102 results on '"Luis D Giavedoni"'
Search Results
2. A prophylactic multivalent vaccine against different filovirus species is immunogenic and provides protection from lethal infections with Ebolavirus and Marburgvirus species in non-human primates.
- Author
-
Benoit Callendret, Jort Vellinga, Kerstin Wunderlich, Ariane Rodriguez, Robin Steigerwald, Ulrike Dirmeier, Cedric Cheminay, Ariane Volkmann, Trevor Brasel, Ricardo Carrion, Luis D Giavedoni, Jean L Patterson, Chad E Mire, Thomas W Geisbert, Jay W Hooper, Mo Weijtens, Jutta Hartkoorn-Pasma, Jerome Custers, Maria Grazia Pau, Hanneke Schuitemaker, and Roland Zahn
- Subjects
Medicine ,Science - Abstract
The search for a universal filovirus vaccine that provides protection against multiple filovirus species has been prompted by sporadic but highly lethal outbreaks of Ebolavirus and Marburgvirus infections. A good prophylactic vaccine should be able to provide protection to all known filovirus species and as an upside potentially protect from newly emerging virus strains. We investigated the immunogenicity and protection elicited by multivalent vaccines expressing glycoproteins (GP) from Ebola virus (EBOV), Sudan virus (SUDV), Taï Forest virus (TAFV) and Marburg virus (MARV). Immune responses against filovirus GP have been associated with protection from disease. The GP antigens were expressed by adenovirus serotypes 26 and 35 (Ad26 and Ad35) and modified Vaccinia virus Ankara (MVA) vectors, all selected for their strong immunogenicity and good safety profile. Using fully lethal NHP intramuscular challenge models, we assessed different vaccination regimens for immunogenicity and protection from filovirus disease. Heterologous multivalent Ad26-Ad35 prime-boost vaccination regimens could give full protection against MARV (range 75%-100% protection) and EBOV (range 50% to 100%) challenge, and partial protection (75%) against SUDV challenge. Heterologous multivalent Ad26-MVA prime-boost immunization gave full protection against EBOV challenge in a small cohort study. The use of such multivalent vaccines did not show overt immune interference in comparison with monovalent vaccines. Multivalent vaccines induced GP-specific antibody responses and cellular IFNγ responses to each GP expressed by the vaccine, and cross-reactivity to TAFV GP was detected in a trivalent vaccine expressing GP from EBOV, SUDV and MARV. In the EBOV challenge studies, higher humoral EBOV GP-specific immune responses (p = 0.0004) were associated with survival from EBOV challenge and less so for cellular immune responses (p = 0.0320). These results demonstrate that it is feasible to generate a multivalent filovirus vaccine that can protect against lethal infection by multiple members of the filovirus family.
- Published
- 2018
- Full Text
- View/download PDF
3. Metabolic Dysregulation in Hepacivirus Infection of Common Marmosets (Callithrix jacchus).
- Author
-
Cordelia Manickam, Lynn Wachtman, Amanda J Martinot, Luis D Giavedoni, and R Keith Reeves
- Subjects
Medicine ,Science - Abstract
Chronic hepatitis C has been associated with metabolic syndrome that includes insulin resistance, hepatic steatosis and obesity. These metabolic aberrations are risk factors for disease severity and treatment outcome in infected patients. Experimental infection of marmosets with GBV-B serves as a tangible, small animal model for human HCV infection, and while virology and pathology are well described, a full investigation of clinical disease and the metabolic milieu is lacking. In this study six marmosets were infected intravenously with GBV-B and changes in hematologic, serum biochemical and plasma metabolic measures were investigated over the duration of infection. Infected animals exhibited signs of lymphocytopenia, but platelet and RBC counts were generally stable or even increased. Although most animals showed a transient decline in blood glucose, infection resulted in several fold increases in plasma insulin, glucagon and glucagon-like peptide 1 (GLP-1). All infected animals experienced transient weight loss within the first 28 days of infection, but also became hypertriglyceridemic and had up to 10-fold increases in adipocytokines such as resistin and plasminogen activator inhibitor 1 (PAI-1). In liver, moderate to severe cytoplasmic changes associated with steatotic changes was observed microscopically at 168 days post infection. Collectively, these results suggest that GBV-B infection is accompanied by hematologic, biochemical and metabolic abnormalities that could lead to obesity, diabetes, thrombosis and atherosclerosis, even after virus has been cleared. Our findings mirror those found in HCV patients, suggesting that metabolic syndrome could be conserved among hepaciviruses, and both mechanistic and interventional studies for treating HCV-induced metabolic complications could be evaluated in this animal model.
- Published
- 2017
- Full Text
- View/download PDF
4. A Non-Human Primate Model of Severe Pneumococcal Pneumonia.
- Author
-
Luis F Reyes, Marcos I Restrepo, Cecilia A Hinojosa, Nilam J Soni, Anukul T Shenoy, Ryan P Gilley, Norberto Gonzalez-Juarbe, Julio R Noda, Vicki T Winter, Melissa A de la Garza, Robert E Shade, Jacqueline J Coalson, Luis D Giavedoni, Antonio Anzueto, and Carlos J Orihuela
- Subjects
Medicine ,Science - Abstract
RATIONALE:Streptococcus pneumoniae is the leading cause of community-acquired pneumonia and infectious death in adults worldwide. A non-human primate model is needed to study the molecular mechanisms that underlie the development of severe pneumonia, identify diagnostic tools, explore potential therapeutic targets, and test clinical interventions during pneumococcal pneumonia. OBJECTIVE:To develop a non-human primate model of pneumococcal pneumonia. METHODS:Seven adult baboons (Papio cynocephalus) were surgically tethered to a continuous monitoring system that recorded heart rate, temperature, and electrocardiography. Animals were inoculated with 109 colony-forming units of S. pneumoniae using bronchoscopy. Three baboons were rescued with intravenous ampicillin therapy. Pneumonia was diagnosed using lung ultrasonography and ex vivo confirmation by histopathology and immunodetection of pneumococcal capsule. Organ failure, using serum biomarkers and quantification of bacteremia, was assessed daily. RESULTS:Challenged animals developed signs and symptoms of pneumonia 4 days after infection. Infection was characterized by the presence of cough, tachypnea, dyspnea, tachycardia and fever. All animals developed leukocytosis and bacteremia 24 hours after infection. A severe inflammatory reaction was detected by elevation of serum cytokines, including Interleukin (IL)1Ra, IL-6, and IL-8, after infection. Lung ultrasonography precisely detected the lobes with pneumonia that were later confirmed by pathological analysis. Lung pathology positively correlated with disease severity. Antimicrobial therapy rapidly reversed symptomology and reduced serum cytokines. CONCLUSIONS:We have developed a novel animal model for severe pneumococcal pneumonia that mimics the clinical presentation, inflammatory response, and infection kinetics seen in humans. This is a novel model to test vaccines and treatments, measure biomarkers to diagnose pneumonia, and predict outcomes.
- Published
- 2016
- Full Text
- View/download PDF
5. Simian Immunodeficiency Virus Infection of Chimpanzees (Pan troglodytes) Shares Features of Both Pathogenic and Non-pathogenic Lentiviral Infections.
- Author
-
Edward J D Greenwood, Fabian Schmidt, Ivanela Kondova, Henk Niphuis, Vida L Hodara, Leah Clissold, Kirsten McLay, Bernadette Guerra, Sharon Redrobe, Luis D Giavedoni, Robert E Lanford, Krishna K Murthy, François Rouet, and Jonathan L Heeney
- Subjects
Immunologic diseases. Allergy ,RC581-607 ,Biology (General) ,QH301-705.5 - Abstract
The virus-host relationship in simian immunodeficiency virus (SIV) infected chimpanzees is thought to be different from that found in other SIV infected African primates. However, studies of captive SIVcpz infected chimpanzees are limited. Previously, the natural SIVcpz infection of one chimpanzee, and the experimental infection of six chimpanzees was reported, with limited follow-up. Here, we present a long-term study of these seven animals, with a retrospective re-examination of the early stages of infection. The only clinical signs consistent with AIDS or AIDS associated disease was thrombocytopenia in two cases, associated with the development of anti-platelet antibodies. However, compared to uninfected and HIV-1 infected animals, SIVcpz infected animals had significantly lower levels of peripheral blood CD4+ T-cells. Despite this, levels of T-cell activation in chronic infection were not significantly elevated. In addition, while plasma levels of β2 microglobulin, neopterin and soluble TNF-related apoptosis inducing ligand (sTRAIL) were elevated in acute infection, these markers returned to near-normal levels in chronic infection, reminiscent of immune activation patterns in 'natural host' species. Furthermore, plasma soluble CD14 was not elevated in chronic infection. However, examination of the secondary lymphoid environment revealed persistent changes to the lymphoid structure, including follicular hyperplasia in SIVcpz infected animals. In addition, both SIV and HIV-1 infected chimpanzees showed increased levels of deposition of collagen and increased levels of Mx1 expression in the T-cell zones of the lymph node. The outcome of SIVcpz infection of captive chimpanzees therefore shares features of both non-pathogenic and pathogenic lentivirus infections.
- Published
- 2015
- Full Text
- View/download PDF
6. Adaptation of SIVmac to baboon primary cells results in complete absence of in vivo baboon infectivity
- Author
-
Veronica Obregon-Perko, Amanda Mannino, Jason T. Ladner, Vida Hodara, Diako Ebrahimi, Laura Parodi, Jessica Callery, Gustavo Palacios, and Luis D. Giavedoni
- Subjects
viral adaptive changes and evolution ,simian immunodeficiency virus (SIV) ,baboon ,innate immunity ,chronic infection ,Microbiology ,QR1-502 - Abstract
While simian immunodeficiency virus (SIV) infection is non-pathogenic in naturally infected African nonhuman primate hosts, experimental or accidental infection in rhesus macaques often leads to AIDS. Baboons, widely distributed throughout Africa, do not naturally harbor SIV, and experimental infection of baboons with SIVmac results in transient low-level viral replication. Elucidation of mechanisms of natural immunity in baboons could uncover new targets of antiviral intervention. We tested the hypothesis that an SIVmac adapted to replicate in baboon primary cells will gain the capacity to establish chronic infections in vivo. Here, we generated SIVmac variants in baboon cells through serial passage in PBMC from different donors (SIVbn-PBMC s1), in PBMC from the same donors (SIVbn-PBMC s2), or in isolated CD4 cells from the same donors used for series 2 (SIVbn-CD4). While SIVbn-PBMC s1 and SIVbn-CD4 demonstrated increased replication capacity, SIVbn-PBMC s2 did not. Pharmacological blockade of CCR5 revealed SIVbn-PBMC s1 could more efficiently use available CCR5 than SIVmac, a trait we hypothesize arose to circumvent receptor occupation by chemokines. Sequencing analysis showed that all three viruses accumulated different types of mutations, and that more non-synonymous mutations became fixed in SIVbn-PBMC s1 than SIVbn-PBMC s2 and SIVbn-CD4, supporting the notion of stronger fitness pressure in PBMC from different genetic backgrounds. Testing the individual contribution of several newly fixed SIV mutations suggested that is the additive effect of these mutations in SIVbn-PBMC s1 that contributed to its enhanced fitness, as recombinant single mutant viruses showed no difference in replication capacity over the parental SIVmac239 strain. The replicative capacity of SIVbn-PBMC passage 4 (P4) s1 was tested in vivo by infecting baboons intravenously with SIVbn-PBMC P4 s1 or SIVmac251. While animals infected with SIVmac251 showed the known pattern of transient low-level viremia, animals infected with SIVbn-PBMC P4 s1 had undetectable viremia or viral DNA in lymphoid tissue. These studies suggest that adaptation of SIV to grow in baboon primary cells results in mutations that confer increased replicative capacity in the artificial environment of cell culture but make the virus unable to avoid the restrictive factors generated by a complex multicellular organism.
- Published
- 2024
- Full Text
- View/download PDF
7. Type I interferon upregulates Bak and contributes to T cell loss during human immunodeficiency virus (HIV) infection.
- Author
-
Joseph A Fraietta, Yvonne M Mueller, Guibin Yang, Alina C Boesteanu, Donald T Gracias, Duc H Do, Jennifer L Hope, Noshin Kathuria, Shannon E McGettigan, Mark G Lewis, Luis D Giavedoni, Jeffrey M Jacobson, and Peter D Katsikis
- Subjects
Immunologic diseases. Allergy ,RC581-607 ,Biology (General) ,QH301-705.5 - Abstract
The role of Type I interferon (IFN) during pathogenic HIV and SIV infections remains unclear, with conflicting observations suggesting protective versus immunopathological effects. We therefore examined the effect of IFNα/β on T cell death and viremia in HIV infection. Ex vivo analysis of eight pro- and anti-apoptotic molecules in chronic HIV-1 infection revealed that pro-apoptotic Bak was increased in CD4+ T cells and correlated directly with sensitivity to CD95/Fas-mediated apoptosis and inversely with CD4+ T cell counts. Apoptosis sensitivity and Bak expression were primarily increased in effector memory T cells. Knockdown of Bak by RNA interference inhibited CD95/Fas-induced death of T cells from HIV-1-infected individuals. In HIV-1-infected patients, IFNα-stimulated gene expression correlated positively with ex vivo T cell Bak levels, CD95/Fas-mediated apoptosis and viremia and negatively with CD4+ T cell counts. In vitro IFNα/β stimulation enhanced Bak expression, CD95/Fas expression and CD95/Fas-mediated apoptosis in healthy donor T cells and induced death of HIV-specific CD8+ T cells from HIV-1-infected patients. HIV-1 in vitro sensitized T cells to CD95/Fas-induced apoptosis and this was Toll-like receptor (TLR)7/9- and Type I IFN-dependent. This sensitization by HIV-1 was due to an indirect effect on T cells, as it occurred in peripheral blood mononuclear cell cultures but not purified CD4+ T cells. Finally, peak IFNα levels and viral loads correlated negatively during acute SIV infection suggesting a potential antiviral effect, but positively during chronic SIV infection indicating that either the virus drives IFNα production or IFNα may facilitate loss of viral control. The above findings indicate stage-specific opposing effects of Type I IFNs during HIV-1 infection and suggest a novel mechanism by which these cytokines contribute to T cell depletion, dysregulation of cellular immunity and disease progression.
- Published
- 2013
- Full Text
- View/download PDF
8. Urinary cytokine measurements do not reflect surgery‐induced inflammation in rhesus macaques
- Author
-
James P. Higham, Eve B. Cooper, Connor Whalen, Christiane Stahl‐Hennig, Luis D. Giavedoni, and Michael Heistermann
- Subjects
Animal Science and Zoology ,Ecology, Evolution, Behavior and Systematics - Published
- 2023
9. Molecular Approaches for the Validation of the Baboon as a Nonhuman Primate Model for the Study of Zika Virus Infection
- Author
-
Emma Mask, Vida L. Hodara, Jessica E. Callery, Laura M. Parodi, Veronica Obregon-Perko, Shigeo Yagi, Jeremy Glenn, Patrice Frost, Elizabeth Clemmons, Jean L. Patterson, Laura A. Cox, and Luis D. Giavedoni
- Subjects
Male ,Microbiology (medical) ,Immunity, Cellular ,Infectious Diseases ,Zika Virus Infection ,Immunology ,Animals ,Viremia ,Zika Virus ,Microbiology ,Papio - Abstract
Nonhuman primates (NHP) are particularly important for modeling infections with viruses that do not naturally replicate in rodent cells. Zika virus (ZIKV) has been responsible for sporadic epidemics, but in 2015 a disseminated outbreak of ZIKV resulted in the World Health Organization declaring it a global health emergency. Since the advent of this last epidemic, several NHP species, including the baboon, have been utilized for modeling and understanding the complications of ZIKV infection in humans; several health issues related to the outcome of infection have not been resolved yet and require further investigation. This study was designed to validate, in baboons, the molecular signatures that have previously been identified in ZIKV-infected humans and macaque models. We performed a comprehensive molecular analysis of baboons during acute ZIKV infection, including flow cytometry, cytokine, immunological, and transcriptomic analyses. We show here that, similar to most human cases, ZIKV infection of male baboons tends to be subclinical, but is associated with a rapid and transient antiviral interferon-based response signature that induces a detectable humoral and cell-mediated immune response. This immunity against the virus protects animals from challenge with a divergent ZIKV strain, as evidenced by undetectable viremia but clear anamnestic responses. These results provide additional support for the use of baboons as an alternative animal model to macaques and validate omic techniques that could help identify the molecular basis of complications associated with ZIKV infections in humans.
- Published
- 2022
10. Outlining key inflammation‐associated parameters during early phase of an experimental gram‐negative sepsis model in rhesus macaques (Macaca mulatta)
- Author
-
Nicole R Compo, Luis D. Giavedoni, Marcos J Ramos-Benitez, Jose J. Rosado‐Franco, Laura M. Parodi, Derick Rosario, and Ana M. Espino
- Subjects
Chemokine ,Medicine (General) ,Inflammation ,Gram negative sepsis ,chemokines ,Procalcitonin ,Sepsis ,sepsis ,03 medical and health sciences ,0302 clinical medicine ,R5-920 ,medicine ,030304 developmental biology ,0303 health sciences ,biology ,business.industry ,General Medicine ,medicine.disease ,biology.organism_classification ,Macaca mulatta ,cytokines ,Rhesus macaque ,030220 oncology & carcinogenesis ,Bacteremia ,Immunology ,biology.protein ,medicine.symptom ,Early phase ,business - Abstract
The aim of this study was to identify inflammation‐associated markers during the early phase of sepsis in rhesus macaque. Four rhesus macaques were given an intravenous dose of 1010 CFU/kg of E. coli. Blood samples were collected before, or 30 minutes, 2, 4, 6 and 8 hours after E. coli infusion. Physiological parameters, bacteremia, endotoxemia, C‐reactive protein (CRP), procalcitonin (PCT), and plasma cytokines/chemokines were determined for each animal. Bacteremia was present in all animals from 30 minutes to 3 hours after E. coli infusion whereas endotoxin was detected during the full‐time course. CRP and PCT levels remained at detectable levels during the whole experimental window suggesting an ongoing inflammatory process. Signature cytokines and chemokines such as TNF‐α, MIP‐1α, and MIP‐1β peaked about 2 hours after E. coli infusion and decreased thereafter. Plasma IL‐6, IL‐12p40, IFN‐γ, and IL‐1Ra, as well as I‐TAC, MIG, IP‐10 and MCP‐1, remained at detectable levels after 4 hours of E. coli infusion. This nonhuman primate model could be useful for the assessment of new therapeutics aiming to suppress key inflammatory markers throughout sepsis early phases.
- Published
- 2019
11. Marmoset Model for Mycobacterium avium Complex Pulmonary Disease
- Author
-
Ceci Hinojosa, Barbara A. Brown-Elliott, Luis D. Giavedoni, Diego J. Maselli, Edward D. Chan, Mandeep Mangat, Jay I. Peters, Jacqueline J. Coalson, and David E. Griffith
- Subjects
medicine.medical_specialty ,Pathology ,Lung ,Bronchiectasis ,medicine.diagnostic_test ,biology ,business.industry ,medicine.disease ,biology.organism_classification ,Callithrix ,Bronchoalveolar lavage ,medicine.anatomical_structure ,Immune system ,medicine ,Histopathology ,business ,Chest radiograph ,Pneumonitis - Abstract
RationaleMycobacterium avium complex, is the most common nontuberculous mycobacterial respiratory pathogen in humans. Disease mechanisms are poorly understood due to the absence of a reliable animal model for M. avium complex pulmonary disease.ObjectiveAssess the susceptibility, immunologic and histopathologic responses of the common marmoset (Callithrix jacchus) to M. avium complex pulmonary infection.Methods7 adult female marmosets underwent endobronchial inoculation with 108 colony-forming units of M. intracellulare and were monitored for 30 or 60 days. Prior to infection, chest radiograph and serum cytokines were assessed; serum cytokines were also monitored weekly for 30 days. At sacrifice 30 days (3 animals) or 60 days (4 animals) after infection, chest radiograph, serum and bronchoalveolar lavage cytokines, histopathology, and cultures of the bronchoalveolar lavage, lungs, liver, and kidney were analyzed.Measurements and Main ResultsFive of seven animals (two at 30 days and three at 60 days of infection) had positive lung cultures for M. intracellulare. Extra-pulmonary cultures were positive in three animals. All animals appeared healthy throughout the study. All five animals with positive lung cultures had radiographic changes consistent with pneumonitis. At 30 days, those with M. intracellulare lung infection showed granulomatous inflammation while at 60 days there was less inflammatory change, but bronchiectasis was noted. The cytokine response in the bronchoalveolar lavage fluid was uniformly greater in the animals with positive M. intracellulare cultures than those without a productive infection with greater levels at 30-days compared to 60-days. Similarly, serum cytokines were more elevated in the animals that had positive M. intracellulare cultures compared to those without a productive infection, peaking 14-21 days after inoculation.ConclusionEndobronchial instillation of M. intracellulare resulted in pulmonary mycobacterial infection in marmosets with a differential immune response, radiographic and histopathologic abnormalities, and an indolent course consistent with M. avium complex lung infection in humans.
- Published
- 2021
12. Multiplexed Simian Immunodeficiency Virus-Specific Paired RNA-Guided Cas9 Nickases Inactivate Proviral DNA
- Author
-
Jason T. Ladner, Jessica Callery, R. Alan Harris, Zhao Lai, Jeffrey Rogers, Yi Zou, Laura M. Parodi, Lisa M. Smith, Vida L. Hodara, Muthuswamy Raveedran, and Luis D. Giavedoni
- Subjects
viruses ,Immunology ,Biology ,medicine.disease_cause ,Microbiology ,chemistry.chemical_compound ,Plasmid ,Genome editing ,Proviruses ,Virology ,Vaccines and Antiviral Agents ,medicine ,CRISPR ,Animals ,Deoxyribonuclease I ,Humans ,Gene Editing ,Mutation ,DNA ,Simian immunodeficiency virus ,Provirus ,Endonucleases ,Macaca mulatta ,Long terminal repeat ,HEK293 Cells ,chemistry ,Mutagenesis ,Insect Science ,Simian Immunodeficiency Virus ,CRISPR-Cas Systems ,Plasmids ,RNA, Guide, Kinetoplastida - Abstract
Human and simian immunodeficiency virus (HIV and SIV) infections establish lifelong reservoirs of cells harboring an integrated proviral genome. Genome editing CRISPR-associated Cas9 nucleases, combined with SIV-specific guiding RNA (gRNA) molecules, inactivate integrated provirus DNA in vitro and in animal models. We generated RNA-guided Cas9 nucleases (RGNu) and nickases (RGNi) targeting conserved SIV regions with no homology in the human or rhesus macaque genome. Assays in cells cotransfected with SIV provirus and plasmids coding for RGNus identified SIV long terminal repeat (LTR), trans-activation response (TAR) element, and ribosome slip site (RSS) regions as the most effective at virus suppression; RGNi targeting these regions inhibited virus production significantly. Multiplex plasmids that coexpressed these three RGNu (Nu3), or six (three pairs) RGNi (Ni6), were more efficient at virus suppression than any combination of individual RGNu and RGNi plasmids. Both Nu3 and Ni6 plasmids were tested in lymphoid cells chronically infected with SIV(mac239), and whole-genome sequencing was used to determine on- and off-target mutations. Treatment with these all-in-one plasmids resulted in similar levels of mutations of viral sequences from the cellular genome; Nu3 induced indels at the 3 SIV-specific sites, whereas for Ni6 indels were present at the LTR and TAR sites. Levels of off-target effects detected by two different algorithms were indistinguishable from background mutations. In summary, we demonstrate that Cas9 nickase in association with gRNA pairs can specifically eliminate parts of the integrated provirus DNA; also, we show that careful design of an all-in-one plasmid coding for 3 gRNAs and Cas9 nuclease inhibits SIV production with undetectable off-target mutations, making these tools a desirable prospect for moving into animal studies. IMPORTANCE Our approach to HIV cure, utilizing the translatable SIV/rhesus macaque model system, aims at provirus inactivation and its removal with the least possible off-target side effects. We developed single molecules that delivered either three truncated SIV-specific gRNAs along with Cas9 nuclease or three pairs of SIV-specific gRNAs (six individual gRNAs) along with Cas9 nickase to enhance efficacy of on-target mutagenesis. Whole-genome sequencing demonstrated effective SIV sequence mutation and inactivation and the absence of demonstrable off-target mutations. These results open the possibility to employ Cas9 variants that introduce single-strand DNA breaks to eliminate integrated proviral DNA.
- Published
- 2021
13. Time elapsed between Zika and dengue virus infections affects antibody and T cell responses
- Author
-
Laura M. Parodi, Petraleigh Pantoja, Alexandra Ortiz-Rosa, Amelia K. Pinto, Crisanta Serrano-Collazo, Luis D. Giavedoni, Aravinda M. de Silva, Mariah Hassert, Teresa Arana, Laura J. White, Erick X. Pérez-Guzmán, James D. Brien, Daniela Weiskopf, Carlos A. Sariol, Melween I. Martínez, Idia V. Rodríguez, Lorna Cruz, and Vida L. Hodara
- Subjects
Male ,0301 basic medicine ,Time Factors ,T-Lymphocytes ,viruses ,General Physics and Astronomy ,Dengue virus ,Antibodies, Viral ,medicine.disease_cause ,Zika virus ,Dengue ,0302 clinical medicine ,ZikV Infection ,030212 general & internal medicine ,lcsh:Science ,Immunity, Cellular ,0303 health sciences ,Multidisciplinary ,Zika Virus Infection ,virus diseases ,Viral host response ,Cellular immunity ,3. Good health ,medicine.anatomical_structure ,Host-Pathogen Interactions ,Cytokines ,Antibody ,T cell ,Science ,030231 tropical medicine ,Viremia ,Cross Reactions ,Biology ,Article ,General Biochemistry, Genetics and Molecular Biology ,Cell Line ,03 medical and health sciences ,Immune system ,Immunity ,medicine ,Animals ,Humans ,030304 developmental biology ,Zika Virus ,General Chemistry ,Dengue Virus ,biochemical phenomena, metabolism, and nutrition ,Immune modulation ,biology.organism_classification ,medicine.disease ,Antibodies, Neutralizing ,Macaca mulatta ,Virology ,030104 developmental biology ,Viral infection ,biology.protein ,lcsh:Q - Abstract
Zika virus (ZIKV) and dengue virus (DENV) are co-endemic in many parts of the world, but the impact of ZIKV infection on subsequent DENV infection is not well understood. Here we show in rhesus macaques that the time elapsed after ZIKV infection affects the immune response to DENV infection. We show that previous ZIKV exposure increases the magnitude of the antibody and T cell responses against DENV. The time interval between ZIKV and subsequent DENV infection further affects the immune response. A mid-convalescent period of 10 months after ZIKV infection results in higher and more durable antibody and T cell responses to DENV infection than a short period of 2 months. In contrast, previous ZIKV infection does not affect DENV viremia or pro-inflammatory status. Collectively, we find no evidence of a detrimental effect of ZIKV immunity in a subsequent DENV infection. This supports the implementation of ZIKV vaccines that could also boost immunity against future DENV epidemics., Here, the authors show that the time elapsed between Zika infection and subsequent dengue virus infection affects the magnitude and durability of the antibody and cell-mediated immune responses against dengue virus, but not viremia. This research in non-human primates has implications for co-endemic regions and vaccination.
- Published
- 2019
14. Local immune responses to tuberculin skin challenge in Mycobacterium bovis BCG-vaccinated baboons: a pilot study of younger and older animals
- Author
-
Colwyn A. Headley, Tucker J. Piergallini, Olga Gonzalez, Joanne Turner, Vida L. Hodara, Nicole Reuter, Luis D. Giavedoni, James F. Papin, and Julia M. Scordo
- Subjects
lcsh:Immunologic diseases. Allergy ,0301 basic medicine ,Aging ,Immunology ,Short Report ,Tuberculin ,lcsh:Geriatrics ,Peripheral blood mononuclear cell ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Antigen ,biology.animal ,Medicine ,Vaccine immunity ,Tissue recall responses ,Mycobacterium bovis ,Tuberculin skin test ,biology ,business.industry ,biology.organism_classification ,Vaccination ,lcsh:RC952-954.6 ,030104 developmental biology ,biology.protein ,Antibody ,lcsh:RC581-607 ,business ,030217 neurology & neurosurgery ,Baboon - Abstract
Individuals over the age of 65 are highly susceptible to infectious diseases, which account for one-third of deaths in this age group. Vaccines are a primary tool to combat infection, yet they are less effective in the elderly population. While many groups have aimed to address this problem by studying vaccine-induced peripheral blood responses in the elderly, work from our lab and others demonstrate that immune responses to vaccination and infectious challenge may differ between tissue sites and the periphery. In this pilot study, we established an in vivo delayed-type hypersensitivity model of Mycobacterium bovis BCG vaccination and tuberculin skin test (TST) in two adult and two aged baboons. Vaccination generates BCG-specific immune cells that are recruited to the skin upon tuberculin challenge. We tested short term recall responses (8 weeks post-vaccination) and long term recall responses (25 weeks post-vaccination) by performing skin punch biopsies around the site of tuberculin injection. In short term recall responses, we found increased oxidation and decreased production of immune proteins in aged baboon skin at the site of TST challenge, in comparison to adult skin. Differences between adult and aged animals normalized in the long term response to tuberculin. In vitro, aged peripheral blood mononuclear cells had increased migration and functional responses to antigen-specific stimulation, suggesting that age-related changes in the tissue in vivo impairs aged immune recall responses to antigenic challenge. These findings highlight the impact of age-associated changes in the local tissue environment in memory recall responses, which may be more broadly applied to the study of other tissues. Moreover, these findings should be considered in future studies aimed at understanding and improving aging immune responses to vaccination and tissue challenge.
- Published
- 2021
15. Responses to acute infection with SARS-CoV-2 in the lungs of rhesus macaques, baboons and marmosets
- Author
-
Justin Ferdin, Vida L. Hodara, Dhiraj Kumar Singh, Shalini Gautam, Shabaana A. Khader, Shannan Hall-Ursone, Shashank Ganatra, Yenny Goez-Gazi, Tae-Hyung Lee, Benjamin Klaffke, Riti Sharan, Elizabeth Clemmons, Cynthia Alvarez, Olga Gonzalez, Christopher H. Chen, Kathleen M. Brasky, Laura M. Parodi, Kendra J. Alfson, Rajesh Thippeshappa, Alyssa Schami, Joanne Turner, Richard Copin, Andra K. Voges, Angélica Olmo-Fontánez, Makedonka Mitreva, Michal Gazi, Amanda Mannino, Patrice A. Frost, Ruby Escobedo, Alyssa Blakley, Deepak Kaushal, Edward J. Dick, Julia M. Scordo, Dharani Ajithdoss, John Dutton, Journey Cole, Hilary M. Staples, Christos A. Kyratsous, Priscilla Escareno, Luis D. Giavedoni, Ricardo Carrion, Bruce A. Rosa, Maya Gough, Luis Martinez-Sobrido, Mushtaq Ahmed, Bindu Singh, Roy N. Platt, Cory R. A. Hallam, Ken Sayers, Jessica Callery, Jordi B. Torrelles, Adelekan Oyejide, Jean L. Patterson, Andreu G. Vilanova, Carmen Bartley, Tim J. Anderson, Xavier Alvarez, Anna Goodroe, Larry S. Schlesinger, Corinna N. Ross, Alina Baum, and Ayan Chatterjee
- Subjects
Male ,Microbiology (medical) ,viruses ,Immunology ,Adaptive Immunity ,Antibodies, Viral ,Macaque ,Bronchoalveolar Lavage ,Applied Microbiology and Biotechnology ,Microbiology ,03 medical and health sciences ,Immune system ,biology.animal ,medicine ,Genetics ,Animals ,Humans ,Myeloid Cells ,Viral shedding ,Lung ,030304 developmental biology ,Pneumonitis ,Inflammation ,0303 health sciences ,Immunity, Cellular ,biology ,medicine.diagnostic_test ,030306 microbiology ,business.industry ,SARS-CoV-2 ,Monkey Diseases ,COVID-19 ,Callithrix ,Cell Biology ,Viral Load ,medicine.disease ,Acquired immune system ,Macaca mulatta ,Virus Shedding ,Bronchoalveolar lavage ,Immunoglobulin G ,Female ,business ,Viral load ,Bronchoalveolar Lavage Fluid ,Baboon ,Papio - Abstract
Non-human primate models will expedite therapeutics and vaccines for coronavirus disease 2019 (COVID-19) to clinical trials. Here, we compare acute severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection in young and old rhesus macaques, baboons and old marmosets. Macaques had clinical signs of viral infection, mild to moderate pneumonitis and extra-pulmonary pathologies, and both age groups recovered in two weeks. Baboons had prolonged viral RNA shedding and substantially more lung inflammation compared with macaques. Inflammation in bronchoalveolar lavage was increased in old versus young baboons. Using techniques including computed tomography imaging, immunophenotyping, and alveolar/peripheral cytokine response and immunohistochemical analyses, we delineated cellular immune responses to SARS-CoV-2 infection in macaque and baboon lungs, including innate and adaptive immune cells and a prominent type-I interferon response. Macaques developed T-cell memory phenotypes/responses and bystander cytokine production. Old macaques had lower titres of SARS-CoV-2-specific IgG antibody levels compared with young macaques. Acute respiratory distress in macaques and baboons recapitulates the progression of COVID-19 in humans, making them suitable as models to test vaccines and therapies.
- Published
- 2020
- Full Text
- View/download PDF
16. Non-human primate to human immunobridging to infer the protective effect of an Ebola virus vaccine candidate
- Author
-
Bart Spiessens, Ramon Roozendaal, Liesbeth Dekking, Maria Grazia Pau, Cynthia Robinson, Dominika N. Czapska-Casey, Yvonne Wollmann, Thierry Van Effelterre, Viki Bockstal, Eline Dekeyster, Roland Zahn, Jeroen N Stoop, Kerstin Luhn, Jan Serroyen, Luis D. Giavedoni, Jenny Hendriks, Sarah Janssen, Maarten Leyssen, Ben Van Baelen, Hanneke Schuitemaker, Ricardo Carrion, Daniel Splinter, An Vandebosch, Jerry Sadoff, Ariane Volkmann, Nadia Verbruggen, Macaya Douoguih, Laura Solforosi, and Benoit Callendret
- Subjects
0301 basic medicine ,Immunology ,Disease ,medicine.disease_cause ,Article ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Vaccine Immunogenicity ,Medicine ,Pharmacology (medical) ,030212 general & internal medicine ,RC254-282 ,Pharmacology ,Vaccines ,Ebola virus ,biology ,business.industry ,Immunogenicity ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC581-607 ,Virology ,Nonhuman primate ,3. Good health ,Regimen ,030104 developmental biology ,Infectious Diseases ,biology.protein ,Immunologic diseases. Allergy ,Antibody ,business - Abstract
It has been proven challenging to conduct traditional efficacy trials for Ebola virus (EBOV) vaccines. In the absence of efficacy data, immunobridging is an approach to infer the likelihood of a vaccine protective effect, by translating vaccine immunogenicity in humans to a protective effect, using the relationship between vaccine immunogenicity and the desired outcome in a suitable animal model. We here propose to infer the protective effect of the Ad26.ZEBOV, MVA-BN-Filo vaccine regimen with an 8-week interval in humans by immunobridging. Immunogenicity and protective efficacy data were obtained for Ad26.ZEBOV and MVA-BN-Filo vaccine regimens using a fully lethal EBOV Kikwit challenge model in cynomolgus monkeys (nonhuman primates [NHP]). The association between EBOV neutralizing antibodies, glycoprotein (GP)-binding antibodies, and GP-reactive T cells and survival in NHP was assessed by logistic regression analysis. Binding antibodies against the EBOV surface GP were identified as the immune parameter with the strongest correlation to survival post EBOV challenge, and used to infer the predicted protective effect of the vaccine in humans using published data from phase I studies. The human vaccine-elicited EBOV GP-binding antibody levels are in a range associated with significant protection against mortality in NHP. Based on this immunobridging analysis, the EBOV GP-specific-binding antibody levels elicited by the Ad26.ZEBOV, MVA-BN-Filo vaccine regimen in humans will likely provide protection against EBOV disease.
- Published
- 2020
17. Age Impacts the Local Immune Response to Tuberculin Skin Challenge in Mycobacterium Bovis BCG-vaccinated Baboons
- Author
-
Luis D. Giavedoni, James F. Papin, Tucker J. Piergallini, Vida L. Hodara, Olga Gonzalez, Joanne Turner, Colwyn A. Headley, Nicole Reuter, and Julia M. Scordo
- Subjects
Mycobacterium bovis ,Immune system ,biology ,business.industry ,Immunology ,Medicine ,Tuberculin ,business ,biology.organism_classification - Abstract
Background - Individuals over the age of 65 are highly susceptible to infectious diseases, which account for one-third of deaths in this age group. Vaccines are a primary tool to combat infection, yet they are less effective in the elderly population. While many groups have aimed to address this problem by studying vaccine-induced peripheral blood responses in the elderly, work from our lab and others demonstrate that immune responses to vaccination and infectious challenge may differ between tissue sites and the periphery. To improve health outcomes in our aged population, we must study vaccine responses in the tissue. Here we established an in vivo delayed-type hypersensitivity model of Mycobacterium bovis BCG vaccination and tuberculin skin test (TST) in adult and aged baboons. Vaccination generates BCG-specific immune cells that are recruited to the skin upon tuberculin challenge. We tested short-term recall responses (8 weeks post-vaccination) and long term recall responses (25 weeks post-vaccination) by performing skin punch biopsies around the site of tuberculin injection. In parallel, we determined BCG-induced responses in the peripheral blood of vaccinated animals. Results - In short term recall responses, we found increased oxidation and decreased production of immune proteins in aged baboon skin at the site of TST challenge, in comparison to adult skin. Differences between adult and aged animals normalized in the long term response to tuberculin. Phenotypic analysis of aged peripheral blood cells found several age-related changes in immune cell populations, independent of BCG vaccination, and no impairment in functional responses. Moreover, aged peripheral blood mononuclear cells had increased migration in vitro, suggesting that age-related changes in the tissue in vivo impairs aged immune recall responses to antigenic challenge. Conclusions - These findings highlight the impact of age-associated changes in the local tissue environment in memory recall responses, which may be more broadly applied to the study of other tissues. Moreover, these findings should be considered in future studies aimed at understanding and improving aging immune responses to vaccination and tissue challenge.
- Published
- 2020
18. Lethality of SARS-CoV-2 infection in K18 human angiotensin-converting enzyme 2 transgenic mice
- Author
-
Andreu Garcia-Vilanova, Corbett Christie, Edward J. Dick, Jordi B. Torrelles, Hilary M. Staples, Kendra J. Alfson, Luis D. Giavedoni, Jun-Gyu Park, Tim J. Anderson, Kevin Chiem, Olga Gonzalez, Juan Ignacio García, Larry S. Schlesinger, Laura M. Parodi, Jean L. Patterson, Varun Dwivedi, Deepak Kaushal, Shannan Hall-Ursone, Colin Chuba, Ricardo Carrion, Chengjin Ye, Katrina N. Kavelish, Angélica Olmo-Fontánez, Stephanie Earley, Luis Martinez-Sobrido, Xavier Alvarez, Cory R. A. Hallam, Stephanie Davis Mdaki, Anna Allué-Guardia, Roy N. Platt, Renee Escalona, Paula A. Pino, Alyssa Schami, Colwyn A. Headley, Michal Gazi, Jesse Martinez, Shalini Gautam, Oscar H. Rodriguez, Fatai S. Oladunni, Joanne Turner, Alison Whigham, and Anwari Akhter
- Subjects
0301 basic medicine ,Genetically modified mouse ,Chemokine ,Science ,Viral pathogenesis ,Transgene ,General Physics and Astronomy ,Mice, Transgenic ,Respiratory Mucosa ,macromolecular substances ,Article ,General Biochemistry, Genetics and Molecular Biology ,Virus ,Pathogenesis ,Mice ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Animals ,Genetic Predisposition to Disease ,Mortality ,Promoter Regions, Genetic ,Lung ,Multidisciplinary ,Keratin-18 ,biology ,SARS-CoV-2 ,Brain ,COVID-19 ,General Chemistry ,respiratory system ,medicine.disease ,Virology ,respiratory tract diseases ,Disease Models, Animal ,030104 developmental biology ,Virus Diseases ,Viral infection ,Angiotensin-converting enzyme 2 ,biology.protein ,Angiotensin-Converting Enzyme 2 ,Disease Susceptibility ,Cytokine Release Syndrome ,Cytokine storm ,030217 neurology & neurosurgery - Abstract
Vaccine and antiviral development against SARS-CoV-2 infection or COVID-19 disease would benefit from validated small animal models. Here, we show that transgenic mice expressing human angiotensin-converting enzyme 2 (hACE2) by the human cytokeratin 18 promoter (K18 hACE2) represent a susceptible rodent model. K18 hACE2 transgenic mice succumbed to SARS-CoV-2 infection by day 6, with virus detected in lung airway epithelium and brain. K18 ACE2 transgenic mice produced a modest TH1/2/17 cytokine storm in the lung and spleen that peaked by day 2, and an extended chemokine storm that was detected in both lungs and brain. This chemokine storm was also detected in the brain at day 6. K18 hACE2 transgenic mice are, therefore, highly susceptible to SARS-CoV-2 infection and represent a suitable animal model for the study of viral pathogenesis, and for identification and characterization of vaccines (prophylactic) and antivirals (therapeutics) for SARS-CoV-2 infection and associated severe COVID-19 disease., Here, the authors characterize tissue-level SARS-CoV-2 infection and pathogenesis in transgenic mice expressing human angiotensin converting enzyme 2 (hACE2) by the human cytokeratin 18 promoter (K18-hACE2) and show that infection induces lethality, making the K18-hACE2 model suitable for vaccine and therapeutic evaluation.
- Published
- 2020
19. Immune variations throughout the course of tuberculosis treatment and its relationship with adrenal hormone changes in HIV-1 patients co-infected with Mycobacterium tuberculosis
- Author
-
Graciela Ben, Matias Tomas Angerami, María Florencia Quiroga, Natalia Laufer, Luis D. Giavedoni, Omar Sued, Gabriela Turk, Diego Ameri, Patricia Maidana, Diego Gonzalez, Guadalupe Verónica Suarez, Laura M. Parodi, Viviana Mesch, Bibiana Fabre, and María Belén Vecchione
- Subjects
0301 basic medicine ,Male ,Chemokine ,Time Factors ,Hydrocortisone ,Antitubercular Agents ,HIV Infections ,Systemic inflammation ,T-Lymphocytes, Regulatory ,Adrenal Cortex Hormones ,Prospective Studies ,biology ,Coinfection ,Dehydroepiandrosterone Sulfate ,Middle Aged ,Infectious Diseases ,medicine.anatomical_structure ,Treatment Outcome ,Host-Pathogen Interactions ,Cytokines ,Female ,medicine.symptom ,hormones, hormone substitutes, and hormone antagonists ,Microbiology (medical) ,Adult ,Tuberculosis ,Regulatory T cell ,030106 microbiology ,Immunology ,Microbiology ,Article ,Mycobacterium tuberculosis ,03 medical and health sciences ,Immune system ,medicine ,Humans ,Risk factor ,business.industry ,Dehydroepiandrosterone ,medicine.disease ,biology.organism_classification ,030104 developmental biology ,biology.protein ,HIV-1 ,business ,Biomarkers ,Hormone - Abstract
HIV infection is a major risk factor predisposing for Mycobacterium tuberculosis infection and progression to active tuberculosis (TB). As host immune response defines the course of infection, we aimed to identify immuno-endocrine changes over six-months of anti-TB chemotherapy in HIV+ people. Plasma levels of cortisol, DHEA and DHEA-S, percentages of CD4+ regulatory T cell subsets and number of IFN-γ-secreting cells were determined. Several cytokines, chemokines and C-reactive protein levels were measured. Results were correlated with clinical parameters as predictors of infection resolution and compared to similar data from HIV+ individuals, HIV-infected persons with latent TB infection and healthy donors. Throughout the course of anti-TB/HIV treatment, DHEA and DHEA-S plasma levels raised while cortisol diminished, which correlated to predictive factors of infection resolution. Furthermore, the balance between cortisol and DHEA, together with clinical assessment, may be considered as an indicator of clinical outcome after anti-TB treatment in HIV+ individuals. Clinical improvement was associated with reduced frequency of unconventional Tregs, increment in IFN-γ-secreting cells, diminution of systemic inflammation and changes of circulating cytokines and chemokines. This study suggests that the combined anti-HIV/TB therapies result in partial restoration of both, immune function and adrenal hormone plasma levels.
- Published
- 2020
20. Lethality of SARS-CoV-2 infection in K18 human angiotensin converting enzyme 2 transgenic mice
- Author
-
Shannan Hall-Ursone, Luis D. Giavedoni, Jun-Gyu Park, Anwari Akhter, Olga Gonzalez, Deepak Kaushal, Colin Chuba, Stephanie Earley, Colwyn A. Headley, Anna Allué-Guardia, Andreu Garcia-Vilanova, Alyssa Schami, Katrina N. Kavelish, Luis Martinez-Sobrido, Michal Gazi, Edward J. Dick, Jesse Martinez, Stephanie Davis Mdaki, Chengjin Ye, Cory R. A. Hallam, Jean L. Patterson, Fatai S. Oladunni, Joanne Turner, Tim J. Anderson, Kevin Chiem, Paula Pino Tamayo, Xavier Alvarez, Laura M. Parodi, Kendra J. Alfson, Jordi B. Torrelles, Roy N. Platt, Renee Escalona, Hilary M. Staples, Juan Ignacio García, Alison Whigham, Larry S. Schlesinger, Shalini Gautam, Angélica Olmo-Fontánez, Oscar H. Rodriguez, Varun Dwivedi, Ricardo Carrion, and Corbett Christie
- Subjects
Genetically modified mouse ,Chemokine ,biology ,Viral pathogenesis ,Spleen ,macromolecular substances ,medicine.disease ,Virology ,Virus ,medicine.anatomical_structure ,Angiotensin-converting enzyme 2 ,medicine ,biology.protein ,Respiratory epithelium ,Cytokine storm - Abstract
Vaccine and antiviral development against SARS-CoV-2 infection or COVID-19 disease currently lacks a validated small animal model. Here, we show that transgenic mice expressing human angiotensin converting enzyme 2 (hACE2) by the human cytokeratin 18 promoter (K18 hACE2) represent a susceptible rodent model. K18 hACE2-transgenic mice succumbed to SARS-CoV-2 infection by day 6, with virus detected in lung airway epithelium and brain. K18 ACE2-transgenic mice produced a modest TH1/2/17 cytokine storm in the lung and spleen that peaked by day 2, and an extended chemokine storm that was detected in both lungs and brain. This chemokine storm was also detected in the brain at day 4. K18 hACE2-transgenic mice are, therefore, highly susceptible to SARS-CoV-2 infection and represent a suitable animal model for the study of viral pathogenesis, and for identification and characterization of vaccines (prophylactic) and antivirals (therapeutics) for SARS-CoV-2 infection and associated severe COVID-19 disease.
- Published
- 2020
21. SARS-CoV-2 infection leads to acute infection with dynamic cellular and inflammatory flux in the lung that varies across nonhuman primate species
- Author
-
Mushtaq Ahmed, Bindu Singh, Tim J. Anderson, Justin Ferdin, Shannan Hall-Ursone, Ruby Escabedo, Jean L. Patterson, Larry S. Schlesinger, Xavier Alvarez, Dhiraj Kumar Singh, Cory R. A. Hallam, Alina Baum, Michal Gazi, Bruce A. Rosa, Ken Sayers, Andra K. Voges, Corinna N. Ross, Jessica Callery, Amanda Mannino, Adelekan Oyejide, Carmen Bartley, Yenny Goez-Gazi, Ayan Chatterjee, Elizabeth Clemmons, Deepak Kaushal, Rajesh Thippeshappa, Cynthia Alvarez, Richard Copin, Tae-Hyung Lee, Edward J. Dick, Anna Goodroe, Hilary M. Staples, Christopher H. Chen, Julia M. Scordo, Journey Cole, Vida L. Hodara, Laura M. Parodi, Alyssa Blakely, Patrice A. Frost, Priscilla Escareno, Jordi B. Torrelles, Roy N. Platt, Riti Sharan, Benjamin Klaffke, Kendra J. Alfson, Olga Gonzalez, Shashank Ganatra, Makedonka Mitreva, Dharani Ajithdoss, John Dutton, Christos A. Kyratsous, Ricardo Carrion, Maya Gough, Luis Martinez-Sobrido, Shabaana A. Khader, Kathleen M. Brasky, Joanne Turner, and Luis D. Giavedoni
- Subjects
Lung ,Disease ,Biology ,Systemic inflammation ,medicine.disease ,Virus ,Pneumonia ,medicine.anatomical_structure ,Immune system ,Viral replication ,Immunology ,medicine ,medicine.symptom ,Pneumonitis - Abstract
SummaryThere are no known cures or vaccines for COVID-19, the defining pandemic of this era. Animal models are essential to fast track new interventions and nonhuman primate (NHP) models of other infectious diseases have proven extremely valuable. Here we compare SARS-CoV-2 infection in three species of experimentally infected NHPs (rhesus macaques, baboons, and marmosets). During the first 3 days, macaques developed clinical signatures of viral infection and systemic inflammation, coupled with early evidence of viral replication and mild-to-moderate interstitial and alveolar pneumonitis, as well as extra-pulmonary pathologies. Cone-beam CT scans showed evidence of moderate pneumonia, which progressed over 3 days. Longitudinal studies showed that while both young and old macaques developed early signs of COVID-19, both groups recovered within a two-week period. Recovery was characterized by low-levels of viral persistence in the lung, suggesting mechanisms by which individuals with compromised immune systems may be susceptible to prolonged and progressive COVID-19. The lung compartment contained a complex early inflammatory milieu with an influx of innate and adaptive immune cells, particularly interstitial macrophages, neutrophils and plasmacytoid dendritic cells, and a prominent Type I-interferon response. While macaques developed moderate disease, baboons exhibited prolonged shedding of virus and extensive pathology following infection; and marmosets demonstrated a milder form of infection. These results showcase in critical detail, the robust early cellular immune responses to SARS-CoV-2 infection, which are not sterilizing and likely impact development of antibody responses. Thus, various NHP genera recapitulate heterogeneous progression of COVID-19. Rhesus macaques and baboons develop different, quantifiable disease attributes making them immediately available essential models to test new vaccines and therapies.
- Published
- 2020
22. Effective control of early Zika virus replication by Dengue immunity is associated to the length of time between the 2 infections but not mediated by antibodies
- Author
-
Laura J. White, Luis D. Giavedoni, James D. Brien, Amelia K. Pinto, Melween I. Martínez, Teresa Arana, Aravinda M. de Silva, Idia V. Rodríguez, L. Parodi, Petraleigh Pantoja, Crisanta Serrano-Collazo, Lorna Cruz, Vida L. Hodara, Mariah Hassert, Erick X. Pérez-Guzmán, and Carlos A. Sariol
- Subjects
RNA viruses ,Male ,0301 basic medicine ,Viral Diseases ,Time Factors ,Physiology ,T-Lymphocytes ,RC955-962 ,Monkeys ,Dengue virus ,Pathology and Laboratory Medicine ,medicine.disease_cause ,Biochemistry ,Dengue fever ,Zika virus ,Dengue ,White Blood Cells ,0302 clinical medicine ,Animal Cells ,Immune Physiology ,Arctic medicine. Tropical medicine ,Medicine and Health Sciences ,Immune Response ,Mammals ,Immunity, Cellular ,Immune System Proteins ,T Cells ,Zika Virus Infection ,Eukaryota ,Animal Models ,Infectious Diseases ,Experimental Organism Systems ,Medical Microbiology ,Viral Pathogens ,Viruses ,Vertebrates ,Pathogens ,Cellular Types ,Antibody ,Public aspects of medicine ,RA1-1270 ,Macaque ,Research Article ,Primates ,Immune Cells ,Immunology ,030231 tropical medicine ,Cytotoxic T cells ,Viremia ,Biology ,Research and Analysis Methods ,Microbiology ,Antibodies ,03 medical and health sciences ,Immune system ,Immunity ,Old World monkeys ,medicine ,Animals ,Immunologic Factors ,Microbial Pathogens ,Blood Cells ,Innate immune system ,Biology and life sciences ,Flaviviruses ,Rhesus Monkeys ,Organisms ,Public Health, Environmental and Occupational Health ,Proteins ,Zika Virus ,Cell Biology ,Dendritic Cells ,biochemical phenomena, metabolism, and nutrition ,medicine.disease ,biology.organism_classification ,Macaca mulatta ,Virology ,Immunity, Innate ,Immunity, Humoral ,030104 developmental biology ,Amniotes ,Animal Studies ,biology.protein - Abstract
Little is known about the contribution of virus-specific and cross-reacting antibodies (Abs) or the cellular immune response generated by a primary dengue (DENV) infection on the course of a secondary zika (ZIKV) infection in vivo. Here we show that the length of time between DENV/ZIKV infections has a qualitative impact on controlling early ZIKV replication. Depletion of DENV2-specific Abs in sera confirmed that those type-specific Abs do not contribute to ZIKV control. We show that the magnitude and durability of the neutralizing antibodies (nAbs) induced by a secondary ZIKV infection is modest compared to the response induced after a secondary heterologous DENV infection. Our in vivo results are showing a complex interplay between the cellular and innate immune responses characterized by a high frequency of plasmacytoid dendritic cells (pDC) correlating with an increase in the frequency of DENV antigen specific T cells and a significant control of ZIKV replication which is time dependent. Taken together, our results suggest that early after ZIKV infection other mechanisms such as the innate and cellular immune responses may play a predominant role in controlling ZIKV replication. Regardless of the time elapsed between infections there was no evidence of in vivo antibody-dependent enhancement (ADE) of ZIKV by DENV immunity. These findings have pivotal implications while interpreting ZIKV pathogenesis in flavivirus-experimented populations, diagnostic results interpretation and vaccine designs and schedules among others., Author summary From our previous work in non-human primates and others using humans, we believe that previous DENV immunity confers some degree of protection against ZIKV infection. However, at least two highly relevant questions remain unanswered. One is precisely if the time between primary DENV and a subsequent ZIKV infections may play a role in the degree of protection conferred by DENV immunity. The second question is related to the mechanisms of cross-protection. In this work we provide evidences that a period of 12 months between DENV and ZIKV infections has a significant impact controlling ZIKV replication compared to a shorter period of 3 months. We also provide evidences that the pre-existing DENV Abs play no role controlling early ZIKV replication. Our results strongly suggest that the mechanisms controlling ZIKV replication are related to the complex interaction between the innate and the cellular immune responses. Our results have significant implications for vaccine design and schedules.
- Published
- 2020
23. Baboon CD8 T cells suppress SIVmac infection in CD4 T cells through contact-dependent production of MIP-1α, MIP-1β, and RANTES
- Author
-
Vida L. Hodara, Luis D. Giavedoni, Laura M. Parodi, and Veronica Obregon-Perko
- Subjects
CD4-Positive T-Lymphocytes ,0301 basic medicine ,Chemokine ,Receptors, CCR5 ,animal diseases ,viruses ,Immunology ,Simian Acquired Immunodeficiency Syndrome ,Viremia ,CD8-Positive T-Lymphocytes ,Biology ,Virus Replication ,medicine.disease_cause ,Biochemistry ,Article ,03 medical and health sciences ,0302 clinical medicine ,biology.animal ,medicine ,Animals ,Immunology and Allergy ,Cytotoxic T cell ,Chemokine CCL4 ,Chemokine CCL5 ,Molecular Biology ,Chemokine CCL3 ,virus diseases ,hemic and immune systems ,Hematology ,Viral Load ,Simian immunodeficiency virus ,medicine.disease ,biology.organism_classification ,Macaca mulatta ,Virology ,Coculture Techniques ,Rhesus macaque ,030104 developmental biology ,Viral replication ,Leukocytes, Mononuclear ,biology.protein ,Simian Immunodeficiency Virus ,Viral load ,Papio ,030215 immunology ,Baboon - Abstract
Simian immunodeficiency virus (SIV) infection in rhesus macaques is often characterized by high viremia and CD4 T cell depletion. By contrast, SIV infection in African nonhuman primate natural hosts is typically nonpathogenic despite active viral replication. Baboons are abundant in Africa and have a geographical distribution that overlaps with natural hosts, but they do not harbor SIVs. Previous work has demonstrated baboons are resistant to chronic SIV infection and/or disease in vivo but the underlying mechanisms remain unknown. Using in vitro SIVmac infections, we sought to identify SIV restriction factors in baboons by comparing observations to the pathogenic rhesus macaque model. SIVmac replicated in baboon PBMC but had delayed kinetics compared to rhesus PBMC. However, SIVmac replication in baboon and rhesus isolated CD4 cells were similar to the kinetics seen for rhesus PBMC, demonstrating intracellular restriction factors do not play a strong role in baboon inhibition of SIVmac replication. Here, we show CD8 T cells contribute to the innate SIV-suppressive activity seen in naïve baboon PBMC. As one mechanism of restriction, we identified higher production of MIP-1α, MIP-1β, and RANTES by baboon PBMC. Contact between CD4 and CD8 T cells resulted in maximum production of these chemokines and suppression of viral replication, whereas neutralization of CCR5-binding chemokines in baboon PBMC increased viral loads. Our studies indicate baboon natural restriction of SIVmac replication is largely dependent on CD4-extrinsinc mechanisms mediated, in part, by CD8 T cells.
- Published
- 2018
24. Systematic evaluation of monoclonal antibodies and immunoassays for the detection of Interferon-γ and Interleukin-2 in old and new world non-human primates
- Author
-
Luis D. Giavedoni, Niklas Ahlborg, Bartels Zuber, Ankie Höglind, Irene Areström, Cecilia Ehrnfelt, and Khosro Masjedi
- Subjects
0301 basic medicine ,Interleukin 2 ,Cellular immunity ,Enzyme-Linked Immunospot Assay ,medicine.drug_class ,030231 tropical medicine ,Immunology ,ELISpot ,Cross Reactions ,Monoclonal antibody ,Article ,03 medical and health sciences ,Interferon-gamma ,0302 clinical medicine ,Chlorocebus aethiops ,medicine ,Animals ,Humans ,Immunology and Allergy ,Antibody ,Non-human primates ,Immunoassay ,biology ,ELISPOT ,Antibodies, Monoclonal ,Pigtail macaque ,biology.organism_classification ,Flow Cytometry ,Virology ,Macaca mulatta ,humanities ,3. Good health ,Interleukin-10 ,Macaca fascicularis ,030104 developmental biology ,Sooty mangabey ,biology.protein ,Interleukin-2 ,ELISA ,Interferon-γ ,FluoroSpot ,medicine.drug - Abstract
Non-human primates (NHP) provide important animal models for studies on immune responses to infections and vaccines. When assessing cellular immunity in NHP, cytokines are almost exclusively analyzed utilizing cross-reactive anti-human antibodies. The functionality of antibodies has to be empirically established for each assay/application as well as NHP species. A rational approach was employed to identify monoclonal antibodies (mAb) cross-reactive with many NHP species. Panels of new and established mAbs against human Interferon (IFN)-γ and Interleukin (IL)-2 were assessed for reactivity with eukaryotically expressed recombinant IFN-γ and IL-2, respectively, from Old (rhesus, cynomolgus and pigtail macaques, African green monkey, sooty mangabey and baboon) and New World NHP (Ma's night monkey, squirrel monkey and common marmoset). Pan-reactive mAbs, recognizing cytokines from all NHP species, were further analyzed in capture assays and flow cytometry with NHP peripheral blood mononuclear cells (PBMC). Pan-reactive mAb pairs for IFN-γ well as IL-2 were identified and used in ELISA to measure IFN-γ and IL-2, respectively, in Old and New World NHP PBMC supernatants. The same mAb pairs displayed high functionality in ELISpot and FluoroSpot for the measurement of antigen-specific IFN-γ and IL-2 responses using cynomolgus PBMC. Functionality of pan-reactive mAbs in flow cytometry was also verified with cynomolgus PBMC. The development of well-defined immunoassays functional with a panel of NHP species facilitates improved analyses of cellular immunity and enables inclusion in multiplex cytokine assays intended for a variety of NHP.
- Published
- 2017
- Full Text
- View/download PDF
25. Nef-Mediated CD3-TCR Downmodulation Dampens Acute Inflammation and Promotes SIV Immune Evasion
- Author
-
Sydney A. Nelson, Kerstin Mätz-Rensing, Christiane Stahl-Hennig, Simone Joas, Anke Heigele, Luis D. Giavedoni, Ulrike Sauermann, Daniel Sauter, Guido Silvestri, Antonina Klippert, Prachi Mehrotra Gupta, Berit Roshani, Frank Kirchhoff, Gregory K. Tharp, Laura M. Parodi, Maria Daskalaki, Steven E. Bosinger, and Nicole Stolte-Leeb
- Subjects
0301 basic medicine ,Male ,CD3 ,animal diseases ,viruses ,Inflammation ,medicine.disease_cause ,General Biochemistry, Genetics and Molecular Biology ,Gene Products, nef ,Article ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,medicine ,Animals ,Viral Regulatory and Accessory Proteins ,lcsh:QH301-705.5 ,Immune Evasion ,biology ,T-cell receptor ,virus diseases ,Simian immunodeficiency virus ,Virology ,Macaca mulatta ,3. Good health ,030104 developmental biology ,Simian AIDS ,Viral replication ,lcsh:Biology (General) ,Receptor-CD3 Complex, Antigen, T-Cell ,biology.protein ,Female ,Simian Immunodeficiency Virus ,medicine.symptom ,Viral load ,030217 neurology & neurosurgery - Abstract
SUMMARY The inability of Nef to downmodulate the CD3-T cell receptor (TCR) complex distinguishes HIV-1 from other primate lentiviruses and may contribute to its high virulence. However, the role of this Nef function in virus-mediated immune activation and pathogenicity remains speculative. Here, we selectively disrupted this Nef activity in SIVmac239 and analyzed the consequences for the virological, immunological, and clinical outcome of infection in rhesus macaques. The inability to downmodulate CD3-TCR does not impair viral replication during acute infection but is associated with increased immune activation and antiviral gene expression. Subsequent early reversion in three of six animals suggests strong selective pressure for this Nef function and is associated with high viral loads and progression to simian AIDS. In the absence of reversions, however, viral replication and the clinical course of infection are attenuated. Thus, Nef-mediated downmodulation of CD3 dampens the inflammatory response to simian immunodeficiency virus (SIV) infection and seems critical for efficient viral immune evasion., Graphical Abstract, In Brief HIV-1 lacks the CD3 downmodulation function of Nef that is otherwise conserved in primate lentiviruses. Joas et al. disrupted this Nef activity in SIVmac239 and show that Nef-mediated downmodulation of CD3 dampens inflammatory responses to SIV. This promotes effective immune evasion and maintenance of high viral loads in infected rhesus macaques.
- Published
- 2019
26. Significant control of Zika infection in macaques depends on the elapsing time after dengue exposure
- Author
-
Mariah Hassert, de Silva A, James D. Brien, Hodara, Petraleigh Pantoja, Luis D. Giavedoni, Melween I. Martínez, Teresa Arana, Laura J. White, Carlos A. Sariol, Erick X. Pérez-Guzmán, Lorna Cruz, Amelia K. Pinto, Laura M. Parodi, Rodríguez, and Crisanta Serrano-Collazo
- Subjects
Serotype ,0303 health sciences ,viruses ,virus diseases ,biochemical phenomena, metabolism, and nutrition ,Dengue virus ,Biology ,medicine.disease ,medicine.disease_cause ,biology.organism_classification ,Virology ,Virus ,3. Good health ,Dengue fever ,03 medical and health sciences ,Flavivirus ,0302 clinical medicine ,Immune system ,Immunity ,medicine ,biology.protein ,030212 general & internal medicine ,Antibody ,030304 developmental biology - Abstract
Prior exposure to a single serotype of dengue virus (DENV) predisposes individuals to severe disease upon secondary heterologous DENV infection. Here we show that the length of time between DENV/Zika (ZIKV) infections has a qualitative impact on controlling ZIKV replication. We identified limited but significant differences in the magnitude of the early humoral immune response associated with a period of twelve months but not three months of DENV convalescence. However, their role limiting ZIKV replication is not conclusive. There was no evidence of in vivo antibody-dependent amplification of ZIKV by DENV immunity in any group. We are also showing that the significant differences among groups may be linked to a pre-existing polyfunctional CD4+ T cells response (increased IFN-g and Cd107a before ZIKV infection) and to an early and continuous expansion of the CD4+ effector memory cells early on after ZIKV infection. Those significant differences were associated with a period of 12 months after DENV infection that were not observed in a span of 3-months. These results suggest that there is a window of optimal cross-protection between ZIKV and DENV with significant consequences. These results have pivotal implications while interpreting ZIKV pathogenesis in flavivirus-experimented populations, diagnostic results interpretation and vaccine designs among others.Author SummarySince its introduction in the Americas region ZIKV virus has been associated to severe birth defects. One of the questions that remains open is the role of previous dengue or any other flavivirus immunity in the pathogenesis of ZIKV and more important, if the time elapse between DENV and ZIKV play a role enhancing ZIKV pathogenesis as it is the case for subsequent DENV infections. On this work, using NHP as a model we compared the effect of a period of 12 months vs. a period of 3 months of DENV immunity in the outcome of ZIKV infection. We found that previous DENV infection, at any of the tested period of time do not induce ZIKV enhancement. More relevant are showing that when the two infection occurs at least one year apart the preexisting DENV immunity is better at controlling ZIKV replication and that the role of the neutralizing antibodies is very limited. On the contrary our results suggest that early after ZIKV infection the cellular immune response, may plays a predominant role. Our findings have critical relevance to understand the dynamic interaction between these two flavivirus, their pathogenies, diagnosis and vaccine design.
- Published
- 2019
27. The Impact of Advanced Age on Vaccine-Induced Tissue Immune Recall Responses
- Author
-
Julia M Scordo, Tucker J Piergallini, Colwyn A Headley, Nicole Reuter, Vida L Hodara, Olga Gonzalez, Luis D Giavedoni, James F Papin, and Joanne Turner
- Subjects
Immunology ,Immunology and Allergy - Abstract
Infectious diseases account for one-third of deaths in individuals over the age of 65. Vaccines are a primary tool to combat infection, yet they are less effective in the aged population. Many have focused on vaccine-induced peripheral blood responses in old age; however, work from our lab demonstrates that immune responses to vaccination and infection in the periphery and local infection site (tissue) differ. To improve health outcomes in our aged population, we must study both systemic and tissue vaccine responses. We do so using a model of Mycobacterium bovis BCG vaccination and subsequent tuberculin skin test (TST) eight weeks after vaccination in adult and aged baboons. Vaccination generates BCG-specific immune cells that are recruited to the skin upon TST challenge, which we can study in skin biopsies reflecting tissue-specific responses. Three days or one week after TST we examine BCG-specific responses in skin biopsies. We also determine BCG-induced responses in the blood to compare tissue and systemic responses. We find increased oxidation and decreased production of immune proteins in aged baboon skin at the site of TST challenge, despite no alterations in antigen-specific peripheral blood mononuclear cell (PBMC) responses between age groups. Interestingly, cell migration assays show increased migration of PBMCs from aged BCG-vaccinated baboons, reinforcing the notion that systemic responses are not altered and that study of the tissue environment is important to understand responses to antigenic challenge with older age.
- Published
- 2021
28. Increases in NKG2C Expression on T Cells and Higher Levels of Circulating CD8+B Cells Are Associated with Sterilizing Immunity Provided by a Live Attenuated SIV Vaccine
- Author
-
M. Shannon Keckler, Luis D. Giavedoni, Vida L. Hodara, and Laura M. Parodi
- Subjects
0301 basic medicine ,Attenuated vaccine ,medicine.medical_treatment ,Immunology ,Inflammation ,Biology ,Simian immunodeficiency virus ,medicine.disease_cause ,Virology ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Infectious Diseases ,Immune system ,Cytokine ,Immunity ,medicine ,Cytotoxic T cell ,medicine.symptom ,CD8 ,030215 immunology - Abstract
Vaccines based on live attenuated viruses are highly effective immunogens in the simian immunodeficiency virus (SIV)/rhesus macaque animal model and offer the possibility of studying correlates of protection against infection with virulent virus. We utilized a tether system for studying, in naive macaques and animals vaccinated with a live-attenuated vaccine, the acute events after challenge with pathogenic SIV. This approach allowed for the frequent sampling of small blood volumes without sedation or restraining of the animals, thus reducing the confounding effect of sampling stress. Before challenge, vaccinated animals presented significantly higher levels of proliferating and activated B cells than naive macaques, which were manifested by high expression of CD8 on B cells. After SIV challenge, the only changes observed in protected vaccinated macaques were significant increases in expression of the NK marker NKG2C on CD4 and CD8 T cells. We also identified that infection of naive macaques with SIV resulted in a transient peak of expression of CD20 on CD8 T cells and a constant rise in the number of B cells expressing CD8. Finally, analysis of a larger cohort of vaccinated animals identified that, even when circulating levels of vaccine virus are below the limit of detection, live attenuated vaccines induce systemic increases of IP-10 and perforin. These studies indicate that components of both the innate and adaptive immune systems of animals inoculated with a live-attenuated SIV vaccine respond to and control infection with virulent virus. Persistence of the vaccine virus in tissues may explain the elevated cytokine and B-cell activation levels. In addition, our report underpins the utility of the tether system for the intensive study of acute immune responses to viral infections.
- Published
- 2016
29. Acute Liver Damage Associated with Innate Immune Activation in a Small Nonhuman Primate Model of Hepacivirus Infection
- Author
-
Luis D. Giavedoni, Joshua A. Kramer, Amanda J. Martinot, Lynn M. Wachtman, Premeela A. Rajakumar, Cordelia Manickam, R. Keith Reeves, and Valerie Varner
- Subjects
0301 basic medicine ,Hepacivirus ,Hepatitis C virus ,Immunology ,medicine.disease_cause ,Microbiology ,GB virus B ,03 medical and health sciences ,Liver disease ,Immune system ,Immunity ,Virology ,medicine ,Animals ,Immunologic Factors ,Hepatitis ,Innate immune system ,biology ,Callithrix ,Dendritic Cells ,Viral Load ,biology.organism_classification ,medicine.disease ,Immunity, Innate ,Killer Cells, Natural ,030104 developmental biology ,Liver ,Hepatitis, Viral, Animal ,Insect Science ,Pathogenesis and Immunity ,Cytokines ,Viral load - Abstract
Despite its importance in shaping adaptive immune responses, viral clearance, and immune-based inflammation, tissue-specific innate immunity remains poorly characterized for hepatitis C virus (HCV) infection due to the lack of access to acutely infected tissues. In this study, we evaluated the impact of natural killer (NK) cells and myeloid (mDCs) and plasmacytoid (pDCs) dendritic cells on control of virus replication and virus-induced pathology caused by another, more rapidly resolving hepacivirus, GB virus B (GBV-B), in infections of common marmosets. High plasma and liver viral loads and robust hepatitis characterized acute GBV-B infection, and while viremia was generally cleared by 2 to 3 months postinfection, hepatitis and liver fibrosis persisted after clearance. Coinciding with peak viral loads and liver pathology, the levels of NK cells, mDCs, and pDCs in the liver increased up to 3-fold. Although no obvious numerical changes in peripheral innate cells occurred, circulating NK cells exhibited increased perforin and Ki67 expression levels and increased surface expression of CXCR3. These data suggested that increased NK cell arming and proliferation as well as tissue trafficking may be associated with influx into the liver during acute infection. Indeed, NK cell frequencies in the liver positively correlated with plasma ( R = 0.698; P = 0.015) and liver ( R = 0.567; P = 0.057) viral loads. Finally, soluble factors associated with NK cells and DCs, including gamma interferon (IFN-γ) and RANTES, were increased in acute infection and also were associated with viral loads and hepatitis. Collectively, the findings showed that mobilization of local and circulating innate immune responses was linked to acute virus-induced hepatitis, and potentially to resolution of GBV-B infection, and our results may provide insight into similar mechanisms in HCV infection. IMPORTANCE Hepatitis C virus (HCV) infection has created a global health crisis, and despite new effective antivirals, it is still a leading cause of liver disease and death worldwide. Recent evidence suggests that innate immunity may be a potential therapeutic target for HCV, but it may also be a correlate of increased disease. Due to a lack of access to human tissues with acute HCV infection, in this study we evaluated the role of innate immunity in resolving infection with a hepacivirus, GBV-B, in common marmosets. Collectively, our data suggest that NK cell and DC mobilization in acute hepacivirus infection can dampen virus replication but also regulate acute and chronic liver damage. How these two opposing effects on the host may be modulated in future therapeutic and vaccine approaches warrants further study.
- Published
- 2016
30. Env-Specific IgA from Viremic HIV-Infected Subjects Compromises Antibody-Dependent Cellular Cytotoxicity
- Author
-
Juliana Falivene, Yanina Alexandra Ghiglione, María Julia Ruiz, María Pía Holgado, Pedro Cahn, Horacio Salomón, María Eugenia Socías, Ana M. Rodríguez, Luis D. Giavedoni, María Magdalena Gherardi, Natalia Laufer, Gabriela Turk, and Omar Sued
- Subjects
Adult ,Male ,0301 basic medicine ,Immunoglobulin A ,Immunology ,HIV Infections ,chemical and pharmacologic phenomena ,Viremia ,HIV Antibodies ,HIV Envelope Protein gp120 ,Microbiology ,Young Adult ,03 medical and health sciences ,0302 clinical medicine ,Immunity ,Virology ,medicine ,Humans ,Fluorometry ,030212 general & internal medicine ,HIV vaccine ,Young adult ,Aged ,Antibody-dependent cell-mediated cytotoxicity ,biology ,Antibody-Dependent Cell Cytotoxicity ,virus diseases ,hemic and immune systems ,Middle Aged ,medicine.disease ,3. Good health ,Chronic infection ,030104 developmental biology ,Insect Science ,biology.protein ,Pathogenesis and Immunity ,Antibody - Abstract
Elucidating the factors that modulate HIV-specific antibody-dependent cellular cytotoxicity (ADCC) will help in understanding its role in HIV immunity. The aim of this study was to determine whether IgA could modify the magnitude of ADCC in HIV infection, abrogating its protective role. Plasma samples from 20 HIV-positive (HIV + ) subjects enrolled during primary HIV infection (PHI), 10 chronically infected subjects (chronic), and 7 elite controllers (EC) were used. ADCC was determined by using a fluorometric ADCC assay, before and after removal of plasma IgA. Data were analyzed by using nonparametric statistics. ADCC was documented in 80% of PHI enrollment samples and in 100% of PHI 12-month, chronic, and EC samples; it peaked after acute infection, reached a plateau in chronic infection, and decreased after initiation of antiretroviral treatment (ART). Significant associations between ADCC and disease progression were found only after removal of plasma IgA from 12-month PHI samples: the magnitude of ADCC not only increased after IgA removal but also correlated with CD4 + T-cell preservation. This work provides evidence that gp120-specific IgA was capable of modifying ADCC responses during natural HIV infection for the first time and adds to similar evidence provided in other settings. Furthermore, it underscores the complexity of the ADCC phenomenon and will help in an understanding of its underlying mechanisms. IMPORTANCE Although the induction of ADCC-mediating antibodies in HIV-infected subjects has been extensively documented, the association of these antibodies with protection from disease progression is poorly understood. Here, we demonstrate that plasma IgA is a factor capable of modifying the magnitude of IgG-mediated ADCC in HIV infection, mitigating its beneficial effect. These results help in understanding why previous studies failed to demonstrate correlations between ADCC and disease progression, and they also contribute to the notion that an HIV vaccine should stimulate the production of ADCC-mediating IgG antibodies but not IgA.
- Published
- 2016
31. Publisher Correction: Experimental Zika Virus Infection in the Pregnant Common Marmoset Induces Spontaneous Fetal Loss and Neurodevelopmental Abnormalities
- Author
-
Jean L. Patterson, Vida L. Hodara, Calla Martyn, Luis D. Giavedoni, Donna Layne-Colon, Suzette D. Tardif, Eumenia Costa da Cunha Castro, Maxim Seferovic, Charles Y. Chiu, Shigeo Yagi, Julienne N. Rutherford, Kevin Reyes, Melissa J. Suter, Manasi Tamhankar, Tony Li, Laura M. Parodi, Kjersti Aagaard, and Claudia Sanchez San Martin
- Subjects
0301 basic medicine ,Placenta ,lcsh:Medicine ,Gestational Age ,Nervous System Malformations ,Virus Replication ,Zika virus ,03 medical and health sciences ,Interferon-gamma ,Fetus ,Pregnancy ,biology.animal ,Maxim ,Animals ,Humans ,Fetal loss ,Viremia ,Pregnancy Complications, Infectious ,lcsh:Science ,Multidisciplinary ,biology ,Zika Virus Infection ,Calla ,Philosophy ,Published Erratum ,lcsh:R ,Neurosciences ,Marmoset ,Callithrix ,Zika Virus ,biology.organism_classification ,Publisher Correction ,Abortion, Spontaneous ,Disease Models, Animal ,Good Health and Well Being ,030104 developmental biology ,Interferon Type I ,ComputingMethodologies_DOCUMENTANDTEXTPROCESSING ,Embryo Loss ,Cytokines ,lcsh:Q ,Female ,Humanities - Abstract
During its most recent outbreak across the Americas, Zika virus (ZIKV) was surprisingly shown to cause fetal loss and congenital malformations in acutely and chronically infected pregnant women. However, understanding the underlying pathogenesis of ZIKV congenital disease has been hampered by a lack of relevant in vivo experimental models. Here we present a candidate New World monkey model of ZIKV infection in pregnant marmosets that faithfully recapitulates human disease. ZIKV inoculation at the human-equivalent of early gestation caused an asymptomatic seroconversion, induction of type I/II interferon-associated genes and proinflammatory cytokines, and persistent viremia and viruria. Spontaneous pregnancy loss was observed 16-18 days post-infection, with extensive active placental viral replication and fetal neurocellular disorganization similar to that seen in humans. These findings underscore the key role of the placenta as a conduit for fetal infection, and demonstrate the utility of marmosets as a highly relevant model for studying congenital ZIKV disease and pregnancy loss.
- Published
- 2018
32. Preliminary Studies on Immune Response and Viral Pathogenesis of Zika Virus in Rhesus Macaques
- Author
-
Rajnish S. Dave, Robert B. Norgren, Luis D. Giavedoni, Santhi Gorantla, Omalla A. Olwenyi, Debashis Dutta, Siddappa N. Byrareddy, Noel D. Johnson, Shawna M. Woollard, and Saumi Mathews
- Subjects
0301 basic medicine ,Microbiology (medical) ,Sexual transmission ,Viral pathogenesis ,lcsh:Medicine ,Article ,Zika virus ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Zika ,Immunology and Allergy ,030212 general & internal medicine ,Molecular Biology ,intravaginal ,Fetus ,General Immunology and Microbiology ,biology ,Flaviviruses ,lcsh:R ,immunopathogenesis ,biology.organism_classification ,Virology ,sexual transmission ,3. Good health ,Rhesus macaque ,030104 developmental biology ,Infectious Diseases ,In utero ,CD8 ,rhesus macaque - Abstract
Zika Virus (ZIKV) is primarily transmitted through mosquito bites. It can also be transmitted during sexual intercourse and in utero from mother to fetus. To gain preliminary insight into ZIKV pathology and immune responses on route of transmission, rhesus macaques (RMs) were inoculated with ZIKV (PRVABC59) via intravaginal (IVAG) (n = 3) or subcutaneous (sub Q) (n = 2) routes. Systemic ZIKV infection was observed in all RMs, regardless of the route of inoculation. After 9 days postinfection (dpi), ZIKV was not detected in the plasma of IVAG- and sub-Q-inoculated RMs. Importantly, RMs harbored ZIKV up to 60 dpi in various anatomical locations. Of note, ZIKV was also present in several regions of the brain, including the caudate nucleus, parietal lobe, cortex, and amygdala. These observations appear to indicate that ZIKV infection may be systemic and persistent regardless of route of inoculation. In addition, we observed changes in key immune cell populations in response to ZIKV infection. Importantly, IVAG ZIKV infection of RMs is associated with increased depletion of CD11C hi myeloid cells, reduced PD-1 expression in NK cells, and elevated frequencies of Ki67+ CD8+ central memory cells as compared to sub Q ZIKV-infected RMs. These results need to interpreted with caution due to the small number of animals utilized in this study. Future studies involving large groups of animals that have been inoculated through both routes of transmission are needed to confirm our findings.
- Published
- 2018
33. SIV/SHIV-Zika coinfection does not alter disease pathogenesis in adult non-pregnant Rhesus Macaques
- Author
-
Luis D. Giavedoni, Debashis Dutta, Mehdi R. M. Bidokhti, Siddappa N. Byrareddy, Shawna M. Woollard, Robert B. Norgren, and Lepakshe S. V. Madduri
- Subjects
Chemokine ,biology ,viruses ,Simian human immunodeficiency virus ,virus diseases ,Disease pathogenesis ,Simian immunodeficiency virus ,medicine.disease ,biology.organism_classification ,medicine.disease_cause ,Virology ,Non pregnant ,Zika virus ,Coinfection ,medicine ,biology.protein ,Viral load - Abstract
Due to the large geographical overlap of populations exposed to Zika virus (ZIKV) and human immunodeficiency virus (HIV), understanding disease pathogenesis in such coinfections is urgently needed. We used chronically infected simian immunodeficiency virus and chimeric simian human immunodeficiency virus (SIV/SHIV) macaques and inoculated with ZIKV. Plasma viral loads of both SIV/SHIV and ZIKV showed no significant changes as compared to ZIKV alone-infected animals. Tissue clearance of ZIKV was observed similarly. Furthermore, minimal changes in cytokines/chemokines were observed. Collectively, these data suggest that coinfection may not alter disease pathogenesis and warrants large HIV-ZIKV epidemiological studies to validate these findings.Author SummaryThe co-infection incidence of human immunodeficiency virus (HIV) infection and neglected tropical infectious diseases is increasing due to the large geographical overlap of populations exposed to both of these viruses. Thus, researching on such coinfection is of particular importance. In this study, we investigated HIV-ZIKV coinfection dynamics in adult non-pregnant Rhesus Macaques model chronically infected with simian immunodeficiency virus (SIV) - or chimeric simian human immunodeficiency virus (SHIV). We found that post ZIKV inoculation, plasma viral loads were similar to ZIKV alone infected animals in addition to minimal changes of cytokines. Dynamics of SIV and SHIV also did not change. Tissue clearance of ZIKV was found 67 months later. Our findings provide insights into HIV-ZKIV coinfection to determine the alteration of their pathogenesis.
- Published
- 2018
34. Experimental Zika Virus Infection in the Pregnant Common Marmoset Induces Spontaneous Fetal Loss and Neurodevelopmental Abnormalities
- Author
-
Suzette D. Tardif, Claudia Sanchez San Martin, Jean L. Patterson, Luis D. Giavedoni, Vida L. Hodara, Melissa J. Suter, Kjersti Aagaard, Manasi Tamhankar, Tony Li, Eumenia Costa da Cunha Castro, Laura M. Parodi, Donna Layne-Colon, Charles Y. Chiu, Calla Martyn, Shigeo Yagi, Maxim Seferovic, Julienne N. Rutherford, and Kevin Reyes
- Subjects
0301 basic medicine ,Placenta ,lcsh:Medicine ,Disease ,Reproductive health and childbirth ,Virus Replication ,Zika virus ,Pathogenesis ,0302 clinical medicine ,Pregnancy ,2.1 Biological and endogenous factors ,2.2 Factors relating to the physical environment ,030212 general & internal medicine ,Aetiology ,lcsh:Science ,Pediatric ,0303 health sciences ,Multidisciplinary ,biology ,Zika Virus Infection ,Infectious ,Marmoset ,Callithrix ,3. Good health ,medicine.anatomical_structure ,Infectious Diseases ,Interferon Type I ,Embryo Loss ,Cytokines ,Female ,Infection ,Viremia ,Gestational Age ,Nervous System Malformations ,Article ,03 medical and health sciences ,Interferon-gamma ,Fetus ,biology.animal ,medicine ,Animals ,Humans ,Conditions Affecting the Embryonic and Fetal Periods ,Seroconversion ,030304 developmental biology ,business.industry ,Animal ,Spontaneous ,lcsh:R ,Abortion ,Zika Virus ,Perinatal Period - Conditions Originating in Perinatal Period ,medicine.disease ,biology.organism_classification ,Pregnancy Complications ,030104 developmental biology ,Good Health and Well Being ,Immunology ,Disease Models ,lcsh:Q ,business - Abstract
During its most recent outbreak across the Americas, Zika virus (ZIKV) was surprisingly shown to cause fetal loss and congenital malformations in acutely and chronically infected pregnant women. However, understanding the underlying pathogenesis of ZIKV congenital disease has been hampered by a lack of relevant in vivo experimental models. Here we present a candidate New World monkey model of ZIKV infection in pregnant marmosets that faithfully recapitulates human disease. ZIKV inoculation at the human-equivalent of early gestation caused an asymptomatic seroconversion, induction of type I/II interferon-associated genes and proinflammatory cytokines, and persistent viremia and viruria. Spontaneous pregnancy loss was observed 16–18 days post-infection, with extensive active placental viral replication and fetal neurocellular disorganization similar to that seen in humans. These findings underscore the key role of the placenta as a conduit for fetal infection, and demonstrate the utility of marmosets as a highly relevant model for studying congenital ZIKV disease and pregnancy loss.
- Published
- 2018
35. SIV/SHIV-Zika co-infection does not alter disease pathogenesis in adult non-pregnant rhesus macaque model
- Author
-
Lepakshe S. V. Madduri, Luis D. Giavedoni, Shawna M. Woollard, Robert B. Norgren, Siddappa N. Byrareddy, Mehdi R. M. Bidokhti, and Debashis Dutta
- Subjects
0301 basic medicine ,RNA viruses ,Chemokine ,Physiology ,viruses ,Simian Acquired Immunodeficiency Syndrome ,Monkeys ,medicine.disease_cause ,Pathology and Laboratory Medicine ,Macaque ,Zika virus ,Plasma ,0302 clinical medicine ,Immunodeficiency Viruses ,Medicine and Health Sciences ,030212 general & internal medicine ,Mammals ,biology ,Coinfection ,Zika Virus Infection ,lcsh:Public aspects of medicine ,virus diseases ,Eukaryota ,Animal Models ,Viral Load ,3. Good health ,Body Fluids ,Rhesus macaque ,Blood ,SIV ,Experimental Organism Systems ,Medical Microbiology ,Viral Pathogens ,Viruses ,Vertebrates ,Cytokines ,Infectious diseases ,Female ,Pathogens ,Anatomy ,Viral load ,Research Article ,HIV infections ,Primates ,lcsh:Arctic medicine. Tropical medicine ,lcsh:RC955-962 ,Viral diseases ,Disease pathogenesis ,Research and Analysis Methods ,Microbiology ,Blood Plasma ,03 medical and health sciences ,biology.animal ,Virology ,Old World monkeys ,Retroviruses ,medicine ,Animals ,Microbial Pathogens ,Biology and life sciences ,Flaviviruses ,Rhesus Monkeys ,Lentivirus ,Public Health, Environmental and Occupational Health ,Organisms ,lcsh:RA1-1270 ,Zika Virus ,Simian immunodeficiency virus ,biology.organism_classification ,medicine.disease ,Macaca mulatta ,Disease Models, Animal ,030104 developmental biology ,Co-Infections ,Amniotes ,biology.protein ,Animal Studies ,Viral Transmission and Infection - Abstract
Due to the large geographical overlap of populations exposed to Zika virus (ZIKV) and human immunodeficiency virus (HIV), understanding the disease pathogenesis of co-infection is urgently needed. This warrants the development of an animal model for HIV-ZIKV co-infection. In this study, we used adult non-pregnant macaques that were chronically infected with simian immunodeficiency virus/chimeric simian human immunodeficiency virus (SIV/SHIV) and then inoculated with ZIKV. Plasma viral loads of both SIV/SHIV and ZIKV co-infected animals revealed no significant changes as compared to animals that were infected with ZIKV alone or as compared to SIV/SHIV infected animals prior to ZIKV inoculation. ZIKV tissue clearance of co-infected animals was similar to animals that were infected with ZIKV alone. Furthermore, in co-infected macaques, there was no statistically significant difference in plasma cytokines/chemokines levels as compared to prior to ZIKV inoculation. Collectively, these findings suggest that co-infection may not alter disease pathogenesis, thus warranting larger HIV-ZIKV epidemiological studies in order to validate these findings., Author summary The co-infection incidence of human immunodeficiency virus (HIV) infection and neglected tropical infectious diseases such as Zika virus (ZIKV) is on the rise due to the large geographical overlap of populations exposed to both of these viruses. Thus, research on such co-infection is of particular importance. In this study, we investigated SIV/SHIV-ZIKV co-infection dynamics in adult non-pregnant Rhesus Macaques (RMs) chronically infected with simian immunodeficiency virus (SIV)—or chimeric simian human immunodeficiency virus (SHIV). We found that post ZIKV inoculation, ZIKV plasma viral loads in co-infected macaques were similar to ZIKV alone-infected animals, and minimal changes were observed in cytokines/chemokines levels. Viral levels of SIV and SHIV also did not change as compared to pre-ZIKV inoculation levels. These findings thus suggest that co-infection may not alter disease pathogenesis of either HIV or ZIKV infections.
- Published
- 2018
36. Biomarkers of progression after HIV acute/early infection: Nothing compares to CD4+ T-cell count?
- Author
-
María Magdalena Gherardi, Jimena Salido, María Paula Caruso, María A. Pando, Omar Sued, María Julia Ruiz, Luis D. Giavedoni, Roberto Daniel Rabinovich, María Pía Holgado, Horacio Salomón, Juliana Falivene, Yanina Alexandra Ghiglione, César Ariel Trifone, Macarena Hormanstorfer, Natalia Laufer, Gabriela Turk, Romina Soledad Coloccini, María Inés Figueroa, and Pedro A. Pury
- Subjects
CD4-Positive T-Lymphocytes ,Male ,0301 basic medicine ,soluble plasma factors ,IMMUNE RESPONSES ,lcsh:QR1-502 ,HIV Infections ,lcsh:Microbiology ,purl.org/becyt/ford/1 [https] ,0302 clinical medicine ,Interferon ,Medicine ,030212 general & internal medicine ,Macrophage inflammatory protein ,Interleukin ,Viral Load ,SOLUBLE PLASMA FACTORS ,3. Good health ,HLA ,Infectious Diseases ,Acute Disease ,Cytokines ,Female ,Tumor necrosis factor alpha ,Viral load ,CIENCIAS NATURALES Y EXACTAS ,medicine.drug ,Adult ,Receptors, CCR5 ,BIOMARKERS ,Alpha (ethology) ,Human leukocyte antigen ,Article ,Ciencias Biológicas ,03 medical and health sciences ,disease progression ,Immune system ,Virology ,Humans ,purl.org/becyt/ford/1.6 [https] ,DECISION TREES ,decision trees ,business.industry ,DISEASE PROGRESSION ,biomarkers ,HIV ,immune responses ,CD4 Lymphocyte Count ,Chemokine CXCL10 ,030104 developmental biology ,Immunology ,HIV-1 ,ACUTE INFECTION ,acute infection ,business ,Virología - Abstract
Progression of HIV infection is variable among individuals, and definition disease progression biomarkers is still needed. Here, we aimed to categorize the predictive potential of several variables using feature selection methods and decision trees. A total of seventy-five treatment-naïve subjects were enrolled during acute/early HIV infection. CD4+ T-cell counts (CD4TC) and viral load (VL) levels were determined at enrollment and for one year. Immune activation, HIV-specific immune response, Human Leukocyte Antigen (HLA) and C-C chemokine receptor type 5 (CCR5) genotypes, and plasma levels of 39 cytokines were determined. Data were analyzed by machine learning and non-parametric methods. Variable hierarchization was performed by Weka correlation-based feature selection and J48 decision tree. Plasma interleukin (IL)-10, interferon gamma-induced protein (IP)-10, soluble IL-2 receptor alpha (sIL-2Rα) and tumor necrosis factor alpha (TNF-α) levels correlated directly with baseline VL, whereas IL-2, TNF-α, fibroblast growth factor (FGF)-2 and macrophage inflammatory protein (MIP)-1β correlated directly with CD4+ T-cell activation (p < 0.05). However, none of these cytokines had good predictive values to distinguish “progressors” from “non-progressors”. Similarly, immune activation, HIV-specific immune responses and HLA/CCR5 genotypes had low discrimination power. Baseline CD4TC was the most potent discerning variable with a cut-off of 438 cells/μL (accuracy = 0.93, κ-Cohen = 0.85). Limited discerning power of the other factors might be related to frequency, variability and/or sampling time. Future studies based on decision trees to identify biomarkers of post-treatment control are warrantied. Fil: Turk, Gabriela Julia Ana. Consejo Nacional de Investigaciones Científicas y Técnicas. Oficina de Coordinación Administrativa Houssay. Instituto de Investigaciones Biomédicas en Retrovirus y Sida. Universidad de Buenos Aires. Facultad de Medicina. Instituto de Investigaciones Biomédicas en Retrovirus y Sida; Argentina Fil: Ghiglione, Yanina Alexandra. Consejo Nacional de Investigaciones Científicas y Técnicas. Oficina de Coordinación Administrativa Houssay. Instituto de Investigaciones Biomédicas en Retrovirus y Sida. Universidad de Buenos Aires. Facultad de Medicina. Instituto de Investigaciones Biomédicas en Retrovirus y Sida; Argentina Fil: Hormanstorfer, Macarena. Fundación Huésped; Argentina Fil: Laufer, Natalia Lorna. Consejo Nacional de Investigaciones Científicas y Técnicas. Oficina de Coordinación Administrativa Houssay. Instituto de Investigaciones Biomédicas en Retrovirus y Sida. Universidad de Buenos Aires. Facultad de Medicina. Instituto de Investigaciones Biomédicas en Retrovirus y Sida; Argentina. Gobierno de la Ciudad de Buenos Aires. Hospital General de Agudos "Juan A. Fernández"; Argentina Fil: Coloccini, Romina Soledad. Consejo Nacional de Investigaciones Científicas y Técnicas. Oficina de Coordinación Administrativa Houssay. Instituto de Investigaciones Biomédicas en Retrovirus y Sida. Universidad de Buenos Aires. Facultad de Medicina. Instituto de Investigaciones Biomédicas en Retrovirus y Sida; Argentina Fil: Salido, Jimena Patricia. Consejo Nacional de Investigaciones Científicas y Técnicas. Oficina de Coordinación Administrativa Houssay. Instituto de Investigaciones Biomédicas en Retrovirus y Sida. Universidad de Buenos Aires. Facultad de Medicina. Instituto de Investigaciones Biomédicas en Retrovirus y Sida; Argentina Fil: Trifone, César Ariel. Consejo Nacional de Investigaciones Científicas y Técnicas. Oficina de Coordinación Administrativa Houssay. Instituto de Investigaciones Biomédicas en Retrovirus y Sida. Universidad de Buenos Aires. Facultad de Medicina. Instituto de Investigaciones Biomédicas en Retrovirus y Sida; Argentina Fil: Ruiz, María Julia. Consejo Nacional de Investigaciones Científicas y Técnicas. Oficina de Coordinación Administrativa Houssay. Instituto de Investigaciones Biomédicas en Retrovirus y Sida. Universidad de Buenos Aires. Facultad de Medicina. Instituto de Investigaciones Biomédicas en Retrovirus y Sida; Argentina Fil: Falivene, Juliana. Consejo Nacional de Investigaciones Científicas y Técnicas. Oficina de Coordinación Administrativa Houssay. Instituto de Investigaciones Biomédicas en Retrovirus y Sida. Universidad de Buenos Aires. Facultad de Medicina. Instituto de Investigaciones Biomédicas en Retrovirus y Sida; Argentina Fil: Holgado, María Pía. Consejo Nacional de Investigaciones Científicas y Técnicas. Oficina de Coordinación Administrativa Houssay. Instituto de Investigaciones Biomédicas en Retrovirus y Sida. Universidad de Buenos Aires. Facultad de Medicina. Instituto de Investigaciones Biomédicas en Retrovirus y Sida; Argentina Fil: Caruso, María Paula. Consejo Nacional de Investigaciones Científicas y Técnicas. Oficina de Coordinación Administrativa Houssay. Instituto de Investigaciones Biomédicas en Retrovirus y Sida. Universidad de Buenos Aires. Facultad de Medicina. Instituto de Investigaciones Biomédicas en Retrovirus y Sida; Argentina Fil: Figueroa, María Inés. Gobierno de la Ciudad de Buenos Aires. Hospital General de Agudos "Juan A. Fernández"; Argentina. Fundación Huésped; Argentina Fil: Salomon, Horacio Eduardo. Consejo Nacional de Investigaciones Científicas y Técnicas. Oficina de Coordinación Administrativa Houssay. Instituto de Investigaciones Biomédicas en Retrovirus y Sida. Universidad de Buenos Aires. Facultad de Medicina. Instituto de Investigaciones Biomédicas en Retrovirus y Sida; Argentina Fil: Giavedoni, Luis D.. Texas Biomedical Research Institute; Estados Unidos Fil: Pando, María de los Ángeles. Consejo Nacional de Investigaciones Científicas y Técnicas. Oficina de Coordinación Administrativa Houssay. Instituto de Investigaciones Biomédicas en Retrovirus y Sida. Universidad de Buenos Aires. Facultad de Medicina. Instituto de Investigaciones Biomédicas en Retrovirus y Sida; Argentina Fil: Gherardi, Maria Magdalena. Consejo Nacional de Investigaciones Científicas y Técnicas. Oficina de Coordinación Administrativa Houssay. Instituto de Investigaciones Biomédicas en Retrovirus y Sida. Universidad de Buenos Aires. Facultad de Medicina. Instituto de Investigaciones Biomédicas en Retrovirus y Sida; Argentina Fil: Rabinovich, Roberto Daniel. Consejo Nacional de Investigaciones Científicas y Técnicas. Oficina de Coordinación Administrativa Houssay. Instituto de Investigaciones Biomédicas en Retrovirus y Sida. Universidad de Buenos Aires. Facultad de Medicina. Instituto de Investigaciones Biomédicas en Retrovirus y Sida; Argentina Fil: Pury, Pedro Angel. Universidad Nacional de Córdoba; Argentina Fil: Sued, Omar Gustavo. Fundación Huésped; Argentina
- Published
- 2018
37. A prophylactic multivalent vaccine against different filovirus species is immunogenic and provides protection from lethal infections with Ebolavirus and Marburgvirus species in non-human primates
- Author
-
Luis D. Giavedoni, Robin Steigerwald, Jerome Custers, Chad E. Mire, Mo Weijtens, Ulrike Dirmeier, Trevor Brasel, Roland Zahn, Maria Grazia Pau, Jutta Sytske Hartkoorn-Pasma, Ricardo Carrion, Hanneke Schuitemaker, Jort Vellinga, Ariane Rodriguez, Kerstin Wunderlich, Jay W. Hooper, Ariane Volkmann, Cédric Cheminay, Thomas W. Geisbert, Jean L. Patterson, Benoit Callendret, and Academic Medical Center
- Subjects
0301 basic medicine ,Male ,Physiology ,Humoral Immune Response ,lcsh:Medicine ,Antibody Response ,medicine.disease_cause ,Biochemistry ,chemistry.chemical_compound ,Immune Physiology ,Medicine and Health Sciences ,Public and Occupational Health ,Marburg Virus Disease ,Enzyme-Linked Immunoassays ,lcsh:Science ,Immune Response ,Vaccines ,Multidisciplinary ,Immune System Proteins ,biology ,Viral Vaccine ,Immunogenicity ,Ebolavirus ,Vaccination and Immunization ,Vaccination ,Infectious Diseases ,Female ,Research Article ,Infectious Disease Control ,Immunology ,Research and Analysis Methods ,Virus ,Antibodies ,03 medical and health sciences ,medicine ,Animals ,Antigens ,Immunoassays ,Ebola virus ,lcsh:R ,Immunity ,Biology and Life Sciences ,Proteins ,Correction ,Viral Vaccines ,Hemorrhagic Fever, Ebola ,Marburgvirus ,biology.organism_classification ,Virology ,Macaca fascicularis ,030104 developmental biology ,chemistry ,Humoral Immunity ,Immunologic Techniques ,lcsh:Q ,Preventive Medicine ,Vaccinia - Abstract
The search for a universal filovirus vaccine that provides protection against multiple filovirus species has been prompted by sporadic but highly lethal outbreaks of Ebolavirus and Marburgvirus infections. A good prophylactic vaccine should be able to provide protection to all known filovirus species and as an upside potentially protect from newly emerging virus strains. We investigated the immunogenicity and protection elicited by multivalent vaccines expressing glycoproteins (GP) from Ebola virus (EBOV), Sudan virus (SUDV), Tai Forest virus (TAFV) and Marburg virus (MARV). Immune responses against filovirus GP have been associated with protection from disease. The GP antigens were expressed by adenovirus serotypes 26 and 35 (Ad26 and Ad35) and modified Vaccinia virus Ankara (MVA) vectors, all selected for their strong immunogenicity and good safety profile. Using fully lethal NHP intramuscular challenge models, we assessed different vaccination regimens for immunogenicity and protection from filovirus disease. Heterologous multivalent Ad26-Ad35 prime-boost vaccination regimens could give full protection against MARV (range 75%-100% protection) and EBOV (range 50% to 100%) challenge, and partial protection (75%) against SUDV challenge. Heterologous multivalent Ad26-MVA prime-boost immunization gave full protection against EBOV challenge in a small cohort study. The use of such multivalent vaccines did not show overt immune interference in comparison with monovalent vaccines. Multivalent vaccines induced GP-specific antibody responses and cellular IFNγ responses to each GP expressed by the vaccine, and cross-reactivity to TAFV GP was detected in a trivalent vaccine expressing GP from EBOV, SUDV and MARV. In the EBOV challenge studies, higher humoral EBOV GP-specific immune responses (p = 0.0004) were associated with survival from EBOV challenge and less so for cellular immune responses (p = 0.0320). These results demonstrate that it is feasible to generate a multivalent filovirus vaccine that can protect against lethal infection by multiple members of the filovirus family.
- Published
- 2018
38. Experimental Zika Virus Inoculation in a New World Monkey Model Reproduces Key Features of the Human Infection
- Author
-
Manasi Tamhankar, Tony Li, Guixia Yu, Charles Y. Chiu, Suzette D. Tardif, Laura M. Parodi, Sneha Somasekar, Luis D. Giavedoni, Shigeo Yagi, Claudia Sanchez San Martin, Vida L. Hodara, Jean L. Patterson, and Jerome Bouquet
- Subjects
0301 basic medicine ,Male ,Science ,Article ,Virus ,Zika virus ,Cercopithecus aethiops ,Vaccine Related ,03 medical and health sciences ,Rare Diseases ,Immune system ,biology.animal ,Chlorocebus aethiops ,2.1 Biological and endogenous factors ,2.2 Factors relating to the physical environment ,Medicine ,Animals ,Aetiology ,Viral shedding ,Saliva ,Vero Cells ,Multidisciplinary ,biology ,Transmission (medicine) ,business.industry ,Animal ,Zika Virus Infection ,Prevention ,Marmoset ,Callithrix ,Zika Virus ,biology.organism_classification ,Virology ,3. Good health ,Disease Models, Animal ,Emerging Infectious Diseases ,Infectious Diseases ,Good Health and Well Being ,030104 developmental biology ,Disease Models ,biology.protein ,Immunization ,Antibody ,Infection ,business - Abstract
A monkey model of Zika virus (ZIKV) infection is urgently needed to better understand transmission and pathogenesis, given its proven association with fetal brain defects in pregnant women and acute neurological illness. Here we experimentally infected 4 male marmosets with ZIKV (prototype 1947 African strain) and monitored them clinically with sampling of various body fluids and tissues for nearly 3 months. We show that the course of acute infection with ZIKV in these New World monkeys resembles the human illness in many respects, including (1) lack of apparent clinical symptoms in most cases, (2) persistence of the virus in body fluids such as semen and saliva for longer periods of time than in serum, and (3) generation of neutralizing antibodies as well as an antiviral immunological host response. Importantly, ZIKV-infected saliva samples (in addition to serum) were found to be infectious, suggesting potential capacity for viral transmission by the oral route. Re-challenge of a previously infected marmoset with a contemporary outbreak strain SPH2015 from Brazil resulted in continued protection against infection, no viral shedding, and boosting of the immune response. Given the key similarities to human infection, a marmoset model of ZIKV infection may be useful for testing of new drugs and vaccines.
- Published
- 2017
39. Evaluation of Different Parameters of Humoral and Cellular Immune Responses in HIV Serodiscordant Heterosexual Couples: Humoral Response Potentially Implicated in Modulating Transmission Rates
- Author
-
G. Damilano, Natalia Laufer, María Magdalena Gherardi, Gabriela Turk, Jimena Salido, María Julia Ruiz, Omar Sued, Luis D. Giavedoni, Yanina Alexandra Ghiglione, Lorena Abusamra, César Ariel Trifone, Ana M. Rodríguez, Cintia Cevallos, and Horacio Salomón
- Subjects
0301 basic medicine ,Immunoglobulin A ,Chemokine ,HIV-1 transmission ,Otras Ciencias Biológicas ,lcsh:Medicine ,Context (language use) ,General Biochemistry, Genetics and Molecular Biology ,Immunoglobulin G ,purl.org/becyt/ford/1 [https] ,Ciencias Biológicas ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,030212 general & internal medicine ,purl.org/becyt/ford/1.6 [https] ,Adcc ,Antibody-dependent cell-mediated cytotoxicity ,lcsh:R5-920 ,biology ,Serodiscordant couples ,lcsh:R ,Hiv-1 ,Hiv-1 Transmission ,virus diseases ,General Medicine ,Virology ,3. Good health ,030104 developmental biology ,Serodiscordant ,Immunology ,HIV-1 ,biology.protein ,Serodiscordant Couples ,Antibody ,ADCC ,lcsh:Medicine (General) ,CIENCIAS NATURALES Y EXACTAS - Abstract
As the HIV/AIDS pandemic still progresses, understanding the mechanisms governing viral transmission as well as protection from HIV acquisition is fundamental. In this context, cohorts of HIV serodiscordant heterosexual couples (SDC) represent a unique tool. The present study was aimed to evaluate specific parameters of innate, cellular and humoral immune responses in SDC. Specifically, plasma levels of cytokines and chemokines, HIV-specific T-cell responses, gp120-specific IgG and IgA antibodies, and HIV-specific antibody-dependent cellular cytotoxicity (ADCC) activity were assessed in nine HIV-exposed seronegative individuals (ESN) and their corresponding HIV seropositive partners (HIV+-P), in eighteen chronically infected HIV subjects (C), nine chronically infected subjects known to be HIV transmitters (CT) and ten healthy HIV− donors (HD). Very low magnitude HIV-specific cellular responses were found in two out of six ESN. Interestingly, HIV+-P had the highest ADCC magnitude, the lowest IgA levels and the highest IgG/IgA ratio, all compared to CT. Positive correlations between CD4+ T-cell counts and both IgG/IgA ratios and %ADCC killing uniquely distinguished HIV+-P. Additionally, evidence of IgA interference with ADCC responses from HIV+-P and CT is provided. These data suggest for the first time a potential role of ADCC and/or gp120-specific IgG/IgA balance in modulating heterosexual transmission. In sum, this study provides key information to understand the host factors that influence viral transmission, which should be considered in both the development of prophylactic vaccines and novel immunotherapies for HIV-1 infection. Fil: Ruiz, María Julia. Fil: Salido, Jimena. Fil: Abusamra, Lorena. Fil: Ghiglione, Yanina. Fil: Cevallos, Cintia. Fil: Damilano, Gabriel. Fil: Rodriguez, Ana María. Fil: Trifone, César Ariel. Universidad de Buenos Aires; Argentina. Universidad de Buenos Aires; Argentina Fil: Laufer, Natalia Lorna. Universidad de Buenos Aires; Argentina. Universidad de Buenos Aires; Argentina Fil: Giavedoni, Luis D.. Universidad de Buenos Aires; Argentina. Universidad de Buenos Aires; Argentina Fil: Sued, Omar. Gobierno de la Ciudad de Buenos Aires. Hospital General de Agudos ; Argentina. Gobierno de la Ciudad de Buenos Aires. Hospital General de Agudos ; Argentina Fil: Salomón, Horacio. Fil: Turk, Gabriela Julia Ana.
- Published
- 2017
40. Zika virus pathogenesis in rhesus macaques is unaffected by pre-existing immunity to dengue virus
- Author
-
Mariah Hassert, James D. Brien, Lorna Cruz, Aravinda M. de Silva, Laura J. White, Vida L. Hodara, Petraleigh Pantoja, Crisanta Serrano, Carlos A. Sariol, Idia V. Rodríguez, Olga González, Luis D. Giavedoni, Melween I. Martínez, Teresa Arana, Erick X. Pérez-Guzmán, and Amelia K. Pinto
- Subjects
0301 basic medicine ,Male ,Science ,viruses ,030231 tropical medicine ,General Physics and Astronomy ,Viremia ,Dengue virus ,Cross Reactions ,medicine.disease_cause ,Antibodies, Viral ,General Biochemistry, Genetics and Molecular Biology ,Virus ,Article ,Zika virus ,Dengue ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Viral Envelope Proteins ,Immunity ,medicine ,Animals ,Humans ,Antibody-dependent enhancement ,Multidisciplinary ,biology ,Zika Virus Infection ,Immune Sera ,virus diseases ,General Chemistry ,Zika Virus ,biochemical phenomena, metabolism, and nutrition ,Dengue Virus ,biology.organism_classification ,medicine.disease ,Virology ,Antibodies, Neutralizing ,Antibody-Dependent Enhancement ,Macaca mulatta ,3. Good health ,030104 developmental biology ,Immunology ,Models, Animal ,biology.protein ,Cytokines ,Antibody ,K562 Cells - Abstract
Zika virus (ZIKV) is a re-emerging virus that has recently spread into dengue virus (DENV) endemic regions and cross-reactive antibodies (Abs) could potentially affect ZIKV pathogenesis. Using DENV-immune serum, it has been shown in vitro that antibody-dependent enhancement (ADE) of ZIKV infection can occur. Here we study the effects of pre-existing DENV immunity on ZIKV infection in vivo. We infect two cohorts of rhesus macaques with ZIKV; one cohort has been exposed to DENV 2.8 years earlier and a second control cohort is naïve to flaviviral infection. Our results, while confirming ADE in vitro, suggest that pre-existing DENV immunity does not result in more severe ZIKV disease. Rather our results show a reduction in the number of days of ZIKV viremia compared to naïve macaques and that the previous exposure to DENV may result in modulation of the immune response without resulting in enhancement of ZIKV pathogenesis., Antibodies against dengue virus (DENV) can increase Zika virus (ZIKV) infection in vitro, but their role in vivo remains largely unknown. Here, the authors show that pre-existing immunity from a 2.8 years earlier DENV infection does not affect ZIKV pathogenesis in macaques but instead shortens Zika viremia.
- Published
- 2017
41. Experimental Zika Virus Infection in a New World Monkey Model Reproduces Key Features of the Human Disease
- Author
-
Guixia Yu, Charles Y. Chiu, Claudia Sanchez San Martin, Jean L. Patterson, Sneha Somasekar, Manasi Tamhankar, Tony Li, Luis D. Giavedoni, Suzette D. Tardif, Vida L. Hodara, Jerome Bouquet, Shigeo Yagi, and Laura M. Parodi
- Subjects
Infectivity ,0303 health sciences ,biology ,biology.organism_classification ,Virology ,Virus ,3. Good health ,Zika virus ,Pathogenesis ,03 medical and health sciences ,Flavivirus ,0302 clinical medicine ,Immune system ,Interferon ,Immunology ,biology.protein ,medicine ,030212 general & internal medicine ,Neutralizing antibody ,030304 developmental biology ,medicine.drug - Abstract
Human infections by Zika virus (ZIKV), a mosquito-borne flavivirus, are associated with a current widespread outbreak in the Americas, and have been associated with neurological complications and adverse fetal outcomes such as microcephaly in pregnant women. A suitable non-human primate model is urgently needed. To evaluate ZIKV infectivity, pathogenesis, and persistence, we inoculated 4 marmosets with ZIKV and followed them by clinical monitoring and serial sampling of body fluids for up to 11 weeks. We found that marmosets experimentally infected with ZIKV reproduced key features of the human disease, including (1) asymptomatic infection, (2) brief period of detectable virus in serum (
- Published
- 2017
42. Cross-sectional comparison of health-span phenotypes in young versus geriatric marmosets
- Author
-
Jessica Adams, Luis D. Giavedoni, Kimberley A. Phillips, Corinna N. Ross, Vida L. Hodara, Anna D. Rigodanzo, Balakuntalam S. Kasinath, Suzette D. Tardif, Edward J. Dick, and Olga Gonzalez
- Subjects
Male ,Cardiac function curve ,Aging ,endocrine system ,animal structures ,Health Status ,Physiology ,Article ,Human health ,Cognition ,Biomarkers of aging ,biology.animal ,Animals ,Homeostasis ,Medicine ,Mobility Limitation ,Ecology, Evolution, Behavior and Systematics ,Health span ,biology ,business.industry ,Marmoset ,Callithrix ,Phenotype ,body regions ,Cross-Sectional Studies ,Models, Animal ,Female ,Animal Science and Zoology ,business - Abstract
The development of the marmoset as a translational model for healthspan and lifespan studies relies on the characterization of health parameters in young and geriatric marmosets. This cross-sectional study examined health phenotypes in marmosets for five domains of interest for human health and aging: mobility, cognition, metabolism, homeostasis, and immune function. Geriatric marmosets were found to have significant executive function impairment when compared to young animals. While geriatric animals did not show gross abnormalities in mobility and measures of locomotion, their types of movement were altered from young animals. Geriatric marmosets had alterations in cardiac function, with significantly increased mean arterial pressures; metabolism, with significantly lower VO(2); and suppressed immune function. Further, this study sought to characterize and describe histopathology for both young and geriatric healthy marmosets. Overall this study provides a characterization of health parameters for young and geriatric marmosets which will greatly enhance future aging and interventional testing in marmosets.
- Published
- 2019
43. Comparative Characterization of Transfection- and Infection-Derived Simian Immunodeficiency Virus Challenge Stocks for In Vivo Nonhuman Primate Studies
- Author
-
Ronald C. Desrosiers, Brandon F. Keele, Elena Chertova, Gregory Q. Del Prete, Carolyn Reid, Christopher J. Miller, Matthew Scarlotta, Luis D. Giavedoni, Laura Newman, Laura M. Parodi, Dan H. Barouch, James D. Roser, Ranajit Pal, Michael Piatak, Jeffrey D. Lifson, Kelli Oswald, David H. O’Connor, and Preston A. Marx
- Subjects
Virus Cultivation ,viruses ,Immunology ,Simian Acquired Immunodeficiency Syndrome ,Biology ,Transfection ,medicine.disease_cause ,Microbiology ,Virus ,In vivo ,Virology ,medicine ,Animals ,Infectivity ,Primate Diseases ,Genetic Variation ,Sequence Analysis, DNA ,Viral Load ,Simian immunodeficiency virus ,Titer ,Genetic Diversity and Evolution ,Insect Science ,Simian Immunodeficiency Virus ,Viral load - Abstract
Simian immunodeficiency virus (SIV) stocks for in vivo nonhuman primate models of AIDS are typically generated by transfection of 293T cells with molecularly cloned viral genomes or by expansion in productively infected T cells. Although titers of stocks are determined for infectivity in vitro prior to in vivo inoculation, virus production methods may differentially affect stock features that are not routinely analyzed but may impact in vivo infectivity, mucosal transmissibility, and early infection events. We performed a detailed analysis of nine SIV stocks, comprising five infection-derived SIVmac251 viral swarm stocks and paired infection- and transfected-293T-cell-derived stocks of both SIVmac239 and SIVmac766. Representative stocks were evaluated for (i) virus content, (ii) infectious titer, (iii) sequence diversity and polymorphism frequency by single-genome amplification and 454 pyrosequencing, (iv) virion-associated Env content, and (v) cytokine and chemokine content by 36-plex Luminex analysis. Regardless of production method, all stocks had comparable particle/infectivity ratios, with the transfected-293T stocks possessing the highest overall virus content and infectivity titers despite containing markedly lower levels of virion-associated Env than infection-derived viruses. Transfected-293T stocks also contained fewer and lower levels of cytokines and chemokines than infection-derived stocks, which had elevated levels of multiple analytes, with substantial variability among stocks. Sequencing of the infection-derived SIVmac251 stocks revealed variable levels of viral diversity between stocks, with evidence of stock-specific selection and expansion of unique viral lineages. These analyses suggest that there may be underappreciated features of SIV in vivo challenge stocks with the potential to impact early infection events, which may merit consideration when selecting virus stocks for in vivo studies.
- Published
- 2013
44. Impact of Mucosal Inflammation on Oral Simian Immunodeficiency Virus Transmission
- Author
-
Luis D. Giavedoni, Laura M. Parodi, David P. Cappelli, Donald L. Sodora, Vida L. Hodara, Valerie Sexton, Hui Ling Chen, Lianrui Chu, and Lisa M. Smith
- Subjects
Male ,viruses ,animal diseases ,Immunology ,Simian Acquired Immunodeficiency Syndrome ,Alpha interferon ,Biology ,medicine.disease_cause ,Microbiology ,Proinflammatory cytokine ,Pathogenesis ,Gingivitis ,Immune system ,Virology ,medicine ,Animals ,Periodontitis ,Moderate gingivitis ,Mouth Mucosa ,virus diseases ,Simian immunodeficiency virus ,medicine.disease ,Macaca mulatta ,Insect Science ,Cytokines ,Pathogenesis and Immunity ,Simian Immunodeficiency Virus ,medicine.symptom - Abstract
Mucosal tissues are the primary route of transmission for most respiratory and sexually transmitted diseases, including human immunodeficiency virus (HIV). There is epidemiological evidence that genital mucosal inflammation leads to enhanced HIV type 1 (HIV-1) transmission. The objective of this study was to assess the influence of periodontal inflammation on oral HIV transmission using a nonhuman primate model of teeth ligature-induced periodontitis. Simian immunodeficiency virus (SIV) was nontraumatically applied to the gingiva after moderate gingivitis was identified through clinical and immunologic analyses (presence of inflammatory cytokines). Overall oral SIV infection rates were similar in the gingivitis-induced and control groups (5 infections following 12 SIV administrations for each), although more macaques were infected with multiple viral variants in the gingivitis group. SIV infection also affected the levels of antiviral and inflammatory cytokines in the gingival crevicular fluid, and a synergistic effect was observed, with alpha interferon and interferon-inducible protein 10 undergoing significant elevations following SIV infection in macaques with gingivitis compared to controls. These increases in antiviral and inflammatory immune modulators in the SIV-infected gingivitis macaques could also be observed in blood plasma, although the effects at both compartments were generally restricted to the acute phase of the infection. In conclusion, while moderate gingivitis was not associated with increased susceptibility to oral SIV infection, it resulted in elevated levels of cytokines in the oral mucosa and plasma of the SIV-infected macaques. These findings suggest a synergy between mucosal inflammation and SIV infection, creating an immune milieu that impacts the early stages of the SIV infection with potential implications for long-term pathogenesis.
- Published
- 2013
45. Increases in NKG2C Expression on T Cells and Higher Levels of Circulating CD8
- Author
-
Vida L, Hodara, Laura M, Parodi, M Shannon, Keckler, and Luis D, Giavedoni
- Subjects
CD4-Positive T-Lymphocytes ,B-Lymphocytes ,Vaccines ,CD8 Antigens ,SAIDS Vaccines ,Simian Acquired Immunodeficiency Syndrome ,Animals ,Gene Expression ,CD8-Positive T-Lymphocytes ,NK Cell Lectin-Like Receptor Subfamily C ,Vaccines, Attenuated ,Macaca mulatta - Abstract
Vaccines based on live attenuated viruses are highly effective immunogens in the simian immunodeficiency virus (SIV)/rhesus macaque animal model and offer the possibility of studying correlates of protection against infection with virulent virus. We utilized a tether system for studying, in naive macaques and animals vaccinated with a live-attenuated vaccine, the acute events after challenge with pathogenic SIV. This approach allowed for the frequent sampling of small blood volumes without sedation or restraining of the animals, thus reducing the confounding effect of sampling stress. Before challenge, vaccinated animals presented significantly higher levels of proliferating and activated B cells than naive macaques, which were manifested by high expression of CD8 on B cells. After SIV challenge, the only changes observed in protected vaccinated macaques were significant increases in expression of the NK marker NKG2C on CD4 and CD8 T cells. We also identified that infection of naive macaques with SIV resulted in a transient peak of expression of CD20 on CD8 T cells and a constant rise in the number of B cells expressing CD8. Finally, analysis of a larger cohort of vaccinated animals identified that, even when circulating levels of vaccine virus are below the limit of detection, live attenuated vaccines induce systemic increases of IP-10 and perforin. These studies indicate that components of both the innate and adaptive immune systems of animals inoculated with a live-attenuated SIV vaccine respond to and control infection with virulent virus. Persistence of the vaccine virus in tissues may explain the elevated cytokine and B-cell activation levels. In addition, our report underpins the utility of the tether system for the intensive study of acute immune responses to viral infections.
- Published
- 2016
46. A Non-Human Primate Model of Severe Pneumococcal Pneumonia
- Author
-
Carlos J. Orihuela, Luis F. Reyes, Norberto Gonzalez-Juarbe, Antonio Anzueto, Robert E. Shade, Melissa De La Garza, Anukul T. Shenoy, Jacqueline J. Coalson, Cecilia A. Hinojosa, Marcos I. Restrepo, Nilam J. Soni, Luis D. Giavedoni, Ryan P. Gilley, Julio Noda, and Vicki T. Winter
- Subjects
Pulmonology ,Physiology ,Biopsy ,lcsh:Medicine ,Monkeys ,Pathology and Laboratory Medicine ,medicine.disease_cause ,Severity of Illness Index ,Diagnostic Radiology ,0302 clinical medicine ,Baboons ,Immune Physiology ,Ultrasound Imaging ,Medicine and Health Sciences ,030212 general & internal medicine ,Leukocytosis ,lcsh:Science ,Lung ,Ultrasonography ,Mammals ,Innate Immune System ,Multidisciplinary ,Radiology and Imaging ,Chemotaxis ,Pneumococcus ,Pulmonary Imaging ,Bacterial Pathogens ,3. Good health ,Cell Motility ,Phenotype ,Streptococcus pneumoniae ,medicine.anatomical_structure ,Medical Microbiology ,Vertebrates ,Pneumococcal pneumonia ,Cytokines ,Pathogens ,Chemokines ,Inflammation Mediators ,medicine.symptom ,Research Article ,Primates ,medicine.medical_specialty ,Imaging Techniques ,Immunology ,Biology ,Research and Analysis Methods ,Microbiology ,Tachypnea ,03 medical and health sciences ,Diagnostic Medicine ,Old World monkeys ,medicine ,Animals ,Microbial Pathogens ,Biology and life sciences ,Bacteria ,lcsh:R ,Organisms ,Hemodynamics ,Streptococcus ,Pneumonia ,Cell Biology ,Molecular Development ,Pneumonia, Pneumococcal ,medicine.disease ,respiratory tract diseases ,Disease Models, Animal ,030228 respiratory system ,Immune System ,Bacteremia ,Amniotes ,Histopathology ,lcsh:Q ,Biomarkers ,Developmental Biology ,Papio - Abstract
Rationale Streptococcus pneumoniae is the leading cause of community-acquired pneumonia and infectious death in adults worldwide. A non-human primate model is needed to study the molecular mechanisms that underlie the development of severe pneumonia, identify diagnostic tools, explore potential therapeutic targets, and test clinical interventions during pneumococcal pneumonia. Objective To develop a non-human primate model of pneumococcal pneumonia. Methods Seven adult baboons (Papio cynocephalus) were surgically tethered to a continuous monitoring system that recorded heart rate, temperature, and electrocardiography. Animals were inoculated with 109 colony-forming units of S. pneumoniae using bronchoscopy. Three baboons were rescued with intravenous ampicillin therapy. Pneumonia was diagnosed using lung ultrasonography and ex vivo confirmation by histopathology and immunodetection of pneumococcal capsule. Organ failure, using serum biomarkers and quantification of bacteremia, was assessed daily. Results Challenged animals developed signs and symptoms of pneumonia 4 days after infection. Infection was characterized by the presence of cough, tachypnea, dyspnea, tachycardia and fever. All animals developed leukocytosis and bacteremia 24 hours after infection. A severe inflammatory reaction was detected by elevation of serum cytokines, including Interleukin (IL)1Ra, IL-6, and IL-8, after infection. Lung ultrasonography precisely detected the lobes with pneumonia that were later confirmed by pathological analysis. Lung pathology positively correlated with disease severity. Antimicrobial therapy rapidly reversed symptomology and reduced serum cytokines. Conclusions We have developed a novel animal model for severe pneumococcal pneumonia that mimics the clinical presentation, inflammatory response, and infection kinetics seen in humans. This is a novel model to test vaccines and treatments, measure biomarkers to diagnose pneumonia, and predict outcomes.
- Published
- 2016
47. Differential Innate Immune Responses to Low or High Dose Oral SIV Challenge in Rhesus Macaques
- Author
-
Luis D. Giavedoni, Welkin E. Johnson, Donald L. Sodora, Melanie A. Gasper, Guido Silvestri, Jeffrey M. Milush, Hui Ling Chen, Vasudha Sundaravaradan, and Andre Durudas
- Subjects
DNA, Complementary ,Genotype ,T-Lymphocytes ,viruses ,animal diseases ,CD14 ,Simian Acquired Immunodeficiency Syndrome ,Administration, Oral ,Biology ,Real-Time Polymerase Chain Reaction ,medicine.disease_cause ,Article ,Immune system ,Antigen ,Antigens, CD ,Immunity ,Virology ,medicine ,Animals ,RNA, Messenger ,HIV vaccine ,Blood Cells ,Innate immune system ,virus diseases ,Viral Load ,Simian immunodeficiency virus ,Macaca mulatta ,Immunity, Innate ,Up-Regulation ,Disease Models, Animal ,Infectious Diseases ,Immunology ,Leukocytes, Mononuclear ,Cytokines ,RNA, Viral ,Simian Immunodeficiency Virus ,Lymph Nodes ,Viral load - Abstract
Mucosal transmission of HIV predominately occurs during sexual intercourse or breast-feeding and generally results in a successful infection from just one or few founder virions. Here we assessed the impact of viral inoculum size on both viral and immune events within two groups of Rhesus macaques that were non-traumatically, orally inoculated with either multiple low (1000 to 4000 TCID(50)) or high (100,000 TCID(50)) doses of SIV. In agreement with previous studies, more diverse SIV variants were observed in macaques following infection with high dose oral SIV compared to a low dose challenge. In peripheral blood cells, the immune gene transcript levels of CXCL9, IFNγ, TNFα and IL10 remained similar to uninfected macaques. In contrast, OAS and CXCL10 were upregulated following SIV infection in both the high and low dosed macaques, with a more rapid kinetics (detectable by 7 days) following the high SIV dose challenge. In peripheral lymph nodes, an increase in CXCL10 was observed irrespective of viral dose while CXCL9 and OAS were differentially regulated in the two SIV dosed groups. Magnetic bead sorting of CD3+, CD14+ and CD3- /CD14- cells from peripheral blood identified the increase in OAS expression primarily within CD14+ monocytes, whereas the CXCL10 expression was primarily in CD3+ T cells. These findings provide insights into the impact of SIV challenge dose on viral and innate immune factors, which has the potential to inform future SIV/HIV vaccine efficacy trials in which vaccinated hosts have the potential to be infected with a range of viral challenge doses.
- Published
- 2011
48. The baboon as a nonhuman primate model for the study of natural Zika virus infection
- Author
-
L. Parodi, Jessica Callery, Luis D. Giavedoni, Laura A. Cox, Vida L. Hodara, Patrice A. Frost, Elizabeth Clemmons, Veronica Obregon-Perko, Jean L. Patterson, and J. Glenn
- Subjects
Microbiology (medical) ,Infectious Diseases ,biology ,biology.animal ,General Medicine ,biology.organism_classification ,Virology ,Nonhuman primate ,Zika virus ,Baboon - Published
- 2018
49. Novel Application of Nonhuman Primate Tethering System for Evaluation of Acute Phase SIVmac251 Infection in Rhesus Macaques (Macaca mulatta)
- Author
-
Vida L. Hodara, M. Shannon Keckler, Laura M. Parodi, and Luis D. Giavedoni
- Subjects
Male ,Restraint, Physical ,Immunology ,Simian Acquired Immunodeficiency Syndrome ,Epitopes, T-Lymphocyte ,CD8-Positive T-Lymphocytes ,Biology ,Vaccines, Attenuated ,medicine.disease_cause ,Catheters, Indwelling ,Immune system ,Immunity ,Virology ,medicine ,Animals ,Viral Regulatory and Accessory Proteins ,Acute-Phase Reaction ,Blood Specimen Collection ,Immunity, Cellular ,Attenuated vaccine ,Histocompatibility Antigens Class I ,Vaccination ,Viral Vaccines ,Viral Load ,Simian immunodeficiency virus ,Macaca mulatta ,CTL ,Haplotypes ,Antibody Formation ,Molecular Medicine ,Female ,Simian Immunodeficiency Virus ,Viral load ,Blood sampling - Abstract
Infection of rhesus macaques with simian immunodeficiency virus (SIV) is the preferred animal model for the development and testing of human immunodeficiency virus (HIV) vaccines, and animals protected from SIV challenge by live attenuated vaccines are an invaluable tool for determining immune correlates of protection. The acute phase of SIV infection, in which immune responses are most critical for slowing disease progression, occurs within the first 4 weeks of exposure. The small window of time available for observing critical immune responses makes obtaining adequate blood samples with sufficient frequency difficult. This study is the first to apply a previously reported nonhuman primate (NHP) tether system to study viral immunology. The use of the tether allows for frequent blood sampling without using restraints or sedation, thereby reducing the potentially confounding physiological changes induced by stress. We performed comparative analysis of acute phase immune responses in vaccinated and unvaccinated animals challenged with SIV-mac251. Our results demonstrate live attenuated vaccine-induced protection, which is associated with small increases in the cytotoxic T-cell (CTL) response to immunodominant epitopes, but not with increases in antibody titers. Additionally, vaccination was shown to establish a pool of antigen-specific CD8+ memory cells available for expansion after challenge. The confirmatory nature of these data indicates the validity of using the tether system for evaluation of acute phase anti-SIV responses and can be applied to the study of immune responses in other viral infections in which frequent sampling in small windows of time would be useful.
- Published
- 2007
50. Transduction with Human Telomerase Reverse Transcriptase Immortalizes A Rhesus Macaque CD8+T Cell Clone with Maintenance of Surface Marker Phenotype And Function
- Author
-
Jeffrey D. Lifson, David E. Ott, Luis D. Giavedoni, Claes Ohlen, Hanne Andersen, Matthew T. Trivett, Eugene V. Barsov, and Charles M. Trubey
- Subjects
Telomerase ,Cellular differentiation ,T cell ,Immunology ,Streptamer ,CD8-Positive T-Lymphocytes ,Biology ,Cell Line ,Transduction, Genetic ,Virology ,medicine ,Animals ,Humans ,Cytotoxic T cell ,Telomerase reverse transcriptase ,Macaca mulatta ,Clone Cells ,Cell biology ,Telomere ,Infectious Diseases ,medicine.anatomical_structure ,Cell culture ,Female ,Simian Immunodeficiency Virus ,T-Lymphocytes, Cytotoxic - Abstract
T cell lines and clones play a key role in basic studies of cellular immunology, and are also finding applications in adoptive immunotherapy. However, with proliferative expansion, T cells ultimately undergo cellular senescence and death, so that long-term culture of T cell clones is difficult to achieve. Expression of telomerase reverse transcriptase (TERT) in differentiated cells can maintain telomere length over many cell divisions, preventing senescence. We used a retroviral vector that expresses the human TERT (hTERT) gene to transduce a rhesus macaque-derived CD8(+) T cell clone specific for the MamuA*01-restricted immunodominant SIV gag epitope CM9. Extensive in vitro characterization revealed that the untransduced parental cells and the hTERT-transduced cells displayed comparable proliferation capacity, effector memory surface marker profiles, cytolytic activities, and cytokine profiles following antigen stimulation. The hTERT-transduced cells showed improved survival compared to parallel nontransduced cultures during in vitro propagation in long-term culture. Such immortalized T cells may be useful as a source of consistent controls for in vitro assays of cellular immune function, and as a potentially important reagent for autologous adoptive cellular immunotherapy studies in macaques.
- Published
- 2007
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.