Network


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

Hotspot


Dive into the research topics where Adam R. Hersperger is active.

Publication


Featured researches published by Adam R. Hersperger.


PLOS Pathogens | 2010

Perforin Expression Directly Ex Vivo by HIV-Specific CD8+ T-Cells Is a Correlate of HIV Elite Control

Adam R. Hersperger; Florencia Pereyra; Martha Nason; Korey Demers; Prameet M. Sheth; Lucy Y. Shin; Colin Kovacs; Benigno Rodriguez; Scott F. Sieg; Leia Teixeira-Johnson; Debbie Gudonis; Paul A. Goepfert; Michael M. Lederman; Ian Frank; George Makedonas; Rupert Kaul; Bruce D. Walker; Michael R. Betts

Many immune correlates of CD8+ T-cell-mediated control of HIV replication, including polyfunctionality, proliferative ability, and inhibitory receptor expression, have been discovered. However, no functional correlates using ex vivo cells have been identified with the known ability to cause the direct elimination of HIV-infected cells. We have recently discovered the ability of human CD8+ T-cells to rapidly upregulate perforin—an essential molecule for cell-mediated cytotoxicity—following antigen-specific stimulation. Here, we examined perforin expression capability in a large cross-sectional cohort of chronically HIV-infected individuals with varying levels of viral load: elite controllers (n = 35), viremic controllers (n = 29), chronic progressors (n = 27), and viremic nonprogressors (n = 6). Using polychromatic flow cytometry and standard intracellular cytokine staining assays, we measured perforin upregulation, cytokine production, and degranulation following stimulation with overlapping peptide pools encompassing all proteins of HIV. We observed that HIV-specific CD8+ T-cells from elite controllers consistently display an enhanced ability to express perforin directly ex vivo compared to all other groups. This ability is not restricted to protective HLA-B haplotypes, does not require proliferation or the addition of exogenous factors, is not restored by HAART, and primarily originates from effector CD8+ T-cells with otherwise limited functional capability. Notably, we found an inverse relationship between HIV-specific perforin expression and viral load. Thus, the capability of HIV-specific CD8+ T-cells to rapidly express perforin defines a novel correlate of control in HIV infection.


Blood | 2011

Increased HIV-specific CD8 + T-cell cytotoxic potential in HIV elite controllers is associated with T-bet expression

Adam R. Hersperger; Jeffrey N. Martin; Lucy Y. Shin; Prameet M. Sheth; Colin Kovacs; Gabriela L. Cosma; George Makedonas; Florencia Pereyra; Bruce D. Walker; Rupert Kaul; Steven G. Deeks; Michael R. Betts

Recent data suggest that CD8+ T-cell effector activity is an important component in the control of HIV replication in elite controllers (ECs). One critical element of CD8+ T-cell effector function and differentiation is the T-box transcription factor T-bet. In the present study, we assessed T-bet expression, together with the effector proteins perforin, granzyme A (Grz A), granzyme B (Grz B), and granulysin, in HIV-specific CD8+ T cells from ECs (n = 20), chronically infected progressors (CPs; n = 18), and highly active antiretroviral therapy (HAART)-suppressed individuals (n = 19). Compared with the other cohort groups, HIV-specific CD8+ T cells among ECs demonstrated a superior ability to express perforin and Grz B, but with no detectable difference in the levels of Grz A or granulysin. We also observed higher levels of T-bet in HIV-specific CD8+ T cells from ECs, with an ensuing positive correlation between T-bet and levels of both perforin and Grz B. Moreover, HIV-specific CD8+ T cells in ECs up-regulated T-bet to a greater extent than CPs after in vitro expansion, with concomitant up-regulation of perforin and Grz B. These results suggest that T-bet may play an important role in driving effector function, and its modulation may lead to enhanced effector activity against HIV.


