Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Cell Rep ; 43(3): 113868, 2024 Mar 26.
Article in English | MEDLINE | ID: mdl-38421868

ABSTRACT

Modeling tumor metabolism in vitro remains challenging. Here, we used galactose as an in vitro tool compound to mimic glycolytic limitation. In contrast to the established idea that high glycolytic flux reduces pyruvate kinase isozyme M2 (PKM2) activity to support anabolic processes, we have discovered that glycolytic limitation also affects PKM2 activity. Surprisingly, despite limited carbon availability and energetic stress, cells induce a near-complete block of PKM2 to divert carbons toward serine metabolism. Simultaneously, TCA cycle flux is sustained, and oxygen consumption is increased, supported by glutamine. Glutamine not only supports TCA cycle flux but also serine synthesis via distinct mechanisms that are directed through PKM2 inhibition. Finally, deleting mitochondrial one-carbon (1C) cycle reversed the PKM2 block, suggesting a potential formate-dependent crosstalk that coordinates mitochondrial 1C flux and cytosolic glycolysis to support cell survival and proliferation during nutrient-scarce conditions.


Subject(s)
Glutamine , Pyruvate Kinase , Pyruvate Kinase/metabolism , Glutamine/metabolism , Glycolysis , Carbon , Serine/metabolism
3.
Cell Rep ; 42(9): 113034, 2023 09 26.
Article in English | MEDLINE | ID: mdl-37651228

ABSTRACT

Metabolic rewiring is essential for cancer onset and progression. We previously showed that one-carbon metabolism-dependent formate production often exceeds the anabolic demand of cancer cells, resulting in formate overflow. Furthermore, we showed that increased extracellular formate concentrations promote the in vitro invasiveness of glioblastoma cells. Here, we substantiate these initial observations with ex vivo and in vivo experiments. We also show that exposure to exogeneous formate can prime cancer cells toward a pro-invasive phenotype leading to increased metastasis formation in vivo. Our results suggest that the increased local formate concentration within the tumor microenvironment can be one factor to promote metastases. Additionally, we describe a mechanistic interplay between formate-dependent increased invasiveness and adaptations of lipid metabolism and matrix metalloproteinase activity. Our findings consolidate the role of formate as pro-invasive metabolite and warrant further research to better understand the interplay between formate and lipid metabolism.


Subject(s)
Glioblastoma , Lipid Metabolism , Humans , Formates , Neoplasm Invasiveness , Tumor Microenvironment
4.
Nat Commun ; 13(1): 2699, 2022 05 16.
Article in English | MEDLINE | ID: mdl-35577770

ABSTRACT

Metastasis is the most common cause of death in cancer patients. Canonical drugs target mainly the proliferative capacity of cancer cells, which leaves slow-proliferating, persistent cancer cells unaffected. Metabolic determinants that contribute to growth-independent functions are still poorly understood. Here we show that antifolate treatment results in an uncoupled and autarkic mitochondrial one-carbon (1C) metabolism during cytosolic 1C metabolism impairment. Interestingly, antifolate dependent growth-arrest does not correlate with decreased migration capacity. Therefore, using methotrexate as a tool compound allows us to disentangle proliferation and migration to profile the metabolic phenotype of migrating cells. We observe that increased serine de novo synthesis (SSP) supports mitochondrial serine catabolism and inhibition of SSP using the competitive PHGDH-inhibitor BI-4916 reduces cancer cell migration. Furthermore, we show that sole inhibition of mitochondrial serine catabolism does not affect primary breast tumor growth but strongly inhibits pulmonary metastasis. We conclude that mitochondrial 1C metabolism, despite being dispensable for proliferative capacities, confers an advantage to cancer cells by supporting their motility potential.


Subject(s)
Breast Neoplasms , Folic Acid Antagonists , Breast Neoplasms/metabolism , Carbon Cycle , Cell Line, Tumor , Cell Movement , Cell Proliferation , Female , Humans , Mitochondria/metabolism , Serine/metabolism
5.
Nat Metab ; 4(4): 458-475, 2022 04.
Article in English | MEDLINE | ID: mdl-35437333

ABSTRACT

The gut microbiome is a key player in the immunomodulatory and protumorigenic microenvironment during colorectal cancer (CRC), as different gut-derived bacteria can induce tumour growth. However, the crosstalk between the gut microbiome and the host in relation to tumour cell metabolism remains largely unexplored. Here we show that formate, a metabolite produced by the CRC-associated bacterium Fusobacterium nucleatum, promotes CRC development. We describe molecular signatures linking CRC phenotypes with Fusobacterium abundance. Cocultures of F. nucleatum with patient-derived CRC cells display protumorigenic effects, along with a metabolic shift towards increased formate secretion and cancer glutamine metabolism. We further show that microbiome-derived formate drives CRC tumour invasion by triggering AhR signalling, while increasing cancer stemness. Finally, F. nucleatum or formate treatment in mice leads to increased tumour incidence or size, and Th17 cell expansion, which can favour proinflammatory profiles. Moving beyond observational studies, we identify formate as a gut-derived oncometabolite that is relevant for CRC progression.


Subject(s)
Colorectal Neoplasms , Gastrointestinal Microbiome , Animals , Bacteria , Colorectal Neoplasms/metabolism , Formates , Fusobacterium nucleatum , Humans , Mice , Tumor Microenvironment
6.
Article in English | MEDLINE | ID: mdl-32117809

ABSTRACT

Elucidating key factors that regulate immune-mediated pathology in vivo is critical for developing improved strategies to treat autoimmune disease and cancer. NK cells can exhibit regulatory functions against CD8+ T cells following viral infection. Here we show that while low doses of lymphocytic choriomeningitis virus (LCMV-WE) can readily induce strong CD8+ T cell responses and diabetes in mice expressing the LCMV glycoprotein on ß-islet cells (RIP-GP mice), hyperglycemia does not occur after infection with higher doses of LCMV. High-dose LCMV infection induced an impaired CD8+ T cell response, which coincided with increased NK cell activity during early time points following infection. Notably, we observed increased NKp46 expression on NK cells during infection with higher doses, which resulted in an NK cell dependent suppression of T cells. Accordingly, depletion with antibodies specific for NK1.1 as well as NKp46 deficiency (Ncr1gfp/gfp mice) could restore CD8+ T cell immunity and permitted the induction of diabetes even following infection of RIP-GP mice with high-dose LCMV. Therefore, we identify conditions where innate lymphoid cells can play a regulatory role and interfere with CD8+ T cell mediated tissue specific pathology using an NKp46 dependent mechanism.


Subject(s)
Lymphocytic Choriomeningitis , Animals , Autoimmunity , CD8-Positive T-Lymphocytes , Immunity, Innate , Killer Cells, Natural , Mice , Mice, Inbred C57BL
7.
Autophagy ; 16(8): 1436-1452, 2020 08.
Article in English | MEDLINE | ID: mdl-31775562

ABSTRACT

In solid tumors, cancer stem cells (CSCs) or tumor-initiating cells (TICs) are often found in hypoxic niches. Nevertheless, the influence of hypoxia on TICs is poorly understood. Using previously established, TIC-enrichedpatient-derived colorectal cancer (CRC) cultures, we show that hypoxia increases the self-renewal capacity of TICs while inducing proliferation arrest in their more differentiated counterpart cultures. Gene expression data revealed macroautophagy/autophagy as one of the major pathways induced by hypoxia in TICs. Interestingly, hypoxia-induced autophagy was found to induce phosphorylation of EZR (ezrin) at Thr567 residue, which could be reversed by knocking down ATG5, BNIP3, BNIP3L, or BECN1. Furthermore, we identified PRKCA/PKCα as a potential kinase involved in hypoxia-induced autophagy-mediated TIC self-renewal. Genetic targeting of autophagy or pharmacological inhibition of PRKC/PKC and EZR resulted in decreased tumor-initiating potential of TICs. In addition, we observed significantly reduced in vivo tumor initiation and growth after a stable knockdown of ATG5. Analysis of human CRC samples showed that p-EZR is often present in TICs located in the hypoxic and autophagic regions of the tumor. Altogether, our results establish the hypoxia-autophagy-PKC-EZR signaling axis as a novel regulatory mechanism of TIC self-renewal and CRC progression. Autophagy inhibition might thus represent a promising therapeutic strategy for cancer patients. ABBREVIATIONS: ATG: autophagy related; BECN1: beclin 1; BNIP3: BCL2 interacting protein 3; BNIP3L: BCL2 interacting protein 3 like; CQ: chloroquine; CSC: cancer stem cells; CRC: colorectal cancer; HIF1A/HIF-1α: hypoxia inducible factor 1 subunit alpha; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; PRKC/PKC: protein kinase C; SQSTM1/p62: sequestosome 1; TICs: tumor-initiating cells.


