Network


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

Hotspot


Dive into the research topics where Ben Youngblood is active.

Publication


Featured researches published by Ben Youngblood.


Journal of Clinical Investigation | 2013

PD-L1 blockade synergizes with IL-2 therapy in reinvigorating exhausted T cells

Erin E. West; Hyun Tak Jin; Ata Ur Rasheed; Pablo Penaloza-MacMaster; Sang Jun Ha; Wendy G. Tan; Ben Youngblood; Gordon J. Freeman; Kendall A. Smith; Rafi Ahmed

The inhibitory receptor programmed cell death 1 (PD-1) plays a major role in functional exhaustion of T cells during chronic infections and cancer, and recent clinical data suggest that blockade of the PD-1 pathway is an effective immunotherapy in treating certain cancers. Thus, it is important to define combinatorial approaches that increase the efficacy of PD-1 blockade. To address this issue, we examined the effect of IL-2 and PD-1 ligand 1 (PD-L1) blockade in the mouse model of chronic lymphocytic choriomeningitis virus (LCMV) infection. We found that low-dose IL-2 administration alone enhanced CD8+ T cell responses in chronically infected mice. IL-2 treatment also decreased inhibitory receptor levels on virus-specific CD8+ T cells and increased expression of CD127 and CD44, resulting in a phenotype resembling that of memory T cells. Surprisingly, IL-2 therapy had only a minimal effect on reducing viral load. However, combining IL-2 treatment with blockade of the PD-1 inhibitory pathway had striking synergistic effects in enhancing virus-specific CD8+ T cell responses and decreasing viral load. Interestingly, this reduction in viral load occurred despite increased numbers of Tregs. These results suggest that combined IL-2 therapy and PD-L1 blockade merits consideration as a regimen for treating human chronic infections and cancer.


Immunology | 2013

T‐cell memory differentiation: insights from transcriptional signatures and epigenetics

Ben Youngblood; J. Scott Hale; Rafi Ahmed

A critical component of vaccine design is to generate and maintain antigen‐specific memory lymphocytes of sufficient quantity and quality to give the host life‐long protection against re‐infection. Therefore, it is important to understand how memory T cells acquire the ability for self‐renewal while retaining a potential for heightened recall of effector functions. During acute viral infection or following vaccination, antigen‐specific T cells undergo extensive phenotypic and functional changes during differentiation to the effector and memory phases of the immune response. The changes in cell phenotype that accompany memory T‐cell differentiation are predominantly mediated through acquired transcriptional regulatory mechanisms, in part achieved through epigenetic modifications of DNA and histones. Here we review our current understanding of epigenetic mechanisms regulating the off‐on‐off expression of CD8 and CD4 T‐cell effector molecules at naive, effector and memory stages of differentiation, respectively, and how covalent modifications to the genome may serve as a mechanism to preserve ‘poised’ transcriptional states in homeostatically dividing memory cells. We discuss the potential of such mechanisms to control genes that undergo on‐off‐on patterns of expression including homing and pro‐survival genes, and the implications on the development of effector‐memory and central‐memory T‐cell differentiation. Lastly, we review recent studies demonstrating epigenetic modifications as a mechanism for the progressive loss of transcriptional adaptation in antigen‐specific T cells that undergo sustained high levels of T‐cell receptor signalling.


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.


Nature | 2017

Origin and differentiation of human memory CD8 T cells after vaccination

Rama Akondy; Mark Fitch; Srilatha Edupuganti; Shu Yang; Haydn T. Kissick; Kelvin Li; Ben Youngblood; Hossam A. Abdelsamed; Donald J. McGuire; Kristen W. Cohen; Gabriela Alexe; Shashi Nagar; Megan McCausland; Satish Gupta; Pramila Tata; W. Nicholas Haining; M. Juliana McElrath; David D. Zhang; Bin Hu; William J. Greenleaf; Jörg J. Goronzy; Mark Mulligan; Marc K. Hellerstein; Rafi Ahmed

The differentiation of human memory CD8 T cells is not well understood. Here we address this issue using the live yellow fever virus (YFV) vaccine, which induces long-term immunity in humans. We used in vivo deuterium labelling to mark CD8 T cells that proliferated in response to the virus and then assessed cellular turnover and longevity by quantifying deuterium dilution kinetics in YFV-specific CD8 T cells using mass spectrometry. This longitudinal analysis showed that the memory pool originates from CD8 T cells that divided extensively during the first two weeks after infection and is maintained by quiescent cells that divide less than once every year (doubling time of over 450 days). Although these long-lived YFV-specific memory CD8 T cells did not express effector molecules, their epigenetic landscape resembled that of effector CD8 T cells. This open chromatin profile at effector genes was maintained in memory CD8 T cells isolated even a decade after vaccination, indicating that these cells retain an epigenetic fingerprint of their effector history and remain poised to respond rapidly upon re-exposure to the pathogen.


Nature | 2017

Effector CD8 T cells dedifferentiate into long-lived memory cells

Ben Youngblood; J. Scott Hale; Haydn T. Kissick; Eunseon Ahn; Xiaojin Xu; Andreas Wieland; Koichi Araki; Erin E. West; Hazem E. Ghoneim; Yiping Fan; Pranay Dogra; Carl W. Davis; Bogumila T. Konieczny; Rustom Antia; Xiaodong Cheng; Rafi Ahmed

Memory CD8 T cells that circulate in the blood and are present in lymphoid organs are an essential component of long-lived T cell immunity. These memory CD8 T cells remain poised to rapidly elaborate effector functions upon re-exposure to pathogens, but also have many properties in common with naive cells, including pluripotency and the ability to migrate to the lymph nodes and spleen. Thus, memory cells embody features of both naive and effector cells, fuelling a long-standing debate centred on whether memory T cells develop from effector cells or directly from naive cells. Here we show that long-lived memory CD8 T cells are derived from a subset of effector T cells through a process of dedifferentiation. To assess the developmental origin of memory CD8 T cells, we investigated changes in DNA methylation programming at naive and effector cell-associated genes in virus-specific CD8 T cells during acute lymphocytic choriomeningitis virus infection in mice. Methylation profiling of terminal effector versus memory-precursor CD8 T cell subsets showed that, rather than retaining a naive epigenetic state, the subset of cells that gives rise to memory cells acquired de novo DNA methylation programs at naive-associated genes and became demethylated at the loci of classically defined effector molecules. Conditional deletion of the de novo methyltransferase Dnmt3a at an early stage of effector differentiation resulted in reduced methylation and faster re-expression of naive-associated genes, thereby accelerating the development of memory cells. Longitudinal phenotypic and epigenetic characterization of the memory-precursor effector subset of virus-specific CD8 T cells transferred into antigen-free mice revealed that differentiation to memory cells was coupled to erasure of de novo methylation programs and re-expression of naive-associated genes. Thus, epigenetic repression of naive-associated genes in effector CD8 T cells can be reversed in cells that develop into long-lived memory CD8 T cells while key effector genes remain demethylated, demonstrating that memory T cells arise from a subset of fate-permissive effector T cells.


The Journal of Infectious Diseases | 2015

Epigenetic Repression of Interleukin 2 Expression in Senescent CD4+ T Cells During Chronic HIV Type 1 Infection

Kaori Nakayama-Hosoya; Takaomi Ishida; Ben Youngblood; Hitomi Nakamura; Noriaki Hosoya; Michiko Koga; Tomohiko Koibuchi; Aikichi Iwamoto; Ai Kawana-Tachikawa

The molecular mechanisms for IL2 gene-specific dysregulation during chronic human immunodeficiency virus type 1 (HIV-1) infection are unknown. Here, we investigated the role of DNA methylation in suppressing interleukin 2 (IL-2) expression in memory CD4(+) T cells during chronic HIV-1 infection. We observed that CpG sites in the IL2 promoter of CD4(+) T cells were fully methylated in naive CD4(+) T cells and significantly demethylated in the memory populations. Interestingly, we found that the memory cells that had a terminally differentiated phenotype and expressed CD57 had increased IL2 promoter methylation relative to less differentiated memory cells in healthy individuals. Importantly, early effector memory subsets from HIV-1-infected subjects expressed high levels of CD57 and were highly methylated at the IL2 locus. Furthermore, the increased CD57 expression on memory CD4(+) T cells was inversely correlated with IL-2 production. These data suggest that DNA methylation at the IL2 locus in CD4(+) T cells is coupled to immunosenescence and plays a critical role in the broad dysfunction that occurs in polyclonal T cells during HIV-1 infection.


Immunology and Cell Biology | 2017

Maintenance of PD-1 on brain-resident memory CD8 T cells is antigen independent

Shwetank; Hossam A. Abdelsamed; Elizabeth L. Frost; Heather M Schmitz; Taryn E Mockus; Ben Youngblood; Aron E. Lukacher

Infection of the central nervous system (CNS) by murine polyomavirus (MuPyV), a persistent natural mouse pathogen, establishes brain‐resident memory CD8 T cells (bTRM) that uniformly and chronically express programmed cell death protein 1 (PD‐1) irrespective of the expression of αE integrin CD103, a TRM cell marker. In contrast, memory antiviral CD8 T cells in the spleen are PD‐1−, despite viral loads being similar in both the brain and spleen during persistent infection. Repetitive antigen engagement is central to sustained PD‐1 expression by T cells in chronic viral infections; however, recent evidence indicates that expression of inhibitory receptors, including PD‐1, is part of the TRM differentiation program. Here we asked whether PD‐1 expression by CD8 bTRM cells during persistent MuPyV encephalitis is antigen dependent. By transferring MuPyV‐specific CD8 bTRM cells into the brains of naive mice and mice infected with cognate epitope‐sufficient and ‐deficient MuPyVs, we demonstrate that antigen and inflammation are dispensable for PD‐1 maintenance. In vitro and direct ex vivo analyses indicate that CD103− MuPyV‐specific CD8 bTRM retain functional competence. We further show that the Pdcd‐1 promoter of anti‐MuPyV bTRM cells is epigenetically fixed in a demethylated state in the brain. In contrast, the PD‐1 promoter of splenic antiviral memory CD8 T cells undergoes remethylation after being demethylated during acute infection. These data show that PD‐1 expression is an intrinsic property of brain TRM cells in a persistent CNS viral infection.


Immunity | 2013

Distinct memory cd4+ t cells with commitment to t follicular helper- and t helper 1-cell lineages are generated after acute viral infection

J. Scott Hale; Ben Youngblood; Donald R. Latner; Ata Ur Rasheed Mohammed; Lilin Ye; Rama Akondy; Tuoqi Wu; Smita S. Iyer; Rafi Ahmed


Cold Spring Harbor Symposia on Quantitative Biology | 2013

Programmed Cell Death 1-Directed Immunotherapy for Enhancing T-Cell Function

Koichi Araki; Ben Youngblood; Rafi Ahmed


BIO-PROTOCOL | 2017

In vitro Homeostatic Proliferation of Human CD8 T Cells

Hossam A. Abdelsamed; Caitlin C. Zebley; Ben Youngblood

Collaboration


Dive into the Ben Youngblood's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hossam A. Abdelsamed

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hazem E. Ghoneim

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

J. Scott Hale

University of Washington

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge