1
|
Griffin DE. Measles virus persistence and its consequences. Curr Opin Virol 2020; 41:46-51. [PMID: 32387998 PMCID: PMC7492426 DOI: 10.1016/j.coviro.2020.03.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 03/13/2020] [Accepted: 03/19/2020] [Indexed: 11/16/2022]
Abstract
Clearance of measles virus is complex. Infectious virus is cleared by the adaptive immune response manifested by the characteristic maculopapular rash. CD8+ T cells are major effectors of infectious virus clearance, a process that may fail in individuals with compromised cellular immune responses leading to progressive giant cell pneumonia and/or measles inclusion body encephalitis. In contrast to the usual rapid clearance of infectious virus, clearance of viral RNA is slow with persistence in lymphoid tissue for many months. Persistence of MeV RNA may contribute to the late development of the slowly progressive disease subacute sclerosing panencephalitis in children infected at a young age and to measles-associated immune suppression but also to maturation of the immune response and development of life-long immunity.
Collapse
Affiliation(s)
- Diane E Griffin
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA.
| |
Collapse
|
2
|
Ene L. Human Immunodeficiency Virus in the Brain-Culprit or Facilitator? Infect Dis (Lond) 2018; 11:1178633717752687. [PMID: 29467577 PMCID: PMC5815409 DOI: 10.1177/1178633717752687] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Accepted: 12/15/2017] [Indexed: 01/21/2023] Open
Abstract
Introduction: Human immunodeficiency virus (HIV) enters the brain early, where it can persist, evolve, and become compartmentalized. Central nervous system (CNS) disease can be attributed to HIV alone or to the complex interplay between the virus and other neurotropic pathogens. Aim: The current review aims to describe the direct impact of HIV on the brain as well as its relationship with other pathogens from a practitioner’s perspective, to provide a general clinical overview, brief workup, and, whenever possible, treatment guidance. Methods: A review of PubMed was conducted to identify studies on neuropathogenesis of HIV in relation to host responses. Furthermore, the interaction between the CNS pathogens and the host damage responses were revised in the setting of advanced and also well-controlled HIV infection. Results: Similar to other pathogens, HIV leads to CNS immune activation, inflammation, and viral persistence. Therefore, almost half of the infected individuals present with neurocognitive disorders, albeit mild. Compartmentalized HIV in the CNS can be responsible in a minority of cases for the dramatic presentation of symptomatic HIV escape. Disruption of the immune system secondary to HIV may reactivate latent infections or allow new pathogens to enter the CNS. Opportunistic infections with an inflammatory component are associated with elevated HIV loads in the cerebrospinal fluid and also with greater cognitive impairment. The inflammatory immune reconstitution syndrome associated with CNS opportunistic infections can be a life-threatening condition, which needs to be recognized and managed by efficiently controlling the pathogen burden and timely balanced combination antiretroviral therapy. Latent neurotropic pathogens can reactivate in the brain and mimic HIV-associated severe neurological diseases or contribute to neurocognitive impairment in the setting of stable HIV infection. Conclusions: As HIV can be responsible for considerable brain damage directly or by facilitating other pathogens, more effort is needed to recognize and manage HIV-associated CNS disorders and to eventually target HIV eradication from the brain.
Collapse
Affiliation(s)
- Luminita Ene
- HIV Department, "Dr. Victor Babes" Hospital for Infectious and Tropical Diseases, Bucharest, Romania
| |
Collapse
|
3
|
Barrios CS, Castillo L, Giam CZ, Wu L, Beilke MA. Inhibition of HIV type 1 replication by human T lymphotropic virus types 1 and 2 Tax proteins in vitro. AIDS Res Hum Retroviruses 2013; 29:1061-7. [PMID: 23464580 DOI: 10.1089/aid.2013.0027] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Patients with HIV-1 and human T-lymphotropic virus type 2 (HTLV-2) coinfections often exhibit a clinical course similar to that seen in HIV-1-infected individuals who are long-term nonprogressors. These findings have been attributed in part to the ability of HTLV-2 to activate production of antiviral chemokines and to downregulate the CCR5 coreceptor on lymphocytes. To further investigate these observations, we tested the ability of recombinant Tax1 and Tax2 proteins to suppress HIV-1 viral replication in vitro. R5-tropic HIV-1 (NLAD8)-infected peripheral blood mononuclear cells (PBMCs) were treated daily with recombinant Tax1 and Tax2 proteins (dosage range 1-100 pM). Culture supernatants were collected at intervals from days 1 to 22 postinfection and assayed for levels of HIV-1 p24 antigen by ELISA. Treatment of PBMCs with Tax2 protein resulted in a significant reduction in HIV-1 p24 antigen levels (p<0.05) at days 10, 14, and 18 postinfection compared to HIV-1-infected or mock-treated PBMCs. This was preceded by the detection of increased levels of CC-chemokines MIP-1α/CCL3, MIP-1β/CCL4, and RANTES/CCL5 on days 1-7 of infection. Similar, but less robust inhibition was observed in Tax1-treated PBMCs. These results support the contention that Tax1 and Tax2 play a role in generating antiviral responses against HIV-1 in vivo and in vitro.
Collapse
Affiliation(s)
- Christy S. Barrios
- Department of Medicine, Division of Infectious Diseases, Medical College of Wisconsin, Milwaukee, Wisconsin
- Clement J. Zablocki Veterans Affairs Medical Center, Milwaukee, Wisconsin
| | - Laura Castillo
- Department of Medicine, Division of Infectious Diseases, Medical College of Wisconsin, Milwaukee, Wisconsin
- Clement J. Zablocki Veterans Affairs Medical Center, Milwaukee, Wisconsin
| | - Chou-Zen Giam
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | - Li Wu
- Center for Retrovirus Research, Department of Veterinary Biosciences, The Ohio State University, Columbus, Ohio
| | - Mark A. Beilke
- Department of Medicine, Division of Infectious Diseases, Medical College of Wisconsin, Milwaukee, Wisconsin
- Clement J. Zablocki Veterans Affairs Medical Center, Milwaukee, Wisconsin
| |
Collapse
|
4
|
Mitsuki YY, Terahara K, Shibusawa K, Yamamoto T, Tsuchiya T, Mizukoshi F, Ishige M, Okada S, Kobayashi K, Morikawa Y, Nakayama T, Takeda M, Yanagi Y, Tsunetsugu-Yokota Y. HIV-1 infection ex vivo accelerates measles virus infection by upregulating signaling lymphocytic activation molecule (SLAM) in CD4+ T cells. J Virol 2012; 86:7227-7234. [PMID: 22532682 PMCID: PMC3416328 DOI: 10.1128/jvi.06681-11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2011] [Accepted: 04/10/2012] [Indexed: 11/20/2022] Open
Abstract
Measles virus (MV) infection in children harboring human immunodeficiency virus type 1 (HIV-1) is often fatal, even in the presence of neutralizing antibodies; however, the underlying mechanisms are unclear. Therefore, the aim of the present study was to examine the interaction between HIV-1 and wild-type MV (MVwt) or an MV vaccine strain (MVvac) during dual infection. The results showed that the frequencies of MVwt- and MVvac-infected CD4(+) T cells within the resting peripheral blood mononuclear cells (PBMCs) were increased 3- to 4-fold after HIV-1 infection, and this was associated with a marked upregulation of signaling lymphocytic activation molecule (SLAM) expression on CD4(+) T cells but not on CD8(+) T cells. SLAM upregulation was induced by infection with a replication-competent HIV-1 isolate comprising both the X4 and R5 types and to a lesser extent by a pseudotyped HIV-1 infection. Notably, SLAM upregulation was observed in HIV-infected as well as -uninfected CD4(+) T cells and was abrogated by the removal of HLA-DR(+) cells from the PBMC culture. Furthermore, SLAM upregulation did not occur in uninfected PBMCs cultured together with HIV-infected PBMCs in compartments separated by a permeable membrane, indicating that no soluble factors were involved. Rather, CD4(+) T cell activation mediated through direct contact with dendritic cells via leukocyte function-associated molecule 1 (LFA-1)/intercellular adhesion molecule 1 (ICAM-1) and LFA-3/CD2 was critical. Thus, HIV-1 infection induces a high level of SLAM expression on CD4(+) T cells, which may enhance their susceptibility to MV and exacerbate measles in coinfected individuals.
Collapse
Affiliation(s)
- Yu-ya Mitsuki
- Department of Immunology, National Institute of Infectious Diseases, Shinjuku-ku, Tokyo, Japan
| | - Kazutaka Terahara
- Department of Immunology, National Institute of Infectious Diseases, Shinjuku-ku, Tokyo, Japan
| | - Kentaro Shibusawa
- Department of Immunology, National Institute of Infectious Diseases, Shinjuku-ku, Tokyo, Japan
| | - Takuya Yamamoto
- Immunology Laboratory, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Takatsugu Tsuchiya
- Department of Immunology, National Institute of Infectious Diseases, Shinjuku-ku, Tokyo, Japan
| | - Fuminori Mizukoshi
- Department of Immunology, National Institute of Infectious Diseases, Shinjuku-ku, Tokyo, Japan
| | - Masayuki Ishige
- Department of Immunology, National Institute of Infectious Diseases, Shinjuku-ku, Tokyo, Japan
- Center for AIDS Research, Kumamoto University, Kumamoto, Japan
| | - Seiji Okada
- Center for AIDS Research, Kumamoto University, Kumamoto, Japan
| | - Kazuo Kobayashi
- Department of Immunology, National Institute of Infectious Diseases, Shinjuku-ku, Tokyo, Japan
| | - Yuko Morikawa
- Laboratory of Viral Infection II, Kitasato Institute for Life Science, Kitasato University, Tokyo, Japan
| | - Tetsuo Nakayama
- Laboratory of Viral Infection I, Kitasato Institute for Life Science, Kitasato University, Tokyo, Japan
| | - Makoto Takeda
- Department of Virology III, National Institute of Infectious Diseases, Tokyo, Japan
| | - Yusuke Yanagi
- Department of Virology, Faculty of Medicine, Kyushu University, Fukuoka, Japan
| | | |
Collapse
|
5
|
Potent host-directed small-molecule inhibitors of myxovirus RNA-dependent RNA-polymerases. PLoS One 2011; 6:e20069. [PMID: 21603574 PMCID: PMC3095640 DOI: 10.1371/journal.pone.0020069] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2011] [Accepted: 04/15/2011] [Indexed: 12/19/2022] Open
Abstract
Therapeutic targeting of host cell factors required for virus replication rather than of pathogen components opens new perspectives to counteract virus infections. Anticipated advantages of this approach include a heightened barrier against the development of viral resistance and a broadened pathogen target spectrum. Myxoviruses are predominantly associated with acute disease and thus are particularly attractive for this approach since treatment time can be kept limited. To identify inhibitor candidates, we have analyzed hit compounds that emerged from a large-scale high-throughput screen for their ability to block replication of members of both the orthomyxovirus and paramyxovirus families. This has returned a compound class with broad anti-viral activity including potent inhibition of different influenza virus and paramyxovirus strains. After hit-to-lead chemistry, inhibitory concentrations are in the nanomolar range in the context of immortalized cell lines and human PBMCs. The compound shows high metabolic stability when exposed to human S-9 hepatocyte subcellular fractions. Antiviral activity is host-cell species specific and most pronounced in cells of higher mammalian origin, supporting a host-cell target. While the compound induces a temporary cell cycle arrest, host mRNA and protein biosynthesis are largely unaffected and treated cells maintain full metabolic activity. Viral replication is blocked at a post-entry step and resembles the inhibition profile of a known inhibitor of viral RNA-dependent RNA-polymerase (RdRp) activity. Direct assessment of RdRp activity in the presence of the reagent reveals strong inhibition both in the context of viral infection and in reporter-based minireplicon assays. In toto, we have identified a compound class with broad viral target range that blocks host factors required for viral RdRp activity. Viral adaptation attempts did not induce resistance after prolonged exposure, in contrast to rapid adaptation to a pathogen-directed inhibitor of RdRp activity.
Collapse
|
6
|
Interactions between viral and prokaryotic pathogens in a mixed infection with cardiovirus and mycoplasma. J Virol 2009; 83:9940-51. [PMID: 19605479 DOI: 10.1128/jvi.01167-09] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
In the natural environment, animal and plant viruses often share ecological niches with microorganisms, but the interactions between these pathogens, although potentially having important implications, are poorly investigated. The present report demonstrates, in a model system, profound mutual effects of mycoplasma and cardioviruses in animal cell cultures. In contrast to mycoplasma-free cells, cultures contaminated with Mycoplasma hyorhinis responded to infection with encephalomyocarditis virus (EMCV), a picornavirus, but not with poliovirus (also a picornavirus), with a strong activation of a DNase(s), as evidenced by the TUNEL (terminal deoxynucleotidyltransferase-mediated dUTP-biotin nick end labeling) immunofluorescence assay and electrophoretic analysis of host DNA. This degradation was reminiscent of that observed upon apoptosis but was caspase independent, judging by the failure of the specific pan-caspase inhibitor Q-VD-OPh to prevent it. The electrophoretic mobility of the enzyme responsible for DNA degradation and dependence of its activity on ionic conditions strongly suggested that it was represented by a DNase(s) of mycoplasma origin. In cells not infected with EMCV, the relevant DNase was dormant. The possibility is discussed that activation of the mycoplasma DNase might be linked to a relatively early increase in permeability of plasma membrane of the infected cells caused by EMCV. This type of unanticipated virus-mycoplasma "cooperation" may exemplify the complexity of pathogen-host interactions under conditions when viruses and microorganisms are infecting the same host. In the course of the present study, it was also demonstrated that pan-caspase inhibitor zVAD(OMe).fmk strongly suppressed cardiovirus polyprotein processing, illustrating an additional pitfall in investigations of viral effects on the apoptotic system of host cells.
Collapse
|
7
|
González OA, Ebersole JL, Huang CB. Oral infectious diseases: a potential risk factor for HIV virus recrudescence? Oral Dis 2009; 15:313-27. [PMID: 19364391 DOI: 10.1111/j.1601-0825.2009.01533.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
As the highly active antiretroviral therapy (HAART) has transitioned human immunodeficiency virus (HIV) infection into a 'chronic disease' management strategy, there is growing evidence that infection with non-HIV pathogens in HIV+ patients may have important public health implications in undermining HAART success and acquired immunodeficiency syndrome progression. Several bacterial and host cell products during infections with non-HIV pathogens have shown the capacity to regulate HIV replication in latently infected cells. A high prevalence of oral infections caused by bacteria, viruses and fungi has been described in HIV+ patients, including periodontal disease. The oral cavity appears to be a site of HIV pathogenesis and potential reservoir for the disease as HIV RNA and DNA forms are present in saliva as well as in gingival crevicular fluid, and oral epithelial cells are susceptible to either cell free or cell-associated HIV infection. The clinical and biological bases of potential associations between chronic oral inflammatory disorders, such as periodontal disease, and exacerbation of HIV viraemia have received little attention. This review attempts to evaluate the current understanding of HIV reactivation as a result of co-infection and/or inflammation induced by non-HIV pathogens in HIV-infected patients, and presents a hypothetic model about the potential role of periodontitis as a global oral infection that potentially contributes to HIV recrudescence.
Collapse
Affiliation(s)
- O A González
- Center for Oral Health Research, College of Dentistry,University of Kentucky, Lexington, KY 40536, USA
| | | | | |
Collapse
|
8
|
Yanagi Y, Takeda M, Ohno S, Hashiguchi T. Measles virus receptors. Curr Top Microbiol Immunol 2009; 329:13-30. [PMID: 19198560 DOI: 10.1007/978-3-540-70523-9_2] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Measles virus (MV) has two envelope glycoproteins, the hemagglutinin (H) and fusion protein, which are responsible for attachment and membrane fusion, respectively. Signaling lymphocyte activation molecule (SLAM, also called CD150), a membrane glycoprotein expressed on immune cells, acts as the principal cellular receptor for MV, accounting for its lymphotropism and immunosuppressive nature. MV also infects polarized epithelial cells via an as yet unknown receptor molecule, thereby presumably facilitating transmission via aerosol droplets. Vaccine and laboratory-adapted strains of MV use ubiquitously expressed CD46 as an alternate receptor through amino acid substitutions in the H protein. The crystal structure of the H protein indicates that the putative binding sites for SLAM, CD46, and the epithelial cell receptor are strategically located in different positions of the H protein. Other molecules have also been implicated in MV infection, although their relevance remains to be determined. The identification of MV receptors has advanced our understanding of MV tropism and pathogenesis.
Collapse
Affiliation(s)
- Y Yanagi
- Department of Virology, Faculty of Medicine, Kyushu University, 812-8582, Fukuoka, Japan.
| | | | | | | |
Collapse
|
9
|
Moss WJ, Scott S, Ndhlovu Z, Monze M, Cutts FT, Quinn TC, Griffin DE. Suppression of human immunodeficiency virus type 1 viral load during acute measles. Pediatr Infect Dis J 2009; 28:63-5. [PMID: 19057456 PMCID: PMC2753197 DOI: 10.1097/inf.0b013e318184eed2] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Acute measles virus infection can result in a transient decrease in plasma human immunodeficiency virus type 1 (HIV-1) RNA loads. We report the kinetics of plasma HIV-1 RNA loads in 2 Zambian children with confirmed and probable measles, and show that the decline in viral load is of similar magnitude to the first-phase decay rate after initiation of antiretroviral therapy.
Collapse
Affiliation(s)
- William J Moss
- Department of Epidemiology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD.
| | | | | | | | | | | | | |
Collapse
|
10
|
McLinden J, Stapleton J, Chang Q, Xiang J. Expression of the Dengue Virus Type 2 NS5 Protein in a CD4+T Cell Line Inhibits HIV Replication. J Infect Dis 2008; 198:860-3. [DOI: 10.1086/591254] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
11
|
|
12
|
Xiang J, McLinden JH, Chang Q, Jordan EL, Stapleton JT. Characterization of a peptide domain within the GB virus C NS5A phosphoprotein that inhibits HIV replication. PLoS One 2008; 3:e2580. [PMID: 18596910 PMCID: PMC2440355 DOI: 10.1371/journal.pone.0002580] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2008] [Accepted: 05/29/2008] [Indexed: 01/30/2023] Open
Abstract
Background GBV-C infection is associated with prolonged survival in HIV-infected people and GBV-C inhibits HIV replication in co-infection models. Expression of the GBV-C nonstructural phosphoprotein 5A (NS5A) decreases surface levels of the HIV co-receptor CXCR4, induces the release of SDF-1 and inhibits HIV replication in Jurkat CD4+ T cell lines. Methodology/Principal Findings Jurkat cell lines stably expressing NS5A protein and peptides were generated and HIV replication in these cell lines assessed. HIV replication was significantly inhibited in all cell lines expressing NS5A amino acids 152–165. Substitution of an either alanine or glycine for the serine at position 158 (S158A or S158G) resulted in a significant decrease in the HIV inhibitory effect. In contrast, substituting a phosphomimetic amino acid (glutamic acid; S158E) inhibited HIV as well as the parent peptide. HIV inhibition was associated with lower levels of surface expression of the HIV co-receptor CXCR4 and increased release of the CXCR4 ligand, SDF-1 compared to control cells. Incubation of CD4+ T cell lines with synthetic peptides containing amino acids 152–167 or the S158E mutant peptide prior to HIV infection resulted in HIV replication inhibition compared to control peptides. Conclusions/Significance Expression of GBV-C NS5A amino acids 152–165 are sufficient to inhibit HIV replication in vitro, and the serine at position 158 appears important for this effect through either phosphorylation or structural changes in this peptide. The addition of synthetic peptides containing 152–167 or the S158E substitution to Jurkat cells resulted in HIV replication inhibition in vitro. These data suggest that GBV-C peptides or a peptide mimetic may offer a novel, cellular-based approach to antiretroviral therapy.
Collapse
Affiliation(s)
- Jinhua Xiang
- Iowa City VA Medical Center and the University of Iowa, Iowa City, Iowa, United States of America
| | - James H. McLinden
- Iowa City VA Medical Center and the University of Iowa, Iowa City, Iowa, United States of America
| | - Qing Chang
- Iowa City VA Medical Center and the University of Iowa, Iowa City, Iowa, United States of America
| | - Emma L. Jordan
- Iowa City VA Medical Center and the University of Iowa, Iowa City, Iowa, United States of America
| | - Jack T. Stapleton
- Iowa City VA Medical Center and the University of Iowa, Iowa City, Iowa, United States of America
- * E-mail:
| |
Collapse
|
13
|
García M, Yu XF, Griffin DE, Moss WJ. Measles virus inhibits human immunodeficiency virus type 1 reverse transcription and replication by blocking cell-cycle progression of CD4+ T lymphocytes. J Gen Virol 2008; 89:984-993. [PMID: 18343840 DOI: 10.1099/vir.0.83601-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Acute measles virus (MV) infection results in a decrease in plasma human immunodeficiency virus type 1 (HIV-1) RNA levels in co-infected children. An in vitro peripheral blood mononuclear cell (PBMC) culture system was used to assess the mechanisms by which MV blocks HIV-1 replication. MV inhibited proliferation of CD4(+) T lymphocytes, the target cell for HIV-1 replication. In the presence of MV, cells did not progress to G(1b) and S phases, steps critical for the completion of HIV-1 reverse transcription and productive replication. This block in cell-cycle progression was characterized by an increased proportion of CD4(+) and HIV-1-infected cells retained in the parental generation in PBMCs co-cultured with MV and HIV-1, and decreased levels of cyclins and RNA synthesis. Early HIV-1 replication was also inhibited in the presence of MV, as measured by reduced expression of a luciferase reporter gene and lower levels of both early (LTR) and late (LTR-gag) DNA intermediates of HIV-1 reverse transcription in the presence of CCR5-tropic HIV-1. The effects of MV on lymphoproliferation and p24 antigen production were reproduced by n-butyrate and hydroxyurea, drugs that block the cell cycle in G(1a) and G(1)/S, respectively. It was concluded that MV inhibits HIV-1 productive replication in part by blocking the proliferation of CD4(+) T lymphocytes.
Collapse
Affiliation(s)
- Mayra García
- Cellular and Molecular Medicine, School of Medicine; Johns Hopkins University, Baltimore, MD 21205, USA
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Xiao-Fang Yu
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Diane E Griffin
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA
| | - William J Moss
- Department of Epidemiology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA
| |
Collapse
|
14
|
Permar SR, Rao SS, Sun Y, Bao S, Buzby AP, Kang HH, Letvin NL. Clinical measles after measles virus challenge in simian immunodeficiency virus-infected measles virus-vaccinated rhesus monkeys. J Infect Dis 2007; 196:1784-93. [PMID: 18190259 DOI: 10.1086/522967] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Understanding the impact of human immunodeficiency virus (HIV) infection on the clinical manifestations and kinetics of measles virus (MV) replication in MV-vaccinated and unvaccinated individuals is important for developing successful vaccine strategies for measles eradication. To model the pathogenesis of MV infection in MV-vaccinated and unvaccinated individuals infected with HIV, previously vaccinated and unvaccinated rhesus monkeys infected with simian immunodeficiency virus (SIV) were challenged with MV and monitored for clinical, virologic, and immunologic sequelae of infection. The magnitude and duration of MV viremia were unchanged by SIV infection. Nevertheless, clinical manifestations of MV infection were altered in animals with significant CD4(+) T lymphocyte loss. Importantly, 2 of the 3 SIV-infected monkeys with high titers of vaccine-induced MV-neutralizing antibody developed clinical evidence of MV infection. Thus, in this experimental animal model, a high-titer vaccine-induced MV-neutralizing antibody response does not protect against clinical manifestations of measles in the setting of a chronic acquired immunodeficiency syndrome virus infection.
Collapse
Affiliation(s)
- Sallie R Permar
- Division of Viral Pathogenesis, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | | | | | | | | | | | | |
Collapse
|
15
|
Early restoration of mucosal CD4 memory CCR5 T cells in the gut of SIV-infected rhesus predicts long term non-progression. AIDS 2007; 21:2377-85. [PMID: 18025874 DOI: 10.1097/qad.0b013e3282f08b32] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVES : To use SIVmac-infected Chinese-origin rhesus macaques (Ch Rh) to characterize the immunopathology of the long term non-progressor (LTNP) state. The key questions addressed were whether or not LTNP experience an early and rapid loss of mucosal CD4 T cells during the acute infection and the mechanisms by which they maintain the LTNP state. METHODS : Ch Rh were infected with SIVmac239. Polychromatic flow cytometry was used to analyze T lymphocyte subsets from blood, lymph nodes and gut tissues during SIV infection. Plasma viral loads were monitored by bDNA assay. Two LTNP were treated with anti-CD8 antibody to deplete CD8 cells in vivo. RESULTS : Thirty-one percent (5/16) of SIVmac239-infected ChRh having low viral loads for as long as 6 years were LTNP. Both LTNP and progressors had similar levels of gut memory CD4/CCR5 T cells (target cells) before infection and there was an early and profound depletion of target cells in both groups. LTNP were distinguished by gradual restoration of mucosal target cells which was evident by 6 months post infection. In vivo CD8 depletion in two LTNP induced AIDS in one LTNP (V542) post anti-CD8 treatment and the other (AJ07) remained healthy after a transient spike in viremia. CONCLUSIONS : Early destruction of target cells was equivalent in LTNP and progressors and did not predict clinical outcome. Restoration of target cells in the gut is associated with long term non-progression. CD8 T cells may play a critical role on maintaining the LTNP state.
Collapse
|
16
|
Stopping HAART temporarily in the absence of virus rebound: exploring new HIV treatment options. Curr Opin HIV AIDS 2007; 2:14-20. [DOI: 10.1097/coh.0b013e328011aad6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
17
|
Grothe C, Haastert K, Jungnickel J. Physiological function and putative therapeutic impact of the FGF-2 system in peripheral nerve regeneration—Lessons from in vivo studies in mice and rats. ACTA ACUST UNITED AC 2006; 51:293-9. [PMID: 16430964 DOI: 10.1016/j.brainresrev.2005.12.001] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2005] [Accepted: 12/07/2005] [Indexed: 11/23/2022]
Abstract
Diffusible and substratum-bound molecules regulate development and regeneration of the peripheral nervous system. The understanding of physiological function of these factors could have an impact on the development of new therapeutic strategies to stimulate nerve regeneration across long gaps. Within the group of trophic factors, basic fibroblast growth factor (FGF-2) and its high-affinity receptors are expressed in the intact peripheral nervous system and regulated following nerve injury. After exogenous application, FGF-2 promotes neuronal survival and neurite outgrowth in vitro and in vivo. In this review, animal studies on the physiological role of the endogenous FGF-2 system and the regenerative capacity after exogenous FGF-2 administration are summarized. The concept of FGF-2 function is discussed in context with other growth factors that are also physiologically relevant in the peripheral nervous system. Studies of sciatic nerve axotomy in FGF-2- and FGF receptor (R) 3-deleted mice, respectively, strongly suggested that FGF-2 binding to FGFR3 is involved in injury-induced neuronal apoptosis. At the lesion site, inhibition of myelination and stimulation of Schwann cell proliferation by FGF-2 via FGFR1/2 is suggested from rat and mouse studies, whereas neurite formation is very likely enhanced via FGFR3 activation. Additionally to these demonstrated physiological functions of endogenous FGF-2, administration of FGF-2 isoforms in the rat model of nerve regeneration across long gaps revealed a role of the high molecular weight isoforms of FGF-2 on sensory recovery. Within the group of physiologically relevant trophic factors, the FGF-2 system seems to be crucially involved in the scenario of peripheral nerve development and regeneration.
Collapse
Affiliation(s)
- Claudia Grothe
- Hannover Medical School, Department of Neuroanatomy, Hannover, Germany.
| | | | | |
Collapse
|
18
|
Zilliox MJ, Parmigiani G, Griffin DE. Gene expression patterns in dendritic cells infected with measles virus compared with other pathogens. Proc Natl Acad Sci U S A 2006; 103:3363-8. [PMID: 16492729 PMCID: PMC1413941 DOI: 10.1073/pnas.0511345103] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Gene expression patterns supply insight into complex biological networks that provide the organization in which viruses and host cells interact. Measles virus (MV) is an important human pathogen that induces transient immunosuppression followed by life-long immunity in infected individuals. Dendritic cells (DCs) are potent antigen-presenting cells that initiate the immune response to pathogens and are postulated to play a role in MV-induced immunosuppression. To better understand the interaction of MV with DCs, we examined the gene expression changes that occur over the first 24 h after infection and compared these changes to those induced by other viral, bacterial, and fungal pathogens. There were 1,553 significantly regulated genes with nearly 60% of them down-regulated. MV-infected DCs up-regulated a core of genes associated with maturation of antigen-presenting function and migration to lymph nodes but also included genes for IFN-regulatory factors 1 and 7, 2'5' oligoadenylate synthetase, Mx, and TNF superfamily proteins 2, 7, 9, and 10 (TNF-related apoptosis-inducing ligand). MV induced genes for IFNs, ILs, chemokines, antiviral proteins, histones, and metallothioneins, many of which were also induced by influenza virus, whereas genes for protein synthesis and oxidative phosphorylation were down-regulated. Unique to MV were the induction of genes for a broad array of IFN-alphas and the failure to up-regulate dsRNA-dependent protein kinase. These results provide a modular view of common and unique DC responses after infection and suggest mechanisms by which MV may modulate the immune response.
Collapse
Affiliation(s)
- Michael J. Zilliox
- *The W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, and
| | - Giovanni Parmigiani
- Departments of Oncology, Biostatistics, and Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Diane E. Griffin
- *The W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, and
| |
Collapse
|