1
|
Lawrence SP, Elser SE, Torben W, Blair RV, Pahar B, Aye PP, Schiro F, Szeltner D, Doyle-Meyers LA, Haggarty BS, Jordan APO, Romano J, Leslie GJ, Alvarez X, O’Connor DH, Wiseman RW, Fennessey CM, Li Y, Piatak M, Lifson JD, LaBranche CC, Lackner AA, Keele BF, Maness NJ, Marsh M, Hoxie JA. A cellular trafficking signal in the SIV envelope protein cytoplasmic domain is strongly selected for in pathogenic infection. PLoS Pathog 2022; 18:e1010507. [PMID: 35714165 PMCID: PMC9275724 DOI: 10.1371/journal.ppat.1010507] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 07/12/2022] [Accepted: 04/07/2022] [Indexed: 01/01/2023] Open
Abstract
The HIV/SIV envelope glycoprotein (Env) cytoplasmic domain contains a highly conserved Tyr-based trafficking signal that mediates both clathrin-dependent endocytosis and polarized sorting. Despite extensive analysis, the role of these functions in viral infection and pathogenesis is unclear. An SIV molecular clone (SIVmac239) in which this signal is inactivated by deletion of Gly-720 and Tyr-721 (SIVmac239ΔGY), replicates acutely to high levels in pigtail macaques (PTM) but is rapidly controlled. However, we previously reported that rhesus macaques and PTM can progress to AIDS following SIVmac239ΔGY infection in association with novel amino acid changes in the Env cytoplasmic domain. These included an R722G flanking the ΔGY deletion and a nine nucleotide deletion encoding amino acids 734-736 (ΔQTH) that overlaps the rev and tat open reading frames. We show that molecular clones containing these mutations reconstitute signals for both endocytosis and polarized sorting. In one PTM, a novel genotype was selected that generated a new signal for polarized sorting but not endocytosis. This genotype, together with the ΔGY mutation, was conserved in association with high viral loads for several months when introduced into naïve PTMs. For the first time, our findings reveal strong selection pressure for Env endocytosis and particularly for polarized sorting during pathogenic SIV infection in vivo.
Collapse
Affiliation(s)
- Scott P. Lawrence
- MRC Laboratory for Molecular Cell Biology, University College London, London, United Kingdom
| | - Samra E. Elser
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Workineh Torben
- Tulane National Primate Research Center, Covington, Louisiana, United States of America
| | - Robert V. Blair
- Tulane National Primate Research Center, Covington, Louisiana, United States of America
| | - Bapi Pahar
- Tulane National Primate Research Center, Covington, Louisiana, United States of America
| | - Pyone P. Aye
- Tulane National Primate Research Center, Covington, Louisiana, United States of America
| | - Faith Schiro
- Tulane National Primate Research Center, Covington, Louisiana, United States of America
| | - Dawn Szeltner
- Tulane National Primate Research Center, Covington, Louisiana, United States of America
| | - Lara A. Doyle-Meyers
- Tulane National Primate Research Center, Covington, Louisiana, United States of America
| | - Beth S. Haggarty
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Andrea P. O. Jordan
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Josephine Romano
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - George J. Leslie
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Xavier Alvarez
- Tulane National Primate Research Center, Covington, Louisiana, United States of America
| | - David H. O’Connor
- Wisconsin National Primate Research Center, Madison, Wisconsin, United States of America
| | - Roger W. Wiseman
- Wisconsin National Primate Research Center, Madison, Wisconsin, United States of America
| | - Christine M. Fennessey
- AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - Yuan Li
- AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - Michael Piatak
- AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - Jeffrey D. Lifson
- AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - Celia C. LaBranche
- Duke University Medical Center, Durham, North Carolina, United States of America
| | - Andrew A. Lackner
- Tulane National Primate Research Center, Covington, Louisiana, United States of America
| | - Brandon F. Keele
- AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - Nicholas J. Maness
- Tulane National Primate Research Center, Covington, Louisiana, United States of America
| | - Mark Marsh
- MRC Laboratory for Molecular Cell Biology, University College London, London, United Kingdom
| | - James A. Hoxie
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| |
Collapse
|
2
|
Moretti S, Virtuoso S, Sernicola L, Farcomeni S, Maggiorella MT, Borsetti A. Advances in SIV/SHIV Non-Human Primate Models of NeuroAIDS. Pathogens 2021; 10:pathogens10081018. [PMID: 34451482 PMCID: PMC8398602 DOI: 10.3390/pathogens10081018] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Revised: 08/04/2021] [Accepted: 08/11/2021] [Indexed: 01/09/2023] Open
Abstract
Non-human primates (NHPs) are the most relevant model of Acquired Immunodeficiency Syndrome (AIDS) and neuroAIDS, being of great importance in explaining the pathogenesis of HIV-induced nervous system damage. Simian Immunodeficiency Virus (SIV)/ Simian-Human Immunodeficiency Virus (SHIV)-infected monkeys have provided evidence of complex interactions between the virus and host that include host immune response, viral genetic diversity, and genetic susceptibility, which may explain virus-associated central nervous system (CNS) pathology and HIV-associated neurocognitive disorders (HAND). In this article, we review the recent progress contributions obtained using monkey models of HIV infection of the CNS, neuropathogenesis and SIV encephalitis (SIVE), with an emphasis on pharmacologic therapies and dependable markers that predict development of CNS AIDS.
