Try a new search

Format these results:

Searched for:

person:vaethm01

in-biosketch:yes

Total Results:

41


A new form of anhidrotic ectodermal dysplasia with immunodeficiency caused by abolished store-operated Ca2+ entry [Meeting Abstract]

Cuk, M; Lian, J; Kahlfuss, S; Kozhaya, L; Vaeth, M; Rieux-Laucat, F; Picard, C; Benson, M J; Jakovcevic, A; Bilic, K; Martinac, I; Stathopulos, P; Kacskovics, I; Vraetz, T; Speckmann, C; Ehl, S; Issekutz, T; Unutmaz, D; Feske, S
Calcium signaling is fundamental to many cellular processes. An important pathway for increasing intracellular Ca2+ levels is store-operated Ca2+ entry (SOCE) regulated by stromal interaction molecule (STIM1-2), and Ca2+ channels formed by ORAI1-3 proteins. Mutations in the ORAI1 and STIM1 genes that abolish SOCE cause a combined immunodeficiency (CID) syndrome that is accompanied by autoimmunity and nonimmunologic symptoms. We present patients with Anhidrotic Ectodermal Dysplasia with Immunodeficiency (EDA-ID) caused by novel homozygous p.V181SfsX8, p.L194P, and p.G98R mutations in the ORAI1 gene that suppressed ORAI1 protein expression and SOCE in the patients' lymphocytes and fibroblasts. A unifying feature of patients with null mutations in ORAI1 is EDA. Anhidrosis was present in patients P1-P4 and confirmed by pilocarpin iontophoresis. Patients had dry and exfoliate skin. They showed signs of heat intolerance and thermoregulatory instability characterized by several attacks of facial flushing accompanied by tachycardia, tachypnea, and hypertension. A skin biopsy showed the presence of eccrine sweat glands in the dermis demonstrating that anhidrosis is not due to a defect in sweat gland development. Recently, we reported that sweat glands require SOCE for opening of the Ca2+-activated chloride channel TMEM16A and thus chloride secretion and sweat production, pointing that anhidrosis in ORAI1-deficient patients could be functional. ORAI1-deficient patients had severe enamel defects diagnosed as hypocalcified amelogenesis imperfecta type III. In contrast, patients with EDA-ID caused by NF-kB signaling defects also have a tooth defect, which is characterized by hypodontia and conical teeth and thus is morphologically easily distinguishable from the enamel defects in ORAI1-deficient patients. ORAI1-deficient patients showed thin and brittle hair. To date, the diagnosis of EDA-ID is limited to patients with defects in NF-kB signaling who are prone to infections with mycobacteria, P.jirovecii, Candida albicans, and, most frequently, pyogenic bacteria caused by hypogammaglobulinemia and failure to mount a specific antibody response to polysaccharide antigens. In contrast, ORAI1-deficient patients are susceptible to an overlapping spectrum of pathogens, but they are also prone to viral infections, including CMV, EBV, RSV, and rotavirus. In addition, AIHA and autoimmune thrombocytopenia are also common in SOCE deficient patients but not NF-kB; instead, patients with NF-kB defects can have inflammatory bowel disease (NF-kB essential modulator colitis). Here we propose that mutations in ORAI1 that abolish SOCE constitute a new form of EDA-ID and are an important differential diagnosis of EDA-ID caused by defects in NF-kB signaling
EMBASE:630602108
ISSN: 1663-2826
CID: 4286172

Ion channelopathies of the immune system

Vaeth, Martin; Feske, Stefan
Ion channels and transporters move ions across membrane barriers and are essential for a host of cell functions in many organs. They conduct K+, Na+ and Cl-, which are essential for regulating the membrane potential, H+ to control intracellular and extracellular pH and divalent cations such as Ca2+, Mg2+ and Zn2+, which function as second messengers and cofactors for many proteins. Inherited channelopathies due to mutations in ion channels or their accessory proteins cause a variety of diseases in the nervous, cardiovascular and other tissues, but channelopathies that affect immune function are not as well studied. Mutations in ORAI1 and STIM1 genes that encode the Ca2+ release-activated Ca2+ (CRAC) channel in immune cells, the Mg2+ transporter MAGT1 and the Cl- channel LRRC8A all cause immunodeficiency with increased susceptibility to infection. Mutations in the Zn2+ transporters SLC39A4 (ZIP4) and SLC30A2 (ZnT2) result in nutritional Zn2+ deficiency and immune dysfunction. These channels, however, only represent a fraction of ion channels that regulate immunity as demonstrated by immune dysregulation in channel knockout mice. The immune system itself can cause acquired channelopathies that are associated with a variety of diseases of nervous, cardiovascular and endocrine systems resulting from autoantibodies binding to ion channels. These autoantibodies highlight the therapeutic potential of functional anti-ion channel antibodies that are being developed for the treatment of autoimmune, inflammatory and other diseases.
PMCID:6004246
PMID: 29635109
ISSN: 1879-0372
CID: 3037292

NFAT control of immune function: New Frontiers for an Abiding Trooper

Vaeth, Martin; Feske, Stefan
Nuclear factor of activated T cells (NFAT) was first described almost three decades ago as a Ca2+/calcineurin-regulated transcription factor in T cells. Since then, a large body of research uncovered the regulation and physiological function of different NFAT homologues in the immune system and many other tissues. In this review, we will discuss novel roles of NFAT in T cells, focusing mainly on its function in humoral immune responses, immunological tolerance, and the regulation of immune metabolism.
PMCID:5840618
PMID: 29568499
ISSN: 2046-1402
CID: 3001072

Store-Operated Ca2+ Entry Controls Clonal Expansion of T Cells through Metabolic Reprogramming

Vaeth, Martin; Maus, Mate; Klein-Hessling, Stefan; Freinkman, Elizaveta; Yang, Jun; Eckstein, Miriam; Cameron, Scott; Turvey, Stuart E; Serfling, Edgar; Berberich-Siebelt, Friederike; Possemato, Richard; Feske, Stefan
Store-operated Ca2+ entry (SOCE) is the main Ca2+ influx pathway in lymphocytes and is essential for T cell function and adaptive immunity. SOCE is mediated by Ca2+ release-activated Ca2+ (CRAC) channels that are activated by stromal interaction molecule (STIM) 1 and STIM2. SOCE regulates many Ca2+-dependent signaling molecules, including calcineurin, and inhibition of SOCE or calcineurin impairs antigen-dependent T cell proliferation. We here report that SOCE and calcineurin regulate cell cycle entry of quiescent T cells by controlling glycolysis and oxidative phosphorylation. SOCE directs the metabolic reprogramming of naive T cells by regulating the expression of glucose transporters, glycolytic enzymes, and metabolic regulators through the activation of nuclear factor of activated T cells (NFAT) and the PI3K-AKT kinase-mTOR nutrient-sensing pathway. We propose that SOCE controls a critical "metabolic checkpoint" at which T cells assess adequate nutrient supply to support clonal expansion and adaptive immune responses.
PMCID:5683398
PMID: 29030115
ISSN: 1097-4180
CID: 2742062

NFATc1 controls the cytotoxicity of CD8+ T cells

Klein-Hessling, Stefan; Muhammad, Khalid; Klein, Matthias; Pusch, Tobias; Rudolf, Ronald; Floter, Jessica; Qureischi, Musga; Beilhack, Andreas; Vaeth, Martin; Kummerow, Carsten; Backes, Christian; Schoppmeyer, Rouven; Hahn, Ulrike; Hoth, Markus; Bopp, Tobias; Berberich-Siebelt, Friederike; Patra, Amiya; Avots, Andris; Muller, Nora; Schulze, Almut; Serfling, Edgar
Cytotoxic T lymphocytes are effector CD8+ T cells that eradicate infected and malignant cells. Here we show that the transcription factor NFATc1 controls the cytotoxicity of mouse cytotoxic T lymphocytes. Activation of Nfatc1 -/- cytotoxic T lymphocytes showed a defective cytoskeleton organization and recruitment of cytosolic organelles to immunological synapses. These cells have reduced cytotoxicity against tumor cells, and mice with NFATc1-deficient T cells are defective in controlling Listeria infection. Transcriptome analysis shows diminished RNA levels of numerous genes in Nfatc1 -/- CD8+ T cells, including Tbx21, Gzmb and genes encoding cytokines and chemokines, and genes controlling glycolysis. Nfatc1 -/- , but not Nfatc2 -/- CD8+ T cells have an impaired metabolic switch to glycolysis, which can be restored by IL-2. Genome-wide ChIP-seq shows that NFATc1 binds many genes that control cytotoxic T lymphocyte activity. Together these data indicate that NFATc1 is an important regulator of cytotoxic T lymphocyte effector functions.NFAT nuclear translocation has been shown to be required for CD8+ T cell cytokine production in response to viral infection. Here the authors show NFATc1 controls the cytotoxicity and metabolic switching of activated CD8+ T cells required for optimal response to bacteria and tumor cells.
PMCID:5593830
PMID: 28894104
ISSN: 2041-1723
CID: 2701552

Store-operated Ca2+ entry controls ameloblast cell function and enamel development

Eckstein, Miriam; Vaeth, Martin; Fornai, Cinzia; Vinu, Manikandan; Bromage, Timothy G; Nurbaeva, Meerim K; Sorge, Jessica L; Coelho, Paulo G; Idaghdour, Youssef; Feske, Stefan; Lacruz, Rodrigo S
Loss-of-function mutations in stromal interaction molecule 1 (STIM1) impair the activation of Ca2+ release-activated Ca2+ (CRAC) channels and store-operated Ca2+ entry (SOCE), resulting in a disease syndrome called CRAC channelopathy that is characterized by severe dental enamel defects. The cause of these enamel defects has remained unclear given a lack of animal models. We generated Stim1/2K14cre mice to delete STIM1 and its homolog STIM2 in enamel cells. These mice showed impaired SOCE in enamel cells. Enamel in Stim1/2K14cre mice was hypomineralized with decreased Ca content, mechanically weak, and thinner. The morphology of SOCE-deficient ameloblasts was altered, showing loss of the typical ruffled border, resulting in mislocalized mitochondria. Global gene expression analysis of SOCE-deficient ameloblasts revealed strong dysregulation of several pathways. ER stress genes associated with the unfolded protein response were increased in Stim1/2-deficient cells, whereas the expression of components of the glutathione system were decreased. Consistent with increased oxidative stress, we found increased ROS production, decreased mitochondrial function, and abnormal mitochondrial morphology in ameloblasts of Stim1/2K14cre mice. Collectively, these data show that loss of SOCE in enamel cells has substantial detrimental effects on gene expression, cell function, and the mineralization of dental enamel.
PMCID:5358480
PMID: 28352661
ISSN: 2379-3708
CID: 2508342

ORAI2 modulates store-operated calcium entry and T cell-mediated immunity

Vaeth, Martin; Yang, Jun; Yamashita, Megumi; Zee, Isabelle; Eckstein, Miriam; Knosp, Camille; Kaufmann, Ulrike; Karoly Jani, Peter; Lacruz, Rodrigo S; Flockerzi, Veit; Kacskovics, Imre; Prakriya, Murali; Feske, Stefan
Store-operated Ca2+ entry (SOCE) through Ca2+ release-activated Ca2+ (CRAC) channels is critical for lymphocyte function and immune responses. CRAC channels are hexamers of ORAI proteins that form the channel pore, but the contributions of individual ORAI homologues to CRAC channel function are not well understood. Here we show that deletion of Orai1 reduces, whereas deletion of Orai2 increases, SOCE in mouse T cells. These distinct effects are due to the ability of ORAI2 to form heteromeric channels with ORAI1 and to attenuate CRAC channel function. The combined deletion of Orai1 and Orai2 abolishes SOCE and strongly impairs T cell function. In vivo, Orai1/Orai2 double-deficient mice have impaired T cell-dependent antiviral immune responses, and are protected from T cell-mediated autoimmunity and alloimmunity in models of colitis and graft-versus-host disease. Our study demonstrates that ORAI1 and ORAI2 form heteromeric CRAC channels, in which ORAI2 fine-tunes the magnitude of SOCE to modulate immune responses.
PMCID:5355949
PMID: 28294127
ISSN: 2041-1723
CID: 2488632

Store-operated Ca2+ entry regulates Ca2+-activated chloride channels and eccrine sweat gland function

Concepcion, Axel R; Vaeth, Martin; Wagner, Larry E 2nd; Eckstein, Miriam; Hecht, Lee; Yang, Jun; Crottes, David; Seidl, Maximilian; Shin, Hyosup P; Weidinger, Carl; Cameron, Scott; Turvey, Stuart E; Issekutz, Thomas; Meyts, Isabelle; Lacruz, Rodrigo S; Cuk, Mario; Yule, David I; Feske, Stefan
Eccrine sweat glands are essential for sweating and thermoregulation in humans. Loss-of-function mutations in the Ca2+ release-activated Ca2+ (CRAC) channel genes ORAI1 and STIM1 abolish store-operated Ca2+ entry (SOCE), and patients with these CRAC channel mutations suffer from anhidrosis and hyperthermia at high ambient temperatures. Here we have shown that CRAC channel-deficient patients and mice with ectodermal tissue-specific deletion of Orai1 (Orai1K14Cre) or Stim1 and Stim2 (Stim1/2K14Cre) failed to sweat despite normal sweat gland development. SOCE was absent in agonist-stimulated sweat glands from Orai1K14Cre and Stim1/2K14Cre mice and human sweat gland cells lacking ORAI1 or STIM1 expression. In Orai1K14Cre mice, abolishment of SOCE was associated with impaired chloride secretion by primary murine sweat glands. In human sweat gland cells, SOCE mediated by ORAI1 was necessary for agonist-induced chloride secretion and activation of the Ca2+-activated chloride channel (CaCC) anoctamin 1 (ANO1, also known as TMEM16A). By contrast, expression of TMEM16A, the water channel aquaporin 5 (AQP5), and other regulators of sweat gland function was normal in the absence of SOCE. Our findings demonstrate that Ca2+ influx via store-operated CRAC channels is essential for CaCC activation, chloride secretion, and sweat production in humans and mice.
PMCID:5096923
PMID: 27721237
ISSN: 0021-9738
CID: 2311942

Exogenous TNFR2 activation protects from acute GvHD via host T reg cell expansion

Chopra, Martin; Biehl, Marlene; Steinfatt, Tim; Brandl, Andreas; Kums, Juliane; Amich, Jorge; Vaeth, Martin; Kuen, Janina; Holtappels, Rafaela; Podlech, Jurgen; Mottok, Anja; Kraus, Sabrina; Jordan-Garrote, Ana-Laura; Bauerlein, Carina A; Brede, Christian; Ribechini, Eliana; Fick, Andrea; Seher, Axel; Polz, Johannes; Ottmuller, Katja J; Baker, Jeanette; Nishikii, Hidekazu; Ritz, Miriam; Mattenheimer, Katharina; Schwinn, Stefanie; Winter, Thorsten; Schafer, Viktoria; Krappmann, Sven; Einsele, Hermann; Muller, Thomas D; Reddehase, Matthias J; Lutz, Manfred B; Mannel, Daniela N; Berberich-Siebelt, Friederike; Wajant, Harald; Beilhack, Andreas
Donor CD4(+)Foxp3(+) regulatory T cells (T reg cells) suppress graft-versus-host disease (GvHD) after allogeneic hematopoietic stem cell transplantation (HCT [allo-HCT]). Current clinical study protocols rely on the ex vivo expansion of donor T reg cells and their infusion in high numbers. In this study, we present a novel strategy for inhibiting GvHD that is based on the in vivo expansion of recipient T reg cells before allo-HCT, exploiting the crucial role of tumor necrosis factor receptor 2 (TNFR2) in T reg cell biology. Expanding radiation-resistant host T reg cells in recipient mice using a mouse TNFR2-selective agonist before allo-HCT significantly prolonged survival and reduced GvHD severity in a TNFR2- and T reg cell-dependent manner. The beneficial effects of transplanted T cells against leukemia cells and infectious pathogens remained unaffected. A corresponding human TNFR2-specific agonist expanded human T reg cells in vitro. These observations indicate the potential of our strategy to protect allo-HCT patients from acute GvHD by expanding T reg cells via selective TNFR2 activation in vivo.
PMCID:4995078
PMID: 27526711
ISSN: 1540-9538
CID: 2681782

Identification of PGAM5 as a Mammalian Protein Histidine Phosphatase that Plays a Central Role to Negatively Regulate CD4+ T Cells

Panda, Saswati; Srivastava, Shekhar; Li, Zhai; Vaeth, Martin; Fuhs, Stephen R; Hunter, Tony; Skolnik, Edward Y
Whereas phosphorylation of serine, threonine, and tyrosine is exceedingly well characterized, the role of histidine phosphorylation in mammalian signaling is largely unexplored. Here we show that phosphoglycerate mutase family 5 (PGAM5) functions as a phosphohistidine phosphatase that specifically associates with and dephosphorylates the catalytic histidine on nucleoside diphosphate kinase B (NDPK-B). By dephosphorylating NDPK-B, PGAM5 negatively regulates CD4+ T cells by inhibiting NDPK-B-mediated histidine phosphorylation and activation of the K+ channel KCa3.1, which is required for TCR-stimulated Ca2+ influx and cytokine production. Using recently developed monoclonal antibodies that specifically recognize phosphorylation of nitrogens at the N1 (1-pHis) or N3 (3-pHis) positions of the imidazole ring, we detect for the first time phosphoisoform-specific regulation of histidine-phosphorylated proteins in vivo, and we link these modifications to TCR signaling. These results represent an important step forward in studying the role of histidine phosphorylation in mammalian biology and disease.
PMCID:5677525
PMID: 27453048
ISSN: 1097-4164
CID: 2191412