Network


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

Hotspot


Dive into the research topics where Andrew M. Intlekofer is active.

Publication


Featured researches published by Andrew M. Intlekofer.


Nature Immunology | 2005

Effector and memory CD8 + T cell fate coupled by T-bet and eomesodermin

Andrew M. Intlekofer; Naofumi Takemoto; E. John Wherry; Sarah Longworth; John T Northrup; Vikram R. Palanivel; Alan C. Mullen; Christopher R Gasink; Susan M. Kaech; Joseph D. Miller; Laurent Gapin; Kenneth Ryan; Andreas P. Russ; Tullia Lindsten; Jordan S. Orange; Ananda W. Goldrath; Rafi Ahmed; Steven L. Reiner

Two seemingly unrelated hallmarks of memory CD8+ T cells are cytokine-driven proliferative renewal after pathogen clearance and a latent effector program in anticipation of rechallenge. Memory CD8+ T cells and natural killer cells share cytotoxic potential and dependence on the growth factor interleukin 15. We now show that mice with compound mutations of the genes encoding the transcription factors T-bet and eomesodermin were nearly devoid of several lineages dependent on interleukin 15, including memory CD8+ T cells and mature natural killer cells, and that their cells had defective cytotoxic effector programming. Moreover, T-bet and eomesodermin were responsible for inducing enhanced expression of CD122, the receptor specifying interleukin 15 responsiveness. Therefore, these key transcription factors link the long-term renewal of memory CD8+ T cells to their characteristic effector potency.*Note: In the version of this article initially published online, the third sentence of the abstract was incorrect. The correct sentence is as follows: “We now show that mice with compound mutations of the genes encoding the transcription factors T-bet and eomesodermin were nearly devoid of several lineages dependent on interleukin 15, including memory CD8+ T cells and mature natural killer cells, and that their cells had defective cytotoxic effector programming.” The error has been corrected for the HTML and print versions of the article. Additionally, in the print version of this article and the version initially published online, some labels for Tbx21 in Figure 7b are incorrect. This correction has been appended to the PDF version.


Science | 2007

Asymmetric T Lymphocyte Division in the Initiation of Adaptive Immune Responses

John T. Chang; Vikram R. Palanivel; Ichiko Kinjyo; Felix Schambach; Andrew M. Intlekofer; Arnob Banerjee; Sarah Longworth; Kristine E. Vinup; Paul Mrass; Jane Oliaro; Nigel Killeen; Jordan S. Orange; Sarah M. Russell; Wolfgang J. Weninger; Steven L. Reiner

A hallmark of mammalian immunity is the heterogeneity of cell fate that exists among pathogen-experienced lymphocytes. We show that a dividing T lymphocyte initially responding to a microbe exhibits unequal partitioning of proteins that mediate signaling, cell fate specification, and asymmetric cell division. Asymmetric segregation of determinants appears to be coordinated by prolonged interaction between the T cell and its antigen-presenting cell before division. Additionally, the first two daughter T cells displayed phenotypic and functional indicators of being differentially fated toward effector and memory lineages. These results suggest a mechanism by which a single lymphocyte can apportion diverse cell fates necessary for adaptive immunity.


Science | 2008

Anomalous Type 17 Response to Viral Infection by CD8+ T Cells Lacking T-bet and Eomesodermin

Andrew M. Intlekofer; Arnob Banerjee; Naofumi Takemoto; Scott M. Gordon; Caitlin S. DeJong; Haina Shin; Christopher A. Hunter; E. John Wherry; Tullia Lindsten; Steven L. Reiner

When intracellular pathogens invade mammalian hosts, naïve CD8+ T cells differentiate into cytotoxic killers, which lyse infected target cells and secrete cytokines that activate intracellular microbicides. We show that CD8+ T cells deficient in the transcription factors T-bet and eomesodermin (Eomes) fail to differentiate into functional killers required for defense against lymphocytic choriomeningitis virus. Instead, virus-specific CD8+ T cells lacking both T-bet and Eomes differentiate into an interleukin-17–secreting lineage, reminiscent of the helper T cell fate that has been implicated in autoimmunity and extracellular microbial defense. Upon viral infection, mice with T cells lacking both T-bet and Eomes develop a CD8+ T cell–dependent, progressive inflammatory and wasting syndrome characterized by multi-organ infiltration of neutrophils. T-bet and Eomes, thus, ensure that CD8+ T cells adopt an appropriate course of intracellular rather than extracellular destruction.


Journal of Immunology | 2006

Cutting Edge: IL-12 Inversely Regulates T-bet and Eomesodermin Expression during Pathogen-Induced CD8+ T Cell Differentiation

Naofumi Takemoto; Andrew M. Intlekofer; John T Northrup; E. John Wherry; Steven L. Reiner

Cytokines are critical determinants for specification of lineage-defining transcription factors of CD4+ T cell subsets. Little is known, however, about how cytokines regulate expression of T-bet and eomesodermin (Eomes) in effector and memory CD8+ T cells. We now report that IL-12, a signature of cell-mediated immunity, represses Eomes while positively regulating T-bet in effector CD8+ T cells during infection with Listeria monocytogenes. After resolution of infection and abatement of IL-12 signaling, Eomes expression rises whereas T-bet expression declines in memory CD8+ T cells. Eomes becomes derepressed in effector cells by ablation of IL-12 signaling. In the absence of IL-12, the dynamics of clonal expansion and contraction are also perturbed. Together, these results reveal how a pathogen-associated signal, such as IL-12, could act as a switch, regulating appropriate clonal growth and decline while, in parallel, shaping a unique pattern of fate-determining transcription factors.


Immunity | 2009

A Role for the Transcriptional Repressor Blimp-1 in CD8+ T Cell Exhaustion during Chronic Viral Infection

Haina Shin; Shawn D. Blackburn; Andrew M. Intlekofer; Charlly Kao; Jill M. Angelosanto; Steven L. Reiner; E. John Wherry

T cell exhaustion is common during chronic infections and can prevent optimal immunity. Although recent studies have demonstrated the importance of inhibitory receptors and other pathways in T cell exhaustion, the underlying transcriptional mechanisms are unknown. Here, we define a role for the transcription factor Blimp-1 in CD8(+) T cell exhaustion during chronic viral infection. Blimp-1 repressed key aspects of normal memory CD8(+) T cell differentiation and promoted high expression of inhibitory receptors during chronic infection. These cardinal features of CD8(+) T cell exhaustion were corrected by conditionally deleting Blimp-1. Although high expression of Blimp-1 fostered aspects of CD8(+) T cell exhaustion, haploinsufficiency indicated that moderate Blimp-1 expression sustained some effector function during chronic viral infection. Thus, we identify Blimp-1 as a transcriptional regulator of CD8(+) T cell exhaustion during chronic viral infection and propose that Blimp-1 acts as a transcriptional rheostat balancing effector function and T cell exhaustion.


Journal of Immunology | 2010

Cutting Edge: The Transcription Factor Eomesodermin Enables CD8+ T Cells To Compete for the Memory Cell Niche

Arnob Banerjee; Scott M. Gordon; Andrew M. Intlekofer; Michael A. Paley; Erin C. Mooney; Tulia Lindsten; E. John Wherry; Steven L. Reiner

CD8+ T cells responding to intracellular infection give rise to cellular progeny that become terminally differentiated effector cells and self-renewing memory cells. T-bet and eomesodermin (Eomes) are key transcription factors of cytotoxic lymphocyte lineages. We show in this study that CD8+ T cells lacking Eomes compete poorly in contributing to the pool of Ag-specific central memory cells. Eomes-deficient CD8+ T cells undergo primary clonal expansion but are defective in long-term survival, populating the bone marrow niche and re-expanding postrechallenge. The phenotype of Eomes-deficient CD8+ T cells supports the hypothesis that T-bet and Eomes can act redundantly to induce effector functions, but can also act to reciprocally promote terminal differentiation versus self-renewal of Ag-specific memory cells.


Nature Immunology | 2011

Transcription factor T-bet represses expression of the inhibitory receptor PD-1 and sustains virus-specific CD8 + T cell responses during chronic infection

Charlly Kao; Kenneth J. Oestreich; Michael A. Paley; Alison Crawford; Jill M. Angelosanto; Mohammed Alkhatim A Ali; Andrew M. Intlekofer; Jeremy M. Boss; Steven L. Reiner; Amy S. Weinmann; E. John Wherry

T cell exhaustion has a major role in failure to control chronic infection. High expression of inhibitory receptors, including PD-1, and the inability to sustain functional T cell responses contribute to exhaustion. However, the transcriptional control of these processes remains unclear. Here we demonstrate that the transcription factor T-bet regulated the exhaustion of CD8+ T cells and the expression of inhibitory receptors. T-bet directly repressed transcription of the gene encoding PD-1 and resulted in lower expression of other inhibitory receptors. Although a greater abundance of T-bet promoted terminal differentiation after acute infection, high T-bet expression sustained exhausted CD8+ T cells and repressed the expression of inhibitory receptors during chronic viral infection. Persistent antigenic stimulation caused downregulation of T-bet, which resulted in more severe exhaustion of CD8+ T cells. Our observations suggest therapeutic opportunities involving higher T-bet expression during chronic infection.


Journal of Leukocyte Biology | 2013

At the bench: Preclinical rationale for CTLA-4 and PD-1 blockade as cancer immunotherapy

Andrew M. Intlekofer; Craig B. Thompson

Tumors can avoid immune surveillance by stimulating immune inhibitory receptors that function to turn off established immune responses. By blocking the ability of tumors to stimulate inhibitory receptors on T cells, sustained, anti‐tumor immune responses can be generated in animals. Thus, therapeutic blockade of immune inhibitory checkpoints provides a potential method to boost anti‐tumor immunity. The CTLA‐4 and PD‐1Rs represent two T cell‐inhibitory receptors with independent mechanisms of action. Preclinical investigations revealed that CTLA‐4 enforces an activation threshold and attenuates proliferation of tumor‐specific T lymphocytes. In contrast, PD‐1 functions primarily as a stop signal that limits T cell effector function within a tumor. The unique mechanisms and sites of action of CTLA‐4 and PD‐1 suggest that although blockade of either has the potential to promote anti‐tumor immune responses, combined blockade of both might offer even more potent anti‐tumor activity. See related review At the Bedside: CTLA‐4 and PD‐1 blocking antibodies in cancer immunotherapy.


Genes & Development | 2009

Pre-TCR signaling inactivates Notch1 transcription by antagonizing E2A

Yumi Yashiro-Ohtani; Yiping He; Takuya Ohtani; Mary Elizabeth Jones; Olga Shestova; Lanwei Xu; Terry C. Fang; Mark Y. Chiang; Andrew M. Intlekofer; Stephen C. Blacklow; Yuan Zhuang

Precise control of the timing and magnitude of Notch signaling is essential for the normal development of many tissues, but the feedback loops that regulate Notch are poorly understood. Developing T cells provide an excellent context to address this issue. Notch1 signals initiate T-cell development and increase in intensity during maturation of early T-cell progenitors (ETP) to the DN3 stage. As DN3 cells undergo beta-selection, during which cells expressing functionally rearranged TCRbeta proliferate and differentiate into CD4(+)CD8(+) progeny, Notch1 signaling is abruptly down-regulated. In this report, we investigate the mechanisms that control Notch1 expression during thymopoiesis. We show that Notch1 and E2A directly regulate Notch1 transcription in pre-beta-selected thymocytes. Following successful beta-selection, pre-TCR signaling rapidly inhibits Notch1 transcription via signals that up-regulate Id3, an E2A inhibitor. Consistent with a regulatory role for Id3 in Notch1 down-regulation, post-beta-selected Id3-deficient thymocytes maintain Notch1 transcription, whereas enforced Id3 expression decreases Notch1 expression and abrogates Notch1-dependent T-cell survival. These data provide new insights into Notch1 regulation in T-cell progenitors and reveal a direct link between pre-TCR signaling and Notch1 expression during thymocyte development. Our findings also suggest new strategies for inhibiting Notch1 signaling in pathologic conditions.


Genes & Development | 2013

Induction of sarcomas by mutant IDH2

Chao Lu; Sriram Venneti; Altuna Akalin; Fang Fang; Patrick S. Ward; Raymond G. DeMatteo; Andrew M. Intlekofer; Chong Chen; Jiangbin Ye; Meera Hameed; Khedoudja Nafa; Narasimhan P. Agaram; Justin R. Cross; Raya Khanin; Christopher E. Mason; John H. Healey; Scott W. Lowe; Gary K. Schwartz; Ari Melnick; Craig B. Thompson

More than 50% of patients with chondrosarcomas exhibit gain-of-function mutations in either isocitrate dehydrogenase 1 (IDH1) or IDH2. In this study, we performed genome-wide CpG methylation sequencing of chondrosarcoma biopsies and found that IDH mutations were associated with DNA hypermethylation at CpG islands but not other genomic regions. Regions of CpG island hypermethylation were enriched for genes implicated in stem cell maintenance/differentiation and lineage specification. In murine 10T1/2 mesenchymal progenitor cells, expression of mutant IDH2 led to DNA hypermethylation and an impairment in differentiation that could be reversed by treatment with DNA-hypomethylating agents. Introduction of mutant IDH2 also induced loss of contact inhibition and generated undifferentiated sarcomas in vivo. The oncogenic potential of mutant IDH2 correlated with the ability to produce 2-hydroxyglutarate. Together, these data demonstrate that neomorphic IDH2 mutations can be oncogenic in mesenchymal cells.

Collaboration


Dive into the Andrew M. Intlekofer's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ross L. Levine

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Craig B. Thompson

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

E. John Wherry

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Ahmet Dogan

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Jie He

Foundation Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Anas Younes

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Andrew D. Zelenetz

Memorial Sloan Kettering Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge