Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
Vet Pathol ; 51(1): 110-26, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24168861

ABSTRACT

Molecular localization techniques remain important diagnostic and research tools for the pathologist evaluating nonhuman primate tissues. In situ hybridization and immunohistochemistry protocols have been developed for many important pathogens of nonhuman primates, including RNA and DNA viruses, prions, and bacterial, protozoal, and fungal pathogens. Such techniques will remain critical in defining the impact and relevance of novel agents on animal health and disease. A comparative pathology perspective often provides valuable insight to the best strategy for reagent development and can also facilitate interpretation of molecular localization patterns. Such a perspective is grounded in a firm understanding of microbe-host pathobiology. This review summarizes current molecular localization protocols used in the diagnosis of selected primate infectious diseases.


Subject(s)
Communicable Diseases/diagnosis , Pathology, Molecular/methods , Pathology, Veterinary/methods , Primate Diseases/diagnosis , Animals , Antibodies , Communicable Diseases/microbiology , Communicable Diseases/parasitology , Immunohistochemistry/veterinary , In Situ Hybridization/veterinary , Primate Diseases/microbiology , Primate Diseases/parasitology , Primates , Sensitivity and Specificity
2.
Vet Pathol ; 51(1): 281-91, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24091814

ABSTRACT

The identification, application, and qualification of safety biomarkers are becoming increasingly critical to successful drug discovery and development as companies are striving to develop drugs for difficult targets and for novel disease indications in a risk-adverse environment. Translational safety biomarkers that are minimally invasive and monitor drug-induced toxicity during human clinical trials are urgently needed to assess whether toxicities observed in preclinical toxicology studies are relevant to humans at therapeutic doses. The interpretation of data during the biomarker qualification phase should include careful consideration of the analytic method used, the biology, pharmacokinetic and pharmacodynamic properties of the biomarker, and the pathophysiology of the process studied. The purpose of this review is to summarize commonly employed technologies in the development of fluid- and tissue-based safety biomarkers in drug discovery and development and to highlight areas of ongoing novel assay development.


Subject(s)
Biomarkers, Pharmacological/analysis , Drug Discovery/methods , Drug Evaluation, Preclinical , Animals , Clinical Trials as Topic , Drug-Related Side Effects and Adverse Reactions , Humans , Pathology, Clinical , Translational Research, Biomedical
3.
Vet Pathol ; 50(6): 1145-53, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23839235

ABSTRACT

A workshop on Emerging Respiratory Viral Infections and Spontaneous Diseases in nonhuman primates was sponsored by the concurrent Annual Meetings of the American College of Veterinary Pathologists and the American Society for Veterinary Clinical Pathology, held December 1-5, 2012, in Seattle, Washington. The session had platform presentations from Drs Karen Terio, Thijs Kuiken, Guy Boivin, and Robert Palermo that focused on naturally occurring influenza, human respiratory syncytial virus, and metapneumovirus in wild and zoo-housed great apes; the molecular biology and pathology of these viral respiratory diseases in nonhuman primate (NHP) models; and the therapeutic and vaccine approaches to prevention and control of these emerging respiratory viral infections. These formal presentations were followed by presentations of 14 unique case studies of rare or newly observed spontaneous lesions in NHPs (see online files for access to digital whole-slide images corresponding to each case report at http://scanscope.com/ACVP%20Slide%20Seminars/2012/Primate%20Pathology/view.apml). The session was attended by meeting participants that included students, pathology trainees, and experienced pathologists from academia and industry with an interest in respiratory and spontaneous diseases of NHPs.


Subject(s)
Macaca , Pan troglodytes , Papio , Primate Diseases/virology , Respiratory Tract Infections/veterinary , Virus Diseases/veterinary , Animals , Female , Macaca fascicularis , Macaca mulatta , Macaca nemestrina , Male , Respiratory Tract Infections/virology , Virus Diseases/virology
4.
Vet Pathol ; 49(6): 1057-69, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23135296

ABSTRACT

The combination of loss of habitat, human population encroachment, and increased demand of select nonhuman primates for biomedical research has significantly affected populations. There remains a need for knowledge and expertise in understanding background findings as related to the age, source, strain, and disease status of nonhuman primates. In particular, for safety/biomedical studies, a broader understanding and documentation of lesions would help clarify background from drug-related findings. A workshop and a minisymposium on spontaneous lesions and diseases in nonhuman primates were sponsored by the concurrent Annual Meetings of the American College of Veterinary Pathologists and the American Society for Veterinary Clinical Pathology held December 3-4, 2011, in Nashville, Tennessee. The first session had presentations from Drs Lowenstine and Montali, pathologists with extensive experience in wild and zoo populations of nonhuman primates, which was followed by presentations of 20 unique case reports of rare or newly observed spontaneous lesions in nonhuman primates (see online files for access to digital whole-slide images corresponding to each case report at http://www.scanscope.com/ACVP%20Slide%20Seminars/2011/Primate%20Pathology/view.apml). The minisymposium was composed of 5 nonhuman-primate researchers (Drs Bradley, Cline, Sasseville, Miller, Hutto) who concentrated on background and spontaneous lesions in nonhuman primates used in drug safety studies. Cynomolgus and rhesus macaques were emphasized, with some material presented on common marmosets. Congenital, acquired, inflammatory, and neoplastic changes were highlighed with a focus on clinical, macroscopic, and histopathologic findings that could confound the interpretation of drug safety studies.


Subject(s)
Animals, Wild , Animals, Zoo , Primate Diseases/pathology , Primates , Animal Experimentation , Animals , Biomedical Research , Drug Evaluation, Preclinical , Female , Macaca fascicularis , Macaca mulatta , Male , Models, Animal
5.
Vet Pathol ; 48(4): 885-95, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21149847

ABSTRACT

Glucocorticoids (GCs), despite having many undesirable side effects, remain effective for the treatment of many inflammatory diseases and are commonly used as benchmark drugs in animal models of disease. However, the molecular mechanisms underling systemic GC effects in these models are poorly characterized. In this study, prednisolone and dexamethasone were evaluated in the fully established Lewis rat adjuvant-induced arthritis (AIA) model. In AIA, adjuvant administration induced polyarticular and systemic inflammation, which included spleen and liver. In the liver, multifocal hepatic granulomas were observed. To characterize the systemic response and the pathways responsible for GC effects, histology, transcriptional profiling, and immunohistochemistry (IHC) were performed. There was a decrease in the incidence and histologic severity score for granulomas with GC treatment. There was no effect on cellular composition of granulomas as assessed by IHC for CD3+ lymphocytes, macrophages, and B cells, but there was a significant reduction in infiltrating lymphocytes in the hepatic parenchyma. By Affymetrix microarray analysis, 10% of hepatic transcripts were altered (P<.01) in livers from AIA rats, with ~31% of them partially reversed with treatment with dexamethasone and ~13% with prednisolone. Many of these altered hepatic transcripts correspond to human genes that are dysregulated in the synovium in human rheumatoid arthritis (RA), indicating that the rat AIA model shares features with human RA. These data establish molecular changes in the liver and the effect of GCs in rat AIA, which can be used to aid in understanding the mechanism of action of novel anti-inflammatory compounds in this animal model.


Subject(s)
Arthritis, Experimental/drug therapy , Dexamethasone/therapeutic use , Glucocorticoids/therapeutic use , Liver/metabolism , Prednisolone/therapeutic use , Transcription, Genetic/drug effects , Animals , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/therapeutic use , Arthritis, Experimental/metabolism , Arthritis, Experimental/pathology , Dexamethasone/administration & dosage , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Administration Schedule , Gene Expression Profiling , Glucocorticoids/administration & dosage , Male , Prednisolone/administration & dosage , Rats , Rats, Inbred Lew
6.
Vet Pathol ; 44(3): 407-10, 2007 May.
Article in English | MEDLINE | ID: mdl-17491089

ABSTRACT

Subendothelial heart valve angiectasis has been reported in cows, dogs, pigs, rats, mice, and in human fetuses and newborns. We observed a high incidence (62 in 208 animals examined) of spontaneous angiectasis on the atrioventricular (AV) valves in 10- to 40-week-old Sprague-Dawley rats. The angiectasis was observed predominately on the septal cusp of the right AV valve and located near the AV ostium in 57 of 62 animals. Of the remaining 5 valvular angiectases, 2 were present on the parietal cusp of the right AV valve and 3 were on the left AV valve. The angiectases were single or multiple, ranging from 40 to 300 microm in diameter and were characterized by light microscopy as blood-filled dilatations lined by endothelium. Spontaneously occurring abnormalities in normal laboratory animals, such as the spontaneous valvular angiectasis reported here, need to be differentiated from drug-related lesions.


Subject(s)
Heart Valve Diseases/veterinary , Rodent Diseases/pathology , Vascular Diseases/veterinary , Animals , Female , Heart Valve Diseases/pathology , Male , Rats , Rats, Sprague-Dawley , Vascular Diseases/pathology
7.
Chem Biol Interact ; 150(1): 9-25, 2004 Nov 01.
Article in English | MEDLINE | ID: mdl-15522258

ABSTRACT

The financial investment grows exponentially as a new chemical entity advances through each stage of discovery and development. The opportunity exists for the modern toxicologist to significantly impact expenditures by the early prediction of potential toxicity/side effect barriers to development by aggressive evaluation of development-limiting liabilities early in drug discovery. Improved efficiency in pharmaceutical research and development lies both in leveraging "best in class" technology and integration with pharmacologic activities during hit-to-lead and early lead optimization stages. To meet this challenge, a discovery assay by stage (DABS) paradigm should be adopted. The DABS clearly delineates to discovery project teams the timing and type of assay required for advancement of compounds to each subsequent level of discovery and development. An integrative core pathology function unifying Drug Safety Evaluation, Molecular Technologies and Clinical Research groups that effectively spans all phases of drug discovery and development is encouraged to drive the DABS. The ultimate goal of such improved efficiency being the accurate prediction of toxicity and side effects that would occur in development before commitment of the large prerequisite resource. Good justification of this approach is that every reduction of development attrition by 10% results in an estimated increase in net present value by $100 million.


Subject(s)
Toxicology/methods , Animals , Chemistry, Pharmaceutical , Drug Design , Drug Evaluation, Preclinical , Humans , Liver/pathology , Models, Animal , Reproducibility of Results , Research Design
8.
Nat Med ; 5(9): 997-103, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10470075

ABSTRACT

Infection of macrophage lineage cells is a feature of primate lentivirus replication, and several properties of primate lentiviruses seem to have evolved to promote the infection of macrophages. Here we demonstrate that the accessory gene product Nef induces the production of two CC-chemokines, macrophage inflammatory proteins 1alpha and 1beta, by HIV-1-infected macrophages. Adenovirus-mediated expression of Nef in primary macrophages was sufficient for chemokine induction. Supernatants from Nef-expressing macrophages induced both the chemotaxis and activation of resting T lymphocytes, permitting productive HIV-1 infection. These results indicate a role for Nef in lymphocyte recruitment and activation at sites of virus replication.


Subject(s)
Chemotaxis , Gene Products, nef/physiology , HIV-1/physiology , Lymphocyte Activation , Macrophages/virology , T-Lymphocytes/immunology , Adenoviridae/genetics , Animals , Cell Line , Cells, Cultured , Chemokine CCL4 , Chemokines/biosynthesis , Chemokines/genetics , Chemotaxis/drug effects , Culture Media, Conditioned , Cytokines/biosynthesis , Encephalitis, Viral/immunology , Encephalitis, Viral/virology , Gene Products, nef/genetics , HIV-1/drug effects , HIV-1/genetics , HIV-1/growth & development , Humans , Lymphocyte Activation/drug effects , Macaca , Macrophage Inflammatory Proteins/biosynthesis , Macrophage Inflammatory Proteins/genetics , Macrophages/immunology , Macrophages/metabolism , Mutation , Simian Immunodeficiency Virus/drug effects , Simian Immunodeficiency Virus/growth & development , Simian Immunodeficiency Virus/physiology , T-Lymphocytes/drug effects , T-Lymphocytes/virology , Virus Replication/drug effects , nef Gene Products, Human Immunodeficiency Virus
9.
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
10.
Science ; 283(5403): 857-60, 1999 Feb 05.
Article in English | MEDLINE | ID: mdl-9933172

ABSTRACT

Clinical evidence suggests that cellular immunity is involved in controlling human immunodeficiency virus-1 (HIV-1) replication. An animal model of acquired immune deficiency syndrome (AIDS), the simian immunodeficiency virus (SIV)-infected rhesus monkey, was used to show that virus replication is not controlled in monkeys depleted of CD8+ lymphocytes during primary SIV infection. Eliminating CD8+ lymphocytes from monkeys during chronic SIV infection resulted in a rapid and marked increase in viremia that was again suppressed coincident with the reappearance of SIV-specific CD8+ T cells. These results confirm the importance of cell-mediated immunity in controlling HIV-1 infection and support the exploration of vaccination approaches for preventing infection that will elicit these immune responses.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/immunology , Acquired Immunodeficiency Syndrome/immunology , Acquired Immunodeficiency Syndrome/virology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Viral/blood , Disease Progression , Gene Products, gag/blood , Humans , Lymphocyte Count , Lymphocyte Depletion , Macaca mulatta , Neutralization Tests , RNA, Viral/blood , Simian Immunodeficiency Virus/physiology , T-Lymphocytes, Cytotoxic/immunology , Time Factors , Viral Load , Viremia/immunology , Viremia/virology , Virus Replication
11.
AIDS Res Hum Retroviruses ; 15(1): 11-21, 1999 Jan 01.
Article in English | MEDLINE | ID: mdl-10024048

ABSTRACT

We engineered recombinant derivatives of simian immunodeficiency virus (SIV) to express enhanced green fluorescent protein (EGFP). Replacement of vpr sequences with EGFP resulted in a genome that did not produce detectable levels of replication-competent virus. Replication-competent virus and bright fluorescence of infected cells were obtained with two other constructs, one in which SIV nef sequences were replaced by EGFP and another in which EGFP was inserted into the SIV nef locus and HIV-1 nef sequences were expressed by downstream placement of an internal ribosomal entry site. These strains were infectious in rhesus monkeys and green fluorescing cells were detected in the tissues of infected monkeys by FACS analysis and by direct microscopic visualization. EGFP sequences were absent from recovered virus by 8 weeks following infection. We conclude that recombinant SIV that is engineered to express EGFP can be used to directly detect productively infected cells and aid in the immunophenotypic characterization of these cells within the first 2 weeks of infection of rhesus monkeys.


Subject(s)
Luminescent Proteins/genetics , Recombinant Fusion Proteins/metabolism , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/genetics , Animals , Base Sequence , Cell Line , Flow Cytometry , Green Fluorescent Proteins , Immunohistochemistry , In Situ Hybridization , Indicators and Reagents/metabolism , Luminescent Proteins/metabolism , Macaca mulatta , Microscopy, Fluorescence , Molecular Sequence Data , Polymerase Chain Reaction/methods , Recombinant Fusion Proteins/genetics , Simian Immunodeficiency Virus/metabolism , Simian Immunodeficiency Virus/physiology , Transfection , Viral Load , Virus Replication
12.
Vet Pathol ; 35(6): 499-505, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9823591

ABSTRACT

A captive-born juvenile female rhesus macaque (Macaca mulatta) was acquired from a commercial breeder and placed in quarantine. Within 8 days of arrival, the animal became anorexic, inactive, and dehydrated. Subsequently, generalized edema and facial ecchymoses developed, and despite supportive therapy, the animal became moribund and was euthanatized. Macroscopic examination showed diffuse stippling and streaking of the myocardium. Histopathologic examination revealed multifocal to coalescing myocardial edema, necrosis, lymphohistiocytic inflammation, and generalized endothelial infection with Sarcocystis sp. Immature and mature schizonts within endothelial cells were most prevalent in the heart. Fewer schizonts were present in the vasculature of other tissues, including skeletal muscle, smooth muscle, adipose tissue, brain, and retina. Mature tissue cysts within muscle fibers were not found in the myocardium but were occasionally seen in skeletal muscle. Analysis of polymerase-chain-reaction-amplified 18s ribosomal RNA gene sequences revealed 96% identity to published sequences of S. hirsuta, S. hominis, and S. fusiformis and 95% identity to S. cruzi and S. tenella. However, sequences did not show complete identity with any organism in the GenBank database. Sequence homology suggests that this is a newly described Sarcocystis sp. Results of antibody tests for simian retrovirus, simian T-lymphotropic virus 1, and simian immunodeficiency virus were negative, suggesting that viral immunosuppression was unlikely to have augmented the pathogenicity of sarcosporidial infection. Clinical and histopathologic findings in this case of fulminant sarcosporidiosis are similar to those described in Dalmeny disease in cattle, which is associated with ingestion of massive numbers of infective Sarcocystis oocysts.


Subject(s)
Macaca mulatta , Monkey Diseases/parasitology , Sarcocystis/isolation & purification , Sarcocystosis/veterinary , Acute Disease , Animals , Base Sequence , Cattle , DNA Primers/chemistry , Ecchymosis/pathology , Ecchymosis/veterinary , Female , Heart/parasitology , Macaca mulatta/parasitology , Molecular Sequence Data , Monkey Diseases/pathology , Muscle, Skeletal/pathology , Muscle, Skeletal/ultrastructure , Myocardium/pathology , Polymerase Chain Reaction/veterinary , RNA, Protozoan/analysis , RNA, Ribosomal, 18S/analysis , Sarcocystis/genetics , Sarcocystis/ultrastructure , Sarcocystosis/parasitology , Sarcocystosis/pathology , Sequence Homology, Nucleic Acid
13.
J Invest Dermatol ; 110(6): 894-901, 1998 Jun.
Article in English | MEDLINE | ID: mdl-9620296

ABSTRACT

The molecularly cloned viruses known as SIVmac239/R17Y and SIVmac239/YEnef cause extensive lymphocyte activation and induce an acute disease syndrome in macaque monkeys. One manifestation of this syndrome is a severe diffuse cutaneous maculopapular exanthem that is similar to the exanthem associated with HIV-1 infection. To examine the pathogenesis of this exanthem, biopsies obtained throughout the course of clinically evident rash were examined for the presence of virus by in situ hybridization and immunohistochemistry, and the cellular infiltrate was characterized with respect to cellular immunophenotype and chemokine receptor expression. The onset of rash was associated with abundant simian immunodeficiency virus nucleic acid and protein within perivascular dermal infiltrates and occasionally within intraepithelial cells. Analysis of cellular infiltrates showed that biopsies, obtained on the day of rash onset, were composed of equal numbers of CD4+ and CD8+ lymphocytes and abundant alphaEbeta7 positive cells surrounding vessels with upregulated endothelial E-selectin. Moreover, by examining virus expression in sequential skin biopsies from the same animal, the clearance of virus and the resolution of rash were associated with an increase in the percentage of cells expressing CD8, the chemokine receptor CXCR3, and GMP-17, a marker of cytotoxic granules. These results suggest that activated cytotoxic T cells are trafficking to sites of inflammation in the skin and directly or indirectly affect levels of viral replication at these sites.


Subject(s)
Exanthema/virology , Genes, nef/genetics , Macaca mulatta/virology , Simian Acquired Immunodeficiency Syndrome , Simian Immunodeficiency Virus/genetics , Animals , Antigens, Differentiation, T-Lymphocyte , Antigens, Neoplasm , Biopsy , Blood Vessels/pathology , Blood Vessels/virology , CD8-Positive T-Lymphocytes/chemistry , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/virology , Cell Division , E-Selectin/analysis , Exanthema/pathology , Genes, Viral/genetics , Genetic Variation/genetics , HIV Infections/pathology , HIV Infections/virology , Immunohistochemistry , Integrins/analysis , Ki-67 Antigen/analysis , Leukocyte Count , Lymphocyte Count , Membrane Glycoproteins/analysis , Neutrophils/chemistry , Neutrophils/cytology , Neutrophils/virology , RNA, Viral/analysis , RNA, Viral/genetics , Receptors, CCR5/analysis , Receptors, CXCR3 , Receptors, Chemokine/analysis , Skin/blood supply , Skin/pathology , Skin/virology , T-Lymphocytes, Cytotoxic/chemistry , T-Lymphocytes, Cytotoxic/cytology , T-Lymphocytes, Cytotoxic/virology , Time Factors , Vascular Cell Adhesion Molecule-1/analysis
14.
Am J Pathol ; 152(3): 659-65, 1998 Mar.
Article in English | MEDLINE | ID: mdl-9502406

ABSTRACT

Although the mechanisms of human immunodeficiency virus (HIV) neuroinvasion, neuronal injury, and subsequent development of HIV-1-associated AIDS dementia complex are not fully understood, a correlation between monocyte/macrophage infiltrates in the brain and neurological disease exists. In light of the many potential roles that chemokines and chemokine receptors may play in HIV neuropathogenesis, we sought to describe their pattern of expression in the SIV-infected rhesus macaque model of HIV encephalitis. We previously demonstrated elevated expression of the chemokines macrophage inflammatory protein (MIP)-1alpha, MIP-1beta, RANTES, and interferon-inducible protein (IP)-10 in brain of macaque monkeys with SIV encephalitis. In this study, we demonstrate that the corresponding chemokine receptors CCR3, CCR5, CXCR3, and CXCR4 are expressed in perivascular infiltrates in these same tissues. In addition, we detected CCR3, CCR5, and CXCR4 on subpopulations of large hippocampal and neocortical pyramidal neurons and on glial cells in both normal and encephalitic brain. These findings suggest that multiple chemokines and their receptors contribute to monocyte and lymphocyte recruitment to the brain in SIV encephalitis. Furthermore, the expression of known HIV/SIV co-receptors on neurons suggests a possible mechanism whereby HIV or SIV can directly interact with these cells, disrupting their normal physiological function and contributing to the pathogenesis of AIDS dementia complex.


Subject(s)
Brain/metabolism , Encephalitis, Viral/metabolism , Receptors, Chemokine/metabolism , Simian Acquired Immunodeficiency Syndrome/metabolism , Simian Immunodeficiency Virus , Animals , Brain/pathology , Brain/virology , Chemokines/metabolism , Encephalitis, Viral/etiology , Encephalitis, Viral/pathology , Immunohistochemistry , Macaca mulatta , Simian Acquired Immunodeficiency Syndrome/etiology , Simian Acquired Immunodeficiency Syndrome/pathology
15.
J Virol ; 71(12): 9764-9, 1997 Dec.
Article in English | MEDLINE | ID: mdl-9371642

ABSTRACT

A herpesvirus that is related to but distinct from the Kaposi's sarcoma-associated herpesvirus (KSHV, or human herpesvirus 8) was isolated from rhesus monkeys. The sequence of 10.6 kbp from virion DNA revealed the presence of an interleukin-6 homolog similar to what is present in KSHV and a closer relatedness of the DNA polymerase and glycoprotein B reading frames to those of KSHV than to those of any other herpesvirus. This rhesus monkey herpesvirus replicated lytically and to high titers in cultured rhesus monkey fibroblasts. Antibody testing revealed a high prevalence for at least 10 years in our rhesus monkey colony and a high prevalence in two other colonies that were tested. Thus, rhesus monkeys naturally harbor a virus related to KSHV, which we have called RRV, for rhesus monkey rhadinovirus.


Subject(s)
Herpesviridae Infections/veterinary , Herpesvirus 8, Human , Macaca mulatta/virology , Rhadinovirus/isolation & purification , Sarcoma, Kaposi/virology , Tumor Virus Infections/veterinary , Animals , Antibodies, Viral/blood , Base Sequence , Cells, Cultured , Cytopathogenic Effect, Viral , DNA, Viral , DNA-Directed DNA Polymerase/genetics , Glycoproteins/genetics , Herpesviridae , Herpesviridae Infections/immunology , Herpesviridae Infections/virology , Humans , Interleukin-6/genetics , Molecular Sequence Data , Rhadinovirus/genetics , Rhadinovirus/immunology , Tumor Virus Infections/immunology , Tumor Virus Infections/virology , Viral Proteins/genetics
16.
J Neurovirol ; 3(1): 1-9, 1997 Feb.
Article in English | MEDLINE | ID: mdl-9147816

ABSTRACT

Patients infected with human immunodeficiency virus (HIV) develop immunologic dysfunction and multiorgan inflammatory diseases directly associated with HIV-1 infection. Of these inflammatory diseases, the most devastating to the HIV-infected patient is involvement of the central nervous system (CNS). The pathogenesis of the clinical syndrome observed in these patients, termed HIV-associated dementia, remains poorly understood. However, as most of the detectable virus in the CNS is in cells of monocyte/macrophage lineage, it is clear that penetration of the blood-brain barrier by HIV-1 and the subsequent influx of monocytes into the brain are crucial components in the neuropathogenesis of HIV-associated dementia. Using the SIV-infected macaque model of acquired immunodeficiency disease, much has been learned about viral neuroinvasion occurring soon after experimental infection. The aim of this review is to discuss these recent advances and provide insight into plausible mechanisms for monocyte entry into the CNS.


Subject(s)
Dementia/virology , Monocytes/virology , Simian Acquired Immunodeficiency Syndrome/complications , AIDS Dementia Complex , Animals , Blood-Brain Barrier , Cell Adhesion Molecules/physiology , Cytokines/physiology , Dementia/immunology , Dementia/pathology , Disease Models, Animal , Encephalomyelitis/immunology , Encephalomyelitis/pathology , Encephalomyelitis/virology , Macaca , Models, Neurological , Monocytes/immunology , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/pathology
17.
J Neurovirol ; 2(6): 423-32, 1996 Dec.
Article in English | MEDLINE | ID: mdl-8972425

ABSTRACT

During peak viremia and initial antibody response, rhesus macaques infected with pathogenic and nonpathogenic isolates of SIV show distinct differences in viral load and tissue distribution. Animals infected with pathogenic isolates of SIV invariably have virus in the CSF and brain parenchyma by two weeks postinoculation, whereas animals infected with nonpathogenic isolates do not. Mechanisms underlying neuroinvasion by SIV and HIV are unknown, but recruitment of latently infected mononuclear cells from the peripheral circulation (Trojan horse theory) is frequently proposed. Circulating monocytes, from which perivascular macrophage/microglia are derived, are a likely vehicle for cell-associated transport of virus across the blood-brain barrier. This transport and the kinetics of perivascular macrophage/microglial turnover in the CNS likely depend on endothelial and leukocyte adhesion molecules such as vascular cell adhesion molecule-1 (VCAM-1), which has previously been shown to be upregulated on cerebrovascular endothelium in SIV encephalitis. To investigate the role of peripheral monocyte recruitment into the perivascular macrophage/microglial cell pool at the time of initial viral neuroinvasion, we examined the temporal relationships among perivascular macrophage/microglia density, endothelial VCAM-1 expression and localization of viral nucleic acid in the CNS of macaques acutely infected with pathogenic and nonpathogenic molecular clones of SIV. The concentration of CSF quinolinic acid, a marker of intrathecal immune and macrophage activation, was examined concurrently. We found that significant increases in the density of perivascular macrophages/microglia coincided with viral neuroinvasion and marked elevations in CSF quinolinic acid. Furthermore, combined in situ hybridization and immunohistochemistry demonstrated that infected perivascular cells were macrophages/microglia. These findings provide evidence suggesting that neuroinvasion occurs through an influx of infected monocytes which take up residence in the CNS as perivascular macrophages/microglia. VCAM-1 expression, however, was not clearly correlated with these events, thus its contribution to initial viral neuroinvasion is unclear.


Subject(s)
Central Nervous System Diseases/virology , Macrophages/virology , Microglia/virology , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Immunodeficiency Virus/isolation & purification , Animals , Brain/blood supply , Brain/pathology , Capillaries/chemistry , Central Nervous System Diseases/immunology , Cerebrovascular Circulation , DNA, Viral/analysis , Image Processing, Computer-Assisted , Immunohistochemistry , In Situ Hybridization , Leukocyte Count , Lymphocyte Activation/immunology , Macaca mulatta , Macrophages/chemistry , Macrophages/cytology , Male , Microglia/chemistry , Microglia/cytology , Quinolinic Acid/analysis , Simian Immunodeficiency Virus/genetics , Vascular Cell Adhesion Molecule-1/analysis
18.
Am J Pathol ; 149(5): 1459-67, 1996 Nov.
Article in English | MEDLINE | ID: mdl-8909235

ABSTRACT

The pathogenesis of neurological dysfunction associated with human immunodeficiency (HIV)-1 infection is uncertain. However, the presence of macrophage infiltrates in the central nervous system is a key feature of HIV encephalitis and is correlated with HIV-associated dementia. Moreover, it has been demonstrated that HIV-infected monocyte/macrophages can produce toxic substances that may play a critical role in the development of HIV-associated dementia. However, the exact mechanisms responsible for HIV infection and leukocyte recruitment to the central nervous system remain speculative. Similar to HIV-infected patients, simian immunodeficiency virus (SIV)-infected macaque monkeys develop immunosuppression and acquired immune deficiency syndrome (AIDS)-related inflammatory disorders, including AIDS encephalitis. In this study, we demonstrate that encephalitic brain from SIV-infected animals has elevated immunohistochemical expression of the C-C chemokines, macrophage inflammatory protein-1 alpha and -beta, RANTES, and monocyte chemotactic protein-3, and the C-X-C chemokine interferon-inducible protein-10. These findings suggest that one or all of of these chemokines could be involved in leukocyte recruitment to the brain in SIV-infected macaque monkeys.


Subject(s)
AIDS Dementia Complex/metabolism , Chemokines, CXC , Chemokines/biosynthesis , Encephalitis, Viral/etiology , Encephalitis, Viral/metabolism , Simian Acquired Immunodeficiency Syndrome/metabolism , Simian Immunodeficiency Virus/pathogenicity , AIDS Dementia Complex/etiology , Animals , Chemokine CCL4 , Chemokine CCL5/biosynthesis , Chemokine CCL7 , Chemokine CXCL10 , Cytokines/biosynthesis , Encephalomyelitis, Autoimmune, Experimental/metabolism , Immunohistochemistry , Macaca , Macrophage Inflammatory Proteins/biosynthesis , Monocyte Chemoattractant Proteins/biosynthesis , Simian Acquired Immunodeficiency Syndrome/etiology
19.
Blood ; 88(5): 1718-24, 1996 Sep 01.
Article in English | MEDLINE | ID: mdl-8829474

ABSTRACT

New world nonhuman primates of the genus Aotus (owl monkeys) can be categorized by 11 distinct karyotypes (K). It has been demonstrated that monkeys of K-VI persistently have one order of magnitude more eosinophils (EOS) in the peripheral blood than K-I monkeys. The purpose of this study was to investigate the basis for this difference and examine EOS recruitment using two cutaneous models of inflammation. Peripheral blood EOS were isolated on metrizamide gradients to > or = 95% purity and then used for phenotypic studies. There were no significant differences when comparing karyotypes in the ratio of normodense (K-I, 6.4% +/- 3.8%; K-VI, 21.1% +/- 8.8%) EOS or their survival in culture (K-I, 5.3% +/- 2.9% at 72 hours; K-VI, 2.8% +/- 0.7% at 72 hours) (P > .05). Examination of bone marrow revealed that K-VI monkeys had greater than fivefold more EOS and EOS precursors than K-I animals. To examine EOS function in recruitment, monkeys of each karyotype were given a single intradermal injection of Escherichia coli lipopolysaccharide (LPS) or human recombinant (PMN) and mononuclear cells occurred in response to LPS as early as 4 hours after injection; the severity of infiltration was similar in both karyotypes and at all time points up to 24 hours. In contrast, by 8 hours after intradermal injection of RANTES, leukocyte infiltration in K-I monkeys consisted mostly of PMN (94.8% +/- 0.7%) that were predominantly EOS. In comparison, there was essentially no infiltrate in K-VI animals at all time points. There was no difference in VCAM-1 expression in response to intradermal LPS or RANTES between the two karyotypes. These results suggest that the genetic basis of peripheralblood eosinophilia in K-VI owl monkeys is likely a function of heightened eosinophilopoiesis and depressed recruitment kinetics from the peripheral circulatory pool in response to RANTES.


Subject(s)
Aotidae/blood , Chemokine CCL5/pharmacology , Chemotactic Factors/pharmacology , Chemotaxis, Leukocyte/drug effects , Eosinophilia/physiopathology , Eosinophils/drug effects , Hematopoiesis , Receptors, Chemokine , Animals , Aotidae/classification , Aotidae/genetics , Bone Marrow/pathology , Drug Resistance , Female , Humans , Leukocyte Count , Lipopolysaccharides/pharmacology , Male , Monocytes/physiology , Neutrophils/physiology , Receptors, CCR5 , Receptors, Cytokine/genetics , Receptors, Cytokine/metabolism , Recombinant Fusion Proteins/pharmacology , Splenectomy , Vascular Cell Adhesion Molecule-1/metabolism
20.
J Med Primatol ; 25(4): 251-6, 1996 Aug.
Article in English | MEDLINE | ID: mdl-8906603

ABSTRACT

The larvae of Mesocestoides are rarely encountered in nonhuman primates, with most cases reported in baboons. Infection of macaques has been occasionally diagnosed, but Mesocestoides in the lung parenchyma is extremely rare. We have previously demonstrated that in macaques with terminal AIDS, simian immunodeficiency virus (SIV)-infected leukocytes are rarely found in cellular infiltrates associated with opportunistic infections or preexisting disease. Here we describe larvae (tetrathyridia) of the cestode Mesocestoides in the lung of an adult, pigtailed macaque (Macaca nemestrina) during acute SIV infection in which virus-positive cells are present within the cellular infiltrates. These results describe a rare parasitic disease in pigtailed macaques and demonstrate that lentivirus-infected leukocytes can be associated with inflammatory sites during acute infection.


Subject(s)
Cestode Infections/veterinary , Lung Diseases, Parasitic/veterinary , Lung/parasitology , Mesocestoides , Primate Diseases , Simian Acquired Immunodeficiency Syndrome/complications , Simian Immunodeficiency Virus/isolation & purification , Animals , Cestode Infections/complications , Cestode Infections/pathology , Larva , Lung/pathology , Lung/virology , Lung Diseases, Parasitic/complications , Lung Diseases, Parasitic/pathology , Macaca nemestrina , Male , Mesocestoides/isolation & purification , Skin/pathology
SELECTION OF CITATIONS
SEARCH DETAIL