Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Rama Akondy is active.

Publication


Featured researches published by Rama Akondy.


The Lancet | 2008

HIV-1 vaccine-induced immunity in the test-of-concept Step Study: a case–cohort analysis

M. Juliana McElrath; Stephen C. De Rosa; Zoe Moodie; Sheri A. Dubey; Lisa Kierstead; Holly Janes; Olivier D. Defawe; Donald K. Carter; John Hural; Rama Akondy; Susan Buchbinder; Michael N. Robertson; Devan V. Mehrotra; Steven G. Self; Lawrence Corey; John W. Shiver; Danilo R. Casimiro

BACKGROUND In the Step Study, the MRKAd5 HIV-1 gag/pol/nef vaccine did not reduce plasma viraemia after infection, and HIV-1 incidence was higher in vaccine-treated than in placebo-treated men with pre-existing adenovirus serotype 5 (Ad5) immunity. We assessed vaccine-induced immunity and its potential contributions to infection risk. METHODS To assess immunogenicity, we characterised HIV-specific T cells ex vivo with validated interferon-gamma ELISPOT and intracellular cytokine staining assays, using a case-cohort design. To establish effects of vaccine and pre-existing Ad5 immunity on infection risk, we undertook flow cytometric studies to measure Ad5-specific T cells and circulating activated (Ki-67+/BcL-2(lo)) CD4+ T cells expressing CCR5. FINDINGS We detected interferon-gamma-secreting HIV-specific T cells (range 163/10(6) to 686/10(6) peripheral blood mononuclear cells) ex vivo by ELISPOT in 77% (258/354) of people receiving vaccine; 218 of 354 (62%) recognised two to three HIV proteins. We identified HIV-specific CD4+ T cells by intracellular cytokine staining in 58 of 142 (41%) people. In those with reactive CD4+ T cells, the median percentage of CD4+ T cells expressing interleukin 2 was 88%, and the median co-expression of interferon gamma or tumor necrosis factor alpha (TNFalpha), or both, was 72%. We noted HIV-specific CD8+ T cells (range 0.4-1.0%) in 117 of 160 (73%) participants, expressing predominantly either interferon gamma alone or with TNFalpha. Vaccine-induced HIV-specific immunity, including response rate, magnitude, and cytokine profile, did not differ between vaccinated male cases (before infection) and non-cases. Ad5-specific T cells were lower in cases than in non-cases in several subgroup analyses. The percentage of circulating Ki-67+BcL-2(lo)/CCR5+CD4+ T cells did not differ between cases and non-cases. INTERPRETATION Consistent with previous trials, the MRKAd5 HIV-1 gag/pol/nef vaccine was highly immunogenic for inducing HIV-specific CD8+ T cells. Our findings suggest that future candidate vaccines have to elicit responses that either exceed in magnitude or differ in breadth or function from those recorded in this trial.


Journal of Experimental Medicine | 2008

Yellow fever vaccine induces integrated multilineage and polyfunctional immune responses

Denis Gaucher; René Therrien; Nadia Kettaf; Bastian R. Angermann; Geneviève Boucher; Abdelali Filali-Mouhim; Janice M. Moser; Riyaz Mehta; Donald R. Drake; Erika Castro; Rama Akondy; Aline Rinfret; Bader Yassine-Diab; Elias A. Said; Younes Chouikh; Mark J. Cameron; Robert Clum; David J. Kelvin; Roland Somogyi; Robert S. Balderas; Peter Wilkinson; Giuseppe Pantaleo; Jim Tartaglia; Elias K. Haddad; Rafick Pierre Sekaly

Correlates of immune-mediated protection to most viral and cancer vaccines are still unknown. This impedes the development of novel vaccines to incurable diseases such as HIV and cancer. In this study, we have used functional genomics and polychromatic flow cytometry to define the signature of the immune response to the yellow fever (YF) vaccine 17D (YF17D) in a cohort of 40 volunteers followed for up to 1 yr after vaccination. We show that immunization with YF17D leads to an integrated immune response that includes several effector arms of innate immunity, including complement, the inflammasome, and interferons, as well as adaptive immunity as shown by an early T cell response followed by a brisk and variable B cell response. Development of these responses is preceded, as demonstrated in three independent vaccination trials and in a novel in vitro system of primary immune responses (modular immune in vitro construct [MIMIC] system), by the coordinated up-regulation of transcripts for specific transcription factors, including STAT1, IRF7, and ETS2, which are upstream of the different effector arms of the immune response. These results clearly show that the immune response to a strong vaccine is preceded by coordinated induction of master transcription factors that lead to the development of a broad, polyfunctional, and persistent immune response that integrates all effector cells of the immune system.


Immunity | 2008

Human effector and memory CD8+ T cell responses to smallpox and yellow fever vaccines.

Joseph D. Miller; Robbert G. van der Most; Rama Akondy; John Glidewell; Sophia Albott; David Masopust; Kaja Murali-Krishna; Patryce L. Mahar; Srilatha Edupuganti; Susan Lalor; Stephanie Germon; Carlos del Rio; Mark J. Mulligan; Silvija I. Staprans; John D. Altman; Mark B. Feinberg; Rafi Ahmed

To explore the human T cell response to acute viral infection, we performed a longitudinal analysis of CD8(+) T cells responding to the live yellow fever virus and smallpox vaccines--two highly successful human vaccines. Our results show that both vaccines generated a brisk primary effector CD8(+) T cell response of substantial magnitude that could be readily quantitated with a simple set of four phenotypic markers. Secondly, the vaccine-induced T cell response was highly specific with minimal bystander effects. Thirdly, virus-specific CD8(+) T cells passed through an obligate effector phase, contracted more than 90% and gradually differentiated into long-lived memory cells. Finally, these memory cells were highly functional and underwent a memory differentiation program distinct from that described for human CD8(+) T cells specific for persistent viruses. These results provide a benchmark for CD8(+) T cell responses induced by two of the most effective vaccines ever developed.


Journal of Experimental Medicine | 2010

Dynamic T cell migration program provides resident memory within intestinal epithelium

David Masopust; Daniel Choo; Vaiva Vezys; E. John Wherry; Jaikumar Duraiswamy; Rama Akondy; Jun Wang; Kerry A. Casey; Daniel L. Barber; Kim S. Kawamura; Kathryn A. Fraser; Richard J. Webby; Volker Brinkmann; Eugene C. Butcher; Kenneth A. Newell; Rafi Ahmed

Migration to intestinal mucosa putatively depends on local activation because gastrointestinal lymphoid tissue induces expression of intestinal homing molecules, whereas skin-draining lymph nodes do not. This paradigm is difficult to reconcile with reports of intestinal T cell responses after alternative routes of immunization. We reconcile this discrepancy by demonstrating that activation within spleen results in intermediate induction of homing potential to the intestinal mucosa. We further demonstrate that memory T cells within small intestine epithelium do not routinely recirculate with memory T cells in other tissues, and we provide evidence that homing is similarly dynamic in humans after subcutaneous live yellow fever vaccine immunization. These data explain why systemic immunization routes induce local cell-mediated immunity within the intestine and indicate that this tissue must be seeded with memory T cell precursors shortly after activation.


Journal of Immunology | 2009

The Yellow Fever Virus Vaccine Induces a Broad and Polyfunctional Human Memory CD8+ T Cell Response

Rama Akondy; Nathan Monson; Joseph D. Miller; Srilatha Edupuganti; Dirk E. Teuwen; Hong Wu; Farah Quyyumi; Seema Garg; John D. Altman; Carlos del Rio; Harry L. Keyserling; Alexander Ploss; Charles M. Rice; Walter A. Orenstein; Mark J. Mulligan; Rafi Ahmed

The live yellow fever vaccine (YF-17D) offers a unique opportunity to study memory CD8+ T cell differentiation in humans following an acute viral infection. We have performed a comprehensive analysis of the virus-specific CD8+ T cell response using overlapping peptides spanning the entire viral genome. Our results showed that the YF-17D vaccine induces a broad CD8+ T cell response targeting several epitopes within each viral protein. We identified a dominant HLA-A2-restricted epitope in the NS4B protein and used tetramers specific for this epitope to track the CD8+ T cell response over a 2 year period. This longitudinal analysis showed the following. 1) Memory CD8+ T cells appear to pass through an effector phase and then gradually down-regulate expression of activation markers and effector molecules. 2) This effector phase was characterized by down-regulation of CD127, Bcl-2, CCR7, and CD45RA and was followed by a substantial contraction resulting in a pool of memory T cells that re-expressed CD127, Bcl-2, and CD45RA. 3) These memory cells were polyfunctional in terms of degranulation and production of the cytokines IFN-γ, TNF-α, IL-2, and MIP-1β. 4) The YF-17D-specific memory CD8+ T cells had a phenotype (CCR7−CD45RA+) that is typically associated with terminally differentiated cells with limited proliferative capacity (TEMRA). However, these cells exhibited robust proliferative potential showing that expression of CD45RA may not always associate with terminal differentiation and, in fact, may be an indicator of highly functional memory CD8+ T cells generated after acute viral infections.


Journal of Virology | 2012

Rapid and Massive Virus-Specific Plasmablast Responses during Acute Dengue Virus Infection in Humans

Jens Wrammert; Nattawat Onlamoon; Rama Akondy; Guey Chuen Perng; Korakot Polsrila; Anmol Chandele; Marcin Kwissa; Bali Pulendran; Patrick C. Wilson; Orasri Wittawatmongkol; Sutee Yoksan; Nasikarn Angkasekwinai; Kovit Pattanapanyasat; Kulkanya Chokephaibulkit; Rafi Ahmed

ABSTRACT Humoral immune responses are thought to play a major role in dengue virus-induced immunopathology; however, little is known about the plasmablasts producing these antibodies during an ongoing infection. Herein we present an analysis of plasmablast responses in patients with acute dengue virus infection. We found very potent plasmablast responses that often increased more than 1,000-fold over the baseline levels in healthy volunteers. In many patients, these responses made up as much 30% of the peripheral lymphocyte population. These responses were largely dengue virus specific and almost entirely made up of IgG-secreting cells, and plasmablasts reached very high numbers at a time after fever onset that generally coincided with the window where the most serious dengue virus-induced pathology is observed. The presence of these large, rapid, and virus-specific plasmablast responses raises the question as to whether these cells might have a role in dengue immunopathology during the ongoing infection. These findings clearly illustrate the need for a detailed understanding of the repertoire and specificity of the antibodies that these plasmablasts produce.


The Journal of Infectious Diseases | 2008

Case of Yellow Fever Vaccine-associated Viscerotropic Disease with Prolonged Viremia, Robust Adaptive Immune Responses, and Polymorphisms in CCR5 and RANTES Genes

Bali Pulendran; Joseph D. Miller; Troy D. Querec; Rama Akondy; Nelson B. Moseley; Oskar Laur; John Glidewell; Nathan Monson; Tuofu Zhu; Haiying Zhu; Sylvija Staprans; David Lee; Margo A. Brinton; Andrey A. Perelygin; Claudia J. Vellozzi; Philip Sigmund Brachman; Susan Lalor; Dirk E. Teuwen; Rachel B. Eidex; Martin S. Cetron; Frances Priddy; Carlos del Rio; John D. Altman; Rafi Ahmed

BACKGROUND The live attenuated yellow fever vaccine 17D (YF-17D) is one of the most effective vaccines. Despite its excellent safety record, some cases of viscerotropic adverse events develop, which are sometimes fatal. The mechanisms underlying such events remain a mystery. Here, we present an analysis of the immunologic and genetic factors driving disease in a 64-year-old male who developed viscerotropic symptoms. METHODS We obtained clinical, serologic, virologic, immunologic and genetic data on this case patient. RESULTS Viral RNA was detected in the blood 33 days after vaccination, in contrast to the expected clearance of virus by day 7 after vaccination in healthy vaccinees. Vaccination induced robust antigen-specific T and B cell responses, which suggested that persistent virus was not due to adaptive immunity of suboptimal magnitude. The genes encoding OAS1, OAS2, TLR3, and DC-SIGN, which mediate antiviral innate immunity, were wild type. However, there were heterozygous genetic polymorphisms in chemokine receptor CCR5, and its ligand RANTES, which influence the migration of effector T cells and CD14+CD16bright monocytes to tissues. Consistent with this, there was a 200-fold increase in the number of CD14+CD16bright monocytes in the blood during viremia and even several months after virus clearance. CONCLUSION In this patient, viscerotropic disease was not due to the impaired magnitude of adaptive immunity but instead to anomalies in the innate immune system and a possible disruption of the CCR5-RANTES axis.


Proceedings of the National Academy of Sciences of the United States of America | 2015

Human Ebola virus infection results in substantial immune activation

Anita K. McElroy; Rama Akondy; Carl W. Davis; Ali H. Ellebedy; Aneesh K. Mehta; Colleen S. Kraft; G. Marshall Lyon; Bruce S. Ribner; Jay B. Varkey; John Sidney; Alessandro Sette; Shelley Campbell; Ute Ströher; Inger K. Damon; Stuart T. Nichol; Christina F. Spiropoulou; Rafi Ahmed

Significance In 2014, Ebola virus became a household term. The ongoing outbreak in West Africa is the largest Ebola virus outbreak ever recorded, with over 20,000 cases and over 8,000 deaths to date. Very little is known about the human cellular immune response to Ebola virus infection, and this lack of knowledge has hindered development of effective therapies and vaccines. In this study, we characterize the human immune response to Ebola virus infection in four patients. We define the kinetics of T- and B-cell activation, and determine which viral proteins are targets of the Ebola virus-specific T-cell response in humans. Four Ebola patients received care at Emory University Hospital, presenting a unique opportunity to examine the cellular immune responses during acute Ebola virus infection. We found striking activation of both B and T cells in all four patients. Plasmablast frequencies were 10–50% of B cells, compared with less than 1% in healthy individuals. Many of these proliferating plasmablasts were IgG-positive, and this finding coincided with the presence of Ebola virus-specific IgG in the serum. Activated CD4 T cells ranged from 5 to 30%, compared with 1–2% in healthy controls. The most pronounced responses were seen in CD8 T cells, with over 50% of the CD8 T cells expressing markers of activation and proliferation. Taken together, these results suggest that all four patients developed robust immune responses during the acute phase of Ebola virus infection, a finding that would not have been predicted based on our current assumptions about the highly immunosuppressive nature of Ebola virus. Also, quite surprisingly, we found sustained immune activation after the virus was cleared from the plasma, observed most strikingly in the persistence of activated CD8 T cells, even 1 mo after the patients’ discharge from the hospital. These results suggest continued antigen stimulation after resolution of the disease. From these convalescent time points, we identified CD4 and CD8 T-cell responses to several Ebola virus proteins, most notably the viral nucleoprotein. Knowledge of the viral proteins targeted by T cells during natural infection should be useful in designing vaccines against Ebola virus.


Journal of Immunology | 2013

Cutting Edge: Prolonged Exposure to HIV Reinforces a Poised Epigenetic Program for PD-1 Expression in Virus-Specific CD8 T Cells

Ben Youngblood; Alessandra Noto; Filippos Porichis; Rama Akondy; Zaza M. Ndhlovu; James W. Austin; Rebeka Bordi; Francesco A. Procopio; Toshiyuki Miura; Todd M. Allen; John Sidney; Alessandro Sette; Bruce D. Walker; Rafi Ahmed; Jeremy M. Boss; Rafick-Pierre Sekaly; Daniel E. Kaufmann

Ag-specific CD8 T cells play a critical role in controlling HIV infection but eventually lose antiviral functions in part because of expression and signaling through the inhibitory programmed death-1 (PD-1) receptor. To better understand the impact of prolonged TCR ligation on regulation of PD-1 expression in HIV-specific CD8 T cells, we investigated the capacity of virus-specific CD8 T cells to modify the PD-1 epigenetic program after reduction in viral load. We observed that the transcriptional regulatory region was unmethylated in the PD-1hi HIV-specific CD8 T cells, whereas it remained methylated in donor-matched naive cells at acute and chronic stages of infection. Surprisingly, the PD-1 promoter remained unmethylated in HIV-specific CD8 T cells from subjects with a viral load controlled by antiviral therapy for >2 y or from elite controllers. Together, these data demonstrate that the epigenetic program at the PD-1 locus becomes fixed after prolonged exposure to HIV virus.


Proceedings of the National Academy of Sciences of the United States of America | 2016

CXCL13 is a plasma biomarker of germinal center activity

Colin Havenar-Daughton; Madelene Lindqvist; Antje Heit; Jennifer E. Wu; Samantha M. Reiss; Kayla Kendric; Simon Bélanger; Sudhir Pai Kasturi; Elise Landais; Rama Akondy; Helen M. McGuire; Marcella Bothwell; Parsia A. Vagefi; Eileen Scully; Georgia D. Tomaras; Mark M. Davis; Pascal Poignard; Rafi Ahmed; Bruce D. Walker; Bali Pulendran; M. Juliana McElrath; Daniel E. Kaufmann; Shane Crotty

Significance A major challenge for vaccine science is that there is no way to measure germinal center activity in humans. This challenge is particularly acute for human clinical trials of candidate vaccines (and most nonhuman primate studies of candidate vaccines), because germinal centers are the engines of Ab affinity maturation, and generation of highly affinity-matured Ab responses is the goal of all Ab-eliciting vaccines. Here, we report that we have identified the chemokine CXCL13 [chemokine (C-X-C motif) ligand 13] as a biomarker of germinal center activity. We show explicit relationships between plasma CXCL13 concentrations and germinal center frequencies in lymph nodes in a series of different conditions, including licensed and experimental vaccines, and in humans, nonhuman primates, and mice. Significantly higher levels of plasma CXCL13 [chemokine (C-X-C motif) ligand 13] were associated with the generation of broadly neutralizing antibodies (bnAbs) against HIV in a large longitudinal cohort of HIV-infected individuals. Germinal centers (GCs) perform the remarkable task of optimizing B-cell Ab responses. GCs are required for almost all B-cell receptor affinity maturation and will be a critical parameter to monitor if HIV bnAbs are to be induced by vaccination. However, lymphoid tissue is rarely available from immunized humans, making the monitoring of GC activity by direct assessment of GC B cells and germinal center CD4+ T follicular helper (GC Tfh) cells problematic. The CXCL13–CXCR5 [chemokine (C-X-C motif) receptor 5] chemokine axis plays a central role in organizing both B-cell follicles and GCs. Because GC Tfh cells can produce CXCL13, we explored the potential use of CXCL13 as a blood biomarker to indicate GC activity. In a series of studies, we found that plasma CXCL13 levels correlated with GC activity in draining lymph nodes of immunized mice, immunized macaques, and HIV-infected humans. Furthermore, plasma CXCL13 levels in immunized humans correlated with the magnitude of Ab responses and the frequency of ICOS+ (inducible T-cell costimulator) Tfh-like cells in blood. Together, these findings support the potential use of CXCL13 as a plasma biomarker of GC activity in human vaccine trials and other clinical settings.

Collaboration


Dive into the Rama Akondy's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Alessandro Sette

La Jolla Institute for Allergy and Immunology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Bali Pulendran

Yerkes National Primate Research Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge