Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 73
Filter
1.
Infection ; 40(4): 441-3, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22002736

ABSTRACT

Dengue virus (DENV) is an arthropod-borne virus (family Flaviviridae) causing dengue fever or dengue hemorrhagic fever. Here, we report the first fatal DENV infection imported into Germany. A female traveler was hospitalized with fever and abdominal pain after returning from Ecuador. Due to a suspected acute acalculous cholecystitis, cholecystectomy was performed. After cholecystectomy, severe spontaneous bleeding from the abdominal wound occurred and the patient died. Postmortem analysis of transudate and tissue demonstrated a DENV secondary infection of the patient and a gallbladder wall thickening (GBWT) due to an extensive edema.


Subject(s)
Severe Dengue/mortality , Travel , Adult , Fatal Outcome , Female , Germany , Humans
2.
Clin Infect Dis ; 50(8): 1112-9, 2010 Apr 15.
Article in English | MEDLINE | ID: mdl-20205588

ABSTRACT

BACKGROUND: This article describes multiple transmissions of rabies via transplanted solid organ from a single infected donor. The empirical Milwaukee treatment regimen was used in the recipients. METHODS: Symptomatic patients were treated by deep sedation (ketamine, midazolam, and phenobarbital), ribavirin, interferon, and active and passive vaccination. Viral loads and antibodies were continuously monitored. RESULTS: Recipients of both cornea and liver transplants developed no symptoms. The recipient of the liver transplant had been vaccinated approximately 20 years before transplantation. Two recipients of kidney and lung transplants developed rabies and died within days of symptomatic disease. Another kidney recipient was treated 7 weeks before he died. The cerebrospinal fluid viral load remained at constant low levels (<10,000 copies/mL) for approximately 5 weeks; it increased suddenly by almost 5 orders of magnitude thereafter. After death, no virus was found in peripheral compartments (nerve tissue, heart, liver, or the small intestine) in this patient, in contrast to in patients in the same cohort who died early. CONCLUSIONS: Our report includes, to our knowledge, the longest documented treatment course of symptomatic rabies and the first time that the virus concentration was measured over time and in different body compartments. The postmortem virus concentration in the periphery was low, but there was no evidence of a reduction of virus in the brain.


Subject(s)
Antibodies, Viral/administration & dosage , Antiviral Agents/therapeutic use , Hypnotics and Sedatives/therapeutic use , Organ Transplantation/adverse effects , Rabies Vaccines/administration & dosage , Rabies virus/isolation & purification , Rabies/drug therapy , Adult , Aged , Antibodies, Viral/blood , Female , Humans , Male , Middle Aged , Rabies Vaccines/immunology , Treatment Outcome , Viral Load
3.
Eur J Med Res ; 15(1): 1-12, 2010 Jan 29.
Article in English | MEDLINE | ID: mdl-20159665

ABSTRACT

This manuscript is communicated by the German AIDS Society (DAIG) (www.daignet.de). It summarizes a series of presentations and discussions during a workshop on immune activation due to HIV infection. The workshop was held on November 22nd 2008 in Hamburg, Germany. It was organized by the ICH Hamburg under the auspices of the German AIDS Society (DAIG e.V.).


Subject(s)
HIV Infections/immunology , Immune System/immunology , Immune System/virology , Germany , Humans
4.
Neuropathol Appl Neurobiol ; 33(6): 644-57, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17573813

ABSTRACT

We studied the central nervous system (CNS) of rhesus macaques during series of vaccination experiments in which attenuated simian immunodeficiency virus (SIV), SIVmac239Deltanef, was applied to the tonsils and the animals were later challenged with pathogenic SIVmac251 or SHIV/89.6P via tonsils or rectum. The pathologic lesions were graded on a scale of 0-5. The lesions were in general very mild, with a score of 0.5, except for one case, in which the animal had progressed to simian AIDS (SAIDS) and had severe lesions of grade 4. Except for the SAIDS case, the most common lesions were meningitis, ependymitis, inflammation of choroid plexus, and astrocytosis. Invasion of the challenge virus, SIVmac251, and pathologic lesions were detected 4 days post infection. The main features of the pathological lesions were similar during short-term follow-up (4 days to 2 weeks) and long-term follow-up (23 to 56 weeks) after challenge. No significant difference was found between unvaccinated controls infected with the challenge viruses and vaccinated and challenged animals. The pathological lesions in the one SAIDS case consisted of extensive lesions of the white matter in connection with confluent ependymitis, indicating an invasion through the choroid plexus. The lesions were characterized by a myriad of multinucleated giant cells of macrophage origin, which showed, together with individual macrophages, strong labelling for viral RNA and proteins. Productive infection of astrocytes was a very rare finding. In three cases infected via tonsils with SIVmac239Deltanef without challenge, we detected expression of Nef-derived peptides, indicating a selective pressure for Nef functions in the CNS.


Subject(s)
Brain/pathology , Immunity, Mucosal , Palatine Tonsil , SAIDS Vaccines/adverse effects , Simian Acquired Immunodeficiency Syndrome/prevention & control , Animals , Female , Genes, nef , Immunohistochemistry , In Situ Hybridization , Macaca mulatta , Male , Mucous Membrane , RNA, Viral/isolation & purification , SAIDS Vaccines/administration & dosage , Simian Acquired Immunodeficiency Syndrome/pathology , Simian Immunodeficiency Virus , Vaccines, Attenuated/adverse effects
5.
Br J Dermatol ; 157(3): 523-30, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17573882

ABSTRACT

BACKGROUND: Human papillomavirus (HPV)-associated anogenital cancers and their precursor lesions occur in excess in human immunodeficiency virus (HIV)-infected patients despite the initiation of highly active antiretroviral therapy. In this context, a drastically increased relative risk for anal intraepithelial neoplasia (AIN) exists in HIV-infected men having sex with men (MSM). In a pilot study, imiquimod, a topical immune response modifier, has been reported to be beneficial in the treatment of AIN. OBJECTIVES: To investigate the role of several biomarkers as potential adjuncts in the course of imiquimod treatment for AIN, and to determine whether these markers correlate with the course of high-risk HPV DNA load during imiquimod therapy. METHODS: Immunohistochemical staining was performed for p16(ink4a), minichromosome maintenance protein (MCM), Ki67, proliferating cell nuclear antigen (PCNA) and p21(waf1) expression before and after 16 weeks of imiquimod treatment for AIN. High-risk HPV DNA load determinations were performed by real-time polymerase chain reaction with type-specific primers and probes for HPV types 16, 18, 31 and 33. RESULTS: Histopathological and virological analyses were performed in 21 HIV-infected MSM with histologically confirmed AIN. Eighteen (86%) patients had a complete histological clearance of AIN after imiquimod therapy. As previously shown, lesional high-risk HPV DNA load significantly decreased during imiquimod therapy. Moreover, a significant decline of p16(ink4a), Ki67, MCM and PCNA expression after treatment was observed, while p21(waf1) expression changed nonsignificantly after imiquimod therapy. A significant correlation between the course of high-risk HPV DNA load and p16(ink4a) expression was observed during imiquimod treatment of AIN, whereas the decline of high-risk HPV DNA load did not significantly correlate with MCM, Ki67, PCNA or p21(waf1) expression. CONCLUSIONS: The significant decrease in p16(ink4a) expression in correlation with the drop of lesional high-risk HPV load suggests that p16(ink4a) may be a useful adjunct for the evaluation of treatment response in HPV-associated malignancies and their precursor lesions.


Subject(s)
Aminoquinolines/therapeutic use , Antineoplastic Agents/therapeutic use , Anus Neoplasms/drug therapy , Carcinoma in Situ/drug therapy , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Precancerous Conditions/drug therapy , AIDS-Related Opportunistic Infections/complications , Administration, Cutaneous , Adult , Anus Neoplasms/metabolism , Anus Neoplasms/virology , Biomarkers, Tumor/metabolism , Carcinoma in Situ/metabolism , Carcinoma in Situ/virology , DNA, Viral/analysis , Homosexuality, Male , Humans , Imiquimod , Ki-67 Antigen/metabolism , Male , Papillomaviridae/genetics , Papillomavirus Infections/complications , Papillomavirus Infections/metabolism , Pilot Projects , Precancerous Conditions/metabolism , Precancerous Conditions/virology , Viral Load
6.
Curr Top Microbiol Immunol ; 276: 1-30, 2003.
Article in English | MEDLINE | ID: mdl-12797441

ABSTRACT

Dendritic cells (DCs) can influence HIV-1 and SIV pathogenesis and protective mechanisms at several levels. First, HIV-1 productively infects select populations of DCs in culture, particularly immature DCs derived from blood monocytes and skin (Langerhans cells). However, there exist only a few instances in which HIV-1- or SIV-infected DCs have been identified in vivo in tissue sections. Second, different types of DCs reliably sequester and transmit infectious HIV-1 and SIV in culture, setting up a productive infection in T cells interacting with the DCs. This stimulation of infection in T cells may explain the observation that CD4+ T lymphocytes are the principal cell type observed to be infected with HIV-1 in lymphoid tissues in vivo. DCs express a C-type lectin, DC-SIGN/CD209, that functions to bind HIV-1 (and other infectious agents) and transmit virus to T cells. When transfected into the THP-1 cell line, the cytosolic domain of DC-SIGN is needed for HIV-1 sequestration and transmission. However, DCs lacking DC-SIGN (Langerhans cells) or expressing very low levels of DC-SIGN (rhesus macaque monocyte-derived DCs) may use additional molecules to bind and transmit immunodeficiency viruses to T cells. Third, DCs are efficient antigen-presenting cells for HIV-1 and SIV antigens. Infection with several recombinant viral vectors as well as attenuated virus is followed by antigen presentation to CD4+ and CD8+ T cells. An intriguing pathway that is well developed in DCs is the exogenous pathway for nonreplicating viral antigens to be presented on class I MHC products. This should allow DCs to stimulate CD8+ T cells after uptake of antibody-coated HIV-1 and dying infected T cells. It has been proposed that DCs, in addition to expanding effector helper and killer T cells, induce tolerance through T cell deletion and suppressor T cell formation, but this must be evaluated directly. Fourth, DCs are likely to be valuable in improving vaccine design. Increasing DC uptake of a vaccine, as well as increasing their numbers and maturation, should enhance efficacy. However, DCs can also capture antigens from other cells that are initially transduced with a DNA vaccine or a recombinant viral vector. The interaction of HIV-1 and SIV with DCs is therefore intricate but pertinent to understanding how these viruses disrupt immune function and elicit immune responses.


Subject(s)
Dendritic Cells/virology , HIV-1/physiology , T-Lymphocytes/virology , Animals , Antigen Presentation/immunology , Antigens, Viral/immunology , Biological Transport , Dendritic Cells/immunology , HIV Infections/immunology , HIV Infections/pathology , HIV Infections/virology , Humans , Monocytes/immunology , Simian Immunodeficiency Virus/physiology , T-Lymphocytes/immunology , Vaccination
7.
Mol Immunol ; 38(2-3): 241-7, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11532285

ABSTRACT

Since the first contact with the host, human immunodeficiency virus (HIV) exploits the complement system to reach maximal spread of infection. HIV has adapted many strategies to avoid complement-mediated lysis and uses the opsonization with complement fragments for attachment to complement receptors (CR). From the pathogen's perspective, binding to CR-expressing cells is remarkably beneficial, bringing together virus and activated target cells that are highly susceptible to infection. Moreover, complement-mediated trapping on CR+ cells permits HIV to infect surrounding cells even in the presence of an excess of neutralizing antibodies. Thus, complement activation initiates the assumption of power over the host's immune system by HIV and thus augments viral spread and replication throughout the body. On the other hand, natural hosts of primate lentiviruses, such as sooty mangabeys, African green monkeys and chimpanzees, are generally considered to be resistant to the development of AIDS, despite persistent viral replication. This review focuses on the possible link between the resistance to disease and species-specific diversity in function of human and monkey complement system.


Subject(s)
Complement System Proteins , HIV Infections/immunology , Lentiviruses, Primate/pathogenicity , Simian Acquired Immunodeficiency Syndrome/immunology , Animals , HIV Infections/etiology , Haplorhini , Humans , Immunity, Innate , Simian Acquired Immunodeficiency Syndrome/etiology , Species Specificity
8.
J Virol ; 75(12): 5646-55, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11356972

ABSTRACT

The mechanism of the progressive loss of CD4+ T lymphocytes, which underlies the development of AIDS in human immunodeficiency virus (HIV-1)-infected individuals, is unknown. Animal models, such as the infection of Old World monkeys by simian-human immunodeficiency virus (SHIV) chimerae, can assist studies of HIV-1 pathogenesis. Serial in vivo passage of the nonpathogenic SHIV-89.6 generated a virus, SHIV-89.6P, that causes rapid depletion of CD4+ T lymphocytes and AIDS-like illness in monkeys. SHIV-KB9, a molecularly cloned virus derived from SHIV-89.6P, also caused CD4+ T-cell decline and AIDS in inoculated monkeys. It has been demonstrated that changes in the envelope glycoproteins of SHIV-89.6 and SHIV-KB9 determine the degree of CD4+ T-cell loss that accompanies a given level of virus replication in the host animals (G. B. Karlsson et. al., J. Exp. Med. 188:1159-1171, 1998). The envelope glycoproteins of the pathogenic SHIV mediated membrane fusion more efficiently than those of the parental, nonpathogenic virus. Here we show that the minimal envelope glycoprotein region that specifies this increase in membrane-fusing capacity is sufficient to convert SHIV-89.6 into a virus that causes profound CD4+ T-lymphocyte depletion in monkeys. We also studied two single amino acid changes that decrease the membrane-fusing ability of the SHIV-KB9 envelope glycoproteins by different mechanisms. Each of these changes attenuated the CD4+ T-cell destruction that accompanied a given level of virus replication in SHIV-infected monkeys. Thus, the ability of the HIV-1 envelope glycoproteins to fuse membranes, which has been implicated in the induction of viral cytopathic effects in vitro, contributes to the capacity of the pathogenic SHIV to deplete CD4+ T lymphocytes in vivo.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , HIV Envelope Protein gp120/metabolism , HIV Envelope Protein gp41/metabolism , HIV-1/pathogenicity , Lymphocyte Depletion , Simian Immunodeficiency Virus/pathogenicity , Animals , HIV Envelope Protein gp120/genetics , HIV Envelope Protein gp41/genetics , HIV Infections/immunology , HIV-1/physiology , Humans , Lymph Nodes/cytology , Lymph Nodes/immunology , Macaca , Membrane Fusion , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Immunodeficiency Virus/physiology
9.
Virology ; 279(1): 221-32, 2001 Jan 05.
Article in English | MEDLINE | ID: mdl-11145904

ABSTRACT

Losing the decisive virus-specific functions of both CD4(+) and CD8(+) T lymphocytes in the first weeks after immunodeficiency virus infection ultimately leads to AIDS. The SIV/rhesus monkey model for AIDS was used to demonstrate that a 4-week chemotherapeutic reduction of viral load during acute SIV infection of macaques allowed the development of a competent immune response able to control virus replication after discontinuation of treatment in two of five monkeys. Increasing SIV-specific CD4(+) T-helper-cell proliferation was found in all macaques several weeks after treatment, independent of their viral load. However, only macaques with low viral loads showed persistent T-cell reactivity of lymph node cells. In contrast to animals with higher viral loads, T-helper-cell counts and memory T-helper cells did not decline in the two macaques controlling viral replication. Lymphocyte apoptosis was consistently low in all treated macaques. In contrast, high CD8(+) lymphocyte death but only slightly increased CD4(+) lymphocyte apoptosis were observed during the first weeks after infection in untreated control animals, indicating that early apoptotic death of virus-specific CTL could be an important factor for disease development. Antiretroviral treatment early after infection obviously retained virus-specific and competent T lymphocytes, whereby a virus-specific immune response could develop in two animals able to control the viral replication after cessation of treatment.


Subject(s)
Adenine/analogs & derivatives , Adenine/therapeutic use , Anti-HIV Agents/therapeutic use , CD8-Positive T-Lymphocytes/physiology , Organophosphonates , Organophosphorus Compounds/therapeutic use , Simian Acquired Immunodeficiency Syndrome/drug therapy , Simian Acquired Immunodeficiency Syndrome/immunology , T-Lymphocytes, Helper-Inducer/immunology , Animals , Apoptosis , Disease Models, Animal , Humans , Immunologic Memory , Macaca mulatta , Reverse Transcriptase Inhibitors/therapeutic use , Simian Immunodeficiency Virus/physiology , Tenofovir , Viral Load
10.
Immunol Rev ; 183: 127-40, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11782253

ABSTRACT

Baboons (Papio cynocephalus) provide a valuable animal model for the study of human immunodeficiency virus (HIV) pathogenesis because HIV-2 infection of baboons causes a chronic viral disease that progresses over several years before clinical signs of acquired immunodeficiency syndrome (AIDS) appear. Since HIV-2-infected baboons develop a chronic viral infection, insights into the immuno-biology of viral latency, clinical stages of disease, virus infection of lymphatic tissue and HIV transmission can be gained using this animal model. The development of an AIDS-like disease in baboons is viral isolate and baboon subspecies dependent. Thus, viral virulence factors and host resistance can be studied as well as the mechanisms of innate and acquired immunity. The control of virus infection is dependent upon cytotoxic and non-cytotoxic antiviral activity of CD8+ T cells. In this regard, some of the HIV-2-infected baboons develop potent antiviral cellular immune responses that have a similar magnitude to that found in HIV-1-infected long-term survivors (or non-progressors). In our laboratory, baboons have been used to study DNA vaccine strategies using new cationic liposome formulations and granulocyte macrophage-colony stimulating factor and B7-2 as genetic adjuvants. The results demonstrate the value of using baboons as an animal model of AIDS pathogenesis and vaccine development.


Subject(s)
Acquired Immunodeficiency Syndrome/microbiology , HIV Infections/immunology , HIV-2/pathogenicity , Models, Animal , Papio , AIDS Vaccines/immunology , Acquired Immunodeficiency Syndrome/etiology , Acquired Immunodeficiency Syndrome/immunology , Animals , Female , HIV Infections/etiology , HIV-2/immunology , Humans , Immunity, Innate/immunology , Male
11.
Blood ; 96(4): 1474-9, 2000 Aug 15.
Article in English | MEDLINE | ID: mdl-10942394

ABSTRACT

Major histocompatibility class I-peptide tetramer technology and simian immunodeficiency virus of macaques (SIVmac)-infected rhesus monkeys were used to clarify the distribution of acquired immunodeficiency syndrome virus-specific cytotoxic T lymphocytes (CTL) in secondary lymphoid organs and to assess the relationship between these CTL and the extent of viral replication in the various anatomic compartments. SIVmac Gag epitope-specific CD8(+) T cells were evaluated in the spleen, bone marrow, tonsils, thymus, and 5 different lymph node compartments of 4 SIVmac-infected rhesus monkeys. The average percentage of CD8(+) T lymphocytes that bound this tetramer in all the different lymph node compartments was similar to that in peripheral blood lymphocytes in individual monkeys. The percentage of CD8(+) T cells that bound the tetramer in the thymus was uniformly low in the monkeys. However, the percentage of CD8(+) T cells that bound the tetramer in bone marrow and spleen was consistently higher than that seen in lymph nodes and peripheral blood. The phenotypic profile of the tetramer-binding CD8(+) T lymphocytes in the different lymphoid compartments was similar, showing a high expression of activation-associated adhesion molecules and a low level expression of naive T-cell-associated molecules. Surprisingly, no correlation was evident between the percentage of tetramer-binding CD8(+) T lymphocytes and the magnitude of the cell-associated SIV RNA level in each lymphoid compartment of individual monkeys. These studies suggest that a dynamic process of trafficking may obscure the tendency of CTL to localize in particular regional lymph nodes or that some lymphoid organs may provide milieus that are particularly conducive to CTL expansion. (Blood. 2000;96:1474-1479)


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/virology , Lymphoid Tissue/immunology , Lymphoid Tissue/virology , RNA, Viral/immunology , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Immunodeficiency Virus/immunology , Animals , Cytotoxicity, Immunologic , In Situ Hybridization , Macaca mulatta , RNA, Viral/analysis
12.
J Virol ; 74(16): 7485-95, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10906202

ABSTRACT

The potential contribution of a plasmid DNA construct to vaccine-elicited protective immunity was explored in the simian immunodeficiency virus (SIV)/macaque model of AIDS. Making use of soluble major histocompatibility class I/peptide tetramers and peptide-specific killing assays to monitor CD8(+) T-lymphocyte responses to a dominant SIV Gag epitope in genetically selected rhesus monkeys, a codon-optimized SIV gag DNA vaccine construct was shown to elicit a high-frequency SIV-specific cytotoxic T-lymphocyte (CTL) response. This CTL response was demonstrable in both peripheral blood and lymph node lymphocytes. Following an intravenous challenge with the highly pathogenic viral isolate SIVsm E660, these vaccinated monkeys developed a secondary CTL response that arose with more rapid kinetics and reached a higher frequency than did the postchallenge CTL response in control plasmid-vaccinated monkeys. While peak plasma SIV RNA levels were comparable in the experimentally and control-vaccinated monkeys during the period of primary infection, the gag plasmid DNA-vaccinated monkeys demonstrated better containment of viral replication by 50 days following SIV challenge. These findings indicate that a plasmid DNA vaccine can elicit SIV-specific CTL responses in rhesus monkeys, and this vaccine-elicited immunity can facilitate the generation of secondary CTL responses and control of viral replication following a pathogenic SIV challenge. These observations suggest that plasmid DNA may prove a useful component of a human immunodeficiency virus type 1 vaccine.


Subject(s)
Gene Products, gag/immunology , SAIDS Vaccines/immunology , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Immunodeficiency Virus/immunology , T-Lymphocytes, Cytotoxic/immunology , Vaccines, DNA/immunology , Animals , CD4 Lymphocyte Count , Gene Products, gag/genetics , Gene Products, gag/metabolism , Humans , Lymph Nodes/immunology , Lymph Nodes/virology , Macaca mulatta , RNA, Viral/analysis , SAIDS Vaccines/administration & dosage , Simian Acquired Immunodeficiency Syndrome/prevention & control , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/physiology , Vaccination , Vaccines, DNA/administration & dosage , Viral Load , Virus Replication
13.
J Immunol ; 164(11): 6015-9, 2000 Jun 01.
Article in English | MEDLINE | ID: mdl-10820285

ABSTRACT

The immunopathogenesis of AIDS-associated hepatitis was explored in the SIV/rhesus monkey model. The livers of SIV-infected monkeys showed a mild hepatitis, with a predominantly CD8+ T lymphocyte infiltration in the periportal fields and sinusoids. These liver-associated CD8+ T cells were comprised of a high percentage of SIV-specific CTL as defined by MHC class I/Gag peptide tetramer binding and Gag peptide epitope-specific lytic activity. There was insufficient viral replication in these livers to account for attracting this large number of functional virus-specific CTL to the liver. There was also no evidence that the predominant population of CTL were functionally end-stage cells trapped in the liver and destined to undergo apoptotic cell death in that organ. Interestingly, we noted that liver tetramer-binding cells showed an increased expression of CD62L, an adhesion molecule usually only rarely expressed on tetramer-binding cells. This observation suggests that the expression of specific adhesion molecules by CTL might facilitate the capture of these cells in the liver. These results demonstrate that functional SIV-specific CD8+ T cells are present in large numbers in the liver of chronically SIV-infected monkeys. Thus, the liver may be a trap for virus-specific cytotoxic T cells.


Subject(s)
Epitopes, T-Lymphocyte/analysis , Liver/immunology , Liver/virology , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/pathology , Simian Immunodeficiency Virus/immunology , T-Lymphocytes, Cytotoxic/pathology , T-Lymphocytes, Cytotoxic/virology , Animals , Apoptosis/immunology , Cell Movement/immunology , Epitopes, T-Lymphocyte/genetics , Gene Products, gag/immunology , Hepatitis, Animal/immunology , Hepatitis, Animal/pathology , Histocompatibility Antigens Class I/genetics , Immunophenotyping , L-Selectin/biosynthesis , L-Selectin/blood , Liver/pathology , Lymphocyte Count , Macaca mulatta , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocyte Subsets/pathology , T-Lymphocyte Subsets/virology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism
14.
J Gen Virol ; 81(Pt 2): 507-13, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10644850

ABSTRACT

The pathogenic properties of four primary human immunodeficiency virus type 2 (HIV-2) isolates and two primary HIV-2 biological clones were studied in an in vivo human-to-mouse chimeric model. The cell-associated viral load and the ability to reduce the severity of the induced graft-versus-host disease symptoms, the CD4/CD8 ratio and the level of repopulation of the mouse tissues by the graft, were determined. All HIV-2 strains, irrespective of their in vitro biological phenotype, replicated to high titres and significantly reduced graft-versus-host disease symptoms as well as the CD4/CD8 ratios. Reduction of graft repopulation caused by infection with the respective HIV-2 strains showed that the in vitro replication rate, syncytium-inducing capacity and ability to infect human macrophages did influence the in vivo pathogenic potential whereas broadening of coreceptor usage did not.


Subject(s)
HIV Infections/etiology , HIV-2/pathogenicity , Receptors, HIV/physiology , Acute Disease , Animals , CD4-CD8 Ratio , Chimera , Disease Models, Animal , Graft vs Host Disease/immunology , HIV Infections/immunology , HIV Infections/virology , HIV-2/physiology , Humans , Immunohistochemistry , Leukocytes, Mononuclear/transplantation , Mice , Transplantation, Heterologous , Virus Replication
15.
Science ; 286(5443): 1353-7, 1999 Nov 12.
Article in English | MEDLINE | ID: mdl-10558989

ABSTRACT

In sexual transmission of simian immunodeficiency virus, and early and later stages of human immunodeficiency virus-type 1 (HIV-1) infection, both viruses were found to replicate predominantly in CD4(+) T cells at the portal of entry and in lymphoid tissues. Infection was propagated not only in activated and proliferating T cells but also, surprisingly, in resting T cells. The infected proliferating cells correspond to the short-lived population that produces the bulk of HIV-1. Most of the HIV-1-infected resting T cells persisted after antiretroviral therapy. Latently and chronically infected cells that may be derived from this population pose challenges to eradicating infection and developing an effective vaccine.


Subject(s)
CD4-Positive T-Lymphocytes/virology , HIV Infections/transmission , HIV-1/physiology , Lymphocyte Activation , Simian Acquired Immunodeficiency Syndrome/transmission , Simian Immunodeficiency Virus/physiology , Animals , Anti-HIV Agents/therapeutic use , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , Cell Cycle , Cervix Uteri/virology , Epithelial Cells/virology , Female , HIV Infections/drug therapy , HIV Infections/virology , Lymph Nodes/virology , Macaca mulatta , RNA, Viral/analysis , Simian Acquired Immunodeficiency Syndrome/virology , Time Factors , Virus Replication
16.
Science ; 285(5431): 1261-5, 1999 Aug 20.
Article in English | MEDLINE | ID: mdl-10455052

ABSTRACT

The early events during infection with an immunodeficiency virus were followed by application of pathogenic simian immunodeficiency virus atraumatically to the tonsils of macaques. Analyses by virologic assays and in situ hybridization revealed that the infection started locally in the tonsils, a mucosal-associated lymphoid organ, and quickly spread to other lymphoid tissues. At day 3, there were few infected cells, but then the number increased rapidly, reaching a high plateau between days 4 and 7. The infection was not detected in the dendritic cell-rich squamous epithelium to which the virus was applied; instead, it was primarily in CD4+ tonsillar T cells, close to the specialized antigen-transporting epithelium of the tonsillar crypts. Transport of the virus and immune-activating stimuli across this epithelium would allow mucosal lymphoid tissue to function in the atraumatic transmission of immunodeficiency viruses.


Subject(s)
Lymphoid Tissue/virology , Mouth Mucosa/virology , Palatine Tonsil/virology , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/physiology , Animals , CD4-Positive T-Lymphocytes/virology , Epithelium/virology , Female , In Situ Hybridization , Leukocytes, Mononuclear/virology , Lymph Nodes/virology , Macaca mulatta , Male , Simian Acquired Immunodeficiency Syndrome/transmission , Viral Load , Virus Replication
17.
AIDS ; 13(9): 1015-9, 1999 Jun 18.
Article in English | MEDLINE | ID: mdl-10397529

ABSTRACT

OBJECTIVE: To assess the source of HIV-1 production in lymphoid tissue biopsies from HIV-infected patients, with no prior anti-retroviral protease inhibitor treatment, with a CD4 cell count > 150 x 10(6)/l (group I) or < 50 x 10(6)/l (group II), co-infected with Mycobacterium tuberculosis or Mycobacterium avium complex. DESIGN AND METHODS: Lymphoid tissue biopsies from 11 HIV-1-infected patients, taken for diagnostic purposes, were studied by HIV-1 RNA in situ hybridization and immunohistochemistry. RESULTS: Patients of group I showed well organized granulomas, in contrast with patients of group II, in which granuloma formation was absent. HIV-1 RNA-positive cells in group I patients were found mainly around the granulomas, whereas in group II HIV-1-producing cells were confined to areas with remaining intact lymphoid tissue. Despite the abundant presence of macrophages, the productively infected HIV-1-positive cells in both groups were almost exclusively CD4 T cells. CONCLUSION: In contrast with previously published data, CD4 T cells appear to remain the major source of HIV-1 production in end-stage disease.


Subject(s)
AIDS-Related Opportunistic Infections/immunology , CD4-Positive T-Lymphocytes/virology , HIV-1/isolation & purification , Mycobacterium avium-intracellulare Infection/immunology , Tuberculosis/immunology , AIDS-Related Opportunistic Infections/microbiology , AIDS-Related Opportunistic Infections/virology , Biopsy , HIV-1/physiology , Humans , Immunohistochemistry , In Situ Hybridization , Lymph Nodes/microbiology , Lymph Nodes/pathology , Lymph Nodes/virology , Mycobacterium avium Complex/isolation & purification , Mycobacterium tuberculosis/isolation & purification , RNA, Viral/analysis
18.
Am J Pathol ; 154(6): 1923-32, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10362819

ABSTRACT

Nonhuman primates provide valuable animal models for human diseases. However, studies assessing the role of cell-mediated immune responses have been difficult to perform in nonhuman primates. We have shown that CD8+ lymphocyte-mediated immunity in rhesus monkeys can be selectively eliminated using the mouse-human chimeric anti-CD8 monoclonal antibody cM-T807. In vitro, this antibody completely blocked antigen-specific expansion of cytotoxic T cells and decreased major histocompatibility complex class I-restricted, antigen-specific lysis of target cells but did not mediate complement-dependent cell lysis. In vivo administration of cM-T807 in rhesus monkeys resulted in near total depletion of CD8+ T cells from the blood and lymph nodes for up to 6 weeks. This depletion was not solely complement-dependent and persisted longer in adults than in juveniles. Preservation of B cell and CD4+ T cell function in monkeys depleted of CD8+ lymphocytes was demonstrated by their ability to develop humoral immune responses to the administered chimeric monoclonal antibody. Furthermore, during CD8+ lymphocyte depletion, monkeys developed delayed-type hypersensitivity reactions comprised only of CD4+ T cells but not CD8+ T cells. This CD8+ lymphocyte depletion model should prove useful in defining the role of cell-mediated immune responses in controlling infectious diseases in nonhuman primates.


Subject(s)
Antibodies, Monoclonal/administration & dosage , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/drug effects , Lymphocyte Depletion , Models, Immunological , Recombinant Fusion Proteins/administration & dosage , Animals , Antibodies/blood , Antibody Formation/drug effects , Antigens, CD/immunology , CD8-Positive T-Lymphocytes/immunology , Complement Activation/drug effects , Elapid Venoms/pharmacology , Humans , Hypersensitivity, Delayed/blood , Hypersensitivity, Delayed/immunology , Immunity, Cellular/drug effects , Macaca mulatta , Mice , Simian Acquired Immunodeficiency Syndrome/immunology , Tetanus Toxoid/immunology
19.
N Engl J Med ; 340(21): 1605-13, 1999 May 27.
Article in English | MEDLINE | ID: mdl-10341272

ABSTRACT

BACKGROUND: In patients infected with human immunodeficiency virus type 1 (HIV-1), combination antiretroviral therapy can result in sustained suppression of plasma levels of the virus. However, replication-competent virus can still be recovered from latently infected resting memory CD4 lymphocytes; this finding raises serious doubts about whether antiviral treatment can eradicate HIV-1. METHODS: We looked for evidence of residual HIV-1 replication in eight patients who began treatment soon after infection and in whom plasma levels of HIV-1 RNA were undetectable after two to three years of antiretroviral therapy. We examined whether there had been changes over time in HIV-1 proviral sequences in peripheral-blood mononuclear cells, which would indicate residual viral replication. We also performed in situ hybridization studies on tissues from one patient to identify cells actively expressing HIV-1 RNA. We estimated the rate of decrease of latent, replication-competent HIV-1 in resting CD4 lymphocytes on the basis of the decrease in the numbers of proviral sequences identified during primary infection and direct sequential measurements of the size of the latent reservoir. RESULTS: Six of the eight patients had no significant variations in proviral sequences during treatment. However, in two patients there was sequence evolution but no evidence of drug-resistant viral genotypes. In one patient, extensive in situ studies provided additional evidence of persistent viral replication in lymphoid tissues. Using two independent approaches, we estimated that the half-life of the latent, replication-competent virus in resting CD4 lymphocytes was approximately six months. CONCLUSIONS: These findings suggest that combination antiretroviral regimens suppress HIV-1 replication in some but not all patients. Given the half-life of latently infected CD4 lymphocytes of about six months, it may require many years of effective antiretroviral treatment to eliminate this reservoir of HIV-1.


Subject(s)
Anti-HIV Agents/therapeutic use , HIV Infections/drug therapy , HIV-1/drug effects , Virus Replication/drug effects , Adult , Anti-HIV Agents/pharmacology , Base Sequence , Drug Therapy, Combination , Genes, env , HIV Infections/virology , HIV-1/genetics , HIV-1/growth & development , HIV-1/isolation & purification , Humans , In Situ Hybridization , Least-Squares Analysis , Male , Molecular Sequence Data , Phylogeny , RNA, Viral/blood , Viral Load
20.
AIDS ; 13(1): F1-8, 1999 Jan 14.
Article in English | MEDLINE | ID: mdl-10207538

ABSTRACT

OBJECTIVE: Lymphoid tissue provides a reservoir where HIV can persist. However, therapies incorporating a protease inhibitor can target this reservoir. This study was designed to investigate the relative long-term effects on lymph-node viral load and cellular architecture of regimens containing multiple nucleosides alone or in combination with protease inhibitors. METHODS: Axillary lymph-node biopsies from 12 patients with undetectable viraemia (viral load < 20 copies/ml: mean CD4 cells 525 x 10(6)/l) for a mean period of 25 months (range, 10-52 months) were investigated for the presence of HIV by in situ hybridization and coculture. Four patients were receiving multiple nucleoside analogues alone or in one case with a suboptimally dosed protease inhibitor (group I). Protease inhibitor was added to the regimen of seven patients at least 6 months prior to lymph-node biopsy (group II). Standard flow cytometry and virological data were obtained from peripheral blood every 3 months. RESULTS: By in situ hybridization, more productively infected CD4+ T cells were found in the lymph nodes of group I patients treated with nucleoside analogues alone. Very low numbers of productively infected lymph node cells were detected in the protease inhibitor-treated group II. No trapping of virions on the follicular dendritic cell (FDC) network was detectable in protease inhibitor-treated patients. In contrast, large deposits of FDC-bound virions were observed in three out of five patients from group I. Virus cultures from lymph node cells were positive in these three group I patients compared with only one out of seven patients from group II. Sequencing reverse transcriptase and protease genes from these isolates revealed typical mutations conferring resistance to the previously administered nucleoside analogue. A more preserved lymph node architecture and less signs of immunopathological change were also observed in protease inhibitor-treated patients. CONCLUSIONS: Undetectable plasma viraemia using the ultrasensitive PCR assay for prolonged periods of time does not always reflect complete HIV-1 suppression within the lymphoid compartment. Our results suggest that protease inhibitor-containing regimens target HIV reservoirs in lymphoid tissue more effectively and preserve or restore lymph node architecture.


Subject(s)
Anti-HIV Agents/therapeutic use , HIV Infections/virology , HIV Protease Inhibitors/therapeutic use , HIV-1/physiology , Nucleosides/therapeutic use , Virus Replication , CD4 Lymphocyte Count , CD4-CD8 Ratio , Coculture Techniques , Dendritic Cells , Drug Resistance, Microbial , Drug Therapy, Combination , HIV Infections/immunology , HIV Infections/pathology , HIV-1/drug effects , HIV-1/genetics , Humans , Lymphoid Tissue/virology , RNA, Viral , Viral Load
SELECTION OF CITATIONS
SEARCH DETAIL
...