Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters











Publication year range
1.
Vet Pathol ; 61(4): 664-674, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38197423

ABSTRACT

NSG-SGM3 and NOG-EXL mice combine severe immunodeficiency with transgenic expression of human myeloid stimulatory cytokines, resulting in marked expansion of myeloid populations upon humanization with CD34+ hematopoietic stem cells (HSCs). Humanized NSG-SGM3 mice typically develop a lethal macrophage activation syndrome and mast cell hyperplasia that limit their use in long-term studies (e.g., humanization followed by tumor xenotransplantation). It is currently unclear to what extent humanized NOG-EXL mice suffer from the same condition observed in humanized NSG-SGM3 mice. We compared the effects of human CD34+ HSC engraftment in these two strains in an orthotopic patient-derived glioblastoma model. NSG-SGM3 mice humanized in-house were compared to NOG-EXL mice humanized in-house and commercially available humanized NOG-EXL mice. Mice were euthanized at humane or study endpoints, and complete pathological assessments were performed. A semiquantitative multiparametric clinicopathological scoring system was developed to characterize chimeric myeloid cell hyperactivation (MCH) syndrome. NSG-SGM3 mice were euthanized at 16 weeks after humanization because of severe deterioration of clinical conditions. Humanized NOG-EXL mice survived to the study endpoint at 22 weeks after humanization and showed less-severe MCH phenotypes than NSG-SGM3 mice. Major differences included the lack of mast cell expansion and limited tissue/organ involvement in NOG-EXL mice compared to NSG-SGM3 mice. Engraftment of human lymphocytes, assessed by immunohistochemistry, was similar in the two strains. The longer survival and decreased MCH phenotype severity in NOG-EXL mice enabled their use in a tumor xenotransplantation study. The NOG-EXL model is better suited than the NSG-SGM3 model for immuno-oncology studies requiring long-term survival after humanization.


Subject(s)
Antigens, CD34 , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells , Mice, Transgenic , Myeloid Cells , Animals , Mice , Humans , Hematopoietic Stem Cells/pathology , Antigens, CD34/metabolism , Myeloid Cells/pathology , Phenotype , Disease Models, Animal
2.
Mol Ther Methods Clin Dev ; 31: 101159, 2023 Dec 14.
Article in English | MEDLINE | ID: mdl-38094200

ABSTRACT

Adeno-associated viral (AAV) vectors have traditionally been viewed as predominantly nonintegrating, with limited concerns for oncogenesis. However, accumulating preclinical data have shown that AAV vectors integrate more often than previously appreciated, with the potential for genotoxicity. To understand the consequences of AAV vector integration, vigilance for rare genotoxic events after vector administration is essential. Here, we investigate the development of multicentric lymphoma in a privately owned dog, PC9, with severe hemophilia A that was treated with an AAV8 vector encapsidating a B domain-deleted canine coagulation F8 gene. PC9 developed an aggressive B cell lineage multicentric lymphoma 3.5 years after AAV treatment. Postmortem analysis of the liver, spleen, and lymph nodes showed the expected biodistribution of the AAV genome. Integration events were found both in PC9 and a second privately owned hemophilia A dog treated similarly with canine F8 gene transfer, which died of a bleeding event without evidence of malignancy. However, we found no evidence of expanded clones harboring a single integration event, indicating that AAV genome integrations were unlikely to have contributed to PC9's cancer. These findings suggest AAV integrations occur but are mostly not genotoxic and support the safety profile of AAV gene therapy.

3.
J Vet Intern Med ; 37(2): 735-739, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36821075

ABSTRACT

A 5-year-old imported Zangersheide gelding was evaluated for SC swellings over both forelimbs and lameness localized to the distal metacarpus. Ultrasound examination of the SC masses was compatible with verminous granulomas. Linear hyperechoic foci were present within the suspensory ligament branches of both forelimbs, suggestive of ligamentous parasitic infiltrates. A diagnosis of onchocerciasis was confirmed on biopsy of a SC mass. The gelding was treated with ivermectin and a tapering course of PO dexamethasone but was eventually euthanized. Necropsy confirmed the presence of SC eosinophilic granulomas and degenerative suspensory ligament desmitis, both with intralesional nematodes. Given the location and appearance of the nematode, a diagnosis of Onchocerca sp., most likely O. reticulata, was made. Onchocerciasis should be included as a differential diagnosis for multifocal suspensory ligament desmitis with these sonographic characteristics when paired with SC masses in imported European Warmbloods.


Subject(s)
Arthritis , Horse Diseases , Muscular Diseases , Onchocerciasis , Animals , Horses , Male , Onchocerca , Onchocerciasis/diagnosis , Onchocerciasis/parasitology , Onchocerciasis/pathology , Onchocerciasis/veterinary , Ligaments/pathology , Arthritis/veterinary , Muscular Diseases/pathology , Muscular Diseases/veterinary , Horse Diseases/diagnostic imaging , Horse Diseases/drug therapy
4.
Vet Pathol ; 60(3): 374-383, 2023 05.
Article in English | MEDLINE | ID: mdl-36727841

ABSTRACT

The spectrum of background, incidental, and experimentally induced lesions affecting NSG and NOG mice has been the subject of intense investigation. However, comprehensive studies focusing on the spontaneous neuropathological changes of these immunocompromised strains are lacking. This work describes the development of spontaneous early-onset neurodegeneration affecting both juvenile and adult NSG, NOG, and NXG mice. The study cohort consisted of 367 NSG mice of both sexes (including 33 NSG-SGM3), 61 NOG females (including 31 NOG-EXL), and 4 NXG females. These animals were primarily used for preclinical CAR T-cell testing, generation of humanized immune system chimeras, and/or tumor xenograft transplantation. Histopathology of brain and spinal cord and immunohistochemistry (IHC) for AIF-1, GFAP, CD34, and CD45 were performed. Neurodegenerative changes were observed in 57.6% of the examined mice (affected mice age range was 6-36 weeks). The lesions were characterized by foci of vacuolation with neuronal degeneration/death and gliosis distributed throughout the brainstem and spinal cord. IHC confirmed the development of gliosis, overexpression of CD34, and a neuroinflammatory component comprised of CD45-positive monocyte-derived macrophages. Lesions were significantly more frequent and severe in NOG mice. NSG males were considerably more affected than NSG females. Increased lesion frequency and severity in older animals were also identified. These findings suggest that NSG, NOG, and NXG mice are predisposed to the early development of identical neurodegenerative changes. While the cause of these lesions is currently unclear, potential associations with the genetic mutations shared by NSG, NOG, and NXG mice as well as unidentified viral infections are considered.


Subject(s)
Neoplasms , Male , Female , Mice , Animals , Gliosis/veterinary , Neoplasms/veterinary , Brain Stem , Spinal Cord , Mice, SCID
5.
6.
Sci Transl Med ; 12(525)2020 01 08.
Article in English | MEDLINE | ID: mdl-31915303

ABSTRACT

Maternal antibodies provide short-term protection to infants against many infections. However, they can inhibit de novo antibody responses in infants elicited by infections or vaccination, leading to increased long-term susceptibility to infectious diseases. Thus, there is a need to develop vaccines that are able to elicit protective immune responses in the presence of antigen-specific maternal antibodies. Here, we used a mouse model to demonstrate that influenza virus-specific maternal antibodies inhibited de novo antibody responses in mouse pups elicited by influenza virus infection or administration of conventional influenza vaccines. We found that a recently developed influenza vaccine, nucleoside-modified mRNA encapsulated in lipid nanoparticles (mRNA-LNP), partially overcame this inhibition by maternal antibodies. The mRNA-LNP influenza vaccine established long-lived germinal centers in the mouse pups and elicited stronger antibody responses than did a conventional influenza vaccine approved for use in humans. Vaccination with mRNA-LNP vaccines may offer a promising strategy for generating robust immune responses in infants in the presence of maternal antibodies.


Subject(s)
Antibody Formation/immunology , Nucleosides/metabolism , RNA, Messenger/immunology , Vaccination , Adjuvants, Immunologic/pharmacology , Animals , Antibodies, Viral/immunology , Disease Models, Animal , Female , Germinal Center , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Humans , Influenza Vaccines/immunology , Lipids/chemistry , Mice, Inbred BALB C , Mice, Inbred C57BL , Nanoparticles/chemistry , Orthomyxoviridae/immunology , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/prevention & control , Orthomyxoviridae Infections/virology
7.
J Exp Med ; 215(6): 1571-1588, 2018 06 04.
Article in English | MEDLINE | ID: mdl-29739835

ABSTRACT

T follicular helper (Tfh) cells are required to develop germinal center (GC) responses and drive immunoglobulin class switch, affinity maturation, and long-term B cell memory. In this study, we characterize a recently developed vaccine platform, nucleoside-modified, purified mRNA encapsulated in lipid nanoparticles (mRNA-LNPs), that induces high levels of Tfh and GC B cells. Intradermal vaccination with nucleoside-modified mRNA-LNPs encoding various viral surface antigens elicited polyfunctional, antigen-specific, CD4+ T cell responses and potent neutralizing antibody responses in mice and nonhuman primates. Importantly, the strong antigen-specific Tfh cell response and high numbers of GC B cells and plasma cells were associated with long-lived and high-affinity neutralizing antibodies and durable protection. Comparative studies demonstrated that nucleoside-modified mRNA-LNP vaccines outperformed adjuvanted protein and inactivated virus vaccines and pathogen infection. The incorporation of noninflammatory, modified nucleosides in the mRNA is required for the production of large amounts of antigen and for robust immune responses.


Subject(s)
B-Lymphocytes/immunology , Germinal Center/cytology , Nucleosides/metabolism , RNA, Messenger/metabolism , T-Lymphocytes, Helper-Inducer/immunology , Vaccines, Subunit/immunology , Adjuvants, Immunologic/pharmacology , Animals , Antibodies, Neutralizing/immunology , Antibody Formation/immunology , Antigens/metabolism , Lipids/chemistry , Macaca mulatta , Nanoparticles/chemistry , Protein Subunits/metabolism , Time Factors , Vaccination
8.
Virology ; 508: 1-6, 2017 08.
Article in English | MEDLINE | ID: mdl-28475924

ABSTRACT

Zika viruses (ZIKVs) are circulating in parts of the world endemic for other flavivirus infections. Some cross-reactive antibodies (Abs) elicited by prior flavivirus exposures can bind to ZIKV and enhance infection of Fc receptor-bearing cells. Here, we measured ZIKV binding of 54 murine monoclonal Abs (mAbs) elicited by exposure with Dengue virus and West Nile virus antigens. We found that 8 of 54 mAbs recognized the envelope protein of ZIKV in conventional binding assays. These 8 cross-reactive mAbs have different specificities; most recognize the DI/II region of the envelope protein but one mAb recognized the DIII lateral ridge of the envelope protein. Interestingly, only 3 of these cross-reactive mAbs were able to enhance ZIKV infection in vitro, and enhancing potential was not strictly correlated with relative binding ability. These data suggest that the ability of flavivirus Abs to enhance ZIKV is dependent on multiple factors.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Viral/immunology , Flavivirus/immunology , Zika Virus/immunology , Animals , Cross Reactions , Flavivirus Infections/immunology , Flavivirus Infections/virology , Humans , Mice , Viral Envelope Proteins/immunology , Zika Virus Infection/immunology , Zika Virus Infection/virology
9.
Nature ; 543(7644): 248-251, 2017 03 09.
Article in English | MEDLINE | ID: mdl-28151488

ABSTRACT

Zika virus (ZIKV) has recently emerged as a pandemic associated with severe neuropathology in newborns and adults. There are no ZIKV-specific treatments or preventatives. Therefore, the development of a safe and effective vaccine is a high priority. Messenger RNA (mRNA) has emerged as a versatile and highly effective platform to deliver vaccine antigens and therapeutic proteins. Here we demonstrate that a single low-dose intradermal immunization with lipid-nanoparticle-encapsulated nucleoside-modified mRNA (mRNA-LNP) encoding the pre-membrane and envelope glycoproteins of a strain from the ZIKV outbreak in 2013 elicited potent and durable neutralizing antibody responses in mice and non-human primates. Immunization with 30 µg of nucleoside-modified ZIKV mRNA-LNP protected mice against ZIKV challenges at 2 weeks or 5 months after vaccination, and a single dose of 50 µg was sufficient to protect non-human primates against a challenge at 5 weeks after vaccination. These data demonstrate that nucleoside-modified mRNA-LNP elicits rapid and durable protective immunity and therefore represents a new and promising vaccine candidate for the global fight against ZIKV.


Subject(s)
RNA, Messenger/administration & dosage , RNA, Messenger/chemistry , Viral Vaccines/immunology , Zika Virus Infection/prevention & control , Zika Virus/immunology , Animals , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Antigens, Viral/genetics , Antigens, Viral/immunology , Female , Glycoproteins/genetics , Glycoproteins/immunology , Injections, Intradermal , Macaca mulatta/immunology , Macaca mulatta/virology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Nanoparticles/administration & dosage , Nanoparticles/chemistry , RNA Stability , RNA, Messenger/genetics , RNA, Viral/administration & dosage , RNA, Viral/chemistry , RNA, Viral/genetics , Time Factors , Vaccination , Viral Envelope Proteins/genetics , Viral Envelope Proteins/immunology , Viral Vaccines/administration & dosage , Zika Virus/chemistry , Zika Virus/genetics , Zika Virus Infection/immunology
10.
Vaccine ; 34(46): 5483-5487, 2016 11 04.
Article in English | MEDLINE | ID: mdl-27670073

ABSTRACT

Influenza A viruses infect many species and cross-species transmission occurs occasionally. An equine H3N8 influenza virus began circulating in dogs in 1999 and an avian H3N2 influenza virus began circulating in dogs in 2006. The canine H3N8 (cH3N8) viral strain has become endemic in parts of the United States and there is a commercially available vaccine against this strain. The canine H3N2 (cH3N2) strain did not circulate widely in the United States until 2015. Here, we used a mouse model to determine if the cH3N8 and cH3N2 strains are antigenically related and if a commercially available cH3N8 vaccine protects animals against the cH3N2 outbreak strain. We find that the cH3N8 vaccine elicits antibodies that react to internal viral proteins and the hemagglutinin stalk region of cH3N2 viruses. These antibodies do not provide sterilizing immunity against cH3N2 infection, but these antibodies limit cH3N2 replication in the lung.


Subject(s)
Dog Diseases/prevention & control , Influenza A Virus, H3N2 Subtype/immunology , Influenza A Virus, H3N8 Subtype/immunology , Influenza Vaccines/immunology , Orthomyxoviridae Infections/veterinary , Animals , Antibodies, Viral/blood , Cross Protection , Disease Models, Animal , Dog Diseases/epidemiology , Dog Diseases/virology , Dogs , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Influenza A Virus, H3N2 Subtype/genetics , Influenza A Virus, H3N8 Subtype/genetics , Lung/virology , Mice , Orthomyxoviridae Infections/epidemiology , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/prevention & control , United States/epidemiology
11.
PLoS One ; 9(2): e88847, 2014.
Article in English | MEDLINE | ID: mdl-24551179

ABSTRACT

The specification of primordial germ cells (PGCs) and subsequent maintenance of germ-line identity in Drosophila embryos has long been thought to occur solely under the control of cell-autonomous factors deposited in the posterior pole plasm during oogenesis. However, here we document a novel role for somatic BMP signaling in the maintenance of PGC fate during the period leading up to embryonic gonad coalescence. We find that PGCs fail to maintain their germline identity when BMP signaling is compromised. They initiate but are unable to properly assemble the germline stem cell-specific organelle, the spectrosome, and they lose expression of the germline-specific gene Vasa. BMP signaling must, however, be finely tuned as there are deleterious consequences to PGCs when the pathway is excessively active. We show that one mechanism used to calibrate the effects of BMP signals is dependent on the Ubc9 homolog Lesswright (Lwr).


Subject(s)
Bone Morphogenetic Proteins/metabolism , Drosophila melanogaster/cytology , Drosophila melanogaster/embryology , Embryo, Nonmammalian/cytology , Germ Cells/cytology , Germ Cells/metabolism , Signal Transduction , Animals , Autocrine Communication , Cell Nucleus/metabolism , Down-Regulation , Drosophila Proteins/metabolism , Embryo, Nonmammalian/metabolism , Embryonic Development , Phosphorylation
SELECTION OF CITATIONS
SEARCH DETAIL