, USA
| | - Yan Chen
- Institute for Molecular Virology, University of Minnesota, Minneapolis, MN 55455, USA; School of Physics and Astronomy, University of Minnesota, Minneapolis, MN 55455, USA
| | - Louis M Mansky
- Institute for Molecular Virology, University of Minnesota, Minneapolis, MN 55455, USA; Department of Diagnostic and Biological Sciences, School of Dentistry, University of Minnesota, Minneapolis, MN 55455, USA; Department of Microbiology, University of Minnesota, Minneapolis, MN 55455, USA.
| | - Joachim D Mueller
- Institute for Molecular Virology, University of Minnesota, Minneapolis, MN 55455, USA; School of Physics and Astronomy, University of Minnesota, Minneapolis, MN 55455, USA; Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
22
|
Mazurov D, Barbashova L, Filatov A. Tetraspanin protein CD9 interacts with metalloprotease CD10 and enhances its release via exosomes. FEBS J 2013; 280:1200-13. [PMID: 23289620 DOI: 10.1111/febs.12110] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2012] [Revised: 12/27/2012] [Accepted: 01/01/2013] [Indexed: 12/11/2022]
Abstract
Tetraspanins interact with a wide variety of transmembrane and intracellular proteins called molecular partners, and modulate their function. In this article, we describe a new partner of tetraspanin web, membrane metalloprotease CD10, which is selectively associated with CD9. By constructing chimeras between tetraspanins CD9 and CD82 (the latter does not interact with CD10) or by using site-directed mutagenesis, we determined that a portion of the large extracellular loop from the CCG motif to transmembrane domain 4, as well as the C-terminal tail of CD9, are involved in the interaction with CD10. The stable expression of wild-type CD9 in K562 CD10-positive cells enhanced the level of CD10 released with exosomes five-fold. In contrast, the expression of chimeric CD9, which contained the cytoplasmic C-terminal domain from CD82, had little effect on CD10 release. Short hairpin RNA knockdown of CD9 expression in Nalm-6 pre-B cells resulted in a two-fold reduction in the amount of endogenous CD10 released with microvesicles. The peptidase activity of CD10 measured either on cells or on exosomes correlated with the level of CD10 expression, and was not significantly modulated by CD9 expression as such. Our data suggest that the interaction of CD10 with tetraspanin CD9 can play an important role in the redistribution of peptidase activity from the cell surface to outer microenvironments. In bone marrow, where CD10 presumably contributes to the maturation of pre-B cells and migration of B cells to the blood circulation, release of CD10 peptidase activity with exosomes may effectively regulate extracellular matrix microenvironments.
Collapse
Affiliation(s)
- Dmitriy Mazurov
- Laboratory of Immunochemistry, Institute of Immunology, Moscow, Russia.
| | | | | |
Collapse
|
23
|
Wang F, Vandepoele K, Van Lijsebettens M. Tetraspanin genes in plants. PLANT SCIENCE : AN INTERNATIONAL JOURNAL OF EXPERIMENTAL PLANT BIOLOGY 2012; 190:9-15. [PMID: 22608515 DOI: 10.1016/j.plantsci.2012.03.005] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2012] [Revised: 03/14/2012] [Accepted: 03/16/2012] [Indexed: 05/19/2023]
Abstract
Tetraspanins represent a four-transmembrane protein superfamily with a conserved structure and amino acid residues that are present in mammals, insects, fungi and plants. Tetraspanins interact with each other or with other membrane proteins to form tetraspanin-enriched microdomains that play important roles in development, pathogenesis and immune responses via facilitating cell-cell adhesion and fusion, ligand binding and intracellular trafficking. Here, we emphasize evolutionary aspects within the plant kingdom based on genomic sequence information. A phylogenetic tree based on 155 tetraspanin genes of 11 plant species revealed ancient and fast evolving clades. Tetraspanins were only present in multicellular plants, were often duplicated in the plant genomes and predicted by the electronic Fluorescent Pictograph for gene expression analysis to be either functionally redundant or divergent. Tetraspanins contain a large extracellular loop with conserved cysteines that provide the binding sites for the interactions. The Arabidopsis thaliana TETRASPANIN1/TORNADO2/EKEKO has a function in leaf and root patterning and TETRASPANIN3 was identified in the plasmodesmatal proteome, suggesting a role in cell-cell communication during plant development.
Collapse
Affiliation(s)
- Feng Wang
- Department of Plant Systems Biology, VIB, Technologiepark 927, B-9052 Gent, Belgium
| | | | | |
Collapse
|
24
|
Gui L, Wang B, Li FH, Sun YM, Luo Z, Xiang JH. Blocking the large extracellular loop (LEL) domain of FcTetraspanin-3 could inhibit the infection of white spot syndrome virus (WSSV) in Chinese shrimp, Fenneropenaeus chinensis. FISH & SHELLFISH IMMUNOLOGY 2012; 32:1008-15. [PMID: 22406449 DOI: 10.1016/j.fsi.2012.02.022] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2011] [Revised: 02/18/2012] [Accepted: 02/19/2012] [Indexed: 05/23/2023]
Abstract
Tetraspanins belong to the transmembrane 4 superfamily (TM(4)SF), which span the cell membrane 4 times and act as bridges or connectors. Increasing evidences have shown that tetraspanins play important role in virus infection. The large extracellular loop (LEL) of a tetraspanin is considered as a possible target of some virus. Tetraspanins are widely found in invertebrates, but the functional roles of most invertebrate tetraspanins have remained unknown. Recently, a tetraspanin, called FcTetraspanin-3, was cloned from the cDNA library of Chinese shrimp, Fenneropenaeus chinensis. The FcTetraspanin-3 constitutive expression in all examined tissues and the expression of the gene were highly induced in hepatopancreas, lymphoid organ and intestine by white spot syndrome virus (WSSV) challenge. In this study, we expressed and purified the recombinant peptide containing the LEL domain of FcTetraspanin-3, and produced the anti-LEL polyclone antibody. The expression of FcTetraspanin-3 was observed by real-time PCR and Western blot. Also, the localization of FcTetraspanin-3-positive cells in intestine and hepatopancreas were revealed by immunofluorescence. The results of anti-LEL antibody blocking experiments shown that the antibody can significantly reduce the mortality of shrimp challenged by WSSV. Additionally, dsRNA interference was utilized to examine the functional role of FcTetraspanin-3 in response to WSSV infection, and a sensible decrease of the viral copy number in the tetraspanin knockdown shrimp. These results suggested the blocking of LEL domain of FcTetraspanin-3 could inhibit the infection of WSSV. FcTetraspanin-3 might play an important role in response to WSSV infection, and the LEL domain of FcTetraspanin-3 might mediate the entry of WSSV.
Collapse
Affiliation(s)
- Lang Gui
- Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, PR China
| | | | | | | | | | | |
Collapse
|
25
|
Role of O-glycosylation and expression of CD43 and CD45 on the surfaces of effector T cells in human T cell leukemia virus type 1 cell-to-cell infection. J Virol 2011; 86:2447-58. [PMID: 22171268 DOI: 10.1128/jvi.06993-11] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
We used replication-dependent retroviral vectors to identify cell surface antigens involved in the cell-to-cell transmission of human T cell leukemia virus type 1 (HTLV-1). We generated monoclonal antibodies (MAbs) against Jurkat T cells and selected several IgM MAbs that strongly inhibited HTLV-1 but not human immune deficiency virus type 1 (HIV-1) cell-to-cell infection. These MAbs recognized the so-called Tn antigen (GalNAcα1-O-Ser/Thr) that arises on Jurkat cells from a mutation in the T-synthase-specific chaperone Cosmc and the consequent loss of O-glycan elongation. Anti-Tn MAbs precipitated two major O-glycan carrier proteins, CD43 and CD45, and caused a strong aggregation of Jurkat cells. The restoration of O-glycosylation in Jurkat cells by stably transducing the wild-type Cosmc gene resulted in a 3- to 4-fold increase in the level of surface expression of CD43 and enhanced HTLV-1 transmission 10-fold in comparison to that of parental cells. The short hairpin RNA (shRNA) knockdown of CD43 or CD45 expression in Jurkat-Cosmc, HBP-ALL, and CEM T cells decreased HTLV-1 infection severalfold. The knockdown of CD45 in Jurkat cells severely reduced both HTLV-1 and HIV-1 infections, but Cosmc coexpression partially rescued infection. HTLV-1 proteins, which assembled in small patches on Jurkat cells, formed large clusters on the surface of Jurkat-Cosmc cells. These data indicate that large aggregates of HTLV-1 assemblies are more infectious than multiple clustered virions. We suggest that heavily O-glycosylated CD43 and CD45 molecules render cells less adhesive, prevent inappropriate cell-cell contacts, and favor the assembly of HTLV-1 particles into large, highly infectious structures on the surface of T cells.
Collapse
|
26
|
Abstract
Cells secrete various membrane-enclosed microvesicles from their cell surface (shedding microvesicles) and from internal, endosome-derived membranes (exosomes). Intriguingly, these vesicles have many characteristics in common with enveloped viruses, including biophysical properties, biogenesis, and uptake by cells. Recent discoveries describing the microvesicle-mediated intercellular transfer of functional cellular proteins, RNAs, and mRNAs have revealed additional similarities between viruses and cellular microvesicles. Apparent differences include the complexity of viral entry, temporally regulated viral expression, and self-replication proceeding to infection of new cells. Interestingly, many virally infected cells secrete microvesicles that differ in content from their virion counterparts but may contain various viral proteins and RNAs. For the most part, these particles have not been analyzed for their content or functions during viral infection. However, early studies of microvesicles (L-particles) secreted from herpes simplex virus-infected cells provided the first evidence of microvesicle-mediated intercellular communication. In the case of Epstein-Barr virus, recent evidence suggests that this tumorigenic herpesvirus also utilizes exosomes as a mechanism of cell-to-cell communication through the transfer of signaling competent proteins and functional microRNAs to uninfected cells. This review focuses on aspects of the biology of microvesicles with an emphasis on their potential contributions to viral infection and pathogenesis.
Collapse
|
27
|
Fogarty KH, Zhang W, Grigsby IF, Johnson JL, Chen Y, Mueller JD, Mansky LM. New insights into HTLV-1 particle structure, assembly, and Gag-Gag interactions in living cells. Viruses 2011; 3:770-93. [PMID: 21994753 PMCID: PMC3185773 DOI: 10.3390/v3060770] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2011] [Revised: 05/20/2011] [Accepted: 05/20/2011] [Indexed: 11/16/2022] Open
Abstract
Human T-cell leukemia virus type 1 (HTLV-1) has a reputation for being extremely difficult to study in cell culture. The challenges in propagating HTLV-1 has prevented a rigorous analysis of how these viruses replicate in cells, including the detailed steps involved in virus assembly. The details for how retrovirus particle assembly occurs are poorly understood, even for other more tractable retroviral systems. Recent studies on HTLV-1 using state-of-the-art cryo-electron microscopy and fluorescence-based biophysical approaches explored questions related to HTLV-1 particle size, Gag stoichiometry in virions, and Gag-Gag interactions in living cells. These results provided new and exciting insights into fundamental aspects of HTLV-1 particle assembly-which are distinct from those of other retroviruses, including HIV-1. The application of these and other novel biophysical approaches promise to provide exciting new insights into HTLV-1 replication.
Collapse
Affiliation(s)
- Keir H. Fogarty
- Institute for Molecular Virology, University of Minnesota, Minneapolis, 18-242 Moos Tower, 515 Delaware St. SE, Minneapolis, MN 55455, USA; E-Mails: (K.H.F.); (W.Z.); (I.F.G.); (Y.C.); (J.D.M.)
- School of Physics and Astronomy, University of Minnesota, Minneapolis, MN 55455, USA; E-Mail: (J.L.J.)
| | - Wei Zhang
- Institute for Molecular Virology, University of Minnesota, Minneapolis, 18-242 Moos Tower, 515 Delaware St. SE, Minneapolis, MN 55455, USA; E-Mails: (K.H.F.); (W.Z.); (I.F.G.); (Y.C.); (J.D.M.)
- Department of Diagnostic and Biological Sciences, School of Dentistry, University of Minnesota, Minneapolis, MN 55455, USA
| | - Iwen F. Grigsby
- Institute for Molecular Virology, University of Minnesota, Minneapolis, 18-242 Moos Tower, 515 Delaware St. SE, Minneapolis, MN 55455, USA; E-Mails: (K.H.F.); (W.Z.); (I.F.G.); (Y.C.); (J.D.M.)
- Department of Diagnostic and Biological Sciences, School of Dentistry, University of Minnesota, Minneapolis, MN 55455, USA
| | - Jolene L. Johnson
- School of Physics and Astronomy, University of Minnesota, Minneapolis, MN 55455, USA; E-Mail: (J.L.J.)
| | - Yan Chen
- Institute for Molecular Virology, University of Minnesota, Minneapolis, 18-242 Moos Tower, 515 Delaware St. SE, Minneapolis, MN 55455, USA; E-Mails: (K.H.F.); (W.Z.); (I.F.G.); (Y.C.); (J.D.M.)
- School of Physics and Astronomy, University of Minnesota, Minneapolis, MN 55455, USA; E-Mail: (J.L.J.)
| | - Joachim D. Mueller
- Institute for Molecular Virology, University of Minnesota, Minneapolis, 18-242 Moos Tower, 515 Delaware St. SE, Minneapolis, MN 55455, USA; E-Mails: (K.H.F.); (W.Z.); (I.F.G.); (Y.C.); (J.D.M.)
- School of Physics and Astronomy, University of Minnesota, Minneapolis, MN 55455, USA; E-Mail: (J.L.J.)
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | - Louis M. Mansky
- Institute for Molecular Virology, University of Minnesota, Minneapolis, 18-242 Moos Tower, 515 Delaware St. SE, Minneapolis, MN 55455, USA; E-Mails: (K.H.F.); (W.Z.); (I.F.G.); (Y.C.); (J.D.M.)
- Department of Diagnostic and Biological Sciences, School of Dentistry, University of Minnesota, Minneapolis, MN 55455, USA
- Department of Microbiology, Medical School, University of Minnesota, Minneapolis, MN 55455, USA
| |
Collapse
|
28
|
Inlora J, Chukkapalli V, Derse D, Ono A. Gag localization and virus-like particle release mediated by the matrix domain of human T-lymphotropic virus type 1 Gag are less dependent on phosphatidylinositol-(4,5)-bisphosphate than those mediated by the matrix domain of HIV-1 Gag. J Virol 2011; 85:3802-10. [PMID: 21289126 PMCID: PMC3126146 DOI: 10.1128/jvi.02383-10] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2010] [Accepted: 01/26/2011] [Indexed: 12/16/2022] Open
Abstract
The human immunodeficiency virus type 1 (HIV-1) Gag matrix (MA) domain facilitates Gag targeting and binding to the plasma membrane (PM) during virus assembly. Interaction with a PM phospholipid, phosphatidylinositol-(4,5)-bisphosphate [PI(4,5)P(2)], plays a key role in these MA functions. Previous studies showed that overexpression of polyphosphoinositide 5-phosphatase IV (5ptaseIV), which depletes cellular PI(4,5)P(2), mislocalizes HIV-1 Gag to the cytosol and greatly reduces HIV-1 release efficiency. In this study, we sought to determine the role of the MA-PI(4,5)P(2) interaction in Gag localization and membrane binding of a deltaretrovirus, human T-lymphotropic virus type 1 (HTLV-1). We compared the chimeric HIV-1 Gag (HTMA), in which MA was replaced with HTLV-1 MA, with wild-type HIV-1 and HTLV-1 Gag for PI(4,5)P(2) dependence. Our results demonstrate that, unlike HIV-1 Gag, subcellular localization of and VLP release by HTLV-1 and HTMA Gag were minimally sensitive to 5ptaseIV overexpression. These results suggest that the interaction of HTLV-1 MA with PI(4,5)P(2) is not essential for HTLV-1 particle assembly. Furthermore, liposome-binding analyses showed that both HTLV-1 and HTMA Gag can bind membrane efficiently even in the absence of PI(4,5)P(2). Efficient HTLV-1 Gag binding to liposomes was largely driven by electrostatic interaction, unlike that of HIV-1 Gag, which required specific interaction with PI(4,5)P(2). Furthermore, membrane binding of HTLV-1 Gag in vitro was not suppressed by RNA, in contrast to HIV-1 Gag. Altogether, our data suggest that Gag targeting and membrane binding mediated by HTLV-1 MA does not require PI(4,5)P(2) and that distinct mechanisms regulate HIV-1 and HTLV-1 Gag membrane binding.
Collapse
Affiliation(s)
- Jingga Inlora
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Vineela Chukkapalli
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan
| | - David Derse
- National Cancer Institute at Frederick, HIV Drug Resistance Program, Frederick, Maryland
| | - Akira Ono
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan
| |
Collapse
|
29
|
Hamard-Peron E, Muriaux D. Retroviral matrix and lipids, the intimate interaction. Retrovirology 2011; 8:15. [PMID: 21385335 PMCID: PMC3059298 DOI: 10.1186/1742-4690-8-15] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2010] [Accepted: 03/07/2011] [Indexed: 11/30/2022] Open
Abstract
Retroviruses are enveloped viruses that assemble on the inner leaflet of cellular membranes. Improving biophysical techniques has recently unveiled many molecular aspects of the interaction between the retroviral structural protein Gag and the cellular membrane lipids. This interaction is driven by the N-terminal matrix domain of the protein, which probably undergoes important structural modifications during this process, and could induce membrane lipid distribution changes as well. This review aims at describing the molecular events occurring during MA-membrane interaction, and pointing out their consequences in terms of viral assembly. The striking conservation of the matrix membrane binding mode among retroviruses indicates that this particular step is most probably a relevant target for antiviral research.
Collapse
Affiliation(s)
- Elise Hamard-Peron
- Human Virology Department, Inserm U758, Ecole Normale Superieure de Lyon, 36 Allee d'Italie, IFR128, Universite de Lyon, Lyon, France
| | | |
Collapse
|
30
|
Grigsby IF, Zhang W, Johnson JL, Fogarty KH, Chen Y, Rawson JM, Crosby AJ, Mueller JD, Mansky LM. Biophysical analysis of HTLV-1 particles reveals novel insights into particle morphology and Gag stochiometry. Retrovirology 2010; 7:75. [PMID: 20854688 PMCID: PMC2954917 DOI: 10.1186/1742-4690-7-75] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2010] [Accepted: 09/20/2010] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Human T-lymphotropic virus type 1 (HTLV-1) is an important human retrovirus that is a cause of adult T-cell leukemia/lymphoma. While an important human pathogen, the details regarding virus replication cycle, including the nature of HTLV-1 particles, remain largely unknown due to the difficulties in propagating the virus in tissue culture. In this study, we created a codon-optimized HTLV-1 Gag fused to an EYFP reporter as a model system to quantitatively analyze HTLV-1 particles released from producer cells. RESULTS The codon-optimized Gag led to a dramatic and highly robust level of Gag expression as well as virus-like particle (VLP) production. The robust level of particle production overcomes previous technical difficulties with authentic particles and allowed for detailed analysis of particle architecture using two novel methodologies. We quantitatively measured the diameter and morphology of HTLV-1 VLPs in their native, hydrated state using cryo-transmission electron microscopy (cryo-TEM). Furthermore, we were able to determine HTLV-1 Gag stoichiometry as well as particle size with the novel biophysical technique of fluorescence fluctuation spectroscopy (FFS). The average HTLV-1 particle diameter determined by cryo-TEM and FFS was 71 ± 20 nm and 75 ± 4 nm, respectively. These values are significantly smaller than previous estimates made of HTLV-1 particles by negative staining TEM. Furthermore, cryo-TEM reveals that the majority of HTLV-1 VLPs lacks an ordered structure of the Gag lattice, suggesting that the HTLV-1 Gag shell is very likely to be organized differently compared to that observed with HIV-1 Gag in immature particles. This conclusion is supported by our observation that the average copy number of HTLV-1 Gag per particle is estimated to be 510 based on FFS, which is significantly lower than that found for HIV-1 immature virions. CONCLUSIONS In summary, our studies represent the first quantitative biophysical analysis of HTLV-1-like particles and reveal novel insights into particle morphology and Gag stochiometry.
Collapse
Affiliation(s)
- Iwen F Grigsby
- Institute for Molecular Virology, University of Minnesota, Minneapolis, MN 55455, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Krementsov DN, Rassam P, Margeat E, Roy NH, Schneider-Schaulies J, Milhiet PE, Thali M. HIV-1 Assembly Differentially Alters Dynamics and Partitioning of Tetraspanins and Raft Components. Traffic 2010; 11:1401-14. [DOI: 10.1111/j.1600-0854.2010.01111.x] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
32
|
Hassuna N, Monk PN, Moseley GW, Partridge LJ. Strategies for targeting tetraspanin proteins: potential therapeutic applications in microbial infections. BioDrugs 2010; 23:341-59. [PMID: 19894777 PMCID: PMC7100176 DOI: 10.2165/11315650-000000000-00000] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
The identification of novel targets and strategies for therapy of microbial infections is an area of intensive research due to the failure of conventional vaccines or antibiotics to combat both newly emerging diseases (e.g. viruses such as severe acute respiratory syndrome (SARS) and new influenza strains, and antibiotic-resistant bacteria) and entrenched, pandemic diseases exemplified by HIV. One clear approach to this problem is to target processes of the host organism rather than the microbe. Recent data have indicated that members of the tetraspanin superfamily, proteins with a widespread distribution in eukaryotic organisms and 33 members in humans, may provide such an approach. Tetraspanins traverse the membrane four times, but are distinguished from other four-pass membrane proteins by the presence of conserved charged residues in the transmembrane domains and a defining ‘signature’ motif in the larger of the two extracellular domains (the EC2). They characteristically form promiscuous associations with one another and with other membrane proteins and lipids to generate a specialized type of microdomain: the tetraspanin-enriched microdomain (TEM). TEMs are integral to the main role of tetraspanins as ‘molecular organizers’ involved in functions such as membrane trafficking, cell-cell fusion, motility, and signaling. Increasing evidence demonstrates that tetraspanins are used by intracellular pathogens as a means of entering and replicating within human cells. Although previous investigations focused mainly on viruses such as hepatitis C and HIV, it is now becoming clear that other microbes associate with tetraspanins, using TEMs as a ‘gateway’ to infection. In this article we review the properties and functions of tetraspanins/TEMs that are relevant to infective processes and discuss the accumulating evidence that shows how different pathogens exploit these properties in infection and in the pathogenesis of disease. We then investigate the novel and exciting possibilities of targeting tetraspanins for the treatment of infectious disease, using specific antibodies, recombinant EC2 domains, small-molecule mimetics, and small interfering RNA. Such therapies, directed at host-cell molecules, may provide alternative options for combating fast-mutating or newly emerging pathogens, where conventional approaches face difficulties.
Collapse
Affiliation(s)
- Noha Hassuna
- Department of Molecular Biology and Biotechnology, Krebs Institute, University of Sheffield, Sheffield, UK
| | | | | | | |
Collapse
|
33
|
Wang L, Liu L, Che Y, Wang L, Jiang L, Dong C, Zhang Y, Li Q. Egress of HSV-1 capsid requires the interaction of VP26 and a cellular tetraspanin membrane protein. Virol J 2010; 7:156. [PMID: 20630051 PMCID: PMC2913958 DOI: 10.1186/1743-422x-7-156] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2010] [Accepted: 07/14/2010] [Indexed: 01/03/2023] Open
Abstract
HSV-1 viral capsid maturation and egress from the nucleus constitutes a self-controlled process of interactions between host cytoplasmic membrane proteins and viral capsid proteins. In this study, a member of the tetraspanin superfamily, CTMP-7, was shown to physically interact with HSV-1 protein VP26, and the VP26-CTMP-7 complex was detected both in vivo and in vitro. The interaction of VP26 with CTMP-7 plays an essential role in normal HSV-1 replication. Additionally, analysis of a recombinant virus HSV-1-UG showed that mutating VP26 resulted in a decreased viral replication rate and in aggregation of viral mutant capsids in the nucleus. Together, our data support the notion that biological events mediated by a VP26 - CTMP-7 interaction aid in viral capsid enveloping and egress from the cell during the HSV-1 infectious process.
Collapse
Affiliation(s)
- Lei Wang
- Institute Of Medical Biology, Chinese Academy of Medicine Science, Peking Union Medical College, Kunming 650118, P. R. China
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Abstract
Tetraspanins are small integral membrane proteins that are known to control a variety of cellular processes, including signaling, migration and cell-cell fusion. Research over the past few years established that they are also regulators of various steps in the HIV-1 replication cycle, but the mechanisms through which these proteins either enhance or repress virus spread remain largely unknown.
Collapse
Affiliation(s)
- Markus Thali
- Department of Microbiology and Molecular Genetics, College of Medicine and CALS, University of Vermont, 318 Stafford Hall, 95 Carrigan Drive, Burlington, VT 05405-0084, USA.
| |
Collapse
|
35
|
Mazurov D, Ilinskaya A, Heidecker G, Lloyd P, Derse D. Quantitative comparison of HTLV-1 and HIV-1 cell-to-cell infection with new replication dependent vectors. PLoS Pathog 2010; 6:e1000788. [PMID: 20195464 PMCID: PMC2829072 DOI: 10.1371/journal.ppat.1000788] [Citation(s) in RCA: 169] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2008] [Accepted: 01/22/2010] [Indexed: 02/03/2023] Open
Abstract
We have developed an efficient method to quantify cell-to-cell infection with single-cycle, replication dependent reporter vectors. This system was used to examine the mechanisms of infection with HTLV-1 and HIV-1 vectors in lymphocyte cell lines. Effector cells transfected with reporter vector, packaging vector, and Env expression plasmid produced virus-like particles that transduced reporter gene activity into cocultured target cells with zero background. Reporter gene expression was detected exclusively in target cells and required an Env-expression plasmid and a viral packaging vector, which provided essential structural and enzymatic proteins for virus replication. Cell-cell fusion did not contribute to infection, as reporter protein was rarely detected in syncytia. Coculture of transfected Jurkat T cells and target Raji/CD4 B cells enhanced HIV-1 infection two fold and HTLV-1 infection ten thousand fold in comparison with cell-free infection of Raji/CD4 cells. Agents that interfere with actin and tubulin polymerization strongly inhibited HTLV-1 and modestly decreased HIV-1 cell-to-cell infection, an indication that cytoskeletal remodeling was more important for HTLV-1 transmission. Time course studies showed that HTLV-1 transmission occurred very rapidly after cell mixing, whereas slower kinetics of HIV-1 coculture infection implies a different mechanism of infectious transmission. HTLV-1 Tax was demonstrated to play an important role in altering cell-cell interactions that enhance virus infection and replication. Interestingly, superantigen-induced synapses between Jurkat cells and Raji/CD4 cells did not enhance infection for either HTLV-1 or HIV-1. In general, the dependence on cell-to-cell infection was determined by the virus, the effector and target cell types, and by the nature of the cell-cell interaction.
Collapse
Affiliation(s)
- Dmitriy Mazurov
- HIV Drug Resistance Program, National Cancer Institute and SAIC-Frederick, NCI-Frederick, Frederick, Maryland, United States of America.
| | | | | | | | | |
Collapse
|
36
|
Qualley DF, Stewart-Maynard KM, Wang F, Mitra M, Gorelick RJ, Rouzina I, Williams MC, Musier-Forsyth K. C-terminal domain modulates the nucleic acid chaperone activity of human T-cell leukemia virus type 1 nucleocapsid protein via an electrostatic mechanism. J Biol Chem 2010; 285:295-307. [PMID: 19887455 PMCID: PMC2804176 DOI: 10.1074/jbc.m109.051334] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2009] [Revised: 10/30/2009] [Indexed: 12/14/2022] Open
Abstract
Retroviral nucleocapsid (NC) proteins are molecular chaperones that facilitate nucleic acid (NA) remodeling events critical in viral replication processes such as reverse transcription. Surprisingly, the NC protein from human T-cell leukemia virus type 1 (HTLV-1) is an extremely poor NA chaperone. Using bulk and single molecule methods, we find that removal of the anionic C-terminal domain (CTD) of HTLV-1 NC results in a protein with chaperone properties comparable with that of other retroviral NCs. Increasing the ionic strength of the solution also improves the chaperone activity of full-length HTLV-1 NC. To determine how the CTD negatively modulates the chaperone activity of HTLV-1 NC, we quantified the thermodynamics and kinetics of wild-type and mutant HTLV-1 NC/NA interactions. The wild-type protein exhibits very slow dissociation kinetics, and removal of the CTD or mutations that eliminate acidic residues dramatically increase the protein/DNA interaction kinetics. Taken together, these results suggest that the anionic CTD interacts with the cationic N-terminal domain intramolecularly when HTLV-1 NC is not bound to nucleic acids, and similar interactions occur between neighboring molecules when NC is NA-bound. The intramolecular N-terminal domain-CTD attraction slows down the association of the HTLV-1 NC with NA, whereas the intermolecular interaction leads to multimerization of HTLV-1 NC on the NA. The latter inhibits both NA/NC aggregation and rapid protein dissociation from single-stranded DNA. These features make HTLV-1 NC a poor NA chaperone, despite its robust duplex destabilizing capability.
Collapse
Affiliation(s)
- Dominic F. Qualley
- From the Departments of Chemistry and Biochemistry, Center for Retrovirus Research, and Center for RNA Biology, Ohio State University, Columbus, Ohio 43210
| | | | - Fei Wang
- the Department of Physics, Northeastern University, Boston, Massachusetts 02115, and
| | - Mithun Mitra
- From the Departments of Chemistry and Biochemistry, Center for Retrovirus Research, and Center for RNA Biology, Ohio State University, Columbus, Ohio 43210
| | - Robert J. Gorelick
- the AIDS and Cancer Virus Program, Science Applications International Corporation-Frederick, Inc., NCI-Frederick, National Institutes of Health, Frederick, Maryland 21702
| | - Ioulia Rouzina
- the Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, Minnesota 55455
| | - Mark C. Williams
- the Department of Physics, Northeastern University, Boston, Massachusetts 02115, and
| | - Karin Musier-Forsyth
- From the Departments of Chemistry and Biochemistry, Center for Retrovirus Research, and Center for RNA Biology, Ohio State University, Columbus, Ohio 43210
| |
Collapse
|
37
|
Yáñez-Mó M, Barreiro O, Gordon-Alonso M, Sala-Valdés M, Sánchez-Madrid F. Tetraspanin-enriched microdomains: a functional unit in cell plasma membranes. Trends Cell Biol 2009; 19:434-46. [DOI: 10.1016/j.tcb.2009.06.004] [Citation(s) in RCA: 439] [Impact Index Per Article: 27.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2009] [Revised: 06/16/2009] [Accepted: 06/17/2009] [Indexed: 12/14/2022]
|
38
|
Krementsov DN, Weng J, Lambelé M, Roy NH, Thali M. Tetraspanins regulate cell-to-cell transmission of HIV-1. Retrovirology 2009; 6:64. [PMID: 19602278 PMCID: PMC2714829 DOI: 10.1186/1742-4690-6-64] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2009] [Accepted: 07/14/2009] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The presence of the tetraspanins CD9, CD63, CD81 and CD82 at HIV-1 budding sites, at the virological synapse (VS), and their enrichment in HIV-1 virions has been well-documented, but it remained unclear if these proteins play a role in the late phase of the viral replication cycle. Here we used overexpression and knockdown approaches to address this question. RESULTS Neither ablation of CD9, CD63 and/or CD81, nor overexpression of these tetraspanins was found to affect the efficiency of virus release. However, confirming recently reported data, tetraspanin overexpression in virus-producing cells resulted in the release of virions with substantially reduced infectivity. We also investigated the roles of these tetraspanins in cell-to-cell transmission of HIV-1. Overexpression of CD9 and CD63 led to reduced cell-to-cell transmission of this virus. Interestingly, in knockdown experiments we found that ablation of CD63, CD9 and/or CD81 had no effect on cell-free infectivity. However, knockdown of CD81, but not CD9 and CD63, enhanced productive particle transmission to target cells, suggesting additional roles for tetraspanins in the transmission process. Finally, tetraspanins were found to be downregulated in HIV-1-infected T lymphocytes, suggesting that HIV-1 modulates the levels of these proteins in order to maximize the efficiency of its transmission within the host. CONCLUSION Altogether, these results establish an active role of tetraspanins in HIV-1 producer cells.
Collapse
Affiliation(s)
- Dimitry N Krementsov
- Department of Microbiology and Molecular Genetics, University of Vermont, Burlington, VT 05405, USA.
| | | | | | | | | |
Collapse
|
39
|
Grigorov B, Attuil-Audenis V, Perugi F, Nedelec M, Watson S, Pique C, Darlix JL, Conjeaud H, Muriaux D. A role for CD81 on the late steps of HIV-1 replication in a chronically infected T cell line. Retrovirology 2009; 6:28. [PMID: 19284574 PMCID: PMC2657109 DOI: 10.1186/1742-4690-6-28] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2008] [Accepted: 03/11/2009] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND HIV-1 uses cellular co-factors for virion formation and release. The virus is able to incorporate into the viral particles host cellular proteins, such as tetraspanins which could serve to facilitate HIV-1 egress. Here, we investigated the implication of several tetraspanins on HIV-1 formation and release in chronically infected T-lymphoblastic cells, a model that permits the study of the late steps of HIV-1 replication. RESULTS Our data revealed that HIV-1 Gag and Env structural proteins co-localized with tetraspanins in the form of clusters. Co-immunoprecipitation experiments showed that Gag proteins interact, directly or indirectly, with CD81, and less with CD82, in tetraspanin-enriched microdomains composed of CD81/CD82/CD63. In addition, when HIV-1 producing cells were treated with anti-CD81 antibodies, or upon CD81 silencing by RNA interference, HIV-1 release was significantly impaired, and its infectivity was modulated. Finally, CD81 downregulation resulted in Gag redistribution at the cell surface. CONCLUSION Our findings not only extend the notion that HIV-1 assembly can occur on tetraspanin-enriched microdomains in T cells, but also highlight a critical role for the tetraspanin CD81 on the late steps of HIV replication.
Collapse
Affiliation(s)
- Boyan Grigorov
- LaboRetro, Unité de Virologie Humaine INSERM U758, Ecole Normale Supérieure de Lyon, Lyon, France.
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Hemler ME. Targeting of tetraspanin proteins--potential benefits and strategies. Nat Rev Drug Discov 2009; 7:747-58. [PMID: 18758472 DOI: 10.1038/nrd2659] [Citation(s) in RCA: 268] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The tetraspanin transmembrane proteins have emerged as key players in malignancy, the immune system, during fertilization and infectious disease processes. Tetraspanins engage in a wide range of specific molecular interactions, occurring through the formation of tetraspanin-enriched microdomains (TEMs). TEMs therefore serve as a starting point for understanding how tetraspanins affect cell signalling, adhesion, morphology, motility, fusion and virus infection. An abundance of recent evidence suggests that targeting tetraspanins, for example, by monoclonal antibodies, soluble large-loop proteins or RNAi technology, should be therapeutically beneficial.
Collapse
Affiliation(s)
- Martin E Hemler
- Dana-Farber Cancer Institute, 44 Binney Street, Boston, Massachussetts 02115, USA.
| |
Collapse
|
41
|
Abstract
Virus particle formation of HIV-1 is a multi-step process driven by a viral structural protein Gag. This process takes place at the plasma membrane in most cell types. However, the pathway that directs Gag to the plasma membrane has recently come under intense scrutiny because of its importance in production of progeny virions as well as virus transmission at cell-cell contacts. This review highlights recent advances in our current understanding of mechanisms that traffic and localize Gag to the plasma membrane. In addition, findings on Gag association with specific plasma membrane domains are discussed in light of potential roles in cell-to-cell transmission.
Collapse
|
42
|
Griffiths RE, Heesom KJ, Anstee DJ. Normal prion protein trafficking in cultured human erythroblasts. Blood 2007; 110:4518-25. [PMID: 17827389 DOI: 10.1182/blood-2007-04-085183] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Normal prion protein (PrPc), an essential substrate for development of prion disease, is widely distributed in hematopoietic cells. Recent evidence that variant Creutzfeldt-Jakob disease can be transmitted by transfusion of red cell preparations has highlighted the need for a greater understanding of the biology of PrPc in blood and blood-forming tissues. Here, we show that in contrast to another glycosylphosphoinositol-anchored protein CD59, PrPc at the cell surface of cultured human erythroblasts is rapidly internalized through the endosomal pathway, where it colocalizes with the tetraspanin CD63. In the plasma membrane, PrPc colocalizes with the tetraspanin CD81. Cross-linking with anti-PrPc or anti-CD81 causes clustering of PrPc and CD81, suggesting they can share the same microdomain. These data are consistent with a role for tetraspanin-enriched microdomains in trafficking of PrPc. These results, when taken together with recent evidence that exosomes released from cells as a result of endosomal-mediated recycling to the plasma membrane contain prion infectivity, provide a pathway for the propagation of prion diseases.
Collapse
Affiliation(s)
- Rebecca E Griffiths
- Bristol Institute for Transfusion Sciences, National Blood Service, Bristol, UK
| | | | | |
Collapse
|
43
|
Fang Y, Wu N, Gan X, Yan W, Morrell JC, Gould SJ. Higher-order oligomerization targets plasma membrane proteins and HIV gag to exosomes. PLoS Biol 2007; 5:e158. [PMID: 17550307 PMCID: PMC1885833 DOI: 10.1371/journal.pbio.0050158] [Citation(s) in RCA: 297] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2006] [Accepted: 04/10/2007] [Indexed: 11/19/2022] Open
Abstract
Exosomes are secreted organelles that have the same topology as the cell and bud outward (outward is defined as away from the cytoplasm) from endosome membranes or endosome-like domains of plasma membrane. Here we describe an exosomal protein-sorting pathway in Jurkat T cells that selects cargo proteins on the basis of both higher-order oligomerization (the oligomerization of oligomers) and plasma membrane association, acts on proteins seemingly without regard to their function, sequence, topology, or mechanism of membrane association, and appears to operate independently of class E vacuolar protein-sorting (VPS) function. We also show that higher-order oligomerization is sufficient to target plasma membrane proteins to HIV virus-like particles, that diverse Gag proteins possess exosomal-sorting information, and that higher-order oligomerization is a primary determinant of HIV Gag budding/exosomal sorting. In addition, we provide evidence that both the HIV late domain and class E VPS function promote HIV budding by unexpectedly complex, seemingly indirect mechanisms. These results support the hypothesis that HIV and other retroviruses are generated by a normal, nonviral pathway of exosome biogenesis.
Collapse
Affiliation(s)
- Yi Fang
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Ning Wu
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Xin Gan
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Wanhua Yan
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - James C Morrell
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Stephen J Gould
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- * To whom correspondence should be addressed. E-mail:
| |
Collapse
|
44
|
Khurana S, Krementsov DN, de Parseval A, Elder JH, Foti M, Thali M. Human immunodeficiency virus type 1 and influenza virus exit via different membrane microdomains. J Virol 2007; 81:12630-40. [PMID: 17855546 PMCID: PMC2168970 DOI: 10.1128/jvi.01255-07] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Directed release of human immunodeficiency virus type 1 (HIV-1) into the cleft of the virological synapse that can form between infected and uninfected T cells, for example, in lymph nodes, is thought to contribute to the systemic spread of this virus. In contrast, influenza virus, which causes local infections, is shed into the airways of the respiratory tract from free surfaces of epithelial cells. We now demonstrate that such differential release of HIV-1 and influenza virus is paralleled, at the subcellular level, by viral assembly at different microsegments of the plasma membrane of HeLa cells. HIV-1, but not influenza virus, buds through microdomains containing the tetraspanins CD9 and CD63. Consequently, the anti-CD9 antibody K41, which redistributes its antigen and also other tetraspanins to cell-cell adhesion sites, interferes with HIV-1 but not with influenza virus release. Altogether, these data strongly suggest that the bimodal egress of these two pathogenic viruses, like their entry into target cells, is guided by specific sets of host cell proteins.
Collapse
Affiliation(s)
- Sandhya Khurana
- Department of Microbiology and Molecular Genetics, University of Vermont, Burlington, VT 05405, USA
| | | | | | | | | | | |
Collapse
|