1
|
Castell NJ, Abreu CM, Shirk EN, Queen SE, Mankowski JL, Clements JE, Veenhuis RT. SIV-specific antibodies protect against inflammasome-driven encephalitis in untreated macaques. Cell Rep 2024; 43:114833. [PMID: 39383041 PMCID: PMC11552693 DOI: 10.1016/j.celrep.2024.114833] [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: 04/26/2024] [Revised: 08/26/2024] [Accepted: 09/19/2024] [Indexed: 10/11/2024] Open
Abstract
Viral encephalitis is a growing public health threat with limited diagnostic and treatment options. Simian immunodeficiency virus (SIV)-infected macaques are an established model for human immunodeficiency virus (HIV), and approximately 60% of untreated pigtail macaques rapidly progress to characteristic SIV encephalitis (SIVE). The immune responses of SIV-infected macaques are investigated in plasma, cerebrospinal fluid (CSF), and brain tissue to determine correlates with SIVE pathology. Macaques with SIVE show myeloid-dominant brain lesions with inflammasome activation in infected and bystander cells, as assessed by interleukin (IL)-1β, IL-18, and apoptosis-associated speck-like protein containing a caspase activation and recruitment domain (ASC), and elevations in monocyte chemoattractant protein (MCP)-1, macrophage inflammatory protein (MIP)-1α, and tumor necrosis factor alpha (TNF-α). SIV-specific immunoglobulin (Ig)G in plasma and CSF is predictive of SIVE as early as 21 days post-inoculation; animals with SIVE continue to show negligible seroconversion 3 months after infection. This dichotomy in immune responses, wherein some macaques fail to initiate robust IgG responses and subsequently develop SIVE, provides insight into the pathogenesis and heterogeneous outcomes in viral encephalitis.
Collapse
Affiliation(s)
- Natalie J Castell
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Celina M Abreu
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Erin N Shirk
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Suzanne E Queen
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Joseph L Mankowski
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Janice E Clements
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Rebecca T Veenhuis
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
2
|
Deshetty UM, Ray S, Singh S, Buch S, Periyasamy P. Opioid abuse and SIV infection in non-human primates. J Neurovirol 2023; 29:377-388. [PMID: 37418108 PMCID: PMC10729652 DOI: 10.1007/s13365-023-01153-z] [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: 03/08/2023] [Revised: 06/06/2023] [Accepted: 06/14/2023] [Indexed: 07/08/2023]
Abstract
Human immunodeficiency virus (HIV) and drug abuse are intertwined epidemics, leading to compromised adherence to combined antiretroviral therapy (cART) and exacerbation of NeuroHIV. As opioid abuse causes increased viral replication and load, leading to a further compromised immune system in people living with HIV (PLWH), it is paramount to address this comorbidity to reduce the NeuroHIV pathogenesis. Non-human primates are well-suited models to study mechanisms involved in HIV neuropathogenesis and provide a better understanding of the underlying mechanisms involved in the comorbidity of HIV and drug abuse, leading to the development of more effective treatments for PLWH. Additionally, using broader behavioral tests in these models can mimic mild NeuroHIV and aid in studying other neurocognitive diseases without encephalitis. The simian immunodeficiency virus (SIV)-infected rhesus macaque model is instrumental in studying the effects of opioid abuse on PLWH due to its similarity to HIV infection. The review highlights the importance of using non-human primate models to study the comorbidity of opioid abuse and HIV infection. It also emphasizes the need to consider modifiable risk factors such as gut homeostasis and pulmonary pathogenesis associated with SIV infection and opioid abuse in this model. Moreover, the review suggests that these non-human primate models can also be used in developing effective treatment strategies for NeuroHIV and opioid addiction. Therefore, non-human primate models can significantly contribute to understanding the complex interplay between HIV infection, opioid abuse, and associated comorbidities.
Collapse
Affiliation(s)
- Uma Maheswari Deshetty
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198-5880, USA
| | - Sudipta Ray
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198-5880, USA
| | - Seema Singh
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198-5880, USA
| | - Shilpa Buch
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198-5880, USA.
| | - Palsamy Periyasamy
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198-5880, USA.
| |
Collapse
|
3
|
Harvey W, Hutto EH, Chilton JA, Chamanza R, Mysore JV, Parry NM, Dick E, Wojcinski ZW, Piaia A, Garcia B, Flandre TD, Pardo ID, Cramer S, Wright JA, Bradley AE. Infectious diseases of non-human primates. SPONTANEOUS PATHOLOGY OF THE LABORATORY NON-HUMAN PRIMATE 2023:15-69. [DOI: 10.1016/b978-0-12-813088-9.00020-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
|
4
|
Haddad A, Voth B, Brooks J, Swang M, Carryl H, Algarzae N, Taylor S, Parker C, Van Rompay KKA, De Paris K, Burke MW. Reduced neuronal population in the dorsolateral prefrontal cortex in infant macaques infected with simian immunodeficiency virus (SIV). J Neurovirol 2021; 27:923-935. [PMID: 34554407 PMCID: PMC8901521 DOI: 10.1007/s13365-021-01019-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 08/11/2021] [Accepted: 08/26/2021] [Indexed: 11/29/2022]
Abstract
Pediatric HIV infection remains a global health crisis with an estimated 150,000 new mother-to-child (MTCT) infections each year. Antiretroviral therapy (ART) has improved childhood survival, but only an estimated 53% of children worldwide have access to treatment. Adding to the health crisis is the neurological impact of HIV on the developing brain, in particular cognitive and executive function, which persists even when ART is available. Imaging studies suggest structural, connectivity, and functional alterations in perinatally HIV-infected youth. However, the paucity of histological data limits our ability to identify specific cortical regions that may underlie the clinical manifestations. Utilizing the pediatric simian immunodeficiency virus (SIV) infection model in infant macaques, we have previously shown that early-life SIV infection depletes the neuronal population in the hippocampus. Here, we expand on these previous studies to investigate the dorsolateral prefrontal cortex (dlPFC). A total of 11 ART-naïve infant rhesus macaques (Macaca mulatta) from previous studies were retrospectively analyzed. Infant macaques were either intravenously (IV) inoculated with highly virulent SIVmac251 at ~1 week of age and monitored for 6-10 weeks or orally challenged with SIVmac251 from week 9 of age onwards with a monitoring period of 10-23 weeks post-infection (19-34 weeks of age), and SIV-uninfected controls were euthanized at 16-17 weeks of age. Both SIV-infected groups show a significant loss of neurons along with evidence of ongoing neuronal death. Oral- and IV-infected animals showed a similar neuronal loss which was negatively correlated to chronic viremia levels as assessed by an area under the curve (AUC) analysis. The loss of dlPFC neurons may contribute to the rapid neurocognitive decline associated with pediatric HIV infection.
Collapse
Affiliation(s)
- Alexandra Haddad
- Department of Physiology and Biophysics, Howard University, Washington, DC, 20059, USA
| | - Brittany Voth
- Department of Physiology and Biophysics, Howard University, Washington, DC, 20059, USA
| | - Janiya Brooks
- Department of Physiology and Biophysics, Howard University, Washington, DC, 20059, USA
| | - Melanie Swang
- Department of Physiology and Biophysics, Howard University, Washington, DC, 20059, USA
| | - Heather Carryl
- Department of Physiology and Biophysics, Howard University, Washington, DC, 20059, USA
| | - Norah Algarzae
- Department of Physiology and Biophysics, Howard University, Washington, DC, 20059, USA
- King Saudi University, Riyadh, Riyadh, Kingdom of Saudi Arabia
| | - Shane Taylor
- Department of Physiology and Biophysics, Howard University, Washington, DC, 20059, USA
| | - Camryn Parker
- Department of Physiology and Biophysics, Howard University, Washington, DC, 20059, USA
| | - Koen K A Van Rompay
- California National Primate Research Center, University of California Davis, Davis, CA, 95616, USA
| | - Kristina De Paris
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Mark W Burke
- Department of Physiology and Biophysics, Howard University, Washington, DC, 20059, USA.
| |
Collapse
|
5
|
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: 15] [Impact Index Per Article: 3.8] [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
|
6
|
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
|
7
|
Knight AC, Brill SA, Solis CV, Richardson MR, McCarron ME, Queen SE, Bailey CC, Mankowski JL. Differential regulation of TREM2 and CSF1R in CNS macrophages in an SIV/macaque model of HIV CNS disease. J Neurovirol 2020; 26:511-519. [PMID: 32488843 PMCID: PMC7442592 DOI: 10.1007/s13365-020-00844-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 02/11/2020] [Accepted: 04/13/2020] [Indexed: 11/26/2022]
Abstract
HIV-associated neuroinflammation is primarily driven by CNS macrophages including microglia. Regulation of these immune responses, however, remains to be characterized in detail. Using the SIV/macaque model of HIV, we evaluated CNS expression of triggering receptor expressed on myeloid cells 2 (TREM2) which is constitutively expressed by microglia and contributes to cell survival, proliferation, and differentiation. Loss-of-function mutations in TREM2 are recognized risk factors for neurodegenerative diseases including Alzheimer's disease (AD), amyotrophic lateral sclerosis (ALS), and Nasu-Hakola disease (NHD); recent reports have also indicated a role for TREM2 in HIV-associated neuroinflammation. Using in situ hybridization (ISH) and qRT-PCR, TREM2 mRNA levels were found to be significantly elevated in frontal cortex of macaques with SIV encephalitis compared with uninfected controls (P = 0.02). TREM2 protein levels were also elevated as measured by ELISA of frontal cortex tissue homogenates in these animals. Previously, we characterized the expression of CSF1R (colony-stimulating factor 1 receptor) in this model; the TREM2 and CSF1R promoters both contain a PU.1 binding site. While TREM2 and CSF1R mRNA levels in the frontal cortex were highly correlated (Spearman R = 0.79, P < 0.001), protein levels were not well correlated. In SIV-infected macaques released from ART to study viral rebound, neither TREM2 nor CSF1R mRNA increased with rebound viremia. However, CSF1R protein levels remained significantly elevated unlike TREM2 (P = 0.02). This differential expression suggests that TREM2 and CSF1R play unique, distinct roles in the pathogenesis of HIV CNS disease.
Collapse
MESH Headings
- Animals
- Antiretroviral Therapy, Highly Active/methods
- Antiviral Agents/pharmacology
- Drug Administration Schedule
- Encephalitis, Viral/drug therapy
- Encephalitis, Viral/genetics
- Encephalitis, Viral/immunology
- Encephalitis, Viral/virology
- Frontal Lobe/drug effects
- Frontal Lobe/immunology
- Frontal Lobe/virology
- Gene Expression Regulation
- Host-Pathogen Interactions/genetics
- Host-Pathogen Interactions/immunology
- Macaca nemestrina/genetics
- Macaca nemestrina/immunology
- Macaca nemestrina/virology
- Macrophages/drug effects
- Macrophages/immunology
- Macrophages/virology
- Male
- Membrane Glycoproteins/genetics
- Membrane Glycoproteins/immunology
- Microglia/drug effects
- Microglia/immunology
- Microglia/virology
- Promoter Regions, Genetic
- Protein Binding
- Proto-Oncogene Proteins/genetics
- Proto-Oncogene Proteins/immunology
- RNA, Messenger/genetics
- RNA, Messenger/immunology
- Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/genetics
- Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/immunology
- Simian Acquired Immunodeficiency Syndrome/drug therapy
- Simian Acquired Immunodeficiency Syndrome/genetics
- Simian Acquired Immunodeficiency Syndrome/immunology
- Simian Acquired Immunodeficiency Syndrome/virology
- Simian Immunodeficiency Virus/drug effects
- Simian Immunodeficiency Virus/growth & development
- Simian Immunodeficiency Virus/immunology
- Trans-Activators/genetics
- Trans-Activators/immunology
Collapse
Affiliation(s)
- Audrey C Knight
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Samuel A Brill
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Clarisse V Solis
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Morgan R Richardson
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Megan E McCarron
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Suzanne E Queen
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Charles C Bailey
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
- Emmune, Inc., 130 Scripps Way, Jupiter, Florida, USA
| | - Joseph L Mankowski
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University, School of Medicine, Baltimore, MD, USA.
- Department of Neurology, Johns Hopkins University, School of Medicine, Baltimore, MD, USA.
