51
|
Silva JN, Polesskaya O, Wei HS, Rasheed IYD, Chamberlain JM, Nishimura C, Feng C, Dewhurst S. Chronic central nervous system expression of HIV-1 Tat leads to accelerated rarefaction of neocortical capillaries and loss of red blood cell velocity heterogeneity. Microcirculation 2015; 21:664-76. [PMID: 24813724 DOI: 10.1111/micc.12145] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Accepted: 05/06/2014] [Indexed: 12/24/2022]
Abstract
OBJECTIVES HIV-1 infection of the CNS is associated with impairment of CBF and neurocognitive function, and accelerated signs of aging. As normal aging is associated with rarefaction of the cerebral vasculature, we set out to examine chronic viral effects on the cerebral vasculature. METHODS DOX-inducible HIV-1 Tat-tg and WT control mice were used. Animals were treated with DOX for three weeks or five to seven months. Cerebral vessel density and capillary segment length were determined from quantitative image analyses of sectioned cortical tissue. In addition, movement of red blood cells in individual capillaries was imaged in vivo using multiphoton microscopy, to determine RBCV and flux. RESULTS Mean RBCV was not different between Tat-tg mice and age-matched WT controls. However, cortical capillaries from Tat-tg mice showed a significant loss of RBCV heterogeneity and increased RBCF that was attributed to a marked decrease in total cortical capillary length (35-40%) compared to WT mice. CONCLUSIONS Cerebrovascular rarefaction is accelerated in HIV-1 Tat-transgenic mice, and this is associated with alterations in red cell blood velocity. These changes may have relevance to the pathogenesis of HIV-associated neurocognitive disorders in an aging HIV-positive population.
Collapse
Affiliation(s)
- Jharon N Silva
- Departments of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York, USA
| | | | | | | | | | | | | | | |
Collapse
|
52
|
Atluri VSR, Hidalgo M, Samikkannu T, Kurapati KRV, Jayant RD, Sagar V, Nair MPN. Effect of human immunodeficiency virus on blood-brain barrier integrity and function: an update. Front Cell Neurosci 2015; 9:212. [PMID: 26113810 PMCID: PMC4461820 DOI: 10.3389/fncel.2015.00212] [Citation(s) in RCA: 98] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2015] [Accepted: 05/17/2015] [Indexed: 02/02/2023] Open
Abstract
The blood-brain barrier (BBB) is a diffusion barrier that has an important role in maintaining a precisely regulated microenvironment protecting the neural tissue from infectious agents and toxins in the circulating system. Compromised BBB integrity plays a major role in the pathogenesis of retroviral associated neurological diseases. Human Immunodeficiency Virus (HIV) infection in the Central Nervous System (CNS) is an early event even before the serodiagnosis for HIV positivity or the initiation of antiretroviral therapy (ART), resulting in neurological complications in many of the infected patients. Macrophages, microglia and astrocytes (in low levels) are the most productively/latently infected cell types within the CNS. In this brief review, we have discussed about the effect of HIV infection and viral proteins on the integrity and function of BBB, which may contribute to the progression of HIV associated neurocognitive disorders.
Collapse
Affiliation(s)
- Venkata Subba Rao Atluri
- Department of Immunology, Institute of NeuroImmune Pharmacology, Herbert Wertheim College of Medicine, Florida International University Miami, FL, USA
| | - Melissa Hidalgo
- Department of Immunology, Institute of NeuroImmune Pharmacology, Herbert Wertheim College of Medicine, Florida International University Miami, FL, USA
| | - Thangavel Samikkannu
- Department of Immunology, Institute of NeuroImmune Pharmacology, Herbert Wertheim College of Medicine, Florida International University Miami, FL, USA
| | - Kesava Rao Venkata Kurapati
- Department of Immunology, Institute of NeuroImmune Pharmacology, Herbert Wertheim College of Medicine, Florida International University Miami, FL, USA
| | - Rahul Dev Jayant
- Department of Immunology, Institute of NeuroImmune Pharmacology, Herbert Wertheim College of Medicine, Florida International University Miami, FL, USA
| | - Vidya Sagar
- Department of Immunology, Institute of NeuroImmune Pharmacology, Herbert Wertheim College of Medicine, Florida International University Miami, FL, USA
| | - Madhavan P N Nair
- Department of Immunology, Institute of NeuroImmune Pharmacology, Herbert Wertheim College of Medicine, Florida International University Miami, FL, USA
| |
Collapse
|
53
|
Zhou Y, Ge R, Wang R, Liu F, Huang Y, Liu H, Hao Y, Zhou Q, Wang C. UXT potentiates angiogenesis by attenuating Notch signaling. Development 2015; 142:774-86. [PMID: 25617435 PMCID: PMC4325377 DOI: 10.1242/dev.112532] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Angiogenesis is spatially and temporally orchestrated by a myriad of signaling pathways, including the Notch signaling pathway. Here, we identified UXT as an evolutionarily conserved and developmentally expressed protein, indispensable for intersegmental vessel (ISV) formation in zebrafish. Deficiency of UXT in zebrafish embryos results in shorter ISVs, loss of tip cell behavior, and impairment of endothelial cell migration and division. Significantly, UXT attenuates the expression of the Notch-responsive genes in vitro and in vivo. Mechanistically, UXT binds to the promoters of the Notch signaling target genes and specifically interacts with the transactivation region domain of the Notch intracellular domain (NICD), impairing the interaction between NICD and the transcription factor RBP-Jκ endogenously. This prevents RBP-Jκ/CSL from activation and thus inhibits the consequent gene inductions. Furthermore, blockade of Notch signaling rescues the angiogenesis defect caused by UXT knockdown both in vitro and in vivo. Taken together, the data presented in this study characterize UXT as a novel repressor of Notch signaling, shedding new light on the molecular regulation of angiogenesis. Summary: UXT/ART27 is a novel repressor of Notch signaling that impairs the interaction between NICD and RBP-Jκ/CSL and is indispensable for zebrafish intersegmental vessel formation.
Collapse
Affiliation(s)
- Yi Zhou
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Rui Ge
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Rui Wang
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Feng Liu
- State Key Laboratory of Biomembrane and Membrane Biotechnology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Yuefeng Huang
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Heng Liu
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yan Hao
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Qin Zhou
- The Division of Molecular Nephrology and the Creative Training Center for Undergraduates, Chongqing Medical University, Chongqing 400016, China
| | - Chen Wang
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| |
Collapse
|
54
|
Abstract
Antiretroviral therapy (ART) potently suppresses HIV-1 replication, but the virus persists in quiescent infected CD4(+)T cells as a latent integrated provirus, and patients must indefinitely remain on therapy. If ART is terminated, these integrated proviruses can reactivate, driving new rounds of infection. A functional cure for HIV requires eliminating low-level ongoing viral replication that persists in certain tissue sanctuaries and preventing viral reactivation. The HIV Tat protein plays an essential role in HIV transcription by recruiting the kinase activity of the P-TEFb complex to the viral mRNA's stem-bulge-loop structure, TAR, activating transcriptional elongation. Because the Tat-mediated transactivation cascade is critical for robust HIV replication, the Tat/TAR/P-TEFb complex is one of the most attractive targets for drug development. Importantly, compounds that interfere with transcription could impair viral reactivation, low-level ongoing replication, and replenishment of the latent reservoir, thereby reducing the size of the latent reservoir pool. Here, we discuss the potential importance of transcriptional inhibitors in the treatment of latent HIV-1 disease and review recent findings on targeting Tat, TAR, and P-TEFb individually or as part of a complex. Finally, we discuss the impact of extracellular Tat in HIV-associated neurocognitive disorders and cancers.
Collapse
|
55
|
Bouwman RD, Palser A, Parry CM, Coulter E, Rasaiyaah J, Kellam P, Jenner RG. Human immunodeficiency virus Tat associates with a specific set of cellular RNAs. Retrovirology 2014; 11:53. [PMID: 24990269 PMCID: PMC4086691 DOI: 10.1186/1742-4690-11-53] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2013] [Accepted: 06/18/2014] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Human Immunodeficiency Virus 1 (HIV-1) exhibits a wide range of interactions with the host cell but whether viral proteins interact with cellular RNA is not clear. A candidate interacting factor is the trans-activator of transcription (Tat) protein. Tat is required for expression of virus genes but activates transcription through an unusual mechanism; binding to an RNA stem-loop, the transactivation response element (TAR), with the host elongation factor P-TEFb. HIV-1 Tat has also been shown to alter the expression of host genes during infection, contributing to viral pathogenesis but, whether Tat also interacts with cellular RNAs is unknown. RESULTS Using RNA immunoprecipitation coupled with microarray analysis, we have discovered that HIV-1 Tat is associated with a specific set of human mRNAs in T cells. mRNAs bound by Tat share a stem-loop structural element and encode proteins with common biological roles. In contrast, we do not find evidence that Tat associates with microRNAs or the RNA-induced silencing complex (RISC). The interaction of Tat with cellular RNA requires an intact RNA binding domain and Tat RNA binding is linked to an increase in RNA abundance in cell lines and during infection of primary CD4+ T cells by HIV. CONCLUSIONS We conclude that Tat interacts with a specific set of human mRNAs in T cells, many of which show changes in abundance in response to Tat and HIV infection. This work uncovers a previously unrecognised interaction between HIV and its host that may contribute to viral alteration of the host cellular environment.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Richard G Jenner
- MRC Centre for Medical Molecular Virology, Division of Infection and Immunity, University College London, London WC1E 6BT, UK.
| |
Collapse
|
56
|
HIV-1 Tat protein induces PD-L1 (B7-H1) expression on dendritic cells through tumor necrosis factor alpha- and toll-like receptor 4-mediated mechanisms. J Virol 2014; 88:6672-89. [PMID: 24696476 DOI: 10.1128/jvi.00825-14] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
UNLABELLED Chronic human immunodeficiency virus type 1 (HIV-1) infection is associated with induction of T-cell coinhibitory pathways. However, the mechanisms by which HIV-1 induces upregulation of coinhibitory molecules remain to be fully elucidated. The aim of the present study was to determine whether and how HIV-1 Tat protein, an immunosuppressive viral factor, induces the PD-1/PD-L1 coinhibitory pathway on human dendritic cells (DCs). We found that treatment of DCs with whole HIV-1 Tat protein significantly upregulated the level of expression of PD-L1. This PD-L1 upregulation was observed in monocyte-derived dendritic cells (MoDCs) obtained from either uninfected or HIV-1-infected patients as well as in primary myeloid DCs from HIV-negative donors. In contrast, no effect on the expression of PD-L2 or PD-1 molecules was detected. The induction of PD-L1 on MoDCs by HIV-1 Tat (i) occurred in dose- and time-dependent manners, (ii) was mediated by the N-terminal 1-45 fragment of Tat, (iii) did not require direct cell-cell contact but appeared rather to be mediated by soluble factor(s), (iv) was abrogated following neutralization of tumor necrosis factor alpha (TNF-α) or blocking of Toll-like receptor 4 (TLR4), (v) was absent in TLR4-knockoout (KO) mice but could be restored following incubation with Tat-conditioned medium from wild-type DCs, (vi) impaired the capacity of MoDCs to functionally stimulate T cells, and (vii) was not reversed functionally following PD-1/PD-L1 pathway blockade, suggesting the implication of other Tat-mediated coinhibitory pathways. Our results demonstrate that HIV-1 Tat protein upregulates PD-L1 expression on MoDCs through TNF-α- and TLR4-mediated mechanisms, functionally compromising the ability of DCs to stimulate T cells. The findings offer a novel potential molecular target for the development of an anti-HIV-1 treatment. IMPORTANCE The objective of this study was to investigate the effect of human immunodeficiency virus type 1 (HIV-1) Tat on the PD-1/PD-L1 coinhibitory pathway on human monocyte-derived dendritic cells (MoDCs). We found that treatment of MoDCs from either healthy or HIV-1-infected patients with HIV-1 Tat protein stimulated the expression of PD-L1. We demonstrate that this stimulation was mediated through an indirect mechanism, involving tumor necrosis factor alpha (TNF-α) and Toll-like receptor 4 (TLR4) pathways, and resulted in compromised ability of Tat-treated MoDCs to functionally stimulate T-cell proliferation.
