1
|
Crater JM, Dunn D, Nixon DF, O’Brien RLF. HIV-1 Mediated Cortical Actin Disruption Mirrors ARP2/3 Defects Found in Primary T Cell Immunodeficiencies. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.07.27.550856. [PMID: 38405733 PMCID: PMC10888893 DOI: 10.1101/2023.07.27.550856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
During cell movement, cortical actin balances mechanical and osmotic forces to maintain cell function while providing the scaffold for cell shape. Migrating CD4+ T cells have a polarized structure with a leading edge containing dynamic branched and linear F-actin structures that bridge intracellular components to surface adhesion molecules. These actin structures are complemented with a microtubular network beaded with membrane bound organelles in the trailing uropod. Disruption of actin structures leads to dysregulated migration and changes in morphology of affected cells. In HIV-1 infection, CD4+ T cells have dysregulated movement. However, the precise mechanisms by which HIV-1 affects CD4+ T cell movement are unknown. Here, we show that HIV-1 infection of primary CD4+ T cells causes at least four progressive morphological differences as a result of virally induced cortical cytoskeleton disruption, shown by ultrastructural and time lapse imaging. Infection with a ΔNef virus partially abrogated the dysfunctional phenotype in infected cells and partially restored a wild-type shape. The pathological morphologies after HIV-1 infection phenocopy leukocytes which contain genetic determinants of specific T cell Inborn Errors of Immunity (IEI) or Primary Immunodeficiencies (PID) that affect the actin cytoskeleton. To identify potential actin regulatory pathways that may be linked to the morphological deformities, uninfected CD4+ T cell morphology was characterized following addition of small molecule chemical inhibitors. The ARP2/3 inhibitor CK-666 recapitulated three of the four abnormal morphologies we observed in HIV-1 infected cells. Restoring ARP2/3 function and cortical actin integrity in people living with HIV-1 infection is a new avenue of investigation to eradicate HIV-1 infected cells from the body.
Collapse
Affiliation(s)
- Jacqueline M. Crater
- Department of Medicine, Division of Infectious Diseases, Weill Cornell Medicine, New York, NY, USA
| | - Daniel Dunn
- Department of Medicine, Division of Infectious Diseases, Weill Cornell Medicine, New York, NY, USA
| | - Douglas F. Nixon
- Department of Medicine, Division of Infectious Diseases, Weill Cornell Medicine, New York, NY, USA
| | - Robert L. Furler O’Brien
- Department of Medicine, Division of Infectious Diseases, Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
2
|
Ghaly M, Proulx J, Borgmann K, Park IW. Novel role of HIV-1 Nef in regulating the ubiquitination of cellular proteins. Front Cell Infect Microbiol 2023; 13:1106591. [PMID: 36968110 PMCID: PMC10031067 DOI: 10.3389/fcimb.2023.1106591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 01/16/2023] [Indexed: 03/10/2023] Open
Abstract
Our recent data established that HIV-1 Nef is pivotal in determining the fate of cellular proteins by modulating ubiquitination. However, it is unknown which proteins are ubiquitinated in the presence of Nef, a question critical for understanding the proliferation/restriction strategies of HIV-1 in infected cells. To identify cellular proteins ubiquitinated by Nef, we conducted a proteomic analysis of cellular proteins in the presence and absence of Nef. Proteomic analysis in HEK293T cells indicated that 93 proteins were upregulated and 232 were downregulated in their ubiquitination status by Nef. Computational analysis classified these proteins based on molecular function, biological process, subcellular localization, and biological pathway. Of those proteins, we found a majority of molecular functions to be involved in binding and catalytic activity. With respect to biological processes, a significant portion of the proteins identified were related to cellular and metabolic processes. Subcellular localization analysis showed the bulk of proteins to be localized to the cytosol and cytosolic compartments, which is consistent with the known function and location of Nef during HIV-1 infection. As for biological pathways, the wide range of affected proteins was denoted by the multiple modes to fulfill function, as distinguished from a strictly singular means, which was not detected. Among these ubiquitinated proteins, six were found to directly interact with Nef, wherein two were upregulated and four downregulated. We also identified 14 proteins involved in protein stability through directly participating in the Ubiquitin Proteasome System (UPS)-mediated proteasomal degradation pathway. Of those proteins, we found six upregulated and eight downregulated. Taken together, these analyses indicate that HIV-1 Nef is integral to regulating the stability of various cellular proteins via modulating ubiquitination. The molecular mechanisms directing Nef-triggered regulation of cellular protein ubiquitination are currently under investigation.
Collapse
|
3
|
Tanabe R, Morikawa Y. Efficient Transendothelial Migration of Latently HIV-1-Infected Cells. Viruses 2021; 13:v13081589. [PMID: 34452453 PMCID: PMC8402846 DOI: 10.3390/v13081589] [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: 07/07/2021] [Revised: 08/07/2021] [Accepted: 08/09/2021] [Indexed: 11/16/2022] Open
Abstract
A small fraction of HIV-1-infected T cells forms populations of latently infected cells when they are a naive T-cell subset or in transit to a resting memory state. Latently HIV-1-infected cells reside in lymphoid tissues and serve as viral reservoirs. However, whether they systemically recirculate in the body and re-enter the lymphoid nodes are unknown. Here, we employed two in-vitro cell coculture systems mimicking the lymphatic endothelium in lymph nodes and investigated the homing potential, specifically the transendothelial migration (TEM), of two latently HIV-1-infected cell lines (J1.1 and ACH-2). In trans-well coculture systems, J1.1 and ACH-2 showed higher TEM efficiencies than their parental uninfected and acutely infected cells. The efficiency of TEM was enhanced by the presence of stromal cells, such as HS-5 and fibroblastic reticular cells. In an in-vitro reconstituted, three-dimensional coculture system in which stromal cells are embedded in collagen matrices, J1.1 showed slightly higher TEM efficiency in the presence of HS-5. In accordance with these phenotypes, latently infected cells adhered to the endothelial cells more efficiently than uninfected cells. Together, our study showed that latently HIV-1-infected cells enhanced cell adhesion and TEM abilities, suggesting their potential for efficient homing to lymph nodes.
Collapse
|
4
|
Blanco A, Mahajan T, Coronado RA, Ma K, Demma DR, Dar RD. Synergistic Chromatin-Modifying Treatments Reactivate Latent HIV and Decrease Migration of Multiple Host-Cell Types. Viruses 2021; 13:v13061097. [PMID: 34201394 PMCID: PMC8228244 DOI: 10.3390/v13061097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 05/29/2021] [Accepted: 06/02/2021] [Indexed: 11/29/2022] Open
Abstract
Upon infection of its host cell, human immunodeficiency virus (HIV) establishes a quiescent and non-productive state capable of spontaneous reactivation. Diverse cell types harboring the provirus form a latent reservoir, constituting a major obstacle to curing HIV. Here, we investigate the effects of latency reversal agents (LRAs) in an HIV-infected THP-1 monocyte cell line in vitro. We demonstrate that leading drug treatments synergize activation of the HIV long terminal repeat (LTR) promoter. We establish a latency model in THP-1 monocytes using a replication incompetent HIV reporter vector with functional Tat, and show that chromatin modifiers synergize with a potent transcriptional activator to enhance HIV reactivation, similar to T-cells. Furthermore, leading reactivation cocktails are shown to differentially affect latency reactivation and surface expression of chemokine receptor type 4 (CXCR4), leading to altered host cell migration. This study investigates the effect of chromatin-modifying LRA treatments on HIV latent reactivation and cell migration in monocytes. As previously reported in T-cells, epigenetic mechanisms in monocytes contribute to controlling the relationship between latent reactivation and cell migration. Ultimately, advanced “Shock and Kill” therapy needs to successfully target and account for all host cell types represented in a complex and composite latency milieu.
Collapse
Affiliation(s)
- Alexandra Blanco
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; (A.B.); (T.M.); (R.A.C.); (K.M.); (D.R.D.)
| | - Tarun Mahajan
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; (A.B.); (T.M.); (R.A.C.); (K.M.); (D.R.D.)
| | - Robert A. Coronado
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; (A.B.); (T.M.); (R.A.C.); (K.M.); (D.R.D.)
| | - Kelly Ma
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; (A.B.); (T.M.); (R.A.C.); (K.M.); (D.R.D.)
| | - Dominic R. Demma
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; (A.B.); (T.M.); (R.A.C.); (K.M.); (D.R.D.)
| | - Roy D. Dar
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; (A.B.); (T.M.); (R.A.C.); (K.M.); (D.R.D.)
- Center for Biophysics and Quantitative Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Correspondence: ; Tel.: +1-(217)-265-0708
| |
Collapse
|
5
|
Multifunctional Roles of the N-Terminal Region of HIV-1 SF2Nef Are Mediated by Three Independent Protein Interaction Sites. J Virol 2019; 94:JVI.01398-19. [PMID: 31597760 DOI: 10.1128/jvi.01398-19] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 10/01/2019] [Indexed: 01/23/2023] Open
Abstract
HIV-1 Nef promotes virus spread and disease progression by altering host cell transport and signaling processes through interaction with multiple host cell proteins. The N-terminal region in HIV-1 Nef encompassing residues 12 to 39 has been implicated in many Nef activities, including disruption of CD4 T lymphocyte polarization and homing to lymph nodes, antagonism of SERINC5 restriction to virion infectivity, downregulation of cell surface CD4 and major histocompatibility complex class I (MHC-I), release of Nef-containing extracellular vesicles, and phosphorylation of Nef by recruitment of the Nef-associated kinase complex (NAKC). How this region mediates these pleiotropic functions is unclear. Characterization of a panel of alanine mutants spanning the N-terminal region to identify specific functional determinants revealed this region to be dispensable for effects of Nef from HIV-1 strain SF2 (HIV-1SF2Nef) on T cell actin organization and chemotaxis, retargeting of the host cell kinase Lck to the trans-Golgi network, and incorporation of Nef into extracellular vesicles. MHC-I downmodulation was specific to residue M20, and inhibition of T cell polarization by Nef required the integrity of the entire region. In contrast, downmodulation of cell surface CD4 and SERINC5 antagonism were mediated by a specific motif encompassing residues 32 to 39 that was also essential for efficient HIV replication in primary CD4 T lymphocytes. Finally, Nef phosphorylation via association with the NAKC was mediated by two EP repeats within residues 24 to 29 but was dispensable for other functions. These results identify the N-terminal region as a multifunctional interaction module for at least three different host cell ligands that mediate independent functions of HIV-1SF2Nef to facilitate immune evasion and virus spread.IMPORTANCE HIV-1 Nef critically determines virus spread and disease progression in infected individuals by acting as a protein interaction adaptor via incompletely defined mechanisms and ligands. Residues 12 to 39 near the N terminus of Nef have been described as an interaction platform for the Nef-associated kinase complex (NAKC) and were recently identified as essential determinants for a broad range of Nef activities. Here, we report a systematic mapping of this amino acid stretch that revealed the presence of three independent interaction motifs with specific ligands and activities. While downmodulation of cell surface MHC-I depends on M20, two EP repeats are the minimal binding site for the NAKC, and residues 32 to 39 mediate antagonism of the host cell restriction factor SERINC5 as well as downmodulation of cell surface CD4. These results reveal that the N-terminal region of HIV-1SF2Nef is a versatile and multifunctional protein interaction module that exerts essential functions of the pathogenicity factor via independent mechanisms.
Collapse
|
6
|
Rivera J, Isidro RA, Loucil-Alicea RY, Cruz ML, Appleyard CB, Isidro AA, Chompre G, Colon-Rivera K, Noel RJ. Infusion of HIV-1 Nef-expressing astrocytes into the rat hippocampus induces enteropathy and interstitial pneumonitis and increases blood-brain-barrier permeability. PLoS One 2019; 14:e0225760. [PMID: 31774879 PMCID: PMC6881014 DOI: 10.1371/journal.pone.0225760] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 11/12/2019] [Indexed: 12/16/2022] Open
Abstract
Even though HIV-1 replication can be suppressed by combination antiretroviral therapy (cART) inflammatory processes still occur, contributing to comorbidities. Comorbidities are attributed to variety of factors, including HIV-1 mediated inflammation. Several HIV-1 proteins mediate central nervous system (CNS) inflammation, including Nef. Nef is an early HIV-1 protein, toxic to neurons and glia and is sufficient to cause learning impairment similar to some deficits observed in HIV-1 associated neurocognitive disorders. To determine whether hippocampal Nef expression by astrocytes contributes to comorbidities, specifically peripheral inflammation, we infused Sprague Dawley rats with GFP- (control) or Nef-transfected astrocytes into the right hippocampus. Brain, lung, and ileum were collected postmortem for the measurement of inflammatory markers. Increased blood-brain-barrier permeability and serum IL-1β levels were detected in the Nef-treated rats. The lungs of Nef-treated rats demonstrated leukocyte infiltration, macrophage upregulation, and enhanced vascular permeability. Ileal tissue showed reactive follicular lymphoid hyperplasia, increased permeability and macrophage infiltration. The intracerebroventricular application of IL-1 receptor antagonist reduced infiltration of immune cells into ileum and lung, indicating the important role of IL-1β in mediating the spread of inflammation from the brain to other tissues. This suggests that localized expression of a single viral protein, HIV-1 Nef, can contribute to a broader inflammatory response by upregulation of IL-1β. Further, these results suggest that Nef contributes to the chronic inflammation seen in HIV patients, even in those whose viremia is controlled by cART.
Collapse
Affiliation(s)
- Jocelyn Rivera
- HIV-1 Immunopathogenesis Laboratory, The Wistar Institute, Philadelphia, PA, United States of America
| | - Raymond A. Isidro
- Department of Basic Sciences, Ponce Health Sciences University, Ponce Research Institute, Ponce, Puerto Rico, United States of America
| | - Raisa Y. Loucil-Alicea
- Department of Basic Sciences, Ponce Health Sciences University, Ponce Research Institute, Ponce, Puerto Rico, United States of America
| | - Myrella L. Cruz
- Department of Basic Sciences, Ponce Health Sciences University, Ponce Research Institute, Ponce, Puerto Rico, United States of America
| | - Caroline B. Appleyard
- Department of Basic Sciences, Ponce Health Sciences University, Ponce Research Institute, Ponce, Puerto Rico, United States of America
| | - Angel A. Isidro
- Department of Basic Sciences, Ponce Health Sciences University, Ponce Research Institute, Ponce, Puerto Rico, United States of America
| | - Gladys Chompre
- Department of Biology, Pontifical Catholic University of Puerto Rico, Ponce, Puerto Rico, United States of America
| | - Krystal Colon-Rivera
- HIV-1 Immunopathogenesis Laboratory, The Wistar Institute, Philadelphia, PA, United States of America
| | - Richard J. Noel
- Department of Basic Sciences, Ponce Health Sciences University, Ponce Research Institute, Ponce, Puerto Rico, United States of America
- * E-mail:
| |
Collapse
|
7
|
Lamas-Murua M, Stolp B, Kaw S, Thoma J, Tsopoulidis N, Trautz B, Ambiel I, Reif T, Arora S, Imle A, Tibroni N, Wu J, Cui G, Stein JV, Tanaka M, Lyck R, Fackler OT. HIV-1 Nef Disrupts CD4 + T Lymphocyte Polarity, Extravasation, and Homing to Lymph Nodes via Its Nef-Associated Kinase Complex Interface. THE JOURNAL OF IMMUNOLOGY 2018; 201:2731-2743. [PMID: 30257886 DOI: 10.4049/jimmunol.1701420] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Accepted: 08/21/2018] [Indexed: 12/31/2022]
Abstract
HIV-1 Nef is a multifunctional protein that optimizes virus spread and promotes immune evasion of infected cells to accelerate disease progression in AIDS patients. As one of its activities, Nef reduces the motility of infected CD4+ T lymphocytes in confined space. In vivo, Nef restricts T lymphocyte homing to lymph nodes as it reduces the ability for extravasation at the diapedesis step. Effects of Nef on T lymphocyte motility are typically mediated by its ability to reduce actin remodeling. However, interference with diapedesis does not depend on residues in Nef required for inhibition of host cell actin dynamics. In search for an alternative mechanism by which Nef could alter T lymphocyte extravasation, we noted that the viral protein interferes with the polarization of primary human CD4+ T lymphocytes upon infection with HIV-1. Expression of Nef alone is sufficient to disrupt T cell polarization, and this effect is conserved among lentiviral Nef proteins. Nef acts by arresting the oscillation of CD4+ T cells between polarized and nonpolarized morphologies. Mapping studies identified the binding site for the Nef-associated kinase complex (NAKC) as critical determinant of this Nef activity and a NAKC-binding-deficient Nef variant fails to impair CD4+ T lymphocyte extravasation and homing to lymph nodes. These results thus imply the disruption of T lymphocyte polarity via its NAKC binding site as a novel mechanism by which lentiviral Nef proteins alter T lymphocyte migration in vivo.
Collapse
Affiliation(s)
- Miguel Lamas-Murua
- Department of Infectious Diseases, Integrative Virology, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Bettina Stolp
- Department of Infectious Diseases, Integrative Virology, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Sheetal Kaw
- Department of Infectious Diseases, Integrative Virology, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Judith Thoma
- Physical Chemistry of Biosystems, University of Heidelberg, 69120 Heidelberg, Germany
| | - Nikolaos Tsopoulidis
- Department of Infectious Diseases, Integrative Virology, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Birthe Trautz
- Department of Infectious Diseases, Integrative Virology, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Ina Ambiel
- Department of Infectious Diseases, Integrative Virology, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Tatjana Reif
- Department of Infectious Diseases, Integrative Virology, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Sakshi Arora
- Department of Infectious Diseases, Integrative Virology, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Andrea Imle
- Department of Infectious Diseases, Integrative Virology, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Nadine Tibroni
- Department of Infectious Diseases, Integrative Virology, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Jingxia Wu
- T Cell Metabolism (D140), German Cancer Research Centre, 69120 Heidelberg, Germany
| | - Guoliang Cui
- T Cell Metabolism (D140), German Cancer Research Centre, 69120 Heidelberg, Germany
| | - Jens V Stein
- Theodor Kocher Institute, University of Bern, 3012 Bern, Switzerland; and
| | - Motomu Tanaka
- Physical Chemistry of Biosystems, University of Heidelberg, 69120 Heidelberg, Germany.,Center for Integrative Medicine and Physics, Institute for Advanced Study, Kyoto University, Kyoto 606-8501, Japan
| | - Ruth Lyck
- Theodor Kocher Institute, University of Bern, 3012 Bern, Switzerland; and
| | - Oliver T Fackler
- Department of Infectious Diseases, Integrative Virology, University Hospital Heidelberg, 69120 Heidelberg, Germany;
| |
Collapse
|
8
|
Visualizing Viral Infection In Vivo by Multi-Photon Intravital Microscopy. Viruses 2018; 10:v10060337. [PMID: 29925766 PMCID: PMC6024644 DOI: 10.3390/v10060337] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 06/12/2018] [Accepted: 06/19/2018] [Indexed: 12/11/2022] Open
Abstract
Viral pathogens have adapted to the host organism to exploit the cellular machinery for virus replication and to modulate the host cells for efficient systemic dissemination and immune evasion. Much of our knowledge of the effects that virus infections have on cells originates from in vitro imaging studies using experimental culture systems consisting of cell lines and primary cells. Recently, intravital microscopy using multi-photon excitation of fluorophores has been applied to observe virus dissemination and pathogenesis in real-time under physiological conditions in living organisms. Critical steps during viral infection and pathogenesis could be studied by direct visualization of fluorescent virus particles, virus-infected cells, and the immune response to viral infection. In this review, I summarize the latest research on in vivo studies of viral infections using multi-photon intravital microscopy (MP-IVM). Initially, the underlying principle of multi-photon microscopy is introduced and experimental challenges during microsurgical animal preparation and fluorescent labeling strategies for intravital imaging are discussed. I will further highlight recent studies that combine MP-IVM with optogenetic tools and transcriptional analysis as a powerful approach to extend the significance of in vivo imaging studies of viral pathogens.
Collapse
|
9
|
Ospina Stella A, Turville S. All-Round Manipulation of the Actin Cytoskeleton by HIV. Viruses 2018; 10:v10020063. [PMID: 29401736 PMCID: PMC5850370 DOI: 10.3390/v10020063] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Revised: 01/24/2018] [Accepted: 01/29/2018] [Indexed: 12/21/2022] Open
Abstract
While significant progress has been made in terms of human immunodeficiency virus (HIV) therapy, treatment does not represent a cure and remains inaccessible to many people living with HIV. Continued mechanistic research into the viral life cycle and its intersection with many aspects of cellular biology are not only fundamental in the continued fight against HIV, but also provide many key observations of the workings of our immune system. Decades of HIV research have testified to the integral role of the actin cytoskeleton in both establishing and spreading the infection. Here, we review how the virus uses different strategies to manipulate cellular actin networks and increase the efficiency of various stages of its life cycle. While some HIV proteins seem able to bind to actin filaments directly, subversion of the cytoskeleton occurs indirectly by exploiting the power of actin regulatory proteins, which are corrupted at multiple levels. Furthermore, this manipulation is not restricted to a discrete class of proteins, but rather extends throughout all layers of the cytoskeleton. We discuss prominent examples of actin regulators that are exploited, neutralized or hijacked by the virus, and address how their coordinated deregulation can lead to changes in cellular behavior that promote viral spreading.
Collapse
Affiliation(s)
- Alberto Ospina Stella
- The Kirby Institute, University of New South Wales (UNSW), Sydney NSW 2052, Australia.
| | - Stuart Turville
- The Kirby Institute, University of New South Wales (UNSW), Sydney NSW 2052, Australia.
| |
Collapse
|
10
|
HIV-1 Nef CAWLEAQ motif: a regulator of monocytes invasion through ENO1 modulation. Mol Cell Biochem 2018; 447:151-164. [DOI: 10.1007/s11010-018-3300-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Accepted: 01/24/2018] [Indexed: 10/18/2022]
|
11
|
Trautz B, Wiedemann H, Lüchtenborg C, Pierini V, Kranich J, Glass B, Kräusslich HG, Brocker T, Pizzato M, Ruggieri A, Brügger B, Fackler OT. The host-cell restriction factor SERINC5 restricts HIV-1 infectivity without altering the lipid composition and organization of viral particles. J Biol Chem 2017; 292:13702-13713. [PMID: 28659343 DOI: 10.1074/jbc.m117.797332] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Revised: 06/21/2017] [Indexed: 12/14/2022] Open
Abstract
The host-cell restriction factor SERINC5 potently suppresses the infectivity of HIV, type 1 (HIV-1) particles, and is counteracted by the viral pathogenesis factor Nef. However, the molecular mechanism by which SERINC5 restricts HIV-1 particle infectivity is still unclear. Because SERINC proteins have been suggested to facilitate the incorporation of serine during the biosynthesis of membrane lipids and because lipid composition of HIV particles is a major determinant of the infectious potential of the particles, we tested whether SERINC5-mediated restriction of HIV particle infectivity involves alterations of membrane lipid composition. We produced and purified HIV-1 particles from SERINC5293T cells with very low endogenous SERINC5 levels under conditions in which ectopically expressed SERINC5 restricts HIV-1 infectivity and is antagonized by Nef and analyzed both virions and producer cells with quantitative lipid MS. SERINC5 restriction and Nef antagonism were not associated with significant alterations in steady-state lipid composition of producer cells and HIV particles. Sphingosine metabolism kinetics were also unaltered by SERINC5 expression. Moreover, the levels of phosphatidylserine on the surface of HIV-1 particles, which may trigger uptake into non-productive internalization pathways in target cells, did not change upon expression of SERINC5 or Nef. Finally, saturating the phosphatidylserine-binding sites on HIV target cells did not affect SERINC5 restriction or Nef antagonism. These results demonstrate that the restriction of HIV-1 particle infectivity by SERINC5 does not depend on alterations in lipid composition and organization of HIV-1 particles and suggest that channeling serine into lipid biosynthesis may not be a cardinal cellular function of SERINC5.
Collapse
Affiliation(s)
- Birthe Trautz
- From the Department of Infectious Diseases, Integrative Virology, and
| | - Hannah Wiedemann
- the Heidelberg University Biochemistry Center, INF 328, 69120 Heidelberg, Germany
| | | | - Virginia Pierini
- From the Department of Infectious Diseases, Integrative Virology, and
| | - Jan Kranich
- the Institute for Immunology, Ludwig-Maximilians-Universität München, Groβhardener Straße 9, 82152 Planegg-Martinsried, Germany
| | - Bärbel Glass
- Department of Infectious Diseases, Virology, University Hospital Heidelberg, INF 324, 69120 Heidelberg, Germany
| | - Hans-Georg Kräusslich
- Department of Infectious Diseases, Virology, University Hospital Heidelberg, INF 324, 69120 Heidelberg, Germany
| | - Thomas Brocker
- the Institute for Immunology, Ludwig-Maximilians-Universität München, Groβhardener Straße 9, 82152 Planegg-Martinsried, Germany
| | - Massimo Pizzato
- the University of Trento, Centre for Integrative Biology, 38122 Trento, Italy, and
| | - Alessia Ruggieri
- the Department of Infectious Diseases, Molecular Virology, University Hospital Heidelberg, INF 345, 69120 Heidelberg, Germany
| | - Britta Brügger
- the Heidelberg University Biochemistry Center, INF 328, 69120 Heidelberg, Germany,
| | - Oliver T Fackler
- From the Department of Infectious Diseases, Integrative Virology, and
| |
Collapse
|
12
|
The Antagonism of HIV-1 Nef to SERINC5 Particle Infectivity Restriction Involves the Counteraction of Virion-Associated Pools of the Restriction Factor. J Virol 2016; 90:10915-10927. [PMID: 27681140 DOI: 10.1128/jvi.01246-16] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 09/21/2016] [Indexed: 11/20/2022] Open
Abstract
SERINC3 (serine incorporator 3) and SERINC5 are recently identified host cell inhibitors of HIV-1 particle infectivity that are counteracted by the viral pathogenesis factor Nef. Here we confirm that HIV-1 Nef, but not HIV-1 Vpu, antagonizes the particle infectivity restriction of SERINC5. SERINC5 antagonism occurred in parallel with other Nef activities, including cell surface receptor downregulation, trans-Golgi network targeting of Lck, and inhibition of host cell actin dynamics. Interaction motifs with host cell endocytic machinery and the Nef-associated kinase complex, as well as CD4 cytoplasmic tail/HIV-1 protease, were identified as essential Nef determinants for SERINC5 antagonism. Characterization of antagonism-deficient Nef mutants revealed that counteraction of SERINC5 occurs in the absence of retargeting of the restriction factor to intracellular compartments and reduction of SERINC5 cell surface density is insufficient for antagonism. Consistent with virion incorporation of SERINC5 being a prerequisite for its antiviral activity, the infectivity of HIV-1 particles produced in the absence of a SERINC5 antagonist decreased with increasing amounts of virion SERINC5. At low levels of SERINC5 expression, enhancement of virion infectivity by Nef was associated with reduced virion incorporation of SERINC5 and antagonism-defective Nef mutants failed to exclude SERINC5 from virions. However, at elevated levels of SERINC5 expression, Nef maintained infectious HIV particles, despite significant virion incorporation of the restriction factor. These results suggest that in addition to virion exclusion, Nef employs a cryptic mechanism to antagonize virion-associated SERINC5. The involvement of common determinants suggests that the antagonism of Nef to SERINC5 and the downregulation of cell surface CD4 by Nef involve related molecular mechanisms. IMPORTANCE HIV-1 Nef critically determines virus spread and disease progression in infected individuals by incompletely defined mechanisms. SERINC3 and SERINC5 were recently identified as potent inhibitors of HIV particle infectivity whose antiviral activity is antagonized by HIV-1 Nef. To address the mechanism of SERINC5 antagonism, we identified four molecular determinants of Nef antagonism that are all linked to the mechanism by which Nef downregulates cell surface CD4. Functional characterization of these mutants revealed that endosomal targeting and cell surface downregulation of SERINC5 are dispensable and insufficient for antagonism, respectively. In contrast, virion exclusion and antagonism of SERINC5 were correlated; however, Nef was also able to enhance the infectivity of virions that incorporated robust levels of SERINC5. These results suggest that the antagonism of HIV-1 Nef to SERINC5 restriction of virion infectivity is mediated by a dual mechanism that is related to CD4 downregulation.
Collapse
|
13
|
Silva JG, Martins NP, Henriques R, Soares H. HIV-1 Nef Impairs the Formation of Calcium Membrane Territories Controlling the Signaling Nanoarchitecture at the Immunological Synapse. THE JOURNAL OF IMMUNOLOGY 2016; 197:4042-4052. [PMID: 27798165 DOI: 10.4049/jimmunol.1601132] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Accepted: 09/16/2016] [Indexed: 12/21/2022]
Abstract
The ability of HIV-1 to replicate and to establish long-term reservoirs is strongly influenced by T cell activation. Through the use of membrane-tethered, genetically encoded calcium (Ca2+) indicators, we were able to detect for the first time, to our knowledge, the formation of Ca2+ territories and determine their role in coordinating the functional signaling nanostructure of the synaptic membrane. Consequently, we report a previously unknown immune subversion mechanism involving HIV-1 exploitation, through its Nef accessory protein, of the interconnectivity among three evolutionarily conserved cellular processes: vesicle traffic, signaling compartmentalization, and the second messenger Ca2+ We found that HIV-1 Nef specifically associates with the traffic regulators MAL and Rab11b compelling the vesicular accumulation of Lck. Through its association with MAL and Rab11b, Nef co-opts Lck switchlike function driving the formation Ca2+ membrane territories, which, in turn, control the fusion of LAT-transporting Rab27 and Rab37 vesicles and the formation of LAT nanoclusters at the immunological synapse. Consequently, HIV-1 Nef disengages TCR triggering from the generation of p-LAT and p-SLP nanoclusters driving TCR signal amplification and diversification. Altogether our results indicate that HIV-1 exploits the interconnectivity among vesicle traffic, Ca2+ membrane territories, and signaling nanoclusters to modulate T cell signaling and function.
Collapse
Affiliation(s)
- Joana G Silva
- Immunobiology and Pathogenesis Group, Chronic Diseases Research Center, NOVA Medical School, NOVA University of Lisbon, 1150-082 Lisbon, Portugal
| | - Nuno P Martins
- Unit of Imaging and Cytometry, Gulbenkian Institute for Science, 2780-156 Oeiras, Portugal
| | - Ricardo Henriques
- Quantitative Imaging and Nanobiophysics Group, Medical Research Council Laboratory for Molecular Cell Biology, University College London, London, United Kingdom; and.,Department of Cell and Developmental Biology, University College London, London WC1E 6BT, United Kingdom
| | - Helena Soares
- Immunobiology and Pathogenesis Group, Chronic Diseases Research Center, NOVA Medical School, NOVA University of Lisbon, 1150-082 Lisbon, Portugal;
| |
Collapse
|
14
|
D186/D190 is an allele-dependent determinant of HIV-1 Nef function. Virology 2016; 498:44-56. [PMID: 27560372 DOI: 10.1016/j.virol.2016.08.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Revised: 08/12/2016] [Accepted: 08/13/2016] [Indexed: 01/23/2023]
Abstract
The HIV-1 pathogenesis factor Nef interacts with numerous ligands to affect cellular vesicular transport, signal transduction and cytoskeletal dynamics. While most Nef functions depend on multivalent protein interaction motifs, disrupting actin dynamics requires a motif that specifically recruits the host kinase PAK2. An adjacent aspartate was recently predicted to mediate Nef-β-catenin interactions. We report here that β-catenin can be co-immunoprecipitated with Nef.GFP from Jurkat T cell lysates. This association is conserved among lentiviral Nef proteins but does not involve classical Nef protein interaction motifs, including the critical aspartate. While aspartate-to-alanine mutations impaired cell surface receptor downregulation and interference with actin dynamics and cell motility by HIV-1 NA7 Nef, analogous mutations did not affect HIV-1 SF2 Nef function. These allelic differences were determined by a proximal lysine/arginine polymorphism. These results emphasize differences between Nef alleles regarding the functional role of individual residues and underscore the need for allele-specific structure-function analyses.
Collapse
|
15
|
Vérollet C, Le Cabec V, Maridonneau-Parini I. HIV-1 Infection of T Lymphocytes and Macrophages Affects Their Migration via Nef. Front Immunol 2015; 6:514. [PMID: 26500651 PMCID: PMC4594015 DOI: 10.3389/fimmu.2015.00514] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Accepted: 09/22/2015] [Indexed: 12/17/2022] Open
Abstract
The human immunodeficiency virus (HIV-1) disseminates in the body and is found in several organs and tissues. Although HIV-1 mainly targets both CD4+ T lymphocytes and macrophages, it has contrasting effects between these cell populations. HIV-1 infection namely reduces the viability of CD4+ T cells, whereas infected macrophages are long-lived. In addition, the migration of T cells is reduced by the infection, whereas HIV-1 differentially modulates the migration modes of macrophages. In 2-dimensions (2D) assays, infected macrophages are less motile compared to the control counterparts. In 3D environments, macrophages use two migration modes that are dependent on the matrix architecture: amoeboid and mesenchymal migration. HIV-1-infected macrophages exhibit a reduced amoeboid migration but an enhanced mesenchymal migration, via the viral protein Nef. Indeed, the mesenchymal migration involves podosomes, and Nef stabilizes these cell structures through the activation of the tyrosine kinase Hck, which in turn phosphorylates the Wiskott–Aldrich syndrome protein (WASP). WASP is a key player in actin remodeling and cell migration. The reprogramed motility of infected macrophages observed in vitro correlates in vivo with enhanced macrophage infiltration in experimental tumors in Nef-transgenic mice compared to control mice. In conclusion, HIV infection of host target cells modifies their migration capacity; we infer that HIV-1 enhances virus spreading in confined environments by reducing T cells migration, and facilitates virus dissemination into different organs and tissues of the human body by enhancing macrophage mesenchymal migration.
Collapse
Affiliation(s)
- Christel Vérollet
- CNRS UMR 5089, Institut de Pharmacologie et de Biologie Structurale (IPBS) , Toulouse , France ; Institut de Pharmacologie et de Biologie Structurale (IPBS), Université Toulouse III - Paul Sabatier , Toulouse , France
| | - Véronique Le Cabec
- CNRS UMR 5089, Institut de Pharmacologie et de Biologie Structurale (IPBS) , Toulouse , France ; Institut de Pharmacologie et de Biologie Structurale (IPBS), Université Toulouse III - Paul Sabatier , Toulouse , France
| | - Isabelle Maridonneau-Parini
- CNRS UMR 5089, Institut de Pharmacologie et de Biologie Structurale (IPBS) , Toulouse , France ; Institut de Pharmacologie et de Biologie Structurale (IPBS), Université Toulouse III - Paul Sabatier , Toulouse , France
| |
Collapse
|
16
|
Association with PAK2 Enables Functional Interactions of Lentiviral Nef Proteins with the Exocyst Complex. mBio 2015; 6:e01309-15. [PMID: 26350970 PMCID: PMC4600113 DOI: 10.1128/mbio.01309-15] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
UNLABELLED Human immunodeficiency virus type 1 (HIV-1) Nef enhances virus replication and contributes to immune evasion in vivo, but the underlying molecular mechanisms remain incompletely defined. Nef interferes with host cell actin dynamics to restrict T lymphocyte responses to chemokine stimulation and T cell receptor engagement. This relies on the assembly of a labile multiprotein complex including the host kinase PAK2 that Nef usurps to phosphorylate and inactivate the actin-severing factor cofilin. Components of the exocyst complex (EXOC), an octameric protein complex involved in vesicular transport and actin remodeling, were recently reported to interact with Nef via the same molecular surface that mediates PAK2 association. Exploring the functional relevance of EXOC in Nef-PAK2 complex assembly/function, we found Nef-EXOC interactions to be specifically mediated by the PAK2 interface of Nef, to occur in infected human T lymphocytes, and to be conserved among lentiviral Nef proteins. In turn, EXOC was dispensable for direct downstream effector functions of Nef-associated PAK2. Surprisingly, PAK2 was essential for Nef-EXOC association, which required a functional Rac1/Cdc42 binding site but not the catalytic activity of PAK2. EXOC was dispensable for Nef functions in vesicular transport but critical for inhibition of actin remodeling and proximal signaling upon T cell receptor engagement. Thus, Nef exploits PAK2 in a stepwise mechanism in which its kinase activity cooperates with an adaptor function for EXOC to inhibit host cell actin dynamics. IMPORTANCE Human immunodeficiency virus type 1 (HIV-1) Nef contributes to AIDS pathogenesis, but the underlying molecular mechanisms remain incompletely understood. An important aspect of Nef function is to facilitate virus replication by disrupting T lymphocyte actin dynamics in response to stimulation via its association with the host cell kinase PAK2. We report here that the molecular surface of Nef for PAK2 association also mediates interaction of Nef with EXOC and establish that PAK2 provides an essential adaptor function for the subsequent formation of Nef-EXOC complexes. PAK2 and EXOC specifically cooperate in the inhibition of actin dynamics and proximal signaling induced by T cell receptor engagement by Nef. These results establish EXOC as a functionally relevant Nef interaction partner, emphasize the suitability of the PAK2 interaction surface for future therapeutic interference with Nef function, and show that such strategies need to target activity-independent PAK2 functions.
Collapse
|
17
|
Saxena R, Gupta S, Singh K, Mitra K, Tripathi AK, Tripathi RK. Proteomic profiling of SupT1 cells reveal modulation of host proteins by HIV-1 Nef variants. PLoS One 2015; 10:e0122994. [PMID: 25874870 PMCID: PMC4395413 DOI: 10.1371/journal.pone.0122994] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Accepted: 02/26/2015] [Indexed: 01/14/2023] Open
Abstract
Nef is an accessory viral protein that promotes HIV-1 replication, facilitating alterations in cellular pathways via multiple protein-protein interactions. The advent of proteomics has expanded the focus on better identification of novel molecular pathways regulating disease progression. In this study, nef was sequenced from randomly selected patients, however, sequence variability identified did not elicited any specific mutation that could have segregated HIV-1 patients in different stages of disease progression. To explore the difference in Nef functionality based on sequence variability we used proteomics approach. Proteomic profiling was done to compare the effect of Nef variants in host cell protein expression. 2DGE in control and Nef transfected SupT1 cells demonstrated several differentially expressed proteins. Fourteen protein spots were detected with more than 1.5 fold difference. Significant down regulation was seen in six unique protein spots in the Nef treated cells. Proteins were identified as Cyclophilin A, EIF5A-1 isoform B, Rho GDI 1 isoform a, VDAC1, OTUB1 and α-enolase isoform 1 (ENO1) through LC-MS/MS. The differential expression of the 6 proteins was analyzed by Real time PCR, Western blotting and Immunofluorescence studies with two Nef variants (RP14 and RP01) in SupT1 cells. There was contrasting difference between the effect of these Nef variants upon the expression of these six proteins. Downregulation of α-enolase (ENO1), VDAC1 and OTUB1 was more significant by Nef RP01 whereas Cyclophilin A and RhoGDI were found to be more downregulated by Nef RP14. This difference in Nef variants upon host protein expression was also studied through a site directed mutant of Nef RP01 (55AAAAAAA61) and the effect was found to be reversed. Deciphering the role of these proteins mediated by Nef variants will open a new avenue of research in understanding Nef mediated pathogenesis. Overall study determines modulation of cellular protein expression in T cells by HIV-1 Nef variants.
Collapse
Affiliation(s)
- Reshu Saxena
- Toxicology division, CSIR-Central Drug Research Institute, Sector-10, Janakipuram Extension, Sitapur Road, Lucknow, India
| | - Sudipti Gupta
- Toxicology division, CSIR-Central Drug Research Institute, Sector-10, Janakipuram Extension, Sitapur Road, Lucknow, India
| | - Kavita Singh
- Electron Microscopy Lab, Sophisticated Analytical Instrument Facility, CSIR-Central Drug Research Institute, Sector-10, Janakipuram Extension, Sitapur Road, Lucknow, India
| | - Kalyan Mitra
- Electron Microscopy Lab, Sophisticated Analytical Instrument Facility, CSIR-Central Drug Research Institute, Sector-10, Janakipuram Extension, Sitapur Road, Lucknow, India
| | - Anil Kumar Tripathi
- Department of Medicine, King George’s Medical University, Chowk, Lucknow, India
| | - Raj Kamal Tripathi
- Toxicology division, CSIR-Central Drug Research Institute, Sector-10, Janakipuram Extension, Sitapur Road, Lucknow, India
- * E-mail:
| |
Collapse
|
18
|
Wyndham-Thomas C, Dirix V, Schepers K, Martin C, Hildebrand M, Goffard JC, Domont F, Libin M, Loyens M, Locht C, Van Vooren JP, Mascart F. Contribution of a heparin-binding haemagglutinin interferon-gamma release assay to the detection of Mycobacterium tuberculosis infection in HIV-infected patients: comparison with the tuberculin skin test and the QuantiFERON-TB Gold In-tube. BMC Infect Dis 2015; 15:59. [PMID: 25886172 PMCID: PMC4337251 DOI: 10.1186/s12879-015-0796-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2014] [Accepted: 02/03/2015] [Indexed: 12/25/2022] Open
Abstract
Background The screening and treatment of latent tuberculosis (TB) infection reduces the risk of progression to active disease and is currently recommended for HIV-infected patients. The aim of this study is to evaluate, in a low TB incidence setting, the potential contribution of an interferon-gamma release assay in response to the mycobacterial latency antigen Heparin-Binding Haemagglutinin (HBHA-IGRA), to the detection of Mycobacterium tuberculosis infection in HIV-infected patients. Methods Treatment-naïve HIV-infected adults were recruited from 4 Brussels-based hospitals. Subjects underwent screening for latent TB using the HBHA-IGRA in parallel to a classical method consisting of medical history, chest X-ray, tuberculin skin test (TST) and QuantiFERON®-TB Gold In-Tube (QFT-GIT). Prospective clinical and biological follow-up ensued, with repeated testing with HBHA-IGRA. A group of HIV-infected patients with clinical suspicion of active TB was also recruited and tested with the HBHA-IGRA. Multiplex analysis was performed on the culture supernatants of this in-house assay to identify test read-outs alternative to interferon-gamma that could increase the sensitivity of the test. Results Among 48 candidates enrolled for screening, 9 were identified with latent TB by TST and/or QFT-GIT results. Four of these 9 patients and an additional 3 screened positive with the HBHA-IGRA. This in-house assay identified all the patients that were positive for the TST and showed the best concordance with the presence of a M. tuberculosis exposure risk. During follow-up (median 14 months) no case of active TB was reported and HBHA-IGRA results remained globally constant. Fourteen HIV-infected patients with clinical suspicion of active TB were recruited. Active TB was confirmed for 6 of them among which 3 were HBHA-IGRA positive, each with very high interferon-gamma concentrations. All patients for whom active TB was finally excluded, including 2 non-tubercular mycobacterial infections, had negative HBHA-IGRA results. Multiplex analysis confirmed interferon-gamma as the best read-out. Conclusions The HBHA-IGRA appears complementary to the QuantiFERON®-TB Gold In-Tube for the screening of latent TB in HIV-infected patients. Large-scale studies are necessary to determine whether this combination offers sufficient sensitivity to dismiss TST, as suggested by our results. Furthermore, HBHA-IGRA may help in the diagnosis work-up of clinical suspicions of active TB.
Collapse
Affiliation(s)
- Chloé Wyndham-Thomas
- Laboratory of Vaccinology and Mucosal Immunology, Université Libre de Bruxelles, Brussels, Belgium. .,Immunodeficiency unit, Hôpital Erasme, Brussels, Belgium.
| | - Violette Dirix
- Laboratory of Vaccinology and Mucosal Immunology, Université Libre de Bruxelles, Brussels, Belgium.
| | - Kinda Schepers
- Immunodeficiency unit, Hôpital Erasme, Brussels, Belgium.
| | | | - Marc Hildebrand
- Infectious disease unit, IRIS SUD hospitals, Brussels, Belgium.
| | | | - Fanny Domont
- Laboratory of Vaccinology and Mucosal Immunology, Université Libre de Bruxelles, Brussels, Belgium.
| | - Myriam Libin
- Laboratory of Vaccinology and Mucosal Immunology, Université Libre de Bruxelles, Brussels, Belgium.
| | - Marc Loyens
- INSERM U1019, Lille, France. .,CNRS UMR8204, Lille, France. .,Université Lille Nord de France, Lille, France. .,Center for Infection and Immunity of Lille, Institut Pasteur de Lille, Lille, France.
| | - Camille Locht
- INSERM U1019, Lille, France. .,CNRS UMR8204, Lille, France. .,Université Lille Nord de France, Lille, France. .,Center for Infection and Immunity of Lille, Institut Pasteur de Lille, Lille, France.
| | | | - Françoise Mascart
- Laboratory of Vaccinology and Mucosal Immunology, Université Libre de Bruxelles, Brussels, Belgium. .,Immunobiology Clinic, Hôpital Erasme, Brussels, Belgium.
| |
Collapse
|
19
|
Adding new dimensions: towards an integrative understanding of HIV-1 spread. Nat Rev Microbiol 2014; 12:563-74. [PMID: 25029025 DOI: 10.1038/nrmicro3309] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
In vitro studies in primary or immortalized cells continue to be used to elucidate the essential principles that govern the interactions between HIV-1 and isolated target cells. However, until recently, substantial technical barriers prevented this information from being efficiently translated to the more complex scenario of HIV-1 spread in the host in vivo, which has limited our understanding of the impact of host physiological parameters on the spread of HIV-1. In this Review, we discuss the recent development of imaging approaches to visualize HIV-1 spread and the adaptation of these approaches to organotypic ex vivo models and animal models. We focus on new concepts, including the mechanisms and in vivo relevance of cell-cell transmission for HIV-1 spread and the function of the HIV-1 pathogenesis factor Nef, which have emerged from the application of these integrative approaches in complex cell systems.
Collapse
|
20
|
Park IW, Fan Y, Luo X, Ryou MG, Liu J, Green L, He JJ. HIV-1 Nef is transferred from expressing T cells to hepatocytic cells through conduits and enhances HCV replication. PLoS One 2014; 9:e99545. [PMID: 24911518 PMCID: PMC4050050 DOI: 10.1371/journal.pone.0099545] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2013] [Accepted: 05/16/2014] [Indexed: 12/15/2022] Open
Abstract
HIV-1 infection enhances HCV replication and as a consequence accelerates HCV-mediated hepatocellular carcinoma (HCC). However, the precise molecular mechanism by which this takes place is currently unknown. Our data showed that infectious HIV-1 failed to replicate in human hepatocytic cell lines. No discernible virus replication was observed, even when the cell lines transfected with HIV-1 proviral DNA were co-cultured with Jurkat T cells, indicating that the problem of liver deterioration in the co-infected patient is not due to the replication of HIV-1 in the hepatocytes of the HCV infected host. Instead, HIV-1 Nef protein was transferred from nef-expressing T cells to hepatocytic cells through conduits, wherein up to 16% (average 10%) of the cells harbored the transferred Nef, when the hepatocytic cells were co-cultured with nef-expressing Jurkat cells for 24 h. Further, Nef altered the size and numbers of lipid droplets (LD), and consistently up-regulated HCV replication by 1.5∼2.5 fold in the target subgenomic replicon cells, which is remarkable in relation to the initially indolent viral replication. Nef also dramatically augmented reactive oxygen species (ROS) production and enhanced ethanol-mediated up-regulation of HCV replication so as to accelerate HCC. Taken together, these data indicate that HIV-1 Nef is a critical element in accelerating progression of liver pathogenesis via enhancing HCV replication and coordinating modulation of key intra- and extra-cellular molecules for liver decay.
Collapse
Affiliation(s)
- In-Woo Park
- Department of Cell Biology and Immunology, University of North Texas Health Science Center, Fort Worth, Texas, United States of America
- Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
- * E-mail:
| | - Yan Fan
- Department of Cell Biology and Immunology, University of North Texas Health Science Center, Fort Worth, Texas, United States of America
- Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Xiaoyu Luo
- Department of Cell Biology and Immunology, University of North Texas Health Science Center, Fort Worth, Texas, United States of America
- Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Myoung-Gwi Ryou
- Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, Texas, United States of America
| | - Jinfeng Liu
- Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Linden Green
- Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Johnny J. He
- Department of Cell Biology and Immunology, University of North Texas Health Science Center, Fort Worth, Texas, United States of America
- Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| |
Collapse
|
21
|
Park IW, He JJ. HIV-1 Nef-mediated T-cell activation and chemotaxis are decoupled using a HIV-1/SIVpbj1.9. chimeric nef variant. Arch Virol 2012; 158:845-52. [PMID: 23224761 DOI: 10.1007/s00705-012-1560-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2012] [Accepted: 10/17/2012] [Indexed: 01/04/2023]
Abstract
HIV-1 Nef is known to activate CD4+ T cells but inhibit their migration toward SDF-1α. However, it is not clear how this protein orchestrates these two seemingly concomitant events. In this study, by comparing these two activities of HIV-1 Nef with those of its primate counterpart, SIVpbj1.9, we found that HIV-1 Nef activated T cells only in the presence of CD3/ CD28 stimulation, while SIVpbj1.9 Nef did even without CD3/CD28. We also observed that HIV-1 Nef inhibited T-cell chemotaxis toward SDF-1α, while SIVpbj1.9 Nef did not. A hybrid between HIV-1 and SIVpbj1.9 Nef completely abrogated the chemotaxis blockade by HIV-1 Nef while failing to activate T cells without CD3/CD28 co-stimulation. Mutations in the myristoylation and SH3-binding site, but not the basic-rich domain, in Nef were unresponsive to CD3/CD28 stimulation but reversed the inhibition of migration. These findings indicate that the signals for T-cell activation by Nef do not necessarily parallel those for T-cell migration.
Collapse
Affiliation(s)
- In-Woo Park
- Department of Cell Biology and Anatomy, Graduate School of Biomedical Sciences, University of North Texas Health Science Center, CBH-316, 3500 Camp Bowie Blvd, Fort Worth, TX 76107, USA.
| | | |
Collapse
|
22
|
HIV-1 Nef interferes with T-lymphocyte circulation through confined environments in vivo. Proc Natl Acad Sci U S A 2012; 109:18541-6. [PMID: 23093676 DOI: 10.1073/pnas.1204322109] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
HIV-1 negative factor (Nef) elevates virus replication and contributes to immune evasion in vivo. As one of its established in vitro activities, Nef interferes with T-lymphocyte chemotaxis by reducing host cell actin dynamics. To explore Nef's influence on in vivo recirculation of T lymphocytes, we assessed lymph-node homing of Nef-expressing primary murine lymphocytes and found a drastic impairment in homing to peripheral lymph nodes. Intravital imaging and 3D immunofluorescence reconstruction of lymph nodes revealed that Nef potently impaired T-lymphocyte extravasation through high endothelial venules and reduced subsequent parenchymal motility. Ex vivo analyses of transendothelial migration revealed that Nef disrupted T-lymphocyte polarization and interfered with diapedesis and migration in the narrow subendothelial space. Consistently, Nef specifically affected T-lymphocyte motility modes used in dense environments that pose high physical barriers to migration. Mechanistically, inhibition of lymph node homing, subendothelial migration and cell polarization, but not diapedesis, depended on Nef's ability to inhibit host cell actin remodeling. Nef-mediated interference with in vivo recirculation of T lymphocytes may compromise T-cell help and thus represents an important mechanism for its function as a HIV pathogenicity factor.
Collapse
|
23
|
Chandrasekaran P, Buckley M, Moore V, Wang LQ, Kehrl JH, Venkatesan S. HIV-1 Nef impairs heterotrimeric G-protein signaling by targeting Gα(i2) for degradation through ubiquitination. J Biol Chem 2012; 287:41481-98. [PMID: 23071112 DOI: 10.1074/jbc.m112.361782] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The HIV Nef protein is an important pathogenic factor that modulates cell surface receptor trafficking and impairs cell motility, presumably by interfering at multiple steps with chemotactic receptor signaling. Here, we report that a dominant effect of Nef is to trigger AIP4 E3 ligase-mediated Gα(i2) ubiquitination, which leads to Gα(i2) endolysosomal sequestration and destruction. The loss of the Gα(i2) subunit was demonstrable in many cell types in the context of gene transfection, HIV infection, or Nef protein transduction. Nef directly interacts with Gα(i2) and ternary complexes containing AIP4, Nef, and Gα(i2) form. A substantial reversal of Gα(i2) loss and a partial recovery of impaired chemotaxis occurred following siRNA knockdown of AIP4 or NEDD4 or by inhibiting dynamin. The N-terminal myristoyl group, (62)EEEE(65) motif, and (72)PXXP(75) motif of Nef are critical for this effect to occur. Nef expression does not affect a Gq(i5) chimera where the five C-terminal residues of Gq are replaced with those of Gα(i2). Lysine at position 296 of Gα(i2) was identified as the critical determinant of Nef-induced degradation. By specifically degrading Gα(i2), Nef directly subverts leukocyte migration and homing. Impaired trafficking and homing of HIV Nef-expressing lymphocytes probably contributes to early immune dysfunction following HIV infection.
Collapse
Affiliation(s)
- Prabha Chandrasekaran
- Laboratory of Molecular Immunology, NIAID, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | | | | | | | | | |
Collapse
|
24
|
Saxena SK, Shrivastava G, Tiwari S, Swamy MA, Nair MP. Modulation of HIV pathogenesis and T-cell signaling by HIV-1 Nef. Future Virol 2012; 7:609-620. [PMID: 22844345 DOI: 10.2217/fvl.12.42] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
HIV-1 Nef protein is an approximately 27-kDa myristoylated protein that is a virulence factor essential for efficient viral replication and infection in CD4(+) T cells. The functions of CD4(+) T cells are directly impeded after HIV infection. HIV-1 Nef plays a crucial role in manipulating host cellular machinery and in HIV pathogenesis by reducing the ability of infected lymphocytes to form immunological synapses by promoting virological synapses with APCs, and by affecting T-cell stimulation. This article reviews the current status of the efficient Nef-mediated spread of virus in the unreceptive environment of the immune system by altering CD4(+) T-lymphocyte signaling, intracellular trafficking, cell migration and apoptotic pathways.
Collapse
Affiliation(s)
- Shailendra K Saxena
- CSIR-Centre for Cellular & Molecular Biology, Uppal Road, Hyderabad 500007 (AP), India
| | | | | | | | | |
Collapse
|
25
|
Cai C, Rodepeter FR, Rossmann A, Teymoortash A, Lee JS, Quint K, Di Fazio P, Ocker M, Werner JA, Mandic R. Nef from SIV(mac239) decreases proliferation and migration of adenoid-cystic carcinoma cells and inhibits angiogenesis. Oral Oncol 2011; 47:847-854. [PMID: 21763177 DOI: 10.1016/j.oraloncology.2011.06.502] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2011] [Revised: 06/20/2011] [Accepted: 06/21/2011] [Indexed: 02/07/2023]
Abstract
The HIV/SIV accessory protein Nef is known to down-modulate cell surface receptors that are required for virus entry such as CD4, CCR5 and CXCR4 to block lethal viral superinfection of the infected cell. The chemokine receptor CXCR4 also plays an important role in promoting cell proliferation, metastasis and tumor angiogenesis. Therefore it was of interest to evaluate if Nef can down-regulate CXCR4 in tumor cells since this could affect these critical prognostic parameters. The CXCR4-expressing cell line ACC3 that was derived from a salivary gland adenoid cystic carcinoma (ACC) of the head and neck was transfected with Nef from SIV(mac239) and cell surface expression of the receptor was monitored by FACS analysis. Real time proliferation of cells was measured with the xCELLigence system (Roche, Mannheim, Germany). Cell migration was detected by an in vitro scratch assay. Similarly, COS-7 cells were co-transfected with CXCR4 and Nef and were treated as described for ACC3. In vitro tube formation was deployed to assess the effect of Nef on angiogenesis. siRNA was used for CXCR4 knockdown. Cell surface down-modulation of endogenous CXCR4 could be observed in ACC3 cells after Nef-transfection as well as in COS-7 cells after co-transfection of CXCR4 and Nef. Proliferation as well as migration of Nef-transfected ACC3 tumor cells appeared significantly reduced. In vitro tube formation was significantly lowered after Nef-transfection or CXCR4 knockdown with siRNA. SIV-Nef could serve as an interesting tool to study the biologic behavior of CXCR4-expressing tumors such as ACC. Deploying SIV-Nef thereby could help in the discovery of new therapeutic approaches for the treatment of ACC and other CXCR4-expressing tumors.
Collapse
Affiliation(s)
- Chengzhong Cai
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Giessen and Marburg, Campus Marburg, Baldingerstrasse, D-35033 Marburg, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Fujii H, Ato M, Takahashi Y, Otake K, Hashimoto SI, Kaji T, Tsunetsugu-Yokota Y, Fujita M, Adachi A, Nakayama T, Taniguchi M, Koyasu S, Takemori T. HIV-1 Nef impairs multiple T-cell functions in antigen-specific immune response in mice. Int Immunol 2011; 23:433-41. [PMID: 21642447 DOI: 10.1093/intimm/dxr031] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The viral protein Nef is a key element for the progression of HIV disease. Previous in vitro studies suggested that Nef expression in T-cell lines enhanced TCR signaling pathways upon stimulation with TCR cross-linking, leading to the proposal that Nef lowers the threshold of T-cell activation, thus increasing susceptibility to viral replication in immune response. Likewise, the in vivo effects of Nef transgenic mouse models supported T-cell hyperresponse by Nef. However, the interpretation is complicated by Nef expression early in the development of T cells in these animal models. Here, we analyzed the consequence of Nef expression in ovalbumin-specific/CD4(+) peripheral T cells by using a novel mouse model and demonstrate that Nef inhibits antigen-specific T-cell proliferation and multiple functions required for immune response in vivo, which includes T-cell helper activity for the primary and memory B-cell response. However, Nef does not completely abrogate T-cell activity, as defined by low levels of cytokine production, which may afford the virus a replicative advantage. These results support a model, in which Nef expression does not cause T-cell hyperresponse in immune reaction, but instead reduces the T-cell activity, that may contribute to a low level of virus spread without viral cytopathic effects.
Collapse
Affiliation(s)
- Hideki Fujii
- Department of Microbiology and Immunology, Keio University School of, Medicine, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Konradt C, Frigimelica E, Nothelfer K, Puhar A, Salgado-Pabon W, di Bartolo V, Scott-Algara D, Rodrigues C, Sansonetti P, Phalipon A. The Shigella flexneri Type Three Secretion System Effector IpgD Inhibits T Cell Migration by Manipulating Host Phosphoinositide Metabolism. Cell Host Microbe 2011; 9:263-72. [DOI: 10.1016/j.chom.2011.03.010] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2010] [Revised: 02/02/2011] [Accepted: 03/18/2011] [Indexed: 11/24/2022]
|
28
|
Stolp B, Fackler OT. How HIV takes advantage of the cytoskeleton in entry and replication. Viruses 2011; 3:293-311. [PMID: 21994733 PMCID: PMC3185699 DOI: 10.3390/v3040293] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2011] [Revised: 03/11/2011] [Accepted: 03/19/2011] [Indexed: 01/08/2023] Open
Abstract
The host cell cytoskeleton plays a key role in the life cycle of viral pathogens whose propagation depends on mandatory intracellular steps. Accordingly, also the human immunodeficiency virus type 1 (HIV-1) has evolved strategies to exploit and modulate in particular the actin cytoskeleton for its purposes. This review will recapitulate recent findings on how HIV-1 hijacks the cytoskeleton to facilitate entry into, transport within and egress from host cells as well as to commandeer communication of infected with uninfected bystander cells.
Collapse
Affiliation(s)
- Bettina Stolp
- Department of Infectious Diseases, Virology, University Hospital Heidelberg, Heidelberg, Germany.
| | | |
Collapse
|
29
|
Laguette N, Brégnard C, Benichou S, Basmaciogullari S. Human immunodeficiency virus (HIV) type-1, HIV-2 and simian immunodeficiency virus Nef proteins. Mol Aspects Med 2010; 31:418-33. [PMID: 20594957 DOI: 10.1016/j.mam.2010.05.003] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2010] [Accepted: 05/26/2010] [Indexed: 11/19/2022]
Abstract
The genomes of all retroviruses encode the Gag Pol and Env structural proteins. Human and simian lentiviruses have acquired non-structural proteins among which Nef plays a major role in the evolution of viral infection towards an immunodeficiency syndrome. Indeed, in the absence of a functional nef gene, primate lentiviruses are far less pathogenic than their wild type counterparts. The multiple protein-protein interactions in which Nef is involved all contribute to explain the role played by Nef in HIV- and SIV-associated disease progression. This review summarizes common and distinct features among Nef proteins and how they contribute to increasing HIV and SIV fitness towards their respective hosts.
Collapse
Affiliation(s)
- Nadine Laguette
- Institut Cochin, CNRS UMR8104, Université Paris Descartes, Paris, France
| | | | | | | |
Collapse
|
30
|
HIV-1 Nef inhibits ruffles, induces filopodia, and modulates migration of infected lymphocytes. J Virol 2009; 84:2282-93. [PMID: 20015995 DOI: 10.1128/jvi.02230-09] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The HIV-1 Nef protein is a pathogenic factor modulating the behavior of infected cells. Nef induces actin cytoskeleton changes and impairs cell migration toward chemokines. We further characterized the morphology, cytoskeleton dynamics, and motility of HIV-1-infected lymphocytes. By using scanning electron microscopy, confocal immunofluorescence microscopy, and ImageStream technology, which combines flow cytometry and automated imaging, we report that HIV-1 induces a characteristic remodeling of the actin cytoskeleton. In infected lymphocytes, ruffle formation is inhibited, whereas long, thin filopodium-like protrusions are induced. Cells infected with HIV with nef deleted display a normal phenotype, and Nef expression alone, in the absence of other viral proteins, induces morphological changes. We also used an innovative imaging system to immobilize and visualize living individual cells in suspension. When combined with confocal "axial tomography," this technique greatly enhances three-dimensional optical resolution. With this technique, we confirmed the induction of long filopodium-like structures in unfixed Nef-expressing lymphocytes. The cytoskeleton reorganization induced by Nef is associated with an important impairment of cell movements. The adhesion and spreading of infected cells to fibronectin, their spontaneous motility, and their migration toward chemokines (CXCL12, CCL3, and CCL19) were all significantly decreased. Therefore, Nef induces complex effects on the lymphocyte actin cytoskeleton and cellular morphology, which likely impacts the capacity of infected cells to circulate and to encounter and communicate with bystander cells.
Collapse
|
31
|
HIV-1 Nef Interferes with Host Cell Motility by Deregulation of Cofilin. Cell Host Microbe 2009; 6:174-86. [DOI: 10.1016/j.chom.2009.06.004] [Citation(s) in RCA: 111] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2009] [Revised: 04/30/2009] [Accepted: 06/01/2009] [Indexed: 11/21/2022]
|
32
|
Park IW, He JJ. HIV-1 Nef-mediated inhibition of T cell migration and its molecular determinants. J Leukoc Biol 2009; 86:1171-8. [PMID: 19641037 DOI: 10.1189/jlb.0409261] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Lymphocyte trafficking is a multistep, intricate process and involves a number of host factors such as integrins and chemokine receptors on lymphocytes, adhesion molecules on endothelial cells, and chemokines present in the local microenvironment. Previous studies have shown that HIV-1 Nef inhibits T cell chemotaxis in response to the physiological ligand SDF-1alpha [( 1) ]. In this study, we aimed to gain a better understanding of the inhibitory mechanisms and to define the molecular determinants of HIV-1 Nef for this phenotype. We showed that HIV-1 Nef inhibited transwell and transendothelial migration of T cells. Specifically, HIV-1 Nef protein impaired T cell chemotaxis toward SDF-1alpha without altering CXCR4 expression. Moreover, we showed that HIV-1 Nef protein down-modulated LFA-1 expression on T lymphocytes and diminished adhesion and polarization of T lymphocytes and as a result, led to decreased migration across the endothelium. Furthermore, we showed that the myristoylation site and DeltaSD domain played important roles in Nef-mediated inhibition of transwell and transendothelial migration and polarization of T lymphocytes; however, different sites or domains were needed for Nef-mediated LFA-1 down-modulation and impaired adhesion of T lymphocyte. Taken together, these results demonstrated that HIV-1 Nef inhibited T lymphocyte migration at multiple steps and suggest that membrane localization and intracellular signaling events likely contribute to the inhibitory effects of Nef on T cell migration and subsequently, the pathobiology of the HIV-1 Nef protein.
Collapse
Affiliation(s)
- In-Woo Park
- Center for AIDS Research and Department of Microbiology and Immunology, Indiana University School of Medicine, R2 302, 950 W. Walnut St., Indianapolis, IN 46202, USA.
| | | |
Collapse
|
33
|
Arhel NJ, Kirchhoff F. Implications of Nef: host cell interactions in viral persistence and progression to AIDS. Curr Top Microbiol Immunol 2009; 339:147-75. [PMID: 20012528 DOI: 10.1007/978-3-642-02175-6_8] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The HIV and SIV Nef accessory proteins are potent enhancers of viral persistence and accelerate progression to AIDS in HIV-1-infected patients and non-human primate models. Although relatively small (27-35 kD), Nef can interact with a multitude of cellular factors and induce complex changes in trafficking, signal transduction, and gene expression that together converge to promote viral replication and immune evasion. In particular, Nef recruits several immunologically relevant cellular receptors to the endocytic machinery to reduce the recognition and elimination of virally infected cells by the host immune system, while simultaneously interacting with various kinases to promote T cell activation and viral replication. This review provides an overview on selected Nef interactions with host cell proteins, and discusses their possible relevance for viral spread and pathogenicity.
Collapse
Affiliation(s)
- Nathalie J Arhel
- Institute of Virology, Universitätsklinikum Ulm, Albert-Einstein-Allee 11, 89081 Ulm, Germany
| | | |
Collapse
|
34
|
Lee CMY, Gala S, Stewart GJ, Williamson P. The Proline-Rich Region of HIV-1 Nef Affects CXCR4-Mediated Chemotaxis in Jurkat T Cells. Viral Immunol 2008; 21:347-54. [DOI: 10.1089/vim.2007.0093] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Carol Man Yuk Lee
- The Institute for Immunology and Allergy Research, Westmead Millennium Institute, Sydney, New South Wales, Australia
- Faculty of Medicine, The Unversity of Sydney, Sydney, New South Wales, Australia
| | - Salvador Gala
- The Institute for Immunology and Allergy Research, Westmead Millennium Institute, Sydney, New South Wales, Australia
| | - Graeme John Stewart
- The Institute for Immunology and Allergy Research, Westmead Millennium Institute, Sydney, New South Wales, Australia
- Faculty of Medicine, The Unversity of Sydney, Sydney, New South Wales, Australia
| | - Peter Williamson
- The Institute for Immunology and Allergy Research, Westmead Millennium Institute, Sydney, New South Wales, Australia
- Centre for Advanced Technologies in Animal Genetics and Reproduction, Faculty of Veterinary Science, The University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
35
|
Hidalgo-Estévez AM, Punzón C, Sanchez-Duffhues G, Muñoz E, Fresno M. HIV-1-Tat potentiates CXCL12/stromal cell-derived factor 1-induced downregulation of membrane CXCR4 in T lymphocytes through protein kinase C zeta. Mol Immunol 2008; 46:106-15. [PMID: 18760839 DOI: 10.1016/j.molimm.2008.07.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2008] [Revised: 07/04/2008] [Accepted: 07/08/2008] [Indexed: 12/25/2022]
Abstract
We have investigated the role of intracellular HIV-1 Tat on CXCR4 expression on T cells. We found that stable or doxycycline-regulated expression of HIV-1 Tat on Jurkat T cells results in lower cell surface expression of CXCR4, but not of other chemokine receptors. This effect was not due to an alteration in CXCR4 transcription, and total CXCR4 levels remained unaltered. Rather, when cells were treated with CXCL12/Stromal Cell-Derived Factor 1, a faster downmodulation of CXCR4 was observed although resurfacing was unaffected. Similar effect was seen in peripheral human T cells transiently transfected with Tat. At the molecular level Tat did not alter cellular levels of G-coupled receptor kinases 2 and 6 and beta-arrestin, proteins involved in CXCR4 downregulation. Neither Tat significantly affected phosphatidylinositol 3-kinase activation in response to CXCL12. Interestingly, in Jurkat cell clones stably expressing both Protein kinase (PK)-Czeta and HIV-1 Tat, CXCL12 induced a faster CXCR4 internalization than in cells only expressing HIV-1 Tat. In contrast in Jurkat cell stably expressing a dominant negative PKCzeta, Tat enhancement of CXCR4 internalization was abrogated. Thus, our results show a new function of HIV-1 Tat, its ability to regulate CXCR4 expression via PKCzeta. The significance of those results is discussed.
Collapse
Affiliation(s)
- Alicia M Hidalgo-Estévez
- Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), Universidad Autónoma de Madrid, c/Nicolás Cabrera, 1. Campus Universitario de Cantoblanco, 28049 Madrid, Spain
| | | | | | | | | |
Collapse
|
36
|
Meditz AL, Schlichtemeier R, Folkvord JM, Givens M, Lesh KC, Ray M, McCarter MD, Connick E. SDF-1alpha is a potent inducer of HIV-1-Specific CD8+ T-cell chemotaxis, but migration of CD8+ T cells is impaired at high viral loads. AIDS Res Hum Retroviruses 2008; 24:977-85. [PMID: 18671480 DOI: 10.1089/aid.2007.0259] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Multiple impairments in HIV-1-specific cytotoxic T cells (CTL) have been reported, but derangements in HIV-1-specific CD8+ T-cell chemotaxis have not been described previously. We assessed migration to SDF-1alpha (stromal cell-derived factor 1-alpha) and CX3CL1 in vitro and expression of cognate receptors, CXCR4 and CX3CR1, by flow cytometry in peripheral blood and lymph node CD8+ T cells from HIV-1-seropositive and -seronegative individuals. Compared with seronegative individuals, percentages of CXCR4+CD8+ T cells were reduced (median, 26% versus 74%, p < 0.001) and percentages of CX3CR1+CD8+ T cells were increased (median, 33% versus 15%, p = 0.03) in seropositive individuals. Robust migration of peripheral blood mononuclear cell (PBMC) CD8+ T cells to SDF-1alpha (1 alphag/ml) was observed in both HIV-1-seropositive (median chemotactic index [CI] 4.9) and -seronegative (median CI 2.8) subjects (p = 0.46). CI to SDF-1alpha was not significantly related to percentage of CXCR4+CD8+ T cells or density of CXCR4, but correlated inversely with plasma HIV-1 RNA concentration (r = -0.82, p = 0.03). Little chemotaxis was observed in response to CX3CL1 and it was unrelated to CX3CR1 expression. Lymph node CD8+ T-cell chemotaxis to SDF-1alpha and CX3CL1 in four subjects demonstrated the same patterns observed in PBMC. HIV-1-specific tetramer-staining CD8+ T cells exhibited chemotaxis of similar magnitude as PBMC CD8+ T cells in a subset of subjects. These data suggest that SDF-1alpha is a potent chemoattractant for HIV-1-specific CTL, but that impairments in migration of HIV-1-specific CTL may exist at high viral loads. Improved understanding of the determinants of CTL localization may provide insight into novel therapies to enhance delivery of CTL to sites of HIV-1 replication.
Collapse
Affiliation(s)
- Amie L. Meditz
- Division of Infectious Diseases, Department of Medicine, University of Colorado Health Sciences Center, Denver, Colorado 80262
| | - Rick Schlichtemeier
- Division of Infectious Diseases, Department of Medicine, University of Colorado Health Sciences Center, Denver, Colorado 80262
| | - Joy M. Folkvord
- Division of Infectious Diseases, Department of Medicine, University of Colorado Health Sciences Center, Denver, Colorado 80262
| | - Monique Givens
- Division of Infectious Diseases, Department of Medicine, University of Colorado Health Sciences Center, Denver, Colorado 80262
- Present address: Research and Development, Gambro BCT, Lakewood, Colorado 80215
| | - Kevin C. Lesh
- Division of Infectious Diseases, Department of Medicine, University of Colorado Health Sciences Center, Denver, Colorado 80262
- Present address: Lynn Institute of the Rockies, Colorado Springs, Colorado 80909
| | - M.G. Ray
- Division of Infectious Diseases, Department of Medicine, University of Colorado Health Sciences Center, Denver, Colorado 80262
| | - Martin D. McCarter
- Department of Surgery, University of Colorado Health Sciences Center, Denver, Colorado 80262
| | - Elizabeth Connick
- Division of Infectious Diseases, Department of Medicine, University of Colorado Health Sciences Center, Denver, Colorado 80262
| |
Collapse
|
37
|
Venzke S, Michel N, Allespach I, Fackler OT, Keppler OT. Expression of Nef downregulates CXCR4, the major coreceptor of human immunodeficiency virus, from the surfaces of target cells and thereby enhances resistance to superinfection. J Virol 2006; 80:11141-52. [PMID: 16928758 PMCID: PMC1642143 DOI: 10.1128/jvi.01556-06] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Lentiviral Nef proteins are key factors for pathogenesis and are known to downregulate functionally important molecules, including CD4 and major histocompatibility complex class I (MHC-I), from the surfaces of infected cells. Recently, we demonstrated that Nef reduces cell surface levels of the human immunodeficiency virus type 1 (HIV-1) entry coreceptor CCR5 (N. Michel, I. Allespach, S. Venzke, O. T. Fackler, and O. T. Keppler, Curr. Biol. 15:714-723, 2005). Here, we report that Nef downregulates the second major HIV-1 coreceptor, CXCR4, from the surfaces of HIV-infected primary CD4 T lymphocytes with efficiencies comparable to those of the natural CXCR4 ligand, stromal cell-derived factor-1 alpha. Analysis of a panel of mutants of HIV-1(SF2) Nef revealed that the viral protein utilized the same signature motifs for downmodulation of CXCR4 and MHC-I, including the proline-rich motif P(73)P(76)P(79)P(82) and the acidic cluster motif E(66)E(67)E(68)E(69.) Expression of wild-type Nef, but not of specific Nef mutants, resulted in a perinuclear accumulation of the coreceptor. Remarkably, the carboxy terminus of CXCR4, which harbors the classical motifs critical for basal and ligand-induced receptor endocytosis, was dispensable for the Nef-mediated reduction of surface exposure. Functionally, the ability of Nef to simultaneously downmodulate CXCR4 and CD4 correlated with maximum-level protection of Nef-expressing target cells from fusion with cells exposing X4 HIV-1 envelopes. Furthermore, the Nef-mediated downregulation of CXCR4 alone on target T lymphocytes was sufficient to diminish cells' susceptibility to X4 HIV-1 virions at the entry step. The downregulation of chemokine coreceptors is a conserved activity of Nef to modulate infected cells, an important functional consequence of which is an enhanced resistance to HIV superinfection.
Collapse
Affiliation(s)
- Stephanie Venzke
- Department of Virology, University of Heidelberg, Im Neuenheimer Feld 324, 69120 Heidelberg, Germany
| | | | | | | | | |
Collapse
|
38
|
Abstract
Infectious disease immunology has largely focused on the effector immune response, changes in the blood and peripheral lymphoid organs of infected individuals, and vaccine development. Studies of the thymus in infected individuals have been neglected, although this is progressively changing. The thymus is a primary lymphoid organ, able to generate mature T cells that eventually colonize secondary lymphoid organs, and is therefore essential for peripheral T cell renewal. Recent data show that normal thymocyte development and export can be altered as a result of an infectious disease. One common feature is the severe atrophy of the infected organ, mainly due to the apoptosis-related depletion of immature CD4+CD8+ thymocytes. Additionally, thymocyte proliferation is frequently diminished. The microenvironmental compartment of the thymus is also affected, particularly in acute infectious diseases, with a densification of the epithelial network and an increase in the deposition of extracellular matrix. In the murine model of Chagas disease, intrathymic chemokine production is also enhanced, and thymocytes from Trypanosoma cruzi-infected mice exhibit greater numbers of cell migration-related receptors for chemokines and extracellular matrix, as well as increased migratory responses to the corresponding ligands. This profile is correlated with the appearance of potentially autoreactive thymus-derived immature CD4+CD8+ T cells in peripheral organs of infected animals. A variety of infectious agents—including viruses, protozoa, and fungi—invade the thymus, raising the hypothesis of the generation of central immunological tolerance for at least some of the infectious agent-derived antigens. It seems clear that the thymus is targeted in a variety of infections, and that such targeting may have consequences on the behavior of peripheral T lymphocytes. In this context, thymus-centered immunotherapeutic approaches potentially represent a new tool for the treatment of severe infectious diseases.
Collapse
Affiliation(s)
- Wilson Savino
- Laboratory on Thymus Research, Department of Immunology, Oswaldo Cruz Institute, Inserm-Fiocruz Associated Laboratory of Immunology, Oswaldo Cruz Foundation, Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
39
|
Abstract
Human immunodeficiency virus generates the accessory proteins Nef, viral infectivity factor (Vif), viral protein R, and viral protein U or viral protein X during viral replication in host cells. Although the significance of these accessory proteins is often lost in vitro, they are essential for viral pathogenesis in vivo. Therefore, these proteins have much potential as antiviral targets. Recent data reveal Vif perturbs an ill-defined antiviral pathway in host cells allowing HIV replication. These data highlight a common feature among HIV accessory proteins in manipulating the host to aid viral pathogenesis. Therefore, these new insights into Vif and other HIV accessory proteins are reviewed, emphasizing host cell interactions and new targets for therapeutic intervention.
Collapse
Affiliation(s)
- Jenny L Anderson
- Department of Microbiology and Immunology, The University of Illinois at Chicago, Chicago, IL 60612, USA.
| | | |
Collapse
|
40
|
Hrecka K, Swigut T, Schindler M, Kirchhoff F, Skowronski J. Nef proteins from diverse groups of primate lentiviruses downmodulate CXCR4 to inhibit migration to the chemokine stromal derived factor 1. J Virol 2005; 79:10650-9. [PMID: 16051857 PMCID: PMC1182621 DOI: 10.1128/jvi.79.16.10650-10659.2005] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2005] [Accepted: 05/20/2005] [Indexed: 11/20/2022] Open
Abstract
Nef proteins of primate lentiviruses promote viral replication, virion infectivity, and evasion of antiviral immune responses by modulating signal transduction pathways and downregulating expression of receptors at the cell surface that are important for efficient antigen-specific responses, such as CD4, CD28, T-cell antigen receptor, and class I and class II major histocompatibility complex. Here we show that Nef proteins from diverse groups of primate lentiviruses which do not require the chemokine receptor CXCR4 for entry into target cells strongly downmodulate the cell surface expression of CXCR4. In contrast, all human immunodeficiency virus type 1 (HIV-1) and the majority of HIV-2 Nef proteins tested did not have such strong effects. SIVmac239 Nef strongly inhibited lymphocyte migration to CXCR4 ligand, the chemokine stromal derived factor 1 (SDF-1). SIVmac239 Nef downregulated CXCR4 by accelerating the rate of its endocytosis. Downmodulation of CXCR4 was abolished by mutations that disrupt the constitutively strong AP-2 clathrin adaptor binding element located in the N-terminal region of the Nef molecule, suggesting that Nef accelerates CXCR4 endocytosis via an AP-2-dependent pathway. Together, these results point to CXCR4 as playing an important role in simian immunodeficiency virus and possibly also HIV-2 persistence in vivo that is unrelated to viral entry into target cells. We speculate that Nef targets CXCR4 to disrupt ordered trafficking of infected leukocytes between local microenvironments in order to facilitate their dissemination and/or impair the antiviral immune response.
Collapse
Affiliation(s)
- Kasia Hrecka
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | | | | | | | | |
Collapse
|
41
|
Abstract
Dendritic cells are critical for host immunity and are involved both in the innate and adaptive immune responses. They are among the first cells targeted by HIV-1 in vivo at mucosal sites. Dendritic cells can sequester HIV-1 in endosomal compartments for several days and transmit infectious HIV-1 to interacting T cells in the lymph node, which is the most important site for viral replication and spread. Initially, the cellular immune response developed against HIV-1 is strong, but eventually it fails to control and resolve the infection. The most dramatic effect seen on the immune system during untreated HIV-1 infection is the destruction of helper CD4(+) T cells, which leads to subsequent immune deficiency. However, the immunomodulatory effects of HIV-1 on different dendritic cell subpopulations may also play an important role in the pathogenesis of HIV-1. This review discusses the effects HIV-1 exerts on dendritic cells in vivo and in vitro, including the binding and uptake of HIV by dendritic cells, the formation of infectious synapses, infection, and the role of dendritic cells in HIV-1 pathogenesis.
Collapse
Affiliation(s)
- Marie Larsson
- New York University, School of Medicine, 550 First Avenue, MSB 507, New York, NY 10016, USA.
| |
Collapse
|
42
|
Poggi A, Carosio R, Fenoglio D, Brenci S, Murdaca G, Setti M, Indiveri F, Scabini S, Ferrero E, Zocchi MR. Migration of V delta 1 and V delta 2 T cells in response to CXCR3 and CXCR4 ligands in healthy donors and HIV-1-infected patients: competition by HIV-1 Tat. Blood 2004; 103:2205-2213. [PMID: 14630801 DOI: 10.1182/blood-2003-08-2928] [Citation(s) in RCA: 100] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
We show that HIV-1-infected patients have increased concentrations of circulating V delta 1 T cells (2.2%-9.0% of T lymphocytes; healthy donors, 1.0%-2%) and, in some instances, V delta 2 T cells (3.5%-4.8% vs 2.0%-3.3%). In these patients, both V delta 1 and V delta 2 T cells are CXCR3+CXCR4+, whereas in healthy donors CXCR4 was preferentially expressed on V delta 1 T lymphocytes. gamma delta T cells transmigrated across endothelial monolayers, in response to interferon-gamma-inducing protein-10 (IP-10/CXCL10), stromal cell-derived factor-1 (SDF-1/CXCL12), or both, according to the expression of the specific receptors CXCR3 and CXCR4. Interestingly, 6Ckine/SLC/CCL21 was more effective than IP-10/CXCL10 on V delta 1 CXCR3+ cells, whereas V delta 2 CXCR3+ cells were driven more efficiently by IP-10/CXCL10. IP-10/CXCL10- and SDF-1/CXCL12-induced transmigration was dependent on phosphoinositide-3 kinase (PI-3K), as demonstrated by the use of the specific blockers wortmannin and LY294002 and by the activation of the downstream serine kinase Akt/PKB on ligation of CXCR3 and CXCR4. Occupancy of CXCR3, but not of CXCR4, led to CAMKII activation; accordingly, the CAMKII inhibitors KN62 and KN93 decreased IP-10/CXCL10- but not SDF-1/CXCL12-driven transmigration. Finally, HIV-1 Tat, which is present in the serum of HIV-1-infected patients, interferes with the chemotactic activity of these chemokines because of the cysteine-rich domain of the protein, which contains CXC and CC chemokine-like sequences.
Collapse
MESH Headings
- Adult
- Calcium-Calmodulin-Dependent Protein Kinase Type 2
- Calcium-Calmodulin-Dependent Protein Kinases/metabolism
- Cell Movement/immunology
- Cells, Cultured
- Chemokine CXCL10
- Chemokine CXCL12
- Chemokines, CXC/metabolism
- Endothelium, Vascular/cytology
- Female
- Gene Products, tat/chemistry
- Gene Products, tat/metabolism
- HIV Infections/immunology
- HIV-1
- Humans
- Ligands
- Male
- Middle Aged
- Phosphatidylinositol 3-Kinases/metabolism
- Protein Binding/immunology
- Protein Structure, Tertiary
- Receptors, CXCR3
- Receptors, CXCR4/metabolism
- Receptors, Chemokine/chemistry
- Receptors, Chemokine/metabolism
- T-Lymphocyte Subsets/cytology
- T-Lymphocyte Subsets/metabolism
- T-Lymphocyte Subsets/virology
- Umbilical Veins/cytology
- tat Gene Products, Human Immunodeficiency Virus
Collapse
Affiliation(s)
- Alessandro Poggi
- Laboratory of Immunology, National Institute fo Cancer Research, Largo R. Benzi 10, I-16132, Genoa, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Abstract
HIV produces structural, regulatory, and accessory proteins during viral replication in host cells. The accessory proteins include Nef, viral infectivity factor (Vif), viral protein R, and viral protein U or viral protein X. Although these accessory proteins are generally dispensable for viral replication in vitro, they are essential for viral pathogenesis in vivo. Consequently, there has been much interest in understanding how these accessory proteins function because this research may yield new antiviral targets to curb HIV pathogenesis in vivo. Therefore, this review highlights recent advances in understanding the HIV accessory proteins and emphasizes breakthrough insights into the elusive Vif protein and potential new targets for therapeutic intervention.
Collapse
Affiliation(s)
- Jenny L. Anderson
- Department of Microbiology and Immunology, The University of Illinois at Chicago, Chicago, IL 60612, USA.
| | | |
Collapse
|
44
|
Fernandis AZ, Cherla RP, Ganju RK. Differential regulation of CXCR4-mediated T-cell chemotaxis and mitogen-activated protein kinase activation by the membrane tyrosine phosphatase, CD45. J Biol Chem 2003; 278:9536-43. [PMID: 12519755 DOI: 10.1074/jbc.m211803200] [Citation(s) in RCA: 73] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The chemokine receptor CXCR4 and its cognate ligand, stromal cell-derived factor-1alpha (CXCL12), regulate lymphocyte trafficking and play an important role in host immune surveillance. However, the molecular mechanisms involved in CXCL12-induced and CXCR4-mediated chemotaxis of T-lymphocytes are not completely elucidated. In the present study, we examined the role of the membrane tyrosine phosphatase CD45, which regulates antigen receptor signaling in CXCR4-mediated chemotaxis and mitogen-activated protein kinase (MAPK) activation in T-cells. We observed a significant reduction in CXCL12-induced chemotaxis in the CD45-negative Jurkat cell line (J45.01) as compared with the CD45-positive control (JE6.1) cells. Expression of a chimeric protein containing the intracellular phosphatase domain of CD45 was able to partially restore CXCL12-induced chemotaxis in the J45.01 cells. However, reconstitution of CD45 into the J45.01 cells restored the CXCL12-induced chemotaxis to about 90%. CD45 had no significant effect on CXCL12 or human immunodeficiency virus gp120-induced internalization of the CXCR4 receptor. Furthermore, J45.01 cells showed a slight enhancement in CXCL12-induced MAP kinase activity as compared with the JE6.1 cells. We also observed that CXCL12 treatment enhanced the tyrosine phosphorylation of CD45 and induced its association with the CXCR4 receptor. Pretreatment of T-cells with the lipid raft inhibitor, methyl-beta-cyclodextrin, blocked the association between CXCR4 and CD45 and markedly abolished CXCL12-induced chemotaxis. Comparisons of signaling pathways induced by CXCL12 in JE6.1 and J45.01 cells revealed that CD45 might moderately regulate the tyrosine phosphorylation of the focal adhesion components the related adhesion focal tyrosine kinase/Pyk2, focal adhesion kinase, p130Cas, and paxillin. CD45 has also been shown to regulate CXCR4-mediated activation and phosphorylation of T-cell receptor downstream effectors Lck, ZAP-70, and SLP-76. Our results show that CD45 differentially regulates CXCR4-mediated chemotactic activity and MAPK activation by modulating the activities of focal adhesion components and the downstream effectors of the T-cell receptor.
Collapse
MESH Headings
- Adaptor Proteins, Signal Transducing
- Blotting, Western
- Cell Line
- Chemokine CXCL12
- Chemokines, CXC/metabolism
- Chemotaxis
- Dose-Response Relationship, Drug
- Enzyme Activation
- Flow Cytometry
- Gene Expression Regulation
- Humans
- Jurkat Cells
- Leukocyte Common Antigens/metabolism
- Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/metabolism
- Lymphocytes/metabolism
- MAP Kinase Signaling System
- Microscopy, Confocal
- Microscopy, Fluorescence
- Phosphoproteins/metabolism
- Phosphorylation
- Precipitin Tests
- Protein Structure, Tertiary
- Protein-Tyrosine Kinases/metabolism
- Receptors, Antigen, T-Cell/metabolism
- Receptors, CXCR4/metabolism
- Signal Transduction
- T-Lymphocytes/metabolism
- Time Factors
- Tyrosine/metabolism
- ZAP-70 Protein-Tyrosine Kinase
Collapse
Affiliation(s)
- Aaron Z Fernandis
- Division of Experimental Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02115, USA
| | | | | |
Collapse
|