Subject(s)
Carcinogenesis/pathology , Colorectal Neoplasms/etiology , Colorectal Neoplasms/pathology , Cytoskeletal Proteins/metabolism , Disease Progression , Hypoxia/complications , Protein Kinase C/metabolism , Signal Transduction , Animals , Autophagosomes/metabolism , Autophagy , Autophagy-Related Protein 5/deficiency , Autophagy-Related Protein 5/metabolism , Cell Self Renewal , Colon/pathology , Humans , Mice, Inbred NOD , Mice, SCID , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Phenotype , Phosphorylation
8.
Front Immunol ; 10: 1848, 2019.
Article in English | MEDLINE | ID: mdl-31440242

ABSTRACT

Cancer cells utilize multiple mechanisms to evade and suppress anticancer immune responses creating a "cold" immunosuppressive tumor microenvironment. Oncolytic virotherapy is emerging as a promising approach to revert tumor immunosuppression and enhance the efficacy of other forms of immunotherapy. Growing evidence indicates that oncolytic viruses (OVs) act in a multimodal fashion, inducing immunogenic cell death and thereby eliciting robust anticancer immune responses. In this review, we summarize information about OV-mediated immune conversion of the tumor microenvironment. As a case study we focus on the rodent protoparvovirus H-1PV and its dual role as an oncolytic and immune modulatory agent. Potential strategies to improve H-1PV anticancer efficacy are also discussed.


Subject(s)
H-1 parvovirus/immunology , Neoplasms/immunology , Neoplasms/therapy , Oncolytic Virotherapy , Oncolytic Viruses/immunology , Tumor Microenvironment/immunology , Humans
9.
Sci Rep ; 8(1): 12179, 2018 08 15.
Article in English | MEDLINE | ID: mdl-30111770

ABSTRACT

Immune responses are critical for defense against pathogens. However, prolonged viral infection can result in defective T cell immunity, leading to chronic viral infection. We studied immune activation in response to arenavirus infection during cholestasis using bile duct ligation (BDL). We monitored T cell responses, virus load and liver pathology markers after infection with lymphocytic choriomeningitis virus (LCMV). BDL mice failed to induce protective anti-viral immunity against LCMV and consequently exhibited chronic viral infection. BDL mice exhibited reduced anti-viral T cell immunity as well as reduced type 1 interferon production early after LCMV infection. Consistently, the presence of serum from BDL mice reduced the responsiveness of dendritic cell (DC) and T cell cultures when compared to Sham controls. Following fractionation and mass spectrometry analyses of sera, we identified several serum factors to be upregulated following BDL including bilirubin, bile acids, 78 kDa Glucose regulated protein (GRP78) and liver enzymes. Bilirubin and GRP78 were capable of inhibiting DC and T cell activation. In this work, we demonstrate that liver damage mediated by cholestasis results in defective immune induction following arenavirus infection.


Subject(s)
Arenaviridae Infections/immunology , Cholestasis/immunology , Liver Diseases/immunology , Liver/immunology , Animals , Arenaviridae Infections/pathology , Arenavirus/immunology , Bile Ducts/immunology , Bile Ducts/pathology , CD4-Positive T-Lymphocytes/immunology , Cholestasis/pathology , Dendritic Cells/immunology , Endoplasmic Reticulum Chaperone BiP , Interferon Type I/immunology , Liver/pathology , Liver Diseases/pathology , Lymphocyte Activation/immunology , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/immunology , Mice , Mice, Inbred C57BL , Signal Transduction
10.
J Virol ; 92(3)2018 02 01.
Article in English | MEDLINE | ID: mdl-29142134

ABSTRACT

Innate immune activation is essential to mount an effective antiviral response and to prime adaptive immunity. Although a crucial role of CD169+ cells during vesicular stomatitis virus (VSV) infections is increasingly recognized, factors regulating CD169+ cells during viral infections remain unclear. Here, we show that tumor necrosis factor is produced by CD11b+ Ly6C+ Ly6G+ cells following infection with VSV. The absence of TNF or TNF receptor 1 (TNFR1) resulted in reduced numbers of CD169+ cells and in reduced type I interferon (IFN-I) production during VSV infection, with a severe disease outcome. Specifically, TNF triggered RelA translocation into the nuclei of CD169+ cells; this translocation was inhibited when the paracaspase MALT-1 was absent. Consequently, MALT1 deficiency resulted in reduced VSV replication, defective innate immune activation, and development of severe disease. These findings indicate that TNF mediates the maintenance of CD169+ cells and innate and adaptive immune activation during VSV infection.IMPORTANCE Over the last decade, strategically placed CD169+ metallophilic macrophages in the marginal zone of the murine spleen and lymph nodes (LN) have been shown to play a very important role in host defense against viral pathogens. CD169+ macrophages have been shown to activate innate and adaptive immunity via "enforced virus replication," a controlled amplification of virus particles. However, the factors regulating the CD169+ macrophages remain to be studied. In this paper, we show that after vesicular stomatitis virus infection, phagocytes produce tumor necrosis factor (TNF), which signals via TNFR1, and promote enforced virus replication in CD169+ macrophages. Consequently, lack of TNF or TNFR1 resulted in defective immune activation and VSV clearance.


Subject(s)
Interferon Type I/immunology , Macrophages/immunology , Tumor Necrosis Factor-alpha/immunology , Vesicular Stomatitis/immunology , Adaptive Immunity , Animals , Immunity, Innate , Macrophages/virology , Mice , Mice, Inbred C57BL , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/genetics , Receptors, Tumor Necrosis Factor, Type I/immunology , Sialic Acid Binding Ig-like Lectin 1 , Transcription Factor RelA/metabolism , Vesiculovirus/physiology , Virus Replication
11.
Sci Rep ; 7(1): 7938, 2017 08 11.
Article in English | MEDLINE | ID: mdl-28801579

ABSTRACT

The devastating consequences of hepatic failure include hepatic encephalopathy, a severe, life threatening impairment of neuronal function. Hepatic encephalopathy is caused by impaired hepatic clearance of NH4+. Cellular NH4+ uptake is accomplished mainly by the Na+,K+,2Cl- cotransporter. Here we show that hepatic clearance of NH4+ is impaired in TNFα deficient as well as TNFR1&TNFR2 double knockout mice, which both develop hyperammonemia. Despite impaired hepatic clearance of NH4+, TNFα deficient mice and TNFR1 deficient mice were protected against acute ammonia intoxication. While 54% of the wild-type mice and 60% of TNFR2 deficient mice survived an NH4+ load, virtually all TNFα deficient mice and TNFR1 deficient mice survived the treatment. Conversely, TNFα treatment of wild type mice sensitized the animals to the toxic effects of an NH4+ load. The protection of TNFα-deficient mice against an NH4+ load was paralleled by decreased cerebral expression of NKCC1. According to the present observations, inhibition of TNFα formation and/or NKCC1 may be strategies to favorably influence the clinical course of hepatic encephalopathy.


Subject(s)
Hyperammonemia/genetics , Receptors, Tumor Necrosis Factor, Type II/genetics , Receptors, Tumor Necrosis Factor, Type I/genetics , Solute Carrier Family 12, Member 2/metabolism , Tumor Necrosis Factor-alpha/deficiency , Ammonia/toxicity , Animals , Brain/metabolism , Hyperammonemia/metabolism , Liver/chemistry , Mice , Mice, Knockout , Solute Carrier Family 12, Member 2/genetics , Tumor Necrosis Factor-alpha/genetics , Up-Regulation
12.
Cell Physiol Biochem ; 39(4): 1271-80, 2016.
Article in English | MEDLINE | ID: mdl-27606466

ABSTRACT

BACKGROUND/AIMS: Viral infections represent a global health problem with the need for new viral therapies and better understanding of the immune response during infection. The most immediate and potent anti-viral defense mechanism is the production of type I interferon (IFN-I) which are activated rapidly following recognition of viral infection by host pathogen recognition receptors (PRR). The mechanisms of innate cellular signaling downstream of PRR activation remain to be fully understood. In the present study, we demonstrate that CASP2 and RIPK1 domain-containing adaptor with death domain (CRADD/RAIDD) is a critical component in type I IFN production. METHODS: The role of RAIDD during IFN-I production was investigated using western blot, shRNA mediated lentiviral knockdown, immunoprecipitation and IFN-I driven dual luciferase assay. RESULTS: Immunoprecipitation analysis revealed the molecular interaction of RAIDD with interferon regulatory factor 7 (IRF7) and its phosphorylating kinase IKKε. Using an IFN-4α driven dual luciferase analysis in RAIDD deficient cells, type I IFN activation by IKKε and IRF7 was dramatically reduced. Furthermore, deletion of either the caspase recruitment domain (CARD) or death domain (DD) of RAIDD inhibited IKKε and IRF7 mediated interferon-4α activation. CONCLUSION: We have identified that the adaptor molecule RAIDD coordinates IKKε and IRF7 interaction to ensure efficient expression of type I interferon.


Subject(s)
CRADD Signaling Adaptor Protein/genetics , I-kappa B Kinase/genetics , Interferon Regulatory Factor-7/genetics , Toll-Like Receptor 3/genetics , Animals , CRADD Signaling Adaptor Protein/immunology , Caspase Activation and Recruitment Domain , Gene Expression Regulation , Genes, Reporter , Genetic Vectors/chemistry , Genetic Vectors/metabolism , HEK293 Cells , Humans , I-kappa B Kinase/immunology , Interferon Regulatory Factor-7/immunology , Interferon-alpha/genetics , Interferon-alpha/immunology , Interferon-beta/genetics , Interferon-beta/immunology , Lentivirus/genetics , Lentivirus/metabolism , Luciferases/genetics , Luciferases/metabolism , Mice , Plasmids/chemistry , Plasmids/metabolism , Poly I-C/pharmacology , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Signal Transduction , Toll-Like Receptor 3/immunology
13.
Cell Physiol Biochem ; 39(2): 668-76, 2016.
Article in English | MEDLINE | ID: mdl-27442519

ABSTRACT

BACKGROUND/AIMS: Similar to apoptosis of nucleated cells, red blood cells (RBC) can undergo suicidal cell death - called eryptosis. It is characterized by cell shrinkage and phosphatidylserine translocation. Eryptosis is triggered by an increase of intracellular calcium concentration due to activation of nonselective cation channels. The cation channels and consequently eryptosis are inhibited by erythropoietin. Eryptotic RBC are engulfed by macrophages and thus rapidly cleared from circulating blood. In this study, we explored whether storage of RBC influences the rate of eryptosis. METHODS: Flow cytometry was employed to quantify phosphatidylserine exposing erythrocytes from annexin V binding and cytosolic Ca2+ activity from Fluo-3 fluorescence. Clearance of stored murine RBC was tested by injection of carboxyfluorescein succinimidyl ester (CFSE)-labelled erythrocytes. RESULTS: Storage for 42 days significantly increased the percentage of phosphatidylserine exposing and haemolytic erythrocytes, an effect blunted by removal of extracellular calcium. Phosphatidylserine exposure could be inhibited by addition of erythropoietin. Upon transfusion, the clearance of murine CFSE-labelled RBC from circulating blood was significantly higher following storage for 10 days when compared to 2 days of storage. CONCLUSION: Storage of RBC triggers eryptosis by Ca2+ and erythropoietin sensitive mechanisms.


Subject(s)
Apoptosis/physiology , Blood Preservation/methods , Eryptosis/physiology , Erythrocytes/metabolism , Animals , Apoptosis/drug effects , Biological Transport/drug effects , Calcium/metabolism , Cell Death/drug effects , Cell Death/physiology , Eryptosis/drug effects , Erythrocytes/chemistry , Erythrocytes/cytology , Erythropoietin/pharmacology , Flow Cytometry/methods , Fluoresceins/chemistry , Humans , Intracellular Space/metabolism , Mice, Inbred C57BL , Phosphatidylserines/metabolism , Succinimides/chemistry , Time Factors
14.
Cell Physiol Biochem ; 38(4): 1500-9, 2016.
Article in English | MEDLINE | ID: mdl-27050423

ABSTRACT

BACKGROUND/AIMS: In nucleated cells, bile acids may activate cation channels subsequently leading to entry of Ca2+. In erythrocytes, increase of cytosolic Ca2+ activity triggers eryptosis, the suicidal death of erythrocytes characterized by phosphatidylserine exposure at the cell surface and cell shrinkage. Eryptosis is triggered by bile duct ligation, an effect partially attributed to conjugated bilirubin. The present study explored, whether bile acids may stimulate eryptosis. METHODS: Phosphatidylserine exposing erythrocytes have been identified utilizing annexin V binding, cell volume estimated from forward scatter, cytosolic Ca2+ activity determined using Fluo-3 fluorescence, and ceramide abundance at the erythrocyte surface utilizing specific antibodies. RESULTS: The exposure of human erythrocytes to glycochenodesoxycholic (GCDC) and taurochenodesoxycholic (TCDC) acid was followed by a significant decrease of forward scatter and significant increase of Fluo-3 fluorescence, ceramide abundance as well as annexin V binding. The effect on annexin V binding was significantly blunted, but not abolished by removal of extracellular Ca2+. CONCLUSION: Bile acids stimulate suicidal cell death, an effect paralleled by and in part due to Ca2+ entry and ceramide. The bile acid induced eryptosis may in turn lead to accelerated clearance of circulating erythrocytes and, thus, may contribute to anemia in cholestatic patients.


Subject(s)
Bile Acids and Salts/toxicity , Eryptosis/drug effects , Aniline Compounds/chemistry , Aniline Compounds/metabolism , Calcium/metabolism , Cells, Cultured , Ceramides/metabolism , Cholagogues and Choleretics/pharmacology , Detergents/pharmacology , Erythrocytes/cytology , Erythrocytes/drug effects , Erythrocytes/metabolism , Flow Cytometry , Glycochenodeoxycholic Acid/toxicity , Hemolysis/drug effects , Humans , Phosphatidylserines/metabolism , Taurochenodeoxycholic Acid/toxicity , Xanthenes/chemistry , Xanthenes/metabolism
15.
Proc Natl Acad Sci U S A ; 112(17): 5521-6, 2015 Apr 28.
Article in English | MEDLINE | ID: mdl-25870278

ABSTRACT

Urea cycle defects and acute or chronic liver failure are linked to systemic hyperammonemia and often result in cerebral dysfunction and encephalopathy. Although an important role of the liver in ammonia metabolism is widely accepted, the role of ammonia metabolizing pathways in the liver for maintenance of whole-body ammonia homeostasis in vivo remains ill-defined. Here, we show by generation of liver-specific Gln synthetase (GS)-deficient mice that GS in the liver is critically involved in systemic ammonia homeostasis in vivo. Hepatic deletion of GS triggered systemic hyperammonemia, which was associated with cerebral oxidative stress as indicated by increased levels of oxidized RNA and enhanced protein Tyr nitration. Liver-specific GS-deficient mice showed increased locomotion, impaired fear memory, and a slightly reduced life span. In conclusion, the present observations highlight the importance of hepatic GS for maintenance of ammonia homeostasis and establish the liver-specific GS KO mouse as a model with which to study effects of chronic hyperammonemia.


Subject(s)
Glutamate-Ammonia Ligase/metabolism , Hyperammonemia/enzymology , Liver/enzymology , Animals , Behavior, Animal , Brain/metabolism , Brain/pathology , Brain/physiopathology , Disease Models, Animal , Gene Targeting , Glutamate-Ammonia Ligase/genetics , Hyperammonemia/genetics , Hyperammonemia/pathology , Hyperammonemia/physiopathology , Liver/metabolism , Liver/physiopathology , Locomotion , Memory , Mice , Mice, Knockout , Oxidative Stress/genetics
16.
Nat Commun ; 6: 6217, 2015 Feb 18.
Article in English | MEDLINE | ID: mdl-25692415

ABSTRACT

B cells are essential for antiviral immune defence because they produce neutralizing antibodies, present antigen and maintain the lymphoid architecture. Here we show that intrinsic signalling of CEACAM1 is essential for generating efficient B-cell responses. Although CEACAM1 exerts limited influence on the proliferation of B cells, expression of CEACAM1 induces survival of proliferating B cells via the BTK/Syk/NF-κB-axis. The absence of this signalling cascade in naive Ceacam1(-/-) mice limits the survival of B cells. During systemic infection with cytopathic vesicular stomatitis virus, Ceacam1(-/-) mice can barely induce neutralizing antibody responses and die early after infection. We find, therefore, that CEACAM1 is a crucial regulator of B-cell survival, influencing B-cell numbers and protective antiviral antibody responses.


Subject(s)
Antibodies, Viral/immunology , B-Lymphocytes/cytology , Carcinoembryonic Antigen/physiology , Animals , Antibodies, Neutralizing/immunology , B-Lymphocytes/immunology , Bone Marrow/metabolism , Bone Marrow Cells/cytology , Cell Differentiation , Cell Proliferation , Cell Separation , Cell Survival , Flow Cytometry , Gene Expression Regulation , Immunoglobulin G/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phosphorylation , Signal Transduction , Spleen/metabolism , Vesiculovirus
17.
Eur J Immunol ; 45(2): 418-27, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25393615

ABSTRACT

STAT3 is a critical transcription factor activated downstream of cytokine signaling and is integral for the function of multiple immune cell types. Human mutations in STAT3 cause primary immunodeficiency resulting in impaired control of a variety of infections, including reactivation of latent viruses. In this study, we investigate how T-cell functions of STAT3 contribute to responses to viral infection by inducing chronic lymphocytic choriomeningitis virus (LCMV) infection in mice lacking STAT3 specifically in T cells. Although mice with conditional disruption of STAT3 in T cells were able to mount early responses to viral infection similar to control animals, including expansion of effector T cells, we found generation of T-follicular helper (Tfh) cells to be impaired. As a result, STAT3 T cell deficient mice produced attenuated germinal center reactions, and did not accumulate bone marrow virus specific IgG-secreting cells, resulting in failure to maintain levels of virus-specific IgG or mount neutralizing responses to LCMV in the serum. These effects were associated with reduced control of viral replication and prolonged infection. Our results demonstrate the importance of STAT3 in T cells for the generation of functional long-term humoral immunity to viral infections.


Subject(s)
Antibodies, Viral/biosynthesis , Immunity, Humoral , Immunoglobulin G/biosynthesis , Lymphocytic Choriomeningitis/immunology , STAT3 Transcription Factor/immunology , T-Lymphocytes, Helper-Inducer/pathology , Animals , B-Lymphocytes/immunology , B-Lymphocytes/pathology , B-Lymphocytes/virology , Chronic Disease , Gene Expression , Immunophenotyping , Lymphocytic Choriomeningitis/genetics , Lymphocytic Choriomeningitis/pathology , Lymphocytic Choriomeningitis/virology , Lymphocytic choriomeningitis virus/immunology , Mice , Mice, Knockout , STAT3 Transcription Factor/deficiency , STAT3 Transcription Factor/genetics , Signal Transduction , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/pathology , T-Lymphocytes, Cytotoxic/virology , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/virology , Virus Replication
18.
Proc Natl Acad Sci U S A ; 110(7): 2593-8, 2013 Feb 12.
Article in English | MEDLINE | ID: mdl-23359703

ABSTRACT

Rapid activation of immune responses is necessary for antibacterial defense, but excessive immune activation can result in life-threatening septic shock. Understanding how these processes are balanced may provide novel therapeutic potential in treating inflammatory disease. Fc receptors are crucial for innate immune activation. However, the role of the putative Fc receptor for IgM, known as Toso/Faim3, has to this point been unclear. In this study, we generated Toso-deficient mice and used them to uncover a critical regulatory function of Toso in innate immune activation. Development of innate immune cells was intact in the absence of Toso, but Toso-deficient neutrophils exhibited more reactive oxygen species production and reduced phagocytosis of pathogens compared with controls. Cytokine production was also decreased in Toso(-/-) mice compared with WT animals, rendering them resistant to septic shock induced by lipopolysaccharide. However, Toso(-/-) mice also displayed limited cytokine production after infection with the bacterium Listeria monocytogenes that was correlated with elevated presence of Listeria throughout the body. Accordingly, Toso(-/-) mice succumbed to infections of L. monocytogenes, whereas WT mice successfully eliminated the infection. Taken together, our data reveal Toso to be a unique regulator of innate immune responses during bacterial infection and septic shock.


Subject(s)
Carrier Proteins/immunology , Granulocytes/immunology , Immunity, Innate/immunology , Listeriosis/immunology , Macrophage Activation/immunology , Membrane Proteins/immunology , Monocytes/immunology , Analysis of Variance , Animals , Carrier Proteins/genetics , Crosses, Genetic , Cytokines/metabolism , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Immunoblotting , Membrane Proteins/genetics , Mice , Mice, Knockout , Peroxidase/metabolism , Phagocytosis/immunology , Reactive Oxygen Species/metabolism , Real-Time Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...