Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
J Clin Invest ; 134(7)2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38557496

ABSTRACT

Programmed cell death protein 1 (PD-1) is an immune checkpoint marker commonly expressed on memory T cells and enriched in latently HIV-infected CD4+ T cells. We engineered an anti-PD-1 chimeric antigen receptor (CAR) to assess the impact of PD-1 depletion on viral reservoirs and rebound dynamics in SIVmac239-infected rhesus macaques (RMs). Adoptive transfer of anti-PD-1 CAR T cells was done in 2 SIV-naive and 4 SIV-infected RMs on antiretroviral therapy (ART). In 3 of 6 RMs, anti-PD-1 CAR T cells expanded and persisted for up to 100 days concomitant with the depletion of PD-1+ memory T cells in blood and tissues, including lymph node CD4+ follicular helper T (TFH) cells. Loss of TFH cells was associated with depletion of detectable SIV RNA from the germinal center (GC). However, following CAR T infusion and ART interruption, there was a marked increase in SIV replication in extrafollicular portions of lymph nodes, a 2-log higher plasma viremia relative to controls, and accelerated disease progression associated with the depletion of CD8+ memory T cells. These data indicate anti-PD-1 CAR T cells depleted PD-1+ T cells, including GC TFH cells, and eradicated SIV from this immunological sanctuary.


Subject(s)
CD4-Positive T-Lymphocytes , Receptors, Chimeric Antigen , Simian Acquired Immunodeficiency Syndrome , Simian Immunodeficiency Virus , Animals , CD4-Positive T-Lymphocytes/immunology , Germinal Center/immunology , HIV Infections/therapy , Macaca mulatta/metabolism , Programmed Cell Death 1 Receptor , Receptors, Chimeric Antigen/genetics , Simian Acquired Immunodeficiency Syndrome/therapy
2.
Front Virol ; 32023.
Article in English | MEDLINE | ID: mdl-37383986

ABSTRACT

Zika virus (ZIKV) is a mosquito-borne flavivirus that causes an acute febrile illness. ZIKV can be transmitted between sexual partners and from mother to fetus. Infection is strongly associated with neurologic complications in adults, including Guillain-Barré syndrome and myelitis, and congenital ZIKV infection can result in fetal injury and congenital Zika syndrome (CZS). Development of an effective vaccine is imperative to protect against ZIKV vertical transmission and CZS. Recombinant Vesicular Stomatitis virus (rVSV) is a highly effective and safe vector for the delivery of foreign immunogens for vaccine purposes. Here, we evaluate an rVSV vaccine expressing the full length pre-membrane (prM) and ZIKV envelope (E) proteins (VSV-ZprME), shown to be immunogenic in murine models of ZIKV infection, for its capacity to induce immune responses in nonhuman primates. Moreover, we assess the efficacy of the rVSVΔM-ZprME vaccine in the protection of pigtail macaques against ZIKV infection. Administration of the rVSVΔM-ZprME vaccine was safe, but it did not induce robust anti-ZIKV T-cell responses, IgM or IgG antibodies, or neutralizing antibodies in most animals. Post ZIKV challenge, animals that received the rVSVΔM control vaccine lacking ZIKV antigen had higher levels of plasma viremia compared to animals that received the rVSVΔM-ZprME vaccine. Anti-ZIKV neutralizing Ab titers were detected in a single animal that received the rVSVΔM-ZprME vaccine that was associated with reduced plasma viremia. The overall suboptimal ZIKV-specific cellular and humoral responses post-immunization indicates the rVSVΔM-ZprME vaccine did not elicit an immune response in this pilot study. However, recall antibody response to the rVSVΔM-ZprME vaccine indicates it may be immunogenic and further developments to the vaccine construct could enhance its potential as a vaccine candidate in a nonhuman primate pre-clinical model.

3.
Hepatol Commun ; 7(2): e0009, 2023 02 01.
Article in English | MEDLINE | ID: mdl-37074875

ABSTRACT

BACKGROUND AND AIMS: Adeno-associated virus (AAV) vectors are widely used to deliver therapeutic transgenes to distinct tissues, including the liver. Vectors based on naturally occurring AAV serotypes as well as vectors using engineered capsids have shown variations in tissue tropism and level of transduction between different mouse models. Moreover, results obtained in rodents frequently lack translatability into large animal studies. In light of the increasing interest in AAV vectors for human gene therapy, an increasing number of studies are being performed in nonhuman primates. To keep animal numbers to a minimum and thus optimize the process of AAV capsid selection, we developed a multiplex barcoding approach to simultaneously evaluate the in vivo vector performance for a set of serotypes and capsid-engineered AAV vectors across multiple organs. APPROACH AND RESULTS: Vector biodistribution and transgene expression were assessed by quantitative PCR, quantitative reverse transcription PCR, vector DNA amplicon Illumina sequencing and vRNAseq in male and female rhesus macaques simultaneously dosed with a mixture of barcoded naturally occurring or engineered AAV vectors encoding the same transgene. As expected, our findings show animal-to-animal variation in both the biodistribution and tissue transduction pattern, which was partly influenced by each animal's distinctive serological status. CONCLUSIONS: This method offers a robust approach to AAV vector optimization that can be used to identify and validate AAV vectors for gene delivery to potentially any anatomical site or cell type.


Subject(s)
Capsid , Dependovirus , Animals , Mice , Female , Male , Humans , Capsid/metabolism , Dependovirus/genetics , Dependovirus/metabolism , Tissue Distribution , Macaca mulatta/genetics , Macaca mulatta/metabolism , Capsid Proteins/genetics , Capsid Proteins/metabolism , Genetic Therapy/methods
4.
Infect Immun ; 90(6): e0013122, 2022 06 16.
Article in English | MEDLINE | ID: mdl-35583346

ABSTRACT

Mycoplasma genitalium is a sexually transmitted bacterial pathogen that causes urogenital disease in men and women. M. genitalium infections can persist for months to years and can ascend to the upper reproductive tract in women where it is associated with serious sequelae including pelvic inflammatory disease, tubal factor infertility, and preterm birth. An animal model is needed to understand immune evasion strategies that allow persistence, mechanisms of ascending infection, and factors associated with clearance. In earlier studies, we determined that pig-tailed macaques are susceptible to cervical infection; however, not all primates were successfully infected, persistence varied between animals, and ascension to the upper reproductive tract was not observed after 4 or 8 weeks of follow-up. Building on our previous findings, we refined our inoculation methods to increase infection rates, extended observation to 18 weeks, and comprehensively sampled the upper reproductive tract to detect ascending infection. With these improvements, we established infection in all (3/3) primates inoculated with M. genitalium and demonstrated lower tract persistence for 16 to 18 weeks. Ascension to the upper reproductive tract at endpoint was observed in two out of three primates. All three primates developed serum and local antibodies reacting primarily to the MgpB and MgpC adherence proteins. Elevated genital polymorphonuclear leukocytes (PMNs) and inflammatory cytokines and chemokines, erythema of the ectocervix in one primate, and histologic evidence of vaginitis and endocervicitis in two primates suggest a mild to moderate inflammatory response to infection. This model will be valuable to understand the natural history of M. genitalium infection including mechanisms of persistence, immune evasion, and ascension to the upper reproductive tract.


Subject(s)
Mycoplasma Infections , Mycoplasma genitalium , Premature Birth , Reproductive Tract Infections , Animals , Female , Humans , Infant, Newborn , Macaca nemestrina , Mycoplasma Infections/microbiology
5.
AIDS Res Hum Retroviruses ; 37(7): 505-509, 2021 07.
Article in English | MEDLINE | ID: mdl-33356854

ABSTRACT

Coccidioidomycosis is a common fungal infection in people living with HIV-1, particularly in southwest regions of the United States where the Coccidioides sp. is endemic, but rates of infection have significantly declined in the era of potent combination antiretroviral therapy (cART). Natural coccidioidomycosis also occurs in outdoor-housed macaques residing in the southwestern states that are utilized in biomedical research. Here, we report on a recrudescent case of previously treated, naturally occurring coccidioidomycosis in a pigtail macaque that was experimentally infected with simian immunodeficiency virus (SIV) and virally suppressed on cART. Coccidioides IgG antibody titer became detectable before discontinuation of cART, but symptomatic coccidioidomycosis developed subsequent to cART withdrawal. This animal was screened and treated in accordance with the guidelines for the prevention and treatment of coccidioidomycosis, suggesting that macaques with a history of coccidioidomycosis should be excluded from enrollment in HIV studies. Continual monitoring for known endemic pathogens based on the colony of origin is also recommended for animals utilized for HIV/AIDS research.


Subject(s)
Coccidioidomycosis , HIV Infections , Simian Acquired Immunodeficiency Syndrome , Simian Immunodeficiency Virus , Animals , Coccidioidomycosis/drug therapy , HIV Infections/complications , HIV Infections/drug therapy , Humans , Macaca nemestrina , Recurrence , Simian Acquired Immunodeficiency Syndrome/complications , Simian Acquired Immunodeficiency Syndrome/drug therapy , Viral Load
6.
Hum Gene Ther ; 32(1-2): 96-112, 2021 01.
Article in English | MEDLINE | ID: mdl-32998579

ABSTRACT

Adeno-associated virus (AAV) vectors such as AAV6, which shows tropism for primary human CD4+ T cells in vitro, are being explored for delivery of anti-HIV therapeutic modalities in vivo. However, pre-existing immunity and sequestration in nontarget organs can significantly hinder their performance. To overcome these challenges, we investigated whether immunosuppression would allow gene delivery by AAV6 or targeted AAV6 derivatives in seropositive rhesus macaques. Animals were immune suppressed with rapamycin before intravenous (IV) or subcutaneous (SC) delivery of AAV, and we monitored vector biodistribution, gene transfer, and safety. Macaques received phosphate-buffered saline, AAV6 alone, or an equal dose of AAV6 and an AAV6-55.2 vector retargeted to CD4 through a direct ankyrin repeat protein (DARPin). AAV6 and AAV6-55.2 vector genomes were found in peripheral blood mononuclear cells and most organs up to 28 days postadministration, with the highest levels seen in liver, spleen, lymph nodes (LNs), and muscle, suggesting that retargeting did not prevent vector sequestration. Despite vector genome detection, gene expression from AAV6-55.2 was not detected in any tissue. SC injection of AAV6 facilitated efficient gene expression in muscle adjacent to the injection site, plus low-level gene expression in spleen, LNs, and liver, whereas gene expression following IV injection of AAV6 was predominantly seen in the spleen. AAV vectors were well tolerated, although elevated liver enzymes were detected in three of four AAV-treated animals 14 days after rapamycin withdrawal. One SC-injected animal had muscle inflammation proximal to the injection site, plus detectable T cell responses against transgene and AAV6 capsid at study finish. Overall, our data suggest that rapamycin treatment may offer a possible strategy to express anti-HIV therapeutics such as broadly neutralizing antibodies from muscle. This study provides important safety and efficacy data that will aid study design for future anti-HIV gene therapies.


Subject(s)
Dependovirus , Genetic Vectors , Animals , Dependovirus/genetics , Designed Ankyrin Repeat Proteins , Genetic Vectors/genetics , Humans , Leukocytes, Mononuclear , Macaca mulatta , Sirolimus/therapeutic use , Tissue Distribution
7.
Stem Cell Reports ; 13(1): 91-104, 2019 07 09.
Article in English | MEDLINE | ID: mdl-31204301

ABSTRACT

Myeloid-differentiated hematopoietic stem cells (HSCs) have contributed to a number of novel treatment approaches for lysosomal storage diseases of the central nervous system (CNS), and may also be applied to patients infected with HIV. We quantified hematopoietic stem and progenitor cell (HSPC) trafficking to 20 tissues including lymph nodes, spleen, liver, gastrointestinal tract, CNS, and reproductive tissues. We observed efficient marking of multiple macrophage subsets, including CNS-associated myeloid cells, suggesting that HSPC-derived macrophages are a viable approach to target gene-modified cells to tissues. Gene-marked cells in the CNS were unique from gene-marked cells at any other physiological sites including peripheral blood. This novel finding suggests that these cells were derived from HSPCs, migrated to the brain, were compartmentalized, established myeloid progeny, and could be targeted for lifelong delivery of therapeutic molecules. Our findings have highly relevant implications for the development of novel therapies for genetic and infectious diseases of the CNS.


Subject(s)
Central Nervous System/cytology , Hematopoietic Stem Cell Transplantation , Myeloid Cells/cytology , Animals , Cell Differentiation , Cell- and Tissue-Based Therapy/methods , Genetic Therapy/methods , Hematopoietic Stem Cells , Longitudinal Studies , Lysosomal Storage Diseases/pathology , Lysosomal Storage Diseases/therapy , Macaca nemestrina , Macrophages/cytology
8.
Nat Commun ; 9(1): 4438, 2018 10 25.
Article in English | MEDLINE | ID: mdl-30361514

ABSTRACT

Allogeneic transplantation (allo-HCT) has led to the cure of HIV in one individual, raising the question of whether transplantation can eradicate the HIV reservoir. To test this, we here present a model of allo-HCT in SHIV-infected, cART-suppressed nonhuman primates. We infect rhesus macaques with SHIV-1157ipd3N4, suppress them with cART, then transplant them using MHC-haploidentical allogeneic donors during continuous cART. Transplant results in ~100% myeloid donor chimerism, and up to 100% T-cell chimerism. Between 9 and 47 days post-transplant, terminal analysis shows that while cell-associated SHIV DNA levels are reduced in the blood and in lymphoid organs post-transplant, the SHIV reservoir persists in multiple organs, including the brain. Sorting of donor-vs.-recipient cells reveals that this reservoir resides in recipient cells. Moreover, tetramer analysis indicates a lack of virus-specific donor immunity post-transplant during continuous cART. These results suggest that early post-transplant, allo-HCT is insufficient for recipient reservoir eradication despite high-level donor chimerism and GVHD.


Subject(s)
Disease Reservoirs/virology , Hematopoietic Stem Cell Transplantation , Major Histocompatibility Complex , Simian Immunodeficiency Virus/physiology , Transplantation, Haploidentical , Animals , Antiretroviral Therapy, Highly Active , CD8-Positive T-Lymphocytes/immunology , DNA, Viral/metabolism , Macaca mulatta , RNA, Viral/metabolism , Simian Acquired Immunodeficiency Syndrome/drug therapy , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/virology , Transplantation, Homologous
9.
JCI Insight ; 2(4): e91230, 2017 02 23.
Article in English | MEDLINE | ID: mdl-28239658

ABSTRACT

The conditioning regimen used as part of the Berlin patient's hematopoietic cell transplant likely contributed to his eradication of HIV infection. We studied the impact of conditioning in simian-human immunodeficiency virus-infected (SHIV-infected) macaques suppressed by combination antiretroviral therapy (cART). The conditioning regimen resulted in a dramatic, but incomplete depletion of CD4+ and CD8+ T cells and CD20+ B cells, increased T cell activation and exhaustion, and a significant loss of SHIV-specific Abs. The disrupted T cell homeostasis and markers of microbial translocation positively correlated with an increased viral rebound after cART interruption. Quantitative viral outgrowth and Tat/rev-induced limiting dilution assays showed that the size of the latent SHIV reservoir did not correlate with viral rebound. These findings identify perturbations of the immune system as a mechanism for the failure of autologous transplantation to eradicate HIV. Thus, transplantation strategies may be improved by incorporating immune modulators to prevent disrupted homeostasis, and gene therapy to protect transplanted cells.


Subject(s)
CD4-Positive T-Lymphocytes/radiation effects , CD8-Positive T-Lymphocytes/radiation effects , HIV Infections/immunology , HIV-1/radiation effects , Hematopoietic Stem Cell Transplantation/methods , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Immunodeficiency Virus/radiation effects , Transplantation Conditioning/methods , Whole-Body Irradiation , Animals , Anti-Retroviral Agents/therapeutic use , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , HIV Infections/drug therapy , Homeostasis/radiation effects , Lentivirus Infections/drug therapy , Lentivirus Infections/immunology , Macaca nemestrina , Simian Acquired Immunodeficiency Syndrome/drug therapy , Transplantation, Autologous , Viral Load/radiation effects
10.
Comp Med ; 64(3): 234-9, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24956217

ABSTRACT

An adult, gravid, female pigtailed macaque (Macaca nemestrina) presented for facial swelling centered on the left mandible that was approximately 5 cm wide. Differential diagnoses included infectious, inflammatory, and neoplastic origins. Definitive antemortem diagnosis was not possible, and the macaque's condition worsened despite supportive care. Necropsy findings included a mandibular mass that was locally invasive and expansile, encompassing approximately 80% of the left mandibular bone. The mass replaced portions of the soft palate, hard palate, sinuses, ear canal, and the caudal-rostral calvarium and masseter muscle. Histologically, the mass was a neoplasm that was poorly circumscribed, unencapsulated, and infiltrative invading regional bone and soft tissue. The mass consisted of polygonal squamous epithelial cells with intercellular bridging that breached the epithelial basement membrane and formed invasive nests, cords, and trabeculae. The mitotic rate averaged 3 per 400× field of view, with occasional bizarre mitotic figures. Epithelial cells often exhibited dyskeratosis, and the nests often contained compact lamellated keratin (keratin pearls). The neoplasm was positive via immunohistochemistry for pancytokeratin, variably positive for S100, and negative for vimentin, smooth muscle actin, and desmin. The gross, histologic, and immunohistochemical findings were consistent with an aggressive oral squamous cell carcinoma. The neoplasm was negative via PCR for papilloma virus. In general, neoplasia in macaques is rare. Although squamous cell carcinomas are one of the most common oral neoplasia in many species, to our knowledge this case represents the first reported oral squamous cell carcinoma in a pigtailed macaque.


Subject(s)
Carcinoma, Squamous Cell/veterinary , Macaca nemestrina , Monkey Diseases/pathology , Mouth Neoplasms/veterinary , Animals , Carcinoma, Squamous Cell/pathology , Fatal Outcome , Female , Immunohistochemistry/veterinary , Keratins/metabolism , Mouth Neoplasms/pathology
11.
Comp Med ; 64(1): 63-7, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24512963

ABSTRACT

A 2.25-y-old male pigtailed macaque (Macaca nemestrina) was experimentally irradiated and received a bone marrow transplant. After transplantation and engraftment, the macaque had unexpected recurring pancytopenia and dependent edema of the prepuce, scrotum, and legs. The diagnostic work-up included a blood smear, which revealed a trypomastigote consistent with Trypanosoma cruzi, the causative agent of Chagas disease (CD). We initially hypothesized that the macaque had acquired the infection when it lived in Georgia. However, because the animal had received multiple blood transfusions, all blood donors were screened for CD. One male pigtailed macaque blood donor, which was previously housed in Louisiana, was positive for T. cruzi antibodies via serology. Due to the low prevalence of infection in Georgia, the blood transfusion was hypothesized to be the source of T. cruzi infection. The transfusion was confirmed as the mechanism of transmission when screening of archived serum revealed seroconversion after blood transfusion from the seropositive blood donor. The macaque made a full clinical recovery, and further follow-up including thoracic radiography, echocardiography, and gross necropsy did not show any abnormalities associated with CD. Other animals that received blood transfusions from the positive blood donor were tested, and one additional pigtailed macaque on the same research protocol was positive for T. cruzi. Although CD has been reported to occur in many nonhuman primate species, especially pigtailed macaques, the transmission of CD via blood transfusion in nonhuman primates has not been reported previously.


Subject(s)
Blood Transfusion/veterinary , Chagas Disease/veterinary , Immunocompromised Host , Macaca nemestrina/parasitology , Monkey Diseases/parasitology , Trypanosoma cruzi/isolation & purification , Animals , Antibodies, Protozoan/blood , Biomarkers/blood , Chagas Disease/blood , Chagas Disease/immunology , Chagas Disease/transmission , Dose Fractionation, Radiation , Genetic Therapy , Macaca nemestrina/blood , Macaca nemestrina/immunology , Male , Models, Animal , Monkey Diseases/blood , Monkey Diseases/immunology , Stem Cell Transplantation , Transfusion Reaction , Trypanosoma cruzi/immunology
12.
PLoS Pathog ; 8(10): e1002962, 2012.
Article in English | MEDLINE | ID: mdl-23055934

ABSTRACT

Two gammaherpesviruses, Epstein-Barr virus (EBV) (Lymphocryptovirus genus) and Kaposi's sarcoma-associated herpesvirus (KSHV) (Rhadinovirus genus) have been implicated in the etiology of AIDS-associated lymphomas. Homologs of these viruses have been identified in macaques and other non-human primates. In order to assess the association of these viruses with non-human primate disease, archived lymphoma samples were screened for the presence of macaque lymphocryptovirus (LCV) homologs of EBV, and macaque rhadinoviruses belonging to the RV1 lineage of KSHV homologs or the more distant RV2 lineage of Old World primate rhadinoviruses. Viral loads were determined by QPCR and infected cells were identified by immunolabeling for different viral proteins. The lymphomas segregated into three groups. The first group (n = 6) was associated with SIV/SHIV infections, contained high levels of LCV (1-25 genomes/cell) and expressed the B-cell antigens CD20 or BLA.36. A strong EBNA-2 signal was detected in the nuclei of the neoplastic cells in one of the LCV-high lymphomas, indicative of a type III latency stage. None of the lymphomas in this group stained for the LCV viral capsid antigen (VCA) lytic marker. The second group (n = 5) was associated with D-type simian retrovirus-2 (SRV-2) infections, contained high levels of RV2 rhadinovirus (9-790 genomes/cell) and expressed the CD3 T-cell marker. The third group (n = 3) was associated with SIV/SHIV infections, contained high levels of RV2 rhadinovirus (2-260 genomes/cell) and was negative for both CD20 and CD3. In both the CD3-positive and CD3/CD20-negative lymphomas, the neoplastic cells stained strongly for markers of RV2 lytic replication. None of the lymphomas had detectable levels of retroperitoneal fibromatosis herpesvirus (RFHV), the macaque RV1 homolog of KSHV. Our data suggest etiological roles for both lymphocryptoviruses and RV2 rhadinoviruses in the development of simian AIDS-associated lymphomas and indicate that the virus-infected neoplastic lymphoid cells are derived from different lymphocyte lineages and differentiation stages.


Subject(s)
Herpesvirus 4, Human , Herpesvirus 8, Human , Lymphocryptovirus/isolation & purification , Lymphoma, AIDS-Related/virology , Simian Acquired Immunodeficiency Syndrome/virology , Animals , Antigens, CD20/biosynthesis , Antigens, Neoplasm/biosynthesis , CD3 Complex/biosynthesis , Epstein-Barr Virus Nuclear Antigens/biosynthesis , Herpesvirus 4, Human/classification , Herpesvirus 4, Human/genetics , Herpesvirus 8, Human/classification , Herpesvirus 8, Human/genetics , Lymphocryptovirus/genetics , Macaca , Mason-Pfizer monkey virus/genetics , Mason-Pfizer monkey virus/isolation & purification , Rhadinovirus/isolation & purification , Simian Acquired Immunodeficiency Syndrome/complications , Simian Immunodeficiency Virus/genetics , Simian Immunodeficiency Virus/isolation & purification , Tumor Cells, Cultured , Viral Load , Viral Proteins/biosynthesis , Viral Proteins/genetics
13.
Mol Cell Proteomics ; 11(9): 605-18, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22580588

ABSTRACT

Morphine has long been known to have immunosuppressive properties in vivo, but the molecular and immunologic changes induced by it are incompletely understood. To explore how these changes interact with lentiviral infections in vivo, animals from two nonhuman primate species (African green monkeys and pigtailed macaques) were provided morphine and studied using a systems biology approach. Biological specimens were obtained from multiple sources (e.g. lymph node, colon, cerebrospinal fluid, and peripheral blood) before and after the administration of morphine (titrated up to a maximum dose of 5 mg/kg over a period of 20 days). Cellular immune, plasma cytokine, and proteome changes were measured and morphine-induced changes in these parameters were assessed on an interorgan, interindividual, and interspecies basis. In both species, morphine was associated with decreased levels of Ki-67(+) T-cell activation but with only minimal changes in overall T-cell counts, neutrophil counts, and NK cell counts. Although changes in T-cell maturation were observed, these varied across the various tissue/fluid compartments studied. Proteomic analysis revealed a morphine-induced suppressive effect in lymph nodes, with decreased abundance of protein mediators involved in the functional categories of energy metabolism, signaling, and maintenance of cell structure. These findings have direct relevance for understanding the impact of heroin addiction and the opioids used to treat addiction as well as on the potential interplay between opioid abuse and the immunological response to an infective agent.


Subject(s)
Immune Tolerance , Immunosuppressive Agents/pharmacology , Lymphocyte Activation/drug effects , Morphine/pharmacology , Proteomics , Animals , Chlorocebus aethiops , Colon/drug effects , Cytokines/blood , Energy Metabolism/drug effects , Ki-67 Antigen , Killer Cells, Natural/drug effects , Lymph Nodes/drug effects , Lymphocyte Count , Macaca nemestrina , Morphine/blood , Morphine/cerebrospinal fluid , Neutrophils/drug effects , Proteome/analysis , Signal Transduction/drug effects , Substance-Related Disorders , T-Lymphocytes/drug effects , T-Lymphocytes/immunology
14.
Comp Med ; 61(1): 86-91, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21819686

ABSTRACT

Fat embolization (FE), the introduction of bone marrow elements into circulation, is a known complication of bone fractures. Although FE has been described in other animal models, this study represents the first reported cases of FE and bone marrow embolism in nonhuman primates. Histopathologic findings from cynomolgus macaques (Macaca fascicularis) indicated that in all 5 cases, fat and bone marrow embolization occurred subsequent to multiple bone marrow biopsies. In the most severe case, extensive embolization was associated pulmonary damage consistent with acute respiratory distress syndrome. Fat embolism syndrome (FES) is an infrequent clinical outcome of FE and is triggered by systemic biochemical and mechanical responses to fat in circulation. Although clinical criteria diagnostic of FES were not investigated at the time of death, this severe case may represent the fulminant form of FES, which occurs within 12 h after trauma. Bone marrow biopsy as an etiology of FES has been reported only once in humans. In addition, the association of embolization with bone marrow biopsies suggests that nonhuman primates may be a useful animal model of FE. FE and FES represent important research confounders and FES should be considered as a differential diagnosis for clinical complications subsequent to skeletal trauma.


Subject(s)
Bone Marrow/pathology , Embolism, Fat/veterinary , Macaca fascicularis , Monkey Diseases/diagnosis , Pulmonary Embolism/veterinary , Animals , Biopsy , Diagnosis, Differential , Embolism, Fat/diagnosis , Female , Postoperative Complications , Pulmonary Embolism/diagnosis
15.
J Am Assoc Lab Anim Sci ; 50(2): 258-62, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21439222

ABSTRACT

An adult, female, pig-tailed macaque (Macaca nemestrina) of Indonesian origin presented with profound weight loss, anemia (PCV, 29%; normal, 36% to 45%), hypoalbuminemia (1.0 g/dL; normal, 3.5 to 5.2 g/dL), elevated alkaline phosphatase (1990 U/L; normal, 26 to 98 U/L), and an elevated erythrocyte sedimentation rate (75 mm/h; normal, less than 20 mm/h). Abdominal ultrasonography demonstrated an enlarged liver with hyperechoic areas. Euthanasia was performed. Grossly, the liver had multifocal, effacing, white masses throughout and was enlarged with rounded edges. There were 2, small nodules in the right lung lobes. Histologically, the hepatic masses were densely fibrous-encapsulated granulomas with vast central necrosis. The lung nodules also were maturing granulomas, and one kidney and one atrium had small, early granulomas. Fite acid-fast stains of liver and lung revealed very few acid-fast bacilli. PCR analysis of paraffin-embedded liver identified Mycobacterium tuberculosis complex. Culture of the liver was negative twice. This macaque had 16 negative intradermal tuberculin skin tests over the course of 6 y. We hypothesize that the animal arrived with a latent hepatic or enteric infection that later recrudesced and disseminated. Primary hepatic mycobacteriosis is not a typical presentation of tuberculosis in macaques. Negative tuberculin skin tests can be seen with latent infections and extrapulmonary tuberculosis such as Pott disease. This case underscores the problems associated with current surveillance procedures and the risks associated with latent mycobacterial infections in macaques.


Subject(s)
Hepatomegaly/veterinary , Macaca nemestrina/microbiology , Monkey Diseases/pathology , Mycobacterium tuberculosis/physiology , Tuberculosis/veterinary , Animals , Female , Hepatomegaly/pathology , Lung/pathology , Lung Diseases/diagnosis , Lung Diseases/pathology , Lung Diseases/veterinary , Monkey Diseases/diagnosis , Tuberculosis/diagnosis , Tuberculosis/pathology
16.
Toxicol Pathol ; 37(6): 799-804, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19773593

ABSTRACT

The systemic inflammatory response syndrome (SIRS) is a clinicopathological manifestation of overexuberant acute-phase inflammation caused by infectious or noninfectious etiologies. The systemic release of pro-inflammatory cytokines, chemokines, and lipid and vasoactive mediators induces endothelial damage and microvascular thrombosis, potentially culminating in disseminated intravascular coagulation (DIC), acute respiratory distress syndrome (ARDS), and multiple organ dysfunction (MOD) or failure (MOF). We present five cases in the pig-tailed macaque and olive baboon where SIRS resulted in MOF, ARDS, DIC, and the Waterhouse-Friderichsen syndrome; each with gross and histological elements manifested as edema, deposition of fibrin, hemorrhage, and thrombosis. In the described cases, SIRS was the end-common pathway for multiple risk factors that parallel those documented in humans: major surgery, obstetric complications, and infection. The diagnosis of SIRS should be considered when evaluating nonhuman primate (NHP) cases of MOF manifesting with histological evidence of vascular leakage. Experimental manipulation of NHP models may be complicated by SIRS and accompanying rapid clinical decompensation. Such adverse events may compromise toxicological studies and should be avoided when possible.


Subject(s)
Acute Lung Injury/veterinary , Disseminated Intravascular Coagulation/veterinary , Multiple Organ Failure/veterinary , Systemic Inflammatory Response Syndrome/veterinary , Acute Lung Injury/blood , Acute Lung Injury/pathology , Animals , Cytokines/metabolism , Disseminated Intravascular Coagulation/blood , Disseminated Intravascular Coagulation/pathology , Female , Macaca nemestrina , Male , Multiple Organ Failure/blood , Multiple Organ Failure/pathology , Papio anubis , Risk Factors , Systemic Inflammatory Response Syndrome/blood , Systemic Inflammatory Response Syndrome/pathology
17.
J Zoo Wildl Med ; 35(4): 525-9, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15732595

ABSTRACT

Three juvenile male Micronesian kingfishers (Halcyon cinnamomina cinnamomina) housed in the same enclosure presented with rapid weight gain and coelomic distension. Physical examination and radiography revealed marked enlargement of the ventriculus and a single, large foreign body within the ventriculus in each individual. Surgical removal by ventriculotomy was attempted in one individual, which died during the procedure. A second individual was treated with natural peanut butter 0.5 ml p.o. b.i.d. for 14 days and recovered, casting the foreign material. The third bird was similarly treated without success and subsequently died during attempts at endoscopic removal of the foreign body. In all three birds, the foreign bodies proved to be phytobezoars. The birds had been observed stripping leaf fragments from live corn plants (Dracaena fragrans) used in the enclosure. Plant fibers from the phytobezoars were compared with D. fragrans leaves and were considered identical. Medical treatment of phytobezoars with peanut butter or similar oil-containing substances in birds should be considered as an alternative to surgical extraction.


Subject(s)
Arachis , Bezoars/veterinary , Bird Diseases/therapy , Stomach, Avian , Animals , Arachis/chemistry , Bezoars/surgery , Bezoars/therapy , Bird Diseases/surgery , Birds , Fatal Outcome , Male , Peanut Oil , Plant Oils/therapeutic use , Stomach, Avian/surgery , Treatment Outcome , Weight Gain
18.
J Vet Diagn Invest ; 14(4): 326-8, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12152813

ABSTRACT

A 2.5-year-old captive female mandrill (Papio sphinx) died following a protracted course of intermittent abdominal bloat, diarrhea, and severe weight loss. Necropsy revealed emaciation and marked gastrointestinal distention with gas and ingesta. Histologic evaluation revealed severe diffuse granulomatous enterocolitis and mesenteric lymphadenitis with massive numbers of 1-2-microm acid-fast bacilli within macrophages. Additionally, there was moderate to severe multifocal myocardial and vascular amyloidosis, moderate multifocal pyogranulomatous interstitial pneumonia with no acid-fast bacteria, and moderate multifocal glossal candidiasis. Samples of feces, ileum, and colon were positive for Mycobacterium avium subsp. paratuberculosis by radiometric culture and a polymerase chain reaction-amplified DNA probe specific for the insertion sequence IS900 of this organism.


Subject(s)
Mycobacterium avium subsp. paratuberculosis/pathogenicity , Papio/microbiology , Paratuberculosis/diagnosis , Animals , Autopsy/veterinary , DNA, Bacterial/analysis , Diarrhea/etiology , Diarrhea/veterinary , Female , Mycobacterium avium subsp. paratuberculosis/genetics , Mycobacterium avium subsp. paratuberculosis/isolation & purification , Paratuberculosis/genetics , Paratuberculosis/pathology , Polymerase Chain Reaction/veterinary , Weight Loss
SELECTION OF CITATIONS
SEARCH DETAIL
...