- Department of Pathology, Johns Hopkins University, School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
8
|
de Boer ECW, van Gils JM, van Gils MJ. Ephrin-Eph signaling usage by a variety of viruses. Pharmacol Res 2020; 159:105038. [PMID: 32565311 DOI: 10.1016/j.phrs.2020.105038] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 06/15/2020] [Accepted: 06/16/2020] [Indexed: 12/20/2022]
Abstract
Ephrin-Eph signaling is a receptor tyrosine kinase signaling pathway involved in a variety of cellular mechanisms, of which many are related to the adhesion or migration of cells. Both the Eph receptor and ephrin ligand are abundantly present on a wide variety of cell types, and strongly evolutionary conserved. This review provides an overview of how 18 genetically diverse viruses utilize the Eph receptor (Eph), ephrin ligand (ephrin) or ephrin-Eph signaling to their advantage in their viral life cycle. Both Ephs and ephrins have been shown to serve as entry receptors for a variety of viruses, via both membrane fusion and endocytosis. Ephs and ephrins are also involved in viral transmission by vectors, associated with viral replication or persistence and lastly to neurological damage caused by viral infection. Although therapeutic opportunities targeting Ephs or ephrins do not seem feasible yet, the current research does propose two models for the viral usage of ephrin-Eph signaling. Firstly, the viral entry model, in which membrane molecules are used for viral entry, leading to cells being used for replication or as a transporter. Secondly, the advantageous expression ephrin-Eph signaling model, where viruses adapt the expression of Ephs or ephrins to change cell-cell interaction to their advantage. These models can guide future research questions on the usage of Ephs or ephrins by viruses and therapeutic opportunities.
Collapse
Affiliation(s)
- Esther C W de Boer
- Department of Medical Microbiology, Amsterdam UMC, Amsterdam Infection & Immunity Institute, University of Amsterdam, Meibergdreef 9, Amsterdam, the Netherlands
| | - Janine M van Gils
- Einthoven Laboratory for Vascular and Regenerative Medicine, Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands.
| | - Marit J van Gils
- Department of Medical Microbiology, Amsterdam UMC, Amsterdam Infection & Immunity Institute, University of Amsterdam, Meibergdreef 9, Amsterdam, the Netherlands.
| |
Collapse
|
9
|
Abreu CM, Veenhuis RT, Avalos CR, Graham S, Parrilla DR, Ferreira EA, Queen SE, Shirk EN, Bullock BT, Li M, Metcalf Pate KA, Beck SE, Mangus LM, Mankowski JL, Mac Gabhann F, O'Connor SL, Gama L, Clements JE. Myeloid and CD4 T Cells Comprise the Latent Reservoir in Antiretroviral Therapy-Suppressed SIVmac251-Infected Macaques. mBio 2019; 10:e01659-19. [PMID: 31431552 PMCID: PMC6703426 DOI: 10.1128/mbio.01659-19] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 07/24/2019] [Indexed: 12/13/2022] Open
Abstract
Human immunodeficiency virus (HIV) eradication or long-term suppression in the absence of antiretroviral therapy (ART) requires an understanding of all viral reservoirs that could contribute to viral rebound after ART interruption. CD4 T cells (CD4s) are recognized as the predominant reservoir in HIV type 1 (HIV-1)-infected individuals. However, macrophages are also infected by HIV-1 and simian immunodeficiency virus (SIV) during acute infection and may persist throughout ART, contributing to the size of the latent reservoir. We sought to determine whether tissue macrophages contribute to the SIVmac251 reservoir in suppressed macaques. Using cell-specific quantitative viral outgrowth assays (CD4-QVOA and MΦ-QVOA), we measured functional latent reservoirs in CD4s and macrophages in ART-suppressed SIVmac251-infected macaques. Spleen, lung, and brain in all suppressed animals contained latently infected macrophages, undetectable or low-level SIV RNA, and detectable SIV DNA. Silent viral genomes with potential for reactivation and viral spread were also identified in blood monocytes, although these cells might not be considered reservoirs due to their short life span. Additionally, virus produced in the MΦ-QVOA was capable of infecting healthy activated CD4s. Our results strongly suggest that functional latent reservoirs in CD4s and macrophages can contribute to viral rebound and reestablishment of productive infection after ART interruption. These findings should be considered in the design and implementation of future HIV cure strategies.IMPORTANCE This study provides further evidence that the latent reservoir is comprised of both CD4+ T cells and myeloid cells. The data presented here suggest that CD4+ T cells and macrophages found throughout tissues in the body can contain replication-competent SIV and contribute to rebound of the virus after treatment interruption. Additionally, we have shown that monocytes in blood contain latent virus and, though not considered a reservoir themselves due to their short life span, could contribute to the size of the latent reservoir upon entering the tissue and differentiating into long-lived macrophages. These new insights into the size and location of the SIV reservoir using a model that is heavily studied in the HIV field could have great implications for HIV-infected individuals and should be taken into consideration with the development of future HIV cure strategies.
Collapse
Affiliation(s)
- Celina M Abreu
- Department of Molecular and Comparative Pathobiology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Rebecca T Veenhuis
- Department of Molecular and Comparative Pathobiology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Claudia R Avalos
- Department of Molecular and Comparative Pathobiology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Shelby Graham
- Department of Molecular and Comparative Pathobiology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Daymond R Parrilla
- Department of Molecular and Comparative Pathobiology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Edna A Ferreira
- Department of Molecular and Comparative Pathobiology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Suzanne E Queen
- Department of Molecular and Comparative Pathobiology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Erin N Shirk
- Department of Molecular and Comparative Pathobiology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Brandon T Bullock
- Department of Molecular and Comparative Pathobiology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Ming Li
- Department of Molecular and Comparative Pathobiology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Kelly A Metcalf Pate
- Department of Molecular and Comparative Pathobiology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Sarah E Beck
- Department of Molecular and Comparative Pathobiology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Lisa M Mangus
- Department of Molecular and Comparative Pathobiology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Joseph L Mankowski
- Department of Molecular and Comparative Pathobiology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Feilim Mac Gabhann
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland, USA
- Institute for Computational Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Shelby L O'Connor
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Lucio Gama
- Department of Molecular and Comparative Pathobiology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, Bethesda, Maryland, USA
| | - Janice E Clements
- Department of Molecular and Comparative Pathobiology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
10
|
The Association of Immune Markers with Cognitive Performance in South African HIV-Positive Patients. J Neuroimmune Pharmacol 2019; 14:679-687. [PMID: 31388873 DOI: 10.1007/s11481-019-09870-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 07/28/2019] [Indexed: 10/26/2022]
Abstract
Dysregulated expression of neuro-immune markers has previously been linked to HIV-associated neurocognitive impairment. We undertook an exploratory approach in a HIV clade-C cohort, investigating the association between eight immune markers and neurocognitive performance in 99 HIV+ and 51 HIV- participants. Markers were selected on preliminary and putative evidence of their link to key neuro-immune functions. Cognitive performance was established using a battery of tests sensitive to HIV-associated neurocognitive impairment, with domain-based scores utilized in analysis. The markers Thymidine phosphorylase (TYMP) and Neutrophil gelatinase-associated lipocalin (NGAL) were significantly higher while Matrix Metalloproteinase (MMP)9 was significantly lower in HIV+ participants. Our results further showed that in the HIV+ group, worse psychomotor processing speed was associated with higher TYMP and NGAL levels and worse motor function was associated with higher NGAL levels. Future studies should explore the underlying mechanisms of these markers in HIV-associated neurocognitive impairment. Graphical Abstract The association of peripheral immune markers with neurocognitive performance in South African HIV-positive patients.
Collapse
|
11
|
Delery E, Bohannon DG, Irons DL, Allers C, Sugimoto C, Cai Y, Merino KM, Amedee AM, Veazey RS, MacLean A, Kuroda MJ, Kim WK. Lack of susceptibility in neonatally infected rhesus macaques to simian immunodeficiency virus-induced encephalitis. J Neurovirol 2019; 25:578-588. [PMID: 31119711 PMCID: PMC6751025 DOI: 10.1007/s13365-019-00755-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 02/15/2019] [Accepted: 04/23/2019] [Indexed: 12/18/2022]
Abstract
Despite combination antiretroviral therapies making HIV a chronic rather than terminal condition for many people, the prevalence of HIV-associated neurocognitive disorders (HAND) is increasing. This is especially problematic for children living with HIV. Children diagnosed HAND rarely display the hallmark pathology of HIV encephalitis in adults, namely infected macrophages and multinucleated giant cells in the brain. This finding has also been documented in rhesus macaques infected perinatally with simian immunodeficiency virus (SIV). However, the extent and mechanisms of lack of susceptibility to encephalitis in perinatally HIV-infected children remain unclear. In the current study, we compared brains of macaques infected with pathogenic strains of SIV at different ages to determine neuropathology, correlates of neuroinflammation, and potential underlying mechanisms. Encephalitis was not found in the macaques infected within 24 h of birth despite similar high plasma viral load and high monocyte turnover. Macaques developed encephalitis only when they were infected after 4 months of age. Lower numbers of CCR5-positive cells in the brain, combined with a less leaky blood-brain barrier, may be responsible for the decreased virus infection in the brain and consequently the absence of encephalitis in newborn macaques infected with SIV.
Collapse
MESH Headings
- Age Factors
- Animals
- Animals, Newborn
- Blood-Brain Barrier/immunology
- Blood-Brain Barrier/pathology
- Blood-Brain Barrier/virology
- Brain Stem/immunology
- Brain Stem/pathology
- Brain Stem/virology
- Capillary Permeability/immunology
- Disease Resistance
- Encephalitis, Viral/genetics
- Encephalitis, Viral/immunology
- Encephalitis, Viral/pathology
- Encephalitis, Viral/virology
- Frontal Lobe/immunology
- Frontal Lobe/pathology
- Frontal Lobe/virology
- Gene Expression
- Macaca mulatta/virology
- Macrophages/immunology
- Macrophages/pathology
- Macrophages/virology
- Monocytes/immunology
- Monocytes/pathology
- Monocytes/virology
- RNA, Viral/genetics
- RNA, Viral/metabolism
- Receptors, CCR5/genetics
- Receptors, CCR5/immunology
- Receptors, Virus/genetics
- Receptors, Virus/immunology
- Simian Acquired Immunodeficiency Syndrome/genetics
- Simian Acquired Immunodeficiency Syndrome/immunology
- Simian Acquired Immunodeficiency Syndrome/pathology
- Simian Acquired Immunodeficiency Syndrome/virology
- Simian Immunodeficiency Virus/pathogenicity
- Simian Immunodeficiency Virus/physiology
- Viral Load
Collapse
Affiliation(s)
- Elizabeth Delery
- Tulane National Primate Research Center, Covington, LA, USA
- Department of Microbiology & Immunology, Tulane Medical School, New Orleans, LA, USA
| | - Diana G Bohannon
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, VA, USA
| | - Derek L Irons
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, VA, USA
| | | | - Chie Sugimoto
- Tulane National Primate Research Center, Covington, LA, USA
- Dokkyo Medical University, Tochigi, Japan
| | - Yanhui Cai
- Tulane National Primate Research Center, Covington, LA, USA
- The Wistar Institute, Philadelphia, PA, USA
| | | | - Angela M Amedee
- Louisiana State University School of Medicine, New Orleans, LA, USA
| | | | - Andrew MacLean
- Tulane National Primate Research Center, Covington, LA, USA
- Department of Microbiology & Immunology, Tulane Medical School, New Orleans, LA, USA
- Tulane Brain Institute, New Orleans, LA, USA
- Tulane Center for Aging, New Orleans, LA, USA
| | - Marcelo J Kuroda
- Tulane National Primate Research Center, Covington, LA, USA
- University of California Davis, Davis, CA, USA
| | - Woong-Ki Kim
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, VA, USA.
| |
Collapse
|
12
|
Knight AC, Brill SA, Queen SE, Tarwater PM, Mankowski JL. Increased Microglial CSF1R Expression in the SIV/Macaque Model of HIV CNS Disease. J Neuropathol Exp Neurol 2019; 77:199-206. [PMID: 29319809 DOI: 10.1093/jnen/nlx115] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Chronic microglial activation and associated neuroinflammation are key factors in neurodegenerative diseases including HIV-associated neurocognitive disorders. Colony stimulating factor 1 receptor (CSF1R)-mediated signaling is constitutive in cells of the myeloid lineage, including microglia, promoting cell survival, proliferation, and differentiation. In amyotrophic lateral sclerosis and Alzheimers disease, CSF1R is upregulated. Inhibiting CSF1R signaling in animal models of these diseases improved disease outcomes. In our studies, CNS expression of the CSF1R ligand, colony-stimulating factor 1 (CSF1) was significantly increased in a SIV/macaque model of HIV CNS disease. Using a Nanostring nCounter immune panel, we found CSF1 overexpression was strongly correlated with upregulation of microglial genes involved in antiviral and oxidative stress responses. Using in situ hybridization, we found that CSF1R mRNA was only present in Iba-1 positive microglia. By ELISA and immunostaining with digital image analysis, SIV-infected macaques had significantly higher CSF1R levels in frontal cortex than uninfected macaques (p = 0.018 and p = 0.02, respectively). SIV-infected macaques treated with suppressive ART also had persistently elevated CSF1R similar to untreated SIV-infected macaques. Coordinate upregulation of CSF1 and CSF1R expression implicates this signaling pathway in progressive HIV CNS disease.
Collapse
Affiliation(s)
- Audrey C Knight
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Samuel A Brill
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Suzanne E Queen
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Patrick M Tarwater
- Department of Biostatistics, UTHealth School of Public Health, El Paso, Texas
| | - Joseph L Mankowski
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
13
|
Abstract
Viral infection in the brain can be acute or chronic, with the responses often producing foci of increasingly cytotoxic inflammation. This can lead to effects beyond the central nervous system (CNS). To stimulate discussion, this commentary addresses four questions: What drives the development of human immunodeficiency virus (HIV)-associated neurocognitive disorders, does the phenotype of macrophages in the CNS spur development of HIV encephalitis (HIVE), does continual activation of astrocytes drive the development of HIV-associated neurocognitive disorders/subclinical disease, and neuroinflammation: friend or foe? A unifying theory that connects each question is the issue of continued activation of glial cells, even in the apparent absence of simian immunodeficiency virus/HIV in the CNS. As the CNS innate immune system is distinct from the rest of the body, it is likely there could be a number of activation profiles not observed elsewhere.
Collapse
Affiliation(s)
- Elizabeth C. Delery
- Tulane National Primate Research Center, Covington, Louisiana
- Tulane Program in Biomedical Sciences, Tulane Medical School, New Orleans, Louisiana
- Department of Microbiology and Immunology, Tulane Medical School, New Orleans, Louisiana
| | - Andrew G. MacLean
- Tulane National Primate Research Center, Covington, Louisiana
- Tulane Program in Biomedical Sciences, Tulane Medical School, New Orleans, Louisiana
- Department of Microbiology and Immunology, Tulane Medical School, New Orleans, Louisiana
- Tulane Brain Institute, Tulane University, New Orleans, Louisiana
- Center for Aging, School of Medicine, Tulane University, New Orleans, Louisiana
| |
Collapse
|
14
|
Bissel SJ, Gurnsey K, Jedema HP, Smith NF, Wang G, Bradberry CW, Wiley CA. Aged Chinese-origin rhesus macaques infected with SIV develop marked viremia in absence of clinical disease, inflammation or cognitive impairment. Retrovirology 2018; 15:17. [PMID: 29391069 PMCID: PMC5796498 DOI: 10.1186/s12977-018-0400-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Accepted: 01/19/2018] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Damage to the central nervous system during HIV infection can lead to variable neurobehavioral dysfunction termed HIV-associated neurocognitive disorders (HAND). There is no clear consensus regarding the neuropathological or cellular basis of HAND. We sought to study the potential contribution of aging to the pathogenesis of HAND. Aged (range = 14.7-24.8 year) rhesus macaques of Chinese origin (RM-Ch) (n = 23) were trained to perform cognitive tasks. Macaques were then divided into four groups to assess the impact of SIVmac251 infection (n = 12) and combined antiretroviral therapy (CART) (5 infected; 5 mock-infected) on the execution of these tasks. RESULTS Aged SIV-infected RM-Ch demonstrated significant plasma viremia and modest CSF viral loads but showed few clinical signs, no elevations of systemic temperature, and no changes in activity levels, platelet counts or weight. Concentrations of biomarkers of acute and chronic inflammation such as soluble CD14, CXCL10, IL-6 and TNF-α are known to be elevated following SIV infection of young adult macaques of several species, but concentrations of these biomarkers did not shift after SIV infection in aged RM-Ch and remained similar to mock-infected macaques. Neither acute nor chronic SIV infection or CART had a significant impact on accuracy, speed or percent completion in a sensorimotor test. CONCLUSIONS Viremia in the absence of a chronic elevated inflammatory response seen in some aged RM-Ch is reminiscent of SIV infection in natural disease resistant hosts. The absence of cognitive impairment during SIV infection in aged RM-Ch might be in part attributed to diminishment of some facets of the immunological response. Additional study encompassing species and age differences is necessary to substantiate this hypothesis.
Collapse
Affiliation(s)
- Stephanie J. Bissel
- University of Pittsburgh, 3550 Terrace Street, S758 Scaife Hall, Pittsburgh, PA 15261 USA
| | - Kate Gurnsey
- University of Pittsburgh, 3550 Terrace Street, S758 Scaife Hall, Pittsburgh, PA 15261 USA
| | - Hank P. Jedema
- University of Pittsburgh, 3550 Terrace Street, S758 Scaife Hall, Pittsburgh, PA 15261 USA
- Present Address: National Institute on Drug Abuse, 251 Bayview Boulevard, Baltimore, MD 21224 USA
| | - Nicholas F. Smith
- University of Pittsburgh, 3550 Terrace Street, S758 Scaife Hall, Pittsburgh, PA 15261 USA
| | - Guoji Wang
- University of Pittsburgh, 3550 Terrace Street, S758 Scaife Hall, Pittsburgh, PA 15261 USA
| | - Charles W. Bradberry
- University of Pittsburgh, 3550 Terrace Street, S758 Scaife Hall, Pittsburgh, PA 15261 USA
- Veterans Affairs Pittsburgh Healthcare System, 4100 Allequippa Street, Pittsburgh, PA 15213 USA
- Present Address: National Institute on Drug Abuse, 251 Bayview Boulevard, Baltimore, MD 21224 USA
| | - Clayton A. Wiley
- University of Pittsburgh, 3550 Terrace Street, S758 Scaife Hall, Pittsburgh, PA 15261 USA
| |
Collapse
|
15
|
Gelman BB, Endsley J, Kolson D. When do models of NeuroAIDS faithfully imitate "the real thing"? J Neurovirol 2017; 24:146-155. [PMID: 29256039 PMCID: PMC5910470 DOI: 10.1007/s13365-017-0601-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 11/03/2017] [Accepted: 11/08/2017] [Indexed: 02/07/2023]
Abstract
HIV-infected patients treated with antiretroviral medicines (ART) still face neurological challenges. HIV-associated neurocognitive disturbances (HAND) can occur, and latent viral DNA persisting in the central nervous system (CNS) prevents eradication of HIV. This communication focuses on how to develop experimental models of HAND and CNS HIV latency that best imitate the CNS pathophysiology in diseased humans, which we take to be “the real thing.” Models of HIV encephalitis (HIVE) with active CNS viral replication were developed in the early years of the AIDS pandemic. The clinical relevancy of such models is in sharp decline because HIVE seldom occurs in virally suppressed patients, while HAND remains common. The search for improved models of HAND should incorporate the neurochemical, neuroimmunological and neuropathological features of virally suppressed patients. Common anomalies in these patients as established in autopsy brain specimens include brain endothelial cell activation and neurochemical imbalances of synaptic transmission; classical neurodegeneration may not be as crucial. With regard to latent HIV with viral suppression, human brain specimens show that the pool of latent proviral HIV DNA in the CNS is relatively small relative to the total body pool and does not change substantially over years. The CNS pool of latent virus probably differs from lymphoid tissues, because the mononuclear phagocyte system sustains productive infection (versus lymphocytes). These and yet-to-be discovered aspects of the human CNS of virally suppressed patients need to be better defined and addressed in experimental models. To maintain clinical relevancy, models of HAND and viral latency should faithfully emulate “the real thing.”
