1
|
Yukselten Y, Wishah H, Li JA, Sutton RE. Targeting CCR5: A central approach to HIV treatment and cure strategies. Virology 2025; 603:110375. [PMID: 39729963 DOI: 10.1016/j.virol.2024.110375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 12/09/2024] [Accepted: 12/20/2024] [Indexed: 12/29/2024]
Abstract
CCR5, a co-receptor critical for R5-tropic HIV entry into host cells, remains a key target for therapeutic interventions. HIV utilizes CCR5, expressed on T cells and macrophages, to facilitate viral entry. Genetic variants, such as the CCR5Δ32 homozygous mutation that confers protection to HIV infection, have made CCR5 a main target for gene-editing technologies, small-molecule inhibitors, and monoclonal antibody-based therapies. Recent studies emphasize the importance of regulating CCR5 expression at transcriptional and post-transcriptional levels and integrating this approach with traditional therapies. Particularly, the role of heterozygous CCR5Δ32 carriers who are HIV seropositive highlights the potential for targeting CCR5 in combination with other immune-regulatory mechanisms. This may lead to more effective treatment strategies and, ultimately, a functional cure for HIV. This minireview discusses the role of CCR5 in HIV pathogenesis and explores the potential of genetic and therapeutic interventions targeting CCR5 as an innovative strategy in the continued battle against HIV.
Collapse
Affiliation(s)
- Yunus Yukselten
- Section of Infectious Diseases, Department of Internal Medicine, Yale University, New Haven, CT, United States
| | - Hanan Wishah
- Section of Infectious Diseases, Department of Internal Medicine, Yale University, New Haven, CT, United States
| | - Jessica A Li
- Section of Infectious Diseases, Department of Internal Medicine, Yale University, New Haven, CT, United States
| | - Richard E Sutton
- Section of Infectious Diseases, Department of Internal Medicine, Yale University, New Haven, CT, United States.
| |
Collapse
|
2
|
Bober A, Mika J, Piotrowska A. A Missing Puzzle in Preclinical Studies-Are CCR2, CCR5, and Their Ligands' Roles Similar in Obesity-Induced Hypersensitivity and Diabetic Neuropathy?-Evidence from Rodent Models and Clinical Studies. Int J Mol Sci 2024; 25:11323. [PMID: 39457105 PMCID: PMC11508617 DOI: 10.3390/ijms252011323] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 10/16/2024] [Accepted: 10/19/2024] [Indexed: 10/28/2024] Open
Abstract
Research has shown that obesity is a low-grade inflammatory disease that is often associated with comorbidities, such as diabetes and chronic pain. Recent data have indicated that chemokines may play a role in these conditions due to their pronociceptive and chemotactic properties, which promote hypersensitivity and inflammation. Accumulating evidence suggests that CCR2, CCR5, and their ligands (CCL2, CCL3, CCL4, CCL5, CCL7, CCL8, CCL11 CCL12, and/or CCL13) play a role in rodent models of pain and obesity, as well as in patients with diabetes and obesity. It was proven that the blockade of CCR2 and CCR5, including the simultaneous blockade of both receptors by dual antagonists, effectively reduces hypersensitivity to thermal and mechanical stimuli in chronic pain states, including diabetic neuropathy. The present review discusses these chemokine receptors and the role of their ligands in diabetes and obesity, as well as their involvement in diabetic neuropathy and obesity-induced hypersensitivity.
Collapse
Affiliation(s)
| | - Joanna Mika
- Department of Pain Pharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, 31-343 Krakow, Poland;
| | - Anna Piotrowska
- Department of Pain Pharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, 31-343 Krakow, Poland;
| |
Collapse
|
3
|
Tufail A, Akkad S, Noble AR, Fascione MA, Signoret N. New insight into a simple high-yielding method for the production of fully folded and functional recombinant human CCL5. Sci Rep 2024; 14:24188. [PMID: 39406925 PMCID: PMC11480376 DOI: 10.1038/s41598-024-75327-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 10/04/2024] [Indexed: 10/19/2024] Open
Abstract
Chemokines are proteins important for a range of biological processes from cell-directed migration (chemotaxis) to cell activation and differentiation. Chemokine C-C ligand 5 (CCL5) is an important pro-inflammatory chemokine attracting immune cells towards inflammatory sites through interaction with its receptors CCR1/3/5. Recombinant production of large quantities of CCL5 in Escherichia coli is challenging due to formation of inclusion bodies which necessitates refolding, often leading to low recovery of biologically active protein. To combat this, we have developed a method for CCL5 production that utilises the purification of SUMO tagged CCL5 from E. coli SHuffle cells avoiding the need to reform disulfide bonds through inclusion body purification and yields high quantities of CCL5 (~ 25 mg/L). We demonstrated that the CCL5 produced was fully functional by assessing well-established cellular changes triggered by CCL5 binding to CCR5, including receptor phosphorylation and internalisation, intracellular signalling leading to calcium flux, as well as cell migration. Overall, we demonstrate that the use of solubility tags, SHuffle cells and low pH dialysis constitutes an approach that increases purification yields of active CCL5 with low endotoxin contamination for biological studies.
Collapse
Affiliation(s)
- Afzaal Tufail
- Hull York Medical School, University of York, York, YO10 5DD, UK
| | - Saeed Akkad
- Department of Chemistry, University of York, York, YO10 5DD, UK
| | - Amanda R Noble
- Department of Chemistry, University of York, York, YO10 5DD, UK
| | | | | |
Collapse
|
4
|
Chen CW, Chen LK, Chung YT, Liu SY, Chen SW, Chang YI, Hsieh PS, Juan CC. Cysteine-cysteine Chemokine Receptor Type 5 Plays a Critical Role in Exercise Performance by Regulating Mitochondrial Content in Skeletal Muscle. Inflammation 2023; 46:2089-2101. [PMID: 37436644 DOI: 10.1007/s10753-023-01864-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 06/21/2023] [Accepted: 06/22/2023] [Indexed: 07/13/2023]
Abstract
Cysteine-cysteine chemokine receptor type 5 (CCR5) is thought to play an important role in the trafficking of lymphoid cells but has recently also been associated with AMPK signaling pathways that are implicated in energy metabolism in skeletal muscle. We hypothesized that genetic deletions of CCR5 would alter mitochondria content and exercise performance in mice. CCR5-/- and wild-type mice with the same genetic background were subjected to endurance exercise and grip strength tests. The soleus muscle was stained with immunofluorescence for myosin heavy chain 7 (MYH7) and succinate dehydrogenase (SDH) analysis as well as the expression of genes associated with muscle atrophy and mitochondrial oxidative phosphorylation were measured using qPCR. Although there were no differences in the weight of the soleus muscle between the CCR5-/- group and the wild-type mice, the CCR5-/- mice showed the following muscular dysfunctions: (i) decreased MYH7 percentage and cross-section area, (ii) higher myostatin and atrogin-1 mRNA levels, (iii) dropped expression of mitochondrial DNA-encoded electron respiratory chain genes (cytochrome B, cytochrome c oxidase subunit III, and ATP synthase subunit 6) as well as mitochondrial generation genes (PPARγ and PGC-1α), and (iv) lower SDH activity and exercise performance when compared with wild-type mice. In addition, genes associated with mitochondrial biogenesis (PGC-1α, PPARγ, and MFN2) and mitochondrial complex (ND4 and Cytb) were upregulated when the skeletal muscle cell line C2C12 was exposed to cysteine-cysteine chemokine ligand 4 (a ligand of CCR5) in vitro. These findings suggested that attenuation of endurance exercise performance is related to the loss of mitochondrial content and lower SDH activity of soleus muscle in CCR5 knockout mice. The present study provides evidence indicating that the chemokine receptor CCR5 might modulate the skeletal muscle metabolic energy system during exercise.
Collapse
Affiliation(s)
- Chien-Wei Chen
- International Sport Science Master's Program, College of Human Development and Health, National Taipei University of Nursing and Health Sciences, Taipei, Taiwan
| | - Luen-Kui Chen
- Institutes of Physiology, College of Medicine, National Yang Ming Chiao Tung University, No. 155, Sec. 2, Li-Nong St., 112304, Taipei, Taiwan
| | - Yi-Ting Chung
- Institutes of Physiology, College of Medicine, National Yang Ming Chiao Tung University, No. 155, Sec. 2, Li-Nong St., 112304, Taipei, Taiwan
| | - Shui-Yu Liu
- Institutes of Physiology, College of Medicine, National Yang Ming Chiao Tung University, No. 155, Sec. 2, Li-Nong St., 112304, Taipei, Taiwan
| | - Shuoh-Wen Chen
- Institutes of Physiology, College of Medicine, National Yang Ming Chiao Tung University, No. 155, Sec. 2, Li-Nong St., 112304, Taipei, Taiwan
| | - Yuan-I Chang
- Institutes of Physiology, College of Medicine, National Yang Ming Chiao Tung University, No. 155, Sec. 2, Li-Nong St., 112304, Taipei, Taiwan
| | - Po-Shiuan Hsieh
- Department of Physiology and Biophysics, National Defense Medical Center, Taipei, Taiwan
- Department of Medical Research, Tri-Service General Hospital, Taipei, Taiwan
| | - Chi-Chang Juan
- Institutes of Physiology, College of Medicine, National Yang Ming Chiao Tung University, No. 155, Sec. 2, Li-Nong St., 112304, Taipei, Taiwan.
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan.
| |
Collapse
|
5
|
Liu S, Liu C, Wang Q, Liu S, Min J. CC Chemokines in Idiopathic Pulmonary Fibrosis: Pathogenic Role and Therapeutic Potential. Biomolecules 2023; 13:biom13020333. [PMID: 36830702 PMCID: PMC9953349 DOI: 10.3390/biom13020333] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 01/03/2023] [Accepted: 01/05/2023] [Indexed: 02/12/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF), characterized by progressive worsening of dyspnea and irreversible decline in lung function, is a chronic and progressive respiratory disease with a poor prognosis. Chronic or repeated lung injury results in inflammation and an excessive injury-repairing response that drives the development of IPF. A number of studies have shown that the development and progression of IPF are associated with dysregulated expression of several chemokines and chemokine receptors, several of which have been used as predictors of IPF outcome. Chemokines of the CC family play significant roles in exacerbating IPF progression by immune cell attraction or fibroblast activation. Modulating levels of detrimental CC chemokines and interrupting the corresponding transduction axis by neutralizing antibodies or antagonists are potential treatment options for IPF. Here, we review the roles of different CC chemokines in the pathogenesis of IPF, and their potential use as biomarkers or therapeutic targets.
Collapse
Affiliation(s)
- Shanshan Liu
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, China
- Correspondence:
| | - Chang Liu
- Drug Clinical Trial Institution, Children’s Hospital, Capital Institute of Pediatrics, Beijing 100020, China
| | - Qianrong Wang
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Suosi Liu
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Jiali Min
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| |
Collapse
|
6
|
Hillman T. A Predictive Model for Identifying the Most Effective Anti-CCR5 Monoclonal Antibody. ARCHIVES OF PHARMACY PRACTICE 2023. [DOI: 10.51847/d9m2zufqr4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
|
7
|
Weichseldorfer M, Tagaya Y, Reitz M, DeVico AL, Latinovic OS. Identifying CCR5 coreceptor populations permissive for HIV-1 entry and productive infection: implications for in vivo studies. J Transl Med 2022; 20:39. [PMID: 35073923 PMCID: PMC8785515 DOI: 10.1186/s12967-022-03243-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 01/10/2022] [Indexed: 12/15/2022] Open
Abstract
Background The chemokine receptor CCR5 is the major coreceptor for HIV-1 cell entry. We previously observed that not all CCR5 mAbs reduce HIV-1 infection, suggesting that only some CCR5 populations are permissive for HIV-1 entry. This study aims to better understand the relevant conformational states of the cellular coreceptor, CCR5, involved in HIV entry. We hypothesized that CCR5 assumes multiple configurations during normal cycling on the plasma membrane, but only particular forms facilitate HIV-1 infection. Methods To this end, we quantified different CCR5 populations using six CCR5 monoclonal antibodies (mAbs) with different epitope specificities and visualized them with super-resolution microscopy. We quantified each surface CCR5 population before and after HIV-1 infection. Results Based on CCR5 conformational changes, down-modulation, and trafficking rates (internalization and recycling kinetics), we were able to distinguish among heterogeneous CCR5 populations and thus which populations might best be targeted to inhibit HIV-1 entry. We assume that a decreased surface presence of a particular CCR5 subpopulation following infection means that it has been internalized due to HIV-1 entry, and that it therefore represents a highly relevant target for future antiviral therapy strategies. Strikingly, this was most true for antibody CTC8, which targets the N-terminal region of CCR5 and blocks viral entry more efficiently than it blocks chemokine binding. Conclusions Defining the virus-host interactions responsible for HIV-1 transmission, including specific coreceptor populations capable of establishing de novo infections, is essential for the development of an HIV-1 vaccine. This study hopefully will facilitate further development of inhibitors to block CCR5 usage by HIV-1, as well as inform future HIV-1 vaccine design. Supplementary Information The online version contains supplementary material available at 10.1186/s12967-022-03243-8.
Collapse
Affiliation(s)
- Matthew Weichseldorfer
- Institute of Human Virology, School of Medicine, University of Maryland, 725 W. Lombard St., Baltimore, MD, 21201, USA
| | - Yutaka Tagaya
- Institute of Human Virology, School of Medicine, University of Maryland, 725 W. Lombard St., Baltimore, MD, 21201, USA.,Department of Medicine, School of Medicine, University of Maryland, Baltimore, MD, 21201, USA
| | - Marvin Reitz
- Institute of Human Virology, School of Medicine, University of Maryland, 725 W. Lombard St., Baltimore, MD, 21201, USA.,Department of Medicine, School of Medicine, University of Maryland, Baltimore, MD, 21201, USA
| | - Anthony L DeVico
- Institute of Human Virology, School of Medicine, University of Maryland, 725 W. Lombard St., Baltimore, MD, 21201, USA.,Department of Medicine, School of Medicine, University of Maryland, Baltimore, MD, 21201, USA
| | - Olga S Latinovic
- Institute of Human Virology, School of Medicine, University of Maryland, 725 W. Lombard St., Baltimore, MD, 21201, USA. .,Department of Microbiology and Immunology, School of Medicine, University of Maryland, Baltimore, MD, 21201, USA.
| |
Collapse
|
8
|
Seow KM, Liu PS, Chen KH, Chen CW, Chen LK, Ho CH, Hwang JL, Wang PH, Juan CC. Cysteine-Cysteine Motif Chemokine Receptor 5 Expression in Letrozole-Induced Polycystic Ovary Syndrome Mice. Int J Mol Sci 2021; 23:134. [PMID: 35008567 PMCID: PMC8745167 DOI: 10.3390/ijms23010134] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 12/20/2021] [Accepted: 12/20/2021] [Indexed: 12/27/2022] Open
Abstract
Polycystic ovary syndrome (PCOS), which affects 5-10% of women of reproductive age, is associated with reproductive and metabolic disorders, such as chronic anovulation, infertility, insulin resistance, and type 2 diabetes. However, the mechanism of PCOS is still unknown. Therefore, this study used a letrozole-exposed mouse model in which mice were orally fed letrozole for 20 weeks to investigate the effects of letrozole on the severity of reproductive and metabolic consequences and the expression of cysteine-cysteine motif chemokine receptor 5 (CCR5) in letrozole-induced PCOS mice. The letrozole-treated mice showed a disrupted estrous cycle and were arrested in the diestrus phase. Letrozole treatment also increased plasma testosterone levels, decreased estradiol levels, and caused multicystic follicle formation. Furthermore, histological analysis of the perigonadal white adipose tissue (pgWAT) showed no significant difference in the size and number of adipocytes between the letrozole-treated mice and the control group. Further, the letrozole-treated mice demonstrated glucose intolerance and insulin resistance during oral glucose and insulin tolerance testing. Additionally, the expression of CCR5 and cysteine-cysteine motif ligand 5 (CCL5) were significantly higher in the pgWAT of the letrozole-treated mice compared with the control group. CCR5 and CCL5 were also significantly correlated with the homeostasis model assessment of insulin resistance (HOMA-IR). Finally, the mechanisms of insulin resistance in PCOS may be caused by an increase in serine phosphorylation and a decrease in Akt phosphorylation.
Collapse
Affiliation(s)
- Kok-Min Seow
- Department of Obstetrics and Gynecology, Shin-Kong Wu Ho-Su Memorial Hospital, Taipei 111, Taiwan; (K.-M.S.); (J.-L.H.)
- Department of Obstetrics and Gynecology, School of Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan; (C.-H.H.); (P.-H.W.)
| | - Pin-Shiou Liu
- Institute of Physiology, National Yang Ming Chiao Tung University, Taipei 112, Taiwan; (P.-S.L.); (L.-K.C.)
| | - Kuo-Hu Chen
- Department of Obstetrics and Gynecology, Taipei Tzu-Chi Hospital, The Buddhist Tzu-Chi Medical Foundation, Taipei 231, Taiwan;
- School of Medicine, Buddhist Tzu-Chi University, Hualien 970, Taiwan
| | - Chien-Wei Chen
- College of Human Development and Health, National Taipei University of Nursing and Health Sciences, Taipei 112, Taiwan;
| | - Luen-Kui Chen
- Institute of Physiology, National Yang Ming Chiao Tung University, Taipei 112, Taiwan; (P.-S.L.); (L.-K.C.)
| | - Chi-Hong Ho
- Department of Obstetrics and Gynecology, School of Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan; (C.-H.H.); (P.-H.W.)
- Department of Obstetrics and Gynecology, Taipei Veterans General Hospital, Taipei 112, Taiwan
| | - Jiann-Loung Hwang
- Department of Obstetrics and Gynecology, Shin-Kong Wu Ho-Su Memorial Hospital, Taipei 111, Taiwan; (K.-M.S.); (J.-L.H.)
- Department of Obstetrics and Gynecology, Taipei Medical University, Taipei 110, Taiwan
- Taipei IVF, Taipei 104, Taiwan
| | - Peng-Hui Wang
- Department of Obstetrics and Gynecology, School of Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan; (C.-H.H.); (P.-H.W.)
- Department of Obstetrics and Gynecology, Taipei Veterans General Hospital, Taipei 112, Taiwan
- Institute of Clinical Medicine, National Yang-Ming University, Taipei 112, Taiwan
- Female Cancer Foundation, Taipei 104, Taiwan
- Department of Medical Research, China Medical University Hospital, Taichung 404, Taiwan
| | - Chi-Chang Juan
- Institute of Physiology, National Yang Ming Chiao Tung University, Taipei 112, Taiwan; (P.-S.L.); (L.-K.C.)
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 112, Taiwan
| |
Collapse
|
9
|
CD4 T Cell-Mediated Immune Control of Cytomegalovirus Infection in Murine Salivary Glands. Pathogens 2021; 10:pathogens10121531. [PMID: 34959486 PMCID: PMC8704252 DOI: 10.3390/pathogens10121531] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 11/17/2021] [Accepted: 11/21/2021] [Indexed: 12/24/2022] Open
Abstract
CD4 T cells are well known for their supportive role in CD8 T cell and B cell responses during viral infection. However, during murine cytomegalovirus (MCMV) infection in the salivary glands (SGs), CD4 T cells exhibit direct antiviral effector functions to control the infection. In this mucosal organ, opposed to other infected tissues, MCMV establishes a sustained lytic replication that lasts for several weeks. While the protective function of CD4 T cells is exerted through the production of the pro-inflammatory cytokines interferon gamma (IFNγ) and tumor necrosis factor alpha (TNF), the reasons for their markedly delayed control of lytic MCMV infection remain elusive. Here, we review the current knowledge on the dynamics and mechanisms of the CD4 T cell-mediated control of MCMV-infected SGs, including their localization in the SG in relation to MCMV infected cells and other immune cells, their mode of action, and their regulation.
Collapse
|
10
|
Nickoloff-Bybel EA, Festa L, Meucci O, Gaskill PJ. Co-receptor signaling in the pathogenesis of neuroHIV. Retrovirology 2021; 18:24. [PMID: 34429135 PMCID: PMC8385912 DOI: 10.1186/s12977-021-00569-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 08/11/2021] [Indexed: 12/13/2022] Open
Abstract
The HIV co-receptors, CCR5 and CXCR4, are necessary for HIV entry into target cells, interacting with the HIV envelope protein, gp120, to initiate several signaling cascades thought to be important to the entry process. Co-receptor signaling may also promote the development of neuroHIV by contributing to both persistent neuroinflammation and indirect neurotoxicity. But despite the critical importance of CXCR4 and CCR5 signaling to HIV pathogenesis, there is only one therapeutic (the CCR5 inhibitor Maraviroc) that targets these receptors. Moreover, our understanding of co-receptor signaling in the specific context of neuroHIV is relatively poor. Research into co-receptor signaling has largely stalled in the past decade, possibly owing to the complexity of the signaling cascades and functions mediated by these receptors. Examining the many signaling pathways triggered by co-receptor activation has been challenging due to the lack of specific molecular tools targeting many of the proteins involved in these pathways and the wide array of model systems used across these experiments. Studies examining the impact of co-receptor signaling on HIV neuropathogenesis often show activation of multiple overlapping pathways by similar stimuli, leading to contradictory data on the effects of co-receptor activation. To address this, we will broadly review HIV infection and neuropathogenesis, examine different co-receptor mediated signaling pathways and functions, then discuss the HIV mediated signaling and the differences between activation induced by HIV and cognate ligands. We will assess the specific effects of co-receptor activation on neuropathogenesis, focusing on neuroinflammation. We will also explore how the use of substances of abuse, which are highly prevalent in people living with HIV, can exacerbate the neuropathogenic effects of co-receptor signaling. Finally, we will discuss the current state of therapeutics targeting co-receptors, highlighting challenges the field has faced and areas in which research into co-receptor signaling would yield the most therapeutic benefit in the context of HIV infection. This discussion will provide a comprehensive overview of what is known and what remains to be explored in regard to co-receptor signaling and HIV infection, and will emphasize the potential value of HIV co-receptors as a target for future therapeutic development. ![]()
Collapse
Affiliation(s)
- E A Nickoloff-Bybel
- Department of Pharmacology and Physiology, Drexel University College of Medicine, 245 N. 15th Street, Philadelphia, PA, 19102, USA
| | - L Festa
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, 240 S. 40th Street, Philadelphia, PA, 19104, USA
| | - O Meucci
- Department of Pharmacology and Physiology, Drexel University College of Medicine, 245 N. 15th Street, Philadelphia, PA, 19102, USA.,Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, 19102, USA
| | - P J Gaskill
- Department of Pharmacology and Physiology, Drexel University College of Medicine, 245 N. 15th Street, Philadelphia, PA, 19102, USA.
| |
Collapse
|
11
|
Scurci I, Akondi KB, Pinheiro I, Paolini-Bertrand M, Borgeat A, Cerini F, Hartley O. CCR5 tyrosine sulfation heterogeneity generates cell surface receptor subpopulations with different ligand binding properties. Biochim Biophys Acta Gen Subj 2020; 1865:129753. [PMID: 32991968 DOI: 10.1016/j.bbagen.2020.129753] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 09/25/2020] [Accepted: 09/25/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND Chemokine receptor tyrosine sulfation plays a key role in the binding of chemokines. It has been suggested that receptor sulfation is heterogeneous, but no experimental evidence has been provided so far. The potent anti-HIV chemokine analog 5P12-RANTES has been proposed to owe its inhibitory activity to a capacity to bind a larger pool of cell surface CCR5 receptors than native chemokines such as CCL5, but the molecular details underlying this phenomenon have not been elucidated. METHODS We investigated the CCR5 sulfation heterogeneity and the sensitivity of CCR5 ligands to receptor sulfation by performing ELISA assays on synthetic N-terminal sulfopeptides and by performing binding assays on CCR5-expressing cells under conditions that modulate CCR5 sulfation levels. RESULTS Two commonly used anti-CCR5 monoclonal antibodies with epitopes in the sulfated N-terminal domain of CCR5 show contrasting binding profiles on CCR5 sulfopeptides, incomplete competition with each other for cell surface CCR5, and opposing sensitivities to cellular treatments that affect CCR5 sulfation levels. 5P12-RANTES is less sensitive than native CCL5 to conditions that affect cellular CCR5 sulfation. CONCLUSIONS CCR5 sulfation is heterogeneous and this affects the binding properties of both native chemokines and antibodies. Enhanced capacity to bind to CCR5 is a component of the inhibitory mechanism of 5P12-RANTES. GENERAL SIGNIFICANCE We provide the first experimental evidence for sulfation heterogeneity of chemokine receptors and its impact on ligand binding, a phenomenon that is important both for the understanding of chemokine cell biology and for the development of drugs that target chemokine receptors.
Collapse
Affiliation(s)
- I Scurci
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Switzerland
| | - K B Akondi
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Switzerland
| | - I Pinheiro
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Switzerland
| | - M Paolini-Bertrand
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Switzerland
| | - A Borgeat
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Switzerland
| | - F Cerini
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Switzerland
| | - O Hartley
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Switzerland; Orion Biotechnology, Avenue de Sécheron 15, 1202 Genève, Switzerland.
| |
Collapse
|
12
|
Purification Methods and the Presence of RNA in Virus Particles and Extracellular Vesicles. Viruses 2020; 12:v12090917. [PMID: 32825599 PMCID: PMC7552034 DOI: 10.3390/v12090917] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Revised: 08/12/2020] [Accepted: 08/18/2020] [Indexed: 12/17/2022] Open
Abstract
The fields of extracellular vesicles (EV) and virus infections are marred in a debate on whether a particular mRNA or non-coding RNA (i.e., miRNA) is packaged into a virus particle or copurifying EV and similarly, whether a particular mRNA or non-coding RNA is contained in meaningful numbers within an EV. Key in settling this debate, is whether the purification methods are adequate to separate virus particles, EV and contaminant soluble RNA and RNA:protein complexes. Differential centrifugation/ultracentrifugation and precipitating agents like polyethylene glycol are widely utilized for both EV and virus purifications. EV are known to co-sediment with virions and other particulates, such as defective interfering particles and protein aggregates. Here, we discuss how encased RNAs from a heterogeneous mixture of particles can be distinguished by different purification methods. This is particularly important for subsequent interpretation of whether the RNA associated phenotype is contributed solely by virus or EV particles or a mixture of both. We also discuss the discrepancy of miRNA abundance in EV from different input material.
Collapse
|
13
|
Yu-Ju Wu C, Chen CH, Lin CY, Feng LY, Lin YC, Wei KC, Huang CY, Fang JY, Chen PY. CCL5 of glioma-associated microglia/macrophages regulates glioma migration and invasion via calcium-dependent matrix metalloproteinase 2. Neuro Oncol 2020; 22:253-266. [PMID: 31593589 PMCID: PMC7032635 DOI: 10.1093/neuonc/noz189] [Citation(s) in RCA: 104] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Accepted: 10/03/2019] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Glioma-associated microglia/macrophages (GAMs) comprise macrophages of peripheral origin and brain-intrinsic microglia, which support tumor progression. Chemokine C-C ligand 5 (CCL5) is an inflammatory mediator produced by immune cells and is involved in tumor growth and migration in several cancers, including glioma. However, the mechanisms detailing how CCL5 facilitates glioma invasion remain largely unresolved. METHODS Glioma migration and invasion were determined by wound healing, transwell assay, and 3D µ-slide chemotaxis assay. The expression levels of CCL5, CD68, matrix metalloproteinase 2 (MMP2), phosphorylated Ca2+/calmodulin-dependent protein kinase II (p-CaMKII), p-Akt, and phosphorylated proline-rich tyrosine kinase 2 were determined by cytokine array, quantitative PCR, western blot, or immunohistochemistry. Zymography and intracellular calcium assays were used to analyze MMP2 activity and intracellular calcium levels, respectively. RESULTS CCL5 modulated the migratory and invasive activities of human glioma cells in association with MMP2 expression. In response to CCL5, glioma cells underwent a synchronized increase in intracellular calcium levels and p-CaMKII and p-Akt expression levels. CCL5-directed glioma invasion and increases in MMP2 were suppressed after inhibition of p-CaMKII. Glioma cells tended to migrate toward GAM-conditioned media activated by granulocyte-macrophage colony-stimulating factor (GM-CSF) in which CCL5 was abundant. This homing effect was associated with MMP2 upregulation, and could be ameliorated either by controlling intracellular and extracellular calcium levels or by CCL5 antagonism. Clinical results also revealed the associations between CCL5 and GAM activation. CONCLUSION Our results suggest that modulation of glioma CaMKII may restrict the effect of CCL5 on glioma invasion and could be a potential therapeutic target for alleviating glioma growth.
Collapse
Affiliation(s)
- Caren Yu-Ju Wu
- Graduate Institute of Biomedical Sciences, Pharmaceutics Laboratory, Graduate Institute of Natural Products, Chang Gung University, Taoyuan, Taiwan
- Department of Neurosurgery, Keelung Chang Gung Memorial Hospital, Keelung, Taiwan
| | - Chia-Hua Chen
- Department of Neurosurgery, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Chun-Yen Lin
- Department of Gastroenterology and Hepatology, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
- School of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Li-Ying Feng
- Department of Neurosurgery, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Yung-Chang Lin
- Department of Gastroenterology and Hepatology, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Kuo-Chen Wei
- Department of Neurosurgery, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
- School of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chiung-Yin Huang
- Department of Neurosurgery, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Jia-You Fang
- Graduate Institute of Biomedical Sciences, Pharmaceutics Laboratory, Graduate Institute of Natural Products, Chang Gung University, Taoyuan, Taiwan
- Department of Anesthesiology, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
- Research Center for Food and Cosmetic Safety and Research Center for Chinese Herbal Medicine, Chang Gung University of Science and Technology, Taoyuan, Taiwan
| | - Pin-Yuan Chen
- Department of Neurosurgery, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
- Department of Neurosurgery, Keelung Chang Gung Memorial Hospital, Keelung, Taiwan
- School of Medicine, Chang Gung University, Taoyuan, Taiwan
| |
Collapse
|
14
|
Kletenkov K, Martynova E, Davidyuk Y, Kabwe E, Shamsutdinov A, Garanina E, Shakirova V, Khaertynova I, Anokhin V, Tarlinton R, Rizvanov A, Khaiboullina S, Morzunov S. Δ ccr5 Genotype Is Associated with Mild Form of Nephropathia Epidemica. Viruses 2019; 11:v11070675. [PMID: 31340562 PMCID: PMC6669606 DOI: 10.3390/v11070675] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 06/22/2019] [Accepted: 07/17/2019] [Indexed: 01/03/2023] Open
Abstract
Nephropathia Epidemica (NE), a mild form of hemorrhagic fever with renal syndrome (HFRS) and linked to hantavirus infection, is endemic in the Republic of Tatarstan. Several genetic markers of HFRS severity have been identified previously, including human leukocyte antigen (HLA) complexes and nucleotide polymorphism in the tumor necrosis factor alpha (TNFα) gene. Still, our understanding of the genetic markers of NE severity remains incomplete. The frequency of the C-C chemokine receptor type 5 (CCR5) gene wild type and gene with 32-base-pair deletion (Δ32CCR5) genotypes in 98 NE samples and 592 controls was analyzed using PCR. Along with the serum levels of 94 analytes, a lack of differences in the CCR5 genotype distribution between NE cases and the general population suggests that the CCR5 genotype does not affect susceptibility to hantavirus infection. However, in NE cases, significant variation in the serum levels of the host matrix metalloproteases between functional CCR5 homozygous and Δ32CCR5 heterozygous patients was detected. Also, the oliguric phase was longer, while thrombocyte counts were lower in functional CCR5 homozygous as compared to heterozygous NE cases. Our data, for the first time, presents the potential role of the CCR5 receptor genotype in NE pathogenesis. Our data suggests that NE pathogenesis in functional CCR5 homozygous and heterozygous NE patients differs, where homozygous cases may have more disintegration of the extracellular matrix and potentially more severe disease.
Collapse
Affiliation(s)
- Konstantin Kletenkov
- Openlab "Gene and Cell Technologies", Institute of Fundamental Medicine and Biology Kazan Federal University, Kazan 420008, Republic of Tatarstan, Russian
| | - Ekaterina Martynova
- Openlab "Gene and Cell Technologies", Institute of Fundamental Medicine and Biology Kazan Federal University, Kazan 420008, Republic of Tatarstan, Russian
| | - Yuriy Davidyuk
- Openlab "Gene and Cell Technologies", Institute of Fundamental Medicine and Biology Kazan Federal University, Kazan 420008, Republic of Tatarstan, Russian
| | - Emmanuel Kabwe
- Openlab "Gene and Cell Technologies", Institute of Fundamental Medicine and Biology Kazan Federal University, Kazan 420008, Republic of Tatarstan, Russian
| | - Anton Shamsutdinov
- Openlab "Gene and Cell Technologies", Institute of Fundamental Medicine and Biology Kazan Federal University, Kazan 420008, Republic of Tatarstan, Russian
| | - Ekaterina Garanina
- Openlab "Gene and Cell Technologies", Institute of Fundamental Medicine and Biology Kazan Federal University, Kazan 420008, Republic of Tatarstan, Russian
| | - Venera Shakirova
- Department of Infectious Diseases, Kazan State Medical Academy, Kazan 420012, the Republic of Tatarstan, Russian
| | - Ilsiyar Khaertynova
- Department of Infectious Diseases, Kazan State Medical Academy, Kazan 420012, the Republic of Tatarstan, Russian
| | - Vladimir Anokhin
- Department of Pediatric Infectious Diseases, Kazan State Medical University, Kazan 420012, Republic of Tatarstan, Russian
| | - Rachael Tarlinton
- School of Veterinary Medicine and Science, University of Nottingham, Loughborough LE12 5RD, UK
| | - Albert Rizvanov
- Openlab "Gene and Cell Technologies", Institute of Fundamental Medicine and Biology Kazan Federal University, Kazan 420008, Republic of Tatarstan, Russian
| | - Svetlana Khaiboullina
- Openlab "Gene and Cell Technologies", Institute of Fundamental Medicine and Biology Kazan Federal University, Kazan 420008, Republic of Tatarstan, Russian
- Department of Microbiology and Immunology, University of Nevada, Reno, NV 89557, USA
| | - Sergey Morzunov
- Department of Pathology, University of Nevada, Reno, NV 89557, USA.
| |
Collapse
|
15
|
Hunegnaw R, Mushtaq Z, Enyindah-Asonye G, Hoang T, Robert-Guroff M. Alveolar Macrophage Dysfunction and Increased PD-1 Expression During Chronic SIV Infection of Rhesus Macaques. Front Immunol 2019; 10:1537. [PMID: 31333668 PMCID: PMC6618664 DOI: 10.3389/fimmu.2019.01537] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 06/19/2019] [Indexed: 01/04/2023] Open
Abstract
HIV infected individuals have been shown to be pre-disposed to pulmonary infections even while receiving anti-retroviral therapy. Alveolar macrophages (AMs) play a critical role in lung innate immunity, but contradictory results have been reported regarding their functionality following HIV infection. Here, using the SIV rhesus macaque model, we document the effect of SIV infection on the phenotypic and functional properties of AMs. Following infection with SIVmac251, AMs in bronchoalveolar lavage (BAL) sampled over 2- to 20-weeks post-infection (wpi) were compared to those in BAL samples from naïve macaques. AM expression of proinflammatory cytokines TNF-α, IL-6, IL-1β, and chemokine RANTES drastically increased 2-wpi compared to AMs of naïve macaques (p < 0.0001 for all), but dropped significantly with progression to chronic infection. Phagocytic activity of AMs 2-and 4-wpi was elevated compared to AMs of naive animals (p = 0.0005, p = 0.0004, respectively) but significantly decreased by 12-wpi (p = 0.0022, p = 0.0019, respectively). By 20-wpi the ability of AMs from chronically infected animals to perform SIV-specific antibody-dependent phagocytosis (ADP) was also diminished (p = 0.028). Acute SIV infection was associated with increased FcγRIII expression which subsequently declined with disease progression. Frequency of FcγRIII+ AMs showed a strong trend toward correlation with SIV-specific ADP, and at 2-wpi FcγRIII expression negatively correlated with viral load (r = -0.6819; p = 0.0013), suggesting a contribution to viremia control. Importantly, PD-1 was found to be expressed on AMs and showed a strong trend toward correlation with plasma viral load (r = 0.8266; p = 0.058), indicating that similar to over-expression on T-cells, PD-1 expression on AMs may also be associated with disease progression. Further, AMs predominantly expressed PD-L2, which remained consistent over the course of infection. PD-1 blockade enhanced SIV-specific ADP by AMs from chronic infection indicating that the PD-1/PD-L2 pathway may modulate functional activity of AMs at that stage. These findings provide new insight into the dynamics of SIV infection leading to AM dysfunction and alteration of pulmonary innate immunity. Our results suggest new pathways to exploit in developing therapies targeting pulmonary disease susceptibility in HIV-infected individuals.
Collapse
Affiliation(s)
- Ruth Hunegnaw
- Immune Biology of Retroviral Infection Section, Vaccine Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Zuena Mushtaq
- Immune Biology of Retroviral Infection Section, Vaccine Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Gospel Enyindah-Asonye
- Immune Biology of Retroviral Infection Section, Vaccine Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Tanya Hoang
- Immune Biology of Retroviral Infection Section, Vaccine Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Marjorie Robert-Guroff
- Immune Biology of Retroviral Infection Section, Vaccine Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
16
|
Hundeyin M, Kurz E, Mishra A, Rossi JAK, Liudahl SM, Leis KR, Mehrotra H, Kim M, Torres LE, Ogunsakin A, Link J, Sears RC, Sivagnanam S, Goecks J, Islam KMS, Dolgalev I, Savadkar S, Wang W, Aykut B, Leinwand J, Diskin B, Adam S, Israr M, Gelas M, Lish J, Chin K, Farooq MS, Wadowski B, Wu J, Shah S, Adeegbe DO, Pushalkar S, Vasudevaraja V, Saxena D, Wong KK, Coussens LM, Miller G. Innate αβ T Cells Mediate Antitumor Immunity by Orchestrating Immunogenic Macrophage Programming. Cancer Discov 2019; 9:1288-1305. [PMID: 31266770 DOI: 10.1158/2159-8290.cd-19-0161] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 05/14/2019] [Accepted: 06/27/2019] [Indexed: 12/16/2022]
Abstract
Unconventional T-lymphocyte populations are emerging as important regulators of tumor immunity. Despite this, the role of TCRαβ+CD4-CD8-NK1.1- innate αβ T cells (iαβT) in pancreatic ductal adenocarcinoma (PDA) has not been explored. We found that iαβTs represent ∼10% of T lymphocytes infiltrating PDA in mice and humans. Intratumoral iαβTs express a distinct T-cell receptor repertoire and profoundly immunogenic phenotype compared with their peripheral counterparts and conventional lymphocytes. iαβTs comprised ∼75% of the total intratumoral IL17+ cells. Moreover, iαβT-cell adoptive transfer is protective in both murine models of PDA and human organotypic systems. We show that iαβT cells induce a CCR5-dependent immunogenic macrophage reprogramming, thereby enabling marked CD4+ and CD8+ T-cell expansion/activation and tumor protection. Collectively, iαβTs govern fundamental intratumoral cross-talk between innate and adaptive immune populations and are attractive therapeutic targets. SIGNIFICANCE: We found that iαβTs are a profoundly activated T-cell subset in PDA that slow tumor growth in murine and human models of disease. iαβTs induce a CCR5-dependent immunogenic tumor-associated macrophage program, T-cell activation and expansion, and should be considered as novel targets for immunotherapy.See related commentary by Banerjee et al., p. 1164.This article is highlighted in the In This Issue feature, p. 1143.
Collapse
Affiliation(s)
- Mautin Hundeyin
- S.A. Localio Laboratory, Department of Surgery, New York University School of Medicine, New York, New York
| | - Emma Kurz
- S.A. Localio Laboratory, Department of Surgery, New York University School of Medicine, New York, New York
| | - Ankita Mishra
- S.A. Localio Laboratory, Department of Surgery, New York University School of Medicine, New York, New York
| | - Juan Andres Kochen Rossi
- S.A. Localio Laboratory, Department of Surgery, New York University School of Medicine, New York, New York
| | - Shannon M Liudahl
- Department of Cell, Developmental and Cancer Biology, Oregon Health and Science University, Portland, Oregon
| | - Kenna R Leis
- Department of Cell, Developmental and Cancer Biology, Oregon Health and Science University, Portland, Oregon
| | - Harshita Mehrotra
- S.A. Localio Laboratory, Department of Surgery, New York University School of Medicine, New York, New York
| | - Mirhee Kim
- S.A. Localio Laboratory, Department of Surgery, New York University School of Medicine, New York, New York
| | - Luisana E Torres
- S.A. Localio Laboratory, Department of Surgery, New York University School of Medicine, New York, New York
| | - Adesola Ogunsakin
- S.A. Localio Laboratory, Department of Surgery, New York University School of Medicine, New York, New York
| | - Jason Link
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, Oregon.,Brenden-Colson Center for Pancreatic Care, Oregon Health and Science University, Portland, Oregon
| | - Rosalie C Sears
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, Oregon.,Brenden-Colson Center for Pancreatic Care, Oregon Health and Science University, Portland, Oregon.,Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon
| | - Shamilene Sivagnanam
- Computational Biology Program, Oregon Health and Science University, Portland, Oregon
| | - Jeremy Goecks
- Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon.,Computational Biology Program, Oregon Health and Science University, Portland, Oregon
| | - K M Sadeq Islam
- S.A. Localio Laboratory, Department of Surgery, New York University School of Medicine, New York, New York
| | - Igor Dolgalev
- Department of Pathology, New York University School of Medicine, New York, New York
| | - Shivraj Savadkar
- S.A. Localio Laboratory, Department of Surgery, New York University School of Medicine, New York, New York
| | - Wei Wang
- S.A. Localio Laboratory, Department of Surgery, New York University School of Medicine, New York, New York
| | - Berk Aykut
- S.A. Localio Laboratory, Department of Surgery, New York University School of Medicine, New York, New York
| | - Joshua Leinwand
- S.A. Localio Laboratory, Department of Surgery, New York University School of Medicine, New York, New York
| | - Brian Diskin
- S.A. Localio Laboratory, Department of Surgery, New York University School of Medicine, New York, New York
| | - Salma Adam
- S.A. Localio Laboratory, Department of Surgery, New York University School of Medicine, New York, New York
| | - Muhammad Israr
- S.A. Localio Laboratory, Department of Surgery, New York University School of Medicine, New York, New York
| | - Maeliss Gelas
- S.A. Localio Laboratory, Department of Surgery, New York University School of Medicine, New York, New York
| | - Justin Lish
- S.A. Localio Laboratory, Department of Surgery, New York University School of Medicine, New York, New York
| | - Kathryn Chin
- S.A. Localio Laboratory, Department of Surgery, New York University School of Medicine, New York, New York
| | - Mohammad Saad Farooq
- S.A. Localio Laboratory, Department of Surgery, New York University School of Medicine, New York, New York
| | - Benjamin Wadowski
- S.A. Localio Laboratory, Department of Surgery, New York University School of Medicine, New York, New York
| | - Jingjing Wu
- S.A. Localio Laboratory, Department of Surgery, New York University School of Medicine, New York, New York
| | - Suhagi Shah
- Department of Medicine, New York University School of Medicine, New York, New York
| | - Dennis O Adeegbe
- Department of Medicine, New York University School of Medicine, New York, New York
| | - Smruti Pushalkar
- Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, New York
| | | | - Deepak Saxena
- Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, New York
| | - Kwok-Kin Wong
- Department of Medicine, New York University School of Medicine, New York, New York
| | - Lisa M Coussens
- Department of Cell, Developmental and Cancer Biology, Oregon Health and Science University, Portland, Oregon.,Brenden-Colson Center for Pancreatic Care, Oregon Health and Science University, Portland, Oregon.,Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon
| | - George Miller
- S.A. Localio Laboratory, Department of Surgery, New York University School of Medicine, New York, New York. .,Department of Cell Biology, New York University School of Medicine, New York, New York
| |
Collapse
|
17
|
Heredia JD, Park J, Brubaker RJ, Szymanski SK, Gill KS, Procko E. Mapping Interaction Sites on Human Chemokine Receptors by Deep Mutational Scanning. THE JOURNAL OF IMMUNOLOGY 2018; 200:3825-3839. [PMID: 29678950 DOI: 10.4049/jimmunol.1800343] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 03/30/2018] [Indexed: 02/02/2023]
Abstract
Chemokine receptors CXCR4 and CCR5 regulate WBC trafficking and are engaged by the HIV-1 envelope glycoprotein gp120 during infection. We combine a selection of human CXCR4 and CCR5 libraries comprising nearly all of ∼7000 single amino acid substitutions with deep sequencing to define sequence-activity landscapes for surface expression and ligand interactions. After consideration of sequence constraints for surface expression, known interaction sites with HIV-1-blocking Abs were appropriately identified as conserved residues following library sorting for Ab binding, validating the use of deep mutational scanning to map functional interaction sites in G protein-coupled receptors. Chemokine CXCL12 was found to interact with residues extending asymmetrically into the CXCR4 ligand-binding cavity, similar to the binding surface of CXCR4 recognized by an antagonistic viral chemokine previously observed crystallographically. CXCR4 mutations distal from the chemokine binding site were identified that enhance chemokine recognition. This included disruptive mutations in the G protein-coupling site that diminished calcium mobilization, as well as conservative mutations to a membrane-exposed site (CXCR4 residues H792.45 and W1614.50) that increased ligand binding without loss of signaling. Compared with CXCR4-CXCL12 interactions, CCR5 residues conserved for gp120 (HIV-1 BaL strain) interactions map to a more expansive surface, mimicking how the cognate chemokine CCL5 makes contacts across the entire CCR5 binding cavity. Acidic substitutions in the CCR5 N terminus and extracellular loops enhanced gp120 binding. This study demonstrates how comprehensive mutational scanning can define functional interaction sites on receptors, and novel mutations that enhance receptor activities can be found simultaneously.
Collapse
Affiliation(s)
- Jeremiah D Heredia
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801
| | - Jihye Park
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801
| | - Riley J Brubaker
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801
| | - Steven K Szymanski
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801
| | - Kevin S Gill
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801
| | - Erik Procko
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801
| |
Collapse
|
18
|
Samson A, Scott KJ, Taggart D, West EJ, Wilson E, Nuovo GJ, Thomson S, Corns R, Mathew RK, Fuller MJ, Kottke TJ, Thompson JM, Ilett EJ, Cockle JV, van Hille P, Sivakumar G, Polson ES, Turnbull SJ, Appleton ES, Migneco G, Rose AS, Coffey MC, Beirne DA, Collinson FJ, Ralph C, Alan Anthoney D, Twelves CJ, Furness AJ, Quezada SA, Wurdak H, Errington-Mais F, Pandha H, Harrington KJ, Selby PJ, Vile RG, Griffin SD, Stead LF, Short SC, Melcher AA. Intravenous delivery of oncolytic reovirus to brain tumor patients immunologically primes for subsequent checkpoint blockade. Sci Transl Med 2018; 10:eaam7577. [PMID: 29298869 PMCID: PMC6276984 DOI: 10.1126/scitranslmed.aam7577] [Citation(s) in RCA: 290] [Impact Index Per Article: 41.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 09/08/2017] [Accepted: 11/27/2017] [Indexed: 12/23/2022]
Abstract
Immune checkpoint inhibitors, including those targeting programmed cell death protein 1 (PD-1), are reshaping cancer therapeutic strategies. Evidence suggests, however, that tumor response and patient survival are determined by tumor programmed death ligand 1 (PD-L1) expression. We hypothesized that preconditioning of the tumor immune microenvironment using targeted, virus-mediated interferon (IFN) stimulation would up-regulate tumor PD-L1 protein expression and increase cytotoxic T cell infiltration, improving the efficacy of subsequent checkpoint blockade. Oncolytic viruses (OVs) represent a promising form of cancer immunotherapy. For brain tumors, almost all studies to date have used direct intralesional injection of OV, because of the largely untested belief that intravenous administration will not deliver virus to this site. We show, in a window-of-opportunity clinical study, that intravenous infusion of oncolytic human Orthoreovirus (referred to herein as reovirus) leads to infection of tumor cells subsequently resected as part of standard clinical care, both in high-grade glioma and in brain metastases, and increases cytotoxic T cell tumor infiltration relative to patients not treated with virus. We further show that reovirus up-regulates IFN-regulated gene expression, as well as the PD-1/PD-L1 axis in tumors, via an IFN-mediated mechanism. Finally, we show that addition of PD-1 blockade to reovirus enhances systemic therapy in a preclinical glioma model. These results support the development of combined systemic immunovirotherapy strategies for the treatment of both primary and secondary tumors in the brain.
Collapse
Affiliation(s)
- Adel Samson
- Leeds Institute of Cancer and Pathology, Faculty of Medicine and Health, University of Leeds, St James's University Hospital, Beckett Street, Leeds, West Yorkshire LS9 7TF, UK.
| | - Karen J Scott
- Leeds Institute of Cancer and Pathology, Faculty of Medicine and Health, University of Leeds, St James's University Hospital, Beckett Street, Leeds, West Yorkshire LS9 7TF, UK
| | - David Taggart
- Leeds Institute of Cancer and Pathology, Faculty of Medicine and Health, University of Leeds, St James's University Hospital, Beckett Street, Leeds, West Yorkshire LS9 7TF, UK
| | - Emma J West
- Leeds Institute of Cancer and Pathology, Faculty of Medicine and Health, University of Leeds, St James's University Hospital, Beckett Street, Leeds, West Yorkshire LS9 7TF, UK
| | - Erica Wilson
- Leeds Institute of Cancer and Pathology, Faculty of Medicine and Health, University of Leeds, St James's University Hospital, Beckett Street, Leeds, West Yorkshire LS9 7TF, UK
| | - Gerard J Nuovo
- Ohio State University, Comprehensive Cancer Centre, Columbus, OH 43210, USA
| | - Simon Thomson
- Leeds Teaching Hospitals National Health Service Trust, St James's University Hospital, Beckett Street, Leeds, West Yorkshire LS9 7TF, UK
| | - Robert Corns
- Leeds Teaching Hospitals National Health Service Trust, St James's University Hospital, Beckett Street, Leeds, West Yorkshire LS9 7TF, UK
| | - Ryan K Mathew
- Leeds Institute of Cancer and Pathology, Faculty of Medicine and Health, University of Leeds, St James's University Hospital, Beckett Street, Leeds, West Yorkshire LS9 7TF, UK
| | - Martin J Fuller
- Leeds Institute of Cancer and Pathology, Faculty of Medicine and Health, University of Leeds, St James's University Hospital, Beckett Street, Leeds, West Yorkshire LS9 7TF, UK
| | | | - Jill M Thompson
- Department of Immunology, Mayo Clinic, Rochester, MN 55905, USA
| | - Elizabeth J Ilett
- Leeds Institute of Cancer and Pathology, Faculty of Medicine and Health, University of Leeds, St James's University Hospital, Beckett Street, Leeds, West Yorkshire LS9 7TF, UK
| | - Julia V Cockle
- Leeds Institute of Cancer and Pathology, Faculty of Medicine and Health, University of Leeds, St James's University Hospital, Beckett Street, Leeds, West Yorkshire LS9 7TF, UK
| | - Philip van Hille
- Leeds Teaching Hospitals National Health Service Trust, St James's University Hospital, Beckett Street, Leeds, West Yorkshire LS9 7TF, UK
| | - Gnanamurthy Sivakumar
- Leeds Teaching Hospitals National Health Service Trust, St James's University Hospital, Beckett Street, Leeds, West Yorkshire LS9 7TF, UK
| | - Euan S Polson
- Leeds Institute of Cancer and Pathology, Faculty of Medicine and Health, University of Leeds, St James's University Hospital, Beckett Street, Leeds, West Yorkshire LS9 7TF, UK
| | - Samantha J Turnbull
- Leeds Institute of Cancer and Pathology, Faculty of Medicine and Health, University of Leeds, St James's University Hospital, Beckett Street, Leeds, West Yorkshire LS9 7TF, UK
| | - Elizabeth S Appleton
- Leeds Institute of Cancer and Pathology, Faculty of Medicine and Health, University of Leeds, St James's University Hospital, Beckett Street, Leeds, West Yorkshire LS9 7TF, UK
| | - Gemma Migneco
- Leeds Institute of Cancer and Pathology, Faculty of Medicine and Health, University of Leeds, St James's University Hospital, Beckett Street, Leeds, West Yorkshire LS9 7TF, UK
| | - Ailsa S Rose
- Leeds Institute of Cancer and Pathology, Faculty of Medicine and Health, University of Leeds, St James's University Hospital, Beckett Street, Leeds, West Yorkshire LS9 7TF, UK
| | | | - Deborah A Beirne
- Leeds Teaching Hospitals National Health Service Trust, St James's University Hospital, Beckett Street, Leeds, West Yorkshire LS9 7TF, UK
| | - Fiona J Collinson
- Leeds Institute of Clinical Trials Research, Faculty of Medicine and Health, University of Leeds, Leeds, West Yorkshire LS2 9JT, UK
| | - Christy Ralph
- Leeds Institute of Cancer and Pathology, Faculty of Medicine and Health, University of Leeds, St James's University Hospital, Beckett Street, Leeds, West Yorkshire LS9 7TF, UK
| | - D Alan Anthoney
- Leeds Institute of Cancer and Pathology, Faculty of Medicine and Health, University of Leeds, St James's University Hospital, Beckett Street, Leeds, West Yorkshire LS9 7TF, UK
| | - Christopher J Twelves
- Leeds Institute of Cancer and Pathology, Faculty of Medicine and Health, University of Leeds, St James's University Hospital, Beckett Street, Leeds, West Yorkshire LS9 7TF, UK
| | | | | | - Heiko Wurdak
- Leeds Institute of Cancer and Pathology, Faculty of Medicine and Health, University of Leeds, St James's University Hospital, Beckett Street, Leeds, West Yorkshire LS9 7TF, UK
| | - Fiona Errington-Mais
- Leeds Institute of Cancer and Pathology, Faculty of Medicine and Health, University of Leeds, St James's University Hospital, Beckett Street, Leeds, West Yorkshire LS9 7TF, UK
| | | | | | - Peter J Selby
- Leeds Institute of Cancer and Pathology, Faculty of Medicine and Health, University of Leeds, St James's University Hospital, Beckett Street, Leeds, West Yorkshire LS9 7TF, UK
| | - Richard G Vile
- Department of Immunology, Mayo Clinic, Rochester, MN 55905, USA
| | - Stephen D Griffin
- Leeds Institute of Cancer and Pathology, Faculty of Medicine and Health, University of Leeds, St James's University Hospital, Beckett Street, Leeds, West Yorkshire LS9 7TF, UK
| | - Lucy F Stead
- Leeds Institute of Cancer and Pathology, Faculty of Medicine and Health, University of Leeds, St James's University Hospital, Beckett Street, Leeds, West Yorkshire LS9 7TF, UK
| | - Susan C Short
- Leeds Institute of Cancer and Pathology, Faculty of Medicine and Health, University of Leeds, St James's University Hospital, Beckett Street, Leeds, West Yorkshire LS9 7TF, UK.
| | - Alan A Melcher
- Institute of Cancer Research, 123 Old Brompton Road, London SW7 3RP, UK.
| |
Collapse
|
19
|
Venuti A, Pastori C, Lopalco L. The Role of Natural Antibodies to CC Chemokine Receptor 5 in HIV Infection. Front Immunol 2017; 8:1358. [PMID: 29163468 PMCID: PMC5670346 DOI: 10.3389/fimmu.2017.01358] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Accepted: 10/03/2017] [Indexed: 12/22/2022] Open
Abstract
The CC chemokine receptor 5 (CCR5) is responsible for immune and inflammatory responses by mediation of chemotactic activity in leukocytes, although it is expressed on different cell types. It has been shown to act as co-receptor for the human and simian immunodeficiency viruses (HIV-1, HIV-2, and SIV). Natural reactive antibodies (Abs) recognizing first loop (ECL1) of CCR5 have been detected in several pools of immunoglobulins from healthy donors and from several cohorts of either HIV-exposed but uninfected subjects (ESN) or HIV-infected individuals who control disease progression (LTNP) as well. The reason of development of anti-CCR5 Abs in the absence of autoimmune disease is still unknown; however, the presence of these Abs specific for CCR5 or for other immune receptors and mediators probably is related to homeostasis maintenance. The majority of anti-CCR5 Abs is directed to HIV binding site (N-terminus and ECL2) of the receptor. Conversely, it is well known that ECL1 of CCR5 does not bind HIV; thus, the anti-CCR5 Abs directed to ECL1 elicit a long-lasting internalization of CCR5 but not interfere with HIV binding directly; these Abs block HIV infection in either epithelial cells or CD4+ T lymphocytes and the mechanism differs from those ones described for all other CCR5-specific ligands. The Ab-mediated CCR5 internalization allows the formation of a stable signalosome by interaction of CCR5, β-arrestin2 and ERK1 proteins. The signalosome degradation and the subsequent de novo proteins synthesis determine the CCR5 reappearance on the cell membrane with a very long-lasting kinetics (8 days). The use of monoclonal Abs to CCR5 with particular characteristics and mode of action may represent a novel mode to fight viral infection in either vaccinal or therapeutic strategies.
Collapse
Affiliation(s)
- Assunta Venuti
- Division of Immunology, Transplantation and Infectious Diseases, DIBIT - San Raffaele Scientific Institute, Milan, Italy
| | - Claudia Pastori
- Division of Immunology, Transplantation and Infectious Diseases, DIBIT - San Raffaele Scientific Institute, Milan, Italy
| | - Lucia Lopalco
- Division of Immunology, Transplantation and Infectious Diseases, DIBIT - San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
20
|
Sun Z, Yan L, Tang J, Qian Q, Lenberg J, Zhu D, Liu W, Wu K, Wang Y, Lu S. Brief introduction of current technologies in isolation of broadly neutralizing HIV-1 antibodies. Virus Res 2017; 243:75-82. [PMID: 29051051 PMCID: PMC7114535 DOI: 10.1016/j.virusres.2017.10.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Revised: 10/13/2017] [Accepted: 10/15/2017] [Indexed: 12/11/2022]
Abstract
HIV/AIDS has become a worldwide pandemic. Before an effective HIV-1 vaccine eliciting broadly neutralizing monoclonal antibodies (bnmAbs) is fully developed, passive immunization for prevention and treatment of HIV-1 infection may alleviate the burden caused by the pandemic. Among HIV-1 infected individuals, about 20% of them generated cross-reactive neutralizing antibodies two to four years after infection, the details of which could provide knowledge for effective vaccine design. Recent progress in techniques for isolation of human broadly neutralizing antibodies has facilitated the study of passive immunization. The isolation and characterization of large panels of potent human broadly neutralizing antibodies has revealed new insights into the principles of antibody-mediated neutralization of HIV. In this paper, we review the current effective techniques in broadly neutralizing antibody isolation.
Collapse
Affiliation(s)
- Zehua Sun
- Department of Medicine, National Jewish Health, 1400 Jackson Street, Denver, CO, 80206, United States.
| | - Lixin Yan
- Harbin Medical University Affiliated 2nd Hospital, 246 Xuefu Road, Harbin, 150086, China.
| | - Jiansong Tang
- Department of Technical Specialist, China Bioengineering Technology Group Limited, Unit 209,Building 16W, Hong Kong Science Park, Shatin, NT, HK, 999077, Hong Kong
| | - Qian Qian
- Department of Medicine, National Jewish Health, 1400 Jackson Street, Denver, CO, 80206, United States
| | - Jerica Lenberg
- Department of Medicine, National Jewish Health, 1400 Jackson Street, Denver, CO, 80206, United States; Augustana University, 2001 S Summit Avenue, Sioux Falls, SD, 571977, United States
| | - Dandan Zhu
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center, Houston, TX, 77030, United States
| | - Wan Liu
- Harbin Medical University Affiliated 2nd Hospital, 246 Xuefu Road, Harbin, 150086, China
| | - Kao Wu
- Glyn O. Philips Hydrocolloid Research Center at HUT, Hubei University of Technology, Wuhan 430068, China
| | - Yilin Wang
- University of California, Irvine. 100 Pacific, Irvine, CA, 92618, United States
| | - Shiqiang Lu
- AIDS Institute, Faculty of Medicine, The University of Hong Kong, No21 Sassoon Road, 999077, Hong Kong, Hong Kong.
| |
Collapse
|
21
|
Tan Y, Tong P, Wang J, Zhao L, Li J, Yu Y, Chen YH, Wang J. The Membrane-Proximal Region of C-C Chemokine Receptor Type 5 Participates in the Infection of HIV-1. Front Immunol 2017; 8:478. [PMID: 28484468 PMCID: PMC5402540 DOI: 10.3389/fimmu.2017.00478] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Accepted: 04/05/2017] [Indexed: 01/22/2023] Open
Abstract
The initial infection and transmission of HIV-1 requires C-C chemokine receptor type 5 (CCR5). Here, we report that the membrane-proximal region (MPR, aa 22-38) of CCR5 participates in the infection of HIV-1. First, MPR-specific antibodies elicited in mice dose-dependently inhibited the infection of CCR5-tropic HIV-1. Second, substituting MPR with the same region from other co-receptors significantly impaired HIV-1 infection, while the key residues identified by alanine scanning mutagenesis formed an exposed leucine zipper-like structure. Moreover, a peptide derived from MPR could block the infection of a number of HIV-1 strains only before the formation of gp41 six-helix bundle, coincide with the early interaction between CCR5 and the gp120 protein during HIV-1 infection. These promising results ensured the potential of this previously uncharacterized domain as a starting point for the development of antiviral drugs, blocking antibodies, and HIV vaccines.
Collapse
Affiliation(s)
- Yue Tan
- Laboratory of Immunology, School of Life Sciences, Beijing Key Laboratory for Protein Therapeutics, Protein Science Laboratory of the Ministry of Education, Tsinghua University, Beijing, China
| | - Pei Tong
- Laboratory of Immunology, School of Life Sciences, Beijing Key Laboratory for Protein Therapeutics, Protein Science Laboratory of the Ministry of Education, Tsinghua University, Beijing, China
| | - Junyi Wang
- Laboratory of Immunology, School of Life Sciences, Beijing Key Laboratory for Protein Therapeutics, Protein Science Laboratory of the Ministry of Education, Tsinghua University, Beijing, China
| | - Lei Zhao
- The Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology of Ministry of Education, Department of Chemistry, Tsinghua University, Beijing, China
| | - Jing Li
- Laboratory of Immunology, School of Life Sciences, Beijing Key Laboratory for Protein Therapeutics, Protein Science Laboratory of the Ministry of Education, Tsinghua University, Beijing, China
| | - Yang Yu
- Laboratory of Immunology, School of Life Sciences, Beijing Key Laboratory for Protein Therapeutics, Protein Science Laboratory of the Ministry of Education, Tsinghua University, Beijing, China
| | - Ying-Hua Chen
- Laboratory of Immunology, School of Life Sciences, Beijing Key Laboratory for Protein Therapeutics, Protein Science Laboratory of the Ministry of Education, Tsinghua University, Beijing, China
| | - Ji Wang
- Laboratory of Immunology, School of Life Sciences, Beijing Key Laboratory for Protein Therapeutics, Protein Science Laboratory of the Ministry of Education, Tsinghua University, Beijing, China.,Wellman Center for Photomedicine, Massachusetts General Hospital, Department of Dermatology, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
22
|
Possible Impact of 190G > A CCR2 and Δ32 CCR5 Mutations on Decrease of the HBV Vaccine Immunogenicity-A Preliminary Report. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2017; 14:ijerph14020166. [PMID: 28208753 PMCID: PMC5334720 DOI: 10.3390/ijerph14020166] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Revised: 01/04/2017] [Accepted: 01/05/2017] [Indexed: 12/14/2022]
Abstract
Background: Chemokine genetic variations are involved in infectious diseases such as hepatitis B virus (HBV). Several allelic variants might, in theory, affect the outcome of vaccination. Objectives: This study was carried out to examine the associations of Δ32 CCR5 and 190G > A CCR2 polymorphisms with a response to a primary course of three HBV vaccinations. Methods: Between December 2014 and December 2016, patients from three randomly selected primary care clinics in the West Pomeranian region (Poland), 1 month after receiving the third dose of HBV vaccine, were enrolled. Enzyme-linked immunosorbent assay (ELISA) system version 3.0 was used to detect anti-HBs and anti-HBc totals. The identification of polymorphisms were performed by a polymerase chain reaction technique using a single primer extension assay. Genotype distributions of responders versus non-responders to HBV vaccination were compared on the basis of anti-HBs level. Results: In 149 patients (mean age 60 years) the mean anti-HBs level was 652.2 ± 425.9 mIU/mL (range: 0–1111.0 mIU/mL). There were 14.1% (n = 21) non-responders to the HBV vaccine (anti-HBs < 10.0 mIU/mL). The wild type/Δ32 genotype of CCR5 gene was found in 18.1% participants, and 1.3% were Δ32/Δ32 homozygotes. The frequency of allele A of the CCR2 gene was 11.1%. Lower anti-HBs levels in Δ32/Δ32 homozygotes were observed (Me = 61 mIU/mL vs. Me = 660.2 mIU/mL; p = 0.048). As age was found to be a correlate to the anti-HBs titer (r = −0.218, p = 0.0075; 95% CI: −0.366–−0.059)—an analysis of a co-variance was performed which found a statistically significant (p = 0.04) difference in anti-HBs titres between Δ32/Δ32 homozygotes and other CCR5 genotypes. The association between anti-HBs titres and CCR2 genotypes was not statistically significant. Conclusions: Our study—which is a preliminary report that suggest this topic deserves further observation with larger sample sizes, different ethnicities, and other single nucleotide poly-morphisms (SNPs)—suggests the possible involvement of CCR5 polymorphism in impairing the immunologic response to HBV vaccination, predominantly in relation to the passage of time.
Collapse
|
23
|
Park RJ, Wang T, Koundakjian D, Hultquist JF, Lamothe-Molina P, Monel B, Schumann K, Yu H, Krupzcak KM, Garcia-Beltran W, Piechocka-Trocha A, Krogan NJ, Marson A, Sabatini DM, Lander ES, Hacohen N, Walker BD. A genome-wide CRISPR screen identifies a restricted set of HIV host dependency factors. Nat Genet 2016; 49:193-203. [PMID: 27992415 DOI: 10.1038/ng.3741] [Citation(s) in RCA: 244] [Impact Index Per Article: 27.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 11/15/2016] [Indexed: 12/14/2022]
Abstract
Host proteins are essential for HIV entry and replication and can be important nonviral therapeutic targets. Large-scale RNA interference (RNAi)-based screens have identified nearly a thousand candidate host factors, but there is little agreement among studies and few factors have been validated. Here we demonstrate that a genome-wide CRISPR-based screen identifies host factors in a physiologically relevant cell system. We identify five factors, including the HIV co-receptors CD4 and CCR5, that are required for HIV infection yet are dispensable for cellular proliferation and viability. Tyrosylprotein sulfotransferase 2 (TPST2) and solute carrier family 35 member B2 (SLC35B2) function in a common pathway to sulfate CCR5 on extracellular tyrosine residues, facilitating CCR5 recognition by the HIV envelope. Activated leukocyte cell adhesion molecule (ALCAM) mediates cell aggregation, which is required for cell-to-cell HIV transmission. We validated these pathways in primary human CD4+ T cells through Cas9-mediated knockout and antibody blockade. Our findings indicate that HIV infection and replication rely on a limited set of host-dispensable genes and suggest that these pathways can be studied for therapeutic intervention.
Collapse
Affiliation(s)
- Ryan J Park
- Ragon Institute of Massachusetts General Hospital (MGH), Massachusetts Institute of Technology (MIT), and Harvard University, Cambridge, Massachusetts, USA.,Harvard-MIT Health Sciences and Technology, Harvard Medical School, Boston, Massachusetts, USA.,Broad Institute of MIT and Harvard University, Cambridge, Massachusetts, USA
| | - Tim Wang
- Broad Institute of MIT and Harvard University, Cambridge, Massachusetts, USA.,Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA.,Whitehead Institute for Biomedical Research, Cambridge, Massachusetts, USA.,David H. Koch Institute for Integrative Cancer Research at MIT, Cambridge, Massachusetts, USA
| | - Dylan Koundakjian
- Ragon Institute of Massachusetts General Hospital (MGH), Massachusetts Institute of Technology (MIT), and Harvard University, Cambridge, Massachusetts, USA
| | - Judd F Hultquist
- Department of Cellular and Molecular Pharmacology, California Institute for Quantitative Biosciences, QB3, University of California at San Francisco (UCSF), San Francisco, California, USA.,Gladstone Institute of Virology and Immunology, J. David Gladstone Institutes, San Francisco, California, USA
| | - Pedro Lamothe-Molina
- Ragon Institute of Massachusetts General Hospital (MGH), Massachusetts Institute of Technology (MIT), and Harvard University, Cambridge, Massachusetts, USA.,Biological Sciences in Public Health, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Blandine Monel
- Ragon Institute of Massachusetts General Hospital (MGH), Massachusetts Institute of Technology (MIT), and Harvard University, Cambridge, Massachusetts, USA.,Howard Hughes Medical Institute, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Kathrin Schumann
- Department of Microbiology and Immunology, University of California at San Francisco, San Francisco, California, USA
| | - Haiyan Yu
- Broad Institute of MIT and Harvard University, Cambridge, Massachusetts, USA
| | - Kevin M Krupzcak
- Whitehead Institute for Biomedical Research, Cambridge, Massachusetts, USA
| | - Wilfredo Garcia-Beltran
- Ragon Institute of Massachusetts General Hospital (MGH), Massachusetts Institute of Technology (MIT), and Harvard University, Cambridge, Massachusetts, USA.,Harvard-MIT Health Sciences and Technology, Harvard Medical School, Boston, Massachusetts, USA
| | - Alicja Piechocka-Trocha
- Ragon Institute of Massachusetts General Hospital (MGH), Massachusetts Institute of Technology (MIT), and Harvard University, Cambridge, Massachusetts, USA
| | - Nevan J Krogan
- Department of Cellular and Molecular Pharmacology, California Institute for Quantitative Biosciences, QB3, University of California at San Francisco (UCSF), San Francisco, California, USA.,Gladstone Institute of Virology and Immunology, J. David Gladstone Institutes, San Francisco, California, USA
| | - Alexander Marson
- Department of Microbiology and Immunology, University of California at San Francisco, San Francisco, California, USA.,Diabetes Center, University of California at San Francisco, San Francisco, California, USA.,Department of Medicine, University of California at San Francisco, San Francisco, California, USA.,Innovative Genomics Initiative (IGI), University of California, Berkeley, Berkeley, California, USA.,UCSF Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, San Francisco, California, USA
| | - David M Sabatini
- Broad Institute of MIT and Harvard University, Cambridge, Massachusetts, USA.,Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA.,Whitehead Institute for Biomedical Research, Cambridge, Massachusetts, USA.,David H. Koch Institute for Integrative Cancer Research at MIT, Cambridge, Massachusetts, USA.,Howard Hughes Medical Institute, Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Eric S Lander
- Broad Institute of MIT and Harvard University, Cambridge, Massachusetts, USA.,Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA.,Department of Systems Biology, Harvard Medical School, Boston, Massachusetts, USA
| | - Nir Hacohen
- Broad Institute of MIT and Harvard University, Cambridge, Massachusetts, USA.,Massachusetts General Hospital Cancer Center, Boston, Massachusetts, USA
| | - Bruce D Walker
- Ragon Institute of Massachusetts General Hospital (MGH), Massachusetts Institute of Technology (MIT), and Harvard University, Cambridge, Massachusetts, USA.,Broad Institute of MIT and Harvard University, Cambridge, Massachusetts, USA.,Howard Hughes Medical Institute, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA.,Institute of Medical Engineering and Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| |
Collapse
|
24
|
Tian H, Fürstenberg A, Huber T. Labeling and Single-Molecule Methods To Monitor G Protein-Coupled Receptor Dynamics. Chem Rev 2016; 117:186-245. [DOI: 10.1021/acs.chemrev.6b00084] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Affiliation(s)
- He Tian
- Laboratory of Chemical Biology
and Signal Transduction, The Rockefeller University, 1230 York
Avenue, New York, New York 10065, United States
| | - Alexandre Fürstenberg
- Laboratory of Chemical Biology
and Signal Transduction, The Rockefeller University, 1230 York
Avenue, New York, New York 10065, United States
| | - Thomas Huber
- Laboratory of Chemical Biology
and Signal Transduction, The Rockefeller University, 1230 York
Avenue, New York, New York 10065, United States
| |
Collapse
|
25
|
Abstract
Objectives: Semen composition is influenced by HIV-1 infection, yet the impact of semen components on HIV infection of primary target cells has only been studied in samples from HIV-uninfected donors. Design: We compared the effect of seminal plasma (SP) from chronically HIV-infected (SP+) versus uninfected donors (SP–) on HIV-1 infection of peripheral blood mononuclear cells (PBMCs) and CD4+ T cells. Methods: Primary cells were infected with HIV-1 in the presence of SP+ or SP– and analyzed for infection level, metabolic activity, HIV receptor expression, proliferation and activation. SP+ and SP– were compared for infection-enhancing peptides, cytokines and prostaglandin E2 levels. Results: SP– efficiently enhanced HIV-1 R5 infection of CD4+ T cells, whereas SP+ enhancing activity was significantly reduced. RANTES (CCL5) concentrations were elevated in SP+ relative to SP–, whereas the concentrations of infectivity-enhancing peptides [semen-derived enhancer of viral infection (SEVI), SEM1, SEM2] were similar. CCR5 membrane expression levels were reduced on CD4+ T cells shortly postexposure to SP+ compared with SP– and correlated to R5-tropic HIV-1 infection levels, and CCR5 ligands’ concentrations in semen. SP+ and SP– displayed similar enhancing activity on PBMC infection by X4-tropic HIV-1. Addition/depletion of RANTES (regulated on activation, normal T-cell expressed and secreted) from SPs modulated their effect on PBMC infection by R5-tropic HIV-1. Conclusion: Semen from HIV-infected donors exhibits a significantly reduced enhancing potential on CD4+ T-cell infection by R5-tropic HIV-1 when compared with semen from uninfected donors. Our data indicate that elevated seminal concentrations of RANTES in HIV-infected men can influence the ability of semen to enhance infection.
Collapse
|
26
|
Pacheco-Martínez E, Figueroa-Medina E, Villarreal C, Cocho G, Medina-Franco JL, Méndez-Lucio O, Huerta L. Statistical correlation of nonconservative substitutions of HIV gp41 variable amino acid residues with the R5X4 HIV-1 phenotype. Virol J 2016; 13:28. [PMID: 26879054 PMCID: PMC4754869 DOI: 10.1186/s12985-016-0486-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Accepted: 02/09/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The interaction of the envelope glycoprotein of HIV-1 (gp120/gp41) with coreceptor molecules has important implications for specific cellular targeting and pathogenesis. Experimental and theoretical evidences have shown a role for gp41 in coreceptor tropism, although there is no consensus about the positions involved. Here we analyze the association of physicochemical properties of gp41 amino acid residues with viral tropism (X4, R5, and R5X4) using a large set of HIV-1 sequences. Under the assumption that conserved regions define the complex structural features essential for protein function, we focused our search only on amino acids in the gp41 variable regions. METHODS Gp41 amino acid sequences of 2823 HIV-1 strains from all clades with known coreceptor tropism were retrieved from Los Alamos HIV Database. Consensus sequences were constructed for homologous sequences (those obtained from the same patient and having the same tropism) in order to avoid bias due to sequence overrepresentation, and the variability (entropy) per site was determined. Comparisons of hydropathy index (HI) and charge (Q) of amino acid residues at highly variable positions between coreceptor groups were performed using two non-parametrical tests and Benjamini-Hochberg correction. Pearson's correlation analysis was performed to determine covariance of HI and Q values. RESULTS Calculation of variability per site rendered 58 highly variable amino acid positions. Of these, statistical analysis rendered significantly different HI or Q only for the R5 vs. R5X4 comparison at twelve positions: 535, 602, 619, 636, 640, 641, 658, 662, 667, 723, 756 and 841. The largest differences in particular amino acid frequencies between coreceptor groups were found at 619, 636, 640, 641, 662, 723 and 756. A hydrophobic tendency of residues 619, 640, 641, 723 and 756, along with a hydrophilic/charged tendency at residues 636 and 662 was observed in R5X4 with respect to R5 sequences. HI of position 640 covariated with that of 602, 619, 636, 662, and 756. CONCLUSIONS Variability and significant correlations of physicochemical properties with viral phenotype suggest that substitutions at residues in the loop (602 and 619), the HR2 (636, 640, 641, 662), and the C-terminal tail (723, 756) of gp41 may contribute to phenotype of R5X4 strains.
Collapse
Affiliation(s)
- Elena Pacheco-Martínez
- Programa de Maestría y Doctorado en Ciencias Bioquímicas, Universidad Nacional Autónoma de México, Ciudad Universitaria, Distrito Federal, 04510, México
| | - Evangelina Figueroa-Medina
- Unidad de Radio Oncología, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Secretaría de Salud, Avenida Vasco de Quiroga No.15, ᅟDistrito Federal, 14080, México
| | - Carlos Villarreal
- Departmento de Física Teórica, Instituto de Física, Universidad Nacional Autónoma de México, Ciudad Universitaria, ᅟDistrito Federal, 04510, México.,Centro de Ciencias de la Complejidad, Universidad Nacional Autónoma de México, Ciudad Univesitaria, ᅟDistrito Federal, 04510, México
| | - Germinal Cocho
- Departmento de Sistemas Complejos, Instituto de Física, Universidad Nacional Autónoma de México, Ciudad Universitaria, ᅟDistrito Federal, 04510, México.,Centro de Ciencias de la Complejidad, Universidad Nacional Autónoma de México, Ciudad Univesitaria, ᅟDistrito Federal, 04510, México
| | - José L Medina-Franco
- Departmento de Farmacia, Facultad de Química, Universidad Nacional Autónoma de México, Ciudad Universitaria, ᅟDistrito Federal, 04510, México
| | - Oscar Méndez-Lucio
- Departmento de Farmacia, Facultad de Química, Universidad Nacional Autónoma de México, Ciudad Universitaria, ᅟDistrito Federal, 04510, México
| | - Leonor Huerta
- Departmento de Immunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad Universitaria, 04510, Distrito Federal, México.
| |
Collapse
|
27
|
Platt EJ, Durnin JP, Kabat D. Short Communication: HIV-1 Variants That Use Mouse CCR5 Reveal Critical Interactions of gp120's V3 Crown with CCR5 Extracellular Loop 1. AIDS Res Hum Retroviruses 2015; 31:992-8. [PMID: 26114311 PMCID: PMC4576943 DOI: 10.1089/aid.2015.0131] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
The CCR5 coreceptor amino terminus and extracellular (ECL) loops 1 and 2 have been implicated in HIV-1 infections, with species differences in these regions inhibiting zoonoses. Interactions of gp120 with CD4 and CCR5 reduce constraints on metastable envelope subunit gp41, enabling gp41 conformational changes needed for infection. We previously selected HIV-1JRCSF variants that efficiently use CCR5(Δ18) with a deleted amino terminus or CCR5(HHMH) with ECL2 from an NIH/Swiss mouse. Unexpectedly, the adaptive gp120 mutations were nearly identical, suggesting that they function by weakening gp120's grip on gp41 and/or by increasing interactions with ECL1. To analyze this and further wean HIV-1 from human CCR5, we selected variants using CCR5(HMMH) with murine ECL1 and 2 sequences. HIV-1JRCSF mutations adaptive for CCR5(Δ18) and CCR5(HHMH) were generally maladaptive for CCR5(HMMH), whereas the converse was true for CCR5(HMMH) adaptations. The HIV-1JRCSF variant adapted to CCR5(HMMH) also weakly used intact NIH/Swiss mouse CCR5. Our results strongly suggest that HIV-1JRCSF makes functionally critical contacts with human ECL1 and that adaptation to murine ECL1 requires multiple mutations in the crown of gp120's V3 loop.
Collapse
Affiliation(s)
- Emily J. Platt
- Department of Biochemistry and Molecular Biology, Oregon Health and Science University, Portland, Oregon
| | - James P. Durnin
- Department of Biochemistry and Molecular Biology, Oregon Health and Science University, Portland, Oregon
| | - David Kabat
- Department of Biochemistry and Molecular Biology, Oregon Health and Science University, Portland, Oregon
| |
Collapse
|
28
|
Lee YH, Kim JH, Song GG. Chemokine receptor 5 Δ32 polymorphism and systemic lupus erythematosus, vasculitis, and primary Sjogren's syndrome. Meta-analysis of possible associations. Z Rheumatol 2015; 73:848-55. [PMID: 24599359 DOI: 10.1007/s00393-014-1356-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
OBJECTIVE The aim of this study was to determine whether the functional chemokine receptor 5 delta32 (CCR5-Δ32) polymorphism is associated with susceptibility to systemic lupus erythematosus (SLE), vasculitis, and primary Sjogren's syndrome (pSS). RESULTS A total of 12 studies were analyzed, including 5 on SLE, 5 on vasculitis, and 2 on pSS, encompassing 1881 patients and 2391 controls. Meta-analysis indicated no association between SLE and the CCR5-Δ32 allele (OR 0.842, 95 % CI 0.793-1.804, p = 0.657), and no association between the CCR5-Δ32 allele and SLE in Europeans (OR 0.647, 95 % CI 0.306-1.368, p = 0.255). Meta-analysis of the CCR5-Δ32 allele and the Δ32Δ32 + Δ32 W genotype showed no association with lupus nephritis (LN; OR 1.771, 95 % CI 0.475-6.595, p = 0.395; OR 2.192, 95 % CI 0.182-26.42, p = 0.537, respectively). In addition, meta-analysis revealed no association between the CCR5-Δ32 allele and vasculitis in all study subjects and in Europeans (OR 1.241, 95 % CI 0.951-1.620, p = 0.111; OR 1.359, 95 % CI 0.803-2.303, p = 0.254, respectively). However, the overall OR for the CCR5-Δ32 allele was significantly higher in Kawasaki disease (KD; OR 1.746, 95 % CI 1.003-2.955, p = 0.038) and the meta-analysis of the Δ32Δ32 + Δ32 W genotype showed a trend indicating an association with KD (OR 1.683, 95 % CI 0.921-3.077, p = 0.091). No association was found between the CCR5-Δ32 polymorphism and pSS. CONCLUSION This meta-analysis demonstrates that the CCR5-Δ32 polymorphism is associated with KD, but does not facilitate susceptibility to SLE, LN, or pSS.
Collapse
Affiliation(s)
- Y H Lee
- Division of Rheumatology, Department of Internal Medicine, Korea University Anam Hospital, Korea University College of Medicine, 126-1, Anam-dong 5-ga, Seongbuk-gu , 136-705, Seoul, Korea,
| | | | | |
Collapse
|
29
|
Fox JM, Kasprowicz R, Hartley O, Signoret N. CCR5 susceptibility to ligand-mediated down-modulation differs between human T lymphocytes and myeloid cells. J Leukoc Biol 2015; 98:59-71. [PMID: 25957306 PMCID: PMC4560160 DOI: 10.1189/jlb.2a0414-193rr] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2014] [Accepted: 04/03/2015] [Indexed: 01/04/2023] Open
Abstract
CCR5 is a chemokine receptor expressed on leukocytes and a coreceptor used by HIV-1 to enter CD4(+) T lymphocytes and macrophages. Stimulation of CCR5 by chemokines triggers internalization of chemokine-bound CCR5 molecules in a process called down-modulation, which contributes to the anti-HIV activity of chemokines. Recent studies have shown that CCR5 conformational heterogeneity influences chemokine-CCR5 interactions and HIV-1 entry in transfected cells or activated CD4(+) T lymphocytes. However, the effect of CCR5 conformations on other cell types and on the process of down-modulation remains unclear. We used mAbs, some already shown to detect distinct CCR5 conformations, to compare the behavior of CCR5 on in vitro generated human T cell blasts, monocytes and MDMs and CHO-CCR5 transfectants. All human cells express distinct antigenic forms of CCR5 not detected on CHO-CCR5 cells. The recognizable populations of CCR5 receptors exhibit different patterns of down-modulation on T lymphocytes compared with myeloid cells. On T cell blasts, CCR5 is recognized by all antibodies and undergoes rapid chemokine-mediated internalization, whereas on monocytes and MDMs, a pool of CCR5 molecules is recognized by a subset of antibodies and is not removed from the cell surface. We demonstrate that this cell surface-retained form of CCR5 responds to prolonged treatment with more-potent chemokine analogs and acts as an HIV-1 coreceptor. Our findings indicate that the regulation of CCR5 is highly specific to cell type and provide a potential explanation for the observation that native chemokines are less-effective HIV-entry inhibitors on macrophages compared with T lymphocytes.
Collapse
Affiliation(s)
- James M Fox
- *Department of Biology and Hull York Medical School, Center for Immunology and Infection, University of York, York, United Kingdom; and Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
| | - Richard Kasprowicz
- *Department of Biology and Hull York Medical School, Center for Immunology and Infection, University of York, York, United Kingdom; and Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
| | - Oliver Hartley
- *Department of Biology and Hull York Medical School, Center for Immunology and Infection, University of York, York, United Kingdom; and Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
| | - Nathalie Signoret
- *Department of Biology and Hull York Medical School, Center for Immunology and Infection, University of York, York, United Kingdom; and Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
| |
Collapse
|
30
|
Lau G, Labrecque J, Metz M, Vaz R, Fricker SP. Specificity for a CCR5 Inhibitor Is Conferred by a Single Amino Acid Residue: ROLE OF ILE198. J Biol Chem 2015; 290:11041-51. [PMID: 25767113 DOI: 10.1074/jbc.m115.640169] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Indexed: 11/06/2022] Open
Abstract
The chemokine receptors CCR5 and CCR2b share 89% amino acid homology. CCR5 is a co-receptor for HIV and CCR5 antagonists have been investigated as inhibitors of HIV infection. We describe the use of two CCR5 antagonists, Schering-C (SCH-C), which is specific for CCR5, and TAK-779, a dual inhibitor of CCR5 and CCR2b, to probe the CCR5 inhibitor binding site using CCR5/CCR2b chimeric receptors. Compound inhibition in the different chimeras was assessed by inhibition of chemokine-induced calcium flux. SCH-C inhibited RANTES (regulated on activation, normal T cell expressed and secreted) (CCL5)-mediated calcium flux on CCR5 with an IC50 of 22.8 nM but was inactive against monocyte chemoattractant protein-1 (CCL2)-mediated calcium flux on CCR2b. However, SCH-C inhibited CCL2-induced calcium flux against a CCR5/CCR2b chimera consisting of transmembrane domains IV-VI of CCR5 with an IC50 of 55 nM. A sequence comparison of CCR5 and CCR2b identified a divergent amino acid sequence located at the junction of transmembrane domain V and second extracellular loop. Transfer of the CCR5 sequence KNFQTLKIV into CCR2b conferred SCH-C inhibition (IC50 of 122 nM) into the predominantly CCR2b chimera. Furthermore, a single substitution, R206I, conferred partial but significant inhibition (IC50 of 1023 nM) by SCH-C. These results show that a limited amino acid sequence is responsible for SCH-C specificity to CCR5, and we propose a model showing the interaction with CCR5 Ile(198).
Collapse
Affiliation(s)
- Gloria Lau
- From Anormed Inc., Langley, British Columbia V2Y 1N5, Canada
| | - Jean Labrecque
- From Anormed Inc., Langley, British Columbia V2Y 1N5, Canada
| | - Markus Metz
- Lead Generation to Candidate Realization, Sanofi, Waltham, Massachusetts 02451, and
| | - Roy Vaz
- Lead Generation to Candidate Realization, Sanofi, Waltham, Massachusetts 02451, and
| | - Simon P Fricker
- Sanofi-Genzyme Research and Development Center, Framingham, Massachusetts 01701
| |
Collapse
|
31
|
Oo Z, Barrios CS, Castillo L, Beilke MA. High levels of CC-chemokine expression and downregulated levels of CCR5 during HIV-1/HTLV-1 and HIV-1/HTLV-2 coinfections. J Med Virol 2015; 87:790-7. [PMID: 25678365 DOI: 10.1002/jmv.24070] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/12/2014] [Indexed: 11/11/2022]
Abstract
The human T-cell lymphotropic virus type 1 (HTLV-1) and HTLV-2 are common copathogens among Human Immunodeficiency Virus (HIV)-infected individuals. HTLV-2 may confer a survival benefit among patients with HIV-1/HTLV-2 coinfections, along with lower plasma HIV-1 levels and delayed rates of CD4(+) T-cell decline. These effects have been attributed to the ability of the HTLV-2 viral transactivating Tax2 protein to induce the production of high levels of antiviral CC-chemokines and to downregulate expression of the CCR5 receptor, resulting in impaired entry of HIV-1 into CD4(+) T-cells. This study investigated the innate immunity of coinfected HIV/HTLV individuals by testing the ability of patient PBMCs to produce CC-chemokines in association CCR5 receptor modulation. The cellular proliferative responses of HIV/HTLV coinfected versus HIV monoinfected individuals were also evaluated. Higher levels of MIP-1α, MIP-1β, and RANTES (P < 0.05) were found in HIV-1/HTLV-2 coinfected group compared to HIV-1 monoinfected population. Upregulated levels of RANTES were shown in HIV-1/HTLV-1 after 1 and 3 days of culture (P < 0.05). Lymphocytes from HIV-1/HTLV-2 coinfected individuals showed significant CCR5 downregulation after 1 and 3 days of culture compared to lymphocytes from HIV-1 and uninfected groups (P < 0.05). Lower percentages of CCR5-positive cells were found in HIV-1/HTLV-1 coinfected after 3 days of incubation (P < 0.05). Levels of proliferation were significantly higher in the HIV-1/HTLV-1 group compared to HIV-1 alone (P < 0.05). HTLV-2 and HTLV-1 infections may induce the involvement of innate immunity against HIV-1 via stimulation of CC-chemokines and receptors, potentially modifying CCR5/HIV-1 binding and HIV-1 progression in coinfected individuals.
Collapse
Affiliation(s)
- Z Oo
- Infectious Diseases Division, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
| | | | | | | |
Collapse
|
32
|
Lee YH, Song GG. Association between chemokine receptor 5 delta32 polymorphism and susceptibility to cancer: a meta-analysis. J Recept Signal Transduct Res 2014; 35:509-15. [PMID: 25203595 DOI: 10.3109/10799893.2014.960934] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
OBJECTIVE To explore whether the functional chemokine receptor 5 delta32 (CCR5-Δ32) polymorphism is associated with susceptibility to cancer. METHODS A meta-analysis was conducted on the association between the CCR5-Δ32 polymorphism and cancer using (i) allele contrast and (ii) the dominant model. RESULTS Thirteen articles, including 16 comparative studies on a total of 3087 patients and 3735 controls, were included in the meta-analysis. These studies encompassed breast cancer (n = 3), bladder cancer (n = 3), cervical cancer (n = 2), pancreatic cancer (n = 2), prostate cancer (n = 2), head and neck cancer (n = 2), lymphoma (n = 1), gallbladder cancer (n = 1), skin cancer (n = 1) and mixed cancer (n = 1). The meta-analysis revealed an association between cancer and the CCR5-Δ32 allele (OR = 1.368, 95% CI = 1.064-1.758, p = 0.014), and stratification by ethnicity showed an association between the CCR5-Δ32 allele and cancer in Indians (OR = 2.480, 95% CI = 1.247-4.932, p = 0.010). The meta-analysis also revealed an association between breast cancer and the CCR5-Δ32 allele (OR = 1.689, 95% CI = 1.012-2.821, p = 0.045). However, allele contrast and the dominant model failed to reveal an association between the CCR5-Δ32 polymorphism and bladder cancer, cervical cancer, pancreatic cancer, prostate cancer, and head and neck cancer. CONCLUSIONS This meta-analysis demonstrates that the CCR5-Δ32 polymorphism is associated with susceptibility to cancer in Indians and is associated with breast cancer.
Collapse
Affiliation(s)
- Young Ho Lee
- a Division of Rheumatology, Department of Internal Medicine , Korea University College of Medicine , Seoul , Korea
| | - Gwan Gyu Song
- a Division of Rheumatology, Department of Internal Medicine , Korea University College of Medicine , Seoul , Korea
| |
Collapse
|
33
|
Chikere K, Webb NE, Chou T, Borm K, Sterjovski J, Gorry PR, Lee B. Distinct HIV-1 entry phenotypes are associated with transmission, subtype specificity, and resistance to broadly neutralizing antibodies. Retrovirology 2014; 11:48. [PMID: 24957778 PMCID: PMC4230403 DOI: 10.1186/1742-4690-11-48] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Accepted: 06/03/2014] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND The efficiency of CD4/CCR5 mediated HIV-1 entry has important implications for pathogenesis and transmission. The HIV-1 receptor affinity profiling (Affinofile) system analyzes and quantifies the infectivity of HIV-1 envelopes (Envs) across a spectrum of CD4/CCR5 expression levels and distills these data into a set of Affinofile metrics. The Affinofile system has shed light on how differential CD4/CCR5 usage efficiencies contributes to an array of Env phenotypes associated with cellular tropism, viral pathogenesis, and CCR5 inhibitor resistance. To facilitate more rapid, convenient, and robust analysis of HIV-1 entry phenotypes, we engineered a reporter Affinofile system containing a Tat- and Rev-dependent Gaussia luciferase-eGFP-Reporter (GGR) that is compatible with the use of pseudotyped or replication competent viruses with or without a virally encoded reporter gene. This GGR Affinofile system enabled a higher throughput characterization of CD4/CCR5 usage efficiencies associated with differential Env phenotypes. RESULTS We first validated our GGR Affinofile system on isogenic JR-CSF Env mutants that differ in their affinity for CD4 and/or CCR5. We established that their GGR Affinofile metrics reflected their differential entry phenotypes on primary PBMCs and CD4+ T-cell subsets. We then applied GGR Affinofile profiling to reveal distinct entry phenotypes associated with transmission, subtype specificity, and resistance to broadly neutralizing antibodies (BNAbs). First, we profiled a panel of reference subtype B transmitted/founder (T/F) and chronic Envs (n = 12) by analyzing the infectivity of each Env across 25 distinct combinations of CD4/CCR5 expression levels. Affinofile metrics revealed that at low CCR5 levels, our panel of subtype B T/F Envs was more dependent on high levels of CD4 for HIV-1 entry compared to chronic Envs. Next, we analyzed a reference panel of 28 acute/early subtype A-D Envs, and noted that subtype C Envs could be distinguished from the other subtypes based on their infectivity profiles and relevant Affinofile metrics. Lastly, mutations known to confer resistance to VRC01 or PG6/PG19 BNAbs, when engineered into subtypes A-D Envs, resulted in significantly decreased CD4/CCR5 usage efficiency. CONCLUSIONS GGR Affinofile profiling reveals pathophysiological phenotypes associated with varying HIV-1 entry efficiencies, and highlight the fitness costs associated with resistance to some broadly neutralizing antibodies.
Collapse
Affiliation(s)
- Kelechi Chikere
- Department of Microbiology, Immunology, and Molecular Genetics, Los Angeles, USA
| | - Nicholas E Webb
- Department of Microbiology, Immunology, and Molecular Genetics, Los Angeles, USA
| | - Tom Chou
- Department of Biomathematics, University of California at Los Angeles, Los Angeles, CA, USA
| | - Katharina Borm
- Center for Biomedical Research, Burnet Institute, Melbourne, VIC, Australia
| | - Jasminka Sterjovski
- Center for Biomedical Research, Burnet Institute, Melbourne, VIC, Australia
- Department of Infectious Diseases, Monash University, Melbourne, VIC, Australia
| | - Paul R Gorry
- Center for Biomedical Research, Burnet Institute, Melbourne, VIC, Australia
- Department of Infectious Diseases, Monash University, Melbourne, VIC, Australia
- Department of Microbiology and Immunology, University of Melbourne, Melbourne, VIC, Australia
| | - Benhur Lee
- Department of Microbiology, Immunology, and Molecular Genetics, Los Angeles, USA
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, #1124, New York, NY 10029, USA
| |
Collapse
|
34
|
Panos G, Watson DC. Effect of HIV-1 subtype and tropism on treatment with chemokine coreceptor entry inhibitors; overview of viral entry inhibition. Crit Rev Microbiol 2014; 41:473-87. [DOI: 10.3109/1040841x.2013.867829] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
35
|
Ray-Saha S, Huber T, Sakmar TP. Antibody epitopes on g protein-coupled receptors mapped with genetically encoded photoactivatable cross-linkers. Biochemistry 2014; 53:1302-10. [PMID: 24490954 PMCID: PMC3985944 DOI: 10.1021/bi401289p] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
![]()
We
developed a strategy for creating epitope maps of monoclonal
antibodies (mAbs) that bind to G protein-coupled receptors (GPCRs)
containing photo-cross-linkers. Using human CXC chemokine receptor
4 (CXCR4) as a model system, we genetically incorporated the photolabile
unnatural amino acid p-azido-l-phenylalanine
(azF) at various positions within extracellular loop 2 (EC2). We then
mapped the interactions of the azF-CXCR4 variants with mAb 12G5 using
targeted loss-of-function studies and photo-cross-linking in whole
cells in a microplate-based format. We used a novel variation of a
whole cell enzyme-linked immunosorbent assay to quantitate cross-linking
efficiency. 12G5 cross-linked primarily to residues 184, 178, and
189 in EC2 of CXCR4. Mapping of the data to the crystal structure
of CXCR4 showed a distinct mAb epitope footprint with the photo-cross-linked
residues clustered around the loss-of-function sites. We also used
the targeted photo-cross-linking approach to study the interaction
of human CC chemokine receptor 5 (CCR5) with PRO 140, a humanized
mAb that inhibits human immunodeficiency virus-1 cellular entry, and
2D7. The mAbs produced distinct cross-linking patterns on EC2 of CCR5.
PRO 140 cross-linked primarily to residues 174 and 175 at the amino-terminal
end of EC2, and 2D7 cross-linked mainly to residues 170, 176, and
184. These results were mapped to the recent crystal structure of
CCR5 in complex with maraviroc, showing cross-linked residues at the
tip of the maraviroc binding crevice formed by EC2. As a strategy
for mapping mAb epitopes on GPCRs, our targeted photo-cross-linking
method is complementary to loss-of-function mutagenesis results and
should be especially useful for studying mAbs with discontinuous epitopes.
Collapse
Affiliation(s)
- Sarmistha Ray-Saha
- Laboratory of Chemical Biology and Signal Transduction, The Rockefeller University , 1230 York Avenue, New York, New York 10065, United States
| | | | | |
Collapse
|
36
|
Lee YH, Bae SC, Song GG. Association between the chemokine receptor 5 delta32 polymorphism and rheumatoid arthritis: a meta-analysis. Mod Rheumatol 2014. [DOI: 10.3109/s10165-012-0665-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
37
|
A vaccine against CCR5 protects a subset of macaques upon intravaginal challenge with simian immunodeficiency virus SIVmac251. J Virol 2013; 88:2011-24. [PMID: 24307581 DOI: 10.1128/jvi.02447-13] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
As an alternative to targeting human immunodeficiency virus (HIV), we have developed vaccines targeting CCR5, a self-protein critically involved in HIV replication and pathogenesis. By displaying peptides derived from CCR5 at high density on the surface of virus-like particles, we can efficiently induce high-titer IgG antibodies against this self-molecule. Here, we investigated whether prophylactic immunization of rhesus macaques with a particle-based vaccine targeting two regions of macaque CCR5 could prevent or suppress vaginal infection with highly virulent SIVmac251. Twelve macaques were vaccinated with a bacteriophage Qß-based vaccine targeting macaque CCR5 (Qß.CCR5). Six control animals were immunized with the Qß platform alone. All animals immunized with Qß.CCR5 developed high-titer anti-CCR5 antibody responses. Macaques were vaginally challenged with a high dose of SIVmac251. The mean peak viral RNA levels in the vaccinated groups were 30-fold lower than in the control group (10(6.8) versus 10(8.3) copies/ml plasma). Three of the 12 vaccinated macaques dramatically suppressed simian immunodeficiency virus (SIV) replication: peak viral loads were low (10(3) to 10(4) RNA copies/ml), and SIV RNA became undetectable from 6 weeks onward. No viral RNA or DNA could be detected in colon and lymph node biopsy specimens collected 13 months after challenge. In vivo depletion of CD8(+) cells failed to induce a viral rebound. However, once anti-CCR5 antibody responses had waned, the 3 animals became infected after intravaginal and/or intravenous rechallenge. In conclusion, vaccination against CCR5 was associated with dramatic suppression of virus replication in a subset (25%) of macaques. These data support further research of vaccination against CCR5 to combat HIV infection.
Collapse
|
38
|
Song GG, Lee YH. A Meta-analysis of the relation between chemokine receptor 5 delta32 polymorphism and multiple sclerosis susceptibility. Immunol Invest 2013; 43:299-311. [DOI: 10.3109/08820139.2013.845204] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
39
|
Song GG, Kim JH, Lee YH. The chemokine receptor 5 delta32 polymorphism and type 1 diabetes, Behcet's disease, and asthma: a meta-analysis. Immunol Invest 2013; 43:123-36. [PMID: 24171669 DOI: 10.3109/08820139.2013.847457] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
OBJECTIVE The aim of this study was to determine whether the functional chemokine receptor 5 delta32 (CCR5-Δ32) polymorphism is associated with susceptibility to type 1 diabetes (T1D), Behcet's disease (BD), and asthma. RESULTS Fourteen studies encompassing 9,656 cases and 12,431 controls, including 6 on T1D, 5 on BD, and 3 on asthma, were available for meta-analysis. The meta-analysis showed a significant negative association between T1D and the CCR5-Δ32 allele (odds ratio [OR] = 0.854, 95% confidence interval [CI] = 0.800-0.912, p = 2.2 × 10⁻⁷). Stratification by ethnicity and analysis of the Δ32Δ32 + Δ32W genotype indicated a significant negative association between the CCR5-Δ32 allele and T1D in Europeans (OR = 0.857, 95% CI = 0.802-0.915, p = 3.5 × 10⁻⁸; OR = 0.896, 95% CI = 0.808-0.932, p = 9.3 × 10⁻⁶, respectively). The meta-analysis showed a positive association between BD and the Δ32Δ32 + Δ32W genotype (OR = 1.403, 95% CI = 1.008-1.954, p = 0.045). Stratification by HLA-B51 status indicated an association between the CCR5-Δ32 allele and HLA-B51-positive BD, but not HLA-B51-negative BD (OR = 1.619, 95% CI = 1.070-2.451, p = 0.023; OR = 1.036, 95% CI = 0.674-1.593, p = 0.872, respectively). No association was found between the CCR5-Δ32 polymorphism and asthma. CONCLUSIONS This meta-analysis demonstrates that the CCR5-Δ32 polymorphism acts as a protective factor in T1D development in Europeans, and a risk factor for BD among HLA-B51 carriers. However, no association was found between the CCR5-Δ32 polymorphism and asthma.
Collapse
Affiliation(s)
- Gwan Gyu Song
- Division of Rheumatology, Department of Internal Medicine, Korea University College of Medicine , Seoul , Korea
| | | | | |
Collapse
|
40
|
Biasin M, De Luca M, Gnudi F, Clerici M. The genetic basis of resistance to HIV infection and disease progression. Expert Rev Clin Immunol 2013; 9:319-34. [PMID: 23557268 DOI: 10.1586/eci.13.16] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Susceptibility to HIV infection and the modulation of disease progression are strictly dependent on inter-individual variability, much of which is secondary to host genetic heterogeneity. The study of host factors that control these phenomena relies not only on candidate gene approaches but also on unbiased genome-wide genetic and functional analyses. Additional new insights stem from the study of mechanisms that control the expression of host and viral genes, such as miRNA. The genetic host factors that have been suggested to be associated either with resistance to HIV-1 infection or with absent/delayed progression to AIDS are nevertheless unable to fully justify the phenomenon of differential susceptibility to HIV. Multidisciplinary approaches are needed to further analyze individuals who deviate from the expected response to HIV exposure/infection. Results of these analyses will facilitate the identification of novel targets that could be exploited in the setting up of innovative therapeutic or vaccine approaches.
Collapse
Affiliation(s)
- Mara Biasin
- Department of Biomedical and Clinical Sciences, University of Milan, Via GB Grassi 74, 20157 Milan, Italy.
| | | | | | | |
Collapse
|
41
|
Wang NX, Sieg SF, Lederman MM, Offord RE, Hartley O, von Recum HA. Using glycosaminoglycan/chemokine interactions for the long-term delivery of 5P12-RANTES in HIV prevention. Mol Pharm 2013; 10:3564-73. [PMID: 23859720 DOI: 10.1021/mp3007242] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
5P12-RANTES is a recently developed chemokine analogue that has shown high level protection from SHIV infection in macaques. However, the feasibility of using 5P12-RANTES as a long-term HIV prevention agent has not been explored partially due to the lack of available delivery devices that can easily be modified for long-term release profiles. Glycosaminoglycans (GAGs) have been known for their affinity for various cytokines and chemokines, including native RANTES, or CCL5. In this work, we investigated used of GAGs in generating a chemokine drug delivery device. Initial studies used surface plasmon resonance analysis to characterize and compare the affinities of different GAGs to 5P12-RANTES. These different GAGs were then incorporated into drug delivery polymeric hydrogels to engineer sustained release of the chemokines. In vitro release studies of 5P12-RANTES from the resulting polymers were performed, and we found that 5P12-RANTES release from these polymers can be controlled by the amount and type of GAG incorporated. Polymer disks containing GAGs with stronger affinity to 5P12-RANTES resulted in more sustained and longer term release than did polymer disks containing GAGs with weaker 5P12-RANTES affinity. Similar trends were observed by varying the amount of GAGs incorporated into the delivery system. 5P12-RANTES released from these polymers demonstrated good levels of CCR5 blocking, retaining activity even after 30 days of incubation.
Collapse
Affiliation(s)
- Nick X Wang
- Department of Biomedical Engineering, Case Western Reserve University , 10900 Euclid Ave, Cleveland, Ohio 44106, United States
| | | | | | | | | | | |
Collapse
|
42
|
Haqqani AA, Tilton JC. Entry inhibitors and their use in the treatment of HIV-1 infection. Antiviral Res 2013; 98:158-70. [PMID: 23541872 DOI: 10.1016/j.antiviral.2013.03.017] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2013] [Revised: 03/18/2013] [Accepted: 03/19/2013] [Indexed: 12/20/2022]
Abstract
Entry of HIV into target cells is a complex, multi-stage process involving sequential attachment and CD4 binding, coreceptor binding, and membrane fusion. HIV entry inhibitors are a complex group of drugs with multiple mechanisms of action depending on the stage of the viral entry process they target. Two entry inhibitors are currently approved for the treatment of HIV-infected patients. Maraviroc, a CCR5 antagonist, blocks interactions between the viral envelope proteins and the CCR5 coreceptor. Enfuvirtide, a fusion inhibitor, disrupts conformational changes in gp41 that drive membrane fusion. A wide array of additional agents are in various stages of development. This review covers the entry inhibitors and their use in the treatment of HIV-infected patients.
Collapse
Affiliation(s)
- Aiman A Haqqani
- Case Center for Proteomics and Bioinformatics, Case Western Reserve University, Cleveland, OH 44106, USA
| | | |
Collapse
|
43
|
Multifaceted mechanisms of HIV inhibition and resistance to CCR5 inhibitors PSC-RANTES and Maraviroc. Antimicrob Agents Chemother 2013; 57:2640-50. [PMID: 23529732 DOI: 10.1128/aac.02511-12] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Small-molecule CCR5 antagonists, such as maraviroc (MVC), likely block HIV-1 through an allosteric, noncompetitive inhibition mechanism, whereas inhibition by agonists such as PSC-RANTES is less defined and may involve receptor removal by cell surface downregulation, competitive inhibition by occluding the HIV-1 envelope binding, and/or allosteric effects by altering CCR5 conformation. We explored the inhibitory mechanisms of maraviroc and PSC-RANTES by employing pairs of virus clones with differential sensitivities to these inhibitors. Intrinsic PSC-RANTES-resistant virus (YA versus RT) or those selected in PSC-RANTES treated macaques (M584 versus P3-4) only displayed resistance in multiple-cycle assays or with a CCR5 mutant that cannot be downregulated. In single-cycle assays, these HIV-1 clones displayed equal sensitivity to PSC-RANTES inhibition, suggesting effective receptor downregulation. Prolonged PSC-RANTES exposure resulted in desensitization of the receptor to internalization such that increasing virus concentration (substrate) could saturate the receptors and overcome PSC-RANTES inhibition. In contrast, resistance to MVC was observed with the MVC-resistant HIV-1 (R3 versus S2) in both multiple- and single-cycle assays and with altered virus concentrations, which is indicative of allosteric inhibition. MVC could also mediate inhibition and possibly resistance through competitive mechanisms.
Collapse
|
44
|
Rummel PC, Arfelt KN, Baumann L, Jenkins TJ, Thiele S, Lüttichau HR, Johnsen A, Pease J, Ghosh S, Kolbeck R, Rosenkilde MM. Molecular requirements for inhibition of the chemokine receptor CCR8--probe-dependent allosteric interactions. Br J Pharmacol 2013; 167:1206-17. [PMID: 22708643 DOI: 10.1111/j.1476-5381.2012.02076.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND AND PURPOSE Here we present a novel series of CCR8 antagonists based on a naphthalene-sulfonamide structure. This structure differs from the predominant pharmacophore for most small-molecule CC-chemokine receptor antagonists, which in fact activate CCR8, suggesting that CCR8 inhibition requires alternative structural probes. EXPERIMENTAL APPROACH The compounds were tested as inverse agonists and as antagonists against CCL1-induced activity in Gα(i) signalling and chemotaxis. Furthermore, they were assessed by heterologous competition binding against two radiolabelled receptor ligands: the endogenous agonist CCL1 and the virus-encoded antagonist MC148. KEY RESULTS All compounds were highly potent inverse agonists with EC(50) values from 1.7 to 23 nM. Their potencies as antagonists were more widely spread (EC(50) values from 5.9 to 1572 nM). Some compounds were balanced antagonists/inverse agonists whereas others were predominantly inverse agonists with >100-fold lower potency as antagonists. A correspondingly broad range of affinities, which followed the antagonist potencies, was disclosed by competition with [(125)I]-CCL1 (K(i) 3.4-842 nM), whereas the affinities measured against [(125)I]-MC148 were less widely spread (K(i) 0.37-27 nM), and matched the inverse agonist potencies. CONCLUSION AND IMPLICATIONS Despite highly potent and direct effects as inverse agonists, competition-binding experiments against radiolabelled agonist and tests for antagonism revealed a probe-dependent allosteric effect of these compounds. Thus, minor chemical changes affected the ability to modify chemokine binding and action, and divided the compounds into two groups: predominantly inverse agonists and balanced antagonists/inverse agonists. These studies have important implications for the design of new inverse agonists with or without antagonist properties.
Collapse
Affiliation(s)
- P C Rummel
- Department of Neuroscience and Pharmacology, Faculty of Health Sciences, Copenhagen University, Copenhagen, Denmark
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Orellana JA, Velasquez S, Williams DW, Sáez JC, Berman JW, Eugenin EA. Pannexin1 hemichannels are critical for HIV infection of human primary CD4+ T lymphocytes. J Leukoc Biol 2013; 94:399-407. [PMID: 23456773 DOI: 10.1189/jlb.0512249] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
HIV is a major public health issue, and infection of CD4(+) T lymphocytes is one of its key features. Whereas several cellular proteins have been identified that facilitate viral infection and replication, the role of hemichannels in these processes has not been fully characterized. We now show that the HIV isolates, R5 and X4, induced a transient-early (5-30 min) and a later, persistent (48-120 h) opening of Panx1 hemichannels, which was dependent on the binding of HIV to CD4 and CCR5/CXCR4 receptors. Blocking Panx1 hemichannels by reducing their opening or protein expression inhibited HIV replication in CD4(+) T lymphocytes. Thus, our findings demonstrate that Panx1 hemichannels play an essential role in HIV infection.
Collapse
Affiliation(s)
- J A Orellana
- Departamento de Neurología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | | | | | | | | | | |
Collapse
|
46
|
Xiang SH, Pacheco B, Bowder D, Yuan W, Sodroski J. Characterization of a dual-tropic human immunodeficiency virus (HIV-1) strain derived from the prototypical X4 isolate HXBc2. Virology 2013; 438:5-13. [PMID: 23369572 DOI: 10.1016/j.virol.2013.01.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2012] [Revised: 12/21/2012] [Accepted: 01/02/2013] [Indexed: 11/25/2022]
Abstract
Human immunodeficiency virus type 1 (HIV-1) coreceptor usage and tropism can be modulated by the V3 loop sequence of the gp120 exterior envelope glycoprotein. For coreceptors, R5 viruses use CCR5, X4 viruses use CXCR4, and dual-tropic (R5X4) viruses use either CCR5 or CXCR4. To understand the requirements for dual tropism, we derived and analyzed a dual-tropic variant of an X4 virus. Changes in the V3 base, which allow gp120 to interact with the tyrosine-sulfated CCR5 N-terminus, and deletion of residues 310/311 in the V3 tip were necessary for efficient CCR5 binding and utilization. Thus, both sets of V3 changes allowed CCR5 utilization with retention of the ability to use CXCR4. We also found that the stable association of gp120 with the trimeric envelope glycoprotein complex in R5X4 viruses, as in X4 viruses, is less sensitive to V3 loop changes than gp120-trimer association in R5 viruses.
Collapse
Affiliation(s)
- Shi-hua Xiang
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute and Department of Microbiology and Immunobiology, Division of AIDS, Harvard Medical School, Boston, MA 02215, USA.
| | | | | | | | | |
Collapse
|
47
|
Knepp AM, Sakmar TP, Huber T. Homogeneous time-resolved fluorescence assay to probe folded G protein-coupled receptors. Methods Enzymol 2013; 522:169-89. [PMID: 23374186 DOI: 10.1016/b978-0-12-407865-9.00010-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/08/2022]
Abstract
Continued advances in G protein-coupled receptor (GPCR) structural biology and biochemistry depend in part on strategies to stabilize these polytopic membrane proteins in purified systems. New methods to measure properly folded GPCRs are needed to facilitate the identification of suitable conditions and ensure sample quality. Most GPCRs do not contain an intrinsic reporter on their functionality, so probes must be introduced. Here, we describe a fluorescence-based approach to quantitatively measure the chemokine receptor CCR5 with labeled antibodies. The assay is exceptionally sensitive and high-throughput. We detail procedures to label antibodies, characterize the system, and process data. We also describe several useful applications, including optimization of incorporation into nanoscale apolipoprotein bound bilayers (NABBs or nanodiscs), measurement of receptor stability, and competition binding assays.
Collapse
Affiliation(s)
- Adam M Knepp
- Laboratory of Chemical Biology and Signal Transduction, The Rockefeller University, New York, New York, USA
| | | | | |
Collapse
|
48
|
Szpakowska M, Fievez V, Arumugan K, van Nuland N, Schmit JC, Chevigné A. Function, diversity and therapeutic potential of the N-terminal domain of human chemokine receptors. Biochem Pharmacol 2012; 84:1366-80. [PMID: 22935450 DOI: 10.1016/j.bcp.2012.08.008] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2012] [Revised: 08/08/2012] [Accepted: 08/09/2012] [Indexed: 11/16/2022]
Abstract
Chemokines and their receptors play fundamental roles in many physiological and pathological processes such as leukocyte trafficking, inflammation, cancer and HIV-1 infection. Chemokine-receptor interactions are particularly intricate and therefore require precise orchestration. The flexible N-terminal domain of human chemokine receptors has regularly been demonstrated to hold a crucial role in the initial recognition and selective binding of the receptor ligands. The length and the amino acid sequences of the N-termini vary considerably among different receptors but they all show a high content of negatively charged residues and are subject to post-translational modifications such as O-sulfation and N- or O-glycosylation. In addition, a conserved cysteine that is most likely engaged in a receptor-stabilizing disulfide bond delimits two functionally distinct parts in the N-terminus, characterized by specific molecular signatures. Structural analyses have shown that the N-terminus of chemokine receptors recognizes a groove on the chemokine surface and that this interaction is stabilized by high-affinity binding to a conserved sulfotyrosine-binding pocket. Altogether, these data provide new insights on the chemokine-receptor molecular interplay and identify the receptor N-terminus-binding site as a new target for the development of therapeutic molecules. This review presents and discusses the diversity and function of human chemokine receptor N-terminal domains and provides a comprehensive annotated inventory of their sequences, laying special emphasis on the presence of post-translational modifications and functional features. Finally, it identifies new molecular signatures and proposes a computational model for the positioning and the conformation of the CXCR4 N-terminus grafted on the first chemokine receptor X-ray structure.
Collapse
Affiliation(s)
- Martyna Szpakowska
- Laboratory of Retrovirology, Public Research Center for Health, Luxembourg, Luxembourg.
| | | | | | | | | | | |
Collapse
|
49
|
An effector phenotype of CD8+ T cells at the junction epithelium during clinical quiescence of herpes simplex virus 2 infection. J Virol 2012; 86:10587-96. [PMID: 22811543 DOI: 10.1128/jvi.01237-12] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Herpes simplex virus 2 infection is characterized by cycles of viral quiescence and reactivation. CD8(+) T cells persist at the site of viral reactivation, at the genital dermal-epidermal junction contiguous to neuronal endings of sensory neurons, for several months after herpes lesion resolution. To evaluate whether these resident CD8(+) T cells frequently encounter HSV antigen even during times of asymptomatic viral infection, we analyzed the transcriptional output of CD8(+) T cells captured by laser microdissection from human genital skin biopsy specimens during the clinically quiescent period of 8 weeks after lesion resolution. These CD8(+) T cells expressed a characteristic set of genes distinct from those of three separate control cell populations, and network and pathway analyses revealed that these T cells significantly upregulated antiviral genes such as GZMB, PRF1, INFG, IL-32, and LTA, carbohydrate and lipid metabolism-related genes such as GLUT-1, and chemotaxis and recruitment genes such as CCL5 and CCR1, suggesting a possible feedback mechanism for the recruitment of CD8(+) T cells to the site of infection. Many of these transcripts are known to have half-lives of <48 h, suggesting that cognate antigen is released frequently into the mucosa and that resident CD8(+) T cells act as functional effectors in controlling viral spread.
Collapse
|
50
|
Association between the chemokine receptor 5 delta32 polymorphism and rheumatoid arthritis: a meta-analysis. Mod Rheumatol 2012; 23:304-10. [PMID: 22638733 DOI: 10.1007/s10165-012-0665-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2012] [Accepted: 04/20/2012] [Indexed: 10/28/2022]
Abstract
OBJECTIVE The aim of this study was to determine whether the functional chemokine receptor 5 delta32 (CCR5-Δ32) polymorphism confers susceptibility to rheumatoid arthritis (RA) and juvenile idiopathic arthritis (JIA). METHODS Meta-analysis was conducted on associations between the CCR5-Δ32 polymorphism and RA and JIA using (1) allele contrast and (2) the recessive, (3) the dominant, and (4) the additive models. RESULTS Eleven population comparisons based on the data obtained from nine studies involving 13,412 subjects (RA 3,848, controls 4,095; JIA 1,599, controls 3,870) were considered. In all study subjects, meta-analysis showed a significant negative association between RA and the CCR5-Δ32 allele (OR = 0.771, 95 % CI = 0.694-0.866, p = 6.5 × 10(-7)). Stratification by ethnicity indicated a significant association between the CCR5-Δ32 allele and RA in Europeans (OR = 0.8001, 95 % CI = 0.709-0.904, p = 3.2 × 10(-5)). Meta-analysis showed associations between the CCR5-Δ32 allele and JIA in Europeans and oligoarticular type (OR = 0.797, 95 % CI = 0.690-0.921, p = 0.002; OR = 0.475, 95 % CI = 0.352-0.693, p = 9.5 × 10(-8)). CONCLUSIONS This meta-analysis demonstrates that the CCR5-Δ32 polymorphism may confer susceptibility to RA and JIA in Europeans, and suggests that the CCR5-Δ32 allele protects against the development of RA and JIA.
Collapse
|