Network


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

Hotspot


Dive into the research topics where Thomas B. Huffaker is active.

Publication


Featured researches published by Thomas B. Huffaker.


Cell Reports | 2012

Epistasis between microRNAs 155 and 146a during T cell-mediated antitumor immunity

Thomas B. Huffaker; Ruozhen Hu; Marah C. Runtsch; Erin Bake; Xinjian Chen; Jimmy L. Zhao; June L. Round; David Baltimore; Ryan M. O'Connell

An increased understanding of antitumor immunity is necessary for improving cell-based immunotherapies against human cancers. Here, we investigated the roles of two immune system-expressed microRNAs (miRNAs), miR-155 and miR-146a, in the regulation of antitumor immune responses. Our results indicate that miR-155 promotes and miR-146a inhibits interferon γ (IFNγ) responses by T cells and reduces solid tumor growth in vivo. Using a double-knockout (DKO) mouse strain deficient in both miR-155 and miR-146a, we have also identified an epistatic relationship between these two miRNAs. DKO mice had defective T cell responses and tumor growth phenotypes similar to miR-155(-/-) mice. Further analysis of the T cell compartment revealed that miR-155 modulates IFNγ expression through a mechanism involving repression of Ship1. Our work reveals critical roles for miRNAs in the reciprocal regulation of CD4(+) and CD8(+) T cell-mediated antitumor immunity and demonstrates the dominant nature of miR-155 during its promotion of immune responses.


Immunity | 2014

miR-155 Promotes T Follicular Helper Cell Accumulation during Chronic, Low-Grade Inflammation

Ruozhen Hu; Dominique A. Kagele; Thomas B. Huffaker; Marah C. Runtsch; Margaret Alexander; Jin Liu; Erin Bake; Wei Su; Matthew A. Williams; Dinesh S. Rao; Thomas Möller; Gwenn A. Garden; June L. Round; Ryan M. O’Connell

Chronic inflammation is a contributing factor to most life-shortening human diseases. However, the molecular and cellular mechanisms that sustain chronic inflammatory responses remain poorly understood, making it difficult to treat this deleterious condition. Using a mouse model of age-dependent inflammation that results from a deficiency in miR-146a, we demonstrate that miR-155 contributed to the progressive inflammatory disease that emerged as Mir146a(-/-) mice grew older. Upon analyzing lymphocytes from inflamed versus healthy middle-aged mice, we found elevated numbers of T follicular helper (Tfh) cells, germinal center (GC) B cells, and autoantibodies, all occurring in a miR-155-dependent manner. Further, Cd4-cre Mir155(fl/fl) mice were generated and demonstrated that miR-155 functions in T cells, in addition to its established role in B cells, to promote humoral immunity in a variety of contexts. Taken together, our study discovers that miR-146a and miR-155 counterregulate Tfh cell development that drives aberrant GC reactions during chronic inflammation.


Journal of Immunology | 2013

MicroRNA-155 Confers Encephalogenic Potential to Th17 Cells by Promoting Effector Gene Expression

Ruozhen Hu; Thomas B. Huffaker; Dominique A. Kagele; Marah C. Runtsch; Erin Bake; Aadel A. Chaudhuri; June L. Round; Ryan M. O'Connell

Th17 cells are central to the pathogenesis of autoimmune disease, and recently specific noncoding microRNAs have been shown to regulate their development. However, it remains unclear whether microRNAs are also involved in modulating Th17 cell effector functions. Consequently, we examined the role of miR-155 in differentiated Th17 cells during their induction of experimental autoimmune encephalomyelitis. Using adoptive transfer experiments, we found that highly purified, myelin oligodendrocyte glycoprotein Ag-specific Th17 cells lacking miR-155 were defective in their capacity to cause experimental autoimmune encephalomyelitis. Gene expression profiling of purified miR-155−/−IL-17F+ Th17 cells identified a subset of effector genes that are dependent on miR-155 for their proper expression through a mechanism involving repression of the transcription factor Ets1. Among the genes reduced in the absence of miR-155 was IL-23R, resulting in miR-155−/− Th17 cells being hyporesponsive to IL-23. Taken together, our study demonstrates a critical role for miR-155 in Th17 cells as they unleash autoimmune inflammation and finds that this occurs through a signaling network involving miR-155, Ets1, and the clinically relevant IL-23–IL-23R pathway.