Collapse
|
57
|
Dalvi P, Wang K, Mermis J, Zeng R, Sanderson M, Johnson S, Dai Y, Sharma G, Ladner AO, Dhillon NK. HIV-1/cocaine induced oxidative stress disrupts tight junction protein-1 in human pulmonary microvascular endothelial cells: role of Ras/ERK1/2 pathway. PLoS One 2014; 9:e85246. [PMID: 24409324 PMCID: PMC3883699 DOI: 10.1371/journal.pone.0085246] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2013] [Accepted: 11/25/2013] [Indexed: 01/08/2023] Open
Abstract
Intravenous drug use (IVDU) is the major risk factor in the development of HIV-related pulmonary arterial hypertension (HRPAH); however, the pathogenesis of HRPAH in association with IVDU has yet to be characterized. Endothelial injury is considered to be an initiating factor for pulmonary vascular remodeling in animal models of PAH. Our previous study shows that simultaneous exposure to HIV-Trans-activator of transcription (Tat) and cocaine exacerbates both disruption of tight junction proteins and permeability of human pulmonary artery endothelial cells compared with either treatment alone. We here now demonstrate that this HIV-Tat and cocaine mediated endothelial dysfunction accompanies with increase in hydrogen peroxide and superoxide radicals generation and involves redox sensitive signaling pathway. Pretreatment with antioxidant cocktail attenuated the cocaine and Tat mediated disassembly of Zonula Occludens (ZO)-1 and enhancement of endothelial monolayer permeability. Furthermore, inhibition of NADPH oxidase by apocynin or siRNA-mediated knockdown of gp-91(phox) abolished the Tat/cocaine-induced reactive oxygen species (ROS) production, suggesting the NADPH oxidase mediated generation of oxidative radicals. In addition, ROS dependent activation of Ras and ERK1/2 Kinase was observed to be mediating the TJP-1 disassembly, and endothelial dysfunction in response to cocaine and Tat exposure. In conclusion, our findings demonstrate that Tat/cocaine -mediated production of ROS activate Ras/Raf/ERK1/2 pathway that contributes to disruption of tight junction protein leading to pulmonary endothelial dysfunction associated with pulmonary vascular remodeling.
Collapse
Affiliation(s)
- Pranjali Dalvi
- Division of Pulmonary and Critical Care Medicine, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Kun Wang
- Division of Pulmonary and Critical Care Medicine, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Joel Mermis
- Division of Pulmonary and Critical Care Medicine, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Ruoxi Zeng
- Division of Pulmonary and Critical Care Medicine, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Miles Sanderson
- Division of Pulmonary and Critical Care Medicine, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Sara Johnson
- Division of Pulmonary and Critical Care Medicine, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Yuqiao Dai
- Division of Pulmonary and Critical Care Medicine, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Garima Sharma
- Division of Pulmonary and Critical Care Medicine, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Amy O’Brien Ladner
- Division of Pulmonary and Critical Care Medicine, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Navneet K. Dhillon
- Division of Pulmonary and Critical Care Medicine, University of Kansas Medical Center, Kansas City, Kansas, United States of America
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas, United States of America
- * E-mail:
| |
Collapse
|
58
|
Kim J, Yoon JH, Kim YS. HIV-1 Tat interacts with and regulates the localization and processing of amyloid precursor protein. PLoS One 2013; 8:e77972. [PMID: 24312169 PMCID: PMC3843664 DOI: 10.1371/journal.pone.0077972] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2013] [Accepted: 09/07/2013] [Indexed: 12/22/2022] Open
Abstract
HIV-1 Tat protein plays various roles in virus proliferation and in the regulation of numerous host cell functions. Accumulating evidence suggests that HIV-1 Tat also plays an important role in HIV-associated neurocognitive disorders (HAND) by disrupting intracellular communication. Amyloid beta (Aβ) is generated from amyloid precursor protein (APP) and accumulates in the senile plaques of Alzheimer's disease patients. This study demonstrates that Tat interacts with APP both in vitro and in vivo, and increases the level of Aβ42 by recruiting APP into lipid rafts. Co-localization of Tat with APP in the cytosol was observed in U-87 MG cells that expressed high levels of Tat, and redistribution of APP into lipid rafts, a site of increased β- and γ-secretase activity, was demonstrated by discontinuous sucrose density gradient ultracentrifugation in the presence of Tat. Furthermore, Tat enhanced the cleavage of APP by β-secretase in vitro, resulting in 5.5-fold higher levels of Aβ42. This was consistent with increased levels of β-C-terminal fragment (β-CTF) and reduced levels of α-CTF. Moreover, stereotaxic injection of a lentiviral Tat expression construct into the hippocampus of APP/presenilin-1 (PS1) transgenic mice resulted in increased Tat-mediated production and processing of Aβ in vivo. Increased levels of Aβ42, as well as an increase in the number and size of Aβ plaques, were observed in the hippocampus following injection of Tat virus compared with mock virus. These results suggest that HIV-1 Tat may contribute to HAND by interacting with and modifying APP processing, thereby increasing Aβ production.
Collapse
Affiliation(s)
- Jiyoung Kim
- Indang Institute of Molecular Biology, Inje University, Jung-Gu, Seoul, Republic of Korea
| | - Jee-Hyun Yoon
- Department of Smart Foods and Drugs, Inje University, Jung-Gu, Seoul, Republic of Korea
| | - Yeon-Soo Kim
- Indang Institute of Molecular Biology, Inje University, Jung-Gu, Seoul, Republic of Korea
- Department of Smart Foods and Drugs, Inje University, Jung-Gu, Seoul, Republic of Korea
- * E-mail:
| |
Collapse
|
59
|
Ait Mebarek M, Wijkhuisen A, Adel-Patient K, Lamourette P, Léonetti M, Volland H. Production of human antibodies by in vitro immunization using a fusion protein containing the transcriptional transactivator of HIV-1. J Immunol Methods 2013; 396:96-106. [PMID: 23954270 DOI: 10.1016/j.jim.2013.07.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2012] [Revised: 07/29/2013] [Accepted: 07/29/2013] [Indexed: 11/16/2022]
Abstract
Antigen-specific activation of human B cells represents a key step for the production of monoclonal antibodies. Several approaches have been developed over the last thirty years in order to improve the process of lymphocyte activation in vitro. In the present study, we investigated whether the transcriptional transactivator (Tat) of human immunodeficiency virus, which possesses numerous biological activities, is able to trigger antibody secretion when incubated with human peripheral blood mononuclear cells. No such effect was observed when using Tat as a free protein. However, we found a significant IgM antibody production when Tat was previously fused to a double domain, called ZZ, derived from protein A of Staphylococcus aureus. The effect was also observed when the fusion protein, called ZZTat101, was incubated with purified B cells, indicating that the phenomenon does not require T-cell help. Antibody secretion was observed in the absence of cytokines that are usually used during in vitro immunization experiments, indicating that ZZTat101 provides the signals required for the initiation of the immune response. Antibody secretion was observed using a ZZTat mutant, containing only the Tat residues 22 to 57, called ZZTat22-57, indicating that this region is sufficient to initiate the immune response. In contrast, the effect was not found with a ZZTat22-57 mutant devoid of the seven Tat cysteines located between residues 22 and 37, demonstrating that these residues play a crucial role in the phenomenon. Our results pave the way to the development of a new in vitro immunization method based on antigens associated with ZZTat.
Collapse
Affiliation(s)
- M Ait Mebarek
- CEA, Institut de Biologie et de Technologie de Saclay (iBiTec-S), Service de Pharmacologie et d'immunoanalyse (SPI), 91191 Gif sur Yvette, France
| | | | | | | | | | | |
Collapse
|
60
|
Abstract
Since the initial hypotheses on the importance of angiogenesis in the pathogenesis of cancer approximately 30 years ago, there have been major advances in the understanding of the cellular and molecular mechanisms involved in the regulation of this complex process of new vessel formation. Among the multitude of factors, vascular endothelial growth factor (VEGF) has emerged as one of the most potent angiogenic factors, being implicated in the initiation of signal transduction responsible for cell proliferation, survival, migration and adhesion. Inhibition of VEGF and its signaling pathway offers a potential new molecular target in cancer therapy. This article reviews the role of angiogenesis and its mediators, particularly vascular endothelial growth factors, in hematological malignancies, as well as the potential use of anti-angiogenic therapies in the management of these conditions.
Collapse
Affiliation(s)
- Soon Thye Lim
- Keck School of Medicine/Norris, university of Southern california, Comprehensive Cancer center, Los Angeles, CA 90033, USA
| | | |
Collapse
|
61
|
Induction of IL-17 and nonclassical T-cell activation by HIV-Tat protein. Proc Natl Acad Sci U S A 2013; 110:13588-93. [PMID: 23898208 DOI: 10.1073/pnas.1308673110] [Citation(s) in RCA: 202] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Chronic immune activation is a major complication of antiretroviral therapy (ART) for HIV infection and can cause a devastating immune reconstitution inflammatory syndrome (IRIS) in the brain. The mechanism of T-cell activation in this population is not well understood. We found HIV-Tat protein and IL-17-expressing mononuclear cells in the brain of an individual with IRIS. Tat was also present in the CSF of individuals virologically controlled on ART. Hence we examined if Tat protein could directly activate T cells. Tat transcriptionally dysregulated 94 genes and induced secretion of 11 cytokines particularly activation of IL-17 signaling pathways supporting the development of a proinflammatory state. Tat increased IL-17 transcription and secretion in T cells. Tat entered the T cells rapidly by clathrin-mediated endocytosis and localized to both the cytoplasm and the nucleus. Tat activated T cells through a nonclassical pathway dependent upon vascular endothelial growth factor receptor-2 and downstream secondary signaling pathways but independent of the T-cell receptor. However, Tat stimulation of T cells did not induce T-cell proliferation but increased viral infectivity. This study demonstrates Tat's role as a virulence factor, by driving T-cell activation and contributing to IRIS pathophysiology. This supports the necessity of an anti-Tat therapy in conjunction with ART and identifies multiple targetable pathways to prevent Tat-mediated T-cell activation.