Collapse
Affiliation(s)
- Benjamin B Gelman
- Department of Pathology, Route 0419, University of Texas Medical Branch, Galveston, TX, 77555-0419, USA.
| | - Janice Endsley
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Dennis Kolson
- Department of Neurology, University of Pennsylvania, Philadelphia, PA, 19104-6140, USA
| |
Collapse
|
16
|
Mangus LM, Beck SE, Queen SE, Brill SA, Shirk EN, Metcalf Pate KA, Muth DC, Adams RJ, Gama L, Clements JE, Mankowski JL. Lymphocyte-Dominant Encephalitis and Meningitis in Simian Immunodeficiency Virus-Infected Macaques Receiving Antiretroviral Therapy. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 188:125-134. [PMID: 29229308 DOI: 10.1016/j.ajpath.2017.08.035] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Revised: 08/17/2017] [Accepted: 08/28/2017] [Indexed: 01/21/2023]
Abstract
A retrospective neuropathologic review of 30 SIV-infected pigtailed macaques receiving combination antiretroviral therapy (cART) was conducted. Seventeen animals with lymphocyte-dominant inflammation in the brain and/or meninges that clearly was morphologically distinct from prototypic SIV encephalitis and human immunodeficiency virus encephalitis were identified. Central nervous system (CNS) infiltrates in cART-treated macaques primarily comprised CD20+ B cells and CD3+ T cells with fewer CD68+ macrophages. Inflammation was associated with low levels of SIV RNA in the brain as shown by in situ hybridization, and generally was observed in animals with episodes of cerebrospinal fluid (CSF) viral rebound or sustained plasma and CSF viremia during treatment. Although the lymphocytic CNS inflammation in these macaques shared morphologic characteristics with uncommon immune-mediated neurologic disorders reported in treated HIV patients, including CNS immune reconstitution inflammatory syndrome and neurosymptomatic CSF escape, the high prevalence of CNS lesions in macaques suggests that persistent adaptive immune responses in the CNS also may develop in neuroasymptomatic or mildly impaired HIV patients yet remain unrecognized given the lack of access to CNS tissue for histopathologic evaluation. Continued investigation into the mechanisms and outcomes of CNS inflammation in cART-treated, SIV-infected macaques will advance our understanding of the consequences of residual CNS HIV replication in patients on cART, including the possible contribution of adaptive immune responses to HIV-associated neurocognitive disorders.
Collapse
Affiliation(s)
- Lisa M Mangus
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Sarah E Beck
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Suzanne E Queen
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Samuel A Brill
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Erin N Shirk
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Kelly A Metcalf Pate
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Dillon C Muth
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Robert J Adams
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Lucio Gama
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Janice E Clements
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Joseph L Mankowski
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland.
| |
Collapse
|
17
|
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
|
18
|
Rahimian P, He JJ. HIV/neuroAIDS biomarkers. Prog Neurobiol 2017; 157:117-132. [PMID: 27084354 PMCID: PMC5705228 DOI: 10.1016/j.pneurobio.2016.04.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Revised: 04/01/2016] [Accepted: 04/08/2016] [Indexed: 12/12/2022]
Abstract
HIV infection often causes neurological symptoms including cognitive and motor dysfunction, which have been collectively termed HIV/neuroAIDS. Neuropsychological assessment and clinical symptoms have been the primary diagnostic criteria for HIV/neuroAIDS, even for the mild cognitive and motor disorder, the most prevalent form of HIV/neuroAIDS in the era of combination antiretroviral therapy. Those performance-based assessments and symptoms are generally descriptive and do not have the sensitivity and specificity to monitor the diagnosis, progression, and treatment response of the disease when compared to objective and quantitative laboratory-based biological markers, or biomarkers. In addition, effects of demographics and comorbidities such as substance abuse, psychiatric disease, nutritional deficiencies, and co-infection on HIV/neuroAIDS could be more readily determined using biomarkers than using neuropsychological assessment and clinical symptoms. Thus, there have been great efforts in identification of HIV/neuroAIDS biomarkers over the past two decades. The need for reliable biomarkers of HIV/neuroAIDS is expected to increase as the HIV-infected population ages and their vulnerability to neurodegenerative diseases, particularly Alzheimer's disease increases. Currently, three classes of HIV/neuroAIDS biomarkers are being pursued to establish objective laboratory-based definitions of HIV-associated neurologic injury: cerebrospinal fluid biomarkers, blood biomarkers, and neuroimaging biomarkers. In this review, we will focus on the current knowledge in the field of HIV/neuroAIDS biomarker discovery.
Collapse
Affiliation(s)
- Pejman Rahimian
- Department of Cell Biology and Immunology, Graduate School of Biomedical Sciences, University of North Texas Health Science Center, Fort Worth, TX 76107, United States
| | - Johnny J He
- Department of Cell Biology and Immunology, Graduate School of Biomedical Sciences, University of North Texas Health Science Center, Fort Worth, TX 76107, United States.
| |
Collapse
|
19
|
Eaton SL, Wishart TM. Bridging the gap: large animal models in neurodegenerative research. Mamm Genome 2017; 28:324-337. [PMID: 28378063 PMCID: PMC5569151 DOI: 10.1007/s00335-017-9687-6] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 03/25/2017] [Indexed: 01/08/2023]
Abstract
The world health organisation has declared neurological disorders as one of the greatest public health risks in the world today. Yet, despite this growing concern, the mechanisms underpinning many of these conditions are still poorly understood. This may in part be due to the seemingly diverse nature of the initiating insults ranging from genetic (such as the Ataxia's and Lysosomal storage disorders) through to protein misfolding and aggregation (i.e. Prions), and those of a predominantly unknown aetiology (i.e. Alzheimer's and Parkinson's disease). However, efforts to elucidate mechanistic regulation are also likely to be hampered because of the complexity of the human nervous system, the apparent selective regional vulnerability and differential degenerative progression. The key to elucidating these aetiologies is determining the regional molecular cascades, which are occurring from the early through to terminal stages of disease progression. Whilst much molecular data have been captured at the end stage of disease from post-mortem analysis in humans, the very early stages of disease are often conspicuously asymptomatic, and even if they were not, repeated sampling from multiple brain regions of "affected" patients and "controls" is neither ethical nor possible. Model systems therefore become fundamental for elucidating the mechanisms governing these complex neurodegenerative conditions. However, finding a model that precisely mimics the human condition can be challenging and expensive. Whilst cellular and invertebrate models are frequently used in neurodegenerative research and have undoubtedly yielded much useful data, the comparatively simplistic nature of these systems makes insights gained from such a stand alone model limited when it comes to translation. Given the recent advances in gene editing technology, the options for novel model generation in higher order species have opened up new and exciting possibilities for the field. In this review, we therefore explain some of the reasons why larger animal models often appear to give a more robust recapitulation of human neurological disorders and why they may be a critical stepping stone for effective therapeutic translation.
Collapse
Affiliation(s)
- S L Eaton
- Roslin Institute and Royal (Dick) Veterinary studies, University of Edinburgh, Easter Bush Campus, Edinburgh, EH25 9RG, UK
| | - T M Wishart
- Roslin Institute and Royal (Dick) Veterinary studies, University of Edinburgh, Easter Bush Campus, Edinburgh, EH25 9RG, UK.
- Euan MacDonald Centre for MND Research, Chancellor's Building, 49 Little France, Edinburgh, EH16 4SB, UK.
| |
Collapse
|
20
|
Carroll A, Brew B. HIV-associated neurocognitive disorders: recent advances in pathogenesis, biomarkers, and treatment. F1000Res 2017; 6:312. [PMID: 28413625 PMCID: PMC5365228 DOI: 10.12688/f1000research.10651.1] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/22/2017] [Indexed: 12/21/2022] Open
Abstract
HIV-associated neurocognitive disorders (HAND) remain prevalent despite plasma viral suppression by antiretroviral agents. In fact, the prevalence of milder subtypes of cognitive impairment is increasing. Neuropsychologic testing remains the "gold standard" of diagnosis; however, this is time consuming and costly in a resource-poor environment. Recently developed screening tools, such as CogState and the revised HIV dementia scale, have very good sensitivity and specificity in the more severe stages of HAND. However, questions remain regarding the utility of, optimal population for, and insensitivity of tests in mild HAND. Recognition of ongoing viral persistence and the inflammatory milieu in the central nervous system (CNS) has advanced our understanding of the pathogenesis of HAND and facilitated the development of biomarkers of CNS disease. The importance of the monocyte-macrophage lineage cell and the astrocyte as viral reservoirs, HIV viral proteins, self-perpetuating CNS inflammation, and CCR5 chemokine receptor neurotropism has been identified. Whilst biomarkers demonstrate monocyte activation, inflammation, and neuronal injury, they remain limited in their clinical utility. The improved understanding of pathogenic mechanisms has led to novel approaches to the treatment of HAND; however, despite these advances, the optimal management is still undefined.
Collapse
Affiliation(s)
- Antonia Carroll
- Department of Neurology, St Vincent’s Hospital, Level 4, Xavier Building, Victoria Street, Darlinghurst, Sydney, Australia
- University of New South Wales, St. Vincent’s Clinical School, Delacy Building, Victoria Street, Darlinghurst, Sydney, Australia
| | - Bruce Brew
- Department of Neurology, St Vincent’s Hospital, Level 4, Xavier Building, Victoria Street, Darlinghurst, Sydney, Australia
- Peter Duncan Neurosciences Unit, St Vincent’s Centre for Applied Medical Research, St Vincent’s Hospital, Sydney, Australia
- Department of HIV Medicine, St Vincent’s Hospital, Level 4, Xavier Building, Victoria Street, Darlinghurst, Sydney, Australia
- University of New South Wales, St. Vincent’s Clinical School, Delacy Building, Victoria Street, Darlinghurst, Sydney, Australia
| |
Collapse
|
21
|
Abstract
OBJECTIVE Resting CD4 T cells have been recognized as the major cell reservoir of latent HIV-1 during antiretroviral therapy (ART). Using an simian immunodeficiency virus (SIV)/macaque model for AIDS and HIV-related neurocognitive disorders we assessed the contribution of the brain to viral latency and reactivation. DESIGN Pigtailed macaques were dual inoculated with SIVDeltaB670 and SIV17E-Fr and treated with an efficacious central nervous system-penetrant ART. After 500 days of viral suppression animals were treated with two cycles of latency reversing agents and increases in viral transcripts were examined. METHODS Longitudinal plasma and cerebrospinal fluid (CSF) viral loads were analyzed by quantitative and digital droplet PCR. After necropsy, viral transcripts in organs were analyzed by PCR, in-situ hybridization, and phylogenetic genotyping based on env V1 loop sequences. Markers for neuronal damage and CSF activation were measured by ELISA. RESULTS Increases in activation markers and plasma and CSF viral loads were observed in one animal treated with latency reversing agents, despite ongoing ART. SIV transcripts were identified in occipital cortex macrophages by in-situ hybridization and CD68 staining. The most abundant SIV genotype in CSF was unique and expanded independent from viruses found in the periphery. CONCLUSION The central nervous system harbors latent SIV genomes after long-term viral suppression by ART, indicating that the brain represents a potential viral reservoir and should be seriously considered during AIDS cure strategies.
Collapse
|
22
|
Non-human primate models of SIV infection and CNS neuropathology. Curr Opin Virol 2016; 19:92-8. [PMID: 27544476 DOI: 10.1016/j.coviro.2016.07.012] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Revised: 07/15/2016] [Accepted: 07/29/2016] [Indexed: 02/03/2023]
Abstract
Non-human primate models of AIDS and neuroAIDS are the premiere model of HIV infection of the CNS and neuropathogenesis. This review discusses current SIV infection models of neuroAIDS emphasizing findings in the last two years. Consistent in these findings is the interplay between host factors that regulate immune responses to virus and viral replication. Several rapid models of AIDS with consistent CNS pathogenesis exist, each of which modulates by antibody treatment or viruses that cause rapid immune suppression and replicate well in macrophages. Consistent in all of these models are data underscoring the importance of monocyte and macrophage activation, infection and accumulation in the CNS.
Collapse
|
23
|
Saylor D, Dickens AM, Sacktor N, Haughey N, Slusher B, Pletnikov M, Mankowski JL, Brown A, Volsky DJ, McArthur JC. HIV-associated neurocognitive disorder--pathogenesis and prospects for treatment. Nat Rev Neurol 2016; 12:234-48. [PMID: 26965674 DOI: 10.1038/nrneurol.2016.27] [Citation(s) in RCA: 628] [Impact Index Per Article: 69.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
In the past two decades, several advancements have improved the care of HIV-infected individuals. Most importantly, the development and deployment of combination antiretroviral therapy (CART) has resulted in a dramatic decline in the rate of deaths from AIDS, so that people living with HIV today have nearly normal life expectancies if treated with CART. The term HIV-associated neurocognitive disorder (HAND) has been used to describe the spectrum of neurocognitive dysfunction associated with HIV infection. HIV can enter the CNS during early stages of infection, and persistent CNS HIV infection and inflammation probably contribute to the development of HAND. The brain can subsequently serve as a sanctuary for ongoing HIV replication, even when systemic viral suppression has been achieved. HAND can remain in patients treated with CART, and its effects on survival, quality of life and everyday functioning make it an important unresolved issue. In this Review, we describe the epidemiology of HAND, the evolving concepts of its neuropathogenesis, novel insights from animal models, and new approaches to treatment. We also discuss how inflammation is sustained in chronic HIV infection. Moreover, we suggest that adjunctive therapies--treatments targeting CNS inflammation and other metabolic processes, including glutamate homeostasis, lipid and energy metabolism--are needed to reverse or improve HAND-related neurological dysfunction.
Collapse
Affiliation(s)
- Deanna Saylor
- Department of Neurology, Johns Hopkins University School of Medicine, Meyer 6113, 600 N Wolfe St, Baltimore, Maryland 21287, USA
| | - Alex M Dickens
- Department of Neurology, Johns Hopkins University School of Medicine, Meyer 6113, 600 N Wolfe St, Baltimore, Maryland 21287, USA
| | - Ned Sacktor
- Department of Neurology, Johns Hopkins University School of Medicine, Meyer 6113, 600 N Wolfe St, Baltimore, Maryland 21287, USA
| | - Norman Haughey
- Department of Neurology, Johns Hopkins University School of Medicine, Meyer 6113, 600 N Wolfe St, Baltimore, Maryland 21287, USA
| | - Barbara Slusher
- Department of Neurology, Johns Hopkins University School of Medicine, Meyer 6113, 600 N Wolfe St, Baltimore, Maryland 21287, USA
| | - Mikhail Pletnikov
- Department of Neurology, Johns Hopkins University School of Medicine, Meyer 6113, 600 N Wolfe St, Baltimore, Maryland 21287, USA
| | - Joseph L Mankowski
- Department of Neurology, Johns Hopkins University School of Medicine, Meyer 6113, 600 N Wolfe St, Baltimore, Maryland 21287, USA
| | - Amanda Brown
- Department of Neurology, Johns Hopkins University School of Medicine, Meyer 6113, 600 N Wolfe St, Baltimore, Maryland 21287, USA
| | - David J Volsky
- The Icahn School of Medicine at Mount Sinai, 1468 Madison Avenue, New York, New York 10029, USA
| | - Justin C McArthur
- Department of Neurology, Johns Hopkins University School of Medicine, Meyer 6113, 600 N Wolfe St, Baltimore, Maryland 21287, USA
| |
Collapse
|
24
|
SAMHD1 transcript upregulation during SIV infection of the central nervous system does not associate with reduced viral load. Sci Rep 2016; 6:22629. [PMID: 26936683 PMCID: PMC4776177 DOI: 10.1038/srep22629] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Accepted: 02/18/2016] [Indexed: 02/01/2023] Open
Abstract
Restriction of HIV-1 in myeloid-lineage cells is attributed in part to the nucleotidase activity of the SAM-domain and HD-domain containing protein (SAMHD1), which depletes free nucleotides, blocking reverse transcription. In the same cells, the Vpx protein of HIV-2 and most SIVs counteracts SAMHD1. Both Type I and II interferons may stimulate SAMHD1 transcription. The contributions of SAMHD1 to retroviral restriction in the central nervous system (CNS) have been the subject of limited study. We hypothesized that SAMHD1 would respond to interferon in the SIV-infected CNS but would not control virus due to SIV Vpx. Accordingly, we investigated SAMHD1 transcript abundance and association with the Type I interferon response in an SIV model. SAMHD1 transcript levels were IFN responsive, increasing during acute phase infection and decreasing during a more quiescent phase, but generally remaining elevated at all post-infection time points. In vitro, SAMHD1 transcript was abundant in macaque astrocytes and further induced by Type I interferon, while IFN produced a weaker response in the more permissive environment of the macrophage. We cannot rule out a contribution of SAMHD1 to retroviral restriction in relatively non-permissive CNS cell types. We encourage additional research in this area, particularly in the context of HIV-1 infection.
Collapse
|
25
|
Witwer KW, Buchanan EL, Myers SL, McAlexander MA. miRNAs and SAMHD1 regulation in vitro and in a model of HIV CNS disease. J Neuroinflammation 2015; 12:159. [PMID: 26337901 PMCID: PMC4559003 DOI: 10.1186/s12974-015-0380-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Accepted: 08/17/2015] [Indexed: 11/17/2022] Open
Abstract
Pilakka-Kanthikeel et al. recently reported higher levels of the retroviral restriction factor sterile alpha motif and histidine/aspartic acid domain-containing protein 1 (SAMHD1) in astrocytes than in microglia, suggesting that SAMHD1 levels might explain in part the relatively refractory nature of astrocytes to retroviral replication. These findings are consistent with our studies of simian and human immunodeficiency virus infection of astrocytes and macrophages. Similarly, a role for two host microRNAs in post-transcriptional regulation of SAMHD1 agrees with our in vitro results and those of others. However, data from an animal model of HIV neurologic disorders may not be consistent with robust miRNA-mediated regulation of SAMHD1 in vivo.
Collapse
Affiliation(s)
- Kenneth W Witwer
- Department of Molecular and Comparative Pathobiology, The Johns Hopkins University School of Medicine, 733 North Broadway, BRB Suite 831, Baltimore, MD, 21025, USA.
| | - Erin L Buchanan
- Department of Molecular and Comparative Pathobiology, The Johns Hopkins University School of Medicine, 733 North Broadway, BRB Suite 831, Baltimore, MD, 21025, USA.
| | - Stephanie L Myers
- Department of Molecular and Comparative Pathobiology, The Johns Hopkins University School of Medicine, 733 North Broadway, BRB Suite 831, Baltimore, MD, 21025, USA.
| | - Melissa A McAlexander
- Department of Molecular and Comparative Pathobiology, The Johns Hopkins University School of Medicine, 733 North Broadway, BRB Suite 831, Baltimore, MD, 21025, USA.
| |
Collapse
|