Blood | 2017

miR-155 promotes FLT3-ITD–induced myeloproliferative disease through inhibition of the interferon response

Jared Wallace; Dominique A. Kagele; Anna M. Eiring; Carissa N. Kim; Ruozhen Hu; Marah C. Runtsch; Margaret Alexander; Thomas B. Huffaker; Soh Hyun Lee; Ami B. Patel; Timothy L. Mosbruger; Dinesh S. Rao; Rodney R. Miles; June L. Round; Michael W. Deininger; Ryan M. O'Connell

FLT3-ITD+ acute myeloid leukemia (AML) accounts for ∼25% of all AML cases and is a subtype that carries a poor prognosis. microRNA-155 (miR-155) is specifically overexpressed in FLT3-ITD+ AML compared with FLT3 wild-type (FLT3-WT) AML and is critical for the growth of FLT3-ITD+ AML cells in vitro. However, miR-155s role in regulating FLT3-ITD-mediated disease in vivo remains unclear. In this study, we used a genetic mouse model to determine whether miR-155 influences the development of FLT3-ITD-induced myeloproliferative disease. Results indicate that miR-155 promotes FLT3-ITD-induced myeloid expansion in the bone marrow, spleen, and peripheral blood. Mechanistically, miR-155 increases proliferation of the hematopoietic stem and progenitor cell compartments by reducing the growth-inhibitory effects of the interferon (IFN) response, and this involves targeting of Cebpb. Consistent with our observations in mice, primary FLT3-ITD+ AML clinical samples have significantly higher miR-155 levels and a lower IFN response compared with FLT3-WT AML samples. Further, inhibition of miR-155 in FLT3-ITD+ AML cell lines using CRISPR/Cas9, or primary FLT3-ITD+ AML samples using locked nucleic acid antisense inhibitors, results in an elevated IFN response and reduces colony formation. Altogether, our data reveal that miR-155 collaborates with FLT3-ITD to promote myeloid cell expansion in vivo and that this involves a multitarget mechanism that includes repression of IFN signaling.


Journal of Neuroimmunology | 2017

MicroRNA 155 and viral-induced neuroinflammation

Laura L. Dickey; Timothy M. Hanley; Thomas B. Huffaker; Andrew G. Ramstead; Ryan M. O'Connell; Thomas E. Lane

MicroRNA (miRNA) regulation of gene expression is becoming an increasingly recognized mechanism by which host immune responses are governed following microbial infection. miRNAs are short, non-coding RNAs that repress translation of target genes, and have been implicated in a number of activities that modulate host immune responses, including the regulation of immune cell proliferation, survival, expansion, differentiation, migration, polarization, and effector function. This review highlights several examples in which mammalian-encoded miR-155 influences immune responses following viral infection of the CNS.


Journal of Immunology | 2017

Rab27-Dependent exosome production inhibits chronic inflammation and enables acute responses to inflammatory stimuli

Margaret Alexander; Andrew G. Ramstead; Kaylyn M. Bauer; Soh-Hyun Lee; Marah C. Runtsch; Jared Wallace; Thomas B. Huffaker; Dane K. Larsen; Tanya Tolmachova; Miguel C. Seabra; June L. Round; Diane M. Ward; Ryan M. O’Connell

Extracellular vesicles, including exosomes, have recently been implicated as novel mediators of immune cell communication in mammals. However, roles for endogenously produced exosomes in regulating immune cell functions in vivo are just beginning to be identified. In this article, we demonstrate that Rab27a and Rab27b double-knockout (Rab27DKO) mice that are deficient in exosome secretion have a chronic, low-grade inflammatory phenotype characterized by elevated inflammatory cytokines and myeloproliferation. Upon further investigation, we found that some of these phenotypes could be complemented by wild-type (WT) hematopoietic cells or administration of exosomes produced by GM-CSF–expanded bone marrow cells. In addition, chronically inflamed Rab27DKO mice had a blunted response to bacterial LPS, resembling endotoxin tolerance. This defect was rescued by bone marrow exosomes from WT, but not miR-155−/−, cells, suggesting that uptake of miR-155–containing exosomes is important for a proper LPS response. Further, we found that SHIP1 and IRAK-M, direct targets of miR-155 that are known negative regulators of the LPS response, were elevated in Rab27DKO mice and decreased after treatment with WT, but not miR-155−/−, exosomes. Together, our study finds that Rab27-dependent exosome production contributes to homeostasis within the hematopoietic system and appropriate responsiveness to inflammatory stimuli.


Journal of Biological Chemistry | 2017

Antitumor immunity is defective in T cell–specific microRNA-155–deficient mice and is rescued by immune checkpoint blockade

Thomas B. Huffaker; Soh-Hyun Lee; William W. Tang; Jared Wallace; Margaret Alexander; Marah C. Runtsch; Dane K. Larsen; Jacob Thompson; Andrew G. Ramstead; Ruozhen Hu; June L. Round; Matthew A. Williams; Ryan M. O'Connell

MicroRNA-155 (miR-155) regulates antitumor immune responses. However, its specific functions within distinct immune cell types have not been delineated in conditional KO mouse models. In this study, we investigated the role of miR-155 specifically within T cells during the immune response to syngeneic tumors. We found that miR-155 expression within T cells is required to limit syngeneic tumor growth and promote IFNγ production by T cells within the tumor microenvironment. Consequently, we found that miR-155 expression by T cells is necessary for proper tumor-associated macrophage expression of IFNγ-inducible genes. We also found that immune checkpoint–blocking (ICB) antibodies against programmed cell death protein 1/programmed death ligand 1 (PD-1/PD-L1) and cytotoxic T lymphocyte–associated protein 4 (CTLA-4) restored antitumor immunity in miR-155 T cell–conditional KO mice. We noted that these ICB antibodies rescued the levels of IFNγ-expressing T cells, expression of multiple activation and effector genes expressed by tumor-infiltrating CD8+ and CD4+ T cells, and tumor-associated macrophage activation. Moreover, the ICB approach partially restored expression of several derepressed miR-155 targets in tumor-infiltrating, miR-155–deficient CD8+ T cells, suggesting that miR-155 and ICB regulate overlapping pathways to promote antitumor immunity. Taken together, our findings highlight the multifaceted role of miR-155 in T cells, in which it promotes antitumor immunity. These results suggest that the augmentation of miR-155 expression could be used to improve anticancer immunotherapies.


Immunity | 2015

miR-155-SOCS1 as a Functional Axis: Satisfying the Burden of Proof

Thomas B. Huffaker; Ryan M. O’Connell


Blood | 2016

MiR-155 Promotes FLT3-ITD-Induced Myeloproliferative Disease through Inhibition of Interferon Signaling

Jared Wallace; Dominique A. Kagele; Ruozhen Hu; Marah C. Runtsch; Margaret Alexander; Thomas B. Huffaker; Timothy L. Mosbruger; Dinesh S. Rao; Rodney R. Miles; June L. Round; Ryan M. O'Connell


Blood | 2015

Genome-Wide Crispr-Cas9 Screen Identifies Functionally Relevant Micro-RNAs in FLT3-ITD+ AML

Jared Wallace; Dominique A. Kagele; Ruozhen Hu; Marah C. Runtsch; Margaret Alexander; Thomas B. Huffaker; Timothy L. Mosbruger; Timothy J. Dahlem; W. Zac Stephens; Dinesh S. Rao; Rodney R. Miles; June L. Round; Ryan M. O'Connell

Collaboration


Dive into the Thomas B. Huffaker'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

Dinesh S. Rao

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge