Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Am J Primatol ; 86(6): e23619, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38482892

ABSTRACT

In apes and humans, neutrophil to lymphocyte ratio (NLR) can be used as a predictive indicator of a variety of clinical conditions, longevity, and physiological stress. In chimpanzees specifically, NLR systematically varies with age, rearing, sex, and premature death, indicating that NLR may be a useful diagnostic tool in assessing primate health. To date, just one very recent study has investigated NLR in old world monkeys and found lower NLR in males and nursery-reared individuals, as well as a negative relationship between NLR and disease outcomes. Given that baboons are increasingly used as research models, we aimed to characterize NLR in baboons by providing descriptive data and examinations of baboon NLR heritability, and of the relationships between NLR, age, rearing, and sex in 387 olive baboons (Papio anubis) between 6 months and 19 years of age. We found that (1) mother-reared baboons had higher NLRs than nursery-reared baboons; (2) females had higher NLRs than males; and (3) there was a quadratic relationship between NLR and age, such that middle-aged individuals had the highest NLR values. We also examined NLR as a function of transport to a new facility using a subset of the data. Baboons exhibited significantly higher transport NLRs compared to routine exam NLRs. More specifically, adult baboons had higher transport NLRs than routine NLRs, whereas juveniles showed no such difference, suggesting that younger animals may experience transport stress differently than older animals. We also found that transport NLR was heritable, whereas routine NLR was not, possibly suggesting that stress responses (as indicated in NLR) have a strong genetic component. Consistent with research in humans and chimpanzees, these findings suggest that NLR varies with important biological and life history variables and that NLR may be a useful health biomarker in baboons.


Subject(s)
Lymphocytes , Neutrophils , Papio anubis , Stress, Physiological , Animals , Female , Papio anubis/genetics , Papio anubis/physiology , Male , Pregnancy , Age Factors , Sex Factors
2.
Proc Natl Acad Sci U S A ; 118(37)2021 09 14.
Article in English | MEDLINE | ID: mdl-34507997

ABSTRACT

Late-stage anthrax infections are characterized by dysregulated immune responses and hematogenous spread of Bacillus anthracis, leading to extreme bacteremia, sepsis, multiple organ failure, and, ultimately, death. Despite the bacterium being nonhemolytic, some fulminant anthrax patients develop a secondary atypical hemolytic uremic syndrome (aHUS) through unknown mechanisms. We recapitulated the pathology in baboons challenged with cell wall peptidoglycan (PGN), a polymeric, pathogen-associated molecular pattern responsible for the hemostatic dysregulation in anthrax sepsis. Similar to aHUS anthrax patients, PGN induces an initial hematocrit elevation followed by progressive hemolytic anemia and associated renal failure. Etiologically, PGN induces erythrolysis through direct excessive activation of all three complement pathways. Blunting terminal complement activation with a C5 neutralizing peptide prevented the progressive deposition of membrane attack complexes on red blood cells (RBC) and subsequent intravascular hemolysis, heme cytotoxicity, and acute kidney injury. Importantly, C5 neutralization did not prevent immune recognition of PGN and shifted the systemic inflammatory responses, consistent with improved survival in sepsis. Whereas PGN-induced hemostatic dysregulation was unchanged, C5 inhibition augmented fibrinolysis and improved the thromboischemic resolution. Overall, our study identifies PGN-driven complement activation as the pathologic mechanism underlying hemolytic anemia in anthrax and likely other gram-positive infections in which PGN is abundantly represented. Neutralization of terminal complement reactions reduces the hemolytic uremic pathology induced by PGN and could alleviate heme cytotoxicity and its associated kidney failure in gram-positive infections.


Subject(s)
Acute Kidney Injury/prevention & control , Anemia, Hemolytic/prevention & control , Bacillus anthracis/chemistry , Cell Wall/chemistry , Complement C5/antagonists & inhibitors , Peptidoglycan/toxicity , Sepsis/complications , Acute Kidney Injury/etiology , Acute Kidney Injury/pathology , Anemia, Hemolytic/etiology , Anemia, Hemolytic/pathology , Animals , Anthrax/microbiology , Anthrax/pathology , Female , Hemolysis , Male , Papio , Sepsis/chemically induced
3.
Blood ; 138(2): 178-189, 2021 07 15.
Article in English | MEDLINE | ID: mdl-33598692

ABSTRACT

Activation of coagulation factor (F) XI promotes multiorgan failure in rodent models of sepsis and in a baboon model of lethal systemic inflammation induced by infusion of heat-inactivated Staphylococcus aureus. Here we used the anticoagulant FXII-neutralizing antibody 5C12 to verify the mechanistic role of FXII in this baboon model. Compared with untreated control animals, repeated 5C12 administration before and at 8 and 24 hours after bacterial challenge prevented the dramatic increase in circulating complexes of contact system enzymes FXIIa, FXIa, and kallikrein with antithrombin or C1 inhibitor, and prevented cleavage and consumption of high-molecular-weight kininogen. Activation of several coagulation factors and fibrinolytic enzymes was also prevented. D-dimer levels exhibited a profound increase in the untreated animals but not in the treated animals. The antibody also blocked the increase in plasma biomarkers of inflammation and cell damage, including tumor necrosis factor, interleukin (IL)-1ß, IL-6, IL-8, IL-10, granulocyte-macrophage colony-stimulating factor, nucleosomes, and myeloperoxidase. Based on clinical presentation and circulating biomarkers, inhibition of FXII prevented fever, terminal hypotension, respiratory distress, and multiorgan failure. All animals receiving 5C12 had milder and transient clinical symptoms and were asymptomatic at day 7, whereas untreated control animals suffered irreversible multiorgan failure and had to be euthanized within 2 days after the bacterial challenge. This study confirms and extends our previous finding that at least 2 enzymes of the contact activation complex, FXIa and FXIIa, play critical roles in the development of an acute and terminal inflammatory response in baboons challenged with heat-inactivated S aureus.


Subject(s)
Factor XII/metabolism , Multiple Organ Failure/metabolism , Multiple Organ Failure/microbiology , Staphylococcus aureus/physiology , Animals , Antibodies/therapeutic use , Blood Coagulation Disorders/complications , Blood Coagulation Disorders/immunology , Blood Coagulation Disorders/microbiology , Blood Platelets/metabolism , Cellular Microenvironment , Complement Activation , Factor XII/immunology , Female , Fibrinogen/metabolism , Hot Temperature , Inflammation/complications , Inflammation/pathology , Male , Multiple Organ Failure/immunology , Papio , Staphylococcal Infections/drug therapy , Staphylococcal Infections/immunology , Staphylococcal Infections/microbiology , Staphylococcal Infections/pathology , Survival Analysis
4.
Immun Inflamm Dis ; 12(6): e1318, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38923761

ABSTRACT

BACKGROUND: Major histocompatibility complex (MHC) class II molecules expressed on B cells, monocytes and dendritic cells present processed peptides to CD4+ T cells as one of the mechanisms to combat infection and inflammation. AIM: To study MHC II expression in a variety of nonhuman primate species, including New World (NWM) squirrel monkeys (Saimiri boliviensis boliviensis), owl monkeys (Aotus nancymae), common marmosets (Callithrix spp.), and Old World (OWM) rhesus (Macaca mulatta), baboons (Papio anubis). METHODS: Two clones of cross-reactive mouse anti-human HLADR monoclonal antibodies (mAb) binding were analyzed by flow cytometry to evaluate MHC II expression on NHP immune cells, including T lymphocytes in whole blood (WB) and peripheral blood mononuclear cells (PBMC). RESULTS: MHC class II antibody reactivity is seen with CD20+ B cells, CD14+ monocytes and CD3+ T lymphocytes. Specific reactivity with both clones was demonstrated in T lymphocytes: this reactivity was not inhibited by purified CD16 antibody but was completely inhibited when pre-blocked with purified unconjugated MHC II antibody. Freshly prepared PBMC also showed reactivity with T lymphocytes without any stimulation. Interestingly, peripheral blood from rhesus macaques and olive baboons (OWM) showed no such T lymphocyte associated MHCII antibody reactivity. DISCUSSION & CONCLUSION: Our results from antibody (MHC II) reactivity clearly show the potential existence of constitutively expressed (with no stimulation) MHC II molecules on T lymphocytes in new world monkeys. These results suggest that additional study is warranted to evaluate the functional and evolutionary significance of these finding and to better understand MHC II expression on T lymphocytes in new world monkeys.


Subject(s)
HLA-DR Antigens , Histocompatibility Antigens Class II , T-Lymphocytes , Animals , Histocompatibility Antigens Class II/immunology , HLA-DR Antigens/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Humans , Macaca mulatta , Antibodies, Monoclonal/immunology , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Saimiri/immunology , Callithrix/immunology , Flow Cytometry , Papio anubis/immunology , Platyrrhini/immunology
5.
Science ; 380(6648): 906-913, 2023 06 02.
Article in English | MEDLINE | ID: mdl-37262161

ABSTRACT

The rich diversity of morphology and behavior displayed across primate species provides an informative context in which to study the impact of genomic diversity on fundamental biological processes. Analysis of that diversity provides insight into long-standing questions in evolutionary and conservation biology and is urgent given severe threats these species are facing. Here, we present high-coverage whole-genome data from 233 primate species representing 86% of genera and all 16 families. This dataset was used, together with fossil calibration, to create a nuclear DNA phylogeny and to reassess evolutionary divergence times among primate clades. We found within-species genetic diversity across families and geographic regions to be associated with climate and sociality, but not with extinction risk. Furthermore, mutation rates differ across species, potentially influenced by effective population sizes. Lastly, we identified extensive recurrence of missense mutations previously thought to be human specific. This study will open a wide range of research avenues for future primate genomic research.


Subject(s)
Biological Evolution , Genetic Variation , Primates , Animals , Humans , Genome , Mutation Rate , Phylogeny , Primates/genetics , Population Density
6.
Comp Med ; 70(2): 160-169, 2020 04 01.
Article in English | MEDLINE | ID: mdl-32014083

ABSTRACT

Olive baboons (P. anubis) have provided a useful model of human diseases and conditions, including cardiac, respiratory, and infectious diseases; diabetes; and involving genetics, immunology, aging, and xenotransplantation. The development of a immunologically defined SPF baboons has advanced research further, especially for studies involving the immune system and immunosuppression. In this study, we compare normal immunologic changes of PBMC subsets, and their function in age-matched conventional and SPF baboons. Our results revealed that both groups have comparable numbers of different lymphocyte subsets, but phenotypic differences in central and effector memory T-cell subsets are more pronounced in CD4+ T cells. Despite equal proportions of CD3+ T cells among the conventional and SPF baboons, PBMC from the conventional group showed greater proliferative responses to phytohemagglutinin and pokeweed mitogen and higher numbers of IFNγ-producing cells after stimulation with concanavalin A or pokeweed mitogen, whereas plasma levels of the inflammatory cytokine TNFα were significantly higher in SPF baboons. Exposure of PBMC from conventional baboons to various Toll-like (TLR) ligands, including TLR3, TLR4, and TLR8, yielded increased numbers of IFNγ producing cells, whereas PBMC from SPF baboons stimulated with TLR5 or TLR6 ligand had more IFNγ-producing cells. These findings suggest that although lymphocyte subsets share many phenotypic and functional similarities in conventional and SPF baboons, specific differences in the immune function of lymphocytes could differentially influence the quality and quantity of their innate and adaptive immune responses. These differences should be considered in interpreting experimental outcomes, specifically in studies measuring immunologic endpoints.


Subject(s)
Immunity, Cellular/immunology , Monkey Diseases/immunology , Animals , Female , Male , Papio , Papio anubis , T-Lymphocytes/immunology
7.
Microb Genom ; 6(9)2020 09.
Article in English | MEDLINE | ID: mdl-32614763

ABSTRACT

Adenoviruses are a frequent cause of acute upper respiratory tract infections that can also cause disseminated disease in immunosuppressed patients. We identified a novel adenovirus, squirrel monkey adenovirus 1 (SqMAdV-1), as the cause of fatal infection in an immunocompromised squirrel monkey (Saimiri boliviensis) at the Keeling Center for Comparative Medicine and Research (KCCMR). Sequencing of SqMAdV-1 revealed that it is most closely related (80.4 % pairwise nucleotide identity) to the titi monkey (Plecturocebus cupreus) adenovirus (TMAdV). Although identified in the titi monkey, TMAdV is highly lethal in these monkeys, and they are not thought to be the natural host. While SqMAdV-1 is similar to other primate adenoviruses in size and genomic characteristics, a nucleotide polymorphism at the expected stop codon of the DNA polymerase gene results in a 126 amino acid extension at the carboxy terminus, a feature not previously observed among other primate adenoviruses. PCR testing and partial sequencing of 95 archived faecal samples from other squirrel monkeys (Saimiri boliviensis and Saimiri sciureus) housed at the KCCMR revealed the presence of three distinct, and apparently endemic species of adenoviruses. A grouping of ten squirrel monkey adenovirus variants has high similarity to SqMAdV-1. A single adenovirus variant (designated SqMAdV-3), detected in five monkeys, has similarity to tufted capuchin (Sapajus apella) adenoviruses. The largest group of adenovirus variants detected (designated SqMAdV-2.0-2.16) has very high similarity (93-99 %) to the TMAdV, suggesting that squirrel monkeys may be the natural host of the TMAdV.


Subject(s)
Adenoviridae Infections/mortality , Adenoviridae/classification , Saimiri/virology , Whole Genome Sequencing/methods , A549 Cells , Adenoviridae/genetics , Adenoviridae/isolation & purification , Adenoviridae Infections/veterinary , Animals , Cell Line , Codon, Terminator , Feces/virology , Female , Genome, Bacterial , Humans , Male , Phylogeny , Polymorphism, Single Nucleotide
8.
Comp Med ; 70(1): 75-82, 2020 02 01.
Article in English | MEDLINE | ID: mdl-31747991

ABSTRACT

Despite the lack of confirmed reports of an exogenous Simian betaretrovirus (SRV) isolated from baboons (Papio sp.), reports of simian endogenous gammaretrovirus (SERV) in baboons with complete genomes suggest that such viruses may be potentially infectious. In addition, serologic tests have repeatedly demonstrated antibody reactivity to SRV in baboons from multiple colonies. These findings complicate the management and use of such animals for research. To provide further insight into this situation, we performed in vitro and in vivo studies to determine if baboons are or can be infected with SRV. In our initial experiment, we were not able to isolate SRV from 6 seropositive or sero-indeterminate baboons by coculturing their peripheral blood mononuclear cells (PBMC) with macaque PBMC or permissive cell lines. In a subsequent experiment, we found that baboon PBMC infected in vitro with high dose SRV were permissive to virus replication. To test in vivo infectibil- ity, groups of naive baboons were infused intravenously with either (i) the same SRV tissue culture virus stocks used for the in vitro studies, (ii) SRV antibody positive and PCR positive macaque blood, (iii) SRV antibody positive or indeterminate, but PCR negative baboon blood, or (iv) SRV antibody and PCR negative baboon blood. Sustained SRV infection, as defined by reproducible PCR detection and/or antibody seroconversion, was confirmed in 2 of 3 baboons receiving tissue culture virus but not in any recipients of transfused blood from seropositive macaques or baboons. In conclusion, the data indicate that even though baboon cells can be infected experimentally with high doses of tissue culture grown SRV, baboons that are repeatedly SRV antibody positive and PCR negative are unlikely to be infected with exogenous SRV and thus are unlikely to transmit a virus that would threaten the SPF status of captive baboon colonies.


Subject(s)
Monkey Diseases/transmission , Papio , Retroviridae Infections/transmission , Animals , Betaretrovirus/isolation & purification , Female , Leukocytes, Mononuclear/virology , Male , Monkey Diseases/blood , Monkey Diseases/virology , Retroviridae Infections/blood , Retroviridae Infections/virology , Virus Replication
9.
Hepatol Commun ; 4(3): 371-386, 2020 Mar.
Article in English | MEDLINE | ID: mdl-32140655

ABSTRACT

Development of curative therapies for chronic hepatitis B virus (HBV) infection will likely require new animal models. Here, we evaluate HBV infection in squirrel monkeys based on the high-sequence homology of the HBV receptor, Na+/taurocholate co-transporting peptide (NTCP), between humans and squirrel monkeys. HBV PreS1 peptide was examined for binding human and squirrel monkey NTCP. Immunodeficient Fah -/- , NOD, Rag1 -/- , Il2Rg null (FNRG) mice engrafted with human or squirrel monkey hepatocytes were challenged with HBV or Woolly Monkey HBV (WMHBV). In addition, adult squirrel monkeys were inoculated with HBV, WMHBV, adeno-associated virus containing an infectious genome of HBV (AAV-HBV), and AAV-WMHBV. Finally, neonate squirrel monkeys were assessed for the potential of chronic infection with WMHBV. PreS1 peptide efficiently bound to human and squirrel monkey NTCP but not to mouse or capuchin NTCP. FNRG mice engrafted with squirrel monkey hepatocytes were susceptible to infection by WMHBV but not human HBV. Similarly, adult squirrel monkeys could be infected with WMHBV but not human HBV, whereas chimeric mice engrafted with human hepatocytes were susceptible to HBV but not WMHBV. Infection of squirrel monkeys with AAV-WMHBV yielded maximum viremia of 108 genomes/mL with detectable virus for up to 8 months. Notably, covalently closed circular DNA was detected in the liver of these animals. Infection of neonates with WMHBV led to detectable viremia for up to 6 months. Conclusions: Adult and neonate squirrel monkeys exhibited prolonged WMHBV viremia lasting 6-8 months. This is greater than twice the duration of viremia achieved in other nonhuman primates and suggests that squirrel monkeys may be a suitable model for testing HBV therapeutics.

10.
ILAR J ; 49(2): 157-69, 2008.
Article in English | MEDLINE | ID: mdl-18323578

ABSTRACT

The careful development, validation, and implementation of serodiagnostic assays can provide reliable results that make them a valuable tool in microbial quality control for nonhuman primates. This article includes identification and description of the components of assay development, including formulas for calculating the number of positive serum samples needed for assay validation and methods for calculating their diagnostic sensitivity and specificity. To ensure that assays are performing within predetermined specifications, there must be a quality control system that includes appropriate system and sample suitability controls as well as mechanisms to track assay performance over time. The section on quality assurance includes definitions of precision and accuracy in assay performance, and how to interpret these two factors using the Levey-Jennings chart, Westgard's rules, and other monitoring methods. Because all serologic assays are prone to false positive and false negative results, it is essential to interpret all diagnostic test results using both the expected prevalence of disease in the population and the population-specific assay performance characteristics that are determined during assay validation. The discussion on interpreting diagnostic test results also includes guidelines for calculating the positive and negative predictive values of an assay and for interpreting results based on the disease prevalence of the test population. A glossary provides definitions of commonly used terms.


Subject(s)
Primates/blood , Serologic Tests/methods , Animals , Quality Control , Reproducibility of Results , Serologic Tests/standards
11.
PLoS One ; 13(12): e0209391, 2018.
Article in English | MEDLINE | ID: mdl-30571742

ABSTRACT

A strain of Zika virus (ZIKV) of Asian origin associated with birth defects and neurological disorders has emerged and spread through the Americas. ZIKV was first isolated in the blood of nonhuman primates in Africa and has been detected in the blood, saliva, and urine of a few catarrhine species in both Africa and Asia, suggesting that nonhuman primates may serve as both a source and a reservoir of the virus. The recent introduction of ZIKV to human populations in the Americas presents the potential for the virus to spread into nonhuman primate reservoirs. Thus, it is critical to develop efficient and noninvasive detection methods to monitor the spread of the virus in wild nonhuman primate populations. Here, we describe a method for ZIKV detection in noninvasively collected fecal samples of a Neotropical primate. Fecal samples were collected from two captive squirrel monkeys (Saimiri boliviensis boliviensis) that were experimentally infected with ZIKV (Strain Mexico_1_44) and an additional two uninfected squirrel monkeys. Nucleic acids were extracted from these samples, and RT-qPCR was used to assay for the presence of ZIKV using primers flanking a 101 bp region of the NS5 gene. In both ZIKV-inoculated animals, ZIKV was detected 5-11 days post-infection, but was not detected in the uninfected animals. We compare the fecal results to ZIKV detection in serum, saliva, and urine samples from the same individuals. Our results indicate that fecal detection is a cost-effective, noninvasive method for monitoring wild populations of Neotropical primates as possible ZIKV reservoirs.


Subject(s)
Disease Reservoirs/virology , Feces/virology , RNA, Viral/isolation & purification , Zika Virus Infection/diagnosis , Zika Virus/isolation & purification , Animals , Disease Models, Animal , Environmental Monitoring/methods , Female , Humans , Pregnancy , Reverse Transcriptase Polymerase Chain Reaction , Saimiri/virology , Saliva/virology , Viral Nonstructural Proteins/genetics , Zika Virus/genetics , Zika Virus Infection/prevention & control , Zika Virus Infection/veterinary , Zika Virus Infection/virology
12.
Am J Trop Med Hyg ; 98(1): 173-177, 2018 01.
Article in English | MEDLINE | ID: mdl-29182145

ABSTRACT

The establishment of a sylvatic reservoir of Zika virus (ZIKV) in the Americas is dependent on the susceptibility of primates of sufficient population density, the duration and magnitude of viremia, and their exposure to the human mosquito-borne transmission cycle. To assess the susceptibility of squirrel (Saimiri sp.) and owl monkeys (Aotus sp.) to infection, we inoculated four animals of each species with ZIKV from the current epidemic. Viremia in the absence of detectible disease was observed in both species and seroconversion occurred by day 28. ZIKV was detected in the spleen of three owl monkeys: one at 7 days postinoculation (dpi) and two at 14 dpi. This study confirms the susceptibility to ZIKV infection of two Neotropical primate species that live in close proximity to humans in South America, suggesting that they could support a widespread sylvatic ZIKV cycle there. Collectively, establishment of a ZIKV sylvatic transmission cycle in South America would imperil eradication efforts and could provide a mechanism for continued exposure of humans to ZIKV infection and disease.


Subject(s)
Aotidae/virology , Primate Diseases/virology , Saimiri/virology , Zika Virus Infection/veterinary , Zika Virus , Animals , Disease Susceptibility/veterinary , Disease Susceptibility/virology , Female , Male , Viral Load/veterinary , Viremia/veterinary , Viremia/virology
13.
Comp Med ; 52(2): 97-110, 2002 Apr.
Article in English | MEDLINE | ID: mdl-12022401

ABSTRACT

Hantaviruses are a newly emerging group of rodent-borne viruses that have significant zoonotic potential. Human infection by hantaviruses can result in profound morbidity and mortality, with death rates as high as 50%, and potentially long-term cardiovascular consequences. Hantaviruses are carried by peridomestic and wild rodents worldwide and have occasionally been linked to infections in laboratory rodents. Because these viruses have been associated with significant human disease, they have become the subject of intense scientific investigation. In this review the reader is introduced to the hantaviruses, including hantavirus diseases and their pathogenesis. A review of the biology, morphology, and molecular biology of the hantaviruses with a brief overview of the ecology and biology of hantavirus-rodent pairs is also included. The risks of occupational exposure to hantaviruses, diagnosis of hantavirus infections, and methods for handling potentially infected rodents and tissues are discussed as well.


Subject(s)
Hantavirus Infections/physiopathology , Orthohantavirus , Animals , Animals, Laboratory/virology , Disease Transmission, Infectious , Disease Vectors , Orthohantavirus/classification , Orthohantavirus/genetics , Orthohantavirus/physiology , Orthohantavirus/ultrastructure , Hantavirus Infections/diagnosis , Hantavirus Infections/epidemiology , Hantavirus Infections/veterinary , Humans , Occupational Exposure , Zoonoses
14.
Comp Med ; 52(6): 548-54, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12540169

ABSTRACT

A novel Respirovirus was isolated from nasopharyngeal swab specimens from clinically normal laboratory guinea pigs, and was characterized and named caviid parainfluenza virus 3 (CavPIV-3). The CavPIV-3 is enveloped, is 100 to 300 nm in diameter, and has a characteristic 15-nm-diameter chevron-shaped virus ribonucleocapsid protein. Sequence analysis of the fusion glycoprotein of CavPIV-3 revealed it to be 94% identical to human and guinea pig parainfluenza 3 (PIV-3) viruses and 80% identical to bovine PIV-3. To determine whether CavPIV-3 causes clinical disease in laboratory guinea pigs and to compare the serologic response of guinea pigs to CavPIV-3 and to other paramyxoviruses, an infection study was performed, in which groups of guinea pigs were inoculated with CavPIV-3, Sendai virus, simian virus 5 (SV-5), murine pneumonia virus (PVM), or bovine PIV-3 virus. During the course of the study, guinea pigs were maintained in an infectious disease suite, housed in Micro-Isolator cages, and were only manipulated under a laminar flow hood. Clinical signs of disease were not observed in any of the paramyxovirus-inoculated guinea pigs during the eight-week course of the study, and histologic signs of disease were not evident at necropsy eight weeks after inoculation. Guinea pigs inoculated with CavPIV-3, Sendai virus, PVM, and bovine PIV-3 developed robust homologous or heterologous serologic responses. In contrast, guinea pigs inoculated with SV-5 developed modest or equivocal serologic responses, as assessed by use of an enzyme-linked immunosorbent assay. Further, use of the SV-5 enzyme-linked immunosorbent assay resulted in the highest degree of non-specific reactivity among all of the paramyxovirus assays. In summary, CavPIV-3 is a novel guinea pig Respirovirus that subclinically infects laboratory guinea pigs, resulting in a robust serologic response, but no observed clinical or histologic disease. The CavPIV-3 fusion glycoprotein gene sequence is available from GenBank as accession No. AF394241, and the CavPIV-3 virus is available from the American Type Culture Collection as accession No. DR-1547.


Subject(s)
Parainfluenza Virus 3, Human/isolation & purification , Respirovirus Infections/veterinary , Animals , Antibodies, Viral/analysis , Base Sequence , Chlorocebus aethiops , DNA Primers/chemistry , Electrophoresis, Agar Gel/veterinary , Enzyme-Linked Immunosorbent Assay/veterinary , Guinea Pigs , Molecular Sequence Data , Parainfluenza Virus 3, Human/pathogenicity , Parainfluenza Virus 3, Human/physiology , Parainfluenza Virus 3, Human/ultrastructure , RNA, Viral/analysis , Respirovirus Infections/physiopathology , Respirovirus Infections/virology , Reverse Transcriptase Polymerase Chain Reaction/veterinary , Sequence Analysis, DNA/veterinary , Vero Cells/virology , Viral Fusion Proteins/analysis
15.
Comp Med ; 61(3): 263-8, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21819697

ABSTRACT

Focally extensive alopecia affecting the distal limbs is a common clinical finding in rhesus macaque (Macaca mulatta) colonies and is both a regulatory and colony-health concern. We performed diagnostic examinations including physical exams, bloodwork, skin scrapes, surface cytology, and surface bacterial-fungal cultures on 17 rhesus macaques with this presentation of alopecia. Skin biopsies from alopecic skin obtained from each macaque were compared with those of normal skin from the same animal. Immunohistochemistry and metachromatic staining for inflammatory cells were performed to compare alopecic and normal skin. In addition, we compared these biopsies with those previously obtained from macaques with generalized alopecia and dermal inflammatory infiltrates consistent with cutaneous hypersensitivity disorders and with those from animals with normal haircoats. Bacterial and fungal cultures, skin scrapes, surface cytology, and bloodwork were unremarkable. Affected skin showed only mild histologic alteration, with rare evidence of trichomalacia and follicular loss. Numbers of mast cells and CD3+ lymphocytes did not differ between alopecic and normally haired skin from the same animal. The number of mast cells in alopecic skin from animals in the current cohort was significantly lower than that in skin of animals previously diagnosed with a cutaneous hypersensitivity disorder. Numbers of both mast cells and CD3+ lymphocytes in alopecic skin from the current cohort were similar to those from biopsies of animals with normal haircoats. Together, the clinical findings and pathology are consistent with a psychogenic origin for this pattern of alopecia in rhesus macaques.


Subject(s)
Alopecia/veterinary , Forearm/pathology , Leg/pathology , Macaca mulatta/psychology , Monkey Diseases/etiology , Alopecia/etiology , Alopecia/pathology , Alopecia/psychology , Animals , Diagnosis, Differential , Monkey Diseases/pathology , Monkey Diseases/psychology
16.
J Immunotoxicol ; 7(2): 102-13, 2010.
Article in English | MEDLINE | ID: mdl-19995244

ABSTRACT

Non-human primates (NHPs), primarily macaques, are commonly used as non-rodent species in pre-clinical safety assessment studies. The use of macaques in such studies is increasing largely due to the development of biopharmaceutical and immunomodulatory therapies that necessitates extensive safety testing. Macaques, commonly available for use in such studies, are infected by a rich flora of herpesviruses that cause persistent, latent, life-long infections. Primary infection of immune competent macaques is typically subclinical with very little associated morbidity and mortality only in very rare cases. A life-long consequence of herpesvirus infection is periodic stochastic and frequently asymptomatic recurrences from latency throughout an infected macaque's lifetime. With immune modulation or suppression, however, immune control of herpesvirus infections can be lost, resulting in significant disease and even death of the affected animals. Since macaques undergo primary infection with herpesviruses starting around 4-6 months-of-age when maternally-derived antibody begins to wane, it is difficult and costly to derive animals that are herpesvirus-free. Further, the herpesvirus flora and prevalence of infection in laboratory macaques mirrors that of the adult human population making the herpesvirus-infected macaque a reasonable model of the general human population. This review is intended to familiarize toxicologists performing preclinical drug safety studies with the basic biology, disease pathogenesis and consequences of immune suppression in herpesvirus-infected laboratory macaques.


Subject(s)
Animals, Laboratory/virology , Herpesviridae Infections/veterinary , Herpesviridae/physiology , Macaca/virology , Monkey Diseases/virology , Animals , Drug Evaluation, Preclinical , Herpesviridae/pathogenicity , Herpesviridae Infections/transmission , Herpesviridae Infections/virology , Immunocompromised Host/drug effects , Immunocompromised Host/immunology , Immunologic Factors/classification , Immunologic Factors/toxicity , Immunosuppression Therapy/veterinary , Models, Animal , Recurrence , Toxicity Tests , Xenobiotics/classification , Xenobiotics/toxicity
17.
J Immunotoxicol ; 7(2): 138-46, 2010.
Article in English | MEDLINE | ID: mdl-20441554

ABSTRACT

Non-human primates (NHP) are used to best understand and address pharmacology and toxicology obligations for human patients with highest and/or unmet need. In order to ensure the most appropriate care and use of NHP, it is important to understand the normal micro flora and fauna of NHP and ensure their utmost health to generate the most valuable and applicable data. There are many infections, including viral, bacterial, parasitic, and fungal that may perturb physiologic endpoints relevant to human health, and are essential to monitor and/or eradicate for NHP health. This publication captures a discussion involving the experience, knowledge and opinion from academic, industry and government experts regarding emerging and normal infections in NHP as they relate to immunotoxicity, and treatment and consequences of known infections.


Subject(s)
Communicable Diseases/microbiology , Immune System/drug effects , Monkey Diseases/microbiology , Opportunistic Infections/microbiology , Veterinary Medicine , Xenobiotics/toxicity , Animals , Communicable Disease Control , Drug Evaluation, Preclinical , Immunosuppression Therapy , Models, Animal , Monkey Diseases/prevention & control , Opportunistic Infections/prevention & control , Toxicity Tests
18.
Comp Med ; 58(1): 8-10, 2008 Feb.
Article in English | MEDLINE | ID: mdl-19793451

ABSTRACT

Macaque models have contributed to key advances in our basic knowledge of behavior, anatomy, and physiology as well as to our understanding of a wide variety of human diseases. This issue of Comparative Medicine focuses on several of the viral agents (members of Retroviridae, Herpesviridae and 2 small DNA viruses) that can infect both nonhuman primates and humans as well as confound research studies. Featured articles also address the challenges of developing colonies of macaques and other nonhuman primates that are truly specific pathogen-free for these and other adventitious infectious agents.


Subject(s)
Macaca/virology , Specific Pathogen-Free Organisms/physiology , Virus Diseases/veterinary , Animals , DNA Virus Infections/genetics , DNA Virus Infections/veterinary , Genetic Predisposition to Disease , Genome, Viral , Herpesviridae Infections/genetics , Herpesviridae Infections/veterinary , Humans , Macaca/genetics , RNA Viruses/genetics , Research/trends , Retroviridae/genetics , Retroviridae Infections/genetics , Retroviridae Infections/veterinary , Virus Diseases/genetics
SELECTION OF CITATIONS
SEARCH DETAIL