Collapse
|
62
|
Del Guerra FB, Fonseca JLI, Figueiredo VM, Ziff EB, Konkiewitz EC. Human immunodeficiency virus-associated depression: contributions of immuno-inflammatory, monoaminergic, neurodegenerative, and neurotrophic pathways. J Neurovirol 2013; 19:314-27. [PMID: 23868513 DOI: 10.1007/s13365-013-0177-7] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2013] [Revised: 05/24/2013] [Accepted: 06/05/2013] [Indexed: 12/14/2022]
Abstract
In the era of greatly improved pharmacological treatment of HIV infection through highly active antiretroviral therapy (HAART), HIV patients experience reduced viral loads, reduced opportunistic infections, increased CD4+ T cell count, and greater life expectancy. Although life expectancy is increased, patients often develop neurological disturbances that may persist for long periods, seriously jeopardizing quality of life and adherence to the medication protocols of HAART. For these reasons, HIV-associated neurological disorders have gained importance in both clinical and basic investigations of HIV infection. Depression is the most prevalent neuropsychiatric disorder among people living with HIV. In this review, we discuss how HIV can predispose infected individuals to depression by several interrelated mechanisms. These include inducing chronic elevation of cytokines through activation of microglia and astrocytes; decreasing monoaminergic function; inducing neurotoxicity, especially in dopaminergic neurons; and reducing brain-derived neurotrophic factor. These viral pathways interact with psychosocial factors to create the depressive state. HIV depression has a great impact on quality of life and implementation of antiretroviral therapy, and thus, recognition of these modes of action is significant for understanding HIV neuropathology and for selecting modalities for pharmacologic treatment.
Collapse
Affiliation(s)
- F B Del Guerra
- Faculdade de Ciências da Saude, Universidade Federal da Grande Dourados Unidade II, Rodovia MS 162, Mato Grosso do Sul, Brazil
| | | | | | | | | |
Collapse
|
63
|
Risk of Cardiovascular Events and Blood Pressure Control in Hypertensive HIV-Infected Patients. J Acquir Immune Defic Syndr 2013; 62:396-404. [DOI: 10.1097/qai.0b013e3182847cd0] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
64
|
Yao H, Duan M, Yang L, Buch S. Nonmuscle myosin light-chain kinase mediates microglial migration induced by HIV Tat: involvement of β1 integrins. FASEB J 2013; 27:1532-48. [PMID: 23292072 DOI: 10.1096/fj.12-219600] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
One of the hallmark features of HIV-associated neurological disease is increased activation and migration of microglia. HIV transactivator of transcription (Tat) is released from infected cells and has the ability to recruit microglia. The purpose of this study was to investigate molecular mechanisms by which recombinant Tat₁₋₇₂, but not heated-inactive Tat₁₋₇₂,induces migration of rat primary microglia. Using primary microglia in Boyden chambers, we demonstrated the role of nonmuscle myosin light-chain kinase (nmMYLK) in Tat₁₋₇₂ (14.4 nM)-mediated increased microglial migration (up to 171.85%). These findings were validated using microglia isolated from wild-type (WT) or nmMYLK(-/-) mice in Dunn chamber assays. Tat₁₋₇₂-mediated activation of nmMYLK resulted in "inside-out" activation of β1 integrin, followed by "outside-in" activation of c-Src, Pyk2, and Cdc42-GTP (using G-LISA in primary and nmMYLK(-/-) microglia) and, subsequently, actin polymerization (flow cytometry and Western blot assays). In vivo corroboration of these findings demonstrated decreased migration of nmMYLK(-/-) microglia (2 × 10(5) cells transplanted into corpus callosum) compared with WT microglia toward microinjected Tat₁₋₇₂ (2 μg/mouse) in hippocampus. Up-regulation of nmMYLK in microglia was also detected in sections of basal ganglia from humans with HIV-encephalitis compared with uninfected controls. nmMYLK is thus critical for eliciting microglial migration during the innate immune response.
Collapse
Affiliation(s)
- Honghong Yao
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska 68198-5880, USA
| | | | | | | |
Collapse
|
65
|
The role of thiamine in HIV infection. Int J Infect Dis 2012; 17:e221-7. [PMID: 23274124 DOI: 10.1016/j.ijid.2012.11.019] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2012] [Revised: 11/21/2012] [Accepted: 11/24/2012] [Indexed: 12/15/2022] Open
Abstract
Patients infected with HIV have a high prevalence of thiamine deficiency. Genetic studies have provided the opportunity to determine which proteins link thiamine to HIV pathology, i.e., renin-angiotensin system, poly(ADP-ribosyl) polymerase 1, Sp1 promoter gene, transcription factor p53, apoptotic factor caspase 3, and glycogen synthetase kinase 3β. Thiamine also affects HIV through non-genomic factors, i.e., matrix metalloproteinase, vascular endothelial growth factor, heme oxygenase 1, the prostaglandins, cyclooxygenase 2, reactive oxygen species, and nitric oxide. In conclusion, thiamine may benefit HIV patients, but further investigation of the role of thiamine in HIV infection is needed.
Collapse
|
66
|
Zhen X, Wang X, Xie C, Wu W, Jiang X. Cellular uptake, antitumor response and tumor penetration of cisplatin-loaded milk protein nanoparticles. Biomaterials 2012; 34:1372-82. [PMID: 23158934 DOI: 10.1016/j.biomaterials.2012.10.061] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2012] [Accepted: 10/25/2012] [Indexed: 01/08/2023]
Abstract
The casein nanoparticles cross-linked by transglutaminase were prepared, and cisplatin (CDDP), as a model antitumor drug, was loaded into the casein nanoparticles. These nanoparticles were characterized by dynamic light scattering (DLS), transmission electron microscopy (TEM), and zeta potential. The uptake and penetration of nanoparticles in 2- and 3-dimensional SH-SY5Y cells were examined at 37 °C and 4 °C. The in vivo biodistribution of the nanoparticles was investigated using near-infrared fluorescence (NIRF) imaging and ion-coupled plasma mass spectrometry (ICP-MS). The antitumor effect of CDDP-loaded nanoparticles was evaluated on hepatic H22 tumor-bearing mice model via intravenous administration. It is found that the obtained nanoparticles showed spherical shape with the size of 257 nm, and drug loading content of 10%. These CDDP-loaded casein nanoparticles have the extraordinary capabilities to penetrate cell membrane barriers, target tumor and inhibit tumor growth. The tumor growth inhibition of CDDP-loaded nanoparticles is 1.5-fold higher than that of free CDDP. Further, the penetration examination of the CDDP-loaded casein nanoparticles in the tumor tissue demonstrated that the nanoparticles had the ability to penetrate the tumor and deliver CDDP to the tumor more deeply and affect the cells far from the vasculature.
Collapse
Affiliation(s)
- Xu Zhen
- Laboratory of Mesoscopic Chemistry and Department of Polymer Science & Engineering, College of Chemistry & Chemical Engineering, Nanjing University, Nanjing 210093, PR China
| | | | | | | | | |
Collapse
|
67
|
Gibellini D, Miserocchi A, Tazzari PL, Ricci F, Clò A, Morini S, Ponti C, Pasquinelli G, Bon I, Pagliaro P, Borderi M, Re MC. Analysis of the effects of HIV-1 Tat on the survival and differentiation of vessel wall-derived mesenchymal stem cells. J Cell Biochem 2012; 113:1132-41. [PMID: 22095559 DOI: 10.1002/jcb.23446] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
HIV infection is an independent risk factor for atherosclerosis development and cardiovascular damage. As vessel wall mesenchymal stem cells (MSCs) are involved in the regulation of vessel structure homeostasis, we investigated the role of Tat, a key factor in HIV replication and pathogenesis, in MSC survival and differentiation. The survival of subconfluent MSCs was impaired when Tat was added at high concentrations (200-1,000 ng/ml), whereas lower Tat concentrations (1-100 ng/ml) did not promote apoptosis. Tat enhanced the differentiation of MSC toward adipogenesis by the transcription and activity upregulation of PPARγ. This Tat-related modulation of adipogenesis was tackled by treatment with antagonists of Tat-specific receptors such as SU5416 and RGD Fc. In contrast, Tat inhibited the differentiation of MSCs to endothelial cells by downregulating the expression of VEGF-induced endothelial markers such as Flt-1, KDR, and vWF. The treatment of MSCs with Tat-derived peptides corresponding to the cysteine-rich, basic, and RGD domains indicated that these Tat regions are involved in the inhibition of endothelial marker expression. The Tat-related impairment of MSC survival and differentiation might play an important role in vessel damage and formation of the atherosclerotic lesions observed in HIV-infected patients.
Collapse
Affiliation(s)
- Davide Gibellini
- Microbiology Section, Department of Haematology and Oncological Sciences, University of Bologna, Bologna, Italy.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
68
|
Deeken JF, Tjen-A-Looi A, Rudek MA, Okuliar C, Young M, Little RF, Dezube BJ. The rising challenge of non-AIDS-defining cancers in HIV-infected patients. Clin Infect Dis 2012; 55:1228-35. [PMID: 22776851 DOI: 10.1093/cid/cis613] [Citation(s) in RCA: 157] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Since the advent of HAART, patients with HIV infection have seen a significant improvement in their morbidity, mortality, and life expectancy. The incidence of AIDS-defining illnesses, including AIDS-defining malignancies, has been on the decline. However, deaths due to non-AIDS-defining illnesses have been on the rise. These so-called non-AIDS-defining cancers (NADCs) include cancers of the lung, liver, kidney, anus, head and neck, and skin, as well as Hodgkin's lymphoma. It is poorly understood why this higher rate of NADCs is occurring. The key challenge facing oncologists is how to administer chemotherapy effectively and safely to patients on antiretroviral therapy. The challenge to clinicians caring for HIV-infected patients is to develop and implement effective means to screen, treat, and prevent NADCs in the future. This review presents data on the epidemiology and etiology of NADCs, as well as ongoing research into this evolving aspect of the HIV epidemic.
Collapse
Affiliation(s)
- John F Deeken
- Division of Hematology/Oncology, Georgetown University Medical Center, 3800 Reservoir Rd NW, Washington, DC 20007, USA.
| | | | | | | | | | | | | |
Collapse
|
69
|
Spikes L, Dalvi P, Tawfik O, Gu H, Voelkel NF, Cheney P, O’Brien-Ladner A, Dhillon NK. Enhanced pulmonary arteriopathy in simian immunodeficiency virus-infected macaques exposed to morphine. Am J Respir Crit Care Med 2012; 185:1235-43. [PMID: 22447963 PMCID: PMC3373069 DOI: 10.1164/rccm.201110-1909oc] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2011] [Accepted: 03/08/2012] [Indexed: 11/16/2022] Open
Abstract
RATIONALE HIV-associated pulmonary arterial hypertension (PAH) is likely a more prevalent noninfectious complication of AIDS than previously recognized. Furthermore, the majority of HIV-PAH cases occur in individuals with a history of intravenous drug use. In this study we used a simian immunodeficiency (SIV) macaque model and a primary cell-culture system to investigate the association between drug abuse and HIV infection in HIV-PAH development. METHODS The archival lung tissues from macaques previously used to study the effect of morphine on SIV infection-associated neuropathogenesis were analyzed for pulmonary vascular changes. The direct effect of HIV proteins and illicit drugs was investigated on oxidative stress, survival, and proliferation of human pulmonary microvascular endothelial cells. MEASUREMENTS AND MAIN RESULTS SIVmacR71/17E-infected rhesus macaques treated with morphine (VM group) demonstrated significant pulmonary vascular remodeling, including the presence of early and advanced complex (plexiform) lesions, when compared with either the SIV-infected (V group) or morphine-treated uninfected (M group) macaques. However, both the V (two of five) and VM (two of six) groups included some animals with Pneumocystis jirovecii pneumonia. The endothelial cells lining the vessels with medial hypertrophy or initial-stage intimal lesions in lung sections from VM macaques demonstrated an increase in positivity for both terminal dUTP nick-end labeling and Ki67. Oxidative stress-mediated enhanced apoptosis followed by enhanced proliferation of endothelial cells was observed on simultaneous treatment with viral proteins and drugs of abuse compared with either treatment alone. CONCLUSIONS Our findings suggest that SIV/HIV protein(s) and morphine interact to cause the proliferation of apoptosis-resistant endothelial cells leading to angio-obliteration.
Collapse
Affiliation(s)
- Leslie Spikes
- Division of Pulmonary and Critical Care Medicine, Department of Medicine
| | - Pranjali Dalvi
- Division of Pulmonary and Critical Care Medicine, Department of Medicine
| | - Ossama Tawfik
- Department of Pathology and Laboratory Medicine, and
| | - Haihua Gu
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas; and
| | | | - Paul Cheney
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas; and
| | - Amy O’Brien-Ladner
- Division of Pulmonary and Critical Care Medicine, Department of Medicine
| | - Navneet K. Dhillon
- Division of Pulmonary and Critical Care Medicine, Department of Medicine
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas; and
| |
Collapse
|
70
|
Urbinati C, Ravelli C, Tanghetti E, Belleri M, Giacopuzzi E, Monti E, Presta M, Rusnati M. Substrate-Immobilized HIV-1 Tat Drives VEGFR2/αvβ3–Integrin Complex Formation and Polarization in Endothelial Cells. Arterioscler Thromb Vasc Biol 2012; 32:e25-34. [DOI: 10.1161/atvbaha.111.242396] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Affiliation(s)
- Chiara Urbinati
- From the Sections of General Pathology and Immunology (C.U., C.R., M.B., M.P., M.R.), Histology (E.T.), Biology and Genetics (E.G.), and Biochemistry (E.M.), Department of Biomedical Sciences and Biotechnology, University of Brescia
| | - Cosetta Ravelli
- From the Sections of General Pathology and Immunology (C.U., C.R., M.B., M.P., M.R.), Histology (E.T.), Biology and Genetics (E.G.), and Biochemistry (E.M.), Department of Biomedical Sciences and Biotechnology, University of Brescia
| | - Elena Tanghetti
- From the Sections of General Pathology and Immunology (C.U., C.R., M.B., M.P., M.R.), Histology (E.T.), Biology and Genetics (E.G.), and Biochemistry (E.M.), Department of Biomedical Sciences and Biotechnology, University of Brescia
| | - Mirella Belleri
- From the Sections of General Pathology and Immunology (C.U., C.R., M.B., M.P., M.R.), Histology (E.T.), Biology and Genetics (E.G.), and Biochemistry (E.M.), Department of Biomedical Sciences and Biotechnology, University of Brescia
| | - Edoardo Giacopuzzi
- From the Sections of General Pathology and Immunology (C.U., C.R., M.B., M.P., M.R.), Histology (E.T.), Biology and Genetics (E.G.), and Biochemistry (E.M.), Department of Biomedical Sciences and Biotechnology, University of Brescia
| | - Eugenio Monti
- From the Sections of General Pathology and Immunology (C.U., C.R., M.B., M.P., M.R.), Histology (E.T.), Biology and Genetics (E.G.), and Biochemistry (E.M.), Department of Biomedical Sciences and Biotechnology, University of Brescia
| | - Marco Presta
- From the Sections of General Pathology and Immunology (C.U., C.R., M.B., M.P., M.R.), Histology (E.T.), Biology and Genetics (E.G.), and Biochemistry (E.M.), Department of Biomedical Sciences and Biotechnology, University of Brescia
| | - Marco Rusnati
- From the Sections of General Pathology and Immunology (C.U., C.R., M.B., M.P., M.R.), Histology (E.T.), Biology and Genetics (E.G.), and Biochemistry (E.M.), Department of Biomedical Sciences and Biotechnology, University of Brescia
| |
Collapse
|
71
|
Chiodelli P, Urbinati C, Mitola S, Tanghetti E, Rusnati M. Sialic acid associated with αvβ3 integrin mediates HIV-1 Tat protein interaction and endothelial cell proangiogenic activation. J Biol Chem 2012; 287:20456-66. [PMID: 22528484 DOI: 10.1074/jbc.m111.337139] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Sialic acid (NeuAc) is a major anion on endothelial cells (ECs) that regulates different biological processes including angiogenesis. NeuAc is present in the oligosaccharidic portion of integrins, receptors that interact with extracellular matrix components and growth factors regulating cell adhesion, migration, and proliferation. Tat is a cationic polypeptide that, once released by HIV-1(+) cells, accumulates in the extracellular matrix, promoting EC adhesion and proangiogenic activation by engaging α(v)β(3). By using two complementary approaches (NeuAc removal by neuraminidase or its masking by NeuAc-binding lectin from Maackia amurensis, MAA), we investigated the presence of NeuAc on endothelial α(v)β(3) and its role in Tat interaction, EC adhesion, and proangiogenic activation. α(v)β(3) immunoprecipitation with biotinylated MAA or Western blot analysis of neuraminidase-treated ECs demonstrated that NeuAc is associated with both the α(v) and the β(3) subunits. Surface plasmon resonance analysis demonstrated that the masking of α(v)β(3)-associated NeuAc by MAA prevents Tat/α(v)β(3) interaction. MAA and neuraminidase prevent α(v)β(3)-dependent EC adhesion to Tat, the consequent FAK and ERK1/2 phosphorylation, and EC proliferation, migration, and regeneration in a wound-healing assay. Finally, MAA inhibits Tat-induced neovascularization in the ex vivo human artery ring sprouting assay. The inhibitions are specific because the NeuAc-unrelated lectin from Ulex europaeus is ineffective on Tat. Also, MAA and neuraminidase affect only weakly integrin-dependent EC adhesion and proangiogenic activation by fibronectin. In conclusion, NeuAc is associated with endothelial α(v)β(3) and mediates Tat-dependent EC adhesion and proangiogenic activation. These data point to the possibility to target integrin glycosylation for the treatment of angiogenesis/AIDS-associated pathologies.
Collapse
Affiliation(s)
- Paola Chiodelli
- Unit of General Pathology and Immunology, School of Medicine, University of Brescia, 25123 Brescia, Italy
| | | | | | | | | |
Collapse
|
72
|
Xue B, Mizianty MJ, Kurgan L, Uversky VN. Protein intrinsic disorder as a flexible armor and a weapon of HIV-1. Cell Mol Life Sci 2012; 69:1211-59. [PMID: 22033837 PMCID: PMC11114566 DOI: 10.1007/s00018-011-0859-3] [Citation(s) in RCA: 84] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2011] [Revised: 09/28/2011] [Accepted: 10/03/2011] [Indexed: 01/19/2023]
Abstract
Many proteins and protein regions are disordered in their native, biologically active states. These proteins/regions are abundant in different organisms and carry out important biological functions that complement the functional repertoire of ordered proteins. Viruses, with their highly compact genomes, small proteomes, and high adaptability for fast change in their biological and physical environment utilize many of the advantages of intrinsic disorder. In fact, viral proteins are generally rich in intrinsic disorder, and intrinsically disordered regions are commonly used by viruses to invade the host organisms, to hijack various host systems, and to help viruses in accommodation to their hostile habitats and to manage their economic usage of genetic material. In this review, we focus on the structural peculiarities of HIV-1 proteins, on the abundance of intrinsic disorder in viral proteins, and on the role of intrinsic disorder in their functions.
Collapse
Affiliation(s)
- Bin Xue
- Department of Molecular Medicine, University of South Florida, College of Medicine, 12901 Bruce B. Downs Blvd, MDC07, Tampa, FL 33612 USA
| | - Marcin J. Mizianty
- Department of Electrical and Computer Engineering, University of Alberta, Edmonton, AB T6G 2V4 Canada
| | - Lukasz Kurgan
- Department of Electrical and Computer Engineering, University of Alberta, Edmonton, AB T6G 2V4 Canada
| | - Vladimir N. Uversky
- Department of Molecular Medicine, University of South Florida, College of Medicine, 12901 Bruce B. Downs Blvd, MDC07, Tampa, FL 33612 USA
- Institute for Biological Instrumentation, Russian Academy of Sciences, 142290 Pushchino, Moscow Region Russia
| |
Collapse
|
73
|
Transfer of the ability of HIV-1 Tat to raise an adjuvant-free humoral immune response to unrelated antigens. Vaccine 2012; 30:2859-68. [DOI: 10.1016/j.vaccine.2012.02.042] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2011] [Revised: 01/24/2012] [Accepted: 02/17/2012] [Indexed: 02/03/2023]
|
74
|
Exploring transduction mechanisms of protein transduction domains (PTDs) in living cells utilizing single-quantum dot tracking (SQT) technology. SENSORS 2012; 12:549-72. [PMID: 22368485 PMCID: PMC3279229 DOI: 10.3390/s120100549] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2011] [Revised: 12/30/2011] [Accepted: 12/31/2011] [Indexed: 12/04/2022]
Abstract
Specific protein domains known as protein transduction domains (PTDs) can permeate cell membranes and deliver proteins or bioactive materials into living cells. Various approaches have been applied for improving their transduction efficacy. It is, therefore, crucial to clarify the entry mechanisms and to identify the rate-limiting steps. Because of technical limitations for imaging PTD behavior on cells with conventional fluorescent-dyes, how PTDs enter the cells has been a topic of much debate. Utilizing quantum dots (QDs), we recently tracked the behavior of PTD that was derived from HIV-1 Tat (TatP) in living cells at the single-molecule level with 7-nm special precision. In this review article, we initially summarize the controversy on TatP entry mechanisms; thereafter, we will focus on our recent findings on single-TatP-QD tracking (SQT), to identify the major sequential steps of intracellular delivery in living cells and to discuss how SQT can easily provide direct information on TatP entry mechanisms. As a primer for SQT study, we also discuss the latest findings on single particle tracking of various molecules on the plasma membrane. Finally, we discuss the problems of QDs and the challenges for the future in utilizing currently available QD probes for SQT. In conclusion, direct identification of the rate-limiting steps of PTD entry with SQT should dramatically improve the methods for enhancing transduction efficiency.
Collapse
|
75
|
Zhang X, Yu J, Kuzontkoski PM, Zhu W, Li DY, Groopman JE. Slit2/Robo4 signaling modulates HIV-1 gp120-induced lymphatic hyperpermeability. PLoS Pathog 2012; 8:e1002461. [PMID: 22241990 PMCID: PMC3252370 DOI: 10.1371/journal.ppat.1002461] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2011] [Accepted: 11/11/2011] [Indexed: 11/25/2022] Open
Abstract
Dissemination of HIV in the host involves transit of the virus and virus-infected cells across the lymphatic endothelium. HIV may alter lymphatic endothelial permeability to foster dissemination, but the mechanism is largely unexplored. Using a primary human lymphatic endothelial cell model, we found that HIV-1 envelope protein gp120 induced lymphatic hyperpermeability by disturbing the normal function of Robo4, a novel regulator of endothelial permeability. HIV-1 gp120 induced fibronectin expression and integrin α₅β₁ phosphorylation, which led to the complexing of these three proteins, and their subsequent interaction with Robo4 through its fibronectin type III repeats. Moreover, pretreatment with an active N-terminus fragment of Slit2, a Robo4 agonist, protected lymphatic endothelial cells from HIV-1 gp120-induced hyperpermeability by inhibiting c-Src kinase activation. Our results indicate that targeting Slit2/Robo4 signaling may protect the integrity of the lymphatic barrier and limit the dissemination of HIV in the host.
Collapse
Affiliation(s)
- Xuefeng Zhang
- Division of Experimental Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Jinlong Yu
- Division of Experimental Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Paula M. Kuzontkoski
- Division of Experimental Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Weiquan Zhu
- Department of Medicine and Molecular Medicine Program, University of Utah, Salt Lake City, Utah, United States of America
| | - Dean Y. Li
- Department of Medicine and Molecular Medicine Program, University of Utah, Salt Lake City, Utah, United States of America
| | - Jerome E. Groopman
- Division of Experimental Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America
| |
Collapse
|
76
|
Fiume G, Vecchio E, De Laurentiis A, Trimboli F, Palmieri C, Pisano A, Falcone C, Pontoriero M, Rossi A, Scialdone A, Fasanella Masci F, Scala G, Quinto I. Human immunodeficiency virus-1 Tat activates NF-κB via physical interaction with IκB-α and p65. Nucleic Acids Res 2011; 40:3548-62. [PMID: 22187158 PMCID: PMC3333881 DOI: 10.1093/nar/gkr1224] [Citation(s) in RCA: 115] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Nuclear factor (NF)-κB is a master regulator of pro-inflammatory genes and is upregulated in human immunodeficiency virus 1 (HIV-1) infection. Mechanisms underlying the NF-κB deregulation by HIV-1 are relevant for immune dysfunction in AIDS. We report that in single round HIV-1 infection, or single-pulse PMA stimulation, the HIV-1 Tat transactivator activated NF-κB by hijacking the inhibitor IκB-α and by preventing the repressor binding to the NF-κB complex. Moreover, Tat associated with the p65 subunit of NF-κB and increased the p65 DNA-binding affinity and transcriptional activity. The arginine- and cysteine-rich domains of Tat were required for IκB-α and p65 association, respectively, and for sustaining the NF-κB activity. Among an array of NF-κB-responsive genes, Tat mostly activated the MIP-1α expression in a p65-dependent manner, and bound to the MIP-1α NF-κB enhancer thus promoting the recruitment of p65 with displacement of IκB-α; similar findings were obtained for the NF-κB-responsive genes CSF3, LTA, NFKBIA and TLR2. Our results support a novel mechanism of NF-κB activation via physical interaction of Tat with IκB-α and p65, and may contribute to further insights into the deregulation of the inflammatory response by HIV-1.
Collapse
Affiliation(s)
- Giuseppe Fiume
- Department of Experimental and Clinical Medicine, University of Catanzaro Magna Graecia, Viale Europa-Germaneto, 88100 Catanzaro, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
77
|
Pathogenesis of HIV-associated pulmonary hypertension: potential role of HIV-1 Nef. Ann Am Thorac Soc 2011; 8:308-12. [PMID: 21653533 DOI: 10.1513/pats.201006-046wr] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Infection with HIV increases the risk for lung diseases, including noninfectious pulmonary hypertension (PH). HIV-associated PH (HIV-PH) is an important lung disease in HIV-infected persons who live longer with antiretrovirals. The early stages of HIV-PH may be overlooked by healthcare providers due to nonspecific symptoms, including progressive dyspnea and nonproductive cough. HIV-PH may be detected via chest radiographs, CT scans, or electrocardiograms, but Doppler echocardiography is the most useful screening test to identify candidates for right heart catheterization. HIV-PH has a poor prognosis with high mortality; improved biomarkers to identify earlier stages of PH would benefit clinical care. The HIV-PH mechanism remains unknown, but HIV proteins such as Tat and Nef may play a role. HIV-1 Nef is a broad-spectrum adaptor protein that may affect HIV-infected and uninfected pulmonary vascular cells. Studies in macaques suggest that Nef is important in HIV-PH pathogenesis because monkeys infected with a chimeric simian immunodeficiency virus (SIV) expressing HIV-nef (SHIVnef) alleles, but not monkeys infected with the native SIV, develop pulmonary vascular remodeling. Four consistent amino acid mutations arose spontaneously in Nef passaged in the monkeys. To translate these findings to humans, one research endeavor of the Lung HIV Study focuses on the identification of HIV nef mutations in HIV-infected individuals with PH compared with HIV-infected normotensive patients. We present some of the preliminary evidence. Ongoing longitudinal studies will establish the connection between Nef mutations and the propensity for HIV-PH.
Collapse
|
78
|
Mocchetti I, Bachis A, Avdoshina V. Neurotoxicity of human immunodeficiency virus-1: viral proteins and axonal transport. Neurotox Res 2011; 21:79-89. [PMID: 21948112 DOI: 10.1007/s12640-011-9279-2] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2011] [Revised: 09/09/2011] [Accepted: 09/19/2011] [Indexed: 12/13/2022]
Abstract
Human immunodeficiency virus-1 (HIV) infection of the central nervous system may cause a neurological syndrome termed HIV-associated neurocognitive disorder (HAND) which includes minor neurocognitive disorders or a more severe form of motor and cognitive impairments. Although treatment with highly active antiretroviral agents decreases the load of HIV in the brain, the prevalence of mild forms of HAND is actually increased due to longer life. Therefore, adjunctive and combined therapies must be developed to prevent and perhaps reverse the neurologic deficits observed in individuals with HAND. Key to developing effective therapies is a better understanding of the molecular and cellular mechanisms by which the virus causes this disorder. A number of HIV proteins has been shown to be released from HIV-infected cells. Moreover, these proteins have been shown to possess neurotoxic properties. This review describes new evidence of a direct interaction of the HIV protein gp120 with neurons, which might play a role in the etiopathology of HAND.
Collapse
Affiliation(s)
- Italo Mocchetti
- Department of Neuroscience, Georgetown University Medical Center, 3970 Reservoir Road, NW, New Research Building WP13, Washington, DC 20057, USA.
| | | | | |
Collapse
|
79
|
Vrancken K, Vervaeke P, Balzarini J, Liekens S. Viruses as key regulators of angiogenesis. Rev Med Virol 2011; 21:181-200. [PMID: 21538666 DOI: 10.1002/rmv.688] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Angiogenesis is an important physiological process that is controlled by a precise balance of growth and inhibitory factors in healthy tissues. However, environmental and genetic factors may disturb this delicate balance, resulting in the development of angiogenic diseases, tumour growth and metastasis. During the past decades, extensive research has led to the identification and characterization of genes, proteins and signalling pathways that are involved in neovascularization. Moreover, increasing evidence indicates that viruses may also regulate angiogenesis either directly, by (i) producing viral chemokines, growth factors and/or receptors or (ii) activating blood vessels as a consequence of endothelial cell tropism, or indirectly, by (iii) modulating the activity of cellular proteins and/or (iv) inducing a local or systemic inflammatory response, thereby creating an angiogenic microenvironment. As such, viruses may modulate several signal transduction pathways involved in angiogenesis leading to changes in endothelial cell proliferation, migration, adhesion, vascular permeability and/or protease production. Here, we will review different mechanisms that may be applied by viruses to deregulate the angiogenic balance in healthy tissues and/or increase the angiogenic potential of tumours.
Collapse
|
80
|
Ramos da Silva S, Elgui de Oliveira D. HIV, EBV and KSHV: Viral cooperation in the pathogenesis of human malignancies. Cancer Lett 2011; 305:175-85. [DOI: 10.1016/j.canlet.2011.02.007] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2010] [Accepted: 02/09/2011] [Indexed: 01/26/2023]
|
81
|
Cotter EJ, Chew N, Powderly WG, Doran PP. HIV type 1 alters mesenchymal stem cell differentiation potential and cell phenotype ex vivo. AIDS Res Hum Retroviruses 2011; 27:187-99. [PMID: 20929345 DOI: 10.1089/aid.2010.0114] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
An increased incidence of bone and lipid toxicities is associated with HIV-1 infection and its treatment. Mesenchymal stem cells (MSCs) are multipotent cells that can differentiate into both osteoblasts (OB) and adipocytes (AC). We hypothesize that the interaction of MSC and HIV-1 underlie these toxicities. Serum was collected from uninfected control and HIV-infected, antiviral-naive patients. Sera were divided into three groups: HIV-negative sera (n = 5), HIV-positive low viral load (LVL) (VL range 120; 4000, n = 5) or high viral load (HVL) (VL range 100,000; 500,000, n = 5). MSCs were exposed to these sera (5%) in an adipogenic/osteogenic condition and in nondifferentiating conditions in acute and chronic exposure models. Markers of adipogenesis/osteogenesis were examined in both MSCs induced to differentiated and nondifferentiating cells. Sera from HVL HIV-1-infected individuals induced a clear proadipogenic phenotype, as evidenced by an increase in adipocyte formation and the induction of increased expression of adipogenic markers including LPL and PPARγ. Both CD4 receptor blockade and treatment with the antiretroviral AZT attenuated these proadipogenic effects, suggesting that an infection event may underlie the observed phenomena. Finally, inhibition of COUP TF-1 by HIV-1 TAT was identified as a potential molecular mechanism for these effects. These results suggest that HIV-1 directly interacts with and may infect MSCs resulting in alterations of their differentiation potential, findings that significantly enhance our understanding of HIV-1-associated bone and fat toxicities.
Collapse
Affiliation(s)
- Eoin J. Cotter
- Clinical Research Center, University College Dublin, Dublin, Ireland
| | - Nicholas Chew
- Clinical Research Center, University College Dublin, Dublin, Ireland
| | - William G. Powderly
- School of Medicine and Medical Sciences, University College Dublin, Dublin, Ireland
| | - Peter P. Doran
- Clinical Research Center, University College Dublin, Dublin, Ireland
| |
Collapse
|
82
|
Imamura J, Suzuki Y, Gonda K, Roy CN, Gatanaga H, Ohuchi N, Higuchi H. Single particle tracking confirms that multivalent Tat protein transduction domain-induced heparan sulfate proteoglycan cross-linkage activates Rac1 for internalization. J Biol Chem 2011; 286:10581-92. [PMID: 21199870 DOI: 10.1074/jbc.m110.187450] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The mechanism by which HIV-1-Tat protein transduction domain (TatP) enters the cell remains unclear because of an insufficient understanding of the initial kinetics of peptide entry. Here, we report the successful visualization and tracking of TatP molecular kinetics on the cell surface with 7-nm spatial precision using quantum dots. Strong cell binding was only observed with a TatP valence of ≥8, whereas monovalent TatP binding was negligible. The requirement of the cell-surface heparan sulfate (HS) chains of HS proteoglycans (HSPGs) for TatP binding and intracellular transport was demonstrated by the enzymatic removal of HS and simultaneous observation of two individual particles. Multivalent TatP induces HSPG cross-linking, recruiting activated Rac1 to adjacent lipid rafts and thereby enhancing the recruitment of TatP/HSPG to actin-associated microdomains and its internalization by macropinocytosis. These findings clarify the initial binding mechanism of TatP to the cell surface and demonstrate the importance of TatP valence for strong surface binding and signal transduction. Our data also shed light on the ability of TatP to exploit the machinery of living cells, using HSPG signaling to activate Rac1 and alter TatP mobility and internalization. This work should guide the future design of TatP-based peptides as therapeutic nanocarriers with efficient transduction.
Collapse
Affiliation(s)
- Junji Imamura
- Department of Emerging Infectious Diseases, Graduate School of Medicine, Tohoku University, Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan
| | | | | | | | | | | | | |
Collapse
|
83
|
Abstract
To cause infections, microbial pathogens elaborate a multitude of factors that interact with host components. Using these host–pathogen interactions to their advantage, pathogens attach, invade, disseminate, and evade host defense mechanisms to promote their survival in the hostile host environment. Many viruses, bacteria, and parasites express adhesins that bind to cell surface heparan sulfate proteoglycans (HSPGs) to facilitate their initial attachment and subsequent cellular entry. Some pathogens also secrete virulence factors that modify HSPG expression. HSPGs are ubiquitously expressed on the cell surface of adherent cells and in the extracellular matrix. HSPGs are composed of one or several heparan sulfate (HS) glycosaminoglycan chains attached covalently to specific core proteins. For most intracellular pathogens, cell surface HSPGs serve as a scaffold that facilitates the interaction of microbes with secondary receptors that mediate host cell entry. Consistent with this mechanism, addition of HS or its pharmaceutical functional mimic, heparin, inhibits microbial attachment and entry into cultured host cells, and HS-binding pathogens can no longer attach or enter cultured host cells whose HS expression has been reduced by enzymatic treatment or chemical mutagenesis. In pathogens where the specific HS adhesin has been identified, mutant strains lacking HS adhesins are viable and show normal growth rates, suggesting that the capacity to interact with HSPGs is strictly a virulence activity. The goal of this chapter is to provide a mechanistic overview of our current understanding of how certain microbial pathogens subvert HSPGs to promote their infection, using specific HSPG–pathogen interactions as representative examples.
Collapse
Affiliation(s)
- Mauro S.G. Pavão
- , Institute of Medical Biochemistry, Federal University of Rio de Janeiro, Av. Prof. Rodolpho Paulo Rocco 255, Rio de Janeiro, 21941-913 Rio de Janeiro Brazil
| |
Collapse
|
84
|
Abstract
HIV-1 crosses the blood-brain barrier (BBB) early in the course of systemic infection and resides in brain macrophages and microglia. The integrity of the brain endothelium is regulated by intercellular tight junctions, which also play a critical role in HIV-1-entry into the brain. Disruption of tight junctions, including changes in claudin-5 expression, is common in HIV-1-infected patients. Recent evidence indicates that both exposure to HIV-1 and HIV-1 specific proteins, such as Tat protein, can contribute to alterations of expression and distribution of claudin-5 in brain endothelial cells and brain microvessels.
Collapse
|
85
|
Vascular endothelial growth factor receptor 1 contributes to Escherichia coli K1 invasion of human brain microvascular endothelial cells through the phosphatidylinositol 3-kinase/Akt signaling pathway. Infect Immun 2010; 78:4809-16. [PMID: 20805333 DOI: 10.1128/iai.00377-10] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Escherichia coli is the most common Gram-negative organism causing neonatal meningitis. Previous studies demonstrated that E. coli K1 invasion of brain microvascular endothelial cells (BMEC) is required for penetration into the central nervous system, but the microbe-host interactions that are involved in this process remain incompletely understood. Here we report the involvement of vascular endothelial growth factor receptor 1 (VEGFR1) expressed on human brain microvascular endothelial cells (HBMEC) in E. coli K1 invasion of HBMEC. Our results showed that treatment of confluent HBMEC with pan-VEGFR inhibitors significantly inhibited E. coli K1 invasion of HBMEC. Immunofluorescence results indicated the colocalization of VEGFR1 with E. coli K1 during bacterial invasion of HBMEC. The E. coli-induced actin cytoskeleton rearrangements in HBMEC were blocked by VEGFR inhibitors but not by VEGFR2-specific inhibitors. The small interfering RNA (siRNA) knockdown of VEGFR1 in HBMEC significantly attenuated E. coli invasion and the concomitant actin filament rearrangement. Furthermore, we found an increased association of VEGFR1 with the p85 subunit of phosphatidylinositol 3-kinase (PI3K) in HBMEC infected with E. coli K1 and that E. coli K1-triggered Akt activation in HBMEC was blocked by VEGFR1 siRNA and VEGFR inhibitors. Taken together, our results demonstrate that VEGFR1 contributes to E. coli K1 invasion of HBMEC via recruitment of the PI3K/Akt signaling pathway.
Collapse
|
86
|
Akhtar LN, Qin H, Muldowney MT, Yanagisawa LL, Kutsch O, Clements JE, Benveniste EN. Suppressor of cytokine signaling 3 inhibits antiviral IFN-beta signaling to enhance HIV-1 replication in macrophages. THE JOURNAL OF IMMUNOLOGY 2010; 185:2393-404. [PMID: 20631305 DOI: 10.4049/jimmunol.0903563] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
HIV-1 replication within macrophages of the CNS often results in cognitive and motor impairment, which is known as HIV-associated dementia (HAD) in its most severe form. IFN-beta suppresses viral replication within these cells during early CNS infection, but the effect is transient. HIV-1 eventually overcomes this protective innate immune response to resume replication through an unknown mechanism, initiating the progression toward HAD. In this article, we show that Suppressor of Cytokine Signaling (SOCS)3, a molecular inhibitor of IFN signaling, may allow HIV-1 to evade innate immunity within the CNS. We found that SOCS3 is elevated in an in vivo SIV/macaque model of HAD and that the pattern of expression correlates with recurrence of viral replication and onset of CNS disease. In vitro, the HIV-1 regulatory protein transactivator of transcription induces SOCS3 in human and murine macrophages in a NF-kappaB-dependent manner. SOCS3 expression attenuates the response of macrophages to IFN-beta at proximal levels of pathway activation and downstream antiviral gene expression and consequently overcomes the inhibitory effect of IFN-beta on HIV-1 replication. These studies indicate that SOCS3 expression, induced by stimuli present in the HIV-1-infected brain, such as transactivator of transcription, inhibits antiviral IFN-beta signaling to enhance HIV-1 replication in macrophages. This consequence of SOCS3 expression in vitro, supported by a correlation with increased viral load and onset of CNS disease in vivo, suggests that SOCS3 may allow HIV-1 to evade the protective innate immune response within the CNS, allowing the recurrence of viral replication and, ultimately, promoting progression toward HAD.
Collapse
|
87
|
Nitzsche B, Gloesenkamp C, Schrader M, Ocker M, Preissner R, Lein M, Zakrzewicz A, Hoffmann B, Höpfner M. Novel compounds with antiangiogenic and antiproliferative potency for growth control of testicular germ cell tumours. Br J Cancer 2010; 103:18-28. [PMID: 20551952 PMCID: PMC2905278 DOI: 10.1038/sj.bjc.6605725] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2010] [Revised: 05/11/2010] [Accepted: 05/14/2010] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Testicular germ cell tumour (TGCT) is the most common cause of death from solid tumours in young men and especially for platinum-refractory patients novel treatment approaches are urgently needed. Using an in silico screening approach for the detection of novel cancer drugs with inhibitory effects on the tyrosine kinase activity of growth factors (e.g., VEGFR, PDGFR), we identified two compounds (HP-2 and HP-14) with antiangiogenic and antiproliferative potency, which were evaluated in endothelial cell models and TGCT cells. RESULTS HP-2 and HP-14 effectively inhibited the growth of VEGFR-2-expressing TGCT cell lines (Tera-1, Tera-2 and 2102EP) and endothelial cell models, while they failed to supress the growth of VEGFR-2-lacking tumour cells. cDNA-microarrays revealed an inhibition of the expression of several growth factor receptors and related signal transduction molecules. Vascular endothelial growth factor (VEGF)-induced cell migration was also potently inhibited. Cell cycle-regulating proteins such as p21 and p27 were upregulated, leading to an S-phase arrest. Additional in vivo evaluations confirmed the antiangiogenic potency and good tolerability of the novel substances. CONCLUSION Our data show that the identified novel compounds inhibit the growth of TGCT cells and decrease angiogenic microvessel formation. The mode of action involves cell cycle arresting effects and changes in the expression pattern of several angiogenic genes. The novel compounds may qualify as new candidates for targeted treatment of TGCT and merit further evaluation.
Collapse
Affiliation(s)
- B Nitzsche
- Department of Physiology, Charité – Universitätsmedizin Berlin, Campus Benjamin Franklin, Arnimallee 22, Berlin 14195, Germany
| | - C Gloesenkamp
- Department of Physiology, Charité – Universitätsmedizin Berlin, Campus Benjamin Franklin, Arnimallee 22, Berlin 14195, Germany
| | - M Schrader
- Department of Urology, University of Ulm, Prittwitzstraße 43, 89075 Ulm, Germany
| | - M Ocker
- Institute for Surgical Research, Philipps University Marburg, Baldingerstrasse, 35043 Marburg, Germany
| | - R Preissner
- Department of Physiology, Charité – Universitätsmedizin Berlin, Campus Benjamin Franklin, Arnimallee 22, Berlin 14195, Germany
| | - M Lein
- Berlin Institute for Urologic Research, Charité –Universitätsmedizin Berlin, Robert Koch -Platz 7, Berlin 10115, Germany
| | - A Zakrzewicz
- Department of Physiology, Charité – Universitätsmedizin Berlin, Campus Benjamin Franklin, Arnimallee 22, Berlin 14195, Germany
| | - B Hoffmann
- Department of Physiology, Charité – Universitätsmedizin Berlin, Campus Benjamin Franklin, Arnimallee 22, Berlin 14195, Germany
| | - M Höpfner
- Department of Physiology, Charité – Universitätsmedizin Berlin, Campus Benjamin Franklin, Arnimallee 22, Berlin 14195, Germany
| |
Collapse
|
88
|
Foucault M, Mayol K, Receveur-Bréchot V, Bussat MC, Klinguer-Hamour C, Verrier B, Beck A, Haser R, Gouet P, Guillon C. UV and X-ray structural studies of a 101-residue long Tat protein from a HIV-1 primary isolate and of its mutated, detoxified, vaccine candidate. Proteins 2010; 78:1441-56. [PMID: 20034112 DOI: 10.1002/prot.22661] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The 101-residue long Tat protein of primary isolate 133 of the human immunodeficiency virus type 1 (HIV-1), wt-Tat(133) displays a high transactivation activity in vitro, whereas the mutant thereof, STLA-Tat(133), a vaccine candidate for HIV-1, has none. These two proteins were chemically synthesized and their biological activity was validated. Their structural properties were characterized using circular dichroism (CD), fluorescence emission, gel filtration, dynamic light scattering, and small angle X-ray scattering (SAXS) techniques. SAXS studies revealed that both proteins were extended and belong to the family of intrinsically unstructured proteins. CD measurements showed that wt-Tat(133) or STLA-Tat(133) underwent limited structural rearrangements when complexed with specific fragments of antibodies. Crystallization trials have been performed on the two forms, assuming that the Tat(133) proteins might have a better propensity to fold in supersaturated conditions, and small crystals have been obtained. These results suggest that biologically active Tat protein is natively unfolded and requires only a limited gain of structure for its function.
Collapse
|
89
|
Yang Y, Wu J, Lu Y. Mechanism of HIV-1-TAT induction of interleukin-1beta from human monocytes: Involvement of the phospholipase C/protein kinase C signaling cascade. J Med Virol 2010; 82:735-46. [PMID: 20336759 PMCID: PMC2849807 DOI: 10.1002/jmv.21720] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Human immunodeficiency virus TAT plays an important role in the disregulation of cytokine production associated with the neurological disorders that follow HIV infection. IL-1beta is one of the important inflammatory cytokines secreted by immune-activated monocytes/macrophages. Previous reports have shown that extracellular TAT stimulates IL-1beta expression in monocytes/macrophages. However, little is known about the mechanisms and possible TAT-responsive elements within the IL-1beta promoter. The present study shows that TAT increases the production of IL-1beta in human monocytes; PLC-PKC pathway-dependent phosphorylation of p44/42 and JNK MAP kinases participates partially in IL-1beta induction by TAT; specific C/EBP and NF-kappaB transcription factor binding elements within the IL-1beta promoter are involved in TAT regulation of IL-1beta production. This study identifies a signaling mechanism for HIV-1-induced IL-1beta production in human monocytes that may be involved in the neuropathogenesis of HIV-associated dementia.
Collapse
Affiliation(s)
- Yongbo Yang
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, 430072, China
- Department of Public Health Science, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI, 96822, U.S.A
| | - Jianguo Wu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, 430072, China
| | - Yuanan Lu
- Department of Public Health Science, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI, 96822, U.S.A
| |
Collapse
|
90
|
Bugatti A, Chiodelli P, Rosenbluh J, Loyter A, Rusnati M. BSA conjugates bearing multiple copies of the basic domain of HIV-1 Tat: Prototype for the development of multitarget inhibitors of extracellular Tat. Antiviral Res 2010; 87:30-9. [PMID: 20398703 DOI: 10.1016/j.antiviral.2010.04.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2009] [Revised: 03/02/2010] [Accepted: 04/06/2010] [Indexed: 11/19/2022]
Abstract
The transactivating factor (Tat) of HIV-1 is involved in AIDS progression and associated pathologies. Tat possesses a basic amino acid sequence implicated in heparan sulfate proteoglycan (HSPG)-mediated internalization, nuclear localization and transactivation by Tat and in the interaction of Tat with integrins and with the vascular endothelial growth factor receptor 2 (KDR) (kinase insert domain receptor). A BSA conjugate bearing an average of four copies of a peptide representing the basic domain/nuclear localization signal of Tat (BSA-Tat-NLS) inhibits transactivation by Tat exogenously added to cells but not by Tat endogenously produced after cell transfection with a tat cDNA, indicating that BSA-Tat-NLS does not interfere with Tat at an intracellular level. Surface plasmon resonance (SPR) experiments revealed that BSA-Tat-NLS binds to the HSPG analogue heparin. Accordingly, BSA-Tat-NLS binds to HSPGs of HL3T1 cell surface and inhibits HSPG-dependent Tat internalization. BSA-Tat-NLS retains its inhibitory potential when pre-incubated with HL3T1 cells before Tat administration, possibly by masking cell-surface HSPGs thus preventing Tat binding and internalization. SPR experiments revealed that BSA-Tat-NLS binds also to integrin alpha(v)beta(3) and KDR. Accordingly, it inhibits pro-angiogenic endothelial cell adhesion to Tat and motogenesis. In conclusion, BSA-Tat-NLS binds/masks three different cell-surface receptors of Tat inhibiting different biological activities. These data point to BSA-Tat-NLS as a prototype for the development of Tat-antagonists endowed with a multitargeted mechanism of action.
Collapse
Affiliation(s)
- Antonella Bugatti
- Department of Biomedical Science and Biotechnology, University of Brescia, Italy
| | | | | | | | | |
Collapse
|
91
|
Raborn ES, Cabral GA. Cannabinoid inhibition of macrophage migration to the trans-activating (Tat) protein of HIV-1 is linked to the CB(2) cannabinoid receptor. J Pharmacol Exp Ther 2010; 333:319-27. [PMID: 20089805 PMCID: PMC2846023 DOI: 10.1124/jpet.109.163055] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2009] [Accepted: 01/19/2010] [Indexed: 12/15/2022] Open
Abstract
Macrophages and macrophage-like cells are important targets of HIV-1 infection at peripheral sites and in the central nervous system. After infection, these cells secrete a plethora of toxic factors, including the viral regulatory trans-activating protein (Tat). This protein is highly immunogenic and also serves as a potent chemoattractant for monocytes. In the present study, the exogenous cannabinoids delta-9-tetrahydrocannabinol (THC) and (-)-cis-3-[2-hydroxy-4-(1,1-dimethylheptyl)phenyl]-trans-4-(3-hydroxypropyl) cyclohexanol (CP55940) were shown to significantly inhibit migration of human U937 macrophage-like cells to the Tat protein in a concentration-related manner. The CB(1) receptor-selective agonist N-(2-chloroethyl)-5Z,8Z,11Z,14Z-eicosatetraenamide (ACEA) had no effect on Tat-mediated migration. In contrast, the CB(2) receptor-selective agonist (1R,3R)-1-[4-(1,1-dimethylheptyl)-2,6-dimethoxyphenyl]-3-methylcyclohexanol (O-2137) exerted a concentration-related inhibition of U937 cell migration in response to Tat. Pharmacological blockage of CB(1) receptor signaling using the antagonist 5-(4-chlorophenyl)-1-(2,4-dichlorophenyl)-4-methyl-N-(1-piperidyl)pyrazole-3-carboxamide hydrochloride (SR141716A) had no effect on CP55940-mediated inhibition of macrophage migration to Tat, whereas treatment with the CB(2) receptor antagonist (1S-endo)-5-(4-chloro-3-methylphenyl)-1-((4-methylphenyl)methyl)-N-(1,3,3-trimethylbicyclo(2.2.1)hept-2-yl)-1H-pyrazole-3-carboxamide (SR144528) reversed the CP55940-mediated inhibition of migration. In addition, THC had no inhibitory effect on U937 migration to Tat after small interfering RNA knockdown of the CB(2) receptor. Collectively, the pharmacological and biochemical knockdown data indicate that cannabinoid-mediated modulation of macrophage migration to the HIV-1 Tat protein is linked to the CB(2) cannabinoid receptor. Furthermore, these results suggest that the CB(2) cannabinoid receptor has potential to serve as a therapeutic target for ablation of HIV-1-associated untoward inflammatory response.
Collapse
Affiliation(s)
- Erinn S Raborn
- Department of Microbiology and Immunology, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA
| | | |
Collapse
|
92
|
Hazleton JE, Berman JW, Eugenin EA. Novel mechanisms of central nervous system damage in HIV infection. HIV AIDS-RESEARCH AND PALLIATIVE CARE 2010; 2:39-49. [PMID: 22096383 PMCID: PMC3218694 DOI: 10.2147/hiv.s9186] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Human immunodeficiency virus-1 infection of the central nervous system is an early event after primary infection, resulting in motor and cognitive defects in a significant number of individuals despite successful antiretroviral therapy. The pathology of the infected brain is characterized by enhanced leukocyte infiltration, microglial activation and nodules, aberrant expression of inflammatory factors, neuronal dysregulation and loss, and blood–brain barrier disruption. Months to years following the primary infection, these central nervous system insults result in a spectrum of motor and cognitive dysfunction, ranging from mild impairment to frank dementia. The mechanisms that mediate impairment are still not fully defined. In this review we discuss the cellular and molecular mechanisms that facilitate impairment and new data that implicate intercellular communication systems, gap junctions and tunneling nanotubes, as mediators of human immunodeficiency virus-1 toxicity and infection within the central nervous system. These data suggest potential targets for novel therapeutics.
Collapse
Affiliation(s)
- Joy E Hazleton
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY, USA
| | | | | |
Collapse
|
93
|
Shojania S, Henry GD, Chen VC, Vo TN, Perreault H, O’Neil JD. High yield expression and purification of HIV-1 Tat1−72 for structural studies. J Virol Methods 2010; 164:35-42. [DOI: 10.1016/j.jviromet.2009.11.021] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2009] [Revised: 11/17/2009] [Accepted: 11/17/2009] [Indexed: 12/11/2022]
|
94
|
Nappi F, Chiozzini C, Bordignon V, Borsetti A, Bellino S, Cippitelli M, Barillari G, Caputo A, Tyagi M, Giacca M, Ensoli B. Immobilized HIV-1 Tat protein promotes gene transfer via a transactivation-independent mechanism which requires binding of Tat to viral particles. J Gene Med 2010; 11:955-65. [PMID: 19653251 DOI: 10.1002/jgm.1381] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Retroviral transduction of cells is improved upon virus adsorption onto immobilized fibronectin (FN) fragments. Because HIV-1 Tat possesses the same functional domains that lead to increased transduction efficiency in FN by colocalization of bound virus and cells, we hypothesized that Tat could enhance gene transfer by a similar mechanism. METHODS Single-cycle replication retro- or lentivirus carrying green fluorescent protein or cloramphenicol acetyltransferase as reporter genes were added to wells coated with Tat or Tat peptides. Wells were extensively washed to remove unbound virus and levels of transduction were detected by measuring reporter gene expression. Virus adsorption to immobilized Tat was measured using a p24 antigen capture assay. RESULTS Immobilized Tat efficiently binds retro- and lentiviral particles and mediates virus transmission at virus input doses that were otherwise unable to transduce susceptible cells. Virus adsorption to Tat is not mediated by envelope glycoprotein (Env) because immobilized Tat binds and retains vesicular stomatitis virus G (VSV-G) pseudotypes as well as envelope-free particles. HIV-1 Env or VSV-G are required for Tat-assisted transduction, which is abrogated by an antibody blocking the HIV-1 Env-CD4 interaction. Tat-assisted transduction is mediated by the cysteine-rich region of Tat, which is known to be essential for Tat transactivation activity. However, Tat transactivation is not required for Tat-assisted transduction, as indicated by the enhancement of transduction by transactivation-silent Tat mutants. CONCLUSIONS Immobilized Tat promotes virus transduction by a transactiva- tion-independent mechanism, which requires binding of virus to Tat. Recombinant Tat or Tat fragments provide a new method to increase efficiency of retro- and lentiviral based gene transfer and gene therapy.
Collapse
Affiliation(s)
- Filomena Nappi
- National AIDS Center, Istituto Superiore Sanità, Rome, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
95
|
Proteoglycans in host-pathogen interactions: molecular mechanisms and therapeutic implications. Expert Rev Mol Med 2010; 12:e5. [PMID: 20113533 DOI: 10.1017/s1462399409001367] [Citation(s) in RCA: 91] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Many microbial pathogens subvert proteoglycans for their adhesion to host tissues, invasion of host cells, infection of neighbouring cells, dissemination into the systemic circulation, and evasion of host defence mechanisms. Where studied, specific virulence factors mediate these proteoglycan-pathogen interactions, which are thus thought to affect the onset, progression and outcome of infection. Proteoglycans are composites of glycosaminoglycan (GAG) chains attached covalently to specific core proteins. Proteoglycans are expressed ubiquitously on the cell surface, in intracellular compartments, and in the extracellular matrix. GAGs mediate the majority of ligand-binding activities of proteoglycans, and many microbial pathogens elaborate cell-surface and secreted factors that interact with GAGs. Some pathogens also modulate the expression and function of proteoglycans through known virulence factors. Several GAG-binding pathogens can no longer attach to and invade host cells whose GAG expression has been reduced by mutagenesis or enzymatic treatment. Furthermore, GAG antagonists have been shown to inhibit microbial attachment and host cell entry in vitro and reduce virulence in vivo. Together, these observations underscore the biological significance of proteoglycan-pathogen interactions in infectious diseases.
Collapse
|
96
|
Huysentruyt LC, McGrath MS. The role of macrophages in the development and progression of AIDS-related non-Hodgkin lymphoma. J Leukoc Biol 2009; 87:627-32. [PMID: 20042471 PMCID: PMC2858304 DOI: 10.1189/jlb.0809564] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Despite HAART, patients infected with HIV develop NHL at a significantly higher level than the noninfected population. The primary difference between lymphoma in non-HIV-infected individuals and those with ARL is that ARL is consistently high-grade and metastatic. The emergence of ARL is associated with the presence of macrophage viral reservoirs, similar to what has been observed for HAD. HIV-infected macrophages, as seen by histology and HIV p24 staining, are present in approximately half of ARLs. Macrophage reservoirs recruit additional immune cells, including monocytes/macrophages, through the release of chemoattractants. Additionally, TAM are known to promote tumor progression for most cancer types, including lymphomas. This review will highlight and discuss the role of macrophage viral reservoirs in the development and progression of ARLs and hopefully, shed light on this new and interesting field.
Collapse
Affiliation(s)
- Leanne C Huysentruyt
- Department of Medicine, Hematology and Oncology, University of California, San Francisco, California, USA.
| | | |
Collapse
|
97
|
Sulfated K5 Escherichia coli polysaccharide derivatives: A novel class of candidate antiviral microbicides. Pharmacol Ther 2009; 123:310-22. [DOI: 10.1016/j.pharmthera.2009.05.001] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2009] [Accepted: 05/06/2009] [Indexed: 10/20/2022]
|
98
|
Yezid H, Konate K, Debaisieux S, Bonhoure A, Beaumelle B. Mechanism for HIV-1 Tat insertion into the endosome membrane. J Biol Chem 2009; 284:22736-46. [PMID: 19549783 DOI: 10.1074/jbc.m109.023705] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The human immunodeficiency virus, type 1, transactivating protein Tat is a small protein that is strictly required for viral transcription and multiplication within infected cells. The infected cells actively secrete Tat using an unconventional secretion pathway. Extracellular Tat can affect different cell types and induce severe cell dysfunctions ranging from cell activation to cell death. To elicit most cell responses, Tat needs to reach the cell cytosol. To this end, Tat is endocytosed, and low endosomal pH will then trigger Tat translocation to the cytosol. Although this translocation step is critical for Tat cytosolic delivery, how Tat could interact with the endosome membrane is unknown, and the key residues involved in this interaction require identification. We found that, upon acidification below pH 6.0 (i.e. within the endosomal pH range), Tat inserts into model membranes such as monolayers or lipid vesicles. This insertion process relies on Tat single Trp, Trp-11, which is not needed for transactivation and could be replaced by another aromatic residue for membrane insertion. Nevertheless, Trp-11 is strictly required for translocation. Tat conformational changes induced by low pH involve a sensor made of its first acidic residue (Glu/Asp-2) and the end of its basic domain (residues 55-57). Mutation of one of these elements results in membrane insertion above pH 6.5. Tat basic domain is also required for efficient Tat endocytosis and membrane insertion. Together with the strict conservation of Tat Trp among different virus isolates, our results point to an important role for Tat-membrane interaction in the multiplication of human immunodeficiency virus type 1.
Collapse
Affiliation(s)
- Hocine Yezid
- Centre d'Etudes d'Agents Pathogènes et Biotechnologies pour la Santé, UMR 5236 CNRS, Université Montpellier II, 34095 Montpellier, France
| | | | | | | | | |
Collapse
|
99
|
Zhong Y, Smart EJ, Weksler B, Couraud PO, Hennig B, Toborek M. Caveolin-1 regulates human immunodeficiency virus-1 Tat-induced alterations of tight junction protein expression via modulation of the Ras signaling. J Neurosci 2008; 28:7788-96. [PMID: 18667611 PMCID: PMC2635104 DOI: 10.1523/jneurosci.0061-08.2008] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2008] [Revised: 06/05/2008] [Accepted: 06/17/2008] [Indexed: 01/06/2023] Open
Abstract
The blood-brain barrier (BBB) is the critical structure for preventing human immunodeficiency virus (HIV) trafficking into the brain. Specific HIV proteins, such as Tat protein, can contribute to the dysfunction of tight junctions at the BBB and HIV entry into the brain. Tat is released by HIV-1-infected cells and can interact with a variety of cell surface receptors activating several signal transduction pathways, including those localized in caveolae. The present study focused on the mechanisms of Tat-induced caveolae-associated Ras signaling at the level of the BBB. Treatment with Tat activated the Ras pathway in human brain microvascular endothelial cells (HBMECs). However, caveolin-1 silencing markedly attenuated these effects. Because the integrity of the brain endothelium is regulated by intercellular tight junctions, these structural elements of the BBB were also evaluated in the present study. Exposure to Tat diminished the expression of several tight junction proteins, namely, occludin, zonula occludens (ZO)-1, and ZO-2 in the caveolar fraction of HBMECs. These effects were effectively protected by pharmacological inhibition of the Ras signaling and by silencing of caveolin-1. The present data indicate the importance of caveolae-associated signaling in the disruption of tight junctions on Tat exposure. They also demonstrate that caveolin-1 may constitute an early and critical modulator that controls signaling pathways leading to the disruption of tight junction proteins. Thus, caveolin-1 may provide an effective target to protect against Tat-induced HBMEC dysfunction and the disruption of the BBB in HIV-1-infected patients.
Collapse
Affiliation(s)
- Yu Zhong
- Molecular Neuroscience and Vascular Biology Laboratory, Department of Neurosurgery
| | | | - Babette Weksler
- Weill Medical College of Cornell University, New York, New York 10021, and
| | - Pierre-Olivier Couraud
- Institut Cochin, Institut National de la Santé et de la Recherche Médicale Unité 567, 75014 Paris, France
| | - Bernhard Hennig
- College of Agriculture, University of Kentucky, Lexington, Kentucky 40536
| | - Michal Toborek
- Molecular Neuroscience and Vascular Biology Laboratory, Department of Neurosurgery
| |
Collapse
|
100
|
Kalantari P, Harandi OF, Hankey PA, Henderson AJ. HIV-1 Tat mediates degradation of RON receptor tyrosine kinase, a regulator of inflammation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2008; 181:1548-55. [PMID: 18606710 PMCID: PMC2536764 DOI: 10.4049/jimmunol.181.2.1548] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
HIV encodes several proteins, including Tat, that have been demonstrated to modulate the expression of receptors critical for innate immunity, including MHC class I, mannose receptor, and beta(2)-microglobulin. We demonstrate that Tat targets the receptor tyrosine kinase recepteur d'origine nantais (RON), which negatively regulates inflammation and HIV transcription, for proteosome degradation. Tat decreases cell surface RON expression in HIV-infected monocytic cells, and Tat-mediated degradation of RON protein is blocked by inhibitors of proteosome activity. Tat specifically induced down-regulation of RON and not other cell surface receptors, such as the transferrin receptor, the receptor tyrosine kinase TrkA, or monocytic markers CD14 and ICAM-1. The Tat trans activation domain is required for RON degradation, and this down-regulation is dependent on the integrity of the kinase domain of RON receptor. We propose that Tat mediates degradation of RON through a ubiquitin-proteosome pathway, and suggest that by targeting signals that modulate inflammation, Tat creates a microenvironment that is optimal for HIV replication and progression of AIDS-associated diseases.
Collapse
Affiliation(s)
- Parisa Kalantari
- Graduate Program in Pathobiology, Center for Molecular Immunology and Infectious Diseases, Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, PA 16802
| | - Omid F. Harandi
- Genetics Program of the Huck Institute for the Life Sciences, Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, PA 16802
| | - Pamela A. Hankey
- Graduate Program in Pathobiology, Center for Molecular Immunology and Infectious Diseases, Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, PA 16802
| | - Andrew J Henderson
- Center for HIV/AIDS Care and Research, Department of Medicine, Section of Infectious Diseases, Boston University School of Medicine, Boston, MA 02118
| |
Collapse
|