91 results on '"Jeremy Smedley"'
Search Results
2. Editorial: Preclinical macaque models of viral diseases
- Author
-
Jeremy Smedley
- Subjects
macaque ,model ,HIV ,SIV ,transplantation ,Zika (ZIKV) ,Immunologic diseases. Allergy ,RC581-607 - Published
- 2023
- Full Text
- View/download PDF
3. Antibody-based CCR5 blockade protects Macaques from mucosal SHIV transmission
- Author
-
Xiao L. Chang, Gabriela M. Webb, Helen L. Wu, Justin M. Greene, Shaheed Abdulhaqq, Katherine B. Bateman, Jason S. Reed, Cleiton Pessoa, Whitney C. Weber, Nicholas Maier, Glen M. Chew, Roxanne M. Gilbride, Lina Gao, Rebecca Agnor, Travis Giobbi, Jeffrey Torgerson, Don Siess, Nicole Burnett, Miranda Fischer, Oriene Shiel, Cassandra Moats, Bruce Patterson, Kush Dhody, Scott Kelly, Nader Pourhassan, Diogo M. Magnani, Jeremy Smedley, Benjamin N. Bimber, Nancy L. Haigwood, Scott G. Hansen, Timothy R. Brown, Lishomwa C. Ndhlovu, and Jonah B. Sacha
- Subjects
Science - Abstract
CCR5 is a co-receptor for many transmitted HIV strains. Here, the authors show that biweekly injection of the CCR5-specific antibody Leronlimab protects rhesus macaques against infection following repeated intrarectal challenges of a CCR5-tropic SHIV.
- Published
- 2021
- Full Text
- View/download PDF
4. Effects of persistent modulation of intestinal microbiota on SIV/HIV vaccination in rhesus macaques
- Author
-
Nichole R. Klatt, Courtney Broedlow, Jessica M. Osborn, Andrew T. Gustin, Sandra Dross, Megan A. O’Connor, Ernesto Coronado, Philip Barnette, Tiffany Hensley-McBain, Alexander S. Zevin, Roshell Muir, Alexander Roederer, Solomon Wangari, Naoto Iwayama, Chul Y. Ahrens, Jeremy Smedley, Cassandra Moats, Rebecca M. Lynch, Elias K. Haddad, Nancy L. Haigwood, Deborah H. Fuller, and Jennifer A. Manuzak
- Subjects
Immunologic diseases. Allergy ,RC581-607 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract An effective vaccine to prevent HIV transmission has not yet been achieved. Modulation of the microbiome via probiotic therapy has been suggested to result in enhanced mucosal immunity. Here, we evaluated whether probiotic therapy could improve the immunogenicity and protective efficacy of SIV/HIV vaccination. Rhesus macaques were co-immunized with an SIV/HIV DNA vaccine via particle-mediated epidermal delivery and an HIV protein vaccine administered intramuscularly with Adjuplex™ adjuvant, while receiving daily oral Visbiome® probiotics. Probiotic therapy alone led to reduced frequencies of colonic CCR5+ and CCR6+ CD4+ T cells. Probiotics with SIV/HIV vaccination led to similar reductions in colonic CCR5+ CD4+ T cell frequencies. SIV/HIV-specific T cell and antibody responses were readily detected in the periphery of vaccinated animals but were not enhanced with probiotic treatment. Combination probiotics and vaccination did not impact rectal SIV/HIV target populations or reduce the rate of heterologous SHIV acquisition during the intrarectal challenge. Finally, post-infection viral kinetics were similar between all groups. Thus, although probiotics were well-tolerated when administered with SIV/HIV vaccination, vaccine-specific responses were not significantly enhanced. Additional work will be necessary to develop more effective strategies of microbiome modulation in order to enhance mucosal vaccine immunogenicity and improve protective immune responses.
- Published
- 2021
- Full Text
- View/download PDF
5. Rapamycin limits CD4+ T cell proliferation in simian immunodeficiency virus–infected rhesus macaques on antiretroviral therapy
- Author
-
Benjamin D. Varco-Merth, William Brantley, Alejandra Marenco, Derick D. Duell, Devin N. Fachko, Brian Richardson, Kathleen Busman-Sahay, Danica Shao, Walter Flores, Kathleen Engelman, Yoshinori Fukazawa, Scott W. Wong, Rebecca L. Skalsky, Jeremy Smedley, Michael K. Axthelm, Jeffrey D. Lifson, Jacob D. Estes, Paul T. Edlefsen, Louis J. Picker, Cheryl M.A. Cameron, Timothy J. Henrich, and Afam A. Okoye
- Subjects
AIDS/HIV ,Medicine - Abstract
Proliferation of latently infected CD4+ T cells with replication-competent proviruses is an important mechanism contributing to HIV persistence during antiretroviral therapy (ART). One approach to targeting this latent cell expansion is to inhibit mTOR, a regulatory kinase involved with cell growth, metabolism, and proliferation. Here, we determined the effects of chronic mTOR inhibition with rapamycin with or without T cell activation in SIV-infected rhesus macaques (RMs) on ART. Rapamycin perturbed the expression of multiple genes and signaling pathways important for cellular proliferation and substantially decreased the frequency of proliferating CD4+ memory T cells (TM cells) in blood and tissues. However, levels of cell-associated SIV DNA and SIV RNA were not markedly different between rapamycin-treated RMs and controls during ART. T cell activation with an anti-CD3LALA antibody induced increases in SIV RNA in plasma of RMs on rapamycin, consistent with SIV production. However, upon ART cessation, both rapamycin and CD3LALA–treated and control-treated RMs rebounded in less than 12 days, with no difference in the time to viral rebound or post-ART viral load set points. These results indicate that, while rapamycin can decrease the proliferation of CD4+ TM cells, chronic mTOR inhibition alone or in combination with T cell activation was not sufficient to disrupt the stability of the SIV reservoir.
- Published
- 2022
- Full Text
- View/download PDF
6. Suppression of human and simian immunodeficiency virus replication with the CCR5-specific antibody Leronlimab in two species.
- Author
-
Xiao L Chang, Jason S Reed, Gabriela M Webb, Helen L Wu, Jimmy Le, Katherine B Bateman, Justin M Greene, Cleiton Pessoa, Courtney Waytashek, Whitney C Weber, Joseph Hwang, Miranda Fischer, Cassandra Moats, Oriene Shiel, Rachele M Bochart, Hugh Crank, Don Siess, Travis Giobbi, Jeffrey Torgerson, Rebecca Agnor, Lina Gao, Kush Dhody, Jacob P Lalezari, Ivo Sah Bandar, Alnor M Carnate, Alina S Pang, Michael J Corley, Scott Kelly, Nader Pourhassan, Jeremy Smedley, Benjamin N Bimber, Scott G Hansen, Lishomwa C Ndhlovu, and Jonah B Sacha
- Subjects
Immunologic diseases. Allergy ,RC581-607 ,Biology (General) ,QH301-705.5 - Abstract
The CCR5-specific antibody Leronlimab is being investigated as a novel immunotherapy that can suppress HIV replication with minimal side effects. Here we studied the virological and immunological consequences of Leronlimab in chronically CCR5-tropic HIV-1 infected humans (n = 5) on suppressive antiretroviral therapy (ART) and in ART-naïve acutely CCR5-tropic SHIV infected rhesus macaques (n = 4). All five human participants transitioned from daily combination ART to self-administered weekly subcutaneous (SC) injections of 350 mg or 700 mg Leronlimab and to date all participants have sustained virologic suppression for over seven years. In all participants, Leronlimab fully occupied CCR5 receptors on peripheral blood CD4+ T cells and monocytes. In ART-naïve rhesus macaques acutely infected with CCR5-tropic SHIV, weekly SC injections of 50 mg/kg Leronlimab fully suppressed plasma viremia in half of the macaques. CCR5 receptor occupancy by Leronlimab occurred concomitant with rebound of CD4+ CCR5+ T-cells in peripheral blood, and full CCR5 receptor occupancy was found in multiple anatomical compartments. Our results demonstrate that weekly, self-administered Leronlimab was safe, well-tolerated, and efficacious for long-term virologic suppression and should be included in the arsenal of safe, easily administered, longer-acting antiretroviral treatments for people living with HIV-1. Trial Registration: ClinicalTrials.gov Identifiers: NCT02175680 and NCT02355184.
- Published
- 2022
- Full Text
- View/download PDF
7. Liver Bacterial Dysbiosis With Non-Tuberculosis Mycobacteria Occurs in SIV-Infected Macaques and Persists During Antiretroviral Therapy
- Author
-
Bridget S. Fisher, Katherine A. Fancher, Andrew T. Gustin, Cole Fisher, Matthew P. Wood, Michael Gale, Benjamin J. Burwitz, Jeremy Smedley, Nichole R. Klatt, Nina Derby, and Donald L. Sodora
- Subjects
HIV/SIV ,microbiome ,liver ,16S rRNA gene ,neutrophils ,Immunologic diseases. Allergy ,RC581-607 - Abstract
Liver disease is a significant contributor to morbidity and mortality in HIV-infected individuals, even during successful viral suppression with combination antiretroviral therapy (cART). Similar to HIV infection, SIV infection of rhesus macaques is associated with gut microbiome dysbiosis and microbial translocation that can be detected systemically in the blood. As microbes leaving the intestines must first pass through the liver via the portal vein, we evaluated the livers of both SIV-infected (SIV+) and SIV-infected cART treated (SIV+cART) rhesus macaques for evidence of microbial changes compared to uninfected macaques. Dysbiosis was observed in both the SIV+ and SIV+cART macaques, encompassing changes in the relative abundance of several genera, including a reduction in the levels of Lactobacillus and Staphylococcus. Most strikingly, we found an increase in the relative abundance and absolute quantity of bacteria within the Mycobacterium genus in both SIV+ and SIV+cART macaques. Multi-gene sequencing identified a species of atypical mycobacteria similar to the opportunistic pathogen M. smegmatis. Phosphatidyl inositol lipoarabinomannan (PILAM) (a glycolipid cell wall component found in atypical mycobacteria) stimulation in primary human hepatocytes resulted in an upregulation of inflammatory transcriptional responses, including an increase in the chemokines associated with neutrophil recruitment (CXCL1, CXCL5, and CXCL6). These studies provide key insights into SIV associated changes in hepatic microbial composition and indicate a link between microbial components and immune cell recruitment in SIV+ and SIV+cART treated macaques.
- Published
- 2022
- Full Text
- View/download PDF
8. The ingenol-based protein kinase C agonist GSK445A is a potent inducer of HIV and SIV RNA transcription.
- Author
-
Afam A Okoye, Rémi Fromentin, Hiroshi Takata, Jessica H Brehm, Yoshinori Fukazawa, Bryan Randall, Marion Pardons, Vincent Tai, Jun Tang, Jeremy Smedley, Michael Axthelm, Jeffrey D Lifson, Louis J Picker, David Favre, Lydie Trautmann, and Nicolas Chomont
- Subjects
Immunologic diseases. Allergy ,RC581-607 ,Biology (General) ,QH301-705.5 - Abstract
Activation of the NF-κB signaling pathway by Protein Kinase C (PKC) agonists is a potent mechanism for human immunodeficiency virus (HIV) latency disruption in vitro. However, significant toxicity risks and the lack of evidence supporting their activity in vivo have limited further evaluation of PKC agonists as HIV latency-reversing agents (LRA) in cure strategies. Here we evaluated whether GSK445A, a stabilized ingenol-B derivative, can induce HIV/simian immunodeficiency virus (SIV) transcription and virus production in vitro and demonstrate pharmacological activity in nonhuman primates (NHP). CD4+ T cells from people living with HIV and from SIV+ rhesus macaques (RM) on antiretroviral therapy (ART) exposed in vitro to 25 nM of GSK445A produced cell-associated viral transcripts as well as viral particles at levels similar to those induced by PMA/Ionomycin, indicating that GSK445A can potently reverse HIV/SIV latency. Importantly, these concentrations of GSK445A did not impair the proliferation or survival of HIV-specific CD8+ T cells, but instead, increased their numbers and enhanced IFN-γ production in response to HIV peptides. In vivo, GSK445A tolerability was established in SIV-naïve RM at 15 μg/kg although tolerability was reduced in SIV-infected RM on ART. Increases in plasma viremia following GSK445A administration were suggestive of increased SIV transcription in vivo. Collectively, these results indicate that GSK445A is a potent HIV/SIV LRA in vitro and has a tolerable safety profile amenable for further evaluation in vivo in NHP models of HIV cure/remission.
- Published
- 2022
- Full Text
- View/download PDF
9. CCR5 Receptor Occupancy Analysis Reveals Increased Peripheral Blood CCR5+CD4+ T Cells Following Treatment With the Anti-CCR5 Antibody Leronlimab
- Author
-
Xiao L. Chang, Helen L. Wu, Gabriela M. Webb, Meenakshi Tiwary, Colette Hughes, Jason S. Reed, Joseph Hwang, Courtney Waytashek, Carla Boyle, Cleiton Pessoa, Andrew W. Sylwester, David Morrow, Karina Belica, Miranda Fischer, Scott Kelly, Nader Pourhassan, Rachele M. Bochart, Jeremy Smedley, Christopher P. Recknor, Scott G. Hansen, and Jonah B. Sacha
- Subjects
CCR5 ,CD4 ,HIV ,receptor occupancy (RO) ,flow cytometry ,antibody ,Immunologic diseases. Allergy ,RC581-607 - Abstract
CCR5 plays a central role in infectious disease, host defense, and cancer progression, thereby making it an ideal target for therapeutic development. Notably, CCR5 is the major HIV entry co-receptor, where its surface density correlates with HIV plasma viremia. The level of CCR5 receptor occupancy (RO) achieved by a CCR5-targeting therapeutic is therefore a critical predictor of its efficacy. However, current methods to measure CCR5 RO lack sensitivity, resulting in high background and overcalculation. Here, we report on two independent, flow cytometric methods of calculating CCR5 RO using the anti-CCR5 antibody, Leronlimab. We show that both methods led to comparable CCR5 RO values, with low background on untreated CCR5+CD4+ T cells and sensitive measurements of occupancy on both blood and tissue-resident CD4+ T cells that correlated longitudinally with plasma concentrations in Leronlimab-treated macaques. Using these assays, we found that Leronlimab stabilized cell surface CCR5, leading to an increase in the levels of circulating and tissue-resident CCR5+CD4+ T cells in vivo in Leronlimab-treated macaques. Weekly Leronlimab treatment in a chronically SIV-infected macaque led to increased CCR5+CD4+ T cells levels and fully suppressed plasma viremia, both concomitant with full CCR5 RO on peripheral blood CD4+ T cells, demonstrating that CCR5+CD4+ T cells were protected from viral replication by Leronlimab binding. Finally, we extended these results to Leronlimab-treated humans and found that weekly 700 mg Leronlimab led to complete CCR5 RO on peripheral blood CD4+ T cells and a statistically significant increase in CCR5+CD4+ T cells in peripheral blood. Collectively, these results establish two RO calculation methods for longitudinal monitoring of anti-CCR5 therapeutic antibody blockade efficacy in both macaques and humans, demonstrate that CCR5+CD4+ T cell levels temporarily increase with Leronlimab treatment, and facilitate future detailed investigations into the immunological impacts of CCR5 inhibition in multiple pathophysiological processes.
- Published
- 2021
- Full Text
- View/download PDF
10. In vitro and in vivo characterization of a recombinant rhesus cytomegalovirus containing a complete genome.
- Author
-
Husam Taher, Eisa Mahyari, Craig Kreklywich, Luke S Uebelhoer, Matthew R McArdle, Matilda J Moström, Amruta Bhusari, Michael Nekorchuk, Xiaofei E, Travis Whitmer, Elizabeth A Scheef, Lesli M Sprehe, Dawn L Roberts, Colette M Hughes, Kerianne A Jackson, Andrea N Selseth, Abigail B Ventura, Hillary C Cleveland-Rubeor, Yujuan Yue, Kimberli A Schmidt, Jason Shao, Paul T Edlefsen, Jeremy Smedley, Timothy F Kowalik, Richard J Stanton, Michael K Axthelm, Jacob D Estes, Scott G Hansen, Amitinder Kaur, Peter A Barry, Benjamin N Bimber, Louis J Picker, Daniel N Streblow, Klaus Früh, and Daniel Malouli
- Subjects
Immunologic diseases. Allergy ,RC581-607 ,Biology (General) ,QH301-705.5 - Abstract
Cytomegaloviruses (CMVs) are highly adapted to their host species resulting in strict species specificity. Hence, in vivo examination of all aspects of CMV biology employs animal models using host-specific CMVs. Infection of rhesus macaques (RM) with rhesus CMV (RhCMV) has been established as a representative model for infection of humans with HCMV due to the close evolutionary relationships of both host and virus. However, the only available RhCMV clone that permits genetic modifications is based on the 68-1 strain which has been passaged in fibroblasts for decades resulting in multiple genomic changes due to tissue culture adaptations. As a result, 68-1 displays reduced viremia in RhCMV-naïve animals and limited shedding compared to non-clonal, low passage isolates. To overcome this limitation, we used sequence information from primary RhCMV isolates to construct a full-length (FL) RhCMV by repairing all mutations affecting open reading frames (ORFs) in the 68-1 bacterial artificial chromosome (BAC). Inoculation of adult, immunocompetent, RhCMV-naïve RM with the reconstituted virus resulted in significant viremia in the blood similar to primary isolates of RhCMV and furthermore led to high viral genome copy numbers in many tissues at day 14 post infection. In contrast, viral dissemination was greatly reduced upon deletion of genes also lacking in 68-1. Transcriptome analysis of infected tissues further revealed that chemokine-like genes deleted in 68-1 are among the most highly expressed viral transcripts both in vitro and in vivo consistent with an important immunomodulatory function of the respective proteins. We conclude that FL-RhCMV displays in vitro and in vivo characteristics of a wildtype virus while being amenable to genetic modifications through BAC recombineering techniques.
- Published
- 2020
- Full Text
- View/download PDF
11. The human IL-15 superagonist N-803 promotes migration of virus-specific CD8+ T and NK cells to B cell follicles but does not reverse latency in ART-suppressed, SHIV-infected macaques.
- Author
-
Gabriela M Webb, Jhomary Molden, Kathleen Busman-Sahay, Shaheed Abdulhaqq, Helen L Wu, Whitney C Weber, Katherine B Bateman, Jason S Reed, Mina Northrup, Nicholas Maier, Shiho Tanaka, Lina Gao, Brianna Davey, Benjamin L Carpenter, Michael K Axthelm, Jeffrey J Stanton, Jeremy Smedley, Justin M Greene, Jeffrey T Safrit, Jacob D Estes, Pamela J Skinner, and Jonah B Sacha
- Subjects
Immunologic diseases. Allergy ,RC581-607 ,Biology (General) ,QH301-705.5 - Abstract
Despite the success of antiretroviral therapy (ART) to halt viral replication and slow disease progression, this treatment is not curative and there remains an urgent need to develop approaches to clear the latent HIV reservoir. The human IL-15 superagonist N-803 (formerly ALT-803) is a promising anti-cancer biologic with potent immunostimulatory properties that has been extended into the field of HIV as a potential "shock and kill" therapeutic for HIV cure. However, the ability of N-803 to reactivate latent virus and modulate anti-viral immunity in vivo under the cover of ART remains undefined. Here, we show that in ART-suppressed, simian-human immunodeficiency virus (SHIV)SF162P3-infected rhesus macaques, subcutaneous administration of N-803 activates and mobilizes both NK cells and SHIV-specific CD8+ T cells from the peripheral blood to lymph node B cell follicles, a sanctuary site for latent virus that normally excludes such effector cells. We observed minimal activation of memory CD4+ T cells and no increase in viral RNA content in lymph node resident CD4+ T cells post N-803 administration. Accordingly, we found no difference in the number or magnitude of plasma viremia timepoints between treated and untreated animals during the N-803 administration period, and no difference in the size of the viral DNA cell-associated reservoir post N-803 treatment. These results substantiate N-803 as a potent immunotherapeutic candidate capable of activating and directing effector CD8+ T and NK cells to the B cell follicle during full ART suppression, and suggest N-803 must be paired with a bona fide latency reversing agent in vivo to facilitate immune-mediated modulation of the latent viral reservoir.
- Published
- 2020
- Full Text
- View/download PDF
12. Oral Immunization with HIV-1 Envelope SOSIP trimers elicits systemic immune responses and cross-reactive anti-V1V2 antibodies in non-human primates.
- Author
-
Bridget S Fisher, Nicholas Dambrauskas, Olesya Trakhimets, Daniela V Andrade, Jeremy Smedley, Donald L Sodora, and D Noah Sather
- Subjects
Medicine ,Science - Abstract
Development of a successful HIV vaccine is dependent upon a determination of the optimum antigen and adjuvant as well as choosing an optimal site for vaccine delivery. The site of delivery is particularly relevant as HIV transmission generally requires that the virus crosses a mucosal membrane to infect a new host. Here we undertake a pilot study comparing three vaccine delivery routes, two to the oral cavity (intraepithelial (iEp) and needle-free (NF-Injex)) as well as intramuscular (IM) delivery. These vaccinations utilized a recombinant HIV-1 Env trimer 10042.05 from an elite neutralizer, subject VC10042, that has previously induced high titers of cross-clade reactive V1V2 antibodies. The 10042.05.SOSIP fused trimer was administered with adjuvants R848 (Resiquimod), MPLA and Alhydrogel to characterize the innate cellular and anti-HIV Envelope (Env) antibody responses following the administration of the vaccine to the oral mucosa. Oral delivery of the 10042.05.SOSIP induced high titers of anti-V1V2 antibodies, which together with previous studies, indicates an immunogenic bias toward the V1V2 regions in 10042-derived Envs. Both types of oral vaccine delivery resulted in immunologic and serologic responses that were comparable to the IM delivery route. Furthermore, induction of anti-V1-V2 specific antibodies was best following iEp delivery of the oral vaccine identifying this as the optimal method to orally deliver this vaccine formulation.
- Published
- 2020
- Full Text
- View/download PDF
13. Early cellular innate immune responses drive Zika viral persistence and tissue tropism in pigtail macaques
- Author
-
Megan A. O’Connor, Jennifer Tisoncik-Go, Thomas B. Lewis, Charlene J. Miller, Debra Bratt, Cassie R. Moats, Paul T. Edlefsen, Jeremy Smedley, Nichole R. Klatt, Michael Gale, and Deborah Heydenburg Fuller
- Subjects
Science - Abstract
The immune response to Zika virus is required to curtail the infection and avoid immunopathology, but may be involved in the associated pathophysiology. Here the authors show that viral persistence and tissue tropism is shaped by an early innate immune response in a pigtail macaque model of infection.
- Published
- 2018
- Full Text
- View/download PDF
14. A Gut Reaction to SIV and SHIV Infection: Lower Dysregulation of Mucosal T Cells during Acute Infection Is Associated with Greater Viral Suppression during cART
- Author
-
Megan A. O’Connor, Paul V. Munson, Sandra E. Dross, Hillary C. Tunggal, Thomas B. Lewis, Jessica Osborn, Christopher W. Peterson, Meei-Li W. Huang, Cassandra Moats, Jeremy Smedley, Keith R. Jerome, Hans-Peter Kiem, Kenneth C. Bagley, James I. Mullins, and Deborah Heydenburg Fuller
- Subjects
non-human primate (NHP) ,SHIV ,SIV ,cART ,colon ,T helper 17 (Th17) ,Microbiology ,QR1-502 - Abstract
Selection of a pre-clinical non-human primate (NHP) model is essential when evaluating therapeutic vaccine and treatment strategies for HIV. SIV and SHIV-infected NHPs exhibit a range of viral burdens, pathologies, and responses to combinatorial antiretroviral therapy (cART) regimens and the choice of the NHP model for AIDS could influence outcomes in studies investigating interventions. Previously, in rhesus macaques (RMs) we showed that maintenance of mucosal Th17/Treg homeostasis during SIV infection correlated with a better virological response to cART. Here, in RMs we compared viral kinetics and dysregulation of gut homeostasis, defined by T cell subset disruption, during highly pathogenic SIVΔB670 compared to SHIV-1157ipd3N4 infection. SHIV infection resulted in lower acute viremia and less disruption to gut CD4 T-cell homeostasis. Additionally, 24/24 SHIV-infected versus 10/19 SIV-infected animals had sustained viral suppression
- Published
- 2021
- Full Text
- View/download PDF
15. Donor T cell chimerism correlates with viral reservoir clearance following allogeneic stem cell transplantation in fully cART-suppressed Mauritian cynomolgus macaques
- Author
-
Helen L. Wu, Whitney Weber, Shaheed A. Abdulhaqq, Christine Shriver-Munsch, Tonya Swanson, Mina Northrup, Kimberly Armantrout, Heidi Price, Mitchell Robertson-LeVay, Jason S. Reed, Katherine B. Bateman, Benjamin N. Bimber, Stephanie L. Junell, Rhonda MacAllister, Alfred W. Legasse, Michael K. Axthelm, Cassandra Moats, Jeremy Smedley, Theodore R. Hobbs, Lauren D. Martin, Gabrielle Meyers, Richard T. Maziarz, Benjamin J. Burwitz, Jeffrey J. Stanton, and Jonah B. Sacha
- Subjects
Microbiology ,QR1-502 ,Public aspects of medicine ,RA1-1270 - Published
- 2019
- Full Text
- View/download PDF
16. Gammaherpesvirus infection and malignant disease in rhesus macaques experimentally infected with SIV or SHIV.
- Author
-
Vickie A Marshall, Nazzarena Labo, Xing-Pei Hao, Benjamin Holdridge, Marshall Thompson, Wendell Miley, Catherine Brands, Vicky Coalter, Rebecca Kiser, Miriam Anver, Yelena Golubeva, Andrew Warner, Elaine S Jaffe, Michael Piatak, Scott W Wong, Claes Ohlen, Rhonda MacAllister, Jeremy Smedley, Claire Deleage, Gregory Q Del Prete, Jeffrey D Lifson, Jacob D Estes, and Denise Whitby
- Subjects
Immunologic diseases. Allergy ,RC581-607 ,Biology (General) ,QH301-705.5 - Abstract
Human gammaherpesviruses are associated with malignancies in HIV infected individuals; in macaques used in non-human primate models of HIV infection, gammaherpesvirus infections also occur. Limited data on prevalence and tumorigenicity of macaque gammaherpesviruses, mostly cross-sectional analyses of small series, are available. We comprehensively examine all three-rhesus macaque gammaherpesviruses -Rhesus rhadinovirus (RRV), Rhesus Lymphocryptovirus (RLCV) and Retroperitoneal Fibromatosis Herpesvirus (RFHV) in macaques experimentally infected with Simian Immunodeficiency Virus or Simian Human Immunodeficiency Virus (SIV/SHIV) in studies spanning 15 years at the AIDS and Cancer Virus Program of the Frederick National Laboratory for Cancer Research. We evaluated 18 animals with malignancies (16 lymphomas, one fibrosarcoma and one carcinoma) and 32 controls. We developed real time quantitative PCR assays for each gammaherpesvirus DNA viral load (VL) in malignant and non-tumor tissues; we also characterized the tumors using immunohistochemistry and in situ hybridization. Furthermore, we retrospectively quantified gammaherpesvirus DNA VL and SIV/SHIV RNA VL in longitudinally-collected PBMCs and plasma, respectively. One or more gammaherpesviruses were detected in 17 tumors; generally, one was predominant, and the relevant DNA VL in the tumor was very high compared to surrounding tissues. RLCV was predominant in tumors resembling diffuse large B cell lymphomas; in a Burkitt-like lymphoma, RRV was predominant; and in the fibrosarcoma, RFHV was predominant. Median RRV and RLCV PBMC DNA VL were significantly higher in cases than controls; SIV/SHIV VL and RLCV VL were independently associated with cancer. Local regressions showed that longitudinal VL patterns in cases and controls, from SIV infection to necropsy, differed for each gammaherpesvirus: while RFHV VL increased only slightly in all animals, RLCV and RRV VL increased significantly and continued to increase steeply in cases; in controls, VL flattened. In conclusion, the data suggest that gammaherpesviruses may play a significant role in tumorogenesis in macaques infected with immunodeficiency viruses.
- Published
- 2018
- Full Text
- View/download PDF
17. Correction: Laparoscopic Technique for Serial Collection of Para-Colonic, Left Colic, and Inferior Mesenteric Lymph Nodes in Macaques.
- Author
-
Jeremy Smedley, Rhonda Macalister, Solomon Wangari, Mercy Gathuka, Joel Ahrens, Naoto Iwayama, Drew May, Debbie Bratt, Megan O'Connor, Paul Munson, Michael Koday, Lifson, and Deborah Heydenburg Fuller
- Subjects
Medicine ,Science - Abstract
[This corrects the article DOI: 10.1371/journal.pone.0157535.].
- Published
- 2018
- Full Text
- View/download PDF
18. Sustained AAV9-mediated expression of a non-self protein in the CNS of non-human primates after immunomodulation.
- Author
-
Arlene I Ramsingh, Steven J Gray, Andrew Reilly, Michael Koday, Debbie Bratt, Merika Treants Koday, Paul Munson, Robert Murnane, Jeremy Smedley, Yuhui Hu, Anne Messer, and Deborah Heydenburg Fuller
- Subjects
Medicine ,Science - Abstract
A critical issue in transgene delivery studies is immune reactivity to the transgene- encoded protein and its impact on sustained gene expression. Here, we test the hypothesis that immunomodulation by rapamycin can decrease immune reactivity after intrathecal AAV9 delivery of a transgene (GFP) in non-human primates, resulting in sustained GFP expression in the CNS. We show that rapamycin treatment clearly reduced the overall immunogenicity of the AAV9/GFP vector by lowering GFP- and AAV9-specific antibody responses, and decreasing T cell responses including cytokine and cytolytic effector responses. Spinal cord GFP protein expression was sustained for twelve weeks, with no toxicity. Immune correlates of robust transgene expression include negligible GFP-specific CD4 and CD8 T cell responses, absence of GFP-specific IFN-γ producing T cells, and absence of GFP-specific cytotoxic T cells, which support the hypothesis that decreased T cell reactivity results in sustained transgene expression. These data strongly support the use of modest doses of rapamycin to modulate immune responses for intrathecal gene therapies, and potentially a much wider range of viral vector-based therapeutics.
- Published
- 2018
- Full Text
- View/download PDF
19. Correction: Sustained AAV9-mediated expression of a non-self protein in the CNS of non-human primates after immunomodulation.
- Author
-
Arlene I Ramsingh, Steven J Gray, Andrew Reilly, Michael Koday, Debbie Bratt, Merika Treants Koday, Paul Munson, Robert Murnane, Jeremy Smedley, Yuhui Hu, Anne Messer, and Deborah Heydenburg Fuller
- Subjects
Medicine ,Science - Abstract
[This corrects the article DOI: 10.1371/journal.pone.0198154.].
- Published
- 2018
- Full Text
- View/download PDF
20. Laparoscopic Technique for Serial Collection of Para-Colonic, Left Colic, and Inferior Mesenteric Lymph Nodes in Macaques.
- Author
-
Jeremy Smedley, Rhonda Macalister, Solomon Wangari, Mercy Gathuka, Joel Ahrens, Naoto Iwayama, Drew May, Debbie Bratt, Megan O'Connor, Paul Munson, Michael Koday, Jeff Lifson, and Deborah Heydenburg Fuller
- Subjects
Medicine ,Science - Abstract
Unlike peripheral lymph nodes (PLN), the mesenteric lymph nodes (MLN) draining the gastrointestinal (GI) tract are exposed to microbes and microbial products from the intestines and as such, are immunologically distinct. GI draining (MLN) have also been shown to be sites of early viral replication and likely impact early events that determine the course of HIV infection. They also are important reservoir sites that harbor latently-infected cells and from which the virus can emerge even after prolonged combination antiretroviral therapy (cART). Changes in the microbial flora and increased permeability of the GI epithelium associated with lentiviral infection can impact the gut associated lymphoid tissue (GALT) and induce changes to secondary lymphoid organs limiting immune reconstitution with cART. Nonhuman primate models for AIDS closely model HIV infection in humans and serial sampling of the GALT and associated secondary lymphoid organs in this model is crucial to gain a better understanding of the critical early events in infection, pathogenesis, and the role of immune responses or drugs in controlling virus at these sites. However, current techniques to sample GI draining (MLN) involve major surgery and/or necropsy, which have, to date, limited the ability to investigate mechanisms mediating the initiation, persistence and control of infection in this compartment. Here, we describe a minimally invasive laparoscopic technique for serial sampling of these sites that can be used with increased sampling frequency, yields greater cell numbers and immune cell subsets than current non-invasive techniques of the GALT and reduces the potential for surgical complications that could complicate interpretation of the results. This procedure has potential to facilitate studies of pathogenesis and evaluation of preventive and treatment interventions, reducing sampling variables that can influence experimental results, and improving animal welfare.
- Published
- 2016
- Full Text
- View/download PDF
21. Tracking the luminal exposure and lymphatic drainage pathways of intravaginal and intrarectal inocula used in nonhuman primate models of HIV transmission.
- Author
-
Jeremy Smedley, Baris Turkbey, Marcelino L Bernardo, Gregory Q Del Prete, Jacob D Estes, Gary L Griffiths, Hisataka Kobayashi, Peter L Choyke, Jeffrey D Lifson, and Brandon F Keele
- Subjects
Medicine ,Science - Abstract
Over 80% of sexual HIV-1 transmissions originate from a single viral variant, but the underlying basis for this transmission bottleneck remains to be elucidated. Nonhuman primate models of mucosal virus transmission allow opportunities to gain insight into the basis of this mucosal bottleneck. We used simulated inocula consisting of either non-infectious vital dye or contrast dye with non-invasive magnetic resonance imaging (MRI) to visualize mucosal exposure and passive lymphatic drainage patterns following vaginal and rectal exposures in Indian origin rhesus macaques. Results revealed a limited overall distance of dye coverage from the anal verge following 1 ml (n = 8) intrarectally administered, which greatly increased with a 3 ml (n = 8) volume. Intravaginal dye exposure using 2 ml revealed complete coverage of the mucosa of the vagina and ectocervix, however dye was not detectable in the endocervix, uterus, fallopian tubes or ovaries in nuliparous sexually mature rhesus macaques (n = 9). In addition, following submucosal and intranodal injections of vital dye or MRI contrast dye in the rectum (n = 9), or distal and proximal vagina (n = 4), the lymphatic drainage pathways were identified as first the internal then common iliac chain followed by para-aortic lymph nodes. Drainage from the distal descending colon (n = 8) was via the para-colonic lymph nodes followed by the inferior mesenteric and para-aortic lymph nodes. Analysis after vaginal challenge with infectious SIVmac239 followed by euthanasia at day 3 revealed a pattern of viral dissemination consistent with the imaging results. These results provide insights into potential patterns of viral dissemination that can help guide efforts to better elucidate the earliest events of virus transmission and potential intervention strategies.
- Published
- 2014
- Full Text
- View/download PDF
22. Persistence of viral reservoirs in multiple tissues after antiretroviral therapy suppression in a macaque RT-SHIV model.
- Author
-
Christopher Kline, Jean Ndjomou, Tamera Franks, Rebecca Kiser, Vicky Coalter, Jeremy Smedley, Michael Piatak, John W Mellors, Jeffrey D Lifson, and Zandrea Ambrose
- Subjects
Medicine ,Science - Abstract
Although antiretroviral therapy (ART) can suppress HIV-1 replication sufficiently to eliminate measurable plasma viremia, infected cells remain and ensure viral recrudescence after discontinuation of ART. We used a macaque model of HIV-1/AIDS to evaluate the location of infected cells during ART. Twelve macaques were infected with RT-SHIVmne, a SIV containing HIV-1 reverse transcriptase, conferring sensitivity to non-nucleoside reverse transcriptase inhibitors (NNRTIs). Ten to fourteen weeks post-infection, 6 animals were treated with 3 or 4 antiretroviral drugs for 17-20 weeks; 6 control animals remained untreated. Viral DNA (vDNA) and RNA (vRNA) were measured in peripheral blood mononuclear cells (PBMC) and at necropsy in multiple tissues by quantitative PCR and RT-PCR. The majority of virally infected cells were located in lymphoid tissues with variable levels in the gastrointestinal tract of both treated and untreated animals. Tissue viral DNA levels correlated with week 1 plasma viremia, suggesting that tissues that harbor proviral DNA are established within the first week of infection. PBMC vDNA levels did not correlate with plasma viremia or tissue levels of vDNA. vRNA levels were high in lymphoid and gastrointestinal tissues of the untreated animals; animals on ART had little vRNA expressed in tissues and virus could not be cultured from lymph node resting CD4+ cells after 17-20 weeks on ART, indicating little or no ongoing viral replication. Strategies for eradication of HIV-1 will need to target residual virus in ART suppressed individuals, which may not be accurately reflected by frequencies of infected cells in blood.
- Published
- 2013
- Full Text
- View/download PDF
23. Damaged intestinal epithelial integrity linked to microbial translocation in pathogenic simian immunodeficiency virus infections.
- Author
-
Jacob D Estes, Levelle D Harris, Nichole R Klatt, Brian Tabb, Stefania Pittaluga, Mirko Paiardini, G Robin Barclay, Jeremy Smedley, Rhonda Pung, Kenneth M Oliveira, Vanessa M Hirsch, Guido Silvestri, Daniel C Douek, Christopher J Miller, Ashley T Haase, Jeffrey Lifson, and Jason M Brenchley
- Subjects
Immunologic diseases. Allergy ,RC581-607 ,Biology (General) ,QH301-705.5 - Abstract
The chronic phase of HIV infection is marked by pathological activation of the immune system, the extent of which better predicts disease progression than either plasma viral load or CD4(+) T cell count. Recently, translocation of microbial products from the gastrointestinal tract has been proposed as an underlying cause of this immune activation, based on indirect evidence including the detection of microbial products and specific immune responses in the plasma of chronically HIV-infected humans or SIV-infected Asian macaques. We analyzed tissues from SIV-infected rhesus macaques (RMs) to provide direct in situ evidence for translocation of microbial constituents from the lumen of the intestine into the lamina propria and to draining and peripheral lymph nodes and liver, accompanied by local immune responses in affected tissues. In chronically SIV-infected RMs this translocation is associated with breakdown of the integrity of the epithelial barrier of the gastrointestinal (GI) tract and apparent inability of lamina propria macrophages to effectively phagocytose translocated microbial constituents. By contrast, in the chronic phase of SIV infection in sooty mangabeys, we found no evidence of epithelial barrier breakdown, no increased microbial translocation and no pathological immune activation. Because immune activation is characteristic of the chronic phase of progressive HIV/SIV infections, these findings suggest that increased microbial translocation from the GI tract, in excess of capacity to clear the translocated microbial constituents, helps drive pathological immune activation. Novel therapeutic approaches to inhibit microbial translocation and/or attenuate chronic immune activation in HIV-infected individuals may complement treatments aimed at direct suppression of viral replication.
- Published
- 2010
- Full Text
- View/download PDF
24. Therapeutic neutralizing monoclonal antibody administration protects against lethal yellow fever virus infection
- Author
-
Michael J. Ricciardi, Lauren N. Rust, Nuria Pedreño-Lopez, Sofiya Yusova, Sreya Biswas, Gabriela M. Webb, Lucas Gonzalez-Nieto, Thomas B. Voigt, Johan J. Louw, Fernanda D. Laurino, John R. DiBello, Hans-Peter Raué, Aaron M. Barber-Axthelm, Kimberly Chun, Samantha Uttke, Lidiane M. S. Raphael, Aaron Yrizarry-Medina, Brandon C. Rosen, Rebecca Agnor, Lina Gao, Caralyn Labriola, Michael Axthelm, Jeremy Smedley, Justin G. Julander, Myrna C. Bonaldo, Laura M. Walker, Ilhem Messaoudi, Mark K. Slifka, Dennis R. Burton, Esper G. Kallas, Jonah B. Sacha, David I. Watkins, and Benjamin J. Burwitz
- Subjects
General Medicine - Abstract
Yellow fever virus (YFV) is a reemerging global health threat, driven by several factors, including increased spread of the mosquito vector and rapid urbanization. Although a prophylactic vaccine exists, vaccine hesitancy, supply deficits, and distribution difficulties leave specific populations at risk of severe YFV disease, as evidenced by recent outbreaks in South America. To establish a treatment for patients with severe YFV infection, we tested 37 YFV-specific monoclonal antibodies isolated from vaccinated humans and identified two capable of potently neutralizing multiple pathogenic primary YFV isolates. Using both hamster and nonhuman primate models of lethal YFV infection, we demonstrate that a single administration of either of these two potently neutralizing antibodies during acute infection fully controlled viremia and prevented severe disease and death in treated animals. Given the potential severity of YFV-induced disease, our results show that these antibodies could be effective in saving lives and fill a much-needed void in managing YFV cases during outbreaks.
- Published
- 2023
- Full Text
- View/download PDF
25. Antibody-based CCR5 blockade protects Macaques from mucosal SHIV transmission
- Author
-
Bruce K. Patterson, Whitney C. Weber, Gabriela M. Webb, Katherine B. Bateman, Cleiton Pessoa, Shaheed A. Abdulhaqq, Lina Gao, Benjamin N. Bimber, Jonah B. Sacha, Scott G. Hansen, Xiao L. Chang, Justin M. Greene, Kush Dhody, Scott Kelly, Nancy L. Haigwood, Rebecca Agnor, Cassandra Moats, Glen M. Chew, Roxanne M. Gilbride, Jeremy Smedley, Nicole D Burnett, Oriene Shiel, Nicholas Maier, Don Siess, Lishomwa C. Ndhlovu, Miranda Fischer, Timothy R. Brown, Diogo M. Magnani, Jason S. Reed, Travis Giobbi, Jeffrey Torgerson, Helen L. Wu, and Nader Pourhassan
- Subjects
CD4-Positive T-Lymphocytes ,Male ,0301 basic medicine ,Adoptive cell transfer ,Receptors, CCR5 ,Chemokine receptor CCR5 ,viruses ,Science ,Simian Acquired Immunodeficiency Syndrome ,General Physics and Astronomy ,HIV Infections ,CD8-Positive T-Lymphocytes ,HIV Antibodies ,Antibodies, Monoclonal, Humanized ,Article ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,0302 clinical medicine ,Animals ,Humans ,Medicine ,030212 general & internal medicine ,Hiv acquisition ,Mucous Membrane ,Multidisciplinary ,biology ,business.industry ,Transmission (medicine) ,virus diseases ,General Chemistry ,Viral Load ,Macaca mulatta ,Antiretroviral therapy ,Virology ,Blockade ,Clinical trial ,030104 developmental biology ,biology.protein ,Female ,Pre-Exposure Prophylaxis ,Simian Immunodeficiency Virus ,Antibody therapy ,Antibody ,business - Abstract
In the absence of a prophylactic vaccine, the use of antiretroviral therapy (ART) as pre-exposure prophylaxis (PrEP) to prevent HIV acquisition by uninfected individuals is a promising approach to slowing the epidemic, but its efficacy is hampered by incomplete patient adherence and ART-resistant variants. Here, we report that competitive inhibition of HIV Env-CCR5 binding via the CCR5-specific antibody Leronlimab protects rhesus macaques against infection following repeated intrarectal challenges of CCR5-tropic SHIVSF162P3. Injection of Leronlimab weekly at 10 mg/kg provides significant but partial protection, while biweekly 50 mg/kg provides complete protection from SHIV acquisition. Tissue biopsies from protected macaques post challenge show complete CCR5 receptor occupancy and an absence of viral nucleic acids. After Leronlimab washout, protected macaques remain aviremic, and adoptive transfer of hematologic cells into naïve macaques does not transmit viral infection. These data identify CCR5 blockade with Leronlimab as a promising approach to HIV prophylaxis and support initiation of clinical trials., CCR5 is a co-receptor for many transmitted HIV strains. Here, the authors show that biweekly injection of the CCR5-specific antibody Leronlimab protects rhesus macaques against infection following repeated intrarectal challenges of a CCR5-tropic SHIV.
- Published
- 2021
26. Myeloid cell tropism enables MHC-E-restricted CD8(+) T cell priming and vaccine efficacy by the RhCMV/SIV vaccine
- Author
-
Scott G. Hansen, Meaghan H. Hancock, Daniel Malouli, Emily E. Marshall, Colette M. Hughes, Kurt T. Randall, David Morrow, Julia C. Ford, Roxanne M. Gilbride, Andrea N. Selseth, Renee Espinosa Trethewy, Lindsey M. Bishop, Kelli Oswald, Rebecca Shoemaker, Brian Berkemeier, William J. Bosche, Michael Hull, Lorna Silipino, Michael Nekorchuk, Kathleen Busman-Sahay, Jacob D. Estes, Michael K. Axthelm, Jeremy Smedley, Danica Shao, Paul T. Edlefsen, Jeffrey D. Lifson, Klaus Früh, Jay A. Nelson, and Louis J. Picker
- Subjects
Immunology ,SAIDS Vaccines ,Simian Acquired Immunodeficiency Syndrome ,Cytomegalovirus ,Vaccine Efficacy ,General Medicine ,CD8-Positive T-Lymphocytes ,Macaca mulatta ,Tropism ,Article ,Major Histocompatibility Complex ,Cytomegalovirus Vaccines ,Epitopes ,MicroRNAs ,Animals ,Myeloid Cells ,Simian Immunodeficiency Virus - Abstract
The strain 68-1 rhesus cytomegalovirus (RhCMV)–based vaccine for simian immunodeficiency virus (SIV) can stringently protect rhesus macaques (RMs) from SIV challenge by arresting viral replication early in primary infection. This vaccine elicits unconventional SIV-specific CD8 + T cells that recognize epitopes presented by major histocompatibility complex (MHC)–II and MHC-E instead of MHC-Ia. Although RhCMV/SIV vaccines based on strains that only elicit MHC-II– and/or MHC-Ia–restricted CD8 + T cells do not protect against SIV, it remains unclear whether MHC-E–restricted T cells are directly responsible for protection and whether these responses can be separated from the MHC-II–restricted component. Using host microRNA (miR)–mediated vector tropism restriction, we show that the priming of MHC-II and MHC-E epitope–targeted responses depended on vector infection of different nonoverlapping cell types in RMs. Selective inhibition of RhCMV infection in myeloid cells with miR-142–mediated tropism restriction eliminated MHC-E epitope–targeted CD8 + T cell priming, yielding an exclusively MHC-II epitope–targeted response. Inhibition with the endothelial cell–selective miR-126 eliminated MHC-II epitope–targeted CD8 + T cell priming, yielding an exclusively MHC-E epitope–targeted response. Dual miR-142 + miR-126–mediated tropism restriction reverted CD8 + T cell responses back to conventional MHC-Ia epitope targeting. Although the magnitude and differentiation state of these CD8 + T cell responses were generally similar, only the vectors programmed to elicit MHC-E–restricted CD8 + T cell responses provided protection against SIV challenge, directly demonstrating the essential role of these responses in RhCMV/SIV vaccine efficacy.
- Published
- 2022
27. Therapeutic neutralizing monoclonal antibody administration protects against lethal Yellow Fever infection
- Author
-
Michael J. Ricciardi, Lauren N. Rust, Nuria Pedreño-Lopez, Sofiya Yusova, Sreya Biswas, Gabriela M. Webb, Lucas Gonsales-Nieto, Thomas B. Voigt, Johan J. Louw, Fernanda D. Laurino, John R. DiBello, Hans-Peter Raué, Aaron M. Barber-Axthelm, Samantha Uttke, Lidiane M.S. Raphael, Aaron Yrizarry-Medina, Brandon C. Rosen, Rebecca Agnor, Lina Gao, Caralyn Labriola, Michael Axthelm, Jeremy Smedley, Justin G. Julander, Myrna C. Bonaldo, Laura M. Walker, Ilhem Messaoudi, Mark K. Slifka, Dennis R. Burton, Esper G. Kallas, Jonah B. Sacha, David I. Watkins, and Benjamin J. Burwitz
- Abstract
Few countermeasures to treat Yellow Fever virus (YFV) infection are under development, because vaccines have helped to limit new infections. Unfortunately, vaccine hesitancy, supply deficits, and a paucity of therapeutic options have left individuals at risk. Here, we tested potent YFV-specific neutralizing monoclonal antibodies in rodents and non-human primates. We administered antibodies during acute pathogenic YFV infection and demonstrate that we can prevent severe disease and death. Given the severity of YFV-induced disease, our results show that these antibodies could be effective in saving lives and fill a much-needed void in managing Yellow Fever cases during outbreaks around the world.One Sentence SummaryTherapeutic monoclonal antibodies prevent death from YFV infection.
- Published
- 2022
- Full Text
- View/download PDF
28. Non-bronchoscopic Bronchoalveolar Lavage as a Refinement for Safely Obtaining High-quality Samples from Macaques
- Author
-
Hugh B. Crank, Kimberly Armantrout, Lauren D. Martin, Cassandra Moats, Kurt T. Randall, Scott G. Hansen, Theodore R. Hobbs, Tonya Swanson, Nicole D Burnett, Roxanne M. Gilbride, Aaron M. Barber-Axthelm, and Jeremy Smedley
- Subjects
General Veterinary ,medicine.diagnostic_test ,040301 veterinary sciences ,business.industry ,medicine.medical_treatment ,04 agricultural and veterinary sciences ,respiratory system ,Bronchoalveolar Lavage ,General Biochemistry, Genetics and Molecular Biology ,respiratory tract diseases ,0403 veterinary science ,Bronchoalveolar lavage ,Anesthesia ,Heart rate ,medicine ,Animals ,Macaca ,business ,Bronchoalveolar Lavage Fluid ,Lung ,Fluid volume ,Saline ,Original Research ,Oxygen saturation (medicine) ,Procedure time - Abstract
Nonbronchoscopic bronchoalveolar lavage (NB-BAL) is a minimally invasive diagnostic and research tool used to sample the cells of lower airways and alveoli without using a bronchoscope. Our study compared NB-BAL and bronchoscopic bronchoalveolar lavage (B-BAL) in terms of costs, cell yields, and the number of post-procedural complications in macaques. We also analyzed procedure times, BAL fluid volume yields, and vital signs in a subset of animals that underwent NB-BAL. Compared with the B-BAL technique, NB-BAL was less expensive to perform, with fewer complications, fewer animals requiring temporary or permanent cessation of BALs, and higher cell yields per mL of recovered saline. The average procedure time for NB-BAL was 6.8 ± 1.6 min, and the average NB-BAL lavage volume yield was 76 ± 9%. We found no significant differences in respiration rate before, during, or after NB-BAL but did find significant differences in heart rate and oxygen saturation (SpO2). This study demonstrates that NB-BAL is a simple, cost-effective, and safe alternative to B-BAL that results in higher cell yields per mL, improved animal welfare, and fewer missed time points, and thus constitutes a refinement over the B-BAL in macaques.
- Published
- 2020
- Full Text
- View/download PDF
29. Terumo spectra optia leukapheresis of cynomolgus macaques for hematopoietic stem cell and T cell collection
- Author
-
Alfred W. Legasse, Mina Northrup, Michael K. Axthelm, Tonya Swanson, Cassandra Moats, Whitney C. Weber, Jeremy Smedley, Kimberly Armantrout, Lauren D. Martin, Christine Shriver-Munsch, Helen L. Wu, Katherine B. Bateman, Justin M. Greene, Richard T. Maziarz, Benjamin J. Burwitz, Theodore R. Hobbs, Jonah B. Sacha, and Nicholas Maier
- Subjects
Male ,Benzylamines ,T-Lymphocytes ,T cell ,030204 cardiovascular system & hematology ,Cyclams ,Article ,03 medical and health sciences ,0302 clinical medicine ,Nucleated cell ,Granulocyte Colony-Stimulating Factor ,medicine ,Animals ,Clinical treatment ,business.industry ,Hematopoietic stem cell ,Hematology ,General Medicine ,Leukapheresis ,Hematopoietic Stem Cells ,Hematopoietic Stem Cell Mobilization ,Transplantation ,Macaca fascicularis ,Haematopoiesis ,medicine.anatomical_structure ,Creatinine ,Immunology ,Blood Component Removal ,Female ,Stem cell ,business ,030215 immunology - Abstract
Macaques are physiologically relevant animal models of human immunology and infectious disease that have provided key insights and advanced clinical treatment in transplantation, vaccinology, and HIV/AIDS. However, the small size of macaques is a stumbling block for studies requiring large numbers of cells, such as hematopoietic stem cells (HSCs) for transplantation, antigen-specific lymphocytes for in-depth immunological analysis, and latently-infected CD4+ T-cells for HIV cure studies. Here, we provide a detailed protocol for collection of large numbers of HSCs and T-cells from cynomolgus macaques as small as 3 kilograms using the Terumo Spectra Optia apheresis system, yielding an average of 5.0 x 10(9) total nucleated cells from mobilized animals and 1.2 x 10(9) total nucleated cells from non-mobilized animals per procedure. This report provides sufficient detail to adapt this apheresis technique at other institutions, which will facilitate more efficient and detailed analysis of HSCs and their progeny blood cells.
- Published
- 2020
- Full Text
- View/download PDF
30. Antibiotic-induced microbiome perturbations are associated with significant alterations to colonic mucosal immunity in rhesus macaques
- Author
-
Ryan Cheu, Michael Cartwright, Alexander S. Zevin, Jeremy Smedley, Ernesto Coronado, Mike Fang, Toni M. Gott, Hans Benjamin Hampel, Drew May, Andrew T. Gustin, Jacob Modesitt, Brian Richardson, Solomon Wangari, Nichole R. Klatt, Jennifer A. Manuzak, Mark J. Cameron, Brian Agricola, Cheryl M. Cameron, Elise Smith, Michael Gale, Charlene Miller, and Tiffany Hensley-McBain
- Subjects
0301 basic medicine ,Colon ,medicine.drug_class ,Immunology ,Antibiotics ,medicine.disease_cause ,Drug Administration Schedule ,Gas Chromatography-Mass Spectrometry ,Immunophenotyping ,Microbiology ,Feces ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,T-Lymphocyte Subsets ,Immunity ,medicine ,Animals ,Immunology and Allergy ,Tissue Distribution ,Microbiome ,Intestinal Mucosa ,Immunity, Mucosal ,Bacteria ,biology ,Clindamycin ,Pathogenic bacteria ,Biodiversity ,Fatty Acids, Volatile ,biology.organism_classification ,Macaca mulatta ,Enterobacteriaceae ,Anti-Bacterial Agents ,Gastrointestinal Microbiome ,030104 developmental biology ,Neutrophil Infiltration ,Drug Monitoring ,Biomarkers ,030215 immunology ,medicine.drug - Abstract
The diverse bacterial communities that colonize the gastrointestinal tract play an essential role in maintaining immune homeostasis through the production of critical metabolites such as short-chain fatty acids (SCFAs) and this can be disrupted by antibiotic use. However, few studies have addressed the effects of specific antibiotics longitudinally on the microbiome and immunity. We evaluated the effects of four specific antibiotics: enrofloxacin, cephalexin, paromomycin, and clindamycin, in healthy female rhesus macaques. All antibiotics disrupted the microbiome, including reduced abundances of fermentative bacteria and increased abundances of potentially pathogenic bacteria, including Enterobacteriaceae in the stool, and decreased Helicobacteraceae in the colon. This was associated with decreased SCFAs, indicating altered bacterial metabolism. Importantly, antibiotic use also substantially altered local immune responses, including increased neutrophils and Th17 cells in the colon. Furthermore, we observed increased soluble CD14 in plasma, indicating microbial translocation. These data provide a longitudinal evaluation of antibiotic-induced changes to the composition and function of colonic bacterial communities associated with specific alterations in mucosal and systemic immunity.
- Published
- 2020
- Full Text
- View/download PDF
31. Suppression of human and simian immunodeficiency virus replication with the CCR5-specific antibody Leronlimab in two species
- Author
-
Xiao L. Chang, Jason S. Reed, Gabriela M. Webb, Helen L. Wu, Jimmy Le, Katherine B. Bateman, Justin M. Greene, Cleiton Pessoa, Courtney Waytashek, Whitney C. Weber, Joseph Hwang, Miranda Fischer, Cassandra Moats, Oriene Shiel, Rachele M. Bochart, Hugh Crank, Don Siess, Travis Giobbi, Jeffrey Torgerson, Rebecca Agnor, Lina Gao, Kush Dhody, Jacob P. Lalezari, Ivo Sah Bandar, Alnor M. Carnate, Alina S. Pang, Michael J. Corley, Scott Kelly, Nader Pourhassan, Jeremy Smedley, Benjamin N. Bimber, Scott G. Hansen, Lishomwa C. Ndhlovu, and Jonah B. Sacha
- Subjects
Receptors, CCR5 ,Immunology ,virus diseases ,HIV Antibodies ,Antibodies, Monoclonal, Humanized ,Microbiology ,Macaca mulatta ,Virology ,Genetics ,Animals ,Humans ,Parasitology ,Simian Immunodeficiency Virus ,Molecular Biology - Abstract
The CCR5-specific antibody Leronlimab is being investigated as a novel immunotherapy that can suppress HIV replication with minimal side effects. Here we studied the virological and immunological consequences of Leronlimab in chronically CCR5-tropic HIV-1 infected humans (n = 5) on suppressive antiretroviral therapy (ART) and in ART-naïve acutely CCR5-tropic SHIV infected rhesus macaques (n = 4). All five human participants transitioned from daily combination ART to self-administered weekly subcutaneous (SC) injections of 350 mg or 700 mg Leronlimab and to date all participants have sustained virologic suppression for over seven years. In all participants, Leronlimab fully occupied CCR5 receptors on peripheral blood CD4+ T cells and monocytes. In ART-naïve rhesus macaques acutely infected with CCR5-tropic SHIV, weekly SC injections of 50 mg/kg Leronlimab fully suppressed plasma viremia in half of the macaques. CCR5 receptor occupancy by Leronlimab occurred concomitant with rebound of CD4+ CCR5+ T-cells in peripheral blood, and full CCR5 receptor occupancy was found in multiple anatomical compartments. Our results demonstrate that weekly, self-administered Leronlimab was safe, well-tolerated, and efficacious for long-term virologic suppression and should be included in the arsenal of safe, easily administered, longer-acting antiretroviral treatments for people living with HIV-1. Trial Registration: ClinicalTrials.gov Identifiers: NCT02175680 and NCT02355184.
- Published
- 2021
32. CCR5 Receptor Occupancy Analysis Reveals Increased Peripheral Blood CCR5+CD4+ T Cells Following Treatment With the Anti-CCR5 Antibody Leronlimab
- Author
-
Joseph Hwang, Andrew W. Sylwester, Meenakshi Tiwary, Rachele M. Bochart, Gabriela M. Webb, Courtney Waytashek, Colette M. Hughes, Scott Kelly, Xiao L. Chang, Jason S. Reed, Carla Boyle, Scott G. Hansen, Miranda Fischer, Jeremy Smedley, Helen L. Wu, Nader Pourhassan, Cleiton Pessoa, Jonah B. Sacha, Christopher P. Recknor, David Morrow, and Karina Belica
- Subjects
CD4-Positive T-Lymphocytes ,Primates ,Receptors, CCR5 ,Chemokine receptor CCR5 ,T cell ,viruses ,Cell ,Immunology ,Simian Acquired Immunodeficiency Syndrome ,Viremia ,HIV Infections ,Pharmacology ,HIV Antibodies ,Antibodies, Monoclonal, Humanized ,Flow cytometry ,In vivo ,antibody ,medicine ,Immunology and Allergy ,Animals ,Humans ,Original Research ,medicine.diagnostic_test ,biology ,Chemistry ,SARS-CoV-2 ,flow cytometry ,virus diseases ,HIV ,RC581-607 ,medicine.disease ,CD4 ,CD4 Lymphocyte Count ,COVID-19 Drug Treatment ,medicine.anatomical_structure ,Treatment Outcome ,Viral replication ,receptor occupancy (RO) ,biology.protein ,HIV-1 ,Female ,Simian Immunodeficiency Virus ,Antibody ,Immunologic diseases. Allergy ,CCR5 ,Protein Binding - Abstract
CCR5 plays a central role in infectious disease, host defense, and cancer progression, thereby making it an ideal target for therapeutic development. Notably, CCR5 is the major HIV entry co-receptor, where its surface density correlates with HIV plasma viremia. The level of CCR5 receptor occupancy (RO) achieved by a CCR5-targeting therapeutic is therefore a critical predictor of its efficacy. However, current methods to measure CCR5 RO lack sensitivity, resulting in high background and overcalculation. Here, we report on two independent, flow cytometric methods of calculating CCR5 RO using the anti-CCR5 antibody, Leronlimab. We show that both methods led to comparable CCR5 RO values, with low background on untreated CCR5+CD4+ T cells and sensitive measurements of occupancy on both blood and tissue-resident CD4+ T cells that correlated longitudinally with plasma concentrations in Leronlimab-treated macaques. Using these assays, we found that Leronlimab stabilized cell surface CCR5, leading to an increase in the levels of circulating and tissue-resident CCR5+CD4+ T cells in vivo in Leronlimab-treated macaques. Weekly Leronlimab treatment in a chronically SIV-infected macaque led to increased CCR5+CD4+ T cells levels and fully suppressed plasma viremia, both concomitant with full CCR5 RO on peripheral blood CD4+ T cells, demonstrating that CCR5+CD4+ T cells were protected from viral replication by Leronlimab binding. Finally, we extended these results to Leronlimab-treated humans and found that weekly 700 mg Leronlimab led to complete CCR5 RO on peripheral blood CD4+ T cells and a statistically significant increase in CCR5+CD4+ T cells in peripheral blood. Collectively, these results establish two RO calculation methods for longitudinal monitoring of anti-CCR5 therapeutic antibody blockade efficacy in both macaques and humans, demonstrate that CCR5+CD4+ T cell levels temporarily increase with Leronlimab treatment, and facilitate future detailed investigations into the immunological impacts of CCR5 inhibition in multiple pathophysiological processes.
- Published
- 2021
- Full Text
- View/download PDF
33. Protection of Newborn Macaques by Plant-Derived HIV Broadly Neutralizing Antibodies: a Model for Passive Immunotherapy during Breastfeeding
- Author
-
Jeffrey J. Stanton, Nancy L. Haigwood, Jonathan Lees, Lori Urban, Yvonne J. Rosenberg, Tracy Cheever, Miranda Fischer, Lingjun Mao, Markus Sack, Heather M. Sidener, Shilpi Pandey, Jeremy Smedley, Felicity J Coulter, and Xiaoming Jiang
- Subjects
Male ,Immunology ,Simian Acquired Immunodeficiency Syndrome ,Viremia ,HIV Infections ,Microbiology ,Macaque ,Virus ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Virology ,biology.animal ,Tobacco ,Vaccines and Antiviral Agents ,medicine ,Animals ,Neutralizing antibody ,030304 developmental biology ,0303 health sciences ,biology ,Immunization, Passive ,medicine.disease ,Macaca mulatta ,Breast Feeding ,Viral replication ,Animals, Newborn ,030220 oncology & carcinogenesis ,Insect Science ,biology.protein ,HIV-1 ,Leukocytes, Mononuclear ,Female ,Simian Immunodeficiency Virus ,Antibody ,Viral load ,Broadly Neutralizing Antibodies - Abstract
Preventing human immunodeficiency virus (HIV) infection in newborns by vertical transmission remains an important unmet medical need in resource-poor areas where antiretroviral therapy (ART) is not available and mothers and infants cannot be treated prepartum or during the breastfeeding period. In the present study, the protective efficacy of the potent HIV-neutralizing antibodies PGT121 and VRC07-523, both produced in plants, were assessed in a multiple-SHIV (simian-human immunodeficiency virus)-challenge breastfeeding macaque model. Newborn macaques received either six weekly subcutaneous injections with PGT121 alone or as a cocktail of PGT121-LS plus VRC07-523-LS injected three times every 2 weeks. Viral challenge with SHIV(SF162P3) was twice weekly over 5.5 weeks using 11 exposures. Despite the transient presence of plasma viral RNA either immediately after the first challenge or as single-point blips, the antibodies prevented a productive infection in all babies with no sustained plasma viremia, compared to viral loads ranging from 10(3) to 5 × 10(8) virions/ml in four untreated controls. No virus was detected in peripheral blood mononuclear cells (PBMCs), and only 3 of 159 tissue samples were weakly positive in the treated babies. Newborn macaques proved to be immunocompetent, producing transient anti-Env antibodies and anti-drug antibody (ADA), which were maintained in the circulation after passive broadly neutralizing antibody clearance. ADA responses were directed to the IgG1 Fc CH2-CH3 domains, which has not been observed to date in adult monkeys passively treated with PGT121 or VRC01. In addition, high levels of VRC07-523 anti-idiotypic antibodies in the circulation of one newborn was concomitant with the rapid elimination of VRC07. Plant-expressed antibodies show promise as passive immunoprophylaxis in a breastfeeding model in newborns. IMPORTANCE Plant-produced human neutralizing antibody prophylaxis is highly effective in preventing infection in newborn monkeys during repeated oral exposure, modeling virus in breastmilk, and offers advantages in cost of production and safety. These findings raise the possibility that anti-Env antibodies may contribute to the control of viral replication in this newborn model and that the observed immune responsiveness may be driven by the long-lived presence of immune complexes.
- Published
- 2021
34. Mucosal T Helper 17 and T Regulatory Cell Homeostasis Correlate with Acute Simian Immunodeficiency Virus Viremia and Responsiveness to Antiretroviral Therapy in Macaques
- Author
-
Nika Hajari, Deborah H. Fuller, James I. Mullins, Hillary C. Tunggal, Thomas B. Lewis, Paul V. Munson, Kenneth C. Bagley, Cassie Moats, Debra Bratt, Jeremy Smedley, and Megan A. O'Connor
- Subjects
Male ,0301 basic medicine ,Cart ,Colon ,T cell ,Immunology ,Simian Acquired Immunodeficiency Syndrome ,HIV Infections ,chemical and pharmacologic phenomena ,Inflammation ,Viremia ,Pathogenesis ,medicine.disease_cause ,T-Lymphocytes, Regulatory ,Virus ,03 medical and health sciences ,0302 clinical medicine ,TIGIT ,Virology ,Animals ,Homeostasis ,Medicine ,Mesenteric lymph nodes ,Mesentery ,030212 general & internal medicine ,Intestinal Mucosa ,business.industry ,Monkey Diseases ,virus diseases ,hemic and immune systems ,Viral Load ,Simian immunodeficiency virus ,medicine.disease ,Macaca mulatta ,Disease Models, Animal ,Treatment Outcome ,030104 developmental biology ,Infectious Diseases ,medicine.anatomical_structure ,Anti-Retroviral Agents ,Th17 Cells ,Simian Immunodeficiency Virus ,Lymph Nodes ,medicine.symptom ,business - Abstract
Depletion of gut T helper 17 (Th17) cells during HIV infection leads to decreased mucosal integrity and increased disease progression. Conversely, T regulatory (Treg) cells may inhibit antiviral responses or immune activation. In HIV elite controllers, a balanced Th17/Treg ratio is maintained in the blood, suggesting a role for these responses in controlling inflammation and viral replication. HIV-infected individuals exhibit a range in responsiveness to combination antiretroviral therapy (cART). Given the link between the Th17/Treg ratio and HIV disease, we reasoned these responses may play a role in cART responsiveness. In this study, we investigated the relationship between the mucosal Th17/Treg ratio to acute simian immunodeficiency virus (SIV) viremia and the response to cART. Nineteen rhesus macaques were infected with highly pathogenic SIVΔB670 virus and cART was initiated 6 weeks postinfection. Mucosal CD4 T cell subsets were assessed by intracellular cytokine staining in the colon and mesenteric lymph nodes. Higher baseline Th17/Treg ratios corresponded with increased acute SIV viremia. Th17/Treg ratios decreased during acute SIV infection and were not restored during cART, and this corresponded to increased gut immune activation (Ki67(+)), markers of microbial translocation (sCD14), and T cell exhaustion (TIGIT(+)). Animals that maintained a more balanced mucosal Th17/Treg ratio at the time of cART initiation exhibited a better virological response to cART and maintained higher peripheral CD4 counts. These results suggest mucosal Th17 and Treg homeostasis influences acute viremia and the response to cART, a result that suggests therapeutic interventions that improve the Th17/Treg ratio before or during cART may improve treatment of HIV.
- Published
- 2019
- Full Text
- View/download PDF
35. Cytomegalovirus-vaccine-induced unconventional T cell priming and control of SIV replication is conserved between primate species
- Author
-
Daniel Malouli, Roxanne M. Gilbride, Helen L. Wu, Joseph M. Hwang, Nicholas Maier, Colette M. Hughes, Daniel Newhouse, David Morrow, Abigail B. Ventura, Lynn Law, Jennifer Tisoncik-Go, Leanne Whitmore, Elise Smith, Inah Golez, Jean Chang, Jason S. Reed, Courtney Waytashek, Whitney Weber, Husam Taher, Luke S. Uebelhoer, Jennie L. Womack, Matthew R. McArdle, Junwei Gao, Courtney R. Papen, Jeffrey D. Lifson, Benjamin J. Burwitz, Michael K. Axthelm, Jeremy Smedley, Klaus Früh, Michael Gale, Louis J. Picker, Scott G. Hansen, and Jonah B. Sacha
- Subjects
AIDS Vaccines ,Interleukin-15 ,SAIDS Vaccines ,Simian Acquired Immunodeficiency Syndrome ,Cytomegalovirus ,CD8-Positive T-Lymphocytes ,Macaca mulatta ,Microbiology ,Cytomegalovirus Vaccines ,Macaca fascicularis ,Virology ,Cytomegalovirus Infections ,Animals ,Simian Immunodeficiency Virus ,Parasitology - Abstract
Strain 68-1 rhesus cytomegalovirus expressing simian immunodeficiency virus (SIV) antigens (RhCMV/SIV) primes MHC-E-restricted CD8+ T cells that control SIV replication in 50%-60% of the vaccinated rhesus macaques. Whether this unconventional SIV-specific immunity and protection is unique to rhesus macaques or RhCMV or is intrinsic to CMV remains unknown. Here, using cynomolgus CMV vectors expressing SIV antigens (CyCMV/SIV) and Mauritian cynomolgus macaques, we demonstrate that the induction of MHC-E-restricted CD8+ T cells requires matching CMV to its host species. RhCMV does not elicit MHC-E-restricted CD8+ T cells in cynomolgus macaques. However, cynomolgus macaques vaccinated with species-matched 68-1-like CyCMV/SIV mounted MHC-E-restricted CD8+ T cells, and half of the vaccinees stringently controlled SIV post-challenge. Protected animals manifested a vaccine-induced IL-15 transcriptomic signature that is associated with efficacy in rhesus macaques. These findings demonstrate that the ability of species-matched CMV vectors to elicit MHC-E-restricted CD8+ T cells that are required for anti-SIV efficacy is conserved in nonhuman primates, and these data support the development of HCMV/HIV for a prophylactic HIV vaccine.
- Published
- 2022
- Full Text
- View/download PDF
36. A Gut Reaction to SIV and SHIV Infection: Lower Dysregulation of Mucosal T Cells during Acute Infection Is Associated with Greater Viral Suppression during cART
- Author
-
Cassandra Moats, Christopher W. Peterson, Thomas B. Lewis, Deborah H. Fuller, Jessica M. Osborn, Paul V. Munson, Jeremy Smedley, Kenneth C. Bagley, Hillary C. Tunggal, Megan A. O'Connor, Sandra Dross, Hans-Peter Kiem, Keith R. Jerome, James I. Mullins, and Meei Li W. Huang
- Subjects
Cart ,Male ,Sustained Virologic Response ,animal diseases ,viruses ,Simian Acquired Immunodeficiency Syndrome ,Acute infection ,Viremia ,cART ,medicine.disease_cause ,Virus Replication ,Microbiology ,T-Lymphocytes, Regulatory ,Article ,Virological response ,Acquired immunodeficiency syndrome (AIDS) ,Virology ,medicine ,Animals ,Homeostasis ,Viral suppression ,Immunity, Mucosal ,Intraepithelial Lymphocytes ,T helper 17 (Th17) ,mucosal dysfunction ,colon ,business.industry ,virus diseases ,Immune dysregulation ,Viral Load ,medicine.disease ,Macaca mulatta ,QR1-502 ,Gastrointestinal Tract ,AIDS models ,Kinetics ,non-human primate (NHP) ,Infectious Diseases ,Anti-Retroviral Agents ,SHIV ,SIV ,Immunology ,Acute Disease ,Models, Animal ,Th17 Cells ,Simian Immunodeficiency Virus ,business ,T regulatory - Abstract
Selection of a pre-clinical non-human primate (NHP) model is essential when evaluating therapeutic vaccine and treatment strategies for HIV. SIV and SHIV-infected NHPs exhibit a range of viral burdens, pathologies, and responses to combinatorial antiretroviral therapy (cART) regimens and the choice of the NHP model for AIDS could influence outcomes in studies investigating interventions. Previously, in rhesus macaques (RMs) we showed that maintenance of mucosal Th17/Treg homeostasis during SIV infection correlated with a better virological response to cART. Here, in RMs we compared viral kinetics and dysregulation of gut homeostasis, defined by T cell subset disruption, during highly pathogenic SIVΔB670 compared to SHIV-1157ipd3N4 infection. SHIV infection resulted in lower acute viremia and less disruption to gut CD4 T-cell homeostasis. Additionally, 24/24 SHIV-infected versus 10/19 SIV-infected animals had sustained viral suppression <, 100 copies/mL of plasma after 5 months of cART. Significantly, the more profound viral suppression during cART in a subset of SIV and all SHIV-infected RMs corresponded with less gut immune dysregulation during acute SIV/SHIV infection, defined by maintenance of the Th17/Treg ratio. These results highlight significant differences in viral control during cART and gut dysregulation in NHP AIDS models and suggest that selection of a model may impact the evaluation of candidate therapeutic interventions for HIV treatment and cure strategies.
- Published
- 2021
37. CD8+ T cells fail to limit SIV reactivation following ART withdrawal until after viral amplification
- Author
-
Paul T. Edlefsen, Brandon F. Keele, Louis J. Picker, Alejandra Marenco, Jeremy Smedley, Jacob D. Estes, Kathleen Busman-Sahay, Roxanne M. Gilbride, Haesun Park, Miles P. Davenport, Mykola Pinkevych, Michael Nekorchuk, Yoshinori Fukazawa, Afam A. Okoye, Benjamin Varco-Merth, Morgan Chaunzwa, Andrea N. Selseth, Hannah Behrens, Michael K. Axthelm, Jeffery D. Lifson, Jason Shao, Scott G. Hansen, Derick D. Duell, and Romas Geleziunas
- Subjects
Male ,0301 basic medicine ,medicine.drug_class ,T cell ,Simian Acquired Immunodeficiency Syndrome ,Viremia ,CD8-Positive T-Lymphocytes ,Biology ,Monoclonal antibody ,Lymphocyte Depletion ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Animals ,Cytotoxic T cell ,RNA ,General Medicine ,Acquired immune system ,medicine.disease ,Macaca mulatta ,Virology ,030104 developmental biology ,medicine.anatomical_structure ,Anti-Retroviral Agents ,Viral replication ,030220 oncology & carcinogenesis ,Female ,Simian Immunodeficiency Virus ,Virus Activation ,CD8 ,Research Article - Abstract
To define the contribution of CD8(+) T cell responses to control of SIV reactivation during and following antiretroviral therapy (ART), we determined the effect of long-term CD8(+) T cell depletion using a rhesusized anti-CD8β monoclonal antibody on barcoded SIVmac239 dynamics on stable ART and after ART cessation in rhesus macaques (RMs). Among the RMs with full CD8(+) T cell depletion in both blood and tissue, there were no significant differences in the frequency of viral blips in plasma, the number of SIV RNA(+) cells and the average number of RNA copies/infected cell in tissue, and levels of cell-associated SIV RNA and DNA in blood and tissue relative to control-treated RMs during ART. Upon ART cessation, both CD8(+) T cell–depleted and control RMs rebounded in fewer than 12 days, with no difference in the time to viral rebound or in either the number or growth rate of rebounding SIVmac239M barcode clonotypes. However, effectively CD8(+) T cell–depleted RMs showed a stable, approximately 2-log increase in post-ART plasma viremia relative to controls. These results indicate that while potent antiviral CD8(+) T cell responses can develop during ART-suppressed SIV infection, these responses effectively intercept post-ART SIV rebound only after systemic viral replication, too late to limit reactivation frequency or the early spread of reactivating SIV reservoirs.
- Published
- 2021
- Full Text
- View/download PDF
38. Effects of persistent modulation of intestinal microbiota on SIV/HIV vaccination in rhesus macaques
- Author
-
Andrew T. Gustin, Jessica M. Osborn, Philip Barnette, Alexander S. Zevin, Rebecca M. Lynch, Tiffany Hensley-McBain, Elias K. Haddad, Jeremy Smedley, Chul Y. Ahrens, Naoto Iwayama, Deborah H. Fuller, Nancy L. Haigwood, Alex Roederer, Solomon Wangari, Cassandra Moats, Megan A. O'Connor, Sandra Dross, Jennifer A. Manuzak, Nichole R. Klatt, Roshell Muir, Courtney Broedlow, and Ernesto Coronado
- Subjects
0301 basic medicine ,Colon ,animal diseases ,medicine.medical_treatment ,T cell ,Immunology ,Heterologous ,Article ,law.invention ,03 medical and health sciences ,Probiotic ,0302 clinical medicine ,Immune system ,law ,Medicine ,Pharmacology (medical) ,Microbiome ,RC254-282 ,Pharmacology ,business.industry ,Microbiota ,Immunogenicity ,Rectum ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,virus diseases ,RC581-607 ,Vaccination ,030104 developmental biology ,Infectious Diseases ,medicine.anatomical_structure ,Preclinical research ,Immunologic diseases. Allergy ,business ,Adjuvant ,HIV infections ,030215 immunology - Abstract
An effective vaccine to prevent HIV transmission has not yet been achieved. Modulation of the microbiome via probiotic therapy has been suggested to result in enhanced mucosal immunity. Here, we evaluated whether probiotic therapy could improve the immunogenicity and protective efficacy of SIV/HIV vaccination. Rhesus macaques were co-immunized with an SIV/HIV DNA vaccine via particle-mediated epidermal delivery and an HIV protein vaccine administered intramuscularly with Adjuplex™ adjuvant, while receiving daily oral Visbiome® probiotics. Probiotic therapy alone led to reduced frequencies of colonic CCR5+ and CCR6+ CD4+ T cells. Probiotics with SIV/HIV vaccination led to similar reductions in colonic CCR5+ CD4+ T cell frequencies. SIV/HIV-specific T cell and antibody responses were readily detected in the periphery of vaccinated animals but were not enhanced with probiotic treatment. Combination probiotics and vaccination did not impact rectal SIV/HIV target populations or reduce the rate of heterologous SHIV acquisition during the intrarectal challenge. Finally, post-infection viral kinetics were similar between all groups. Thus, although probiotics were well-tolerated when administered with SIV/HIV vaccination, vaccine-specific responses were not significantly enhanced. Additional work will be necessary to develop more effective strategies of microbiome modulation in order to enhance mucosal vaccine immunogenicity and improve protective immune responses.
- Published
- 2021
- Full Text
- View/download PDF
39. Immune inactivation of anti-simian immunodeficiency virus chimeric antigen receptor T cells in rhesus macaques
- Author
-
Rosemarie D. Mason, Lawrence Corey, Louis J. Picker, Blake J. Rust, Karsten Eichholz, Françoise Haeseleer, Benjamin Varco-Merth, Yoshinori Fukazawa, Haesun Park, Jeremy Smedley, Hans-Peter Kiem, Afam A. Okoye, Christopher W. Peterson, and Mario Roederer
- Subjects
medicine.drug_class ,Human immunodeficiency virus (HIV) ,QH426-470 ,medicine.disease_cause ,Monoclonal antibody ,infectious diseases ,CAR T cells ,Epitope ,Immune system ,medicine ,Genetics ,Effector functions ,Molecular Biology ,autologous cell therapy ,biology ,QH573-671 ,HIV ,Simian immunodeficiency virus ,Virology ,Chimeric antigen receptor ,humoral immune response ,SIV ,biology.protein ,Molecular Medicine ,Original Article ,Antibody ,Cytology ,human activities - Abstract
Chimeric antigen receptor (CAR) T cell therapies are being investigated as potential HIV cures and designed to target HIV reservoirs. Monoclonal antibodies (mAbs) targeting the simian immunodeficiency virus (SIV) envelope allowed us to investigate the potency of single-chain variable fragment (scFv)-based anti-SIV CAR T cells. In vitro, CAR T cells expressing the scFv to both the variable loop 1 (V1) or V3 of the SIV envelope were highly potent at eliminating SIV-infected T cells. However, in preclinical studies, in vivo infusion of these CAR T cells in rhesus macaques (RMs) resulted in lack of expansion and no detectable in vivo antiviral activity. Injection of envelope-expressing antigen-presenting cells (APCs) 1 week post-CAR T cell infusion also failed to stimulate CAR T cell expansion in vivo. To investigate this in vitro versus in vivo discrepancy, we examined host immune responses directed at CAR T cells. A humoral immune response against the CAR scFv was detected post-infusion of the anti-SIV CAR T cells; anti-SIV IgG antibodies present in plasma of SIV-infected animals were associated with inhibited CAR T cell effector functions. These data indicate that lack of in vivo expansion and efficacy of CAR T cells might be due to antibodies blocking the interaction between the CAR scFv and its epitope., Graphical abstract, Corey and colleagues evaluated CAR T cells targeting the envelope of simian immunodeficiency virus. They show that antibodies blocking the interaction between the CAR and its epitope might be responsible for the lack of in vivo proliferation of anti-SIV CAR T cells and killing of SIV-infected cells after infusion in rhesus macaques.
- Published
- 2021
40. Liver Biopsies Reveal Temporal Changes in Pathology and Macrophage Function following SIV Infection of Rhesus Macaques
- Author
-
Nina R Derby, Katherine A Fancher, Sreya Biswas, Sofiya Yusova, Cristina Luevano-Santos, Jeremy Smedley, Cristina Pacheco, Benjamin Burwitz, and Donald L Sodora
- Subjects
Immunology ,Immunology and Allergy - Abstract
Non-alcoholic fatty liver disease (NAFLD) contributes to morbidity and mortality in HIV-infected individuals. To evaluate liver immune changes during SIV infection of macaques, we obtained liver biopsies laparoscopically at 2, 6, and 16 weeks post-infection as well as at necropsy (32 weeks). SIV infection was associated with liver dysfunction that included elevated liver enzymes during acute infection, higher liver weight at necropsy, and histologic changes within the liver lobule (functional unit of the liver) including sinusoidal dilatation near central veins and portal expansion. The type-1 interferon (IFN-1) response (MX1 expression) increased over time post-SIV infection, initially being observed in macrophages (week 2) with a switch to a mixed hepatocyte/monocyte/macrophage response by 16–32 weeks. Over the course of SIV infection, macrophage frequencies also increased – both infiltrating (CD68+CD163+CD206-) and resident (CD68+CD163+CD206+) tissue macrophages – and these macrophages were found more near central veins over time. In contrast, CD68+CD163-CD206− trafficking myeloid cells/monocytes were detected predominantly in the periportal zone of the lobule and associated with elevated MX1 expression in that zone. Our findings identify pathologic changes within the livers of SIV infected macaques as early as 2 weeks post-infection, IFN-1 responses that are initially observed in liver macrophages, and an accumulation of macrophages associated with signs of inflammation by week 32. These studies offer quantitative and spatial insights to inform macrophage-targeted therapeutic approaches that may improve liver function during HIV infection. Supported by grants from NIH (R01AI134630)
- Published
- 2022
- Full Text
- View/download PDF
41. RhCMV/SIV tropism modulation programs unconventional CD8+ T cell priming and vaccine efficacy
- Author
-
Meaghan Hancock, Scott Hansen, Daniel Malouli, Emily Marshall, Collette Hughes, Kurt T. Randall, David Morrow, Julia Ford, Roxanne Gilbride, Andrea Selseth, Renee Espinosa Trethewy, Lindsey Bishop, Kelli Oswald, Rebecca Shoemaker, Brian Berkemeier, William Bosche, Michael Hull, Michael Nekorchuk, Kathleen Busman-Sahay, Jacob Estes, Michael Axthelm, Jeremy Smedley, Danica Shao, Paul Edlefsen, Jeffrey Lifson, Klaus Fruh, Jay Nelson, and Louis J Picker
- Subjects
Immunology ,Immunology and Allergy - Abstract
Strain 68-1 rhesus cytomegalovirus (RhCMV) vectors expressing simian immunodeficiency virus (SIV) antigens demonstrate a vaccine efficacy where 50–60% of vaccinated rhesus macaques are protected from SIV challenge. Intriguingly, RhCMV/SIV vectors elicit CD8+ T cells recognizing epitopes presented by MHC-II and MHC-E instead of MHC-Ia. We are studying how these unconventional T cell responses are elicited and contribute to the efficacy against SIV challenge. Here we utilize host microRNA (miRNA)-mediated vector tropism restriction to show that MHC-II- and MHC-E-restricted responses are primed by directly infected, non-overlapping cell types in rhesus macaques. Targeting essential RhCMV genes with myeloid cell-selective miR-142-3p eliminated MHC-E-restricted CD8+ T cell priming, yielding an exclusively MHC-II-restricted response, whereas endothelial cell-selective miR-126-3p targeting eliminated MHC-II-restricted CD8+ T cell priming, yielding an exclusively MHC-E-restricted response. Incorporation of both restriction elements reverts CD8+ T cell responses back to conventional MHC-Ia restriction. Using these otherwise isogenic vectors we show that although they demonstrate similar overall immunogenicity, only the vectors programmed to elicit MHC-E-restricted CD8+ T cell responses provided protection against SIV challenge. The MHC-E-only RhCMV/SIV vaccine efficacy did not exceed that of the parental 68-1 RhCMV/SIV vectors (that elicits both MHC-II and MHC-E responses) indicating that while the MHC-II-restricted CD8+ T cell responses are neutral to overall vaccine efficacy, an additional component of 68-1 RhCMV/SIV-induced immunity contributes to overall vaccine efficacy. This work was supported by the National Institute of Allergy and Infectious Diseases (NIAID) grants UM1 AI124377 and U19 AI128741 to LJP; the Oregon National Primate Research Center Core grant from the National Institutes of Health, Office of the Director (P51 OD011092); contracts from the National Cancer Institute (# HHSN261200800001E) to JDL; and the Bill and Melinda Gates Foundation grant OPP1107409.
- Published
- 2022
- Full Text
- View/download PDF
42. Liver bacterial dysbiosis occurs in SIV-infected macaques and persists during antiretroviral therapy
- Author
-
Nichole R. Klatt, Cole Fisher, Jeremy Smedley, Katherine A. Fancher, Donald L. Sodora, Matthew P. Wood, Bridget S. Fisher, Andrew T. Gustin, Nina Derby, Benjamin J. Burwitz, and Michael Gale
- Subjects
business.industry ,virus diseases ,Medicine ,business ,medicine.disease ,Antiretroviral therapy ,Virology ,Dysbiosis - Abstract
Background: Liver disease remains a significant contributor to morbidity and mortality in HIV-infected individuals, even during successful treatment with combination antiretroviral therapy (cART). In non-human primates, SIV infection is associated with gut microbiome dysbiosis as well as bacterial translocation into the colonic lamina propria and liver via the portal vein. Here the liver microbiome was evaluated in rhesus macaques to discern the influence of SIV infection alone (SIV+) and during cART administration (SIV+cART) on liver bacterial dysbiosis and neutrophil infiltration.Results: Dysbiosis in liver bacterial composition was observed, encompassing changes in a number of genera, during SIV infection in the absence and presence of cART. The most striking finding was an increase in the level of Mycobacterium, which while barely detectable in the uninfected macaques, was the most abundant genus observed in the livers of a majority SIV+ and SIV+cART macaques. Multi-gene sequencing analyses identified a species of environmental mycobacteria similar to the opportunistic pathogen M. smegmatis. The effect of M. smegmatis on host gene expression in primary hepatocytes was evaluated in vitro utilizing PILAM, a glycolipid cell wall component found in atypical Mycobacteria. PILAM induced an upregulation of inflammatory responses, including an increase in the chemokines associated with neutrophil chemotaxis (CXCL1, CXCL5, and CXCL6). Assessment of the macaque livers by microscopy determined that neutrophil levels were reduced in SIV+cART macaques, suggesting that the SIV infection and/or cART treatment influence the liver-associated neutrophil response. Conclusions: A number of liver bacteria genera were altered following SIV infection even in the context of cART, possibly as a consequence of reduced neutrophil recruitment. Mycobacteria became a major component of the SIV infected macaque liver microbiome, raising the possibility that bacteria of this genus might contribute to liver disease in HIV infected patients.
- Published
- 2020
- Full Text
- View/download PDF
43. In vitro and in vivo characterization of a recombinant rhesus cytomegalovirus containing a complete genome
- Author
-
Travis Whitmer, Jason Shao, Scott G. Hansen, Richard J. Stanton, Xiaofei E, Paul T. Edlefsen, Lesli M. Sprehe, Hillary C. Cleveland-Rubeor, Amitinder Kaur, Matilda J. Moström, Matthew R. McArdle, Colette M. Hughes, Peter A. Barry, Daniel Malouli, Andrea N. Selseth, Michael Nekorchuk, Eisa Mahyari, Jacob D. Estes, Michael K. Axthelm, Kerianne A. Jackson, Craig N. Kreklywich, Daniel N. Streblow, Yujuan Yue, Timothy F. Kowalik, Jeremy Smedley, Klaus Früh, Husam Taher, Benjamin N. Bimber, Luke S. Uebelhoer, Amruta Bhusari, Dawn L. Roberts, Louis J. Picker, Kimberli A. Schmidt, Elizabeth A. Scheef, Abigail B. Ventura, and Kalejta, Robert F
- Subjects
Cytomegalovirus Infection ,Male ,Viral Diseases ,Chromosomes, Artificial, Bacterial ,Physiology ,Cytomegalovirus ,Urine ,Monkeys ,Genome ,Biochemistry ,Recombineering ,Medical Conditions ,Animal Cells ,Medicine and Health Sciences ,2.2 Factors relating to the physical environment ,Viral ,Biology (General) ,Aetiology ,Phylogeny ,Connective Tissue Cells ,Mammals ,Recombinant ,Mammalian Genomics ,Bacterial ,Eukaryota ,Genomics ,Body Fluids ,Nucleic acids ,Infectious Diseases ,Medical Microbiology ,Connective Tissue ,Artificial ,Vertebrates ,Cytomegalovirus Infections ,Female ,Cellular Types ,Anatomy ,Infection ,Macaque ,Biotechnology ,Research Article ,Primates ,QH301-705.5 ,Gene prediction ,Immunology ,DNA, Recombinant ,DNA repair ,Viremia ,Genome, Viral ,Biology ,Microbiology ,Virus ,Chromosomes ,Cell Line ,Open Reading Frames ,Species Specificity ,In vivo ,Virology ,Old World monkeys ,medicine ,Genetics ,Animals ,Humans ,Gene Prediction ,Molecular Biology ,Gene ,Bacterial artificial chromosome ,Animal ,Organisms ,Biology and Life Sciences ,Computational Biology ,Cell Biology ,DNA ,RC581-607 ,Fibroblasts ,medicine.disease ,Genome Analysis ,Macaca mulatta ,Disease Models, Animal ,Biological Tissue ,Animal Genomics ,Disease Models ,Amniotes ,Mutation ,Parasitology ,Immunologic diseases. Allergy ,Zoology - Abstract
Cytomegaloviruses (CMVs) are highly adapted to their host species resulting in strict species specificity. Hence, in vivo examination of all aspects of CMV biology employs animal models using host-specific CMVs. Infection of rhesus macaques (RM) with rhesus CMV (RhCMV) has been established as a representative model for infection of humans with HCMV due to the close evolutionary relationships of both host and virus. However, the only available RhCMV clone that permits genetic modifications is based on the 68–1 strain which has been passaged in fibroblasts for decades resulting in multiple genomic changes due to tissue culture adaptations. As a result, 68–1 displays reduced viremia in RhCMV-naïve animals and limited shedding compared to non-clonal, low passage isolates. To overcome this limitation, we used sequence information from primary RhCMV isolates to construct a full-length (FL) RhCMV by repairing all mutations affecting open reading frames (ORFs) in the 68–1 bacterial artificial chromosome (BAC). Inoculation of adult, immunocompetent, RhCMV-naïve RM with the reconstituted virus resulted in significant viremia in the blood similar to primary isolates of RhCMV and furthermore led to high viral genome copy numbers in many tissues at day 14 post infection. In contrast, viral dissemination was greatly reduced upon deletion of genes also lacking in 68–1. Transcriptome analysis of infected tissues further revealed that chemokine-like genes deleted in 68–1 are among the most highly expressed viral transcripts both in vitro and in vivo consistent with an important immunomodulatory function of the respective proteins. We conclude that FL-RhCMV displays in vitro and in vivo characteristics of a wildtype virus while being amenable to genetic modifications through BAC recombineering techniques., Author summary Human cytomegalovirus (HCMV) infections are generally asymptomatic in healthy immunocompetent individuals, but HCMV can cause serious disease after congenital infection and in individuals with immunocompromised immune systems. Since HCMV is highly species specific and cannot productively infect immunocompetent laboratory animals, experimental infection of rhesus macaques (RM) with rhesus CMV (RhCMV) has been established as a closely related animal model for HCMV. By employing the unique ability of CMV to elicit robust and lasting cellular immunity, this model has also been instrumental in developing novel CMV-based vaccines against chronic and recurring infections with pathogens such as the human immunodeficiency virus (HIV) and Mycobacterium tuberculosis (Mtb). However, most of this work was conducted with derivatives of the 68–1 strain of RhCMV which has acquired multiple genomic alterations in tissue culture. To model pathogenesis and immunology of clinical HCMV isolates we generated a full-length (FL) RhCMV clone representative of low passage isolates. Infection of RhCMV-naïve RM with FL-RhCMV demonstrated viremia and tissue dissemination that was comparable to that of non-clonal low passage isolates. We further demonstrate that FL-RhCMV is strongly attenuated upon deletion of gene regions absent in 68–1 thus demonstrating the usefulness of FL-RhCMV to study RhCMV pathogenesis.
- Published
- 2020
44. Oral Immunization with HIV-1 Envelope SOSIP trimers elicits systemic immune responses and cross-reactive anti-V1V2 antibodies in non-human primates
- Author
-
Daniela V. Andrade, Nicholas Dambrauskas, D. Noah Sather, Bridget S. Fisher, Donald L. Sodora, Jeremy Smedley, and Olesya Trakhimets
- Subjects
0301 basic medicine ,B Cells ,Physiology ,medicine.medical_treatment ,Antibody Response ,Administration, Oral ,HIV Infections ,Pilot Projects ,Monkeys ,Biochemistry ,chemistry.chemical_compound ,White Blood Cells ,0302 clinical medicine ,Viral Envelope Proteins ,Animal Cells ,Immune Physiology ,Medicine and Health Sciences ,Public and Occupational Health ,HIV vaccine ,Enzyme-Linked Immunoassays ,Immune Response ,Mammals ,AIDS Vaccines ,Vaccines ,Multidisciplinary ,Immune System Proteins ,biology ,Imidazoles ,Eukaryota ,Vaccination and Immunization ,Vaccination ,Infectious Diseases ,030220 oncology & carcinogenesis ,Vertebrates ,Medicine ,Resiquimod ,Antibody ,Cellular Types ,Adjuvant ,Macaque ,Research Article ,Primates ,Infectious Disease Control ,Science ,Immune Cells ,Immunology ,Cross Reactions ,Research and Analysis Methods ,Virus ,Antibodies ,03 medical and health sciences ,Immune system ,Antigen ,Adjuvants, Immunologic ,Old World monkeys ,medicine ,Animals ,Immunoassays ,Antibody-Producing Cells ,Blood Cells ,business.industry ,Organisms ,Biology and Life Sciences ,Proteins ,Cell Biology ,Macaca mulatta ,030104 developmental biology ,chemistry ,Amniotes ,biology.protein ,Immunologic Techniques ,HIV-1 ,Preventive Medicine ,business - Abstract
Development of a successful HIV vaccine is dependent upon a determination of the optimum antigen and adjuvant as well as choosing an optimal site for vaccine delivery. The site of delivery is particularly relevant as HIV transmission generally requires that the virus crosses a mucosal membrane to infect a new host. Here we undertake a pilot study comparing three vaccine delivery routes, two to the oral cavity (intraepithelial (iEp) and needle-free (NF-Injex)) as well as intramuscular (IM) delivery. These vaccinations utilized a recombinant HIV-1 Env trimer 10042.05 from an elite neutralizer, subject VC10042, that has previously induced high titers of cross-clade reactive V1V2 antibodies. The 10042.05.SOSIP fused trimer was administered with adjuvants R848 (Resiquimod), MPLA and Alhydrogel to characterize the innate cellular and anti-HIV Envelope (Env) antibody responses following the administration of the vaccine to the oral mucosa. Oral delivery of the 10042.05.SOSIP induced high titers of anti-V1V2 antibodies, which together with previous studies, indicates an immunogenic bias toward the V1V2 regions in 10042-derived Envs. Both types of oral vaccine delivery resulted in immunologic and serologic responses that were comparable to the IM delivery route. Furthermore, induction of anti-V1-V2 specific antibodies was best following iEp delivery of the oral vaccine identifying this as the optimal method to orally deliver this vaccine formulation.
- Published
- 2020
45. The human IL-15 superagonist N-803 promotes migration of virus-specific CD8+ T and NK cells to B cell follicles but does not reverse latency in ART-suppressed, SHIV-infected macaques
- Author
-
Jacob D. Estes, Jason S. Reed, Katherine B. Bateman, Justin M. Greene, Lina Gao, Pamela J. Skinner, Gabriela M. Webb, Nicholas Maier, Kathleen Busman-Sahay, Helen L. Wu, Jeffrey T. Safrit, Jeremy Smedley, Brianna C Davey, Jeffrey J. Stanton, Whitney C. Weber, Jonah B. Sacha, Jhomary S Molden, Shaheed A. Abdulhaqq, Benjamin L. Carpenter, Michael K. Axthelm, Mina Northrup, and Shiho Tanaka
- Subjects
RNA viruses ,B Cells ,Simian Acquired Immunodeficiency Syndrome ,HIV Infections ,NK cells ,Monkeys ,CD8-Positive T-Lymphocytes ,Pathology and Laboratory Medicine ,White Blood Cells ,Latent Virus ,Immunodeficiency Viruses ,Animal Cells ,Cell Movement ,Medicine and Health Sciences ,Cytotoxic T cell ,Biology (General) ,Mammals ,Interleukin-15 ,B-Lymphocytes ,0303 health sciences ,T Cells ,030302 biochemistry & molecular biology ,Eukaryota ,Viral Load ,Virus Latency ,3. Good health ,Killer Cells, Natural ,medicine.anatomical_structure ,Anti-Retroviral Agents ,Medical Microbiology ,Interleukin 15 ,Viral Pathogens ,Viruses ,Vertebrates ,Simian Immunodeficiency Virus ,Cellular Types ,Anatomy ,Pathogens ,Viral load ,Macaque ,Research Article ,Primates ,QH301-705.5 ,Immune Cells ,Recombinant Fusion Proteins ,Immunology ,Cytotoxic T cells ,Biology ,Microbiology ,Virus ,Lymphatic System ,03 medical and health sciences ,Virology ,Retroviruses ,Old World monkeys ,Genetics ,medicine ,Animals ,Humans ,Antibody-Producing Cells ,Microbial Pathogens ,Molecular Biology ,B cell ,030304 developmental biology ,Blood Cells ,Lentivirus ,Organisms ,Biology and Life Sciences ,HIV ,Proteins ,Cell Biology ,RC581-607 ,Macaca mulatta ,Disease Models, Animal ,Viral replication ,Amniotes ,HIV-1 ,Parasitology ,Lymph Nodes ,Immunologic diseases. Allergy ,Viral Transmission and Infection ,CD8 - Abstract
Despite the success of antiretroviral therapy (ART) to halt viral replication and slow disease progression, this treatment is not curative and there remains an urgent need to develop approaches to clear the latent HIV reservoir. The human IL-15 superagonist N-803 (formerly ALT-803) is a promising anti-cancer biologic with potent immunostimulatory properties that has been extended into the field of HIV as a potential “shock and kill” therapeutic for HIV cure. However, the ability of N-803 to reactivate latent virus and modulate anti-viral immunity in vivo under the cover of ART remains undefined. Here, we show that in ART-suppressed, simian-human immunodeficiency virus (SHIV)SF162P3-infected rhesus macaques, subcutaneous administration of N-803 activates and mobilizes both NK cells and SHIV-specific CD8+ T cells from the peripheral blood to lymph node B cell follicles, a sanctuary site for latent virus that normally excludes such effector cells. We observed minimal activation of memory CD4+ T cells and no increase in viral RNA content in lymph node resident CD4+ T cells post N-803 administration. Accordingly, we found no difference in the number or magnitude of plasma viremia timepoints between treated and untreated animals during the N-803 administration period, and no difference in the size of the viral DNA cell-associated reservoir post N-803 treatment. These results substantiate N-803 as a potent immunotherapeutic candidate capable of activating and directing effector CD8+ T and NK cells to the B cell follicle during full ART suppression, and suggest N-803 must be paired with a bona fide latency reversing agent in vivo to facilitate immune-mediated modulation of the latent viral reservoir., Author summary IL-15 regulates NK and memory T cell homeostasis and is therefore being explored for clinical immunotherapy of chronic diseases like cancer and HIV. To explore the applicability of the clinical grade IL-15 superagonist N-803 to HIV cure strategies we tested the impact of N-803 on host immunity and latent virus in SHIV-infected rhesus macaques. Our results suggest that N-803 beneficially modulates effector NK and CD8+ T cells by expanding the numbers of these cells and redistributing them to lymph node B cell follicles, a site known to harbor persistent latent virus during ART. However, our results further suggest that N-803 does not perturb the viral reservoir present in memory CD4+ T cells and that in order to fully unlock the immunotherapeutic potential of N-803 it must be paired with latency reversal agents.
- Published
- 2020
46. Early cellular innate immune responses drive Zika viral persistence and tissue tropism in pigtail macaques
- Author
-
Thomas B. Lewis, Charlene Miller, Cassie Moats, Paul T. Edlefsen, Megan A. O'Connor, Michael Gale, Debra Bratt, Jennifer Tisoncik-Go, Deborah H. Fuller, Jeremy Smedley, and Nichole R. Klatt
- Subjects
0301 basic medicine ,Science ,Viral pathogenesis ,viruses ,General Physics and Astronomy ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Article ,Zika virus ,Pathogenesis ,03 medical and health sciences ,0302 clinical medicine ,Immunity ,In vivo ,Animals ,030212 general & internal medicine ,lcsh:Science ,Chemokine CCL2 ,Multidisciplinary ,Innate immune system ,Zika Virus Infection ,General Chemistry ,Pigtail macaque ,Zika Virus ,biochemical phenomena, metabolism, and nutrition ,biology.organism_classification ,Immunity, Innate ,3. Good health ,030104 developmental biology ,Immunology ,Tissue tropism ,lcsh:Q ,Macaca nemestrina - Abstract
The immunological and virological events that contribute to the establishment of Zika virus (ZIKV) infection in humans are unclear. Here, we show that robust cellular innate immune responses arising early in the blood and tissues in response to ZIKV infection are significantly stronger in males and correlate with increased viral persistence. In particular, early peripheral blood recruitment of plasmacytoid dendritic cells and higher production of monocyte chemoattractant protein (MCP-1) correspond with greater viral persistence and tissue dissemination. We also identify non-classical monocytes as primary in vivo targets of ZIKV infection in the blood and peripheral lymph node. These results demonstrate the potential differences in ZIKV pathogenesis between males and females and a key role for early cellular innate immune responses in the blood in viral dissemination and ZIKV pathogenesis., The immune response to Zika virus is required to curtail the infection and avoid immunopathology, but may be involved in the associated pathophysiology. Here the authors show that viral persistence and tissue tropism is shaped by an early innate immune response in a pigtail macaque model of infection.
- Published
- 2018
- Full Text
- View/download PDF
47. Evaluating latency reactivation synergies between the bromodomain inhibitor iBET-151 and the SMAC mimetic AZD5582 in SIV-infected macaques on ART
- Author
-
Afam A. Okoye, B.E. Randall, Jeremy Smedley, Richard M. Dunham, R. Lum, Shane D. Falcinelli, Louis J. Picker, Jeffrey D. Lifson, B. Varco-Merth, and Yoshinori Fukazawa
- Subjects
Epidemiology ,Immunology ,Public Health, Environmental and Occupational Health ,Biology ,Smac mimetics ,Microbiology ,QR1-502 ,Bromodomain ,Infectious Diseases ,Virology ,Cancer research ,Latency (engineering) ,Public aspects of medicine ,RA1-1270 - Published
- 2019
48. Viral opportunistic infections in Mauritian cynomolgus macaques undergoing allogeneic stem cell transplantation mirror human transplant infectious disease complications
- Author
-
Heidi Price, Lois M. A. Colgin, Tonya Swanson, Mina Northrup, Gabrielle Meyers, Helen L. Wu, Rhonda MacAllister, Katherine B. Bateman, Kimberly Armantrout, Jeffrey J. Stanton, Michael K. Axthelm, Benjamin N. Bimber, Mark K. Slifka, Richard T. Maziarz, Lauren D. Martin, Archana Thomas, Jeremy Smedley, Cassandra Moats, Christine Shriver-Munsch, Stephanie L. Junell, Anne D. Lewis, Whitney C. Weber, Benjamin J. Burwitz, Theodore R. Hobbs, Jonah B. Sacha, Eisa Mahyari, Jason S. Reed, Mitchell Robertson-LeVay, and Alfred W. Legasse
- Subjects
0301 basic medicine ,Human cytomegalovirus ,medicine.medical_treatment ,Immunology ,Hematopoietic stem cell transplantation ,030230 surgery ,Opportunistic Infections ,medicine.disease_cause ,Macaque ,Virus ,Article ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,biology.animal ,medicine ,Animals ,Humans ,Transplantation ,biology ,business.industry ,Hematopoietic Stem Cell Transplantation ,medicine.disease ,Allografts ,BK virus ,Disease Models, Animal ,Macaca fascicularis ,030104 developmental biology ,surgical procedures, operative ,Virus Diseases ,business ,Hemorrhagic cystitis - Abstract
Allogeneic hematopoietic stem cell transplantation (HSCT) and xenotransplantation are accompanied by viral reactivations and virus-associated complications resulting from immune deficiency. Here, in a Mauritian cynomolgus macaque model of fully MHC-matched allogeneic HSCT, we report reactivations of cynomolgus polyomavirus, lymphocryptovirus, and cytomegalovirus, macaque viruses analogous to HSCT-associated human counterparts BK virus, Epstein-Barr virus, and human Cytomegalovirus. Viral replication in recipient macaques resulted in characteristic disease manifestations observed in HSCT patients, such as polyomavirus-associated hemorrhagic cystitis and tubulointerstitial nephritis or lymphocryptovirus-associated post-transplant lymphoproliferative disorder. However, in most cases, the reconstituted immune system, alone or in combination with short-term pharmacological intervention, exerted control over viral replication, suggesting engraftment of functional donor-derived immunity. Indeed, the donor-derived reconstituted immune systems of two long-term engrafted HSCT recipient macaques responded to live attenuated yellow fever 17D vaccine (YFV 17D) indistinguishably from untransplanted controls, mounting 17D-targeted neutralizing antibody responses and clearing YFV 17D within 14 days. Together, these data demonstrate that this macaque model of allogeneic HSCT recapitulates clinical situations of opportunistic viral infections in transplant patients, and provides a pre-clinical model to test novel prophylactic and therapeutic modalities.
- Published
- 2019
49. Simian-Human Immunodeficiency Virus SHIV.CH505 Infection of Rhesus Macaques Results in Persistent Viral Replication and Induces Intestinal Immunopathology
- Author
-
Nichole R. Klatt, Cassie Moats, Elias K. Haddad, Naoto Iwayama, Nancy L. Haigwood, Jessica M. Osborn, Katharine J. Bar, Chul Y. Ahrens, Ernesto Coronado, George M. Shaw, Toni M. Gott, Deborah H. Fuller, Megan A. O'Connor, Jeremy Smedley, Solomon Wangari, Charlene Miller, Tiffany Hensley-McBain, Rebecca M. Lynch, and Jennifer A. Manuzak
- Subjects
animal diseases ,viruses ,Immunology ,Simian Acquired Immunodeficiency Syndrome ,HIV Infections ,Virus Replication ,Models, Biological ,Microbiology ,Macaque ,Virus ,03 medical and health sciences ,0302 clinical medicine ,Intestinal mucosa ,Virology ,biology.animal ,Immunopathology ,medicine ,Animals ,Humans ,Intestinal Mucosa ,Immunodeficiency ,030304 developmental biology ,0303 health sciences ,Innate immune system ,biology ,env Gene Products, Human Immunodeficiency Virus ,HIV ,virus diseases ,Viral Load ,medicine.disease ,Macaca mulatta ,Disease Models, Animal ,Viral replication ,Insect Science ,HIV-1 ,Pathogenesis and Immunity ,Simian Immunodeficiency Virus ,Genetic Engineering ,CD8 ,030215 immunology - Abstract
Simian-human immunodeficiency viruses (SHIVs) have been utilized to test vaccine efficacy and characterize mechanisms of viral transmission and pathogenesis. However, the majority of SHIVs currently available have significant limitations in that they were developed using sequences from chronically HIV-infected individuals or uncommon HIV subtypes or were optimized for the macaque model by serially passaging the engineered virus in vitro or in vivo. Recently, a newly developed SHIV, SHIV.C.CH505.375H.dCT (SHIV.CH505), which incorporates vpu-env (gp140) sequences from a transmitted/founder HIV-1 subtype C strain, was shown to retain attributes of primary HIV-1 strains. However, a comprehensive analysis of the immunopathology that results from infection with this virus, especially in critical tissue compartments like the intestinal mucosa, has not been completed. In this study, we evaluated the viral dynamics and immunopathology of SHIV.CH505 in rhesus macaques. In line with previous findings, we found that SHIV.CH505 is capable of infecting and replicating efficiently in rhesus macaques, resulting in peripheral viral kinetics similar to that seen in pathogenic SIV and HIV infection. Furthermore, we observed significant and persistent depletions of CCR5(+) and CCR6(+) CD4(+) T cells in mucosal tissues, decreases in CD4(+) T cells producing Th17 cell-associated cytokines, CD8(+) T cell dysfunction, and alterations of B cell and innate immune cell function, indicating that SHIV.CH505 elicits intestinal immunopathology typical of SIV/HIV infection. These findings suggest that SHIV.CH505 recapitulates the early viral replication dynamics and immunopathogenesis of HIV-1 infection of humans and thus can serve as a new model for HIV-1 pathogenesis, treatment, and prevention research. IMPORTANCE The development of chimeric SHIVs has been instrumental in advancing our understanding of HIV-host interactions and allowing for in vivo testing of novel treatments. However, many of the currently available SHIVs have distinct drawbacks and are unable to fully reflect the features characteristic of primary SIV and HIV strains. Here, we utilize rhesus macaques to define the immunopathogenesis of the recently developed SHIV.CH505, which was designed without many of the limitations of previous SHIVs. We observed that infection with SHIV.CH505 leads to peripheral viral kinetics and mucosal immunopathogenesis comparable with those caused by pathogenic SIV and HIV. Overall, these data provide evidence of the value of SHIV.CH505 as an effective model of SIV/HIV infection and an important tool that can be used in future studies, including preclinical testing of new therapies or prevention strategies.
- Published
- 2019
- Full Text
- View/download PDF
50. Defining early SIV replication and dissemination dynamics following vaginal transmission
- Author
-
Celine Camus, Jeremy Smedley, Arnold Reynaldi, Christine M. Fennessey, Sean P. O’Brien, Laura Newman, Taina T. Immonen, Gregory Q. Del Prete, Miles P. Davenport, Jessica M. Conway, Claire Deleage, Brandon F. Keele, Carolyn Reid, Jeffrey D. Lifson, Timothy E. Schlub, Jacob D. Estes, and Leslie Lipkey
- Subjects
Female circumcision ,CD4-Positive T-Lymphocytes ,Time Factors ,animal diseases ,viruses ,Simian Acquired Immunodeficiency Syndrome ,Biology ,Virus Replication ,03 medical and health sciences ,0302 clinical medicine ,Animals ,Health and Medicine ,Research Articles ,030304 developmental biology ,0303 health sciences ,Multidisciplinary ,Transmission (medicine) ,virus diseases ,SciAdv r-articles ,Genitalia, Female ,Viral Load ,Phenotype ,Virology ,Macaca mulatta ,Replication (computing) ,Nonhuman primate ,3. Good health ,Lymphatic system ,Viral replication ,030220 oncology & carcinogenesis ,Vagina ,Female ,Simian Immunodeficiency Virus ,Viral load ,Research Article - Abstract
Viral dynamics and host responses to vaginal infection of SIV in nonhuman primates may identify viral vulnerabilities for HIV., Understanding HIV transmission is critical to guide the development of prophylactic interventions to prevent infection. We used a nonhuman primate (NHP) model with a synthetic swarm of sequence-tagged variants of SIVmac239 (“SIVmac239X”) and scheduled necropsy during primary infection (days 3 to 14 after challenge) to study viral dynamics and host responses to the establishment and dissemination of infection following vaginal challenge. We demonstrate that local replication was initiated at multiple sites within the female genital tract (FGT), with each site having multiple viral variants. Local replication and spread in the FGT preceded lymphatic dissemination. Innate viral restriction factors were observed but appeared to follow viral replication and were ineffective at blocking initial viral establishment and dissemination. However, major delays were observed in time to dissemination in animals and among different viral variants within the same animal. It will be important to assess how phenotypic differences affect early viral dynamics.
- Published
- 2019
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.