Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Am J Pathol ; 193(12): 2031-2046, 2023 12.
Article in English | MEDLINE | ID: mdl-37689386

ABSTRACT

The pathophysiology of long-recognized hematologic abnormalities in Ebolavirus (EBOV) disease (EVD) is unknown. From limited human sampling (of peripheral blood), it has been postulated that emergency hematopoiesis plays a role in severe EVD, but the systematic characterization of the bone marrow (BM) has not occurred in human disease or in nonhuman primate models. In a lethal rhesus macaque model of EVD, 18 sternal BM samples exposed to the Kikwit strain of EBOV were compared to those from uninfected controls (n = 3). Immunohistochemistry, RNAscope in situ hybridization, transmission electron microscopy, and confocal microscopy showed that EBOV infects BM monocytes/macrophages and megakaryocytes. EBOV exposure was associated with severe BM hypocellularity, including depletion of myeloid, erythroid, and megakaryocyte hematopoietic cells. These depletions were negatively correlated with cell proliferation (Ki67 expression) and were not associated with BM apoptosis during disease progression. In EBOV-infected rhesus macaques with terminal disease, BM showed marked hemophagocytosis, megakaryocyte emperipolesis, and the release of immature hematopoietic cells into the sinusoids. Collectively, these data demonstrate not only direct EBOV infection of BM monocytes/macrophages and megakaryocytes but also that disease progression is associated with hematopoietic failure, notably in peripheral cytopenia. These findings inform current pathophysiologic unknowns and suggest a crucial role for BM dysfunction and/or failure, including emergency hematopoiesis, as part of the natural history of severe human disease.


Subject(s)
Ebolavirus , Hemorrhagic Fever, Ebola , Animals , Humans , Ebolavirus/physiology , Macaca mulatta , Bone Marrow , Disease Progression
2.
J Infect Dis ; 228(4): 371-382, 2023 08 16.
Article in English | MEDLINE | ID: mdl-37279544

ABSTRACT

BACKGROUND: Ebola virus (EBOV) disease (EVD) is one of the most severe and fatal viral hemorrhagic fevers and appears to mimic many clinical and laboratory manifestations of hemophagocytic lymphohistiocytosis syndrome (HLS), also known as macrophage activation syndrome. However, a clear association is yet to be firmly established for effective host-targeted, immunomodulatory therapeutic approaches to improve outcomes in patients with severe EVD. METHODS: Twenty-four rhesus monkeys were exposed intramuscularly to the EBOV Kikwit isolate and euthanized at prescheduled time points or when they reached the end-stage disease criteria. Three additional monkeys were mock-exposed and used as uninfected controls. RESULTS: EBOV-exposed monkeys presented with clinicopathologic features of HLS, including fever, multiple organomegaly, pancytopenia, hemophagocytosis, hyperfibrinogenemia with disseminated intravascular coagulation, hypertriglyceridemia, hypercytokinemia, increased concentrations of soluble CD163 and CD25 in serum, and the loss of activated natural killer cells. CONCLUSIONS: Our data suggest that EVD in the rhesus macaque model mimics pathophysiologic features of HLS/macrophage activation syndrome. Hence, regulating inflammation and immune function might provide an effective treatment for controlling the pathogenesis of acute EVD.


Subject(s)
Ebolavirus , Hemorrhagic Fever, Ebola , Lymphohistiocytosis, Hemophagocytic , Macrophage Activation Syndrome , Animals , Macrophage Activation Syndrome/therapy , Macaca mulatta
3.
Am J Pathol ; 192(1): 121-129, 2022 01.
Article in English | MEDLINE | ID: mdl-34626576

ABSTRACT

The pathogenesis of Ebola virus disease (EVD) is still incomplete, in spite of the availability of a nonhuman primate modelfor more than 4 decades. To further investigate EVD pathogenesis, a natural history study was conducted using 27 Chinese-origin rhesus macaques. Of these, 24 macaques were exposed intramuscularly to Kikwit Ebola virus and euthanized at predetermined time points or when end-stage clinical disease criteria were met, and 3 sham-exposed macaques were euthanized on study day 0. This study showed for the first time that Ebola virus causes uterine cervicitis, vaginitis, posthitis, and medullary adrenalitis. Not only was Ebola virus detected in the interstitial stromal cells of the genital tract, but it was also present in the epididymal and seminal vesicular tubular epithelial cells, ectocervical and vaginal squamous epithelial cells, and seminal fluid. Furthermore, as early as day 3 after exposure, Ebola virus replicative intermediate RNA was detected in Kupffer cells and hepatocytes. These findings in the nonhuman model provide additional insight into potential sexual transmission, possible disruption of sympathetic hormone production, and early virus replication sites in human EVD patients.


Subject(s)
Ebolavirus/physiology , Hormones/metabolism , Liver/virology , Tropism/physiology , Virus Replication/physiology , Animals , Chromaffin Cells/pathology , Chromaffin Cells/virology , Disease Models, Animal , Epididymis/pathology , Epididymis/virology , Epithelial Cells/pathology , Epithelial Cells/virology , Female , Hepatocytes/pathology , Hepatocytes/virology , Kupffer Cells/pathology , Kupffer Cells/virology , Macaca mulatta , Male , Uterine Cervicitis/pathology , Uterine Cervicitis/virology , Vaginitis/pathology , Vaginitis/virology
4.
Am J Pathol ; 190(9): 1867-1880, 2020 09.
Article in English | MEDLINE | ID: mdl-32479821

ABSTRACT

The most commonly reported symptom of post-Ebola virus disease syndrome in survivors is arthralgia, yet involvement of the joints in acute or convalescent Ebola virus infection is not well characterized in human patients or animal models. Through immunohistochemistry, we found that the lining synovial intima of the stifle (knee) is a target for acute infection by Ebola virus/Kikwit, Ebola virus/Makona-C05, and Marburg virus/Angola in the rhesus macaque model. Furthermore, histologic analysis, immunohistochemistry, RNAscope in situ hybridization, and transmission electron microscopy showed that synoviocytes of the stifle, shoulder, and hip are a target for mouse-adapted Ebola virus/Yambuku-Mayinga infection during acute disease in rhesus macaques. A time course of infection study with Ebola virus/Kikwit found that the large joint synovium became immunopositive beginning on postinfection day 6. In total, the synovium of 28 of 30 rhesus macaques with terminal filovirus disease had evidence of infection (64 of 96 joints examined). On the basis of immunofluorescence, infected cell types included CD68+ type A (macrophage-like) synoviocytes and CD44+ type B (fibroblast-like) synoviocytes. Cultured primary human fibroblast-like synoviocytes were permissive to infection with Ebola and Marburg viruses in vitro. Because synovial joints include immune privileged sites, these findings are significant for future investigations of filovirus pathogenesis and persistence as well as arthralgias in acute and convalescent filovirus disease.


Subject(s)
Filoviridae Infections/virology , Synoviocytes/virology , Animals , Cells, Cultured , Filoviridae , Humans , Macaca mulatta
5.
J Infect Dis ; 222(10): 1745-1755, 2020 10 13.
Article in English | MEDLINE | ID: mdl-32498080

ABSTRACT

Neurological signs and symptoms are the most common complications of Ebola virus disease. However, the mechanisms underlying the neurologic manifestations in Ebola patients are not known. In this study, peripheral ganglia were collected from 12 rhesus macaques that succumbed to Ebola virus (EBOV) disease from 5 to 8 days post exposure. Ganglionitis, characterized by neuronal degeneration, necrosis, and mononuclear leukocyte infiltrates, was observed in the dorsal root, autonomic, and enteric ganglia. By immunohistochemistry, RNAscope in situ hybridization, transmission electron microscopy, and confocal microscopy, we confirmed that CD68+ macrophages are the target cells for EBOV in affected ganglia. Further, we demonstrated that EBOV can induce satellite cell and neuronal apoptosis and microglial activation in infected ganglia. Our results demonstrate that EBOV can infect peripheral ganglia and results in ganglionopathy in rhesus macaques, which may contribute to the neurological signs and symptoms observed in acute and convalescent Ebola virus disease in human patients.


Subject(s)
Hemorrhagic Fever, Ebola/complications , Hemorrhagic Fever, Ebola/pathology , Nerve Degeneration/complications , Nerve Degeneration/pathology , Peripheral Nervous System Diseases/complications , Peripheral Nervous System Diseases/pathology , Animals , Antigens, CD , Antigens, Differentiation, Myelomonocytic , Disease Models, Animal , Ebolavirus , Female , Ganglia , Ganglia, Spinal/pathology , Ganglia, Spinal/virology , Ganglion Cysts/pathology , Hemorrhagic Fever, Ebola/virology , Humans , Immunohistochemistry , Leukocytes, Mononuclear , Macaca mulatta , Macrophages/pathology , Male , Microglia/pathology , Microglia/virology , Necrosis , Parasympathetic Nervous System/pathology , Peripheral Nervous System Diseases/virology , Sensory Receptor Cells/pathology , Sensory Receptor Cells/virology , Sympathetic Nervous System/pathology
6.
Am J Pathol ; 190(7): 1449-1460, 2020 07.
Article in English | MEDLINE | ID: mdl-32275904

ABSTRACT

Zaire ebolavirus (EBOV) causes Ebola virus disease (EVD), which carries a fatality rate between 25% and 90% in humans. Liver pathology is a hallmark of terminal EVD; however, little is known about temporal disease progression. We used multiplexed fluorescent immunohistochemistry and in situ hybridization in combination with whole slide imaging and image analysis (IA) to quantitatively characterize temporospatial signatures of viral and host factors as related to EBOV pathogenesis. Eighteen rhesus monkeys euthanized between 3 and 8 days post-infection, and 3 uninfected controls were enrolled in this study. Compared with semiquantitative histomorphologic ordinal scoring, quantitative IA detected subtle and progressive features of early and terminal EVD that was not feasible with routine approaches. Sinusoidal macrophages were the earliest cells to respond to infection, expressing proinflammatory cytokine interleukin 6 (IL6) mRNA, which was subsequently also observed in fibrovascular compartments. The mRNA of interferon-stimulated gene-15 (ISG-15), also known as ISG15 ubiquitin like modifier (ISG15), was observed early, with a progressive and ubiquitous hybridization signature involving mesenchymal and epithelial compartments. ISG-15 mRNA was prominent near infected cells, but not in infected cells, supporting the hypothesis that bystander cells produce a robust interferon gene response. This study contributes to our current understanding of early EVD progression and illustrates the value that digital pathology and quantitative IA serve in infectious disease research.


Subject(s)
Biomarkers/analysis , Hemorrhagic Fever, Ebola/pathology , Hemorrhagic Fever, Ebola/virology , Host-Pathogen Interactions/physiology , Liver/virology , Animals , Ebolavirus , Female , Hemorrhagic Fever, Ebola/immunology , Liver/immunology , Liver/pathology , Longitudinal Studies , Macaca mulatta , Male
SELECTION OF CITATIONS
SEARCH DETAIL
...