PLOS Pathogens | 2010

Perforin and IL-2 Upregulation Define Qualitative Differences among Highly Functional Virus-Specific Human CD8(+) T Cells

George Makedonas; Natalie A. Hutnick; Danielle Haney; Alexandra C. Amick; Jay Gardner; Gabriela L. Cosma; Adam R. Hersperger; Douglas V. Dolfi; E. John Wherry; Guido Ferrari; Michael R. Betts

The prevailing paradigm of T lymphocyte control of viral replication is that the protective capacity of virus-specific CD8+ T cells is directly proportional to the number of functions they can perform, with IL-2 production capacity considered critical. Having recently defined rapid perforin upregulation as a novel effector function of antigen-specific CD8+ T cells, here we sought to determine whether new perforin production is a component of polyfunctional CD8+ T cell responses that contributes to the control of several human viral infections: cytomegalovirus (CMV), Epstein-Barr virus (EBV), influenza (flu), and adenovirus (Ad). We stimulated normal human donor PBMC with synthetic peptides whose amino acid sequences correspond to defined CTL epitopes in the aforementioned viruses, and then used polychromatic flow cytometry to measure the functional capacity and the phenotype of the responding CD8+ T cells. While EBV and flu-specific CD8+ T cells rarely upregulate perforin, CMV-specific cells often do and Ad stimulates an exceptionally strong perforin response. The differential propensity of CD8+ T cells to produce either IL-2 or perforin is in part related to levels of CD28 and the transcription factor T-bet, as CD8+ T cells that rapidly upregulate perforin harbor high levels of T-bet and those producing IL-2 express high amounts of CD28. Thus, “polyfunctional” profiling of antigen-specific CD8+ T cells must not be limited to simply the number of functions the cell can perform, or one particular memory phenotype, but should actually define which combinations of memory markers and functions are relevant in each pathogenic context.


Current Opinion in Hiv and Aids | 2011

Qualitative features of the Hiv-specific Cd8+ T-cell response associated with immunologic control

Adam R. Hersperger; Stephen A. Migueles; Michael R. Betts; Mark Connors

Purpose of reviewOver the past 2 years, a clearer picture has emerged regarding the properties of HIV-specific CD8+ T cells associated with immunologic control of HIV replication. These properties represent a potential mechanism by which rare patients might control HIV replication in the absence of antiretroviral therapy. This review addresses the background and recent findings that have lead to our current understanding of these mechanism(s). Recent findingsPatients with immunologic control of HIV are not distinguished by targeted specificities, or greater numbers or breadth of their HIV-specific CD8+ T-cell response. For this reason, recent work has focused greater attention on qualitative features of this response. The qualitative features most closely associated with immunologic control of HIV are related to the granule-exocytosis-mediated elimination of HIV-infected CD4+ T cells. The ability of HIV-specific CD8+ T cells to increase their contents of proteins known to mediate cytotoxicity, such as granzyme B and perforin, appears to be a critical means by which HIV-specific cytotoxic capacity is regulated. SummaryInvestigation from multiple groups has now focused upon HIV-specific CD8+ T-cell granule-exocytosis-mediated cytotoxicity as a correlate of immunologic control of HIV. In the near future, a more detailed understanding of the qualities associated with immunologic control may provide critical insights regarding the necessary features of a response that should be stimulated by immunotherapies or T-cell-based vaccines.


Journal of Immunology | 2009

Rapid Up-Regulation and Granule-Independent Transport of Perforin to the Immunological Synapse Define a Novel Mechanism of Antigen-Specific CD8+ T Cell Cytotoxic Activity

George Makedonas; Pinaki P. Banerjee; Rahul Pandey; Adam R. Hersperger; Keri B. Sanborn; Gareth A. D. Hardy; Jordan S. Orange; Michael R. Betts

CTL are endowed with the ability to eliminate pathogens through perforin-mediated cytotoxic activity. The mechanism for perforin-mediated Ag-specific killing has been solely attributed to cytotoxic granule exocytosis from activated CD8+ T cells. In this study, we redefine this mechanism, demonstrating that virus-specific CD8+ T cells rapidly up-regulate perforin in response to stimulation temporally with IFN-γ and CD107a expression. Following Ag-specific activation, newly synthesized perforin rapidly appears at the immunological synapse, both in association with and independent of cytotoxic granules, where it functions to promote cytotoxicity. Our work suggests a novel mechanism of CTL cytotoxicity and identifies a novel correlate of CD8+ T cell-mediated immunity.


Cytometry Part A | 2008

Flow cytometric detection of perforin upregulation in human CD8 T cells

Adam R. Hersperger; George Makedonas; Michael R. Betts

Perforin and granzymes work synergistically to induce apoptosis in target cells recognized by cytotoxic T lymphocytes. While perforin is readily detectable by flow cytometry in resting CD8 T cells, upregulation of perforin in activated cells is thought to require proliferation. However, perforin undergoes numerous conformational changes during its maturation, which may affect the ability of conventional antibodies to recognize newly synthesized perforin. Polychromatic flow cytometry was used to detect perforin and cytokine production following stimulation of ex vivo human CD8 T cells. Two different anti‐perforin antibodies, clones B‐D48 and δG9, were used to discriminate various forms of perforin after cellular activation. We provide evidence for the rapid upregulation of perforin protein, which may contribute to the ability of CD8 T cells to kill multiple targets over time. The δG9 clone recognizes the granule‐associated conformation of perforin, while the B‐D48 clone is able to detect perforin in multiple forms. Finally, we show there is variability in the ability of CD8 T cells to upregulate perforin. Human CD8 T cells are capable of new perforin production immediately following activation. This work defines a novel flow cytometric procedure that can be used to more completely assess the cytotoxic capacity of human CD8 T cells.


Proceedings of the National Academy of Sciences of the United States of America | 2012

Perforin-dependent CD4+ T-cell cytotoxicity contributes to control a murine poxvirus infection

Min Fang; Nicholas A. Siciliano; Adam R. Hersperger; Felicia Roscoe; Angela Hu; Xueying Ma; Ahamed R. Shamsedeen; Laurence C. Eisenlohr; Luis J. Sigal

CD4+ T cells are generally regarded as helpers and regulators of the immune response. Although cytolytic CD4+ T cells have been described, whether those generated during the course of a viral infection play a role in virus control remains unknown. Here we show that during acute infection with ectromelia virus, the mouse homolog of the human virus of smallpox, large numbers of CD4+ T cells in the draining lymph node and liver of resistant mice have a cytotoxic phenotype. We also show that these cells kill targets in vivo in a perforin-dependent manner and that mice with specific deficiency of perforin in CD4+ T cells have impaired virus control. Thus, perforin-dependent CD4+ T-cell killing of infected cells is an important mechanism of antiviral defense.


PLOS ONE | 2015

Ectopic Expression of Vaccinia Virus E3 and K3 Cannot Rescue Ectromelia Virus Replication in Rabbit RK13 Cells

Erin S. Hand; Sherry L. Haller; Chen Peng; Stefan Rothenburg; Adam R. Hersperger

As a group, poxviruses have been shown to infect a wide variety of animal species. However, there is individual variability in the range of species able to be productively infected. In this study, we observed that ectromelia virus (ECTV) does not replicate efficiently in cultured rabbit RK13 cells. Conversely, vaccinia virus (VACV) replicates well in these cells. Upon infection of RK13 cells, the replication cycle of ECTV is abortive in nature, resulting in a greatly reduced ability to spread among cells in culture. We observed ample levels of early gene expression but reduced detection of virus factories and severely blunted production of enveloped virus at the cell surface. This work focused on two important host range genes, named E3L and K3L, in VACV. Both VACV and ECTV express a functional protein product from the E3L gene, but only VACV contains an intact K3L gene. To better understand the discrepancy in replication capacity of these viruses, we examined the ability of ECTV to replicate in wild-type RK13 cells compared to cells that constitutively express E3 and K3 from VACV. The role these proteins play in the ability of VACV to replicate in RK13 cells was also analyzed to determine their individual contribution to viral replication and PKR activation. Since E3L and K3L are two relevant host range genes, we hypothesized that expression of one or both of them may have a positive impact on the ability of ECTV to replicate in RK13 cells. Using various methods to assess virus growth, we did not detect any significant differences with respect to the replication of ECTV between wild-type RK13 compared to versions of this cell line that stably expressed VACV E3 alone or in combination with K3. Therefore, there remain unanswered questions related to the factors that limit the host range of ECTV.


Journal of Virology | 2014

Epithelial immunization induces polyfunctional CD8+ T-cells and optimal mousepox protection

Adam R. Hersperger; Nicholas A. Siciliano; Brian C. DeHaven; Adam E. Snook; Laurence C. Eisenlohr

ABSTRACT We assessed several routes of immunization with vaccinia virus (VACV) in protecting mice against ectromelia virus (ECTV). By a wide margin, skin scarification provided the greatest protection. Humoral immunity and resident-memory T cells notwithstanding, several approaches revealed that circulating, memory CD8+ T cells primed via scarification were functionally superior and conferred enhanced virus control. Immunization via the epithelial route warrants further investigation, as it may also provide enhanced defense against other infectious agents.


Journal of Virology | 2014

Impact of Distinct Poxvirus Infections on the Specificities and Functionalities of CD4+ T Cell Responses

Nicholas A. Siciliano; Adam R. Hersperger; Aimee M. Lacuanan; Ren-Huan Xu; John Sidney; Alessandro Sette; Luis J. Sigal; Laurence C. Eisenlohr

ABSTRACT The factors that determine CD4+ T cell (TCD4+ ) specificities, functional capacity, and memory persistence in response to complex pathogens remain unclear. We explored these parameters in the C57BL/6 mouse through comparison of two highly related (>92% homology) poxviruses: ectromelia virus (ECTV), a natural mouse pathogen, and vaccinia virus (VACV), a heterologous virus that nevertheless elicits potent immune responses. In addition to elucidating several previously unidentified major histocompatibility complex class II (MHC-II)-restricted epitopes, we observed many qualitative and quantitative differences between the TCD4+ repertoires, including responses not elicited by VACV despite complete sequence conservation. In addition, we observed functional heterogeneity between ECTV- and VACV-specific TCD4+ at both a global and individual epitope level, particularly greater expression of the cytolytic marker CD107a from TCD4+ following ECTV infection. Most striking were differences during the late memory phase where, in contrast to ECTV, VACV infection failed to elicit measurable epitope-specific TCD4+ as determined by intracellular cytokine staining. These findings illustrate the strong influence of epitope-extrinsic factors on TCD4+ responses and memory. IMPORTANCE Much of our understanding concerning host-pathogen relationships in the context of poxvirus infections stems from studies of VACV in mice. However, VACV is not a natural mouse pathogen, and therefore, the relevance of results obtained using this model may be limited. Here, we explored the MHC class II-restricted TCD4+ repertoire induced by mousepox (ECTV) infection and the functional profile of the responding epitope-specific TCD4+ , comparing these results to those induced by VACV infection under matched conditions. Despite a high degree of homology between the two viruses, we observed distinct specificity and functional profiles of TCD4+ responses at both acute and memory time points, with VACV-specific TCD4+ memory being notably compromised. These data offer insight into the impact of epitope-extrinsic factors on the resulting TCD4+ responses.

Collaboration


Dive into the Adam R. Hersperger's collaboration.

Top Co-Authors

Avatar

George Makedonas

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Michael R. Betts

University of Pennsylvania

View shared research outputs
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

Benigno Rodriguez

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge