Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
Toxicol Pathol ; 51(6): 375-389, 2023 08.
Article in English | MEDLINE | ID: mdl-38179962

ABSTRACT

Direct delivery of therapeutics to the central nervous system (CNS) greatly expands opportunities to treat neurological diseases but is technically challenging. This opinion outlines principal technical aspects of direct CNS delivery via intracerebroventricular (ICV) or intrathecal (IT) injection to common nonclinical test species (rodents, dogs, and nonhuman primates) and describes procedure-related clinical and histopathological effects that confound interpretation of test article-related effects. Direct dosing is by ICV injection in mice due to their small body size, while other species are dosed IT in the lumbar cistern. The most frequent procedure-related functional effects are transient absence of lower spinal reflexes after IT injection or death soon after ICV dosing. Common procedure-related microscopic findings in all species include leukocyte infiltrates in CNS meninges or perivascular (Virchow-Robin) spaces; nerve fiber degeneration in the spinal cord white matter (especially dorsal and lateral tracts compressed by dosing needles or indwelling catheters), spinal nerve roots, and sciatic nerve; meningeal fibrosis at or near IT injection sites; hemorrhage; and gliosis. Findings typically are minimal to occasionally mild. Findings tend to be more severe and/or have a higher incidence in the spinal cord segments and spinal nerve roots at or close to the site of administration.


Subject(s)
Oligonucleotides , Rodentia , Dogs , Mice , Animals , Central Nervous System/pathology , Spinal Cord/pathology , Nerve Degeneration/pathology , Primates
2.
Toxicol Pathol ; 51(7-8): 414-431, 2023 Oct.
Article in English | MEDLINE | ID: mdl-38380881

ABSTRACT

Biotherapeutic modalities such as cell therapies, gene therapies, nucleic acids, and proteins are increasingly investigated as disease-modifying treatments for severe and life-threatening neurodegenerative disorders. Such diverse bio-derived test articles are fraught with unique and often unpredictable biological consequences, while guidance regarding nonclinical experimental design, neuropathology evaluation, and interpretation is often limited. This paper summarizes key messages offered during a half-day continuing education course on toxicologic neuropathology of neuro-targeted biotherapeutics. Topics included fundamental neurobiology concepts, pharmacology, frequent toxicological findings, and their interpretation including adversity decisions. Covered biotherapeutic classes included cell therapies, gene editing and gene therapy vectors, nucleic acids, and proteins. If agents are administered directly into the central nervous system, initial screening using hematoxylin and eosin (H&E)-stained sections of currently recommended neural organs (brain [7 levels], spinal cord [3 levels], and sciatic nerve) may need to expand to include other components (e.g., more brain levels, ganglia, and/or additional nerves) and/or special neurohistological procedures to characterize possible neural effects (e.g., cell type-specific markers for reactive glial cells). Scientists who evaluate the safety of novel biologics will find this paper to be a practical reference for preclinical safety testing and risk assessment.


Subject(s)
Neuropathology , Nucleic Acids , Brain , Spinal Cord , Sciatic Nerve
3.
Proc Natl Acad Sci U S A ; 112(16): 5117-22, 2015 Apr 21.
Article in English | MEDLINE | ID: mdl-25848017

ABSTRACT

The TREX1 gene encodes a potent DNA exonuclease, and mutations in TREX1 cause a spectrum of lupus-like autoimmune diseases. Most lupus patients develop autoantibodies to double-stranded DNA (dsDNA), but the source of DNA antigen is unknown. The TREX1 D18N mutation causes a monogenic, cutaneous form of lupus called familial chilblain lupus, and the TREX1 D18N enzyme exhibits dysfunctional dsDNA-degrading activity, providing a link between dsDNA degradation and nucleic acid-mediated autoimmune disease. We determined the structure of the TREX1 D18N protein in complex with dsDNA, revealing how this exonuclease uses a novel DNA-unwinding mechanism to separate the polynucleotide strands for single-stranded DNA (ssDNA) loading into the active site. The TREX1 D18N dsDNA interactions coupled with catalytic deficiency explain how this mutant nuclease prevents dsDNA degradation. We tested the effects of TREX1 D18N in vivo by replacing the TREX1 WT gene in mice with the TREX1 D18N allele. The TREX1 D18N mice exhibit systemic inflammation, lymphoid hyperplasia, vasculitis, and kidney disease. The observed lupus-like inflammatory disease is associated with immune activation, production of autoantibodies to dsDNA, and deposition of immune complexes in the kidney. Thus, dysfunctional dsDNA degradation by TREX1 D18N induces disease in mice that recapitulates many characteristics of human lupus. Failure to clear DNA has long been linked to lupus in humans, and these data point to dsDNA as a key substrate for TREX1 and a major antigen source in mice with dysfunctional TREX1 enzyme.


Subject(s)
Chilblains/enzymology , Chilblains/genetics , DNA Damage , DNA/metabolism , Exodeoxyribonucleases/genetics , Inflammation/pathology , Lupus Erythematosus, Cutaneous/enzymology , Lupus Erythematosus, Cutaneous/genetics , Phosphoproteins/genetics , Alleles , Animals , Antibodies/immunology , Autoimmunity/immunology , Base Sequence , Chilblains/pathology , DNA/chemistry , DNA/genetics , Exodeoxyribonucleases/chemistry , Humans , Lupus Erythematosus, Cutaneous/pathology , Mice , Molecular Sequence Data , Mutation/genetics , Nucleic Acid Conformation , Phenotype , Phosphoproteins/chemistry , Protein Biosynthesis
4.
Mol Ther ; 20(11): 2098-110, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22990674

ABSTRACT

Recombinant adeno-associated virus (rAAV) vectors have gained an extensive record of safety and efficacy in animal models of human disease. Infrequent reports of genotoxicity have been limited to specific vectors associated with excess hepatocellular carcinomas (HCC) in mice. In order to understand potential mechanisms of genotoxicity, and identify patterns of insertion that could promote tumor formation, we compared a self-complementary AAV (scAAV) vector designed to promote insertional activation (scAAV-CBA-null) to a conventional scAAV-CMV-GFP vector. HCC-prone C3H/HeJ mice and severe combined immunodeficiency (SCID) mice were infected with vector plus secondary treatments including partial hepatectomy (HPX) and camptothecin (CPT) to determine the effects of cell cycling and DNA damage on tumor incidence. Infection with either vector led to a significant increase in HCC incidence in male C3H/HeJ mice. Partial HPX after infection reduced HCC incidence in the cytomegalovirus-green fluorescent protein (CMV-GFP)-infected mice, but not in the cognate chicken ß-actin (CBA)-null infected group. Tumors from CBA-null infected, hepatectomized mice were more likely to contain significant levels of vector DNA than tumors from the corresponding CMV-GFP-infected group. Most CBA-null vector insertions recovered from tumors were associated with known proto-oncogenes or tumor suppressors. Specific patterns of insertion suggested read-through transcription, enhancer effects, and disruption of tumor suppressors as likely mechanisms for genotoxicity.


Subject(s)
Carcinoma, Hepatocellular/virology , Dependovirus/genetics , Liver Neoplasms, Experimental/virology , Mutagenesis, Insertional , Virus Integration , Animals , Base Sequence , Camptothecin , Carcinoma, Hepatocellular/chemically induced , Carcinoma, Hepatocellular/genetics , DNA Damage , Dependovirus/physiology , Female , Fibroblast Growth Factor 10/genetics , Fibroblast Growth Factor 3/genetics , Genetic Vectors , Genome, Viral , Hepatectomy , Liver Neoplasms, Experimental/chemically induced , Liver Neoplasms, Experimental/genetics , Male , Mice , Mice, Inbred C3H , Mice, SCID , Molecular Sequence Data , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogenes , SOS1 Protein/genetics , Transcriptional Activation
5.
J Virol ; 84(21): 11515-22, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20739515

ABSTRACT

The type I alpha/beta interferons (IFN-α/ß) are known to play an important role in host defense against influenza A virus infection, but we have now discovered that the recently identified type III IFNs (IFN-λ) constitute the major response to intranasal infection with this virus. Type III IFNs were present at much higher levels than type I IFNs in the lungs of infected mice, and the enhanced susceptibility of STAT2-/- animals demonstrated that only signaling through the IFN-α/ß or IFN-λ pathways was sufficient to mediate protection. This finding offers a possible explanation for the similar levels of antiviral protection found in wild-type (WT) mice and in animals lacking a functional type I IFN receptor (IFNAR-/-) but also argues that our current understanding of type III IFN induction is incomplete. While murine IFN-λ production is thought to depend on signaling through the type I IFN receptor, we demonstrate that intranasal influenza A virus infection leads to the robust type III IFN induction in the lungs of both WT and IFNAR-/- mice. This is consistent with previous studies showing that IFNAR-mediated protection is redundant for mucosal influenza virus infection and with data showing that the type III IFN receptor is expressed primarily by epithelial cells. However, the overlapping effects of these two cytokine families are limited by their differential receptor expression, with a requirement for IFN-α/ß signaling in combating systemic disease.


Subject(s)
Cytokines/genetics , Interferons/genetics , Orthomyxoviridae Infections/immunology , Transcriptional Activation , Animals , Epithelial Cells/metabolism , Humans , Influenza A virus , Mice , Mice, Inbred BALB C , Mice, Knockout , Receptor, Interferon alpha-beta/deficiency
6.
Vaccine ; 36(26): 3842-3852, 2018 06 18.
Article in English | MEDLINE | ID: mdl-29779923

ABSTRACT

Human respiratory syncytial virus (RSV) is the leading cause of lower airway disease in infants worldwide and repeatedly infects immunocompetent individuals throughout life. Severe lower airway RSV infection during infancy can be life-threatening, but is also associated with important sequelae including development of asthma and recurrent wheezing in later childhood. The basis for the inadequate, short-lived adaptive immune response to RSV infection is poorly understood, but it is widely recognized that RSV actively antagonizes Type I interferon (IFN) production. In addition to the induction of the anti-viral state, IFN production during viral infection is critical for downstream development of robust, long-lived immunity. Based on the hypothesis that a vaccine that induced robust IFN production would be protective, we previously constructed a Newcastle disease virus-vectored vaccine that expresses the F glycoprotein of RSV (NDV-F) and demonstrated that vaccinated mice had reduced lung viral loads and an enhanced IFN-γ response after RSV challenge. Here we show that vaccination also protected cotton rats from RSV challenge and induced long-lived neutralizing antibody production, even in RSV immune animals. Finally, pulmonary eosinophilia induced by RSV infection of unvaccinated cotton rats was prevented by vaccination. Overall, these data demonstrate enhanced protective immunity to RSV F when this protein is presented in the context of an abortive NDV infection.


Subject(s)
Immunity, Humoral , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Virus Vaccines/immunology , Respiratory Syncytial Viruses/immunology , Animals , Disease Models, Animal , Female , Interferon-gamma/metabolism , Lung/virology , Respiratory Syncytial Virus Vaccines/administration & dosage , Respiratory Syncytial Virus Vaccines/isolation & purification , Respiratory Syncytial Viruses/isolation & purification , Sigmodontinae , Time Factors , Viral Load
7.
Comp Med ; 65(4): 315-26, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26310461

ABSTRACT

Infection with respiratory syncytial virus (RSV) generally presents as a mild, upper airway disease in human patients but may cause severe lower airway disease in the very young and very old. Progress toward understanding the mechanisms of RSV pathogenesis has been hampered by a lack of relevant rodent models. Mice, the species most commonly used in RSV research, are resistant to upper respiratory infection and do not recapitulate the pattern of virus spread in the human host. To address the need for better rodent models of RSV infection, we have characterized the acute and chronic pathology of RSV infection of a relatively permissive host, cotton rats (Sigmodon hispidus). We demonstrate that virus delivered to the upper airway results in widespread RSV replication in the ciliated respiratory epithelial cells of the nasal cavity and, to a lesser extent, of the lung. Although acute inflammation is relatively mild and rapidly eliminated after viral clearance, chronic, eosinophilic lung pathology persists. These data support the use of cotton rats as a robust rodent model of human RSV disease, including the association between RSV pneumonia and subsequent development of allergic asthma.


Subject(s)
Asthma/virology , Lung/virology , Pneumonia, Viral/virology , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Viruses/pathogenicity , Sigmodontinae/virology , Animals , Asthma/immunology , Asthma/pathology , Bronchiolitis/virology , Bronchoalveolar Lavage Fluid/virology , Disease Models, Animal , Inhalation Exposure , Lung/immunology , Lung/pathology , Nasal Mucosa/virology , Pneumonia, Viral/immunology , Pneumonia, Viral/pathology , Pulmonary Eosinophilia/virology , Respiratory Mucosa/virology , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Infections/pathology , Respiratory Syncytial Viruses/immunology , Time Factors , Virus Replication
8.
Comp Med ; 60(3): 225-32, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20579438

ABSTRACT

Although most viral infections of the upper respiratory tract can predispose to bacterial otitis media, human respiratory syncytial virus (HRSV) is the predominant viral copathogen of this highly prevalent pediatric polymicrobial disease. Rigorous study of the specific mechanisms by which HRSV predisposes to otitis media has been hindered by lack of a relevant animal model. We recently reported that the chinchilla, the preferred rodent host for studying otitis media, is semipermissive for upper-airway HRSV infection. In the current study, we defined the anatomy and kinetics of HRSV infection and spread in the upper airway of chinchilla hosts. Chinchillas were challenged intranasally with a fluorescent-protein-expressing HRSV. Upper-airway tissues were recovered at multiple time points after viral challenge and examined by confocal microscopy and immunohistochemistry. HRSV replication was observed from the rostral- to caudalmost regions of the nasal cavity as well as throughout the Eustachian tube in a time-dependent manner. Although fluorescence was not observed and virus was not detected in nasopharyngeal lavage fluids 14 d after infection, the latest time point examined in this study, occasional clusters of immunopositive cells were present, suggesting that the nasal cavity may serve as a reservoir for HRSV. These data provide important new information concerning the time course of HRSV infection of the uppermost airway and suggest that chinchillas may be useful for modeling the HRSV-induced changes that predispose to secondary bacterial infection.


Subject(s)
Chinchilla , Disease Models, Animal , Respiratory Syncytial Virus Infections , Respiratory Syncytial Virus, Human/pathogenicity , Respiratory System , Animals , Chinchilla/anatomy & histology , Chinchilla/virology , Female , Humans , Male , Mice , Nasal Cavity/anatomy & histology , Nasal Cavity/virology , Nasal Lavage Fluid/virology , Otitis Media/veterinary , Otitis Media/virology , Respiratory Syncytial Virus Infections/veterinary , Respiratory Syncytial Virus Infections/virology , Respiratory System/anatomy & histology , Respiratory System/virology , Viral Load
9.
J Med Primatol ; 37(6): 318-28, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18671769

ABSTRACT

BACKGROUND: Congenital transmission of Trypanosoma cruzi has been described in humans and experimental work has been conducted with mice, but not with non-human primates (NHPs). METHODS: We conducted a retrospective study of female baboons (Papio hamadryas spp.) naturally seropositive or seronegative for T. cruzi with history of fetal loss, and we report a stillbirth in a cynomolgus macaque (Macaca fascicularis) with placental T. cruzi amastigotes. RESULTS: There were no differences in menstrual cycle parameters and the number of fetal losses between seropositive and seronegative baboons with history of fetal loss. The amount of parasite DNA detected using quantitative polymerase chain reaction (Q-PCR) in M. fascicularis placenta was within the range detected in infected baboon tissues. CONCLUSIONS: There is no evidence that chronic maternal T. cruzi infection causes fetal loss in baboons. Q-PCR is a useful diagnostic tool to study archived NHP placentas.


Subject(s)
Ape Diseases/parasitology , Chagas Disease/veterinary , Papio , Stillbirth/veterinary , Trypanosoma cruzi/growth & development , Animals , Chagas Disease/parasitology , DNA, Protozoan/chemistry , DNA, Protozoan/genetics , Fatal Outcome , Female , Immunohistochemistry , Macaca fascicularis , Male , Placenta/parasitology , Polymerase Chain Reaction/veterinary , Pregnancy , Retrospective Studies
SELECTION OF CITATIONS
SEARCH DETAIL