%0 Journal Article %J PLoS Pathog %D 2015 %T CD39 Expression Identifies Terminally Exhausted CD8+ T Cells. %A Gupta, Prakash K %A Godec, Jernej %A Wolski, David %A Adland, Emily %A Yates, Kathleen %A Pauken, Kristen E %A Cosgrove, Cormac %A Ledderose, Carola %A Junger, Wolfgang G %A Robson, Simon C %A Wherry, E John %A Alter, Galit %A Goulder, Philip J R %A Klenerman, Paul %A Sharpe, Arlene H %A Lauer, Georg M %A Haining, W Nicholas %K Animals %K Antigens, CD %K Apyrase %K Arenaviridae Infections %K Biomarkers %K CD8-Positive T-Lymphocytes %K Chromatography, High Pressure Liquid %K Chronic Disease %K Disease Models, Animal %K Flow Cytometry %K Hepatitis C, Chronic %K HIV Infections %K Humans %K Lymphocytic Choriomeningitis %K Lymphocytic choriomeningitis virus %K Mice %K Mice, Inbred C57BL %K Oligonucleotide Array Sequence Analysis %K RNA Virus Infections %K T-Lymphocyte Subsets %X Exhausted T cells express multiple co-inhibitory molecules that impair their function and limit immunity to chronic viral infection. Defining novel markers of exhaustion is important both for identifying and potentially reversing T cell exhaustion. Herein, we show that the ectonucleotidse CD39 is a marker of exhausted CD8+ T cells. CD8+ T cells specific for HCV or HIV express high levels of CD39, but those specific for EBV and CMV do not. CD39 expressed by CD8+ T cells in chronic infection is enzymatically active, co-expressed with PD-1, marks cells with a transcriptional signature of T cell exhaustion and correlates with viral load in HIV and HCV. In the mouse model of chronic Lymphocytic Choriomeningitis Virus infection, virus-specific CD8+ T cells contain a population of CD39high CD8+ T cells that is absent in functional memory cells elicited by acute infection. This CD39high CD8+ T cell population is enriched for cells with the phenotypic and functional profile of terminal exhaustion. These findings provide a new marker of T cell exhaustion, and implicate the purinergic pathway in the regulation of T cell exhaustion. %B PLoS Pathog %V 11 %P e1005177 %8 2015 Oct %G eng %N 10 %1 http://www.ncbi.nlm.nih.gov/pubmed/26485519?dopt=Abstract %R 10.1371/journal.ppat.1005177 %0 Journal Article %J Cancer Immunol Res %D 2015 %T PD-L1 Antibodies to Its Cytoplasmic Domain Most Clearly Delineate Cell Membranes in Immunohistochemical Staining of Tumor Cells. %A Mahoney, Kathleen M %A Sun, Heather %A Liao, Xiaoyun %A Hua, Ping %A Callea, Marcella %A Greenfield, Edward A %A Hodi, F Stephen %A Sharpe, Arlene H %A Signoretti, Sabina %A Rodig, Scott J %A Freeman, Gordon J %X Blocking the programmed death-1 (PD-1) pathway has clinical benefit in metastatic cancer and has led to the approval of the mAbs pembrolizumab and nivolumab to treat melanoma and nivolumab for non-small cell lung cancer. Expression of PD-L1 on the cell surface of either tumor cells or infiltrating immune cells is associated with a higher likelihood of response to PD-1 blockade in multiple studies. Most mAbs to PD-L1 in use are directed to its extracellular domain and immunohistochemically stain tumor tissue with a mixture of cytoplasmic and membrane staining. Cytoplasmic staining obscures the interpretation of a positive reaction on the tumor cell membrane, and thus affects the accuracy of PD-L1 scoring systems. We developed a mAb to the cytoplasmic domain of PD-L1, 405.9A11 (9A11), which is both more selective for membranous PD-L1 and more sensitive in IHC and Western blotting, compared with previous mAbs specific for the PD-L1 extracellular domain. Here, we compare immunohistochemical staining patterns of PD-L1 expression in five types of tumors, using five PD-L1 mAbs: 9A11, 7G11, and three commercially available mAbs. We demonstrate that 9A11, as well as two other cytoplasmic domain-specific mAbs, E1L3N and SP142, can clearly delineate the membrane of PD-L1-positive cells in formalin-fixed paraffin-embedded tissue and facilitate interpretation of staining results. %B Cancer Immunol Res %V 3 %P 1308-15 %8 2015 Dec %G eng %N 12 %1 http://www.ncbi.nlm.nih.gov/pubmed/26546452?dopt=Abstract %R 10.1158/2326-6066.CIR-15-0116 %0 Journal Article %J Sci Adv %D 2015 %T The PTEN pathway in Tregs is a critical driver of the suppressive tumor microenvironment. %A Sharma, Madhav D %A Shinde, Rahul %A McGaha, Tracy L %A Huang, Lei %A Holmgaard, Rikke B %A Wolchok, Jedd D %A Mautino, Mario R %A Celis, Esteban %A Sharpe, Arlene H %A Francisco, Loise M %A Powell, Jonathan D %A Yagita, Hideo %A Mellor, Andrew L %A Blazar, Bruce R %A Munn, David H %X The tumor microenvironment is profoundly immunosuppressive. We show that multiple tumor types create intratumoral immune suppression driven by a specialized form of regulatory T cell (Treg) activation dependent on the PTEN (phosphatase and tensin homolog) lipid phosphatase. PTEN acted to stabilize Tregs in tumors, preventing them from reprogramming into inflammatory effector cells. In mice with a Treg-specific deletion of PTEN, tumors grew slowly, were inflamed, and could not create an immunosuppressive tumor microenvironment. In normal mice, exposure to apoptotic tumor cells rapidly elicited PTEN-expressing Tregs, and PTEN-deficient mice were unable to maintain tolerance to apoptotic cells. In wild-type mice with large established tumors, pharmacologic inhibition of PTEN after chemotherapy or immunotherapy profoundly reconfigured the tumor microenvironment, changing it from a suppressive to an inflammatory milieu, and tumors underwent rapid regression. Thus, the immunosuppressive milieu in tumors must be actively maintained, and tumors become susceptible to immune attack if the PTEN pathway in Tregs is disrupted. %B Sci Adv %V 1 %P e1500845 %8 2015 Nov %G eng %N 10 %1 http://www.ncbi.nlm.nih.gov/pubmed/26601142?dopt=Abstract %R 10.1126/sciadv.1500845 %0 Journal Article %J J Exp Med %D 2015 %T Genetic absence of PD-1 promotes accumulation of terminally differentiated exhausted CD8+ T cells. %A Odorizzi, Pamela M %A Pauken, Kristen E %A Paley, Michael A %A Sharpe, Arlene %A Wherry, E John %K Adoptive Transfer %K Animals %K Bromodeoxyuridine %K CD8-Positive T-Lymphocytes %K Cell Differentiation %K Cell Proliferation %K Flow Cytometry %K Lymphocytic Choriomeningitis %K Mice %K Programmed Cell Death 1 Receptor %X Programmed Death-1 (PD-1) has received considerable attention as a key regulator of CD8(+) T cell exhaustion during chronic infection and cancer because blockade of this pathway partially reverses T cell dysfunction. Although the PD-1 pathway is critical in regulating established "exhausted" CD8(+) T cells (TEX cells), it is unclear whether PD-1 directly causes T cell exhaustion. We show that PD-1 is not required for the induction of exhaustion in mice with chronic lymphocytic choriomeningitis virus (LCMV) infection. In fact, some aspects of exhaustion are more severe with genetic deletion of PD-1 from the onset of infection. Increased proliferation between days 8 and 14 postinfection is associated with subsequent decreased CD8(+) T cell survival and disruption of a critical proliferative hierarchy necessary to maintain exhausted populations long term. Ultimately, the absence of PD-1 leads to the accumulation of more cytotoxic, but terminally differentiated, CD8(+) TEX cells. These results demonstrate that CD8(+) T cell exhaustion can occur in the absence of PD-1. They also highlight a novel role for PD-1 in preserving TEX cell populations from overstimulation, excessive proliferation, and terminal differentiation. %B J Exp Med %V 212 %P 1125-37 %8 2015 Jun 29 %G eng %N 7 %1 http://www.ncbi.nlm.nih.gov/pubmed/26034050?dopt=Abstract %R 10.1084/jem.20142237 %0 Journal Article %J Curr Opin Immunol %D 2015 %T Hepatic immune regulation by stromal cells. %A Schildberg, Frank A %A Sharpe, Arlene H %A Turley, Shannon J %X A metabolic organ, the liver also has a central role in tolerance induction. Stromal cells lining the hepatic sinusoids, such as liver sinusoidal endothelial cells (LSECs) and hepatic stellate cells (HSCs), are the first liver cells to encounter gut-derived and systemic antigens, thereby shaping local and systemic tolerance. Recent studies have demonstrated that stromal cells can modulate immune responses by antigen-dependent and independent mechanisms. Stromal cells interfere with the function of other antigen-presenting cells (APCs) and induce non-responsive T cells as well as regulatory T cells and myeloid-derived suppressor cells (MDSCs). The immunosuppressive microenvironment thus created provides a means to protect the liver from tissue damage. Such tolerized surroundings, however, can be exploited by certain pathogens, promoting persistent liver infections. %B Curr Opin Immunol %V 32 %P 1-6 %8 2015 Feb %G eng %1 http://www.ncbi.nlm.nih.gov/pubmed/25463592?dopt=Abstract %R 10.1016/j.coi.2014.10.002 %0 Journal Article %J Proc Natl Acad Sci U S A %D 2015 %T Inducible RNAi in vivo reveals that the transcription factor BATF is required to initiate but not maintain CD8+ T-cell effector differentiation. %A Godec, Jernej %A Cowley, Glenn S %A Barnitz, R Anthony %A Root, David E %A Sharpe, Arlene H %A Haining, W Nicholas %K Animals %K Basic-Leucine Zipper Transcription Factors %K CD8-Positive T-Lymphocytes %K Cell Differentiation %K Gene Knockdown Techniques %K Hematopoietic Stem Cells %K Lentivirus %K Mice %K Mice, Inbred C57BL %K Mice, Transgenic %K RNA Interference %K RNA, Small Interfering %K Transduction, Genetic %K Transplantation Chimera %X The differentiation of effector CD8(+) T cells is critical for the development of protective responses to pathogens and for effective vaccines. In the first few hours after activation, naive CD8(+) T cells initiate a transcriptional program that leads to the formation of effector and memory T cells, but the regulation of this process is poorly understood. Investigating the role of specific transcription factors (TFs) in determining CD8(+) effector T-cell fate by gene knockdown with RNAi is challenging because naive T cells are refractory to transduction with viral vectors without extensive ex vivo stimulation, which obscures the earliest events in effector differentiation. To overcome this obstacle, we developed a novel strategy to test the function of genes in naive CD8(+) T cells in vivo by creating bone marrow chimera from hematopoietic progenitors transduced with an inducible shRNA construct. Following hematopoietic reconstitution, this approach allowed inducible in vivo gene knockdown in any cell type that developed from this transduced progenitor pool. We demonstrated that lentivirus-transduced progenitor cells could reconstitute normal hematopoiesis and develop into naive CD8(+) T cells that were indistinguishable from wild-type naive T cells. This experimental system enabled induction of efficient gene knockdown in vivo without subsequent manipulation. We applied this strategy to show that the TF BATF is essential for initial commitment of naive CD8(+) T cells to effector development but becomes dispensable by 72h. This approach makes possible the study of gene function in vivo in unperturbed cells of hematopoietic origin that are refractory to viral transduction. %B Proc Natl Acad Sci U S A %V 112 %P 512-7 %8 2015 Jan 13 %G eng %N 2 %1 http://www.ncbi.nlm.nih.gov/pubmed/25548173?dopt=Abstract %R 10.1073/pnas.1413291112 %0 Journal Article %J Gerontology %D 2015 %T Mitochondrial metabolism in T cell activation and senescence: a mini-review. %A Ron-Harel, Noga %A Sharpe, Arlene H %A Haigis, Marcia C %X The aging immune system is unable to optimally respond to pathogens and generate long-term immunological memory against encountered antigens. Amongst the immune components most affected by aging are T lymphocytes. T lymphocytes are cells of the cell-mediated immune system, which can recognize microbial antigens and either directly kill infected cells or support the maturation and activation of other immune cells. When activated, T cells undergo a metabolic switch to accommodate their changing needs at every stage of the immune response. Here we review the different aspects of metabolic regulation of T cell activation, focusing on the emerging role of mitochondrial metabolism, and discuss changes that may contribute to age-related decline in T cell potency. Better understanding of the role of mitochondrial metabolism in immune cell function could provide insights into mechanisms of immune senescence with the potential for developing novel therapeutic approaches to improve immune responses in aged individuals. %B Gerontology %V 61 %P 131-8 %8 2015 %G eng %N 2 %1 http://www.ncbi.nlm.nih.gov/pubmed/25402204?dopt=Abstract %R 10.1159/000362502 %0 Journal Article %J Exp Parasitol %D 2015 %T Ox40L-Ox40 pathway plays distinct roles in regulating Th2 responses but does not determine outcome of cutaneous leishmaniasis caused by Leishmania mexicana and Leishmania major. %A Tuladhar, Rashmi %A Oghumu, Steve %A Dong, Ran %A Peterson, Allison %A Sharpe, Arlene H %A Satoskar, Abhay R %K Animals %K Antibodies, Protozoan %K Cytokines %K Female %K Leishmania major %K Leishmania mexicana %K Leishmaniasis, Cutaneous %K Mice %K Mice, Inbred BALB C %K OX40 Ligand %K Receptors, OX40 %K Th2 Cells %K Virulence %X Ox40 ligand (Ox40L)-Ox40 pathway has been shown to enhance Th2 responses and play a role in pathogenesis of cutaneous leishmaniasis (CL) caused by Leishmania major. Using Ox40l(-/-) BALB/c mice we analyzed the role of this pathway in determining the outcome to CL caused by L. mexicana and compared to L. major. Contrary to our expectations, Ox40l(-/-) mice were highly susceptible to both L. major (LV39) and L. mexicana (M379) and developed large non-healing lesions containing parasites comparable to Ox40l(+/+) BALB/c mice. Interestingly, upon in vitro stimulation with Leishmania antigen (LmAg), the lymph node cells from L. major infected Ox40l(-/-) mice produced significantly less IL-4 and IL-10 compared to Ox40l(+/+) mice. L. mexicana infected Ox40l(-/-) and Ox40l(+/+) mice did not show any difference in the production of IL-4 and IL-10. No difference was noted in the amount of Th1 cytokines IFN-ү and IL-12 produced by Ox40l(-/-) and Ox40l(+/+) mice infected with either parasite. These results indicate that the Ox40L-Ox40 pathway promotes Th2 bias only in L. major infection but not L. mexicana infection and this pathway is not critical for susceptibility to CL. %B Exp Parasitol %V 148 %P 49-55 %8 2015 Jan %G eng %1 http://www.ncbi.nlm.nih.gov/pubmed/25447125?dopt=Abstract %R 10.1016/j.exppara.2014.11.002 %0 Journal Article %J Cell Rep %D 2015 %T ABCB5 Identifies Immunoregulatory Dermal Cells. %A Schatton, Tobias %A Yang, Jun %A Kleffel, Sonja %A Uehara, Mayuko %A Barthel, Steven R %A Schlapbach, Christoph %A Zhan, Qian %A Dudeney, Stephen %A Mueller, Hansgeorg %A Lee, Nayoung %A de Vries, Juliane C %A Meier, Barbara %A Vander Beken, Seppe %A Kluth, Mark A %A Ganss, Christoph %A Sharpe, Arlene H %A Waaga-Gasser, Ana Maria %A Sayegh, Mohamed H %A Abdi, Reza %A Scharffetter-Kochanek, Karin %A Murphy, George F %A Kupper, Thomas S %A Frank, Natasha Y %A Frank, Markus H %X Cell-based strategies represent a new frontier in the treatment of immune-mediated disorders. However, the paucity of markers for isolation of molecularly defined immunomodulatory cell populations poses a barrier to this field. Here, we show that ATP-binding cassette member B5 (ABCB5) identifies dermal immunoregulatory cells (DIRCs) capable of exerting therapeutic immunoregulatory functions through engagement of programmed cell death 1 (PD-1). Purified Abcb5(+) DIRCs suppressed T cell proliferation, evaded immune rejection, homed to recipient immune tissues, and induced Tregs in vivo. In fully major-histocompatibility-complex-mismatched cardiac allotransplantation models, allogeneic DIRCs significantly prolonged allograft survival. Blockade of DIRC-expressed PD-1 reversed the inhibitory effects of DIRCs on T cell activation, inhibited DIRC-dependent Treg induction, and attenuated DIRC-induced prolongation of cardiac allograft survival, indicating that DIRC immunoregulatory function is mediated, at least in part, through PD-1. Our results identify ABCB5(+) DIRCs as a distinct immunoregulatory cell population and suggest promising roles of this expandable cell subset in cellular immunotherapy. %B Cell Rep %V 12 %P 1564-74 %8 2015 Sep 8 %G eng %N 10 %1 http://www.ncbi.nlm.nih.gov/pubmed/26321644?dopt=Abstract %R 10.1016/j.celrep.2015.08.010 %0 Journal Article %J Nat Immunol %D 2015 %T Control of PI(3) kinase in Treg cells maintains homeostasis and lineage stability. %A Huynh, Alexandria %A DuPage, Michel %A Priyadharshini, Bhavana %A Sage, Peter T %A Quiros, Jason %A Borges, Christopher M %A Townamchai, Natavudh %A Gerriets, Valerie A %A Rathmell, Jeffrey C %A Sharpe, Arlene H %A Bluestone, Jeffrey A %A Turka, Laurence A %K Animals %K Cell Lineage %K Enzyme-Linked Immunosorbent Assay %K Flow Cytometry %K Gene Deletion %K Homeostasis %K Mice %K Phosphatidylinositol 3-Kinases %K PTEN Phosphohydrolase %K Signal Transduction %K T-Lymphocytes, Regulatory %X Foxp3(+) regulatory T cells (Treg cells) are required for immunological homeostasis. One notable distinction between conventional T cells (Tconv cells) and Treg cells is differences in the activity of phosphatidylinositol-3-OH kinase (PI(3)K); only Tconv cells downregulate PTEN, the main negative regulator of PI(3)K, upon activation. Here we found that control of PI(3)K in Treg cells was essential for lineage homeostasis and stability. Mice lacking Pten in Treg cells developed an autoimmune-lymphoproliferative disease characterized by excessive T helper type 1 (TH1) responses and B cell activation. Diminished control of PI(3)K activity in Treg cells led to reduced expression of the interleukin-2 (IL-2) receptor α subunit CD25, accumulation of Foxp3(+)CD25(-) cells and, ultimately, loss of expression of the transcription factor Foxp3 in these cells. Collectively, our data demonstrate that control of PI(3)K signaling by PTEN in Treg cells is critical for maintaining their homeostasis, function and stability. %B Nat Immunol %V 16 %P 188-96 %8 2015 Feb %G eng %N 2 %1 http://www.ncbi.nlm.nih.gov/pubmed/25559257?dopt=Abstract %R 10.1038/ni.3077 %0 Journal Article %J Cell Rep %D 2015 %T Defective TFH Cell Function and Increased TFR Cells Contribute to Defective Antibody Production in Aging. %A Sage, Peter T %A Tan, Catherine L %A Freeman, Gordon J %A Haigis, Marcia %A Sharpe, Arlene H %X Defective antibody production in aging is broadly attributed to immunosenescence. However, the precise immunological mechanisms remain unclear. Here, we demonstrate an increase in the ratio of inhibitory T follicular regulatory (TFR) cells to stimulatory T follicular helper (TFH) cells in aged mice. Aged TFH and TFR cells are phenotypically distinct from those in young mice, exhibiting increased programmed cell death protein-1 expression but decreased ICOS expression. Aged TFH cells exhibit defective antigen-specific responses, and programmed cell death protein-ligand 1 blockade can partially rescue TFH cell function. In contrast, young and aged TFR cells have similar suppressive capacity on a per-cell basis in vitro and in vivo. Together, these studies reveal mechanisms contributing to defective humoral immunity in aging: an increase in suppressive TFR cells combined with impaired function of aged TFH cells results in reduced T-cell-dependent antibody responses in aged mice. %B Cell Rep %V 12 %P 163-71 %8 2015 Jul 14 %G eng %N 2 %1 http://www.ncbi.nlm.nih.gov/pubmed/26146074?dopt=Abstract %R 10.1016/j.celrep.2015.06.015 %0 Journal Article %J J Exp Med %D 2015 %T Deletion of CTLA-4 on regulatory T cells during adulthood leads to resistance to autoimmunity. %A Paterson, Alison M %A Lovitch, Scott B %A Sage, Peter T %A Juneja, Vikram R %A Lee, Youjin %A Trombley, Justin D %A Arancibia-Cárcamo, Carolina V %A Sobel, Raymond A %A Rudensky, Alexander Y %A Kuchroo, Vijay K %A Freeman, Gordon J %A Sharpe, Arlene H %X Cytotoxic T lymphocyte antigen-4 (CTLA-4) is an essential negative regulator of T cell responses. Germline Ctla4 deficiency is lethal, making investigation of the function of CTLA-4 on mature T cells challenging. To elucidate the function of CTLA-4 on mature T cells, we have conditionally ablated Ctla4 in adult mice. We show that, in contrast to germline knockout mice, deletion of Ctla4 during adulthood does not precipitate systemic autoimmunity, but surprisingly confers protection from experimental autoimmune encephalomyelitis (EAE) and does not lead to increased resistance to MC38 tumors. Deletion of Ctla4 during adulthood was accompanied by activation and expansion of both conventional CD4(+)Foxp3(-) (T conv) and regulatory Foxp3(+) (T reg cells) T cell subsets; however, deletion of CTLA-4 on T reg cells was necessary and sufficient for protection from EAE. CTLA-4 deleted T reg cells remained functionally suppressive. Deletion of Ctla4 on T reg cells alone or on all adult T cells led to major changes in the Ctla4 sufficient T conv cell compartment, including up-regulation of immunoinhibitory molecules IL-10, LAG-3 and PD-1, thereby providing a compensatory immunosuppressive mechanism. Collectively, our findings point to a profound role for CTLA-4 on T reg cells in limiting their peripheral expansion and activation, thereby regulating the phenotype and function of T conv cells. %B J Exp Med %8 2015 Sep 14 %G ENG %1 http://www.ncbi.nlm.nih.gov/pubmed/26371185?dopt=Abstract %R 10.1084/jem.20141030 %0 Journal Article %J PLoS One %D 2015 %T Helicobacter pylori cag pathogenicity island's role in B7-H1 induction and immune evasion. %A Lina, Taslima T %A Alzahrani, Shatha %A House, Jennifer %A Yamaoka, Yoshio %A Sharpe, Arlene H %A Rampy, Bill A %A Pinchuk, Irina V %A Reyes, Victor E %X During Helicobacter pylori (H. pylori) infection CD4+ T cells in the gastric lamina propria are hyporesponsive and polarized by Th1/Th17 cell responses controlled by Treg cells. We have previously shown that H. pylori upregulates B7-H1 expression on GEC, which, in turn, suppress T cell proliferation, effector function, and induce Treg cells in vitro. In this study, we investigated the underlying mechanisms and the functional relevance of B7-H1 induction by H. pylori infection to chronic infection. Using H. pylori wild type (WT), cag pathogenicity island (cag PAI-) and cagA- isogenic mutant strains we demonstrated that H. pylori requires its type 4 secretion system (T4SS) as well as its effector protein CagA and peptidoglycan (PG) fragments for B7-H1 upregulation on GEC. Our study also showed that H. pylori uses the p38 MAPK pathway to upregulate B7-H1 expression in GEC. In vivo confirmation was obtained when infection of C57BL/6 mice with H. pylori PMSS1 strain, which has a functional T4SS delivery system, but not with H. pylori SS1 strain lacking a functional T4SS, led to a strong upregulation of B7-H1 expression in the gastric mucosa, increased bacterial load, induction of Treg cells in the stomach, increased IL-10 in the serum. Interestingly, B7-H1-/- mice showed less Treg cells and reduced bacterial loads after infection. These studies demonstrate how H. pylori T4SS components activate the p38 MAPK pathway, upregulate B7-H1 expression by GEC, and cause Treg cell induction; thus, contribute to establishing a persistent infection characteristic of H. pylori. %B PLoS One %V 10 %P e0121841 %8 2015 %G eng %N 3 %1 http://www.ncbi.nlm.nih.gov/pubmed/25807464?dopt=Abstract %R 10.1371/journal.pone.0121841 %0 Journal Article %J Immunity %D 2015 %T ICOS:ICOS-ligand interaction is required for type 2 innate lymphoid cell function, homeostasis, and induction of airway hyperreactivity. %A Maazi, Hadi %A Patel, Nisheel %A Sankaranarayanan, Ishwarya %A Suzuki, Yuzo %A Rigas, Diamanda %A Soroosh, Pejman %A Freeman, Gordon J %A Sharpe, Arlene H %A Akbari, Omid %K Animals %K Asthma %K Female %K Gene Expression Regulation %K Homeostasis %K Humans %K Immunity, Innate %K Inducible T-Cell Co-Stimulator Ligand %K Inducible T-Cell Co-Stimulator Protein %K Interleukin-13 %K Interleukin-2 %K Interleukin-5 %K Interleukins %K Lymphocytes %K Mice, Transgenic %K Respiratory System %K Signal Transduction %K STAT5 Transcription Factor %X Allergic asthma is caused by Th2-cell-type cytokines in response to allergen exposure. Type 2 innate lymphoid cells (ILC2s) are a newly identified subset of immune cells that, along with Th2 cells, contribute to the pathogenesis of asthma by producing copious amounts of IL-5 and IL-13, which cause eosinophilia and airway hyperreactivity (AHR), a cardinal feature of asthma. ILC2s express ICOS, a T cell costimulatory molecule with a currently unknown function. Here we showed that a lack of ICOS on murine ILC2s and blocking the ICOS:ICOS-ligand interaction in human ILC2s reduced AHR and lung inflammation. ILC2s expressed both ICOS and ICOS-ligand, and the ICOS:ICOS-ligand interaction promoted cytokine production and survival in ILC2s through STAT5 signaling. Thus, ICOS:ICOS-ligand signaling pathway is critically involved in ILC2 function and homeostasis. %B Immunity %V 42 %P 538-51 %8 2015 Mar 17 %G eng %N 3 %1 http://www.ncbi.nlm.nih.gov/pubmed/25769613?dopt=Abstract %R 10.1016/j.immuni.2015.02.007 %0 Journal Article %J Methods Mol Biol %D 2015 %T In vitro assay to sensitively measure T(FR) suppressive capacity and T(FH) stimulation of B cell responses. %A Sage, Peter T %A Sharpe, Arlene H %X T follicular helper (T(FH)) cells stimulate, whereas T follicular regulatory (T(FR)) cells inhibit, B cell responses. Despite the potent immunoregulatory roles for TFR cells in controlling the magnitude of antibody production, the precise mechanisms by which T(FR) cells exert their suppressive effects are not yet clear. The lack of specific assays to assess T(FR) cell function separately from differentiation has hindered progress in elucidating T(FR) cell function. This is due, in part, to difficulty in separating T(FR) cells from phenotypically similar, but functionally different, T(FH) cells. Here we describe an in vitro approach for sensitively and quantitatively assessing the capacity of T(FR) cells to suppress T(FH)-mediated B cell antibody production utilizing both ELISA and flow cytometry to measure B cell responses. Beyond assessing T(FR) function, this assay system can also be used to sensitively measure T(FH) stimulatory capacity as well as B cell function. %B Methods Mol Biol %V 1291 %P 151-60 %8 2015 %G eng %1 http://www.ncbi.nlm.nih.gov/pubmed/25836309?dopt=Abstract %R 10.1007/978-1-4939-2498-1_13 %0 Journal Article %J Elife %D 2015 %T The kinase DYRK1A reciprocally regulates the differentiation of Th17 and regulatory T cells. %A Khor, Bernard %A Gagnon, John D %A Goel, Gautam %A Roche, Marly I %A Conway, Kara L %A Tran, Khoa %A Aldrich, Leslie N %A Sundberg, Thomas B %A Paterson, Alison M %A Mordecai, Scott %A Dombkowski, David %A Schirmer, Melanie %A Tan, Pauline H %A Bhan, Atul K %A Roychoudhuri, Rahul %A Restifo, Nicholas P %A O'Shea, John J %A Medoff, Benjamin D %A Shamji, Alykhan F %A Schreiber, Stuart L %A Sharpe, Arlene H %A Shaw, Stanley Y %A Xavier, Ramnik J %X The balance between Th17 and T regulatory (Treg) cells critically modulates immune homeostasis, with an inadequate Treg response contributing to inflammatory disease. Using an unbiased chemical biology approach, we identified a novel role for the dual specificity tyrosine-phosphorylation-regulated kinase DYRK1A in regulating this balance. Inhibition of DYRK1A enhances Treg differentiation and impairs Th17 differentiation without affecting known pathways of Treg/Th17 differentiation. Thus, DYRK1A represents a novel mechanistic node at the branch point between commitment to either Treg or Th17 lineages. Importantly, both Treg cells generated using the DYRK1A inhibitor harmine and direct administration of harmine itself potently attenuate inflammation in multiple experimental models of systemic autoimmunity and mucosal inflammation. Our results identify DYRK1A as a physiologically relevant regulator of Treg cell differentiation and suggest a broader role for other DYRK family members in immune homeostasis. These results are discussed in the context of human diseases associated with dysregulated DYRK activity. %B Elife %V 4 %8 2015 %G eng %1 http://www.ncbi.nlm.nih.gov/pubmed/25998054?dopt=Abstract %R 10.7554/eLife.05920 %0 Journal Article %J Cell %D 2015 %T Melanoma Cell-Intrinsic PD-1 Receptor Functions Promote Tumor Growth. %A Kleffel, Sonja %A Posch, Christian %A Barthel, Steven R %A Mueller, Hansgeorg %A Schlapbach, Christoph %A Guenova, Emmanuella %A Elco, Christopher P %A Lee, Nayoung %A Juneja, Vikram R %A Zhan, Qian %A Lian, Christine G %A Thomi, Rahel %A Hoetzenecker, Wolfram %A Cozzio, Antonio %A Dummer, Reinhard %A Mihm, Martin C %A Flaherty, Keith T %A Frank, Markus H %A Murphy, George F %A Sharpe, Arlene H %A Kupper, Thomas S %A Schatton, Tobias %X Therapeutic antibodies targeting programmed cell death 1 (PD-1) activate tumor-specific immunity and have shown remarkable efficacy in the treatment of melanoma. Yet, little is known about tumor cell-intrinsic PD-1 pathway effects. Here, we show that murine and human melanomas contain PD-1-expressing cancer subpopulations and demonstrate that melanoma cell-intrinsic PD-1 promotes tumorigenesis, even in mice lacking adaptive immunity. PD-1 inhibition on melanoma cells by RNAi, blocking antibodies, or mutagenesis of melanoma-PD-1 signaling motifs suppresses tumor growth in immunocompetent, immunocompromised, and PD-1-deficient tumor graft recipient mice. Conversely, melanoma-specific PD-1 overexpression enhances tumorigenicity, as does engagement of melanoma-PD-1 by its ligand, PD-L1, whereas melanoma-PD-L1 inhibition or knockout of host-PD-L1 attenuate growth of PD-1-positive melanomas. Mechanistically, the melanoma-PD-1 receptor modulates downstream effectors of mTOR signaling. Our results identify melanoma cell-intrinsic functions of the PD-1:PD-L1 axis in tumor growth and suggest that blocking melanoma-PD-1 might contribute to the striking clinical efficacy of anti-PD-1 therapy. %B Cell %V 162 %P 1242-56 %8 2015 Sep 10 %G eng %N 6 %1 http://www.ncbi.nlm.nih.gov/pubmed/26359984?dopt=Abstract %R 10.1016/j.cell.2015.08.052 %0 Journal Article %J Trends Immunol %D 2015 %T T follicular regulatory cells in the regulation of B cell responses. %A Sage, Peter T %A Sharpe, Arlene H %X High affinity antibodies result from interactions between B cells and T follicular helper (Tfh) cells in germinal centers (GCs). Recent studies have identified an effector subset of T regulatory cells termed T follicular regulatory (Tfr) cells that specifically controls GC responses by suppressing Tfh and B cells. The discovery of Tfr cells has shed new light on pathways regulating humoral immunity that enable potent and specific responses to pathogens while restricting autoimmunity. Here, we review the current understanding of the cellular and molecular mechanisms underlying the differentiation and function of Tfr cells. In this context we discuss recent insights into the role of Tfh cells in disease, how this knowledge may be translated therapeutically, and important areas of further research. %B Trends Immunol %V 36 %P 410-8 %8 2015 Jul %G eng %N 7 %1 http://www.ncbi.nlm.nih.gov/pubmed/26091728?dopt=Abstract %R 10.1016/j.it.2015.05.005