Collapse
|
3
|
Braxton AM, Chalmin AL, Najarro KM, Brockhurst JK, Johnson KT, Lyons CE, Daly B, Cryer CG, Vijay S, Cyphers G, Guerrero-Martin SM, Aston SA, McGee K, Su YP, Arav-Boger R, Metcalf Pate KA. Platelet-endothelial associations may promote cytomegalovirus replication in the salivary gland in mice. Platelets 2020; 31:860-868. [PMID: 31726921 PMCID: PMC7220825 DOI: 10.1080/09537104.2019.1689383] [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: 09/13/2019] [Revised: 10/24/2019] [Accepted: 10/31/2019] [Indexed: 10/25/2022]
Abstract
Platelet decline is a feature of many acute viral infections, including cytomegalovirus (CMV) infection in humans and mice. Platelet sequestration in association with other cells, including endothelium and circulating leukocytes, can contribute to this decline and influence the immune response to and pathogenesis of viral infection. We sought to determine if platelet-endothelial associations (PEAs) contribute to platelet decline during acute murine CMV (mCMV) infection, and if these associations affect viral load and production. Male BALB/c mice were infected with mCMV (Smith strain), euthanized at timepoints throughout acute infection and compared to uninfected controls. An increase in PEA formation was confirmed in the salivary gland at all post-inoculation timepoints using immunohistochemistry for CD41+ platelets co-localizing with CD34+ vessels. Platelet depletion did not change amount of viral DNA or timecourse of infection, as measured by qPCR. However, platelet depletion reduced viral titer of mCMV in the salivary glands while undepleted controls demonstrated robust replication in the tissue by plaque assay. Thus, platelet associations with endothelium may enhance the ability of mCMV to replicate within the salivary gland. Further work is needed to determine the mechanisms behind this effect and if pharmacologic inhibition of PEAs may reduce CMV production in acutely infected patients.
Collapse
Affiliation(s)
- Alicia M. Braxton
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, USA
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Alyssa L. Chalmin
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Kevin M. Najarro
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Jacqueline K. Brockhurst
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, USA
- University of Pennsylvania School of Veterinary Medicine, Philadelphia, USA
| | - Karl T. Johnson
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Claire E. Lyons
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Brenna Daly
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, USA
- Cummings School of Veterinary Medicine, Tufts University, North Grafton, USA
| | - Catherine G. Cryer
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, USA
- University of Pennsylvania School of Veterinary Medicine, Philadelphia, USA
| | - Shefali Vijay
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Griffin Cyphers
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Selena M. Guerrero-Martin
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, USA
| | - S. Andrew Aston
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, USA
- Department of Psychiatry of Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Kirstin McGee
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Yu-Pin Su
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Ravit Arav-Boger
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, USA
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, USA
| | - Kelly A. Metcalf Pate
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, USA
| |
Collapse
|
4
|
Beck SE, Queen SE, Metcalf Pate KA, Mangus LM, Abreu CM, Gama L, Witwer KW, Adams RJ, Zink MC, Clements JE, Mankowski JL. An SIV/macaque model targeted to study HIV-associated neurocognitive disorders. J Neurovirol 2017; 24:204-212. [PMID: 28975505 DOI: 10.1007/s13365-017-0582-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 08/25/2017] [Accepted: 09/14/2017] [Indexed: 02/01/2023]
Abstract
Simian immunodeficiency virus (SIV) infection of pigtailed macaques is a highly representative and well-characterized animal model for HIV neuropathogenesis studies that provides an excellent opportunity to study and develop prognostic markers of HIV-associated neurocognitive disorders (HAND) for HIV-infected individuals. SIV studies can be performed in a controlled setting that enhances reproducibility and offers high-translational value. Similar to observations in HIV-infected patients receiving antiretroviral therapy (ART), ongoing neurodegeneration and inflammation are present in SIV-infected pigtailed macaques treated with suppressive ART. By developing quantitative viral outgrowth assays that measure both CD4+ T cells and macrophages harboring replication competent SIV as well as a highly sensitive mouse-based viral outgrowth assay, we have positioned the SIV/pigtailed macaque model to advance our understanding of latent cellular reservoirs, including potential CNS reservoirs, to promote HIV cure. In addition to contributing to our understanding of the pathogenesis of HAND, the SIV/pigtailed macaque model also provides an excellent opportunity to test innovative approaches to eliminate the latent HIV reservoir in the brain.
Collapse
Affiliation(s)
- Sarah E Beck
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Suzanne E Queen
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Kelly A Metcalf Pate
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Lisa M Mangus
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Celina M Abreu
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Lucio Gama
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Kenneth W Witwer
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University, Baltimore, MD, 21205, USA
- Department of Neurology, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Robert J Adams
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - M Christine Zink
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Janice E Clements
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University, Baltimore, MD, 21205, USA
- Department of Neurology, Johns Hopkins University, Baltimore, MD, 21205, USA
- Department of Pathology, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Joseph L Mankowski
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University, Baltimore, MD, 21205, USA.
- Department of Neurology, Johns Hopkins University, Baltimore, MD, 21205, USA.
- Department of Pathology, Johns Hopkins University, Baltimore, MD, 21205, USA.
| |
Collapse
|
5
|
Lipopolysaccharide Increases Immune Activation and Alters T Cell Homeostasis in SHIVB'WHU Chronically Infected Chinese Rhesus Macaque. J Immunol Res 2015; 2015:202738. [PMID: 26713320 PMCID: PMC4680103 DOI: 10.1155/2015/202738] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Accepted: 11/05/2015] [Indexed: 11/18/2022] Open
Abstract
Immune activation plays a significant role in the disease progression of HIV. Microbial products, especially bacterial lipopolysaccharide (LPS), contribute to immune activation. Increasing evidence indicates that T lymphocyte homeostasis disruptions are associated with immune activation. However, the mechanism by which LPS affects disruption of immune response is still not fully understood. Chronically SHIVB'WHU-infected Chinese rhesus macaques received 50 μg/kg body weight LPS in this study. LPS administration affected the virus/host equilibrium by elevating the levels of viral replication and activating T lymphocytes. LPS induced upregulation of CD8+ naïve T cells and downregulated the number of CD4+ and CD8+ T effector memory cells. The downregulated effector memory cells are associated with a lower frequency of monofunctional and polyfunctional cells, and an upregulated programmed cell death-1 (PD-1) expression on CD4+ and CD8+ T cells was observed in monkeys after LPS stimulation. Our data provide new insights into the function of LPS in the immune activation in SHIV/HIV infection.
Collapse
|
6
|
Kiebala M, Singh MV, Piepenbrink MS, Qiu X, Kobie JJ, Maggirwar SB. Platelet Activation in Human Immunodeficiency Virus Type-1 Patients Is Not Altered with Cocaine Abuse. PLoS One 2015; 10:e0130061. [PMID: 26076359 PMCID: PMC4467977 DOI: 10.1371/journal.pone.0130061] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Accepted: 05/15/2015] [Indexed: 12/30/2022] Open
Abstract
Recent work has indicated that platelets, which are anucleate blood cells, significantly contribute to inflammatory disorders. Importantly, platelets also likely contribute to various inflammatory secondary disorders that are increasingly associated with Human Immunodeficiency Virus Type-1 (HIV) infection including neurological impairments and cardiovascular complications. Indeed, HIV infection is often associated with increased levels of platelet activators. Additionally, cocaine, a drug commonly abused by HIV-infected individuals, leads to increased platelet activation in humans. Considering that orchestrated signaling mechanisms are essential for platelet activation, and that nuclear factor-kappa B (NF-κB) inhibitors can alter platelet function, the role of NF-κB signaling in platelet activation during HIV infection warrants further investigation. Here we tested the hypothesis that inhibitory kappa B kinase complex (IKK) activation would be central for platelet activation induced by HIV and cocaine. Whole blood from HIV-positive and HIV-negative individuals, with or without cocaine abuse was used to assess platelet activation via flow cytometry whereas IKK activation was analyzed by performing immunoblotting and in vitro kinase assays. We demonstrate that increased platelet activation in HIV patients, as measured by CD62P expression, is not altered with reported cocaine use. Furthermore, cocaine and HIV do not activate platelets in whole blood when treated ex vivo. Finally, HIV-induced platelet activation does not involve the NF-κB signaling intermediate, IKKβ. Platelet activation in HIV patients is not altered with cocaine abuse. These results support the notion that non-IKK targeting approaches will be better suited for the treatment of HIV-associated inflammatory disorders.
Collapse
Affiliation(s)
- Michelle Kiebala
- Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, New York, United States of America
- * E-mail:
| | - Meera V. Singh
- Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, New York, United States of America
| | - Michael S. Piepenbrink
- Division of Infectious Diseases, University of Rochester School of Medicine and Dentistry, Rochester, New York, United States of America
| | - Xing Qiu
- Department of Biostatistics and Computational Biology, University of Rochester School of Medicine and Dentistry, Rochester, New York, United States of America
| | - James J. Kobie
- Division of Infectious Diseases, University of Rochester School of Medicine and Dentistry, Rochester, New York, United States of America
| | - Sanjay B. Maggirwar
- Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, New York, United States of America
| |
Collapse
|
7
|
Rafatpanah H, Essmailian L, Hedayati-Moghaddam MR, Vakili R, Norouzi M, Sarvghad MR, Hosseinpour AM, Sharebiani H, Rezaee SAR. Evaluation of Non-Viral Surrogate Markers as Predictive Indicators for Monitoring Progression of Human Immunodeficiency Virus Infection: An Eight-Year Analysis in a Regional Center. Jpn J Infect Dis 2015; 69:39-44. [PMID: 25971319 DOI: 10.7883/yoken.jjid.2014.261] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Suitable methods for clinical monitoring of HIV-infected patients are crucial in resource-poor settings. Demographic data, clinical staging, and laboratory findings for 112 asymptomatic subjects positive for HIV were assessed at the first admission and the last visit from 2002 to 2010. Cox regression analysis showed hemoglobin (Hb) (HR = 0.643, P = 0.021) to be a predictive indicator for disease progression, while CD4, CD8, and platelet counts showed low HRs, despite having significant probability values. Hb and total lymphocyte count (TLC) rapidly declined from stage II to III (10.9 and 29.6%, respectively). Reduced CD4 and platelet counts and Hb during stage I were associated with disease progression, and TLC was correlated with CD4 counts at the last follow-up (P < 0.001). However, WHO TLC cutoff of 1,200 cell/mm(3) had 26.1% sensitivity and 98.6% specificity. ROC curve analysis suggested that a TLC cutoff of 1,800 cell/mm(3) was more reliable in this region. Statistical analysis and data mining findings showed that Hb and TLC, and their rapid decline from stage II to III, in addition to reduced platelet count, could be valuable markers for a surrogate algorithm for monitoring of HIV-infected subjects and starting anti-viral therapy in the absence of sophisticated detection assays.
Collapse
|
8
|
Beck SE, Queen SE, Witwer KW, Metcalf Pate KA, Mangus LM, Gama L, Adams RJ, Clements JE, Christine Zink M, Mankowski JL. Paving the path to HIV neurotherapy: Predicting SIV CNS disease. Eur J Pharmacol 2015; 759:303-12. [PMID: 25818747 DOI: 10.1016/j.ejphar.2015.03.018] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Revised: 01/23/2015] [Accepted: 03/12/2015] [Indexed: 12/31/2022]
Abstract
HIV-induced damage to the CNS remains a major challenge for over 30 million people in the world despite the successes of combined antiretroviral therapy in limiting viral replication. Predicting development and progression of HIV-associated CNS disease is crucial because prevention and early intervention could be more effective than attempts to promote repair. The SIV/macaque model is the premier platform to study HIV neuropathogenesis, including discovery of predictive factors such as neuroprotective host genes and both blood and CSF biomarkers that precede and predict development of SIV CNS disease. This report details the role of macaque MHC class I genes, longitudinal alterations in biomarkers in the circulation, and expression of inflammatory and neuronal damage markers in CSF using samples from SIV-inoculated pigtailed macaques collected during acute, asymptomatic, and terminal stages of infection.
Collapse
Affiliation(s)
- Sarah E Beck
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States
| | - Suzanne E Queen
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States
| | - Kenneth W Witwer
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States
| | - Kelly A Metcalf Pate
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States
| | - Lisa M Mangus
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States
| | - Lucio Gama
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States
| | - Robert J Adams
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States
| | - Janice E Clements
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States; Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States
| | - M Christine Zink
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States; Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States
| | - Joseph L Mankowski
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States; Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States.
| |
Collapse
|
9
|
TGFβ-Mediated Downregulation of Thrombopoietin Is Associated With Platelet Decline in Asymptomatic SIV Infection. J Acquir Immune Defic Syndr 2014; 65:510-6. [PMID: 24220290 DOI: 10.1097/qai.0000000000000048] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
BACKGROUND Thrombocytopenia is a known consequence of HIV infection, and decreased production of platelets has been previously implicated in the pathogenesis of platelet decline during asymptomatic infection. Thrombopoietin (THPO) drives platelet production by stimulating the maturation of bone marrow megakaryocytes and can be transcriptionally downregulated by cytokines that are increased during infection such as transforming growth factor β (TGFβ) and platelet factor 4 (pf4). DESIGN To determine whether transcriptional downregulation of THPO contributed to decreased platelet production during asymptomatic infection in the simian immunodeficiency virus (SIV)/macaque model of HIV, we compared hepatic THPO mRNA levels to platelet number and megakaryocyte density. To identify potential inhibitory factors that decrease THPO transcription during asymptomatic infection, we measured TGFβ and pf4 plasma levels. To determine whether combined antiretroviral therapy (cART) could correct platelet decline by altering cytokine levels, we measured TGFβ and pf4 in cART-treated SIV-infected macaques and compared these values to cART-untreated SIV-infected macaques. RESULTS Hepatic THPO transcription was downregulated during asymptomatic SIV infection concurrent with platelet decline. Hepatic THPO mRNA levels correlated with bone marrow megakaryocyte density. In contrast, plasma TGFβ levels were inversely correlated with hepatic THPO transcription and bone marrow megakaryocyte density. With cART treatment, plasma TGFβ levels and platelet count returned to values similar to those in uninfected macaques. CONCLUSIONS TGFβ-mediated downregulation of hepatic THPO may lead to decline in platelet number during asymptomatic SIV infection, and cART may prevent platelet decline by normalizing plasma TGFβ levels.
Collapse
|
10
|
Abstract
Despite their small size and anucleate status, platelets have diverse roles in vascular biology. Not only are platelets the cellular mediator of thrombosis, but platelets are also immune cells that initiate and accelerate many vascular inflammatory conditions. Platelets are linked to the pathogenesis of inflammatory diseases such as atherosclerosis, malaria infection, transplant rejection, and rheumatoid arthritis. In some contexts, platelet immune functions are protective, whereas in others platelets contribute to adverse inflammatory outcomes. In this review, we will discuss platelet and platelet-derived mediator interactions with the innate and acquired arms of the immune system and platelet-vessel wall interactions that drive inflammatory disease. There have been many recent publications indicating both important protective and adverse roles for platelets in infectious disease. Because of this new accumulating data, and the fact that infectious disease continues to be a leading cause of death globally, we will also focus on new and emerging concepts related to platelet immune and inflammatory functions in the context of infectious disease.
Collapse
|
11
|
Metcalf Pate KA, Lyons CE, Dorsey JL, Shirk EN, Queen SE, Adams RJ, Gama L, Morrell CN, Mankowski JL. Platelet activation and platelet-monocyte aggregate formation contribute to decreased platelet count during acute simian immunodeficiency virus infection in pig-tailed macaques. J Infect Dis 2013; 208:874-83. [PMID: 23852120 DOI: 10.1093/infdis/jit278] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Platelets are key participants in innate immune responses to pathogens. As a decrease in circulating platelet count is one of the initial hematologic indicators of human immunodeficiency virus (HIV) infection, we sought to determine whether decline in platelet number during acute infection results from decreased production, increased antibody-mediated destruction, or increased platelet activation in a simian immunodeficiency virus (SIV)/macaque model. During acute SIV infection, circulating platelets were activated with increased surface expression of P-selection, CD40L and major histocompatibility complex class I. Platelet production was maintained and platelet autoantibodies were not detected during acute infection. Concurrent with a decrease in platelet numbers and an increase in circulating monocytes, platelets were found sequestered in platelet-monocyte aggregates, thereby contributing to the decline in platelet counts. Because the majority of circulating CD16(+) monocytes formed complexes with platelets during acute SIV infection, a decreased platelet count may represent platelet participation in the innate immune response to HIV.
Collapse
Affiliation(s)
- Kelly A Metcalf Pate
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
12
|
A plasma microRNA signature of acute lentiviral infection: biomarkers of central nervous system disease. AIDS 2011; 25:2057-67. [PMID: 21857495 DOI: 10.1097/qad.0b013e32834b95bf] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
OBJECTIVE Plasma microRNAs (miRNAs) are modulated during disease and are emerging biomarkers; they have not been characterized in HIV infection. Using our macaque/simian immunodeficiency virus (SIV) model of HIV, we sought to identify a plasma miRNA profile of acute lentiviral infection, evaluate its relationship with known cellular and viral determinants of lentivirus-associated central nervous system (CNS) disease, and explore the potential of miRNAs to predict CNS disease. DESIGN Plasma samples were obtained before inoculation and 10 days after inoculation from SIV-infected macaques. METHODS Plasma miRNA expression profiles were determined by TaqMan low-density array for six individuals. miRNA expression was compared with levels of cytokines, virus, and plasma platelet count. miRNA results were confirmed by single miRNA-specific assays for 10 macaques. Nineteen individuals were used to validate a disease prediction test. RESULTS A 45-miRNA signature of acute infection (differential expression with P < 0.05 after multiple comparison correction) classified plasma as infected or not. Several differentially expressed miRNAs correlated with CNS disease-associated cytokines interleukin-6 and CCL2 and included predicted and/or validated regulators of the corresponding mRNAs. miRNAs tracked with viral load and platelet count were also predictors of CNS disease. At least six miRNAs were significantly differentially expressed in individuals with severe versus no CNS disease; in an unweighted expression test, they predicted CNS disease. CONCLUSION Acute-phase differential expression of plasma miRNAs predicts CNS disease and suggests that CNS damage or predisposition to disease progression begins in the earliest phase of infection. Plasma miRNAs should be investigated further as leading indicators of HIV diseases as early as acute infection.
Collapse
|
13
|
Metcalf Pate KA, Mankowski JL. HIV and SIV Associated Thrombocytopenia: An Expanding Role for Platelets in the Pathogenesis of HIV. DRUG DISCOVERY TODAY. DISEASE MECHANISMS 2011; 8:e25-e32. [PMID: 22577463 PMCID: PMC3346281 DOI: 10.1016/j.ddmec.2011.10.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/15/2022]
Abstract
Thrombocytopenia is common in HIV and SIV infection, and is often associated with disease progression. HIV and SIV-associated thrombocytopenia arise through multiple mechanisms, including decreased platelet production, increased platelet destruction due to HIV-mimetic anti-platelet antibodies, and increased use of activated platelets. Activated platelets have the potential to contribute to the pathogenesis of HIV and SIV by interacting directly with inflammatory cells and endothelium and by releasing soluble immunomodulatory cytokines.
Collapse
Affiliation(s)
- Kelly A Metcalf Pate
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore MD, 21205
| | | |
Collapse
|
14
|
Reeves RK, Wei Q, Stallworth J, Fultz PN. Systemic dendritic cell mobilization associated with administration of FLT3 ligand to SIV- and SHIV-infected macaques. AIDS Res Hum Retroviruses 2009; 25:1313-28. [PMID: 20001520 DOI: 10.1089/aid.2009.0053] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Reports indicate that myeloid and plasmacytoid dendritic cells (mDCs and pDCs), which are key effector cells in host innate immune responses, can be infected with HIV-1 and are reduced in number and function during the chronic phase of HIV disease. Furthermore, it was recently demonstrated that a sustained loss of mDCs and pDCs occurs in SIV-infected macaques. Since loss of functional DC populations might impair innate immune responses to opportunistic microorganisms and neoplastic cells, we explored whether inoculation of naive and SIV- or SHIV-infected pigtailed macaques with the hematopoietic cytokine FLT3-ligand (FLT3-L) would expand the number of mDCs and pDCs in vivo. After the macaques received supraphysiologic doses of FLT3-L, mDCs, pDCs, and monocytes increased up to 45-fold in blood, lymph nodes, and bone marrow (BM), with DC expansion in the BM preceding mobilization in blood and lymphoid tissues. FLT3-L also increased serum levels of IL-12, at least transiently, and elicited higher surface expression of HLA-DR and the activation markers CD25 and CD69 on NK and T cells. During and after treatment of infected animals, APCs increased in number and were activated; however, CD4(+) T cell numbers, virion RNA, and anti-SIV/SHIV antibody titers remained relatively stable, suggesting that FLT3-L might be a safe modality to expand DC populations and provide therapeutic benefit during chronic lentivirus infections.
Collapse
Affiliation(s)
- R. Keith Reeves
- Department of Microbiology, University of Alabama at Birmingham (UAB), Birmingham, Alabama 35294
| | - Qing Wei
- Department of Microbiology, University of Alabama at Birmingham (UAB), Birmingham, Alabama 35294
| | - Jackie Stallworth
- Department of Microbiology, University of Alabama at Birmingham (UAB), Birmingham, Alabama 35294
| | - Patricia N. Fultz
- Department of Microbiology, University of Alabama at Birmingham (UAB), Birmingham, Alabama 35294
| |
Collapse
|
15
|
Thrombocytopenia is strongly associated with simian AIDS in pigtail macaques. J Acquir Immune Defic Syndr 2009; 51:374-9. [PMID: 19461525 DOI: 10.1097/qai.0b013e3181a9cbcf] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Simian AIDS has a variable time course and presentation making it difficult to define disease effects of progressive simian immunodeficiency virus (SIV) infection. We commonly observed thrombocytopenia (TCP) associated with progressive SIV infection of pigtail macaques (Macaca nemestrina). We therefore analyzed the relationship between platelet counts, viral load (VL), and CD4 T-cell levels in 44 unselected macaques with chronic SIV infection. Persistent TCP was observed in 70% of pigtail macaques infected with SIVmac251 for up to 77 weeks in the absence of clinically significant bleeding. The presence of TCP correlated with higher SIV plasma VLs and depressed total and memory CD4 T cells. TCP was more common in macaques requiring euthanasia for incipient AIDS than macaques that survived to the end of the studies, although VL and CD4 T-cell decline were stronger independent predictors of AIDS-free survival. There was however no clear correlation between the development of TCP and immune activation as measured by plasma soluble CD14. We conclude that TCP is a useful end point to analyze SIV studies in pigtail macaques.
Collapse
|
16
|
Míguez-Burbano MJ, Nair M, Lewis JE, Fishman J. The role of alcohol on platelets, thymus and cognitive performance among HIV-infected subjects: are they related? Platelets 2009; 20:260-7. [PMID: 19459132 PMCID: PMC4933293 DOI: 10.1080/09537100902964759] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Our objective was to evaluate whether thrombocytopenia and small thymus volume, which may be associated with hazardous alcohol consumption, are predictors of cognitive performance after highly-active antiretroviral treatment (HAART). To achieve this goal 165 people living with HIV starting HAART underwent thymus magnetic resonance imaging, cognitive (HIV Dementia Score [HDS] and the California Verbal Learning Test [CVLT]), immune and laboratory assessments at baseline and after 6 months of HAART. At baseline, hazardous alcohol consumption was significantly correlated with both thymus size (r = -0.44, p = 0.003) and thrombocytopenia (r = 0.28, p = 0.001). Of interest, thrombocytopenic patients were characterized by a smaller thymus size. Individuals with and without cognitive impairment differed in alcohol consumption, platelet counts and thymus size, suggesting that they may be risk factors for neurological abnormalities. In fact, after HAART hazardous alcohol use associations with thrombocytopenia were related to cognitive decline (learning = -0.2 +/- 0.8, recall = -0.3 +/- 0.1 and HDS = -0.5). This contrasted with improvements on every cognitive measure (learning = 1.6 +/- 0.3, p = 0.0001, recall = 2.2 +/- 0.4, p = 0.0001 and HDS = 1.0, p = 0.05) in those with neither alcohol use nor thrombocytopenia. In adjusted analyses for sociodemographics, adherence and immune measurements, reduced thymus size was associated with a 90% and thrombocytopenia with a 70% increase in the risk of scoring in the demented range after HAART (RR = 1.9, p < 0.05 and RR = 1.7, p = 0.03) and with low CVLT scores (thymus volume RR = 2.0, p = 0.04, chronic alcohol use p = 0.05 and thrombocytopenia p = 0.06). Thymus volume and platelet counts were negatively affected by alcohol and were predictors of cognitive performance and improvements after HAART. These results could have important clinical and therapeutic implications.
Collapse
|
17
|
Venneti S, Bonneh-Barkay D, Lopresti BJ, Bissel SJ, Wang G, Mathis CA, Piatak M, Lifson JD, Nyaundi JO, Murphey-Corb M, Wiley CA. Longitudinal in vivo positron emission tomography imaging of infected and activated brain macrophages in a macaque model of human immunodeficiency virus encephalitis correlates with central and peripheral markers of encephalitis and areas of synaptic degeneration. THE AMERICAN JOURNAL OF PATHOLOGY 2008; 172:1603-16. [PMID: 18467697 DOI: 10.2353/ajpath.2008.070967] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Human immunodeficiency virus encephalitis is characterized by infiltration of the brain with infected and activated macrophages; however, it is not known why disease occurs after variable lengths of infection in 25% of immunosuppressed acquired immune deficiency syndrome patients. We determined in vivo correlates (in peripheral blood and the central nervous system) for the development and progression of lentiviral encephalitis by longitudinally following infected and activated macrophages in the brain using positron emission tomography (PET). Using human postmortem brain tissues from both lentivirus-infected encephalitic patients and cell culture systems, we showed that the PET ligand [(3)H](R)-PK11195 bound specifically to virus-infected and activated macrophages. We longitudinally imaged infected and activated brain macrophages in a cohort of macaques infected with simian immunodeficiency virus using [(11)C](R)-PK11195. [(11)C](R)-PK11195 retention in vivo in the brain correlated with viral burden in the brain and cerebrospinal fluid, and with regions of both presynaptic and postsynaptic damage. Finally, longitudinal changes in [(11)C](R)-PK11195 retention in the brain in vivo correlated with changes in circulating monocytes as well as in both natural killer and memory CD4(+) T cells in the periphery. Our results suggest that development and progression of simian immunodeficiency virus encephalitis in vivo correlates with changes in specific cell subtypes in the periphery. A combination of PET imaging and the assessment of these peripheral immune parameters may facilitate longitudinal assessment of lentiviral encephalitis in living patients as well as evaluation of therapeutic efficacies.
Collapse
Affiliation(s)
- Sriram Venneti
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Clay CC, Rodrigues DS, Ho YS, Fallert BA, Janatpour K, Reinhart TA, Esser U. Neuroinvasion of fluorescein-positive monocytes in acute simian immunodeficiency virus infection. J Virol 2007; 81:12040-8. [PMID: 17715237 PMCID: PMC2168770 DOI: 10.1128/jvi.00133-07] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Monocytes and macrophages play a central role in the pathogenesis of human immunodeficiency virus (HIV)-associated dementia. They represent prominent targets for HIV infection and are thought to facilitate viral neuroinvasion and neuroinflammatory processes. However, many aspects regarding monocyte brain recruitment in HIV infection remain undefined. The nonhuman primate model of AIDS is uniquely suited for examination of the role of monocytes in the pathogenesis of AIDS-associated encephalitis. Nevertheless, an approach to monitor cell migration from peripheral blood into the central nervous system (CNS) in primates had been lacking. Here, upon autologous transfer of fluorescein dye-labeled leukocytes, we demonstrate the trafficking of dye-positive monocytes into the choroid plexus stromata and perivascular spaces in the cerebra of rhesus macaques acutely infected with simian immunodeficiency virus between days 12 and 14 postinfection (p.i.). Dye-positive cells that had migrated expressed the monocyte activation marker CD16 and the macrophage marker CD68. Monocyte neuroinvasion coincided with the presence of the virus in brain tissue and cerebrospinal fluid and with the induction of the proinflammatory mediators CXCL9/MIG and CCL2/MCP-1 in the CNS. Prior to neuroinfiltration, plasma viral load levels peaked on day 11 p.i. Furthermore, the numbers of peripheral blood monocytes rapidly increased between days 4 and 8 p.i., and circulating monocytes exhibited increased functional capacity to produce CCL2/MCP-1. Our findings demonstrate acute monocyte brain infiltration in an animal model of AIDS. Such studies facilitate future examinations of the migratory profile of CNS-homing monocytes, the role of monocytes in virus import into the brain, and the disruption of blood-cerebrospinal fluid and blood-brain barrier functions in primates.
Collapse
Affiliation(s)
- Candice C Clay
- Department of Pathology and Laboratory Medicine, Research III Building, Room 3400A, University of California-Davis Medical Center, 4645 2nd Avenue, Sacramento, CA 95817, USA
| | | | | | | | | | | | | |
Collapse
|
19
|
Sui Z, Sniderhan LF, Schifitto G, Phipps RP, Gelbard HA, Dewhurst S, Maggirwar SB. Functional Synergy between CD40 Ligand and HIV-1 Tat Contributes to Inflammation: Implications in HIV Type 1 Dementia. THE JOURNAL OF IMMUNOLOGY 2007; 178:3226-36. [PMID: 17312171 DOI: 10.4049/jimmunol.178.5.3226] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
HIV type 1 (HIV-1)-associated dementia (HAD) is believed to occur due to aberrant activation of monocyte-derived macrophages and brain-resident microglial cells by viral proteins as well as by the proinflammatory mediators released by infected cells. To investigate the inflammatory aspects of the disease, we examined the levels of soluble CD40L (sCD40L) in paired samples of plasma and cerebrospinal fluid obtained from 25 HIV-infected individuals. A significantly higher level of sCD40L was detected in both cerebrospinal fluid and plasma from HIV-infected patients with cognitive impairment, compared with their nonimpaired counterparts. The contribution of sCD40L to the pathogenesis of HAD was then examined by in vitro experiments. rCD40L synergized with HIV-1 Tat to increase TNF-alpha release from primary human monocytes and microglia, in an NF-kappaB-dependent manner. The mechanistic basis for this synergism was attributed to a Tat-mediated up-regulation of CD40 in monocytes and microglia. Finally, the CD40L-mediated increase in TNF-alpha production by monocytes was shown to be biologically important; immunodepletion experiments revealed that TNF-alpha was essential for the neurotoxic effects of conditioned medium recovered from Tat/CD40L-treated monocytes. Taken together, our results show that CD40 signaling in microglia and monocytes can synergize with the effects of Tat, further amplifying inflammatory processes within the CNS and influencing neuronal survival.
Collapse
Affiliation(s)
- Ziye Sui
- Interdepartmental Program in Neuroscience, University of Rochester School of Medicine, 601 Elmwood Avenue, Rochester, NY 14642, USA
| | | | | | | | | | | | | |
Collapse
|