Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
PLoS Pathog ; 18(4): e1009925, 2022 04.
Article in English | MEDLINE | ID: mdl-35443018

ABSTRACT

Early in the SARS-CoV-2 pandemic, there was a high level of optimism based on observational studies and small controlled trials that treating hospitalized patients with convalescent plasma from COVID-19 survivors (CCP) would be an important immunotherapy. However, as more data from controlled trials became available, the results became disappointing, with at best moderate evidence of efficacy when CCP with high titers of neutralizing antibodies was used early in infection. To better understand the potential therapeutic efficacy of CCP, and to further validate SARS-CoV-2 infection of macaques as a reliable animal model for testing such strategies, we inoculated 12 adult rhesus macaques with SARS-CoV-2 by intratracheal and intranasal routes. One day later, 8 animals were infused with pooled human CCP with a high titer of neutralizing antibodies (RVPN NT50 value of 3,003), while 4 control animals received normal human plasma. Animals were monitored for 7 days. Animals treated with CCP had detectable but low levels of antiviral antibodies after infusion. In comparison to the control animals, CCP-treated animals had similar levels of viral RNA in upper and lower respiratory tract secretions, similar detection of viral RNA in lung tissues by in situ hybridization, but lower amounts of infectious virus in the lungs. CCP-treated animals had a moderate, but statistically significant reduction in interstitial pneumonia, as measured by comprehensive lung histology. Thus overall, therapeutic benefits of CCP were marginal and inferior to results obtained earlier with monoclonal antibodies in this animal model. By highlighting strengths and weaknesses, data of this study can help to further optimize nonhuman primate models to provide proof-of-concept of intervention strategies, and guide the future use of convalescent plasma against SARS-CoV-2 and potentially other newly emerging respiratory viruses.


Subject(s)
COVID-19 , SARS-CoV-2 , Animals , Antibodies, Neutralizing , Antiviral Agents , COVID-19/therapy , Humans , Immunization, Passive , Macaca mulatta , RNA, Viral , COVID-19 Serotherapy
2.
PLoS Pathog ; 17(7): e1009688, 2021 07.
Article in English | MEDLINE | ID: mdl-34228761

ABSTRACT

There is an urgent need for effective therapeutic interventions against SARS-CoV-2, including new variants that continue to arise. Neutralizing monoclonal antibodies have shown promise in clinical studies. We investigated the therapeutic efficacy of a combination of two potent monoclonal antibodies, C135-LS and C144-LS that carry half-life extension mutations, in the rhesus macaque model of COVID-19. Twelve young adult macaques (three groups of four animals) were inoculated intranasally and intra-tracheally with a high dose of SARS-CoV-2 and 24 hours later, treated intravenously with a high (40 mg/kg) or low (12 mg/kg) dose of the C135-LS and C144-LS antibody combination, or a control monoclonal antibody. Animals were monitored for 7 days. Compared to the control animals, animals treated with either dose of the anti-SARS-CoV-2 antibodies showed similarly improved clinical scores, lower levels of virus replication in upper and lower respiratory tract, and significantly reduced interstitial pneumonia, as measured by comprehensive lung histology. In conclusion, this study provides proof-of-concept in support of further clinical development of these monoclonal antibodies against COVID-19 during early infection.


Subject(s)
Antibodies, Neutralizing/therapeutic use , Antibodies, Viral/therapeutic use , COVID-19/therapy , Lung/pathology , SARS-CoV-2/immunology , Virus Replication , Animals , Antibodies, Monoclonal/blood , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/therapeutic use , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Antibodies, Viral/blood , Antibodies, Viral/immunology , COVID-19/pathology , COVID-19/virology , Disease Models, Animal , Female , Lung/diagnostic imaging , Macaca mulatta , Male , Multivariate Analysis , Radiography , Respiratory System/virology , SARS-CoV-2/physiology , Time Factors , Treatment Outcome , Virus Replication/immunology
3.
J Virol ; 95(16): e0040321, 2021 07 26.
Article in English | MEDLINE | ID: mdl-34037419

ABSTRACT

To understand susceptibility of wild California sea lions and Northern elephant seals to influenza A virus (IAV), we developed an ex vivo respiratory explant model and used it to compare infection kinetics for multiple IAV subtypes. We first established the approach using explants from colonized rhesus macaques, a model for human IAV. Trachea, bronchi, and lungs from 11 California sea lions, 2 Northern elephant seals, and 10 rhesus macaques were inoculated within 24 h postmortem with 6 strains representing 4 IAV subtypes. Explants from the 3 species showed similar IAV infection kinetics, with peak viral titers 48 to 72 h post-inoculation that increased by 2 to 4 log10 PFU/explant relative to the inoculum. Immunohistochemistry localized IAV infection to apical epithelial cells. These results demonstrate that respiratory tissue explants from wild marine mammals support IAV infection. In the absence of the ability to perform experimental infections of marine mammals, this ex vivo culture of respiratory tissues mirrors the in vivo environment and serves as a tool to study IAV susceptibility, host range, and tissue tropism. IMPORTANCE Although influenza A virus can infect marine mammals, a dearth of marine mammal cell lines and ethical and logistical challenges prohibiting experimental infections of living marine mammals mean that little is known about IAV infection kinetics in these species. We circumvented these limitations by adapting a respiratory tract explant model first to establish the approach with rhesus macaques and then for use with explants from wild marine mammals euthanized for nonrespiratory medical conditions. We observed that multiple strains representing 4 IAV subtypes infected trachea, bronchi, and lungs of macaques and marine mammals with variable peak titers and kinetics. This ex vivo model can define infection dynamics for IAV in marine mammals. Further, use of explants from animals euthanized for other reasons reduces use of animals in research.


Subject(s)
Influenza A virus/physiology , Orthomyxoviridae Infections/virology , Respiratory Tract Infections/virology , Animals , Dogs , Host Specificity , Influenza A virus/classification , Kinetics , Macaca mulatta , Madin Darby Canine Kidney Cells , Models, Biological , Respiratory System/pathology , Respiratory System/virology , Sea Lions , Seals, Earless , Species Specificity , Viral Load , Viral Tropism
4.
Toxicol Pathol ; 50(5): 638-659, 2022 07.
Article in English | MEDLINE | ID: mdl-35363082

ABSTRACT

Rhesus and cynomolgus macaques are the most frequently used nonhuman primate (NHP) species for biomedical research and toxicology studies of novel therapeutics. In recent years, there has been a shortage of laboratory macaques due to a variety of competing factors. This was most recently exacerbated by the surge in NHP research required to address the severe acute respiratory syndrome (SARS)-coronavirus 2 pandemic. Continued support of these important studies has required the use of more varied cohorts of macaques, including animals with different origins, increased exposure to naturally occurring pathogens, and a wider age range. Diarrhea and diseases of the gastrointestinal tract are the most frequently occurring spontaneous findings in macaques of all origins and ages. The purpose of this review is to alert pathologists and scientists involved in NHP research to these findings and their impact on animal health and study endpoints, which may otherwise confound the interpretation of data generated using macaques.


Subject(s)
COVID-19 , Animals , Gastrointestinal Tract , Macaca fascicularis , Macaca mulatta
5.
J Med Primatol ; 49(1): 60-62, 2020 02.
Article in English | MEDLINE | ID: mdl-31742695

ABSTRACT

A pregnant female rhesus macaque died spontaneously during stage two labor. Gross and histopathologic findings included severe pulmonary edema, with low numbers of blood vessels containing pale basophilic mucinous material (Alcian Blue positive and PTAH negative), consistent with intravascular amniotic fluid-derived mucin resulting in pulmonary amniotic fluid embolism.


Subject(s)
Embolism, Amniotic Fluid/veterinary , Macaca mulatta , Monkey Diseases/pathology , Pregnancy Complications/veterinary , Animals , Embolism, Amniotic Fluid/pathology , Female , Pregnancy , Pregnancy Complications/pathology
6.
J Virol ; 88(13): 7412-25, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24741104

ABSTRACT

UNLABELLED: Influenza is the cause of significant morbidity and mortality in pediatric populations. The contribution of pulmonary host defense mechanisms to viral respiratory infection susceptibility in very young children is poorly understood. As a surrogate to compare mucosal immune responses of infant and adult lungs, rhesus monkey primary airway epithelial cell cultures were infected with pandemic influenza A/H1N1 virus in vitro. Virus replication, cytokine secretion, cell viability, and type I interferon (IFN) pathway PCR array profiles were evaluated for both infant and adult cultures. In comparison with adult cultures, infant cultures showed significantly increased levels of H1N1 replication, reduced alpha interferon (IFN-α) protein synthesis, and no difference in cell death following infection. Age-dependent differences in expression levels of multiple genes associated with the type I IFN pathway were observed in H1N1-infected cultures. To investigate the pulmonary and systemic responses to H1N1 infection in early life, infant monkeys were inoculated with H1N1 by upper airway administration. Animals were monitored for virus and parameters of inflammation over a 14-day period. High H1N1 titers were recovered from airways at day 1, with viral RNA remaining detectable until day 9 postinfection. Despite viral clearance, bronchiolitis and alveolitis persisted at day 14 postinfection; histopathological analysis revealed alveolar septal thickening and intermittent type II pneumocyte hyperplasia. Our overall findings are consistent with the known susceptibility of pediatric populations to respiratory virus infection and suggest that intrinsic developmental differences in airway epithelial cell immune function may contribute to the limited efficacy of host defense during early childhood. IMPORTANCE: To the best of our knowledge, this study represents the first report of intrinsic developmental differences in infant airway epithelial cells that may contribute to the increased susceptibility of the host to respiratory virus infections. Despite the global burden of influenza, there are currently no vaccine formulations approved for children <6 months of age. Given the challenges of conducting experimental studies involving pediatric patients, rhesus monkeys are an ideal laboratory animal model to investigate the maturation of pulmonary mucosal immune mechanisms during early life because they are most similar to those of humans with regard to postnatal maturation of the lung structure and the immune system. Thus, our findings are highly relevant to translational medicine, and these data may ultimately lead to novel approaches that enhance airway immunity in very young children.


Subject(s)
Epithelium/immunology , Immunity, Innate/immunology , Influenza A Virus, H1N1 Subtype/physiology , Lung/immunology , Orthomyxoviridae Infections/immunology , Respiratory System/immunology , Virus Replication/physiology , Animals , Animals, Newborn , Blotting, Western , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Immunoenzyme Techniques , Inflammation/immunology , Inflammation/virology , Interferons/genetics , Macaca mulatta , Orthomyxoviridae Infections/virology , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
7.
Immunohorizons ; 6(12): 851-863, 2022 12 01.
Article in English | MEDLINE | ID: mdl-36547390

ABSTRACT

The global spread of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and its associated coronavirus disease (COVID-19) has led to a pandemic of unprecedented scale. An intriguing feature of the infection is the minimal disease in most children, a demographic at higher risk for other respiratory viral diseases. To investigate age-dependent effects of SARS-CoV-2 pathogenesis, we inoculated two rhesus macaque monkey dam-infant pairs with SARS-CoV-2 and conducted virological and transcriptomic analyses of the respiratory tract and evaluated systemic cytokine and Ab responses. Viral RNA levels in all sampled mucosal secretions were comparable across dam-infant pairs in the respiratory tract. Despite comparable viral loads, adult macaques showed higher IL-6 in serum at day 1 postinfection whereas CXCL10 was induced in all animals. Both groups mounted neutralizing Ab responses, with infants showing a more rapid induction at day 7. Transcriptome analysis of tracheal airway cells isolated at day 14 postinfection revealed significant upregulation of multiple IFN-stimulated genes in infants compared with adults. In contrast, a profibrotic transcriptomic signature with genes associated with cilia structure and function, extracellular matrix composition and metabolism, coagulation, angiogenesis, and hypoxia was induced in adults compared with infants. Our study in rhesus macaque monkey dam-infant pairs suggests age-dependent differential airway responses to SARS-CoV-2 infection and describes a model that can be used to investigate SARS-CoV-2 pathogenesis between infants and adults.


Subject(s)
COVID-19 , Animals , Macaca mulatta , Lung/pathology , SARS-CoV-2 , Virus Replication
8.
Cell Rep ; 37(5): 109942, 2021 11 02.
Article in English | MEDLINE | ID: mdl-34706272

ABSTRACT

Anti-viral monoclonal antibody (mAb) treatments may provide immediate but short-term immunity from coronavirus disease 2019 (COVID-19) in high-risk populations, such as people with diabetes and the elderly; however, data on their efficacy in these populations are limited. We demonstrate that prophylactic mAb treatment blocks viral replication in both the upper and lower respiratory tracts in aged, type 2 diabetic rhesus macaques. mAb infusion dramatically curtails severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-mediated stimulation of interferon-induced chemokines and T cell activation, significantly reducing development of interstitial pneumonia. Furthermore, mAb infusion significantly dampens the greater than 3-fold increase in SARS-CoV-2-induced effector CD4 T cell influx into the cerebrospinal fluid. Our data show that neutralizing mAbs administered preventatively to high-risk populations may mitigate the adverse inflammatory consequences of SARS-CoV-2 exposure.


Subject(s)
Antibodies, Monoclonal/therapeutic use , COVID-19/prevention & control , SARS-CoV-2/immunology , Aging/immunology , Animals , COVID-19/cerebrospinal fluid , COVID-19/complications , COVID-19/immunology , Diabetes Complications/immunology , Diabetes Complications/virology , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/immunology , Female , Humans , Lymphocyte Activation , Macaca mulatta , Male , Neuritis/immunology , Neuritis/prevention & control , Pre-Exposure Prophylaxis , T-Lymphocytes/immunology , Virus Replication/immunology
9.
Front Immunol ; 12: 729845, 2021.
Article in English | MEDLINE | ID: mdl-34938283

ABSTRACT

Non-human primate (NHP) animal models are an integral part of the drug research and development process. For some biothreat pathogens, animal model challenge studies may offer the only possibility to evaluate medical countermeasure efficacy. A thorough understanding of host immune responses in such NHP models is therefore vital. However, applying antibody-based immune characterization techniques to NHP models requires extensive reagent development for species compatibility. In the case of studies involving high consequence pathogens, further optimization for use of inactivated samples may be required. Here, we describe the first optimized CO-Detection by indEXing (CODEX) multiplexed tissue imaging antibody panel for deep profiling of spatially resolved single-cell immune responses in rhesus macaques. This 21-marker panel is composed of a set of 18 antibodies that stratify major immune cell types along with a set three Ebola virus (EBOV)-specific antibodies. We validated these two sets of markers using immunohistochemistry and CODEX in fully inactivated Formalin-Fixed Paraffin-Embedded (FFPE) tissues from mock and EBOV challenged macaques respectively and provide an efficient framework for orthogonal validation of multiple antibody clones using CODEX multiplexed tissue imaging. We also provide the antibody clones and oligonucleotide tag sequences as a valuable resource for other researchers to recreate this reagent set for future studies of tissue immune responses to EBOV infection and other diseases.


Subject(s)
Antibodies, Viral/immunology , Ebolavirus/immunology , Hemorrhagic Fever, Ebola/immunology , Immunity , Immunohistochemistry/methods , Animals , Disease Models, Animal , Hemorrhagic Fever, Ebola/diagnostic imaging , Hemorrhagic Fever, Ebola/pathology , Hemorrhagic Fever, Ebola/virology , Leukocytes/immunology , Macaca mulatta , Microscopy, Fluorescence/methods , Single-Cell Analysis/methods
10.
Microbiol Spectr ; 9(3): e0139721, 2021 12 22.
Article in English | MEDLINE | ID: mdl-34817208

ABSTRACT

Human clinical studies investigating use of convalescent plasma (CP) for treatment of coronavirus disease 2019 (COVID-19) have produced conflicting results. Outcomes in these studies may vary at least partly due to different timing of CP administration relative to symptom onset. The mechanisms of action of CP include neutralizing antibodies but may extend beyond virus neutralization to include normalization of blood clotting and dampening of inflammation. Unresolved questions include the minimum therapeutic titer in the CP units or CP recipient as well as the optimal timing of administration. Here, we show that treatment of macaques with CP within 24 h of infection does not reduce viral shedding in nasal or lung secretions compared to controls and does not detectably improve any clinical endpoint. We also demonstrate that CP administration does not impact viral sequence diversity in vivo, although the selection of a viral sequence variant in both macaques receiving normal human plasma was suggestive of immune pressure. Our results suggest that CP, administered to medium titers, has limited efficacy, even when given very early after infection. Our findings also contribute information important for the continued development of the nonhuman primate model of COVID-19. These results should inform interpretation of clinical studies of CP in addition to providing insights useful for developing other passive immunotherapies and vaccine strategies. IMPORTANCE Antiviral treatment options for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) remain very limited. One treatment that was explored beginning early in the pandemic (and that is likely to be tested early in future pandemics) is plasma collected from people who have recovered from coronavirus disease 2019 (COVID-19), known as convalescent plasma (CP). We tested if CP reduces viral shedding or disease in a nonhuman primate model. Our results demonstrate that administration of CP 1 day after SARS-CoV-2 infection had no significant impact on viral loads, clinical disease, or sequence diversity, although treatment with normal human plasma resulted in selection of a specific viral variant. Our results demonstrate that passive immunization with CP, even during early infection, provided no significant benefit in a nonhuman primate model of SARS-CoV-2 infection.


Subject(s)
COVID-19/therapy , Immunization, Passive/methods , SARS-CoV-2 , Animals , Antibodies, Neutralizing/immunology , Antibodies, Viral/blood , Antiviral Agents/therapeutic use , COVID-19/immunology , Disease Models, Animal , Humans , Immunity , Lung/pathology , Macaca mulatta , Pandemics , Spike Glycoprotein, Coronavirus/immunology , Viral Load , Virus Replication
11.
bioRxiv ; 2021 Sep 01.
Article in English | MEDLINE | ID: mdl-34494025

ABSTRACT

Early in the SARS-CoV-2 pandemic, there was a high level of optimism based on observational studies and small controlled trials that treating hospitalized patients with convalescent plasma from COVID-19 survivors (CCP) would be an important immunotherapy. However, as more data from controlled trials became available, the results became disappointing, with at best moderate evidence of efficacy when CCP with high titers of neutralizing antibodies was used early in infection. To better understand the potential therapeutic efficacy of CCP, and to further validate SARS-CoV-2 infection of macaques as a reliable animal model for testing such strategies, we inoculated 12 adult rhesus macaques with SARS-CoV-2 by intratracheal and intranasal routes. One day later, 8 animals were infused with pooled human CCP with a high titer of neutralizing antibodies (RVPN NT 50 value of 3,003), while 4 control animals received normal human plasma. Animals were monitored for 7 days. Animals treated with CCP had detectable levels of antiviral antibodies after infusion. In comparison to the control animals, they had similar levels of virus replication in the upper and lower respiratory tract, but had significantly reduced interstitial pneumonia, as measured by comprehensive lung histology. By highlighting strengths and weaknesses, data of this study can help to further optimize nonhuman primate models to provide proof-of-concept of intervention strategies, and guide the future use of convalescent plasma against SARS-CoV-2 and potentially other newly emerging respiratory viruses. AUTHOR SUMMARY: The results of treating SARS-CoV-2 infected hospitalized patients with COVID-19 convalescent plasma (CCP), collected from survivors of natural infection, have been disappointing. The available data from various studies indicate at best moderate clinical benefits only when CCP with high titer of neutralizing antibodies was infused early in infection. The macaque model of SARS-CoV-2 infection can be useful to gain further insights in the value of CCP therapy. In this study, animals were infected with SARS-CoV-2 and the next day, were infused with pooled human convalescent plasma, selected to have a very high titer of neutralizing antibodies. While administration of CCP did not result in a detectable reduction in virus replication in the respiratory tract, it significantly reduced lung inflammation. These data, combined with the results of monoclonal antibody studies, emphasize the need to use products with high titers of neutralizing antibodies, and guide the future development of CCP-based therapies.

12.
J Immunol ; 181(10): 6850-8, 2008 Nov 15.
Article in English | MEDLINE | ID: mdl-18981104

ABSTRACT

Polysialic acid (polySia) is a large glycan with restricted expression, typically found attached to the protein scaffold neural cell adhesion molecule (NCAM). PolySia is best known for its proposed role in modulating neuronal development. Its presence and potential functions outside the nervous systems are essentially unexplored. Herein we show the expression of polySia on hematopoietic progenitor cells, and demonstrate a role for this glycan in immune response using both acute inflammatory and tumor models. Specifically, we found that human NK cells modulate expression of NCAM and the degree of polymerization of its polySia glycans according to activation state. This contrasts with the mouse, where polySia and NCAM expression are restricted to multipotent hematopoietic progenitors and cells developing along a myeloid lineage. Sialyltransferase 8Sia IV(-/-) mice, which lacked polySia expression in the immune compartment, demonstrated an increased contact hypersensitivity response and decreased control of tumor growth as compared with wild-type animals. This is the first demonstration of polySia expression and regulation on myeloid cells, and the results in animal models suggest a role for polySia in immune regulation.


Subject(s)
Hematopoietic Stem Cells/immunology , Killer Cells, Natural/immunology , Sialic Acids/immunology , Animals , Cell Differentiation/immunology , Dermatitis, Contact/immunology , Flow Cytometry , Hematopoietic Stem Cells/cytology , Humans , Immunoblotting , Immunoprecipitation , Mice , Multipotent Stem Cells/cytology , Multipotent Stem Cells/immunology , Neoplasms, Experimental/immunology , Neural Cell Adhesion Molecules/immunology , Neural Cell Adhesion Molecules/metabolism
13.
Comp Med ; 70(2): 183-189, 2020 04 01.
Article in English | MEDLINE | ID: mdl-32178759

ABSTRACT

Klebsiella pneumoniae is a gram-negative bacterium found in the environment and as a commensal in humans and animals. In humans, K. pneumoniae is one of the most serious nosocomial infections encountered currently and is characterized by liver abscesses, pneumonia, and bacteremia resulting in meningoencephalitis and endophthalmitis. K. pneumoniae in veterinary medicine is rarely reported in NHP, and recent literature describing this disease is sparse. In our colony of predominantly outdoor-housed rhesus macaques (Macaca mulatta), K. pneumoniae is cultured infrequently from healthy animals during routine screening and is even rarer in sick animals. This report summarizes the clinical and postmortem findings associated with this pathogen in 9 rhesus macaques and compares these results with the disease outcomes reported for humans. In these cases, K. pneumoniae infection was confirmed through culture or PCR testing or both. In our experience, when this bacterium does cause clinical signs, the disease is rapidly progressive and severe. At necropsy of NHP, the findings are strikingly similar to opportunistic Klebsiella-associated syndromes described in humans and include liver abscesses, meningoencephalitis, and endophthalmitis. In addition, many of the affected macaques had similar risk factors to humans that succumb to disease, thus perhaps indicating that rhesus macaques could be a viable model for investigating these syndromes.


Subject(s)
Klebsiella Infections/veterinary , Monkey Diseases/diagnosis , Pneumonia, Bacterial/veterinary , Animals , Anti-Bacterial Agents/therapeutic use , Female , Klebsiella Infections/diagnosis , Klebsiella Infections/drug therapy , Klebsiella Infections/microbiology , Klebsiella pneumoniae/isolation & purification , Macaca mulatta , Male , Monkey Diseases/drug therapy , Monkey Diseases/microbiology , Pneumonia, Bacterial/diagnosis , Pneumonia, Bacterial/drug therapy , Pneumonia, Bacterial/microbiology
14.
Comp Med ; 70(5): 358-367, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32753092

ABSTRACT

In humans, abnormal thickening of the left ventricle of the heart clinically defines hypertrophic cardiomyopathy (HCM), a common inherited cardiovascular disorder that can precede a sudden cardiac death event. The wide range of clinical presentations in HCM obscures genetic variants that may influence an individual's susceptibility to sudden cardiac death. Although exon sequencing of major sarcomere genes can be used to detect high-impact causal mutations, this strategy is successful in only half of patient cases. The incidence of left ventricular hypertrophy (LVH) in a managed research colony of rhesus macaques provides an excellent comparative model in which to explore the genomic etiology of severe HCM and sudden cardiac death. Because no rhesus HCM-associated mutations have been reported, we used a next-generation genotyping assay that targets 7 sarcomeric rhesus genes within 63 genomic sites that are orthologous to human genomic regions known to harbor HCM disease variants. Amplicon sequencing was performed on 52 macaques with confirmed LVH and 42 unrelated, unaffected animals representing both the Indian and Chinese rhesus macaque subspecies. Bias-reduced logistic regression uncovered a risk haplotype in the rhesus MYBPC3 gene, which is frequently disrupted in both human and feline HCM; this haplotype implicates an intronic variant strongly associated with disease in either homozygous or carrier form. Our results highlight that leveraging evolutionary genomic data provides a unique, practical strategy for minimizing population bias in complex disease studies.


Subject(s)
Cardiomyopathy, Hypertrophic , Carrier Proteins , Animals , Cardiomyopathy, Hypertrophic/genetics , Cardiomyopathy, Hypertrophic/veterinary , Carrier Proteins/genetics , Cats , Haplotypes , Humans , Macaca mulatta/genetics , Mutation
15.
Res Sq ; 2020 Aug 14.
Article in English | MEDLINE | ID: mdl-32818217

ABSTRACT

CD4 T follicular helper (T fh ) cells are important for the generation of durable and specific humoral protection against viral infections. The degree to which SARS-CoV-2 infection generates T fh cells and stimulates the germinal center response is an important question as we investigate vaccine options for the current pandemic. Here we report that SARS-CoV-2 infection resulted in transient accumulation of pro-inflammatory monocytes and proliferating T fh cells with a T h 1 profile in peripheral blood. CD4 helper cell responses were skewed predominantly toward a T h 1 response in blood, lung, and lymph nodes. We observed the generation of germinal center T fh cells specific for the SARS-CoV-2 spike (S) and nucleocapsid (N) proteins, and a corresponding early appearance of antiviral serum IgG antibodies. Our data suggest that a vaccine promoting T h 1-type T fh responses that target the S protein may lead to protective immunity.

16.
bioRxiv ; 2020 Jul 08.
Article in English | MEDLINE | ID: mdl-32676606

ABSTRACT

CD4 T follicular helper (T fh ) cells are important for the generation of long-lasting and specific humoral protection against viral infections. The degree to which SARS-CoV-2 infection generates T fh cells and stimulates the germinal center response is an important question as we investigate vaccine options for the current pandemic. Here we report that, following infection with SARS-CoV-2, adult rhesus macaques exhibited transient accumulation of activated, proliferating T fh cells in their peripheral blood on a transitory basis. The CD4 helper cell responses were skewed predominantly toward a T h 1 response in blood, lung, and lymph nodes, reflective of the interferon-rich cytokine environment following infection. We also observed the generation of germinal center T fh cells specific for the SARS-CoV-2 spike (S) and nucleocapsid (N) proteins, and a corresponding early appearance of antiviral serum IgG antibodies but delayed or absent IgA antibodies. Our data suggest that a vaccine promoting Th1-type Tfh responses that target the S protein may lead to protective immunity.

17.
J Med Microbiol ; 57(Pt 2): 164-170, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18201981

ABSTRACT

The Mycobacterium tuberculosis genome contains four copies of an operon called mce (mce1-4). Previously we reported that M. tuberculosis disrupted in the mce1 operon is more virulent than wild-type M. tuberculosis in mice. We generated single deletion mutants in mce3 (Deltamce3) and mce4 (Deltamce4) operons and a double deletion mutant (Deltamce3/4). Similar doubling times and growth characteristics were observed for all mutants and the wild-type (parent) M. tuberculosis H37Rv strain in culture and in macrophages. In addition, similar bacterial burdens were detected in organs from mice infected with Deltamce3 and the parent strain. However, the bacterial burdens of mice infected with Deltamce4 and Deltamce 3/4 were less than those of mice infected with the parent strain. The median survival times of mice infected with wild-type M. tuberculosis, Deltamce3, Deltamce4 and Deltamce3/4 were 40.5, 46, 58 and 62 weeks, respectively. Histopathological examination of lungs at 15 weeks post-infection showed that the extent of the lung lesions was less prominent in mice infected with Deltamce4 and Deltamce 3/4 mutants than in mice infected with the other two strains. These observations suggest that the mce3 and mce4 operons have a role distinct from that of mce1 for in vivo survival of M. tuberculosis.


Subject(s)
Bacterial Proteins/physiology , Mycobacterium Infections/microbiology , Mycobacterium tuberculosis/pathogenicity , Virulence Factors/physiology , Animals , Bacterial Proteins/genetics , Colony Count, Microbial , Gene Deletion , Liver/microbiology , Lung/microbiology , Lung/pathology , Macrophages/microbiology , Mice , Mice, Inbred C57BL , Mutagenesis, Insertional , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/growth & development , Operon , Spleen/microbiology , Survival Analysis , Virulence , Virulence Factors/genetics
18.
Toxicology ; 249(2-3): 123-9, 2008 Jul 30.
Article in English | MEDLINE | ID: mdl-18538461

ABSTRACT

Botulinum neurotoxins (BoNTs) are among the most potent biological toxins for humans. They are primarily produced by the gram-positive, anaerobic spore-forming bacterium, Clostridium botulinum. In bacterial cultures, secreted BoNTs are associated with non-toxic accessory proteins forming large complexes. Neurotoxin-associated proteins have been shown to play an important role in the oral toxicity of BoNTs by protecting them from degradation and digestion by gastric acid and enzymes. Most toxicity studies using BoNTs have been performed using highly purified toxin. In this study, the toxicities of purified and crude BoNT/A toxin preparations were compared. Protein components secreted into culture supernatants along with BoNT/A were identified by mass spectrometry and the contribution of extra proteins found in the soluble crude toxin extracts to the toxicity of BoNTs was determined in mouse models of oral and parenteral botulinum intoxication. Analysis of crude toxin composition permitted assessment of the impact of accessory proteins on the oral bioavailability of BoNT/A toxin in food matrices.


Subject(s)
Botulinum Toxins, Type A/isolation & purification , Botulinum Toxins, Type A/pharmacokinetics , Animals , Botulinum Toxins, Type A/chemistry , Dose-Response Relationship, Drug , Electrophoresis, Polyacrylamide Gel , Food-Drug Interactions , Injections, Intraperitoneal , Intubation, Gastrointestinal , Lethal Dose 50 , Mass Spectrometry , Mice
19.
Nat Commun ; 9(1): 2414, 2018 06 20.
Article in English | MEDLINE | ID: mdl-29925843

ABSTRACT

Zika virus (ZIKV) infection of pregnant women can cause fetal microcephaly and other neurologic defects. We describe the development of a non-human primate model to better understand fetal pathogenesis. To reliably induce fetal infection at defined times, four pregnant rhesus macaques are inoculated intravenously and intraamniotically with ZIKV at gestational day (GD) 41, 50, 64, or 90, corresponding to first and second trimester of gestation. The GD41-inoculated animal, experiencing fetal death 7 days later, has high virus levels in fetal and placental tissues, implicating ZIKV as cause of death. The other three fetuses are carried to near term and euthanized; while none display gross microcephaly, all show ZIKV RNA in many tissues, especially in the brain, which exhibits calcifications and reduced neural precursor cells. Given that this model consistently recapitulates neurologic defects of human congenital Zika syndrome, it is highly relevant to unravel determinants of fetal neuropathogenesis and to explore interventions.


Subject(s)
Disease Models, Animal , Fetal Diseases/pathology , Macaca mulatta , Nervous System Diseases/pathology , Pregnancy Complications, Infectious/pathology , Zika Virus Infection/pathology , Zika Virus/pathogenicity , Animals , Brain/pathology , Brain/virology , Female , Fetal Diseases/virology , Fetus/pathology , Fetus/virology , Humans , Male , Nervous System Diseases/virology , Pregnancy , Pregnancy Complications, Infectious/virology , RNA, Viral/isolation & purification , Zika Virus/genetics , Zika Virus/isolation & purification , Zika Virus Infection/virology
20.
J Med Microbiol ; 56(Pt 4): 454-458, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17374883

ABSTRACT

The emergence of drug-resistant Mycobacterium tuberculosis strains and the widespread occurrence of AIDS demand newer and more efficient control of tuberculosis. The protective efficacy of the current Mycobacterium bovis bacille Calmette-Guérin (BCG) vaccine is highly variable. Therefore, development of an effective new vaccine has gained momentum in recent years. Recently, several M. tuberculosis mutants were tested as potential vaccine candidates in the mouse model of tuberculosis. However, only some of these mutants were able to generate protection equivalent to that of BCG in mice. This study reports the vaccine potential of an attenuated 5'-adenosine phosphosulfate reductase mutant (DeltacysH) of M. tuberculosis. Immunization of mice with either BCG or DeltacysH followed by infection with the virulent M. tuberculosis Erdman strain demonstrated that DeltacysH can generate protection equivalent to that of the BCG vaccine.


Subject(s)
Mutation/genetics , Mycobacterium tuberculosis/immunology , Tuberculosis Vaccines/immunology , Tuberculosis, Pulmonary/prevention & control , Vaccines, Attenuated/immunology , Animals , Bacterial Proteins/genetics , Bacterial Proteins/immunology , Female , Genes, Bacterial/genetics , Lung/microbiology , Lung/pathology , Mice , Mice, Inbred C57BL , Mycobacterium tuberculosis/genetics , Spleen/microbiology , Spleen/pathology , Tuberculosis Vaccines/genetics , Tuberculosis, Pulmonary/immunology , Tuberculosis, Pulmonary/pathology , Vaccines, Attenuated/genetics
SELECTION OF CITATIONS
SEARCH DETAIL