Try a new search

Format these results:

Searched for:

person:feskes01

in-biosketch:yes

Total Results:

131


PGE2 potentiates Orai1-mediated calcium entry contributing to peripheral sensitization

Wei, Dongyu; Birla, Hareram; Dou, Yannong; Mei, Yixiao; Huo, Xiaodong; Whitehead, Victoria; Osei-Owusu, Patrick; Feske, Stefan; Patafio, Giovanna; Tao, YuanXiang; Hu, Huijuan
Peripheral sensitization is one of the primary mechanisms underlying the pathogenesis of chronic pain. However, candidate molecules involved in peripheral sensitization remain incompletely understood. We have shown that store-operated calcium channels (SOCs) are expressed in the dorsal root ganglion (DRG) neurons. Whether SOCs contribute to peripheral sensitization associated with chronic inflammatory pain is elusive. Here we report that global or conditional deletion of Orai1 attenuates CFA-induced pain hypersensitivity in both male and female mice. To further establish the role of Orai1 in inflammatory pain, we performed calcium imaging and patch-clamp recordings in wild-type (WT) and Orai1 knockout (KO) DRG neurons. We found that SOC function was significantly enhanced in WT but not in Orai1 KO DRG neurons from CFA- and carrageenan-injected mice. Interestingly, the Orai1 protein level in L3/4 DRGs was not altered under inflammatory conditions. To understand how Orai1 is modulated under inflammatory pain conditions, prostaglandin E2 (PGE2) was used to sensitize DRG neurons. PGE2-induced increase in neuronal excitability and pain hypersensitivity was significantly reduced in Orai1 KO mice. PGE2-induced potentiation of SOCE was observed in WT, but not in Orai1 KO DRG neurons. This effect was attenuated by a PGE2 receptor 1 (EP1) antagonist and mimicked by an EP1 agonist. Inhibition of Gq/11, PKC or ERK abolished PGE2-induced SOCE increase, indicating PGE2-induced SOCE enhancement is mediated by EP1-mediated downstream cascade. These findings demonstrate that Orai1 plays an important role in peripheral sensitization. Our study also provides new insight into molecular mechanisms underlying PGE2-induced modulation of inflammatory pain.Significance Statement Store-operated calcium channel (SOC) Orai1 is expressed and functional in DRG neurons. Whether Orai1 contributes to peripheral sensitization is unclear. The present demonstrates that Orai1-mediated SOC function is enhanced in DRG neurons under inflammatory conditions. Global and conditional deletion of Orai1 attenuates CFA-induced pain hypersensitivity. We also demonstrate that Prostaglandin E2 (PGE2) potentiates SOC function in DRG neurons, through EP1-mediated signaling pathway. Importantly, we have found that Orai1 deficiency diminishes PGE2-induced SOC function increase and reduces PGE2-induced increase in neuronal excitability and pain hypersensitivity. These findings suggest that Orai1 plays an important role in peripheral sensitization associated with inflammatory pain. Our study reveals a novel mechanism underlying PGE2/EP1-induced peripheral sensitization. Orai1 may serve as a potential target for pathological pain.
PMID: 37952941
ISSN: 1529-2401
CID: 5610862

Orai3 and Orai1 mediate CRAC channel function and metabolic reprogramming in B cells

Emrich, Scott M; Yoast, Ryan E; Zhang, Xuexin; Fike, Adam J; Wang, Yin-Hu; Bricker, Kristen N; Tao, Anthony Y; Xin, Ping; Walter, Vonn; Johnson, Martin T; Pathak, Trayambak; Straub, Adam C; Feske, Stefan; Rahman, Ziaur S M; Trebak, Mohamed
The essential role of store-operated Ca2+ entry (SOCE) through Ca2+ release-activated Ca2+ (CRAC) channels in T cells is well established. In contrast, the contribution of individual Orai isoforms to SOCE and their downstream signaling functions in B cells are poorly understood. Here, we demonstrate changes in the expression of Orai isoforms in response to B cell activation. We show that both Orai3 and Orai1 mediate native CRAC channels in B cells. The combined loss of Orai1 and Orai3, but not Orai3 alone, impairs SOCE, proliferation and survival, nuclear factor of activated T cells (NFAT) activation, mitochondrial respiration, glycolysis, and the metabolic reprogramming of primary B cells in response to antigenic stimulation. Nevertheless, the combined deletion of Orai1 and Orai3 in B cells did not compromise humoral immunity to influenza A virus infection in mice, suggesting that other in vivo co-stimulatory signals can overcome the requirement of BCR-mediated CRAC channel function in B cells. Our results shed important new light on the physiological roles of Orai1 and Orai3 proteins in SOCE and the effector functions of B lymphocytes.
PMID: 36803766
ISSN: 2050-084x
CID: 5432252

Distinct roles of ORAI1 in T cell-mediated allergic airway inflammation and immunity to influenza A virus infection

Wang, Yin-Hu; Noyer, Lucile; Kahlfuss, Sascha; Raphael, Dimitrius; Tao, Anthony Y; Kaufmann, Ulrike; Zhu, Jingjie; Mitchell-Flack, Marisa; Sidhu, Ikjot; Zhou, Fang; Vaeth, Martin; Thomas, Paul G; Saunders, Sean P; Stauderman, Kenneth; Curotto de Lafaille, Maria A; Feske, Stefan
T cell activation and function depend on Ca2+ signals mediated by store-operated Ca2+ entry (SOCE) through Ca2+ release-activated Ca2+ (CRAC) channels formed by ORAI1 proteins. We here investigated how SOCE controls T cell function in pulmonary inflammation during a T helper 1 (TH1) cell-mediated response to influenza A virus (IAV) infection and TH2 cell-mediated allergic airway inflammation. T cell-specific deletion of Orai1 did not exacerbate pulmonary inflammation and viral burdens following IAV infection but protected mice from house dust mite-induced allergic airway inflammation. ORAI1 controlled the expression of genes including p53 and E2F transcription factors that regulate the cell cycle in TH2 cells in response to allergen stimulation and the expression of transcription factors and cytokines that regulate TH2 cell function. Systemic application of a CRAC channel blocker suppressed allergic airway inflammation without compromising immunity to IAV infection, suggesting that inhibition of SOCE is a potential treatment for allergic airway disease.
PMCID:9544339
PMID: 36206339
ISSN: 2375-2548
CID: 5351732

ORAI3 is dispensable for store-operated Ca2+ entry and immune responses by lymphocytes and macrophages

Wang, Liwei; Noyer, Lucile; Wang, Yin-Hu; Tao, Anthony Y; Li, Wenyi; Zhu, Jingjie; Saavedra, Pedro; Hoda, Syed T; Yang, Jun; Feske, Stefan
Ca2+ signals regulate the function of many immune cells and promote immune responses to infection, cancer, and autoantigens. Ca2+ influx in immune cells is mediated by store-operated Ca2+ entry (SOCE) that results from the opening of Ca2+ release-activated Ca2+ (CRAC) channels. The CRAC channel is formed by three plasma membrane proteins, ORAI1, ORAI2, and ORAI3. Of these, ORAI1 is the best studied and plays important roles in immune function. By contrast, the physiological role of ORAI3 in immune cells remains elusive. We show here that ORAI3 is expressed in many immune cells including macrophages, B cells, and T cells. To investigate ORAI3 function in immune cells, we generated Orai3-/- mice. The development of lymphoid and myeloid cells in the thymus and bone marrow was normal in Orai3-/- mice, as was the composition of immune cells in secondary lymphoid organs. Deletion of Orai3 did not affect SOCE in B cells and T cells but moderately enhanced SOCE in macrophages. Orai3-deficient macrophages, B cells, and T cells had normal effector functions in vitro. Immune responses in vivo, including humoral immunity (T cell dependent or independent) and antitumor immunity, were normal in Orai3-/- mice. Moreover, Orai3-/- mice showed no differences in susceptibility to septic shock, experimental autoimmune encephalomyelitis, or collagen-induced arthritis. We conclude that despite its expression in myeloid and lymphoid cells, ORAI3 appears to be dispensable or redundant for physiological and pathological immune responses mediated by these cells.
PMID: 35861698
ISSN: 1540-7748
CID: 5275992

Store-operated calcium entry controls innate and adaptive immune cell function in inflammatory bowel disease

Letizia, Marilena; Wang, Yin-Hu; Kaufmann, Ulrike; Gerbeth, Lorenz; Sand, Annegret; Brunkhorst, Max; Weidner, Patrick; Ziegler, Jörn Felix; Böttcher, Chotima; Schlickeiser, Stephan; Fernández, Camila; Yamashita, Megumi; Stauderman, Kenneth; Sun, Katherine; Kunkel, Désirée; Prakriya, Murali; Sanders, Ashley D; Siegmund, Britta; Feske, Stefan; Weidinger, Carl
Inflammatory bowel disease (IBD) is characterized by dysregulated intestinal immune responses. Using mass cytometry (CyTOF) to analyze the immune cell composition in the lamina propria (LP) of patients with ulcerative colitis (UC) and Crohn's disease (CD), we observed an enrichment of CD4+ effector T cells producing IL-17A and TNF, CD8+ T cells producing IFNγ, T regulatory (Treg) cells, and innate lymphoid cells (ILC). The function of these immune cells is regulated by store-operated Ca2+ entry (SOCE), which results from the opening of Ca2+ release-activated Ca2+ (CRAC) channels formed by ORAI and STIM proteins. We observed that the pharmacologic inhibition of SOCE attenuated the production of proinflammatory cytokines including IL-2, IL-4, IL-6, IL-17A, TNF, and IFNγ by human colonic T cells and ILCs, reduced the production of IL-6 by B cells and the production of IFNγ by myeloid cells, but had no effect on the viability, differentiation, and function of intestinal epithelial cells. T cell-specific deletion of CRAC channel genes in mice showed that Orai1, Stim1, and Stim2-deficient T cells have quantitatively distinct defects in SOCE, which correlate with gradually more pronounced impairment of cytokine production by Th1 and Th17 cells and the severity of IBD. Moreover, the pharmacologic inhibition of SOCE with a selective CRAC channel inhibitor attenuated IBD severity and colitogenic T cell function in mice. Our data indicate that SOCE inhibition may be a suitable new approach for the treatment of IBD.
PMID: 35919953
ISSN: 1757-4684
CID: 5288032

The function of Wtap in N6-adenosine methylation of mRNAs controls T cell receptor signaling and survival of T cells

Ito-Kureha, Taku; Leoni, Cristina; Borland, Kayla; Cantini, Giulia; Bataclan, Marian; Metzger, Rebecca N; Ammann, Gregor; Krug, Anne B; Marsico, Annalisa; Kaiser, Stefanie; Canzar, Stefan; Feske, Stefan; Monticelli, Silvia; König, Julian; Heissmeyer, Vigo
T cell antigen-receptor (TCR) signaling controls the development, activation and survival of T cells by involving several layers and numerous mechanisms of gene regulation. N6-methyladenosine (m6A) is the most prevalent messenger RNA modification affecting splicing, translation and stability of transcripts. In the present study, we describe the Wtap protein as essential for m6A methyltransferase complex function and reveal its crucial role in TCR signaling in mouse T cells. Wtap and m6A methyltransferase functions were required for the differentiation of thymocytes, control of activation-induced death of peripheral T cells and prevention of colitis by enabling gut RORγt+ regulatory T cell function. Transcriptome and epitranscriptomic analyses reveal that m6A modification destabilizes Orai1 and Ripk1 mRNAs. Lack of post-transcriptional repression of the encoded proteins correlated with increased store-operated calcium entry activity and diminished survival of T cells with conditional genetic inactivation of Wtap. These findings uncover how m6A modification impacts on TCR signal transduction and determines activation and survival of T cells.
PMID: 35879451
ISSN: 1529-2916
CID: 5276302

Cavβ1 regulates T cell expansion and apoptosis independently of voltage-gated Ca2+ channel function

Erdogmus, Serap; Concepcion, Axel R; Yamashita, Megumi; Sidhu, Ikjot; Tao, Anthony Y; Li, Wenyi; Rocha, Pedro P; Huang, Bonnie; Garippa, Ralph; Lee, Boram; Lee, Amy; Hell, Johannes W; Lewis, Richard S; Prakriya, Murali; Feske, Stefan
TCR stimulation triggers Ca2+ signals that are critical for T cell function and immunity. Several pore-forming α and auxiliary β subunits of voltage-gated Ca2+ channels (VGCC) were reported in T cells, but their mechanism of activation remains elusive and their contribution to Ca2+ signaling in T cells is controversial. We here identify CaVβ1, encoded by Cacnb1, as a regulator of T cell function. Cacnb1 deletion enhances apoptosis and impairs the clonal expansion of T cells after lymphocytic choriomeningitis virus (LCMV) infection. By contrast, Cacnb1 is dispensable for T cell proliferation, cytokine production and Ca2+ signaling. Using patch clamp electrophysiology and Ca2+ recordings, we are unable to detect voltage-gated Ca2+ currents or Ca2+ influx in human and mouse T cells upon depolarization with or without prior TCR stimulation. mRNAs of several VGCC α1 subunits are detectable in human (CaV3.3, CaV3.2) and mouse (CaV2.1) T cells, but they lack transcription of many 5' exons, likely resulting in N-terminally truncated and non-functional proteins. Our findings demonstrate that although CaVβ1 regulates T cell function, these effects are independent of VGCC channel activity.
PMCID:9018955
PMID: 35440113
ISSN: 2041-1723
CID: 5216862

The volume-regulated anion channel LRRC8C suppresses T cell function by regulating cyclic dinucleotide transport and STING-p53 signaling

Concepcion, Axel R; Wagner, Larry E 2nd; Zhu, Jingjie; Tao, Anthony Y; Yang, Jun; Khodadadi-Jamayran, Alireza; Wang, Yin-Hu; Liu, Menghan; Rose, Rebecca E; Jones, Drew R; Coetzee, William A; Yule, David I; Feske, Stefan
PMID: 35105987
ISSN: 1529-2908
CID: 5147322

Disrupting Roquin-1 interaction with Regnase-1 induces autoimmunity and enhances antitumor responses

Behrens, Gesine; Edelmann, Stephanie L; Raj, Timsse; Kronbeck, Nina; Monecke, Thomas; Davydova, Elena; Wong, Elaine H; Kifinger, Lisa; Giesert, Florian; Kirmaier, Martin E; Hohn, Christine; de Jonge, Laura S; Pisfil, Mariano Gonzalez; Fu, Mingui; Theurich, Sebastian; Feske, Stefan; Kawakami, Naoto; Wurst, Wolfgang; Niessing, Dierk; Heissmeyer, Vigo
Roquin and Regnase-1 proteins bind and post-transcriptionally regulate proinflammatory target messenger RNAs to maintain immune homeostasis. Either the sanroque mutation in Roquin-1 or loss of Regnase-1 cause systemic lupus erythematosus-like phenotypes. Analyzing mice with T cells that lack expression of Roquin-1, its paralog Roquin-2 and Regnase-1 proteins, we detect overlapping or unique phenotypes by comparing individual and combined inactivation. These comprised spontaneous activation, metabolic reprogramming and persistence of T cells leading to autoimmunity. Here, we define an interaction surface in Roquin-1 for binding to Regnase-1 that included the sanroque residue. Mutations in Roquin-1 impairing this interaction and cooperative regulation of targets induced T follicular helper cells, germinal center B cells and autoantibody formation. These mutations also improved the functionality of tumor-specific T cells by promoting their accumulation in the tumor and reducing expression of exhaustion markers. Our data reveal the physical interaction of Roquin-1 with Regnase-1 as a hub to control self-reactivity and effector functions in immune cell therapies.
PMID: 34811541
ISSN: 1529-2916
CID: 5063442

Straight from the channel's mouth: AKAP79 links Ca2+ influx through ORAI1 to NFAT activation

Noyer, Lucile; Feske, Stefan
PMID: 34481158
ISSN: 1532-1991
CID: 5011832