111 results on '"Shelby L O'Connor"'
Search Results
2. Supplementary Figure S1 from Bortezomib-Resistant Nuclear Factor-κB Activity in Multiple Myeloma Cells
- Author
-
Shigeki Miyamoto, KyungMann Kim, Brad S. Kahl, Catherine P. Leith, Martha Raschko, Jae E. Werndli, Jihoon Kim, Shelby L. O'Connor, Natalie S. Callander, and Stephanie Markovina
- Abstract
Supplementary Figure S1 from Bortezomib-Resistant Nuclear Factor-κB Activity in Multiple Myeloma Cells
- Published
- 2023
- Full Text
- View/download PDF
3. IL-15 Superagonist N-803 Enhances IFN-γ Production of MAIT Cells in SIV
- Author
-
Amy L, Ellis-Connell, Alexis J, Balgeman, Nadean M, Kannal, Karigynn, Hansen Chaimson, Anna, Batchenkova, Jeffrey T, Safrit, and Shelby L, O'Connor
- Subjects
Interferon-gamma ,Ki-67 Antigen ,Microbial Immunity and Vaccines ,Leukocytes, Mononuclear ,Animals ,Macaca mulatta ,Mucosal-Associated Invariant T Cells ,Granzymes - Abstract
Mucosal associated invariant T (MAIT) cells are innate T cells that recognize bacterial metabolites and secrete cytokines and cytolytic enzymes to destroy infected target cells. This makes MAIT cells promising targets for immunotherapy to combat bacterial infections. Here, we analyzed the effects of an immunotherapeutic agent, the IL-15 superagonist N-803, on MAIT cell activation, trafficking, and cytolytic function in macaques. We found that N-803 could activate MAIT cells in vitro and increase their ability to produce IFN-γ in response to bacterial stimulation. To expand upon this, we examined the phenotypes and functions of MAIT cells present in samples collected from PBMC, airways (bronchoalveolar lavage [BAL] fluid), and lymph nodes (LN) from rhesus macaques that were treated in vivo with N-803. N-803 treatment led to a transient 6 to 7-fold decrease in the total number of MAIT cells in the peripheral blood, relative to pre N-803 time points. Concurrent with the decrease in cells in the peripheral blood, we observed a rapid decline in the frequency of CXCR3(+)CCR6(+) MAITs. This corresponded with an increase in the frequency of CCR6(+) MAITs in the BAL fluid, and higher frequencies of ki-67(+) and granzyme B(+) MAITs in the blood, LN, and BAL fluid. Finally, N-803 improved the ability of MAIT cells collected from PBMC and airways to produce IFN-γ in response to bacterial stimulation. Overall, N-803 shows the potential to transiently alter the phenotypes and functions of MAIT cells, which could be combined with other strategies to combat bacterial infections.
- Published
- 2023
4. Control of Simian Immunodeficiency Virus Infection in Prophylactically Vaccinated, Antiretroviral Treatment-Naive Macaques Is Required for the Most Efficacious CD8 T Cell Response during Treatment with the Interleukin-15 Superagonist N-803
- Author
-
Amy L. Ellis-Connell, Alexis J. Balgeman, Olivia E. Harwood, Ryan V. Moriarty, Jeffrey T. Safrit, Andrea M. Weiler, Thomas C. Friedrich, and Shelby L. O’Connor
- Subjects
Interleukin-15 ,Immunology ,Simian Acquired Immunodeficiency Syndrome ,HIV Infections ,DNA ,CD8-Positive T-Lymphocytes ,Viral Load ,Microbiology ,Macaca mulatta ,Interleukin-12 ,Granzymes ,Ki-67 Antigen ,Anti-Retroviral Agents ,Virology ,Insect Science ,Vaccines and Antiviral Agents ,Animals ,Simian Immunodeficiency Virus ,Viremia - Abstract
The IL-15 superagonist N-803 has been shown to enhance the function of CD8 T cells and NK cells. We previously found that in a subset of vaccinated, ART-naive, SIV(+) rhesus macaques, N-803 treatment led to a rapid but transient decline in plasma viremia that positively correlated with an increase in the frequency of CD8 T cells. Here, we tested the hypothesis that prophylactic vaccination was required for the N-803 mediated suppression of SIV plasma viremia. We either vaccinated rhesus macaques with a DNA prime/Ad5 boost regimen using vectors expressing SIVmac239 gag with or without a plasmid expressing IL-12 or left them unvaccinated. The animals were then intravenously infected with SIVmac239M. 6 months after infection, the animals were treated with N-803. We found no differences in the control of plasma viremia during N-803 treatment between vaccinated and unvaccinated macaques. Interestingly, when we divided the SIV(+) animals based on their plasma viral load set-points prior to the N-803 treatment, N-803 increased the frequency of SIV-specific T cells expressing ki-67(+) and granzyme B(+) in animals with low plasma viremia (10(4) copies/mL; SIV noncontrollers). In addition, Gag-specific CD8 T cells from the SIV(+) controllers had a greater increase in CD8(+)CD107a(+) T cells in ex vivo functional assays than did the SIV(+) noncontrollers. Overall, our results indicate that N-803 is most effective in SIV(+) animals with a preexisting immunological ability to control SIV replication. IMPORTANCE N-803 is a drug that boosts the immune cells involved in combating HIV/SIV infection. Here, we found that in SIV(+) rhesus macaques that were not on antiretroviral therapy, N-803 increased the proliferation and potential capacity for killing of the SIV-specific immune cells to a greater degree in animals that spontaneously controlled SIV than in animals that did not control SIV. Understanding the mechanism of how N-803 might function differently in individuals that control HIV/SIV (for example, individuals on antiretroviral therapy or spontaneous controllers) compared to settings where HIV/SIV are not controlled, could impact the efficacy of N-803 utilization in the field of HIV cure.
- Published
- 2023
5. Data from Bortezomib-Resistant Nuclear Factor-κB Activity in Multiple Myeloma Cells
- Author
-
Shigeki Miyamoto, KyungMann Kim, Brad S. Kahl, Catherine P. Leith, Martha Raschko, Jae E. Werndli, Jihoon Kim, Shelby L. O'Connor, Natalie S. Callander, and Stephanie Markovina
- Abstract
Bortezomib (Velcade/PS341), a proteasome inhibitor used in the treatment of multiple myeloma (MM), can inhibit activation of nuclear factor-κB (NF-κB), a family of transcription factors often deregulated and constitutively activated in primary MM cells. NF-κB can be activated via several distinct mechanisms, including the proteasome inhibitor–resistant (PIR) pathway. It remains unknown what fraction of primary MM cells harbor constitutive NF-κB activity maintained by proteasome-dependent mechanisms. Here, we report an unexpected finding that constitutive NF-κB activity in 10 of 14 primary MM samples analyzed is refractory to inhibition by bortezomib. Moreover, when MM cells were cocultured with MM patient-derived bone marrow stromal cells (BMSC), microenvironment components critical for MM growth and survival, further increases in NF-κB activity were observed that were also refractory to bortezomib. Similarly, MM-BMSCs caused PIR NF-κB activation in the RPMI8226 MM cell line, leading to increased NF-κB–dependent transcription and resistance to bortezomib-induced apoptosis. Our findings show that primary MM cells frequently harbor PIR NF-κB activity that is further enhanced by the presence of patient-derived BMSCs. They also suggest that this activity is likely relevant to the drug resistance development in some patients. Further elucidation of the mechanism of PIR NF-κB regulation could lead to the identification of novel diagnostic biomarkers and/or therapeutic targets for MM treatment. (Mol Cancer Res 2008;6(8):1356–64)
- Published
- 2023
- Full Text
- View/download PDF
6. Gain without pain: Adaptation and increased virulence of Zika virus in vertebrate host without fitness cost in mosquito vector
- Author
-
Anna S. Jaeger, Jeffrey Marano, Kasen Riemersma, David Castañeda, Elise Pritchard, Julia Pritchard, Ellie K. Bohm, John J. Baczenas, Shelby L. O’Connor, James Weger-Lucarelli, Thomas C. Friedrich, and Matthew T. Aliota
- Subjects
Article - Abstract
Zika virus (ZIKV) is now in a post-pandemic period, for which the potential for re-emergence and future spread is unknown. Adding to this uncertainty is the unique capacity of ZIKV to directly transmit between humans via sexual transmission. Recently, we demonstrated that direct transmission of ZIKV between vertebrate hosts leads to rapid adaptation resulting in enhanced virulence in mice and the emergence of three amino acid substitutions (NS2A-A117V, NS2A- A117T, and NS4A-E19G) shared among all vertebrate-passaged lineages. Here, we further characterized these host-adapted viruses and found that vertebrate-passaged viruses also have enhanced transmission potential in mosquitoes. To understand the contribution of genetic changes to the enhanced virulence and transmission phenotype, we engineered these amino acid substitutions, singly and in combination, into a ZIKV infectious clone. We found that NS4A- E19G contributed to the enhanced virulence and mortality phenotype in mice. Further analyses revealed that NS4A-E19G results in increased neurotropism and distinct innate immune signaling patterns in the brain. None of the substitutions contributed to changes in transmission potential in mosquitoes. Together, these findings suggest that direct transmission chains could enable the emergence of more virulent ZIKV strains without compromising mosquito transmission capacity, although the underlying genetics of these adaptations are complex.
- Published
- 2023
7. CD8+ cells and small viral reservoirs facilitate post-ART control of SIV in Mauritian cynomolgus macaques
- Author
-
Olivia E. Harwood, Lea M. Matschke, Ryan V. Moriarty, Alexis J. Balgeman, Abigail J. Weaver, Amy L. Ellis-Connell, Andrea M. Weiler, Lee C. Winchester, Courtney V. Fletcher, Thomas C. Friedrich, Brandon F. Keele, David H. O’Connor, Jessica D. Lang, Matthew R. Reynolds, and Shelby L. O’Connor
- Abstract
Sustainable HIV remission after antiretroviral therapy (ART) withdrawal, or post-treatment control (PTC), remains a top priority for HIV treatment. We observed surprising PTC in an MHC-haplomatched cohort of MHC-M3+ SIVmac239+ Mauritian cynomolgus macaques (MCMs) initiated on ART at two weeks post-infection (wpi). For six months after ART withdrawal, we observed undetectable or transient viremia in seven of eight MCMs. In vivo depletion of CD8α+ cells induced rebound in all animals, indicating the PTC was mediated, at least in part, by CD8α+ cells. We found that MCMs had smaller acute viral reservoirs than a cohort of identically infected rhesus macaques, a population that rarely develops PTC. The mechanisms by which unusually small viral reservoirs and CD8α+ cell-mediated virus suppression enable PTC can be investigated using this MHC-haplomatched MCM model. Further, defining the immunologic mechanisms that engender PTC in this model may identify therapeutic targets for inducing durable HIV remission in humans.
- Published
- 2023
- Full Text
- View/download PDF
8. Mathematical modeling indicates that regulatory inhibition of CD8+T cell cytotoxicity can limit efficacy of IL-15 immunotherapy in cases of high pre-treatment SIV viral load
- Author
-
Jonathan W. Cody, Amy L. Ellis-Connell, Shelby L. O’Connor, and Elsje Pienaar
- Abstract
Immunotherapeutic cytokines can activate immune cells against cancers and chronic infections. N-803 is an IL-15 superagonist that expands CD8+T cells and increases their cytotoxicity. N-803 also temporarily reduced viral load in a limited subset of non-human primates infected with simian immunodeficiency virus (SIV), a model of HIV. However, viral suppression has not been observed in all SIV cohorts and may depend on pre-treatment viral load and the corresponding effects on CD8+T cells. Starting from an existing mechanistic mathematical model of N-803 immunotherapy of SIV, we develop a model that includes activation of SIV-specific and non-SIV-specific CD8+T cells by antigen, inflammation, and N-803. Also included is a regulatory counter-response that inhibits CD8+T cell proliferation and function, representing the effects of immune checkpoint molecules and immunosuppressive cells. We simultaneously calibrate the model to two separate SIV cohorts. The first cohort had low viral loads prior to treatment (≈3-4 log viral RNA copy equivalents (CEQ)/mL), and N-803 treatment transiently suppressed viral load. The second had higher pre-treatment viral loads (≈5-7 log CEQ/mL) and saw no consistent virus suppression with N-803. The mathematical model can replicate the viral and CD8+T cell dynamics of both cohorts based on different pre-treatment viral loads and different levels of regulatory inhibition of CD8+T cells due to those viral loads (i.e. initial conditions of model). Our predictions are validated by additional data from these and other SIV cohorts. While both cohorts had high numbers of activated SIV-specific CD8+T cells in simulations, viral suppression was precluded in the high viral load cohort due to elevated inhibition of cytotoxicity. Thus, we mathematically demonstrate how the pre-treatment viral load can influence immunotherapeutic efficacy, highlighting the in vivo conditions and combination therapies that could maximize efficacy and improve treatment outcomes.Author SummaryImmunotherapy bolsters and redirects the immune system to fight chronic infections and cancers. However, the effectiveness of some immunotherapies may depend on the level of pre-treatment inflammation and the corresponding presence of regulatory cells and immune checkpoint molecules that normally function to prevent immune overreaction. Here, we consider two previously published cohorts of macaques who were given the immunotherapeutic N-803 to treat Simian Immunodeficiency Virus, an analog of Human Immunodeficiency Virus (HIV). One cohort had low viral loads before treatment, and N-803 temporarily suppressed viral loads. The second cohort had high viral loads that did not consistently decrease with N-803 treatment. In this work, we demonstrate with a mathematical model how these two distinct outcomes can arise due only to the different viral loads and the corresponding immune activation and regulatory response. In the model, we find that the key limiting factor is the direct inhibition of the cytotoxic action of immune cells by immune checkpoint molecules. This model indicates that simultaneous blockade of immune checkpoint molecules may be necessary for effective application of N-803 for the treatment of HIV. This and similar models can inform the design of such combination therapies for cancer and chronic infection.
- Published
- 2023
- Full Text
- View/download PDF
9. Host Immunity toMycobacterium tuberculosisInfection is Similar in Simian Immunodeficiency Virus (SIV)-infected, Antiretroviral Therapy-treated and SIV-naïve Juvenile Macaques
- Author
-
Erica C. Larson, Amy L. Ellis, Mark A. Rodgers, Abigail K. Gubernat, Janelle L. Gleim, Ryan V. Moriarty, Alexis J. Balgeman, Yonne K. Menezes, Cassaundra L. Ameel, Daniel J. Fillmore, Skyler M. Pergalske, Jennifer A. Juno, Pauline Maiello, Alexander G. White, H. Jacob Borish, Dale I. Godfrey, Stephen J. Kent, Lishomwa C. Ndhlovu, Shelby L. O’Connor, and Charles A. Scanga
- Abstract
Pre-existing HIV infection increases tuberculosis (TB) risk in children. Antiretroviral therapy (ART) reduces, but does not abolish, this risk in children with HIV. The immunologic mechanisms involved in TB progression in both HIV-naïve and HIV-infected children have not been explored. Much of our current understanding is based on human studies in adults and adult animal models. In this study, we sought to model childhood HIV/Mycobacterium tuberculosis(Mtb) coinfection in the setting of ART and characterize T cells during TB progression. Macaques equivalent to 4-8 year-old children were intravenously infected with SIVmac239M, treated with ART three months later, and coinfected with Mtb three months after initiating ART. SIV-naïve macaques were similarly infected with Mtb alone. TB pathology and total Mtb burden did not differ between SIV-infected, ART-treated and SIV-naïve macaques, although lung Mtb burden was lower in SIV-infected, ART-treated macaques. No major differences in frequencies of CD4+ and CD8+ T cells and unconventional T cell subsets (Vγ9+ γδ T cells, MAIT cells, and NKT cells) in airways were observed between SIV-infected, ART-treated and SIV-naïve macaques over the course of Mtb infection, with the exception of CCR5+ CD4+ and CD8+ T cells which were slightly lower. CD4+ and CD8+ T cell frequencies did not differ in the lung granulomas obtained at necropsy, nor did they differ in the frequency of immune checkpoint and proliferative markers. Thus, ART treatment of juvenile macaques, three months after SIV infection, resulted in similar progression of Mtb and T cell responses compared to Mtb in SIV-naïve macaques.
- Published
- 2022
- Full Text
- View/download PDF
10. Transient T Cell Expansion, Activation, and Proliferation in Therapeutically Vaccinated Simian Immunodeficiency Virus-Positive Macaques Treated with N-803
- Author
-
Olivia E. Harwood, Alexis J. Balgeman, Abigail J. Weaver, Amy L. Ellis-Connell, Andrea M. Weiler, Katrina N. Erickson, Lea M. Matschke, Athena E. Golfinos, Vaiva Vezys, Pamela J. Skinner, Jeffrey T. Safrit, Paul T. Edlefsen, Matthew R. Reynolds, Thomas C. Friedrich, and Shelby L. O’Connor
- Subjects
Vaccination ,Immunology ,SAIDS Vaccines ,Simian Acquired Immunodeficiency Syndrome ,Vaccinia virus ,CD8-Positive T-Lymphocytes ,Macaca mulatta ,Microbiology ,Virology ,Insect Science ,Vaccines and Antiviral Agents ,Animals ,Simian Immunodeficiency Virus ,Cell Proliferation - Abstract
Vaccine strategies aimed at eliciting human immunodeficiency virus (HIV)-specific CD8(+) T cells are one major target of interest in HIV functional cure strategies. We hypothesized that CD8(+) T cells elicited by therapeutic vaccination during antiretroviral therapy (ART) would be recalled and boosted by treatment with the interleukin 15 (IL-15) superagonist N-803 after ART discontinuation. We intravenously immunized four simian immunodeficiency virus-positive (SIV(+)) Mauritian cynomolgus macaques receiving ART with vesicular stomatitis virus (VSV), modified vaccinia virus Ankara strain (MVA), and recombinant adenovirus serotype 5 (rAd-5) vectors all expressing SIVmac239 Gag. Immediately after ART cessation, these animals received three doses of N-803. Four control animals received no vaccines or N-803. The vaccine regimen generated a high-magnitude response involving Gag-specific CD8(+) T cells that were proliferative and biased toward an effector memory phenotype. We then compared cells elicited by vaccination (Gag specific) to cells elicited by SIV infection and unaffected by vaccination (Nef specific). We found that N-803 treatment enhanced the frequencies of both bulk and proliferating antigen-specific CD8(+) T cells elicited by vaccination and the antigen-specific CD8(+) T cells elicited by SIV infection. In sum, we demonstrate that a therapeutic heterologous prime-boost-boost (HPBB) vaccine can elicit antigen-specific effector memory CD8(+) T cells that are boosted by N-803. IMPORTANCE While antiretroviral therapy (ART) can suppress HIV replication, it is not a cure. It is therefore essential to develop therapeutic strategies to enhance the immune system to better become activated and recognize virus-infected cells. Here, we evaluated a novel therapeutic vaccination strategy delivered to SIV(+) Mauritian cynomolgus macaques receiving ART. ART was then discontinued and we delivered an immunotherapeutic agent (N-803) after ART withdrawal with the goal of eliciting and boosting anti-SIV cellular immunity. Immunologic and virologic analysis of peripheral blood and lymph nodes collected from these animals revealed transient boosts in the frequency, activation, proliferation, and memory phenotype of CD4(+) and CD8(+) T cells following each intervention. Overall, these results are important in educating the field of the transient nature of the immunological responses to this particular therapeutic regimen and the similar effects of N-803 on boosting T cells elicited by vaccination or elicited naturally by infection.
- Published
- 2022
- Full Text
- View/download PDF
11. A New Variant of Hepatitis A Virus Causing Transient Liver Enzyme Elevations in Mauritius-origin Laboratory-housed Cynomolgus macaques
- Author
-
Lars Mecklenburg, Rebecca Ducore, Molly Boyle, Andrew Newell, Laura Boone, Joerg Luft, Annette Romeike, Ann-Kathrin Haverkamp, Keith Mansfield, JJ Baczenas, Nick Minor, Shelby L. O’Connor, and David H. O’Connor
- Abstract
Hepatitis A virus (HAV) infects humans and non-human primates causing an acute self-limited illness. Three HAV genotypes have been described for humans and three genotypes have been described for non-human primates. We observed transiently elevated liver enzymes in Mauritius-origin laboratory-housed macaques in Germany and were not able to demonstrate HAV by serology and PCR. Using deep sequencing, we have identified a new HAV genotype with 86% nucleotide sequence homology to HAV genotype IV capsid proteins and approximately 80% nucleotide homology to other HAV genotypes. In situ hybridization indicates persistence in the biliary epithelium up to 3 months after liver enzymes were elevated. Vaccination using a commercial vaccine against human HAV prevented reoccurrence of liver enzyme elevations. Since available assays for HAV did not detect this new variant, knowledge of its existence may ameliorate potential significant epidemiological and research implications in laboratories globally.Article Summary LineA new genotype of hepatitis A virus, that was not identifiable by available diagnostic assays, caused liver enzyme elevations in laboratory-housed Mauritius-origin Cynomolgus macaques.
- Published
- 2022
- Full Text
- View/download PDF
12. IL-15 Superagonist N-803 Enhances IFN-γ Production of MAIT Cells in SIV + Macaques
- Author
-
Amy L. Ellis-Connell, Alexis J. Balgeman, Nadean M. Kannal, Karigynn Hansen Chaimson, Anna Batchenkova, Jeffrey T. Safrit, and Shelby L. O’Connor
- Subjects
Infectious Diseases ,Immunology ,Parasitology ,Microbiology - Abstract
Mucosal associated invariant T (MAIT) cells are innate T cells that recognize bacterial metabolites and secrete cytokines and cytolytic enzymes to destroy infected target cells. This makes MAIT cells promising targets for immunotherapy to combat bacterial infections.
- Published
- 2022
- Full Text
- View/download PDF
13. Pre-existing Simian Immunodeficiency Virus Infection Increases Expression of T Cell Markers Associated with Activation during Early Mycobacterium tuberculosis Coinfection and Impairs TNF Responses in Granulomas
- Author
-
Pauline Maiello, Chelsea M Causgrove, Cassaundra Ameel, Jaime A Tomko, Alexis J. Balgeman, Tonilynn M Baranowski, Ryan V. Moriarty, Charles A. Scanga, Erica C. Larson, Amy L. Ellis-Connell, Mark Rodgers, and Shelby L. O’Connor
- Subjects
Tuberculosis ,medicine.diagnostic_test ,biology ,business.industry ,animal diseases ,T cell ,Immunology ,virus diseases ,medicine.disease ,biology.organism_classification ,Virology ,Mycobacterium tuberculosis ,03 medical and health sciences ,0302 clinical medicine ,medicine.anatomical_structure ,Bronchoalveolar lavage ,TIGIT ,Coinfection ,medicine ,Immunology and Allergy ,Tumor necrosis factor alpha ,business ,CD8 ,030215 immunology - Abstract
Tuberculosis (TB) is the leading infectious cause of death among people living with HIV. People living with HIV are more susceptible to contracting Mycobacterium tuberculosis and often have worsened TB disease. Understanding the immunologic defects caused by HIV and the consequences it has on M. tuberculosis coinfection is critical in combating this global health epidemic. We previously showed in a model of SIV and M. tuberculosis coinfection in Mauritian cynomolgus macaques (MCM) that SIV/M. tuberculosis–coinfected MCM had rapidly progressive TB. We hypothesized that pre-existing SIV infection impairs early T cell responses to M. tuberculosis infection. We infected MCM with SIVmac239, followed by coinfection with M. tuberculosis Erdman 6 mo later. Although similar, TB progression was observed in both SIV+ and SIV-naive animals at 6 wk post–M. tuberculosis infection; longitudinal sampling of the blood (PBMC) and airways (bronchoalveolar lavage) revealed a significant reduction in circulating CD4+ T cells and an influx of CD8+ T cells in airways of SIV+ animals. At sites of M. tuberculosis infection (i.e., granulomas), SIV/M. tuberculosis–coinfected animals had a higher proportion of CD4+ and CD8+ T cells expressing PD-1 and TIGIT. In addition, there were fewer TNF-producing CD4+ T cells in granulomas of SIV/M. tuberculosis–coinfected animals. Taken together, we show that concurrent SIV infection alters T cell phenotypes in granulomas during the early stages of TB disease. As it is critical to establish control of M. tuberculosis replication soon postinfection, these phenotypic changes may distinguish the immune dysfunction that arises from pre-existing SIV infection, which promotes TB progression.
- Published
- 2021
- Full Text
- View/download PDF
14. Transient T cell expansion, activation, and proliferation in therapeutically vaccinated SIV+ macaques treated with N-803
- Author
-
Olivia E. Harwood, Alexis J. Balgeman, Abigail J. Weaver, Amy L. Ellis-Connell, Andrea M. Weiler, Katrina N. Erickson, Lea M. Matschke, Athena E. Golfinos, Vaiva Vezys, Pamela J. Skinner, Jeffrey T. Safrit, Paul T. Edlefsen, Matthew R. Reynolds, Thomas C. Friedrich, and Shelby L. O’Connor
- Abstract
Vaccine strategies aimed at eliciting HIV-specific CD8+ T cells are one major target of interest in HIV functional cure strategies. We hypothesized that CD8+ T cells elicited by therapeutic vaccination during antiretroviral therapy (ART) would be recalled and boosted by treatment with the IL-15 superagonist N-803 after ART discontinuation. We intravenously immunized four SIV+ Mauritian cynomolgus macaques (MCM) receiving ART with vesicular stomatitis virus (VSV), modified vaccinia virus Ankara strain (MVA), and recombinant adenovirus serotype 5 (rAd-5) vectors all expressing SIVmac239 Gag. Immediately after ART cessation, these animals received three doses of N-803. Four control animals received no vaccines or N-803. The vaccine regimen generated a high magnitude of Gag-specific CD8+ T cells that were proliferative and biased toward an effector memory phenotype. We then compared cells elicited by vaccination (Gag-specific) to cells elicited by SIV infection and unaffected by vaccination (Nef-specific). We found that N-803 treatment enhanced both the frequencies of bulk and proliferating antigen-specific CD8+ T cells elicited by vaccination and the antigen-specific CD8+ T cells elicited by SIV infection. In sum, we demonstrate that a therapeutic heterologous prime-boost-boost (HPBB) vaccine can elicit antigen-specific effector memory CD8+ T cells that are boosted by N-803.
- Published
- 2022
- Full Text
- View/download PDF
15. IL-15 superagonist N-803 enhances IFNγ production and alters the trafficking of MAIT cells in SIV+ macaques
- Author
-
Amy L. Ellis-Connell, Alexis J. Balgeman, Nadean M. Kannal, Karigynn Hansen Chaimson, Anna Batchenkova, Jeffrey T. Safrit, and Shelby L. O’Connor
- Abstract
Mucosal Associated Invariant T cells (MAIT cells) are innate T cells that recognize bacterial metabolites and secrete cytokines and cytolytic enzymes to destroy infected target cells. This makes MAIT cells promising targets for immunotherapy to combat bacterial infections. Here, we analyzed the effects of an immunotherapeutic agent, the IL-15 superagonist N-803, on MAIT cell activation, trafficking, and cytolytic function in macaques. We found that N-803 could activate MAIT cells in vitro and increase their ability to produce IFNγ in response to bacterial stimulation. To expand upon this, we examined the phenotypes and function of MAIT cells present in samples collected from PBMC, airways (BAL), and lymph nodes (LN) from rhesus macaques that were treated in vivo with N-803. N-803 treatment led to a transient 6-7 fold decrease in the total number of MAIT cells in the peripheral blood relative to pre N-803 timepoints. Concurrent with the decrease in cells in the peripheral blood, we observed a rapid decline in the frequency of CXCR3+CCR6+ MAITs. This corresponded with an increase in the frequency of CCR6+ MAITs in BAL, and higher frequencies of ki-67+ and granzyme B+ MAITs in blood, LN, and BAL. Finally, N-803 improved the ability of MAIT cells collected from PBMC and airways to produce IFNγ in response to bacterial stimulation. Overall, N-803 shows the potential to transiently alter the trafficking and enhance the antibacterial activity of MAIT cells which could be combined with other strategies to combat bacterial infections.
- Published
- 2022
- Full Text
- View/download PDF
16. Control of SIV infection in prophylactically vaccinated, ART-naïve macaques is required for the most efficacious CD8 T cell response during treatment with the IL-15 superagonist N-803
- Author
-
Amy L. Ellis-Connell, Alexis J. Balgeman, Olivia E. Harwood, Ryan V. Moriarty, Jeffrey T. Safrit, Andrea M. Weiler, Thomas C. Friedrich, and Shelby L. O’Connor
- Abstract
The IL-15 superagonist N-803 has been shown to enhance the function of CD8 T cells and NK cells. We previously found that in a subset of vaccinated, ART-naïve, SIV+ rhesus macaques, N-803 treatment led to a rapid but transient decline in plasma viremia that positively correlated with an increase in the frequency of CD8 T cells. Here we tested the hypothesis that prophylactic vaccination was required for N-803 mediated suppression of SIV plasma viremia. We vaccinated rhesus macaques with a DNA prime/Ad5 boost regimen using vectors expressing SIVmac239 gag, with or without a plasmid expressing IL-12, or left them unvaccinated. Animals were then intravenously infected with SIVmac239M. Six months after infection, animals were treated with N-803. We found no differences in control of plasma viremia during N-803 treatment between vaccinated and unvaccinated macaques. Furthermore, the SIV-specific CD8 T cells displayed no differences in frequency or ability to traffic to the lymph nodes. Interestingly, when we divided the SIV+ animals based on plasma viral load set-point prior to N-803 treatment, N-803 increased the frequency of SIV-specific T cells expressing ki-67+ and granzyme B+ in animals with low plasma viremia (4 copies/mL; SIV controllers) compared to animals with high plasma viremia (>104 copies/mL;SIV non-controllers). In addition, Gag-specific CD8 T cells from the SIV+ controllers had a greater increase in CD107a+CD8+ T cells when compared to SIV+ non-controllers. Overall, our results indicate that N-803 is most effective in SIV+ animals with a pre-existing immunological ability to control SIV replication.
- Published
- 2022
- Full Text
- View/download PDF
17. Characterization of the SARS-CoV-2 B.1.621 (Mu) variant
- Author
-
Peter J. Halfmann, Makoto Kuroda, Tammy Armbrust, James Theiler, Ariane Balaram, Gage K. Moreno, Molly A. Accola, Kiyoko Iwatsuki-Horimoto, Riccardo Valdez, Emily Stoneman, Katarina Braun, Seiya Yamayoshi, Elizabeth Somsen, John J. Baczenas, Keiko Mitamura, Masao Hagihara, Eisuke Adachi, Michiko Koga, Matthew McLaughlin, William Rehrauer, Masaki Imai, Shinya Yamamoto, Takeya Tsutsumi, Makoto Saito, Thomas C. Friedrich, Shelby L. O’Connor, David H. O’Connor, Aubree Gordon, Bette Korber, and Yoshihiro Kawaoka
- Subjects
Membrane Glycoproteins ,SARS-CoV-2 ,Vaccination ,Immunization, Passive ,COVID-19 ,General Medicine ,Antibodies, Viral ,Antibodies, Neutralizing ,Article ,Viral Envelope Proteins ,Neutralization Tests ,Spike Glycoprotein, Coronavirus ,Humans ,COVID-19 Serotherapy - Abstract
The SARS-CoV-2 B.1.621 (Mu) variant emerged in January 2021 and was categorized as a variant of interest by the World Health Organization in August 2021. This designation prompted us to study the sensitivity of this variant to antibody neutralization. In a live virus neutralization assay with serum samples from individuals vaccinated with the Pfizer/BioNTech or Moderna mRNA vaccines, we measured neutralization antibody titers against B.1.621, an early isolate (spike 614D), and a variant of concern (B.1.351, Beta variant). We observed reduced neutralizing antibody titers against the B.1.621 variant (3.4- to 7-fold reduction, depending on the serum sample and time after the second vaccination) compared to the early isolate and a similar reduction when compared to B.1.351. Likewise, convalescent serum from hamsters previously infected with an early isolate neutralized B.1.621 to a lower degree. Despite this antibody titer reduction, hamsters could not be efficiently rechallenged with the B.1.621 variant, suggesting that the immune response to the first infection is adequate to provide protection against a subsequent infection with the B.1.621 variant.
- Published
- 2022
18. Spontaneous Control of SIV Replication Does Not Prevent T Cell Dysregulation and Bacterial Dissemination in Animals Co-Infected with M. tuberculosis
- Author
-
Ryan V. Moriarty, Mark A. Rodgers, Amy L. Ellis, Alexis J. Balgeman, Erica C. Larson, Forrest Hopkins, Michael R. Chase, Pauline Maiello, Sarah M. Fortune, Charles A. Scanga, and Shelby L. O’Connor
- Subjects
Microbiology (medical) ,CD4-Positive T-Lymphocytes ,Granuloma ,General Immunology and Microbiology ,Ecology ,Physiology ,Coinfection ,T-Lymphocytes ,Simian Acquired Immunodeficiency Syndrome ,virus diseases ,HIV Infections ,Cell Biology ,Mycobacterium tuberculosis ,Macaca fascicularis ,Infectious Diseases ,Genetics ,Animals ,Tuberculosis ,Simian Immunodeficiency Virus - Abstract
Individuals co-infected with HIV and Mycobacterium tuberculosis (Mtb) are more likely to develop severe tuberculosis (TB) disease than HIV-naive individuals. To understand how a chronic pre-existing Simian immunodeficiency virus (SIV) infection impairs the early immune response to Mtb, we used the Mauritian cynomolgus macaque (MCM) model of SIV/Mtb co-infection. We examined the relationship between peripheral viral control and Mtb burden, Mtb dissemination, and T cell function between SIV+ spontaneous controllers, SIV+ non-controllers, and SIV-naive MCM who were challenged with a barcoded Mtb Erdman strain 6 months post-SIV infection and necropsied 6 weeks post-Mtb infection. Mycobacterial burden was highest in the SIV+ non-controllers in all assessed tissues. In lung granulomas, the frequency of TNF-α-producing CD4
- Published
- 2022
19. Mimicking SIV chimpanzee viral evolution toward HIV‐1 during cross‐species transmission
- Author
-
Ramesh Akkina, James Curlin, Leila Remling-Mulder, Ryan V. Moriarty, Kelly Goff, Mark D. Stenglein, Kimberly Schmitt, Shelby L. O’Connor, and Preston A. Marx
- Subjects
Pan troglodytes ,viruses ,Human immunodeficiency virus (HIV) ,Cross-species transmission ,Biology ,medicine.disease_cause ,Genome ,Article ,law.invention ,Evolution, Molecular ,law ,medicine ,Animals ,General Veterinary ,virus diseases ,Virology ,Ape Diseases ,Disease Models, Animal ,Transmission (mechanics) ,Viral evolution ,Mutation ,HIV-1 ,Simian Immunodeficiency Virus ,Animal Science and Zoology ,Adaptation ,Viral load - Abstract
HIV-1 evolved from SIV during cross-species transmission events, though viral genetic changes are not well understood. Here we studied the evolution of SIVcpzLB715 into HIV-1 Group M using humanized mice. High viral loads, rapid CD4(+) T-cell decline, and non-synonymous substitutions were identified throughout the viral genome suggesting viral adaptation.
- Published
- 2020
- Full Text
- View/download PDF
20. Evolution of SIVsm in humanized mice towards HIV‐2
- Author
-
Kimberly Schmitt, Shelby L. O’Connor, Preston A. Marx, Ramesh Akkina, Leila Remling-Mulder, Ryan V. Moriarty, James Curlin, Kelly Goff, and Mark D. Stenglein
- Subjects
040301 veterinary sciences ,Human immunodeficiency virus (HIV) ,Virulence ,Mice, Transgenic ,Biology ,medicine.disease_cause ,Article ,Virus ,Evolution, Molecular ,0403 veterinary science ,Cercocebus atys ,Mice ,medicine ,Animals ,0501 psychology and cognitive sciences ,050102 behavioral science & comparative psychology ,General Veterinary ,Monkey Diseases ,05 social sciences ,04 agricultural and veterinary sciences ,Virology ,CD4 Lymphocyte Count ,Disease Models, Animal ,HIV-2 ,Mutation ,Humanized mouse ,Simian Immunodeficiency Virus ,Animal Science and Zoology - Abstract
Through the accumulation of adaptive mutations, HIV-2 originated from SIVsm. To identify these evolutionary changes, a humanized mouse model recapitulated the process that likely enabled this cross-species transmission event. Various adaptive mutations arose, as well as increased virulence and CD4(+) T-cell decline as the virus was passaged in humanized mice.
- Published
- 2020
- Full Text
- View/download PDF
21. Spondweni virus causes fetal harm in Ifnar1 mice and is transmitted by Aedes aegypti mosquitoes
- Author
-
Shelby L. O’Connor, Anna S. Jaeger, Matthew T. Aliota, Davis M. Seelig, Andrea M. Weiler, Michael K. Fritsch, Thomas C. Friedrich, David H. O’Connor, Sierra Rybarczyk, and Ryan V. Moriarty
- Subjects
Placental histopathology ,Mosquito Vectors ,Receptor, Interferon alpha-beta ,Aedes aegypti ,medicine.disease_cause ,Article ,Flavivirus Infections ,Zika virus ,Mice ,03 medical and health sciences ,Aedes ,Virology ,parasitic diseases ,medicine ,Animals ,Humans ,Congenital zika syndrome ,030304 developmental biology ,Mice, Knockout ,Arbovirus ,Spondweni virus ,0303 health sciences ,Fetus ,biology ,Flavivirus ,fungi ,030302 biochemistry & molecular biology ,virus diseases ,biology.organism_classification ,Culex quinquefasciatus ,Mice, Inbred C57BL ,Disease Models, Animal ,Female ,Vector competence - Abstract
Spondweni virus (SPONV) is the most closely related known flavivirus to Zika virus (ZIKV). Its pathogenic potential and vector specificity have not been well defined. SPONV has been found predominantly in Africa, but was recently detected in a pool of Culex quinquefasciatus mosquitoes in Haiti. Here we show that SPONV can cause significant fetal harm, including demise, comparable to ZIKV, in a mouse model of vertical transmission. Following maternal inoculation, we detected infectious SPONV in placentas and fetuses, along with significant fetal and placental histopathology, together suggesting vertical transmission. To test vector competence, we exposed Aedes aegypti and Culex quinquefasciatus mosquitoes to SPONV-infected bloodmeals. Aedes aegypti could efficiently transmit SPONV, whereas Culex quinquefasciatus could not. Our results suggest that SPONV has the same features that made ZIKV a public health risk.
- Published
- 2020
- Full Text
- View/download PDF
22. SARS-CoV-2 and other respiratory pathogens are detected in continuous air samples from congregate settings
- Author
-
Mitchell D. Ramuta, Christina M. Newman, Savannah F. Brakefield, Miranda R. Stauss, Roger W. Wiseman, Amanda Kita-Yarbro, Eli J. O’Connor, Neeti Dahal, Ailam Lim, Keith P. Poulsen, Nasia Safdar, John A. Marx, Molly A. Accola, William M. Rehrauer, Julia A. Zimmer, Manjeet Khubbar, Lucas J. Beversdorf, Emma C. Boehm, David Castañeda, Clayton Rushford, Devon A. Gregory, Joseph D. Yao, Sanjib Bhattacharyya, Marc C. Johnson, Matthew T. Aliota, Thomas C. Friedrich, David H. O’Connor, and Shelby L. O’Connor
- Subjects
Multidisciplinary ,Wisconsin ,SARS-CoV-2 ,Minnesota ,fungi ,General Physics and Astronomy ,COVID-19 ,Humans ,RNA, Viral ,General Chemistry ,General Biochemistry, Genetics and Molecular Biology - Abstract
Two years after the emergence of SARS-CoV-2, there is still a need for better ways to assess the risk of transmission in congregate spaces. We deployed active air samplers to monitor the presence of SARS-CoV-2 in real-world settings across communities in the Upper Midwestern states of Wisconsin and Minnesota. Over 29 weeks, we collected 527 air samples from 15 congregate settings. We detected 106 samples that were positive for SARS-CoV-2 viral RNA, demonstrating that SARS-CoV-2 can be detected in continuous air samples collected from a variety of real-world settings. We expanded the utility of air surveillance to test for 40 other respiratory pathogens. Surveillance data revealed differences in timing and location of SARS-CoV-2 and influenza A virus detection. In addition, we obtained SARS-CoV-2 genome sequences from air samples to identify variant lineages. Collectively, this shows air sampling is a scalable, high throughput surveillance tool that could be used in conjunction with other methods for detecting respiratory pathogens in congregate settings.
- Published
- 2022
23. Propagation of SARS-CoV-2 in Calu-3 Cells to Eliminate Mutations in the Furin Cleavage Site of Spike
- Author
-
John James Baczenas, Hanne Andersen, Sujatha Rashid, David Yarmosh, Nikhita Puthuveetil, Michael Parker, Rebecca Bradford, Clint Florence, Kimberly J. Stemple, Mark G. Lewis, and Shelby L. O’Connor
- Subjects
Furin ,Calu-3 ,SARS-CoV-2 ,polymorphisms ,stock preparation ,viruses ,COVID-19 ,Microbiology ,Article ,QR1-502 ,Infectious Diseases ,Cell Line, Tumor ,Cricetinae ,Virology ,Chlorocebus aethiops ,Mutation ,Spike Glycoprotein, Coronavirus ,Animals ,Humans ,Serial Passage ,Vero Cells - Abstract
SARS-CoV-2 pathogenesis, vaccine, and therapeutic studies rely on the use of animals challenged with highly pathogenic virus stocks produced in cell cultures. Ideally, these virus stocks should be genetically and functionally similar to the original clinical isolate, retaining wild-type properties to be reliably used in animal model studies. It is well-established that SARS-CoV-2 isolates serially passaged on Vero cell lines accumulate mutations and deletions in the furin cleavage site; however, these can be eliminated when passaged on Calu-3 lung epithelial cell lines, as presented in this study. As numerous stocks of SARS-CoV-2 variants of concern are being grown in cell cultures with the intent for use in animal models, it is essential that propagation methods generate virus stocks that are pathogenic in vivo. Here, we found that the propagation of a B.1.351 SARS-CoV-2 stock on Calu-3 cells eliminated viruses that previously accumulated mutations in the furin cleavage site. Notably, there were alternative variants that accumulated at the same nucleotide positions in virus populations grown on Calu-3 cells at multiple independent facilities. When a Calu-3-derived B.1.351 virus stock was used to infect hamsters, the virus remained pathogenic and the Calu-3-specific variants persisted in the population. These results suggest that Calu-3-derived virus stocks are pathogenic but care should still be taken to evaluate virus stocks for newly arising mutations during propagation.
- Published
- 2021
- Full Text
- View/download PDF
24. SARS-CoV-2 Infection of Rhesus Macaques Treated Early with Human COVID-19 Convalescent Plasma
- Author
-
Smita S. Iyer, Ramya Immareddy, Koen K. A. Van Rompay, Linda Fritts, Jodie Usachenko, Amy Kistler, Brian A. Schmidt, JoAnn L. Yee, Dennis J. Hartigan-O'Connor, Sonny R. Elizaldi, J. Rachel Reader, Joseph L. DeRisi, Yashavanth Shaan Lakshmanappa, Rachel E. Pollard, Clara Di Germanio, Timothy D. Carroll, Katherine J. Olstad, Jesse D. Deere, Jamin W. Roh, Shelby L. O’Connor, Jennifer Watanabe, Michael P. Busch, Christopher J. Miller, Joseph Dutra, Jack Kamm, Graham Simmons, Rebecca L. Sammak, and Martinez, Miguel Angel
- Subjects
Physiology ,Disease ,Passive ,Antibodies, Viral ,Virus Replication ,Clinical endpoint ,Medicine ,Viral ,Lung ,Neutralizing ,Ecology ,biology ,Viral Load ,QR1-502 ,Spike Glycoprotein ,virology ,Titer ,Infectious Diseases ,Spike Glycoprotein, Coronavirus ,convalescent plasma ,Pneumonia & Influenza ,Antibody ,Infection ,Viral load ,Research Article ,Microbiology (medical) ,microbial pathogenesis ,nonhuman primate ,Microbiology ,Antiviral Agents ,Antibodies ,Vaccine Related ,Immune system ,Biodefense ,Genetics ,Animals ,Humans ,Viral shedding ,Pandemics ,animal models of infectious diseases ,General Immunology and Microbiology ,business.industry ,Animal ,SARS-CoV-2 ,Prevention ,Immunization, Passive ,Immunity ,COVID-19 ,Cell Biology ,Pneumonia ,Antibodies, Neutralizing ,Macaca mulatta ,Coronavirus ,Disease Models, Animal ,Emerging Infectious Diseases ,Good Health and Well Being ,Immunization ,Immunology ,Disease Models ,biology.protein ,passive immunization ,business - Abstract
Human clinical studies investigating use of convalescent plasma (CP) for treatment of coronavirus disease 2019 (COVID-19) have produced conflicting results. Outcomes in these studies may vary at least partly due to different timing of CP administration relative to symptom onset. The mechanisms of action of CP include neutralizing antibodies but may extend beyond virus neutralization to include normalization of blood clotting and dampening of inflammation. Unresolved questions include the minimum therapeutic titer in the CP units or CP recipient as well as the optimal timing of administration. Here, we show that treatment of macaques with CP within 24 h of infection does not reduce viral shedding in nasal or lung secretions compared to controls and does not detectably improve any clinical endpoint. We also demonstrate that CP administration does not impact viral sequence diversity in vivo, although the selection of a viral sequence variant in both macaques receiving normal human plasma was suggestive of immune pressure. Our results suggest that CP, administered to medium titers, has limited efficacy, even when given very early after infection. Our findings also contribute information important for the continued development of the nonhuman primate model of COVID-19. These results should inform interpretation of clinical studies of CP in addition to providing insights useful for developing other passive immunotherapies and vaccine strategies. IMPORTANCE Antiviral treatment options for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) remain very limited. One treatment that was explored beginning early in the pandemic (and that is likely to be tested early in future pandemics) is plasma collected from people who have recovered from coronavirus disease 2019 (COVID-19), known as convalescent plasma (CP). We tested if CP reduces viral shedding or disease in a nonhuman primate model. Our results demonstrate that administration of CP 1 day after SARS-CoV-2 infection had no significant impact on viral loads, clinical disease, or sequence diversity, although treatment with normal human plasma resulted in selection of a specific viral variant. Our results demonstrate that passive immunization with CP, even during early infection, provided no significant benefit in a nonhuman primate model of SARS-CoV-2 infection.
- Published
- 2021
25. Prior infection with SARS-CoV-2 WA1/2020 partially protects rhesus macaques against reinfection with B.1.1.7 and B.1.351 variants
- Author
-
Mark G. Lewis, Laurent Pessaint, Tochi Anioke, Lisa H. Tostanoski, Anthony L. Cook, Aiquan Chang, Daniel Valentin, Dan H. Barouch, Abishek Chandrashekar, Katherine McMahan, Owen Sanborn, Lauren Wattay, Hanne Leth Andersen, Jingyou Yu, Shelby L. O’Connor, Deandre Bueno-Wilkerson, Felix Nampanya, David L. Hope, Noe B. Mercado, Victoria M. Giffin, Huahua Wan, Sarah Gardner, Jinyan Liu, Maria Grazia Ferrari, Catherine Jacob-Dolan, John J. Baczenas, Daniel Sellers, Renita Brown, Benjamin Espina, Amanda J. Martinot, Elyse Teow, and Shivani A. Patel
- Subjects
2019-20 coronavirus outbreak ,biology ,Coronavirus disease 2019 (COVID-19) ,business.industry ,SARS-CoV-2 ,viruses ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,virus diseases ,COVID-19 ,General Medicine ,Evasion (ethics) ,Antibodies, Viral ,Virology ,Antibodies, Neutralizing ,Macaca mulatta ,Transmissibility (vibration) ,Reinfection ,biology.protein ,Medicine ,Animals ,Humans ,Antibody ,business ,Re infection - Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants that result in increased transmissibility and partial evasion of neutralizing antibodies have recently emerged. Whether natural immunity induced by the original SARS-CoV-2 WA1/2020 strain protects against rechallenge with these SARS-CoV-2 variants remains a critical unresolved question. In this study, we show that natural immunity induced by the WA1/2020 strain leads to partial but incomplete protection against the SARS-CoV-2 variants B.1.1.7 (alpha) and B.1.351 (beta) in rhesus macaques. We challenged rhesus macaques with B.1.1.7 and B.1.351 and showed that infection with these variants resulted in high viral replication in the upper and lower respiratory tract. We then infected rhesus macaques with the WA1/2020 strain and rechallenged them on day 35 with the WA1/2020, B.1.1.7, or B.1.351 variants. Natural immunity to WA1/2020 led to robust protection against rechallenge with WA1/2020 but only partial protection against rechallenge with B.1.351. An intermediate degree of protection was observed in rhesus macaques against rechallenge with B.1.1.7. These data demonstrate partial but incomplete protective efficacy of natural immunity induced by WA1/2020 against SARS-CoV-2 variants of concern. Our findings have important implications for both vaccination and public health strategies in the context of emerging SARS-CoV-2 variants of concern.
- Published
- 2021
26. Therapeutic Potential of IL-15 and N-803 in HIV/SIV Infection
- Author
-
Shelby L. O’Connor and Olivia E Harwood
- Subjects
Cellular immunity ,Recombinant Fusion Proteins ,T cell ,Simian Acquired Immunodeficiency Syndrome ,HIV Infections ,Context (language use) ,Review ,Virus Replication ,Microbiology ,Proinflammatory cytokine ,Mice ,Immune system ,Virology ,medicine ,Animals ,Humans ,N-803 ,Interleukin-15 ,Clinical Trials as Topic ,business.industry ,HIV ,Viral Load ,Macaca mulatta ,QR1-502 ,Virus Latency ,Infectious Diseases ,medicine.anatomical_structure ,Anti-Retroviral Agents ,IL-15 ,SIV ,Interleukin 15 ,Immunology ,HIV-1 ,Simian Immunodeficiency Virus ,business ,Viral load ,CD8 - Abstract
IL-15, a proinflammatory cytokine critical for the generation, maintenance, and homeostasis of T cell responses, is produced naturally in response to HIV/SIV infection, but has also demonstrated therapeutic potential. IL-15 can boost CD4+ and CD8+ T cell and NK cell proliferation, activation, and function. However, IL-15 treatment may cause aberrant immune activation and accelerated disease progression in certain circumstances. Moreover, the relationship between the timing of IL-15 administration and disease progression remains unclear. The IL-15 superagonist N-803 was developed to expand the therapeutic potential of IL-15 by maximizing its tissue distribution and half-life. N-803 has garnered enthusiasm recently as a way to enhance the innate and cellular immune responses to HIV/SIV by improving CD8+ T cell recognition and killing of virus-infected cells and directing immune cells to mucosal sites and lymph nodes, the primary sites of virus replication. N-803 has also been evaluated in “shock and kill” strategies due to its potential to reverse latency (shock) and enhance antiviral immunity (kill). This review examines the current literature about the effects of IL-15 and N-803 on innate and cellular immunity, viral burden, and latency reversal in the context of HIV/SIV, and their therapeutic potential both alone and combined with additional interventions such as antiretroviral therapy (ART) and vaccination.
- Published
- 2021
27. SIVcpz cross‐species transmission and viral evolution toward HIV‐1 in a humanized mouse model
- Author
-
Kimberly Schmitt, Preston A. Marx, Leila Remling-Mulder, Shelby L. O’Connor, Ramesh Akkina, Mark D. Stenglein, Ryan V. Moriarty, and James Curlin
- Subjects
040301 veterinary sciences ,viruses ,Simian Acquired Immunodeficiency Syndrome ,Cross-species transmission ,HIV Infections ,Mice, Transgenic ,Genome, Viral ,Biology ,Genome ,Article ,Evolution, Molecular ,0403 veterinary science ,Mice ,Animals ,0501 psychology and cognitive sciences ,050102 behavioral science & comparative psychology ,Progenitor ,General Veterinary ,Transmission (medicine) ,05 social sciences ,04 agricultural and veterinary sciences ,Viral Load ,Biological Evolution ,Virology ,CD4 Lymphocyte Count ,Viral evolution ,Humanized mouse ,HIV-1 ,Simian Immunodeficiency Virus ,Animal Science and Zoology ,Adaptation ,Viral load - Abstract
HIV-1 evolved from its progenitor SIV strains, but details are lacking on its adaptation to the human host. We followed the evolution of SIVcpz in humanized mice to mimic cross-species transmission. Increasing viral loads, CD4(+) T-cell decline, and non-synonymous mutations were seen in the entire genome reflecting viral adaptation.
- Published
- 2019
- Full Text
- View/download PDF
28. Characterization of a new SARS-CoV-2 variant that emerged in Brazil
- Author
-
Hiroaki Mitsuya, Tadashi Maemura, Ichiro Nakachi, Rie Baba, Riccardo Valdez, Hiroyuki Nagai, Hideaki Nakajima, Tetsuya Suzuki, Michiko Koga, Junichi Ochi, Tadaki Suzuki, Aubree Gordon, Samantha Loeber, Kensuke Fujita, Kenji Maeda, Maki Kiso, Carmen Gherasim, David A. Baker, Norio Sugaya, Makoto Saito, Noriko Nakajima, Yoshihiro Kawaoka, Takayuki Ogura, John J. Baczenas, Moe Okuda, Hideaki Kato, Kiyoko Iwatsuki-Horimoto, Shuetsu Fukushi, Hiroshi Ueki, Tiago J. S. Lopes, Yudai Kuroda, Shiho Chiba, Tokiko Watanabe, Makoto Kuroda, Norio Ohmagari, Tsuguto Fujimoto, Masaki Imai, Osamu Akasaka, Peter Halfmann, Masato Hatta, Michiko Ujie, Eisuke Adachi, Ken Maeda, Shelby L. O’Connor, Hiroshi Yotsuyanagi, Atsuhiro Yasuhara, Shinya Yamamoto, Amie J. Eisfeld, Yusuke Miyazato, Kenta Takahashi, Keiko Mitamura, Yuko Sakai-Tagawa, Mutsumi Ito, Shin ichiro Hattori, Morio Nakamura, Yuri Furusawa, David H. O’Connor, Seiya Yamayoshi, and Kazuma Yagi
- Subjects
0106 biological sciences ,0301 basic medicine ,Letter ,viruses ,Virus Replication ,Microbiology ,01 natural sciences ,Neutralization ,reinfection ,Mice ,03 medical and health sciences ,Immunogenicity, Vaccine ,Immune system ,Antigen ,Cricetinae ,Animals ,Humans ,skin and connective tissue diseases ,Lung ,Infectivity ,Multidisciplinary ,Mesocricetus ,biology ,SARS-CoV-2 ,fungi ,virus diseases ,COVID-19 ,convalescent human plasma ,X-Ray Microtomography ,Biological Sciences ,biology.organism_classification ,Antibodies, Neutralizing ,Virology ,body regions ,Titer ,030104 developmental biology ,P.1 variant ,Viral replication ,Spike Glycoprotein, Coronavirus ,biology.protein ,Antibody ,Syrian hamsters ,010606 plant biology & botany - Abstract
Significance Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants are of concern, with the P.1 variants dominating in Brazil. Brazil is now seeing a record number of deaths. Here, we report that the pathogenicity in hamsters of a P.1 variant is similar to that of nonvariant SARS-CoV-2. However, it has an expanded host range as shown by its replication in mice. Prior infection with nonvariant SARS-CoV-2 strains efficiently prevented replication of the P.1 variant in the lower respiratory tract of hamsters upon reinfection. Convalescent sera from patients infected with nonvariants or sera from messenger RNA vaccinees showed comparable neutralization titers among the P.1 and previously circulating strains. These results suggest that previous SARS-CoV-2 infection and vaccines based on the original SARS-CoV-2 will provide some protection against P.1 infection., The spike (S) protein of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) plays a key role in viral infectivity. It is also the major antigen stimulating the host's protective immune response, specifically, the production of neutralizing antibodies. Recently, a new variant of SARS-CoV-2 possessing multiple mutations in the S protein, designated P.1, emerged in Brazil. Here, we characterized a P.1 variant isolated in Japan by using Syrian hamsters, a well-established small animal model for the study of SARS-CoV-2 disease (COVID-19). In hamsters, the variant showed replicative abilities and pathogenicity similar to those of early and contemporary strains (i.e., SARS-CoV-2 bearing aspartic acid [D] or glycine [G] at position 614 of the S protein). Sera and/or plasma from convalescent patients and BNT162b2 messenger RNA vaccinees showed comparable neutralization titers across the P.1 variant, S-614D, and S-614G strains. In contrast, the S-614D and S-614G strains were less well recognized than the P.1 variant by serum from a P.1-infected patient. Prior infection with S-614D or S-614G strains efficiently prevented the replication of the P.1 variant in the lower respiratory tract of hamsters upon reinfection. In addition, passive transfer of neutralizing antibodies to hamsters infected with the P.1 variant or the S-614G strain led to reduced virus replication in the lower respiratory tract. However, the effect was less pronounced against the P.1 variant than the S-614G strain. These findings suggest that the P.1 variant may be somewhat antigenically different from the early and contemporary strains of SARS-CoV-2.
- Published
- 2021
- Full Text
- View/download PDF
29. Zika virus infection of pregnant Ifnar1−/− mice triggers strain-specific differences in fetal outcomes
- Author
-
Anna S. Jaeger, Shelby L. O’Connor, Kevin K. Noguchi, Jennifer T. Vangorder-Braid, Ellie K. Bohm, Matthew T. Aliota, Michael K. Fritsch, Natalie C. Bennett, Nicole A. Fuhler, Ryan V. Moriarty, and John J. Baczenas
- Subjects
Microcephaly ,Fetus ,Flavivirus ,Pregnancy ,biology ,Genetic heterogeneity ,medicine ,Physiology ,Viremia ,Abortion ,medicine.disease ,biology.organism_classification ,Zika virus - Abstract
Zika virus (ZIKV) is a flavivirus that causes a constellation of adverse fetal outcomes collectively termed Congenital Zika Syndrome (CZS). However, not all pregnancies exposed to ZIKV result in an infant with apparent defects. During the 2015-2016 American outbreak of ZIKV, CZS rates varied by geographic location. The underlying mechanisms responsible for this heterogeneity in outcomes have not been well defined. Therefore, we sought to characterize and compare the pathogenic potential of multiple Asian/American-lineage ZIKV strains in an established Ifnar1−/− pregnant mouse model. Here, we show significant differences in the rate of fetal demise following maternal inoculation with ZIKV strains from Puerto Rico, Panama, Mexico, Brazil, and Cambodia. Rates of fetal demise broadly correlated with maternal viremia but were independent of fetus and placenta virus titer, indicating that additional underlying factors contribute to fetus outcome. Our results, in concert with those from other studies, suggest that subtle differences in ZIKV strains may have important phenotypic impacts. With ZIKV now endemic in the Americas, greater emphasis needs to be placed on elucidating and understanding the underlying mechanisms that contribute to fetal outcome.IMPORTANCEZika virus (ZIKV) actively circulates in 89 countries and territories around the globe. ZIKV infection during pregnancy is associated with adverse fetal outcomes including birth defects, microcephaly, neurological complications, and even spontaneous abortion. Rates of adverse fetal outcomes vary between regions, and not every pregnancy exposed to ZIKV results in birth defects. Not much is known about how or if the infecting ZIKV strain is linked to fetal outcomes. Our research provides evidence of phenotypic heterogeneity between Asian/American-lineage ZIKV strains and provides insight into the underlying causes of adverse fetal outcomes. Understanding ZIKV strain-dependent pathogenic potential during pregnancy and elucidating underlying causes of diverse clinical sequelae observed during human infections is critical to understanding ZIKV on a global scale.
- Published
- 2021
- Full Text
- View/download PDF
30. Spontaneous control of SIV replication does not prevent immune dysregulation and bacterial dissemination in animals co-infected with M. tuberculosis
- Author
-
Erica C. Larson, Hopkins F, Shelby L. O’Connor, Sarah M. Fortune, Alexis J. Balgeman, Charles A. Scanga, Michael R. Chase, Mark Rodgers, Ryan V. Moriarty, Amy L. Ellis, and Pauline Maiello
- Subjects
education.field_of_study ,Tuberculosis ,biology ,animal diseases ,viruses ,Population ,virus diseases ,Simian immunodeficiency virus ,Immune dysregulation ,medicine.disease_cause ,biology.organism_classification ,medicine.disease ,Virology ,Mycobacterium tuberculosis ,Immune system ,TIGIT ,medicine ,Tumor necrosis factor alpha ,education - Abstract
Individuals infected with both HIV and Mycobacterium tuberculosis (Mtb) are more likely to develop severe Tuberculosis (TB) disease than HIV-naïve individuals. To understand how a chronic pre-existing Simian immunodeficiency virus (SIV) infection impairs the early immune response to Mtb, we used the Mauritian cynomolgus macaque (MCM) model of SIV/Mtb co-infection. We examined the relationship between peripheral viral control and Mtb burden, Mtb dissemination, and immunological function between SIV+ spontaneous controllers, SIV+ non-controllers, and SIV-naïve MCM who were challenged with a barcoded Mtb Erdman strain and necropsied six weeks post infection. Mycobacterial burden was highest in the SIV+ non-controllers in all assessed tissues. In lung granulomas, we found the frequency of CD4+ T cells producing TNFα was reduced in all SIV+ MCM, but CD4+ T cells producing IFNγ were only lower in the SIV+ non-controllers. Further, while all SIV+ MCM had more PD1+ and TIGIT+ T cells in the lung granulomas relative to SIV-naïve MCM, SIV+ controllers exhibited the highest frequency of cells expressing these markers. To measure the effect of SIV infection on within-host bacterial dissemination, we sequenced the molecular barcodes of Mtb present in each tissue and characterized the complexity of the Mtb populations. While Mtb population complexity was not associated with infection group, lymph nodes had increased complexity when compared to lung granulomas across all groups. These results provide evidence SIV+ animals, independent of viral control, exhibit dysregulated immune responses and enhanced dissemination of Mtb, likely contributing to the poor TB disease course across all SIV/Mtb co-infected animals.ImportanceHIV and TB remain significant global health issues, despite the availability of treatments. Individuals with HIV, including those who are virally suppressed, are at an increased risk to develop and succumb to severe TB disease when compared to HIV-naïve individuals. Our study aims to understand the relationship between SIV replication, mycobacterial growth, and immunological function in the tissues of co-infected Mauritian cynomolgus macaques during the early phase of Mtb infection. Here we demonstrate that increased viral replication is associated with increased bacterial burden in the tissues and impaired immunologic responses, and that the damage attributed to virus infection is not fully eliminated when animals spontaneously control virus replication.
- Published
- 2021
- Full Text
- View/download PDF
31. The Mucosal Barrier and Anti-Viral Immune Responses can Eliminate Portions of the Viral Population during Transmission and Early Viral Growth
- Author
-
Thomas C. Friedrich, Hannah Schweigert, Vanessa Venturi, Jaffna Mathiaparanam, Andrea M. Weiler, Athena E. Golfinos, Shelby L. O’Connor, Miles P. Davenport, Brandon F. Keele, Dane D. Gellerup, and Alexis J. Balgeman
- Subjects
education.field_of_study ,viruses ,Population ,Simian immunodeficiency virus ,Biology ,medicine.disease_cause ,Virology ,law.invention ,Immune system ,Transmission (mechanics) ,law ,Viral growth ,medicine ,anatomy_morphology ,education - Abstract
Little is known about how individual virus lineages replicating during acute Human Immunodeficiency Virus or Simian Immunodeficiency Virus (HIV/SIV) infection persist into chronic infection. In this study, we use molecularly barcoded SIV (SIVmac239M) to track distinct viral lineages for 12 weeks after intravenous and intrarectal challenge in macaques. Two Mafa-A1*063+ cynomolgus macaques (Macaca fascicularis) were challenged intravenously (IV), and two Mamu-A1*001+ rhesus macaques (Macaca mulatta) were challenged intrarectally (IR) with 200,000 Infectious Units (IU) of SIVmac239M. We deep sequenced the molecular barcode from all animals over 12 weeks to characterize the diversity and persistence of virus lineages, as well as the sequences of T cell epitopes during acute SIV infection. During the first three weeks post-infection, we found ~175-950 times more unique virus lineages circulating in the animals challenged intravenously than those challenged intrarectally, suggesting that challenge route is the primary driver restricting the transmission of individual viral lineages. Additionally, the emergence of escape variants can occur on multiple virus templates simultaneously, but elimination of some templates is likely a consequence of additional host factors. These data imply that virus lineages present during acute infection can be eliminated from the virus population even after initial T cell selection.
- Published
- 2021
32. Initial evaluation of a mobile SARS-CoV-2 RT-LAMP testing strategy
- Author
-
Miranda R Stauss, Elizabeth D Somsen, Olivia E Harwood, Roger W. Wiseman, Matthew R. Reynolds, Hailey E. Bussan, Cecilia G. Shortreed, Corrie B. Burmeister, Ryan V. Moriarty, Laurel M. Stewart, William M. Rehrauer, Thomas C. Friedrich, Katrina N Fauser, Christina M. Newman, Mason Bliss, Julie A. Karl, Amelia K. Haj, Kristi L Hall, David H. O’Connor, Andrea M. Weiler, Anna S. Heffron, Dawn M. Dudley, Chelsea M. Crooks, Shelby L. O’Connor, Trent M. Prall, Matthew T. McLaughlin, Luis A. Haddock, Robert J Maddox, Mitchell D. Ramuta, Emma K. Neumann, and Molly A. Accola
- Subjects
Test strategy ,Saliva ,Concordance ,Loop-mediated isothermal amplification ,Asymptomatic ,Sensitivity and Specificity ,Article ,law.invention ,COVID-19 Testing ,law ,Medicine ,Humans ,Molecular Biology ,Polymerase chain reaction ,business.industry ,SARS-CoV-2 ,RNA ,COVID-19 ,Articles ,Virology ,Reverse transcriptase ,Molecular Diagnostic Techniques ,RNA, Viral ,medicine.symptom ,business ,Nucleic Acid Amplification Techniques - Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) control in the United States remains hampered, in part, by testing limitations. We evaluated a simple, outdoor, mobile, colorimetric reverse transcription loop-mediated isothermal amplification (RT-LAMP) assay workflow where self-collected saliva is tested for SARS-CoV-2 RNA. From July 16 to November 19, 2020, 4,704 surveillance samples were collected from volunteers and tested for SARS-CoV-2 at 5 sites. A total of 21 samples tested positive for SARS-CoV-2 by RT-LAMP; 12 were confirmed positive by subsequent quantitative reverse-transcription polymerase chain reaction (qRT-PCR) testing, while 8 were negative for SARS-CoV-2 RNA, and 1 could not be confirmed because the donor did not consent to further molecular testing. We estimated the RT-LAMP assay’s false-negative rate from July 16 to September 17, 2020 by pooling residual heat-inactivated saliva that was unambiguously negative by RT-LAMP into groups of 6 or less and testing for SARS-CoV-2 RNA by qRT-PCR. We observed a 98.8% concordance between the RT-LAMP and qRT-PCR assays, with only 5 of 421 RT-LAMP negative pools (2,493 samples) testing positive in the more sensitive qRT-PCR assay. Overall, we demonstrate a rapid testing method that can be implemented outside the traditional laboratory setting by individuals with basic molecular biology skills and can effectively identify asymptomatic individuals who would not typically meet the criteria for symptom-based testing modalities.
- Published
- 2021
33. Pre-existing Simian Immunodeficiency Virus Infection Increases Expression of T Cell Markers Associated with Activation during Early
- Author
-
Erica C, Larson, Amy, Ellis-Connell, Mark A, Rodgers, Alexis J, Balgeman, Ryan V, Moriarty, Cassaundra L, Ameel, Tonilynn M, Baranowski, Jaime A, Tomko, Chelsea M, Causgrove, Pauline, Maiello, Shelby L, O'Connor, and Charles A, Scanga
- Subjects
Granuloma ,viruses ,animal diseases ,T-Lymphocytes ,Simian Acquired Immunodeficiency Syndrome ,virus diseases ,Mycobacterium tuberculosis ,respiratory system ,CD8-Positive T-Lymphocytes ,bacterial infections and mycoses ,Article ,Tumor Necrosis Factors ,Animals ,Macaca ,Tuberculosis ,Simian Immunodeficiency Virus ,Biomarkers - Abstract
Tuberculosis (TB) is the leading infectious cause of death among people living with HIV (PLHIV). PLHIV are more susceptible to contracting Mycobacterium tuberculosis (Mtb) and often have worsened TB disease. Understanding the immunologic defects caused by HIV and the consequences it has on Mtb co-infection is critical in combating this global health epidemic. We previously showed in a model of simian immunodeficiency virus (SIV) and Mtb co-infection in Mauritian cynomolgus macaques (MCM) that SIV/Mtb co-infected MCM had rapidly progressive TB. We hypothesized that pre-existing SIV infection impairs early T cell responses to Mtb infection. We infected MCM with SIVmac239 followed by co-infection with Mtb Erdman 6 months later. While similar TB progression was observed in both SIV+ and SIV-naïve animals at six weeks post-Mtb infection, longitudinal sampling of the blood (PBMC) and airways (BAL) revealed a significant reduction in circulating CD4+ T cells and an influx of CD8+ T cells in airways of SIV+ animals. At sites of Mtb infection (i.e. granulomas), SIV/Mtb co-infected animals had a higher proportion of CD4+ and CD8+ T cells expressing PD-1 and TIGIT. In addition, there were fewer TNF-producing CD4+ T cells in granulomas of SIV/Mtb co-infected animals. Taken together, we show that concurrent SIV infection alters T cell phenotypes in granulomas during the early stages of TB disease. As it is critical to establish control of Mtb replication soon after infection, these phenotypic changes may distinguish the immune dysfunction that arises from pre-existing SIV infection which promotes TB progression.
- Published
- 2021
34. Validation of multiplex PCR sequencing assay of SIV
- Author
-
Shelby L. O’Connor, Ryan V. Moriarty, Nicolas Fesser, Vanessa Venturi, Miles P. Davenport, Timothy E. Schlub, and Matthew S. Sutton
- Subjects
0301 basic medicine ,Deep sequencing ,viruses ,Population ,Simian Acquired Immunodeficiency Syndrome ,Genome, Viral ,Biology ,medicine.disease_cause ,Sensitivity and Specificity ,lcsh:Infectious and parasitic diseases ,03 medical and health sciences ,SNV detection ,Virology ,Complementary DNA ,Multiplex polymerase chain reaction ,medicine ,False positive paradox ,Animals ,Humans ,lcsh:RC109-216 ,education ,education.field_of_study ,030102 biochemistry & molecular biology ,Research ,High-Throughput Nucleotide Sequencing ,Reproducibility of Results ,COVID-19 ,Replicate ,Simian immunodeficiency virus ,Multiplex PCR ,Macaca mulatta ,Coronavirus ,Titer ,030104 developmental biology ,Infectious Diseases ,SIV ,DNA, Viral ,RNA, Viral ,Simian Immunodeficiency Virus ,Multiplex Polymerase Chain Reaction - Abstract
Background The generation of accurate and reproducible viral sequence data is necessary to understand the diversity present in populations of RNA viruses isolated from clinical samples. While various sequencing methods are available, they often require high quality templates and high viral titer to ensure reliable data. Methods We modified a multiplex PCR and sequencing approach to characterize populations of simian immunodeficiency virus (SIV) isolated from nonhuman primates. We chose this approach with the aim of reducing the number of required input templates while maintaining fidelity and sensitivity. We conducted replicate sequencing experiments using different numbers of quantified viral RNA (vRNA) or viral cDNA as input material. We performed assays with clonal SIVmac239 to detect false positives, and we mixed SIVmac239 and a variant with 24 point mutations (SIVmac239-24X) to measure variant detection sensitivity. Results We found that utilizing a starting material of quantified viral cDNA templates had a lower rate of false positives and increased reproducibility when compared to that of quantified vRNA templates. This study identifies the importance of rigorously validating deep sequencing methods and including replicate samples when using a new method to characterize low frequency variants in a population with a small number of templates. Conclusions Because the need to generate reproducible and accurate sequencing data from diverse viruses from low titer samples, we modified a multiplex PCR and sequencing approach to characterize SIV from populations from non-human primates. We found that increasing starting template numbers increased the reproducibility and decreased the number of false positives identified, and this was further seen when cDNA was used as a starting material. Ultimately, we highlight the importance of vigorously validating methods to prevent overinterpretation of low frequency variants in a sample.
- Published
- 2021
- Full Text
- View/download PDF
35. The mucosal barrier and anti-viral immune responses can eliminate portions of the viral population during transmission and early viral growth
- Author
-
Hannah Schweigert, Thomas C. Friedrich, Andrea M. Weiler, Ryan V. Moriarty, Alexis J. Balgeman, Miles P. Davenport, Vanessa Venturi, Brandon F. Keele, Dane D. Gellerup, Athena E. Golfinos, Jaffna Mathiaparanam, and Shelby L. O’Connor
- Subjects
RNA viruses ,T-Lymphocytes ,viruses ,Simian Acquired Immunodeficiency Syndrome ,Artificial Gene Amplification and Extension ,Monkeys ,Pathology and Laboratory Medicine ,medicine.disease_cause ,Polymerase Chain Reaction ,Macaque ,Epitope ,White Blood Cells ,Epitopes ,Immunodeficiency Viruses ,Animal Cells ,Medicine and Health Sciences ,Mammals ,education.field_of_study ,Multidisciplinary ,Ecology ,biology ,T Cells ,Eukaryota ,Animal Models ,Viral Load ,SIV ,Experimental Organism Systems ,Medical Microbiology ,Viral Pathogens ,Vertebrates ,Viruses ,Gene Products, tat ,Injections, Intravenous ,RNA, Viral ,Medicine ,Female ,Simian Immunodeficiency Virus ,Cellular Types ,Pathogens ,Viral load ,Research Article ,Primates ,Ecological Metrics ,Immune Cells ,Science ,Immunology ,Population ,Research and Analysis Methods ,Microbiology ,Virus ,Immune system ,Virology ,biology.animal ,Old World monkeys ,Retroviruses ,medicine ,Animals ,Viremia ,Molecular Biology Techniques ,education ,Microbial Pathogens ,Molecular Biology ,Blood Cells ,Mucous Membrane ,Biology and life sciences ,Rhesus Monkeys ,Lentivirus ,Ecology and Environmental Sciences ,Organisms ,Species Diversity ,Cell Biology ,Reverse Transcriptase-Polymerase Chain Reaction ,Simian immunodeficiency virus ,Macaca mulatta ,Viral Replication ,Macaca fascicularis ,Chronic infection ,Amniotes ,Mutation ,Animal Studies ,Zoology - Abstract
Little is known about how specific individual viral lineages replicating systemically during acute Human Immunodeficiency Virus or Simian Immunodeficiency Virus (HIV/SIV) infection persist into chronic infection. In this study, we use molecularly barcoded SIV (SIVmac239M) to track distinct viral lineages for 12 weeks after intravenous (IV) or intrarectal (IR) challenge in macaques. Two Mafa-A1*063+ cynomolgus macaques (Macaca fascicularis, CM) were challenged IV, and two Mamu-A1*001+ rhesus macaques (Macaca mulatta, RM) were challenged IR with 200,000 Infectious Units (IU) of SIVmac239M. We sequenced the molecular barcode of SIVmac239M from all animals over the 12 weeks of the study to characterize the diversity and persistence of virus lineages. During the first three weeks post-infection, we found ~70–560 times more unique viral lineages circulating in the animals challenged IV compared to those challenged IR, which is consistent with the hypothesis that the challenge route is the primary driver restricting the transmission of individual viral lineages. We also characterized the sequences of T cell epitopes targeted during acute SIV infection, and found that the emergence of escape variants in acutely targeted epitopes can occur on multiple virus templates simultaneously, but that elimination of some of these templates is likely a consequence of additional host factors. These data imply that virus lineages present during acute infection can still be eliminated from the systemic virus population even after initial selection.
- Published
- 2021
36. Pre-existing SIV infection increases expression of T cell markers associated with activation during early Mycobacterium tuberculosis co-infection and impairs TNF responses in granulomas
- Author
-
Erica C. Larson, Amy L. Ellis, Alexis J. Balgeman, Charles A. Scanga, Pauline Maiello, Cassaundra Ameel, Chelsea M Causgrove, Mark Rodgers, Shelby L. O’Connor, Tonilynn M Baranowski, Ryan V. Moriarty, and Jaime A Tomko
- Subjects
Tuberculosis ,biology ,T cell ,respiratory system ,Simian immunodeficiency virus ,biology.organism_classification ,medicine.disease_cause ,Acquired immune system ,medicine.disease ,Virology ,Mycobacterium tuberculosis ,medicine.anatomical_structure ,Immune system ,TIGIT ,medicine ,CD8 - Abstract
Tuberculosis (TB) is the leading infectious cause of death among people living with HIV (PLHIV). PLHIV are more susceptible to contracting Mycobacterium tuberculosis (Mtb) infection and often have worsened TB disease. Understanding the immunologic defects caused by HIV and the consequences it has on Mtb co-infection is critical in combating this global health epidemic. We previously established a model of simian immunodeficiency virus (SIV) and Mtb co-infection in Mauritian cynomolgus macaques (MCM), and showed that SIV/Mtb co-infected MCM had rapidly progressive TB. We hypothesized that pre-existing SIV infection impairs early T cell responses to Mtb infection. To test our hypothesis, we infected MCM with SIVmac239 intrarectally followed by co-infection with a low dose of Mtb Erdman 6 months later. SIV-naïve MCM were infected with Mtb alone as controls. Six weeks after Mtb infection, animals were necropsied and immune responses were measured by multiparameter flow cytometry. While the two groups exhibited similar TB progression at time of necropsy (Nx), longitudinal sampling of the blood (PBMC) and airways (BAL) revealed a significant reduction in circulating CD4+ T cells and an influx of CD8+ T cells in airways following Mtb co-infection of SIV+ animals. Differences in the activation markers CD69, PD-1, and TIGIT were observed. At sites of Mtb infection (i.e. granulomas), SIV/Mtb co-infected animals had a higher proportion of CD4+ and CD8+ T cells expressing PD-1 and TIGIT. In addition, there were fewer TNF-producing CD4+ and CD8+ T cells in granulomas and airways of SIV/Mtb co-infected animals. Taken together, we show that concurrent SIV infection alters T cell phenotypes in granulomas during the early stages of TB disease. As it is critical to establish control of Mtb replication soon after infection, these phenotypic changes may distinguish the immune dysfunction that arises from pre-existing SIV infection which promotes TB progression.Author SummaryPeople living with HIV are incredibly susceptible to TB and, when co-infected with Mtb, often develop serious TB disease. We do not yet understand precisely how HIV infection impairs the early stages of the adaptive immune response against Mtb bacilli. We employed a non-human primate model of HIV, using SIV as a surrogate for HIV, followed by Mtb co-infection to investigate the immunologic defects associated with pre-existing SIV infection over the first six weeks of Mtb co-infection. Our study focused on CD4+ and CD8+ T cells as these cells are known to play an important role in Mtb control. We found more CD8+ T cells in granulomas, the sites of Mtb infection, from SIV/Mtb co-infected animals, with little difference in CD4+ T cells. SIV/Mtb co-infected animals and animals infected with SIV alone had a higher proportion of both CD4+ and CD8+ T cells expressing activation markers compared to SIV-naïve animals, consistent with SIV-dependent immune activation. Notably, we observed a lower proportion of TNF-producing T cells, a cytokine critical for Mtb control, in granulomas and airways of SIV/Mtb co-infected animals. Taken together, these data show that pre-existing SIV alters T cell phenotypes and reduces TNF responses early in Mtb infection.
- Published
- 2020
- Full Text
- View/download PDF
37. Revealing fine-scale spatiotemporal differences in SARS-CoV-2 introduction and spread
- Author
-
Sanjib Bhattacharyya, William M. Rehrauer, David Baker, Yoshihiro Kawaoka, Mitchell D. Ramuta, Molly A. Accola, Trent M. Prall, John J. Baczenas, Manjeet Khubbar, Michael A. Martin, Caitlin S. Pepperell, Peter Halfmann, Kasen K. Riemersma, Katarina M. Braun, Anna S. Heffron, Katia Koelle, Shelby L. O’Connor, Nasia Safdar, Thomas C. Friedrich, Andrea M. Weiler, David H. O’Connor, Gage K. Moreno, Chelsea M. Crooks, and Trivikram Dasu
- Subjects
0301 basic medicine ,Epidemiology ,viruses ,General Physics and Astronomy ,law.invention ,0302 clinical medicine ,law ,Pandemic ,Mass Screening ,030212 general & internal medicine ,Respiratory Protective Devices ,lcsh:Science ,skin and connective tissue diseases ,Socioeconomics ,Molecular Epidemiology ,Multidisciplinary ,Geography ,Viral Epidemiology ,Phylogenetics ,Transmission (mechanics) ,Psychological Distance ,Scale (social sciences) ,Coronavirus Infections ,medicine.medical_specialty ,Science ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,Pneumonia, Viral ,Genome, Viral ,Article ,General Biochemistry, Genetics and Molecular Biology ,Betacoronavirus ,03 medical and health sciences ,Wisconsin ,medicine ,Humans ,Pandemics ,Mass screening ,SARS-CoV-2 ,Public health ,fungi ,COVID-19 ,Outbreak ,General Chemistry ,United States ,body regions ,030104 developmental biology ,Viral infection ,lcsh:Q - Abstract
Evidence-based public health approaches that minimize the introduction and spread of new SARS-CoV-2 transmission clusters are urgently needed in the United States and other countries struggling with expanding epidemics. Here we analyze 247 full-genome SARS-CoV-2 sequences from two nearby communities in Wisconsin, USA, and find surprisingly distinct patterns of viral spread. Dane County had the 12th known introduction of SARS-CoV-2 in the United States, but this did not lead to descendant community spread. Instead, the Dane County outbreak was seeded by multiple later introductions, followed by limited community spread. In contrast, relatively few introductions in Milwaukee County led to extensive community spread. We present evidence for reduced viral spread in both counties following the statewide “Safer at Home” order, which went into effect 25 March 2020. Our results suggest patterns of SARS-CoV-2 transmission may vary substantially even in nearby communities. Understanding these local patterns will enable better targeting of public health interventions., In this study, the authors present an analysis of 247 full-genome SARS-CoV-2 sequences obtained from two communities in Wisconsin, USA, and report distinct patterns of viral spread. Their results suggest that patterns of SARS-CoV-2 transmission and spread may vary substantially, even between neighbouring communities.
- Published
- 2020
- Full Text
- View/download PDF
38. Mathematical modeling of N-803 treatment in SIV-infected non-human primates
- Author
-
Amy L. Ellis-Connell, Jonathan W. Cody, Shelby L. O’Connor, and Elsje Pienaar
- Subjects
RNA viruses ,0301 basic medicine ,Physiology ,viruses ,Lymphocyte ,Cell ,Simian Acquired Immunodeficiency Syndrome ,NK cells ,CD8-Positive T-Lymphocytes ,Virus Replication ,Pathology and Laboratory Medicine ,medicine.disease_cause ,0302 clinical medicine ,Immunodeficiency Viruses ,Drug tolerance ,Cellular types ,Cytotoxic T cell ,Biology (General) ,Interleukin-15 ,Ecology ,Pharmaceutics ,Immune cells ,Viral Load ,Body Fluids ,Killer Cells, Natural ,Blood ,medicine.anatomical_structure ,SIV ,Computational Theory and Mathematics ,Medical Microbiology ,Viral Pathogens ,030220 oncology & carcinogenesis ,Modeling and Simulation ,Viruses ,White blood cells ,Simian Immunodeficiency Virus ,Pathogens ,Anatomy ,Viral load ,Research Article ,Cell biology ,Blood cells ,Drug Research and Development ,QH301-705.5 ,Recombinant Fusion Proteins ,T cell ,Immunology ,T cells ,Cytotoxic T cells ,Biology ,Models, Biological ,Microbiology ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,Immune system ,Drug Therapy ,Virology ,Retroviruses ,Immune Tolerance ,Genetics ,medicine ,Animals ,Humans ,Microbial Pathogens ,Molecular Biology ,Ecology, Evolution, Behavior and Systematics ,Drug Regulation ,Medicine and health sciences ,Pharmacology ,Biology and life sciences ,Lentivirus ,Organisms ,Computational Biology ,Mathematical Concepts ,Simian immunodeficiency virus ,Macaca mulatta ,030104 developmental biology ,Animal cells ,Viral Transmission and Infection ,CD8 - Abstract
Immunomodulatory drugs could contribute to a functional cure for Human Immunodeficiency Virus (HIV). Interleukin-15 (IL-15) promotes expansion and activation of CD8+ T cell and natural killer (NK) cell populations. In one study, an IL-15 superagonist, N-803, suppressed Simian Immunodeficiency Virus (SIV) in non-human primates (NHPs) who had received prior SIV vaccination. However, viral suppression attenuated with continued N-803 treatment, partially returning after long treatment interruption. While there is evidence of concurrent drug tolerance, immune regulation, and viral escape, the relative contributions of these mechanisms to the observed viral dynamics have not been quantified. Here, we utilize mathematical models of N-803 treatment in SIV-infected macaques to estimate contributions of these three key mechanisms to treatment outcomes: 1) drug tolerance, 2) immune regulation, and 3) viral escape. We calibrated our model to viral and lymphocyte responses from the above-mentioned NHP study. Our models track CD8+ T cell and NK cell populations with N-803-dependent proliferation and activation, as well as viral dynamics in response to these immune cell populations. We compared mathematical models with different combinations of the three key mechanisms based on Akaike Information Criterion and important qualitative features of the NHP data. Two minimal models were capable of reproducing the observed SIV response to N-803. In both models, immune regulation strongly reduced cytotoxic cell activation to enable viral rebound. Either long-term drug tolerance or viral escape (or some combination thereof) could account for changes to viral dynamics across long breaks in N-803 treatment. Theoretical explorations with the models showed that less-frequent N-803 dosing and concurrent immune regulation blockade (e.g. PD-L1 inhibition) may improve N-803 efficacy. However, N-803 may need to be combined with other immune therapies to countermand viral escape from the CD8+ T cell response. Our mechanistic model will inform such therapy design and guide future studies., Author summary Immune therapy may be a critical component in the functional cure for Human Immunodeficiency Virus (HIV). N-803 is an immunotherapeutic drug that activates antigen-specific CD8+ T cells of the immune system. These CD8+ T cells eliminate HIV-infected cells in order to limit the spread of infection in the body. In one study, N-803 reduced plasma viremia in macaques that were infected with Simian Immunodeficiency Virus, an analog of HIV. Here, we used mathematical models to analyze the data from this study to better understand the effects of N-803 therapy on the immune system. Our models indicated that inhibitory signals may be reversing the stimulatory effect of N-803. Results also suggested the possibilities that tolerance to N-803 could build up within the CD8+ T cells themselves and that the treatment may be selecting for virus strains that are not targeted by CD8+ T cells. Our models predict that N-803 therapy may be made more effective if the time between doses is increased or if inhibitory signals are blocked by an additional drug. Also, N-803 may need to be combined with other immune therapies to target virus that would otherwise evade CD8+ T cells.
- Published
- 2020
- Full Text
- View/download PDF
39. Distinct patterns of SARS-CoV-2 transmission in two nearby communities in Wisconsin, USA
- Author
-
Gage K. Moreno, Peter Halfmann, John J. Baczenas, Katarina M. Braun, Yoshihiro Kawaoka, David Baker, Andrea M. Weiler, Trent M. Prall, Michael A. Martin, Manjeet Khubbar, Kasen K. Riemersma, Shelby L. O’Connor, William M. Rehrauer, Caitlin S. Pepperell, Chelsea M. Crooks, Molly A. Accola, Anna S. Heffron, Trivikram Dasu, Sanjib Bhattacharyya, Thomas C. Friedrich, Katia Koelle, Nasia Safdar, David H. O’Connor, and Mitchell D. Ramuta
- Subjects
2019-20 coronavirus outbreak ,medicine.medical_specialty ,Wisconsin usa ,Public health ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,Viral transmission ,Outbreak ,law.invention ,Geography ,Transmission (mechanics) ,law ,medicine ,Viral spread ,Socioeconomics - Abstract
Evidence-based public health approaches that minimize the introduction and spread of new SARS-CoV-2 transmission clusters are urgently needed in the United States and other countries struggling with expanding epidemics. Here we analyze 247 full-genome SARS-CoV-2 sequences from two nearby communities in Wisconsin, USA, and find surprisingly distinct patterns of viral spread. Dane County had the 12th known introduction of SARS-CoV-2 in the United States, but this did not lead to descendant community spread. Instead, the Dane County outbreak was seeded by multiple later introductions, followed by limited community spread. In contrast, relatively few introductions in Milwaukee County led to extensive community spread. We present evidence for reduced viral spread in both counties, and limited viral transmission between counties, following the statewide “Safer at Home” public health order, which went into effect 25 March 2020. Our results suggest that early containment efforts suppressed the spread of SARS-CoV-2 within Wisconsin.
- Published
- 2020
- Full Text
- View/download PDF
40. MAIT cells are functionally impaired in a Mauritian cynomolgus macaque model of SIV and Mtb co-infection
- Author
-
Alexis J. Balgeman, Cassaundra Ameel, Shelby L. O’Connor, Jennifer A Juno, Charles A. Scanga, Amy L. Ellis, Pauline Maiello, Tonilynn M Baranowski, Mark Rodgers, Erica C. Larson, and Nadean M Kannal
- Subjects
RNA viruses ,Cell ,Programmed Cell Death 1 Receptor ,Simian Acquired Immunodeficiency Syndrome ,Monkeys ,Pathology and Laboratory Medicine ,White Blood Cells ,Spectrum Analysis Techniques ,Immunodeficiency Viruses ,Animal Cells ,Medicine and Health Sciences ,Cytotoxic T cell ,Biology (General) ,Receptors, Immunologic ,Mammals ,0303 health sciences ,Granuloma ,biology ,medicine.diagnostic_test ,Coinfection ,T Cells ,030302 biochemistry & molecular biology ,Eukaryota ,respiratory system ,Flow Cytometry ,Actinobacteria ,medicine.anatomical_structure ,Infectious Diseases ,SIV ,Medical Microbiology ,Spectrophotometry ,Viral Pathogens ,Viruses ,Vertebrates ,Granulomas ,Tumor necrosis factor alpha ,Simian Immunodeficiency Virus ,Lymph ,Cytophotometry ,Pathogens ,Cellular Types ,Macaque ,Research Article ,Primates ,QH301-705.5 ,T cell ,Immune Cells ,Immunology ,Cytotoxic T cells ,Research and Analysis Methods ,Microbiology ,Mucosal-Associated Invariant T Cells ,Flow cytometry ,Mycobacterium tuberculosis ,03 medical and health sciences ,TIGIT ,Virology ,Retroviruses ,Old World monkeys ,Genetics ,medicine ,Animals ,Molecular Biology ,Tuberculosis, Pulmonary ,Microbial Pathogens ,030304 developmental biology ,Blood Cells ,Bacteria ,Lentivirus ,Organisms ,Biology and Life Sciences ,Cell Biology ,RC581-607 ,biology.organism_classification ,bacterial infections and mycoses ,Macaca fascicularis ,Co-Infections ,Amniotes ,Parasitology ,Lymph Nodes ,Immunologic diseases. Allergy - Abstract
Mucosal-associated invariant T (MAIT) cells can recognize and respond to some bacterially infected cells. Several in vitro and in vivo models of Mycobacterium tuberculosis (Mtb) infection suggest that MAIT cells can contribute to control of Mtb, but these studies are often cross-sectional and use peripheral blood cells. Whether MAIT cells are recruited to Mtb-affected granulomas and lymph nodes (LNs) during early Mtb infection and what purpose they might serve there is less well understood. Furthermore, whether HIV/SIV infection impairs MAIT cell frequency or function at the sites of Mtb replication has not been determined. Using Mauritian cynomolgus macaques (MCM), we phenotyped MAIT cells in the peripheral blood and bronchoalveolar lavage (BAL) before and during infection with SIVmac239. To test the hypothesis that SIV co-infection impairs MAIT cell frequency and function within granulomas, SIV+ and -naïve MCM were infected with a low dose of Mtb Erdman, and necropsied at 6 weeks post Mtb-challenge. MAIT cell frequency and function were examined within the peripheral blood, BAL, and Mtb-affected lymph nodes (LN) and granulomas. MAIT cells did not express markers indicative of T cell activation in response to Mtb in vivo within granulomas in animals infected with Mtb alone. SIV and Mtb co-infection led to increased expression of the activation/exhaustion markers PD-1 and TIGIT, and decreased ability to secrete TNFα when compared to SIV-naïve MCM. Our study provides evidence that SIV infection does not prohibit the recruitment of MAIT cells to sites of Mtb infection, but does functionally impair those MAIT cells. Their impaired function could have impacts, either direct or indirect, on the long-term containment of TB disease., Author summary MAIT cells are a population of immune cells that can directly detect and destroy some bacterially infected cells. Evidence suggests that MAIT cells may play a role in control of Mycobacterium tuberculosis (Mtb) infection, but few studies have examined MAIT cell activity within granulomas, which are the sites of Mtb replication. In addition, chronic HIV infection has been shown to impair the frequency and function of MAIT cells in humans, but these studies focus on peripheral blood and not the sites of Mtb infection. Here, we used a macaque model of SIV and Mtb co-infection to determine whether SIV, as a model for HIV, could dysregulate MAIT cells in tissues where Mtb replication is occurring. SIV co-infection did not affect the absolute numbers of MAIT cells present within granulomas but did impair the ability of the MAIT cells to respond to mycobacteria both in vivo and in vitro. Overall, our study provides evidence that SIV infection alters MAIT cells phenotypically, and impairs MAIT cell function. What effect this might have on antimycobacterial immunity is an avenue for future exploration.
- Published
- 2020
41. MAIT cells are minimally responsive to Mycobacterium tuberculosis within granulomas, but are functionally impaired by SIV in a macaque model of SIV and Mtb co-infection
- Author
-
Jennifer A Juno, Tonilynn M Baranowski, Charles A. Scanga, Amy L. Ellis-Connell, Erica C. Larson, Mark Rodgers, Alexis J. Balgeman, Shelby L. O’Connor, Pauline Maiello, and Cassaundra Ameel
- Subjects
Mycobacterium tuberculosis ,Immune system ,medicine.anatomical_structure ,biology ,Immunology ,Cell ,medicine ,Secretion ,Tumor necrosis factor alpha ,biology.organism_classification ,Cell activation ,Ex vivo ,In vitro - Abstract
Mucosal associated invariant T (MAIT) cells recognize and can directly destroy bacterially infected cells. While a role for MAIT cells has been suggested in several in vitro and in vivo models of M.tuberculosis (Mtb) infection, these studies have often focused on MAIT cells within the peripheral blood or are cross-sectional studies rather than longitudinal studies. The role of MAIT cells within granulomas and other sites of Mtb infection is relatively unknown. Furthermore, how HIV/SIV infection might impair MAIT cells at the sites of Mtb infection has not been determined. Using a Mauritian cynomolgus macaque (MCM) model system, we phenotyped MAIT cells in the peripheral blood and BAL prior to and during infection with SIVmac239. To characterize the role of MAIT cells within granulomas, SIV+ and -naïve MCM were infected with a low dose of Mtb for 6 weeks. MAIT cell frequency and function was examined within the peripheral blood, distal airways, as well as within Mtb-affected lymph nodes (LN) and tissues. Surprisingly, we found no evidence of MAIT cell responsiveness to Mtb within granulomas. Additionally, MAIT cells only minimally responded to mycobacterial stimulus in ex vivo functional assays. In contrast, most MAIT cell activation seemed to occur in samples with highly active SIV replication, including blood and SIV-infected LN. Finally, the ability of MAIT cells to secrete TNFα (TNF) was impaired during SIV and Mtb co-infection, indicating that the two pathogens together could have a synergistically deleterious effect on MAIT cell function. The effect of this functional impairment on overall TB disease burden was unclear, but might be deleterious if MAIT cells are needed to fully activate antimycobacterial immune cells within the granulomas.
- Published
- 2020
- Full Text
- View/download PDF
42. Characterization of major histocompatibility complex-related molecule 1 sequence variants in non-human primates
- Author
-
Shelby L. O’Connor, Nadean M Kannal, Amy L. Ellis-Connell, and Alexis J. Balgeman
- Subjects
Primates ,0301 basic medicine ,Immunology ,Antigen presentation ,Population genetics ,Biology ,Major histocompatibility complex ,Macaque ,Mucosal-Associated Invariant T Cells ,Article ,Cell Line ,Conserved sequence ,Minor Histocompatibility Antigens ,03 medical and health sciences ,0302 clinical medicine ,biology.animal ,Genetics ,Animals ,Humans ,Allele ,Alleles ,Antigen Presentation ,Histocompatibility Antigens Class I ,HEK 293 cells ,Callithrix ,030104 developmental biology ,Cell culture ,biology.protein ,Macaca ,030215 immunology - Abstract
The Major Histocompatibility complex (MHC) class I-related molecule, MR1, presents vitamin B metabolites from bacteria and yeast to Mucosal-associated invariant T (MAIT) cells. Despite the evolutionary conservation of MR1, we do not know whether different allele variants of MR1 exist within the nonhuman primate (NHP) populations that are commonly used for biomedical research. In this study, we identified 21 distinct MR1 nucleotide sequences representing 32 different alleles across five different NHP populations. The majority of the alleles conferring amino acid changes (allele variants) were found in or near the alpha-1 domain of the mature MR1 protein. We expressed four of the most commonly observed MR1 allele variants in 293T cells, and we found that each variant could present bacterial metabolites on the cell surface. We successfully induced cytokine production in macaque MAIT cells stimulated with 293T cells expressing the four most common MR1 allele variants, demonstrating the usefulness of these cell lines to study MAIT cell activity. Our data suggests that MR1 is not monomorphic, but that there are multiple MR1 alleles in NHPs. The materials we describe here will be valuable for characterizing differences in MR1 antigen presentation and MAIT cell function in NHPs.
- Published
- 2018
- Full Text
- View/download PDF
43. SIV progenitor evolution toward HIV: A humanized mouse surrogate model for SIVsm adaptation toward HIV-2
- Author
-
Shelby L. O’Connor, Kimberly Schmitt, Ramesh Akkina, Preston A. Marx, Leila Remling-Mulder, Dipu Mohan Kumar, Stephanie L. Feely, James Curlin, and Mark D. Stenglein
- Subjects
0301 basic medicine ,animal diseases ,Human immunodeficiency virus (HIV) ,Virulence ,Biology ,medicine.disease_cause ,Article ,Cercocebus atys ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Surrogate model ,medicine ,Animals ,Humans ,Serial Passage ,Progenitor cell ,Progenitor ,General Veterinary ,Transmission (medicine) ,virus diseases ,Viral Load ,Virology ,Disease Models, Animal ,030104 developmental biology ,HIV-2 ,Humanized mouse ,Simian Immunodeficiency Virus ,Animal Science and Zoology ,Adaptation ,030215 immunology - Abstract
How SIV progenitors evolved into deadly HIV-1 and HIV-2 following initial cross-species transmission still remains a mystery. Here we used humanized mice as a human surrogate system to evaluate SIVsm evolution into HIV-2. Increased viral virulence to human CD4(+) T cells and adaptive genetic changes were observed during serial passages.
- Published
- 2018
- Full Text
- View/download PDF
44. Using barcoded Zika virus to assess virus population structure in vitro and in Aedes aegypti mosquitoes
- Author
-
David H. O’Connor, Gregory D. Ebel, Thomas C. Friedrich, Selene M. Garcia, Claudia Rückert, Alex D. Byas, Shelby L. O’Connor, Matthew T. Aliota, and James Weger-Lucarelli
- Subjects
0301 basic medicine ,viruses ,Population ,Aedes aegypti ,Genome ,Arbovirus ,Deep sequencing ,Article ,Virus ,Cell Line ,Zika virus ,Flaviviridae ,03 medical and health sciences ,Aedes ,Virology ,medicine ,Animals ,education ,Genetics ,education.field_of_study ,biology ,Genetic Variation ,Haplorhini ,Zika Virus ,biology.organism_classification ,medicine.disease ,Flavivirus ,030104 developmental biology ,Viral evolution - Abstract
Arboviruses such as Zika virus (ZIKV, Flaviviridae; Flavivirus) replicate in both mammalian and insect hosts where they encounter a variety of distinct host defenses. To overcome these pressures, arboviruses exist as diverse populations of distinct genomes. However, transmission between hosts and replication within hosts can involve genetic bottlenecks, during which population size and viral diversity may be significantly reduced, potentially resulting in large fitness losses. Understanding the points at which bottlenecks exist during arbovirus transmission is critical to identifying targets for preventing transmission. To study these bottleneck effects, we constructed 4 “barcoded” ZIKV clones - 2 with an 8-base-pair degenerate insertion in the 3’ UTR and 2 with 8 or 9 degenerate synonymous changes in the coding sequence, theoretically containing thousands of variants each. We passaged these viruses 3 times each in 2 mammalian and 2 mosquito cell lines and characterized selection of the “barcode” populations using deep sequencing. Additionally, the viruses were used to feed three recently field-caught populations of Aedes aegypti mosquitoes to assess bottlenecks in a natural host. The barcoded viruses replicated well in multiple cell lines in vitro and in vivo in mosquitoes and could be characterized using next-generation sequencing. The stochastic nature of mosquito transmission was clearly shown by tracking individual barcodes in Ae. aegypti mosquitoes. Barcoded viruses provide an efficient method to examine bottlenecks during virus infection.AUTHOR SUMMARYIn general, mosquito-borne viruses like ZIKV must replicate in two very different host environments: an insect and a mammalian host. RNA viruses such as ZIKV must maintain genetic diversity in order to adapt to these changing conditions. During this transmission cycle, several barriers exist which can severely restrict viral genetic diversity, causing bottlenecks in the virus population. It is critical to understand these bottlenecks during virus transmission as this will provide important insights into the selective forces shaping arbovirus evolution within and between hots. Here, we employ a set of barcoded ZIKV constructs containing a degenerate stretch of nucleotides that can be tracked using next-generation sequencing. We found that the insertion site in the genome was an important determinant of the resulting diversity of the genetic barcode. We also found that bottlenecks varied between different mosquito populations and patterns of genetic diversity were distinct among individual mosquitoes within a single population, highlighting the randomness of virus dissemination in mosquitoes. Our study characterizes a new tool for tracking bottlenecks during virus transmission in vivo and highlights the importance of both viral and host factors on the maintenance of viral diversity.
- Published
- 2018
- Full Text
- View/download PDF
45. Translating viral vaccines into immunity
- Author
-
Shelby L. O’Connor and Jennifer A Juno
- Subjects
2019-20 coronavirus outbreak ,Multidisciplinary ,Coronavirus disease 2019 (COVID-19) ,business.industry ,Viral Vaccine ,Immunogenicity ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,Viral Vaccines ,Mucosal associated invariant T cell ,biochemical phenomena, metabolism, and nutrition ,medicine.disease_cause ,Virology ,Mucosal-Associated Invariant T Cells ,Article ,Adenoviridae ,Immunity, Humoral ,Immunogenicity, Vaccine ,Immunity ,medicine ,business - Abstract
Mucosal-associated invariant T (MAIT) cells are innate sensors of viruses and can augment early immune responses and contribute to protection. We hypothesized MAIT cells may have inherent adjuvant activity in vaccine platforms that employ replication-incompetent adenovirus vectors. In mice and humans, ChAdOx1 (chimpanzee adenovirus Ox1) immunization robustly activated MAIT cells. Activation required plasmacytoid dendritic cell (pDC)–derived IFN-α and monocyte-derived IL-18. IFN-α-induced, monocyte-derived TNF was also identified as a key secondary signal. All three cytokines were required in vitro and in vivo. Activation of MAIT cells positively correlated with vaccine-induced T cell responses in human volunteers and MAIT cell-deficient mice displayed impaired CD8(+) T cell responses to multiple vaccine-encoded antigens. Thus, MAIT cells contribute to the immunogenicity of adenovirus vectors, with implications for vaccine design.
- Published
- 2021
- Full Text
- View/download PDF
46. Reactivation of simian immunodeficiency virus reservoirs in the brain of virally suppressed macaques
- Author
-
Kelly A. Metcalf Pate, Erin N. Shirk, Ming Li, Janice E. Clements, Greg M. Laird, Robert F. Siliciano, Sally Price, Carine Van Lint, Luiz Francisco Pianowski, Shelby L. O’Connor, Stephen W. Wietgrefe, Lucio Gama, Celina M. Abreu, and Ashley T. Haase
- Subjects
viral reservoir ,Central Nervous System ,0301 basic medicine ,Genotyping Techniques ,viruses ,Simian Acquired Immunodeficiency Syndrome ,HIV Infections ,medicine.disease_cause ,Polymerase Chain Reaction ,Macaque ,Plasma ,0302 clinical medicine ,Cerebrospinal fluid ,Basic Science ,Virus latency ,Immunology and Allergy ,In Situ Hybridization ,Phylogeny ,Cerebrospinal Fluid ,Viral Load ,Virus Latency ,3. Good health ,Infectious Diseases ,medicine.anatomical_structure ,Anti-Retroviral Agents ,vorinostat ,Viral load ,simian immunodeficiency virus ,latency reversing agents ,brain ,Immunology ,Central nervous system ,Neurocognitive Disorders ,Enzyme-Linked Immunosorbent Assay ,In situ hybridization ,Biology ,03 medical and health sciences ,biology.animal ,medicine ,Animals ,Humans ,latency ,Virus Activation ,Gene Products, env ,Sequence Analysis, DNA ,Simian immunodeficiency virus ,medicine.disease ,Virology ,Disease Models, Animal ,030104 developmental biology ,HIV-1 ,ingenol-B ,Macaca ,030217 neurology & neurosurgery - Abstract
Objective Resting CD4 T cells have been recognized as the major cell reservoir of latent HIV-1 during antiretroviral therapy (ART). Using an simian immunodeficiency virus (SIV)/macaque model for AIDS and HIV-related neurocognitive disorders we assessed the contribution of the brain to viral latency and reactivation. Design Pigtailed macaques were dual inoculated with SIVDeltaB670 and SIV17E-Fr and treated with an efficacious central nervous system-penetrant ART. After 500 days of viral suppression animals were treated with two cycles of latency reversing agents and increases in viral transcripts were examined. Methods Longitudinal plasma and cerebrospinal fluid (CSF) viral loads were analyzed by quantitative and digital droplet PCR. After necropsy, viral transcripts in organs were analyzed by PCR, in-situ hybridization, and phylogenetic genotyping based on env V1 loop sequences. Markers for neuronal damage and CSF activation were measured by ELISA. Results Increases in activation markers and plasma and CSF viral loads were observed in one animal treated with latency reversing agents, despite ongoing ART. SIV transcripts were identified in occipital cortex macrophages by in-situ hybridization and CD68 staining. The most abundant SIV genotype in CSF was unique and expanded independent from viruses found in the periphery. Conclusion The central nervous system harbors latent SIV genomes after long-term viral suppression by ART, indicating that the brain represents a potential viral reservoir and should be seriously considered during AIDS cure strategies.
- Published
- 2017
- Full Text
- View/download PDF
47. Spondweni virus causes fetal harm in a mouse model of vertical transmission and is transmitted by Aedes aegypti mosquitoes
- Author
-
Thomas C. Friedrich, Andrea M. Weiler, Ryan V. Moriarty, David H. O’Connor, Anna S. Jaeger, Sierra Rybarczyk, Michael K. Fritsch, Shelby L. O’Connor, Matthew T. Aliota, and Davis M. Seelig
- Subjects
Spondweni virus ,0303 health sciences ,Fetus ,biology ,Placental histopathology ,030231 tropical medicine ,fungi ,virus diseases ,Aedes aegypti ,biology.organism_classification ,medicine.disease_cause ,Virology ,Culex quinquefasciatus ,3. Good health ,Zika virus ,03 medical and health sciences ,Flavivirus ,0302 clinical medicine ,parasitic diseases ,medicine ,030304 developmental biology - Abstract
Spondweni virus (SPONV) is the most closely related known flavivirus to Zika virus (ZIKV). Its pathogenic potential and vector specificity have not been well defined. SPONV has been found predominantly in Africa, but was recently detected in a pool ofCulex quinquefasciatusmosquitoes in Haiti. Here we show that SPONV can cause significant fetal harm, including demise, comparable to ZIKV, in a mouse model of vertical transmission. Following maternal inoculation, we detected infectious SPONV in placentas and fetuses, along with significant fetal and placental histopathology, together indicating vertical transmission. To test vector competence, we exposedAedes aegyptiandCulex quinquefasciatusmosquitoes to SPONV-infected bloodmeals.Aedes aegypticould efficiently transmit SPONV, whereasCulex quinquefasciatuscould not. Our results suggest that SPONV has the same features that made ZIKV a public health risk.
- Published
- 2019
- Full Text
- View/download PDF
48. CD8β Depletion Does Not Prevent Control of Viral Replication or Protection from Challenge in Macaques Chronically Infected with a Live Attenuated Simian Immunodeficiency Virus
- Author
-
Gabrielle L. Barry, Alexis J. Balgeman, Thomas C. Friedrich, Nancy J. Sullivan, Annie W. Lau-Kilby, Shelby L. O’Connor, Andrea M. Weiler, Amy L. Ellis-Connell, John R. Mascola, Yan Zhou, Matthew S. Sutton, Rosemarie D. Mason, Scott Hetzel, Kristin K. Biris, and Mario Roederer
- Subjects
medicine.drug_class ,CD8 Antigens ,T cell ,Immunology ,Simian Acquired Immunodeficiency Syndrome ,Viremia ,CD8-Positive T-Lymphocytes ,Biology ,Virus Replication ,Monoclonal antibody ,medicine.disease_cause ,Microbiology ,Lymphocyte Depletion ,Plasma ,03 medical and health sciences ,0302 clinical medicine ,T-Lymphocyte Subsets ,In vivo ,Virology ,medicine ,Animals ,Lymph node ,030304 developmental biology ,0303 health sciences ,Viral Load ,Simian immunodeficiency virus ,medicine.disease ,medicine.anatomical_structure ,Viral replication ,Insect Science ,Macaca ,Pathogenesis and Immunity ,Simian Immunodeficiency Virus ,CD8 ,030215 immunology - Abstract
We evaluated the contribution of CD8αβ(+) T cells to control of live-attenuated simian immunodeficiency virus (LASIV) replication during chronic infection and subsequent protection from pathogenic SIV challenge. Unlike previous reports with a CD8α-specific depleting monoclonal antibody (mAb), the CD8β-specific mAb CD8β255R1 selectively depleted CD8αβ(+) T cells without also depleting non-CD8(+) T cell populations that express CD8α, such as natural killer (NK) cells and γδ T cells. Following infusion with CD8β255R1, plasma viremia transiently increased coincident with declining peripheral CD8αβ(+) T cells. Interestingly, plasma viremia returned to predepletion levels even when peripheral CD8αβ(+) T cells did not. Although depletion of CD8αβ(+) T cells in the lymph node (LN) was incomplete, frequencies of these cells were 3-fold lower (P = 0.006) in animals that received CD8β255R1 than in those that received control IgG. It is possible that these residual SIV-specific CD8αβ(+) T cells may have contributed to suppression of viremia during chronic infection. We also determined whether infusion of CD8β255R1 in the LASIV-vaccinated animals increased their susceptibility to infection following intravenous challenge with pathogenic SIVmac239. We found that 7/8 animals infused with CD8β255R1, and 3/4 animals infused with the control IgG, were resistant to SIVmac239 infection. These results suggest that infusion with CD8β255R1 did not eliminate the protection afforded to LASIV vaccination. This provides a comprehensive description of the impact of CD8β255R1 infusion on the immunological composition in cynomolgus macaques, compared to an isotype-matched control IgG, while showing that the control of LASIV viremia and protection from challenge can occur even after CD8β255R1 administration. IMPORTANCE Studies of SIV-infected macaques that deplete CD8(+) T cells in vivo with monoclonal antibodies have provided compelling evidence for their direct antiviral role. These studies utilized CD8α-specific mAbs that target both the major (CD8αβ(+)) and minor (CD8αα(+)) populations of CD8(+) T cells but additionally deplete non-CD8(+) T cell populations that express CD8α, such as NK cells and γδ T cells. In the current study, we administered the CD8β-specific depleting mAb CD8β255R1 to cynomolgus macaques chronically infected with a LASIV to selectively deplete CD8αβ(+) T cells without removing CD8αα(+) lymphocytes. We evaluated the impact on control of virus replication and protection from pathogenic SIVmac239 challenge. These results underscore the utility of CD8β255R1 for studying the direct contribution of CD8αβ(+) T cells in various disease states.
- Published
- 2019
- Full Text
- View/download PDF
49. Myeloid and CD4 T Cells Comprise the Latent Reservoir in Antiretroviral Therapy-Suppressed SIVmac251-Infected Macaques
- Author
-
Lucio Gama, Erin N. Shirk, Ming Li, Janice E. Clements, Joseph L. Mankowski, Shelby L. O’Connor, Claudia R. Avalos, Brandon T. Bullock, Edna A. Ferreira, Celina M. Abreu, Shelby Graham, Kelly A. Metcalf Pate, Lisa M. Mangus, Rebecca T. Veenhuis, Daymond R. Parrilla, Feilim Mac Gabhann, Sarah E. Beck, and Suzanne E. Queen
- Subjects
CD4-Positive T-Lymphocytes ,Male ,Myeloid ,viruses ,Simian Acquired Immunodeficiency Syndrome ,Spleen ,HIV Infections ,Genome, Viral ,Biology ,medicine.disease_cause ,Virus Replication ,Microbiology ,Virus ,Monocytes ,Clinical Science and Epidemiology ,03 medical and health sciences ,0302 clinical medicine ,Latent Virus ,Virology ,medicine ,Animals ,Myeloid Cells ,Lung ,latency ,030304 developmental biology ,0303 health sciences ,Macrophages ,RNA ,HIV ,Simian immunodeficiency virus ,Viral Load ,Antiretroviral therapy ,Macaca mulatta ,QR1-502 ,3. Good health ,Virus Latency ,Disease Models, Animal ,medicine.anatomical_structure ,SIV ,Anti-Retroviral Agents ,Simian Immunodeficiency Virus ,030217 neurology & neurosurgery ,Research Article - Abstract
This study provides further evidence that the latent reservoir is comprised of both CD4+ T cells and myeloid cells. The data presented here suggest that CD4+ T cells and macrophages found throughout tissues in the body can contain replication-competent SIV and contribute to rebound of the virus after treatment interruption. Additionally, we have shown that monocytes in blood contain latent virus and, though not considered a reservoir themselves due to their short life span, could contribute to the size of the latent reservoir upon entering the tissue and differentiating into long-lived macrophages. These new insights into the size and location of the SIV reservoir using a model that is heavily studied in the HIV field could have great implications for HIV-infected individuals and should be taken into consideration with the development of future HIV cure strategies., Human immunodeficiency virus (HIV) eradication or long-term suppression in the absence of antiretroviral therapy (ART) requires an understanding of all viral reservoirs that could contribute to viral rebound after ART interruption. CD4 T cells (CD4s) are recognized as the predominant reservoir in HIV type 1 (HIV-1)-infected individuals. However, macrophages are also infected by HIV-1 and simian immunodeficiency virus (SIV) during acute infection and may persist throughout ART, contributing to the size of the latent reservoir. We sought to determine whether tissue macrophages contribute to the SIVmac251 reservoir in suppressed macaques. Using cell-specific quantitative viral outgrowth assays (CD4-QVOA and MΦ-QVOA), we measured functional latent reservoirs in CD4s and macrophages in ART-suppressed SIVmac251-infected macaques. Spleen, lung, and brain in all suppressed animals contained latently infected macrophages, undetectable or low-level SIV RNA, and detectable SIV DNA. Silent viral genomes with potential for reactivation and viral spread were also identified in blood monocytes, although these cells might not be considered reservoirs due to their short life span. Additionally, virus produced in the MΦ-QVOA was capable of infecting healthy activated CD4s. Our results strongly suggest that functional latent reservoirs in CD4s and macrophages can contribute to viral rebound and reestablishment of productive infection after ART interruption. These findings should be considered in the design and implementation of future HIV cure strategies.
- Published
- 2019
50. Extensive CD8β depletion does not prevent control of viral replication or protection from challenge in macaques chronically infected with a live attenuated simian immunodeficiency virus
- Author
-
Yan Zhou, Alexis J. Balgeman, Rosemarie D. Mason, Amy L. Ellis-Connell, Annie Kilby, Andrea M. Weiler, Scott Hetzel, Matthew S. Sutton, Nancy J. Sullivan, John R. Mascola, Thomas C. Friedrich, Gabrielle L. Barry, Shelby L. O’Connor, Mario Roederer, and Kristin K. Biris
- Subjects
0303 health sciences ,medicine.drug_class ,T cell ,Viremia ,Simian immunodeficiency virus ,Biology ,medicine.disease_cause ,medicine.disease ,Monoclonal antibody ,Virology ,Isotype ,3. Good health ,03 medical and health sciences ,0302 clinical medicine ,medicine.anatomical_structure ,Viral replication ,medicine ,Lymph node ,CD8 ,030304 developmental biology ,030215 immunology - Abstract
We evaluated the contribution of CD8αβ+ T cells on control of live-attenuated simian immunodeficiency virus (LASIV) replication during chronic infection and subsequent protection from pathogenic SIV challenge. Unlike previous reports with a CD8α-specific depleting monoclonal antibody (mAb), the CD8β-specific mAb CD8β255R1 selectively depleted CD8αβ+ T cells without also depleting non-CD8+ T cell populations that express CD8α, such as natural killer (NK) cells and γδ T cells. Following infusion with CD8β255R1, plasma viremia transiently increased coincident with declining peripheral CD8αβ+ T cells. Interestingly, plasma viremia returned to pre-depletion levels even when peripheral CD8αβ+ T cells did not. Although depletion of CD8αβ+ T cells in the lymph node (LN) was incomplete, frequencies of these cells were three-fold lower (p=0.006) in animals that received CD8β255R1 compared to control IgG. It is possible that these residual SIV-specific CD8αβ+ T cells may have contributed to suppression of viremia during chronic infection. We also determined whether infusion of CD8β255R1 in the LASIV-vaccinated animals increased their susceptibility to infection following intravenous challenge with pathogenic SIVmac239. We found that 7/8 animals infused with CD8β255R1, and 3/4 animals infused with the control IgG, were resistant to SIVmac239 infection. These results suggest that infusion with CD8β255R1 did not eliminate the protection afforded to LASIV vaccination. This provides a comprehensive description of the impact of CD8β255R1 infusion on the immunological composition of the host, when compared to an isotype matched control IgG, while showing that the control of LASIV viremia and protection from challenge can occur even after CD8β255R1 administration.ImportanceStudies of SIV-infected macaques that deplete CD8+ T cellsin vivowith monoclonal antibodies have provided compelling evidence for their direct antiviral role. These studies utilized CD8α-specific mAbs that target both the major (CD8αβ+) and minor (CD8αα+) populations of CD8+ T cells, but additionally deplete non-CD8+ T cell populations that express CD8α, such as NK cells and γδ T cells. In the current study, we administered the CD8β-specific depleting mAb CD8β255R1 to cynomolgus macaques chronically infected with a LASIV to selectively deplete CD8αβ+ T cells without removing CD8αα+ lymphocytes. We evaluated the impact on control of virus replication and protection from pathogenic SIVmac239 challenge. These results underscore the utility of CD8β255R1 for studying the direct contribution of CD8αβ+ T cells in various disease states.
- Published
- 2019
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.