35 results on '"Lang, Karl Sebastian"'
Search Results
2. Das lymphozytäre Choriomeningitisvirus – Untersucht mittels eines Mausmodells für virusinduzierte Immunpathologie in der Leber
- Author
-
Lang, Karl Sebastian, primary
- Published
- 2021
- Full Text
- View/download PDF
3. Usp22 Deficiency Leads to Downregulation of PD-L1 and Pathological Activation of CD8+ T Cells and Causes Immunopathology in Response to Acute LCMV Infection
- Author
-
Friebus-Kardash, Justa, primary, Christ, Theresa Charlotte, additional, Dietlein, Nikolaus, additional, Elwy, Abdelrahman, additional, Abdelrahman, Hossam, additional, Holnsteiner, Lisa, additional, Hu, Zhongwen, additional, Rodewald, Hans-Reimer, additional, and Lang, Karl Sebastian, additional
- Published
- 2023
- Full Text
- View/download PDF
4. Usp22 Deficiency Switches Antiviral Response into Pathological CD8+ T Cell Activation via Downregulation of PD-L1
- Author
-
Friebus-Kardash, Justa, primary, Christ, Theresa Charlotte, additional, Dietlein, Nikolaus, additional, Elwy, Abdelrahman, additional, Abdelrahman, Hossam, additional, Holnsteiner, Lisa, additional, Hu, Zhongwen, additional, Rodewald3, Hans-Reimer, additional, and Lang, Karl Sebastian, additional
- Published
- 2023
- Full Text
- View/download PDF
5. Expression of Interferon Regulatory Factor 8 (IRF8) and Its Association with Infections in Dialysis Patients
- Author
-
Friebus-Kardash, Justa, primary, Kuang, Fei, additional, Peitz, Tobias, additional, Hamdan, Thamer A., additional, Eisenberger, Ute, additional, Boss, Kristina, additional, Kribben, Andreas, additional, Lang, Karl Sebastian, additional, and Jahn, Michael, additional
- Published
- 2023
- Full Text
- View/download PDF
6. Susceptibility of BAFF-var allele carriers to severe SLE with occurrence of lupus nephritis
- Author
-
Friebus-Kardash, Justa, Trendelenburg, Marten, Eisenberger, Ute, Ribi, Camillo, Chizzolini, Carlo, Huynh-Do, Uyen, Lang, Karl Sebastian, Wilde, Benjamin, Kribben, Andreas, Witzke, Oliver, Dolff, Sebastian, and Hardt, Cornelia
- Published
- 2019
- Full Text
- View/download PDF
7. Immune suppression of vaccine-induced CD8+ T-cell responses by gamma retrovirus envelope is mediated by interleukin-10-producing CD4+ T cells
- Author
-
Podschwadt, Philip, primary, Malyshkina, Anna, additional, Windmann, Sonja, additional, Papadamakis, Athanasios, additional, Kerkmann, Leonie, additional, Lapuente, Dennis, additional, Tenbusch, Matthias, additional, Lu, Mengji, additional, Schindler, Michael, additional, Lang, Karl Sebastian, additional, Hansen, Wiebke, additional, and Bayer, Wibke, additional
- Published
- 2022
- Full Text
- View/download PDF
8. Usp22 Deficiency Leads to Downregulation of PD-L1 and Pathological Activation of CD8 + T Cells and Causes Immunopathology in Response to Acute LCMV Infection.
- Author
-
Friebus-Kardash, Justa, Christ, Theresa Charlotte, Dietlein, Nikolaus, Elwy, Abdelrahman, Abdelrahman, Hossam, Holnsteiner, Lisa, Hu, Zhongwen, Rodewald, Hans-Reimer, and Lang, Karl Sebastian
- Subjects
T cells ,INTERFERON gamma ,TYPE I interferons ,DEUBIQUITINATING enzymes ,PROGRAMMED death-ligand 1 - Abstract
Ubiquitin-specific peptidase 22 (Usp22) cleaves ubiquitin moieties from numerous proteins, including histone H2B and transcription factors. Recently, it was reported that Usp22 acts as a negative regulator of interferon-dependent responses. In the current study, we investigated the role of Usp22 deficiency in acute viral infection with lymphocytic choriomeningitis virus (LCMV). We found that the lack of Usp22 on bone marrow-derived cells (Usp22
fl/fl Vav1-Cre mice) reduced the induction of type I and II interferons. A limited type I interferon response did not influence virus replication. However, restricted expression of PD-L1 led to increased frequencies of functional virus-specific CD8+ T cells and rapid death of Usp22-deficient mice. CD8+ T cell depletion experiments revealed that accelerated CD8+ T cells were responsible for enhanced lethality in Usp22 deficient mice. In conclusion, we found that the lack of Usp22 generated a pathological CD8+ T cell response, which gave rise to severe disease in mice. [ABSTRACT FROM AUTHOR]- Published
- 2023
- Full Text
- View/download PDF
9. The acid ceramidase/ceramide axis controls parasitemia in Plasmodium yoelii-infected mice by regulating erythropoiesis
- Author
-
Günther, Anne, primary, Hose, Matthias, additional, Abberger, Hanna, additional, Schumacher, Fabian, additional, Veith, Ylva, additional, Kleuser, Burkhard, additional, Matuschewski, Kai, additional, Lang, Karl Sebastian, additional, Gulbins, Erich, additional, Buer, Jan, additional, Westendorf, Astrid M, additional, and Hansen, Wiebke, additional
- Published
- 2022
- Full Text
- View/download PDF
10. Author response: The acid ceramidase/ceramide axis controls parasitemia in Plasmodium yoelii-infected mice by regulating erythropoiesis
- Author
-
Günther, Anne, primary, Hose, Matthias, additional, Abberger, Hanna, additional, Schumacher, Fabian, additional, Veith, Ylva, additional, Kleuser, Burkhard, additional, Matuschewski, Kai, additional, Lang, Karl Sebastian, additional, Gulbins, Erich, additional, Buer, Jan, additional, Westendorf, Astrid M, additional, and Hansen, Wiebke, additional
- Published
- 2022
- Full Text
- View/download PDF
11. SIRT1 ‐mediated deacetylation of FOXO3a transcription factor supports pro‐angiogenic activity of interferon‐deficient tumor‐associated neutrophils
- Author
-
Bordbari, Sharareh, primary, Mörchen, Britta, additional, Pylaeva, Ekaterina, additional, Siakaeva, Elena, additional, Spyra, Ilona, additional, Domnich, Maksim, additional, Droege, Freya, additional, Kanaan, Oliver, additional, Lang, Karl Sebastian, additional, Schadendorf, Dirk, additional, Lang, Stephan, additional, Helfrich, Iris, additional, and Jablonska, Jadwiga, additional
- Published
- 2021
- Full Text
- View/download PDF
12. Replication of Influenza A Virus in Secondary Lymphatic Tissue Contributes to Innate Immune Activation
- Author
-
Friedrich, Sarah-Kim, Schmitz, Rosa, Bergerhausen, Michael, Lang, Judith, Duhan, Vikas, Hardt, Cornelia, Tenbusch, Matthias, Prinz, Marco, Asano, Kenichi, Bhat, Hilal, Hamdan, Thamer A., Lang, Philipp Alexander, and Lang, Karl Sebastian
- Subjects
enforced viral replication ,Medizinische Fakultät » Universitätsklinikum Essen » Institut für Immunologie ,innate immune activation ,viruses ,Medizin ,Medicine ,ddc:610 ,Influenza virus ,Article - Abstract
The replication of viruses in secondary lymphoid organs guarantees sufficient amounts of pattern-recognition receptor ligands and antigens to activate the innate and adaptive immune system. Viruses with broad cell tropism usually replicate in lymphoid organs, however, whether a virus with a narrow tropism relies on replication in the secondary lymphoid organs to activate the immune system remains not well studied. In this study, we used the artificial intravenous route of infection to determine whether Influenza A virus (IAV) replication can occur in secondary lymphatic organs (SLO) and whether such replication correlates with innate immune activation. Indeed, we found that IAV replicates in secondary lymphatic tissue. IAV replication was dependent on the expression of Sialic acid residues in antigen-presenting cells and on the expression of the interferon-inhibitor UBP43 (Usp18). The replication of IAV correlated with innate immune activation, resulting in IAV eradication. The genetic deletion of Usp18 curbed IAV replication and limited innate immune activation. In conclusion, we found that IAV replicates in SLO, a mechanism which allows innate immune activation.
- Published
- 2021
13. CD107a⁺ (LAMP-1) Cytotoxic CD8⁺ T-Cells in Lupus Nephritis Patients
- Author
-
Wiechmann, Anika, Wilde, Benjamin, Tyczynski, Bartosz, Amann, Kerstin, Abdulahad, Wayel H., Kribben, Andreas, Lang, Karl Sebastian, Witzke, Oliver, Dolff, Sebastian, and Translational Immunology Groningen (TRIGR)
- Subjects
lupus nephritis ,cytotoxic T-cells ,Medizinische Fakultät » Universitätsklinikum Essen » Institut für Immunologie ,SLE ,Medizin ,Medizinische Fakultät » Universitätsklinikum Essen » Klinik für Infektiologie ,CD107a ,Medizinische Fakultät » Universitätsklinikum Essen » Klinik für Nephrologie ,immune system diseases ,Medicine ,ddc:610 ,LAMP-1 ,skin and connective tissue diseases ,Original Research - Abstract
Cytotoxic CD8(+) T-cells play a pivotal role in the pathogenesis of systemic lupus erythematosus (SLE). The aim of this study was to investigate the role of CD107a (LAMP-1) on cytotoxic CD8(+) T-cells in SLE-patients in particular with lupus nephritis. Peripheral blood of SLE-patients (n = 31) and healthy controls (n = 21) was analyzed for the expression of CD314 and CD107a by flow cytometry. Kidney biopsies of lupus nephritis patients were investigated for the presence of CD8(+) and C107a(+) cells by immunohistochemistry and immunofluorescence staining. The percentages of CD107a(+) on CD8(+) T-cells were significantly decreased in SLE-patients as compared to healthy controls (40.2 +/- 18.5% vs. 47.9 +/- 15.0%, p = 0.02). This was even more significant in SLE-patients with inactive disease. There was a significant correlation between the percentages of CD107a(+)CD8(+) T-cells and SLEDAI. The evaluation of lupus nephritis biopsies showed a significant number of CD107a(+)CD8(+) T-cells mainly located in the peritubular infiltrates. The intrarenal expression of CD107a(+) was significantly correlated with proteinuria. These results demonstrate that CD8(+) T-cells of patients with systemic lupus erythematosus have an altered expression of CD107a which seems to be associated with disease activity. The proof of intrarenal CD107a(+)CD8(+) suggests a role in the pathogenesis of lupus nephritis.
- Published
- 2021
14. Virotherapy in Germany—Recent Activities in Virus Engineering, Preclinical Development, and Clinical Studies
- Author
-
Nettelbeck, Dirk M., primary, Leber, Mathias F., additional, Altomonte, Jennifer, additional, Angelova, Assia, additional, Beil, Julia, additional, Berchtold, Susanne, additional, Delic, Maike, additional, Eberle, Jürgen, additional, Ehrhardt, Anja, additional, Engeland, Christine E., additional, Fechner, Henry, additional, Geletneky, Karsten, additional, Goepfert, Katrin, additional, Holm, Per Sonne, additional, Kochanek, Stefan, additional, Kreppel, Florian, additional, Krutzke, Lea, additional, Kühnel, Florian, additional, Lang, Karl Sebastian, additional, Marchini, Antonio, additional, Moehler, Markus, additional, Mühlebach, Michael D., additional, Naumann, Ulrike, additional, Nawroth, Roman, additional, Nüesch, Jürg, additional, Rommelaere, Jean, additional, Lauer, Ulrich M., additional, and Ungerechts, Guy, additional
- Published
- 2021
- Full Text
- View/download PDF
15. CD107a+ (LAMP-1) Cytotoxic CD8+ T-Cells in Lupus Nephritis Patients
- Author
-
Wiechmann, Anika, primary, Wilde, Benjamin, additional, Tyczynski, Bartosz, additional, Amann, Kerstin, additional, Abdulahad, Wayel H., additional, Kribben, Andreas, additional, Lang, Karl Sebastian, additional, Witzke, Oliver, additional, and Dolff, Sebastian, additional
- Published
- 2021
- Full Text
- View/download PDF
16. Usp18 Expression in CD169+ Macrophages is Important for Strong Immune Response after Vaccination with VSV-EBOV
- Author
-
Friedrich, Sarah-Kim, primary, Schmitz, Rosa, additional, Bergerhausen, Michael, additional, Lang, Judith, additional, Cham, Lamin B., additional, Duhan, Vikas, additional, Häussinger, Dieter, additional, Hardt, Cornelia, additional, Addo, Marylyn, additional, Prinz, Marco, additional, Asano, Kenichi, additional, Lang, Philipp Alexander, additional, and Lang, Karl Sebastian, additional
- Published
- 2020
- Full Text
- View/download PDF
17. SIRT1‐mediated deacetylation of FOXO3a transcription factor supports pro‐angiogenic activity of interferon‐deficient tumor‐associated neutrophils.
- Author
-
Bordbari, Sharareh, Mörchen, Britta, Pylaeva, Ekaterina, Siakaeva, Elena, Spyra, Ilona, Domnich, Maksim, Droege, Freya, Kanaan, Oliver, Lang, Karl Sebastian, Schadendorf, Dirk, Lang, Stephan, Helfrich, Iris, and Jablonska, Jadwiga
- Subjects
FORKHEAD transcription factors ,TRANSCRIPTION factors ,NEUTROPHILS ,TYPE I interferons ,DEACETYLATION ,POST-translational modification - Abstract
Angiogenesis plays an important role during tumor growth and metastasis. We could previously show that Type I interferon (IFN)‐deficient tumor‐associated neutrophils (TANs) show strong pro‐angiogenic activity, and stimulate tumor angiogenesis and growth. However, the exact mechanism responsible for their pro‐angiogenic shift is not clear. Here, we set out to delineate the molecular mechanism and factors regulating pro‐angiogenic properties of neutrophils in the context of Type I IFN availability. We demonstrate that neutrophils from IFN‐deficient (Ifnar1−/−) mice efficiently release pro‐angiogenic factors, such as VEGF, MMP9 or BV8, and thus significantly support the vascular normalization of tumors by increasing the maturation of perivascular cells. Mechanistically, we could show here that the expression of pro‐angiogenic factors in neutrophils is controlled by the transcription factor forkhead box protein O3a (FOXO3a), which activity depends on its post‐translational modifications, such as deacetylation or phosphorylation. In TANs isolated from Ifnar1−/− mice, we observe significantly elevated SIRT1, resulting in SIRT1‐mediated deacetylation of FOXO3a, its nuclear retention and activation. Activated FOXO3a supports in turn the transcription of pro‐angiogenic genes in TANs. In the absence of SIRT1, or after its inhibition in neutrophils, elevated kinase MEK/ERK and PI3K/AKT activity is observed, leading to FOXO3a phosphorylation, cytoplasmic transfer and inactivation. In summary, we have found that FOXO3a is a key transcription factor controlling the angiogenic switch of neutrophils. Post‐translational FOXO3a modifications regulate its transcriptional activity and, as a result, the expression of pro‐angiogenic factors supporting development of vascular network in growing tumors. Therefore, targeting FOXO3a activity could provide a novel strategy of antiangiogenic targeted therapy for cancer. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
18. Sofosbuvir activates EGFR-Dependent pathways in hepatoma cells with implications for liver-related pathological processes
- Author
-
Bojkova, Denisa, Westhaus, Sandra, Costa, Rui, Timmer, Lejla, Funkenberg, Nora, Korencak, Marek, Streeck, Hendrik, Vondran, Florian, Bröring, Ruth, Heinrichs, Stefan, Lang, Karl Sebastian, Ciesek, Sandra, Bojkova, Denisa, Westhaus, Sandra, Costa, Rui, Timmer, Lejla, Funkenberg, Nora, Korencak, Marek, Streeck, Hendrik, Vondran, Florian, Bröring, Ruth, Heinrichs, Stefan, Lang, Karl Sebastian, and Ciesek, Sandra
- Abstract
Direct acting antivirals (DAAs) revolutionized the therapy of chronic hepatitis C infection. However, unexpected high recurrence rates of hepatocellular carcinoma (HCC) after DAA treatment became an issue in patients with advanced cirrhosis and fibrosis. In this study, we aimed to investigate an impact of DAA treatment on the molecular changes related to HCC development and progression in hepatoma cell lines and primary human hepatocytes. We found that treatment with sofosbuvir (SOF), a backbone of DAA therapy, caused an increase in EGFR expression and phosphorylation. As a result, enhanced translocation of EGFR into the nucleus and transactivation of factors associated with cell cycle progression, B-MYB and Cyclin D1, was detected. Serine/threonine kinase profiling identified additional pathways, especially the MAPK pathway, also activated during SOF treatment. Importantly, the blocking of EGFR kinase activity by erlotinib during SOF treatment prevented all downstream events. Altogether, our findings suggest that SOF may have an impact on pathological processes in the liver via the induction of EGFR signaling. Notably, zidovudine, another nucleoside analogue, exerted a similar cell phenotype, suggesting that the observed effects may be induced by additional members of this drug class.
- Published
- 2020
19. HLA-A2 Restricted, Melanocyte-Specific CD8+ T Lymphocytes Detected in Vitiligo Patients are Related to Disease Activity and are Predominantly Directed Against MelanA/MART1
- Author
-
Lang, Karl Sebastian, Caroli, Constanze Charlotte, Muhm, Alexandra, Wernet, Dorothee, Moris, Arnaud, Schittek, Birgit, Knauss-Scherwitz, Evelyn, Stevanovic, Stefan, Rammensee, Hans-Georg, and Garbe, Claus
- Published
- 2001
20. CD107a+ (LAMP-1) cytotoxic CD8+ T-cells in lupus nephritis patients
- Author
-
Wiechmann, Anika, primary, Wilde, Benjamin, additional, Amann, Kerstin, additional, Abdulahad, Wayel H., additional, Kribben, Andreas, additional, Lang, Karl Sebastian, additional, Witzke, Oliver, additional, and Dolff, Sebastian, additional
- Published
- 2020
- Full Text
- View/download PDF
21. Susceptibility of BAFF-var allele carriers to severe SLE with occurrence of lupus nephritis
- Author
-
Friebus-Kardash, Justa, primary, Trendelenburg, Marten, additional, Eisenberger, Ute, additional, Ribi, Camillo, additional, Chizzolini, Carlo, additional, Huynh-Do, Uyen, additional, Lang, Karl Sebastian, additional, Wilde, Benjamin, additional, Kribben, Andreas, additional, Witzke, Oliver, additional, Dolff, Sebastian, additional, and Hardt, Cornelia, additional
- Published
- 2019
- Full Text
- View/download PDF
22. Diverse Immunomodulatory Effects of Individual IFNα Subtypes on Virus-Specific CD8+ T Cell Responses
- Author
-
Dickow, Julia, primary, Francois, Sandra, additional, Kaiserling, Rouven-Luca, additional, Malyshkina, Anna, additional, Drexler, Ingo, additional, Westendorf, Astrid Maria, additional, Lang, Karl Sebastian, additional, Santiago, Mario L., additional, Dittmer, Ulf, additional, and Sutter, Kathrin, additional
- Published
- 2019
- Full Text
- View/download PDF
23. Tamoxifen Protects from Vesicular Stomatitis Virus Infection
- Author
-
Cham, Lamin B., primary, Friedrich, Sarah-Kim, additional, Adomati, Tom, additional, Bhat, Hilal, additional, Schiller, Maximilian, additional, Bergerhausen, Michael, additional, Hamdan, Thamer, additional, Li, Fanghui, additional, Machlah, Yara Maria, additional, Ali, Murtaza, additional, Duhan, Vikas, additional, Lang, Karl Sebastian, additional, Friebus-Kardash, Justa, additional, and Lang, Judith, additional
- Published
- 2019
- Full Text
- View/download PDF
24. Virotherapy Research in Germany: From Engineering to Translation
- Author
-
Ungerechts, Guy, primary, Engeland, Christine E., additional, Buchholz, Christian J., additional, Eberle, Jürgen, additional, Fechner, Henry, additional, Geletneky, Karsten, additional, Holm, Per Sonne, additional, Kreppel, Florian, additional, Kühnel, Florian, additional, Lang, Karl Sebastian, additional, Leber, Mathias F., additional, Marchini, Antonio, additional, Moehler, Markus, additional, Mühlebach, Michael D., additional, Rommelaere, Jean, additional, Springfeld, Christoph, additional, Lauer, Ulrich M., additional, and Nettelbeck, Dirk M., additional
- Published
- 2017
- Full Text
- View/download PDF
25. Diverse Immunomodulatory Effects of Individual IFNα Subtypes on Virus-Specific CD8+ T Cell Responses.
- Author
-
Dickow, Julia, Francois, Sandra, Kaiserling, Rouven-Luca, Malyshkina, Anna, Drexler, Ingo, Westendorf, Astrid Maria, Lang, Karl Sebastian, Santiago, Mario L., Dittmer, Ulf, and Sutter, Kathrin
- Subjects
T cells ,IMMUNOREGULATION ,DENDRITIC cells ,VIRUS diseases ,PSEUDOPOTENTIAL method - Abstract
Clinical administration of Interferon α (IFNα) resulted in limited therapeutic success against some viral infections. Immune modulation of CD8
+ T cell responses during IFNα therapy is believed to play a pivotal role in promoting viral clearance. However, these clinical studies primarily focused on IFNα subtype 2. To date, the immunomodulatory roles of the remaining 10–13 IFNα subtypes remains poorly understood, thereby precluding assessments of their potential for more effective treatments. Here, we report that virus-specific CD8+ T cell responses were influenced to various extents by individual IFNα subtypes. IFNα4, 6, and 9 had the strongest effects on CD8+ T cells, including antiproliferative effects, improved cytokine production and cytotoxicity. Interestingly, augmented cytokine responses were dependent on IFNα subtype stimulation of dendritic cells (DCs), while antiproliferative effects and cytotoxicity were mediated by IFNAR signaling in either CD8+ T cells or DCs. Thus, precise modulation of virus-specific CD8+ T cell responses may be feasible for specific antiviral immunotherapies through careful selection and administration of individual IFNα subtypes. [ABSTRACT FROM AUTHOR]- Published
- 2019
- Full Text
- View/download PDF
26. Die Passagierung von LCMV-WE auf Tumorzellen führt zu sich wiederholenden Mutationen, die Melanomzellen besser infizieren
- Author
-
Schiller, Maximilian and Lang, Karl Sebastian (Akademische Betreuung)
- Subjects
Medizin - Abstract
Dissertation, Universität Duisburg-Essen, 2022
- Published
- 2021
27. Role of tyrosine kinase receptor Mertk in innate and adaptive immune response
- Author
-
Adomati, Tom, Karl Lang, Sebastian, and Lang, Karl Sebastian (Akademische Betreuung)
- Subjects
Fakultät für Biologie ,ddc:610 ,Biologie - Abstract
In this study we showed that the WT (C57BL/6) mice previously infected with VSV (highly cytopathic virus) induces IFN-α anergy after Poly (I.C) rechallenge. We demonstrated that IFN-α anergy was associated with induction of apoptotic cells as infection with VSV enhanced active caspase-3 detection and this correlated with reduced cell viability and increased phosphatidylserine (PS) expression. In line, pretreatment of BDMCs derived from WT mice with dead cells significantly blunted IFN-α induction after VSV challenge. We speculated that Mertk as a sensor of PS on apoptotic cells may be involved in innate anergy after acute viral infection. First, we showed that macrophages and DCs strongly express TAM receptor Mertk. Next, we demonstrated that Mertk deficiency strongly enhanced IFN-1, TNF-α and IL-6 production but lack of Mertk abrogates IFN-α anergy after VSV infection. We proposed that dead cells may activate Mertk signals and this activation can induce other inhibitory signals that dampen antiviral responses. We showed that induction of Mertk signal after VSV challenge results in upregulation of SOCS1, SOCS3, IL-10 and TGF-β. In line, pretreatment of BMDCs derived from WT mice with dead cells strongly induced IL10 expression, which was significantly prevented in Mertk deficient mice. In fact, lack of Mertk limited the expression of these inhibitory proteins and promotes innate antiviral response. We further determined the overall influence of Mertk on antiviral response. We found that genetic deletion or pharmacological inhibition of Mertk reduces VSV titers in the spleen, liver, kidney, lungs, and lymph nodes. Similar to IRF-3 and IRF-7 upregulation, ISG15, OAS1, and MX1 were highly induced in Mertk deficient mice and resulted in better survival of infection. Additionally, deficiency of Mertk enhances IFN-γ production by CD4+ and CD8+ T cells. Though the neutralizing capacities were similar, we suggested that the enhanced IFN-1 production by Mertk–/– mice was sufficient to allow the host to survive the infection. Meanwhile LCMV is completely non cytopathic virus and causes chronic infections, it has been found to result in dramatic CD8 T‐cell expansion and acquisition of effector function and cytotoxic T cells kill their targets by programming them to undergo apoptosis. Based on this finding, we proposed that the apoptotic CD8T cells or CD8 T cell killed target cells might induce inhibitory signals via Mertk that limits CD8T cell function. Indeed, lack of T cells in TCR-beta deficient mice prevented the induction of apoptotic cells as detected by active caspase-3 suggesting that the CD8Tcells T cell response against the virus not the virus itself are inducing the dead cells. The induction of active caspase 3 correlated with upregulation of Mertk in WT mice. WT mice showed reduced Virus specific CD8Tcells and CD8+Tcell expansion, and increased expression of IL10, which is a master regulator of immune response. Similarly, expression of PD1 on CD8+ T cells and virus specific CD8+ T cells was strongly upregulated in WT mice. In fact, lack of Mertk limited PD1 expression and augmented CD8+ T cell function suggesting that Mertk is a promoter of T cells exhaustion. Evaluating immune-pathological consequences of this highly activated CD8+Tcells status, we showed that Mertk–/– mice transiently enhanced liver transaminases. Exploiting dexamethasone as a known inducer of rat thymic apoptosis to enhance Mertk expression in vivo, we proposed that upregulation of Mertk by dexamethasone can limit antiviral response. We showed that pretreatment of WT mice with dexamethasone substantially increased Mertk expression and this was associated with disseminated VSV replication in spleen, liver, kidney, lungs, and lymph nodes. In line, we observed significant increase in SOCS1 and SOCS3 expression and suppressed IFN-1production in dexamethasone-pretreated WT mice. Similarly, after infection with chronic virus, virus specific CD8Tcells and CD8Tcell proliferation was greatly abrogated in dexamethasone pre-treated WT mice. Lack of Mertk abolished the immunosuppressive effect of dexamethasone. In conclusion, dead cells induce tolerogenic state via Mertk. The induction of SOCS1, SOCS3, IL-10, and TGF-β after the activation of Mertk signals extinguishes antiviral mechanisms and results in innate anergy after acute VSV challenge. Mertk regulates adaptive immune response via IL10 and promotes exhausted CD8+Tcell phenotype during chronic LCMV infection. Dissertation, Universität Duisburg-Essen, 2020
- Published
- 2020
- Full Text
- View/download PDF
28. NK cells negatively regulate CD8+ T cells during chronic viral infection in FcεRIγ-dependent manner
- Author
-
Hamdan, Thamer A., Lang, Karl S. (Akademische Betreuung), and Lang, Karl Sebastian
- Subjects
Medizinische Fakultät » Universitätsklinikum Essen » Institut für Immunologie ,Medizin ,ddc:610 - Abstract
Ein großer Anteil der Weltbevölkerung ist von einer chronischen viralen Infektion betroffen. Eine effektive Regulation der CD8+ T-Zellen ist essenziell für die Bekämpfung einer viralen Infektion. Außerdem gelten natürliche Killerzellen (NK-Zellen) als Wächter des angeborenen Immunsystems, welche die antiviralen T-Zellen negativ regulieren. Der Typ I Interferon (IFN-I)-Signalweg der CD8+-T-Zellen ist einer der regulierenden Mediatoren, welche die CD8+T-Zell Antwort vor dem durch NK-Zellen vermittelten Abtöten schützen. Regulierende Faktoren der natürlichen Killerzellen sind jedoch weitgehend unbekannt. Das Lymphozytäre-Choriomeningitis-Virus (LCMV) ist resistent gegen NK-Zellen und stellt ein typisches Modell zur Untersuchung der Interaktion zwischen den NK-Zellen und den CD8+-T-Zellen dar. In dieser Studie haben wir mit Hilfe eines Mausmodells (Fcer1g–/– Mäuse), welchem der Fc-Rezeptor der gemeinsamen Gamma-Kette (FcRγ oder FcεRIγ) fehlt, den Effekt des intrinsischen Fc-Rezeptors der natürlichen Killerzellen auf die Antwort der T-Zellen im Rahmen einer chronischen LCMV-Infektion erforscht. Wir können berichten, dass das Fehlen von FcεRIγ trotz der unveränderten Qualität der natürlichen Killerzellen in den FcεRIγ– Mäusen zu einer starken Immunantwort der CD8+-T-Zellen und einer kontrollierten LCMV-Infektion geführt hat. Außerdem konnten wir bei den NK-Zellen eine hohe intrazelluläre Expression von FcεRIγ feststellen. Darüber hinaus haben wir herausgefunden, dass das Rezeptorprotein NCR1/NKp46, das einen einzigartigen aktivierenden NK-Zell-Rezeptor darstellt und bei ruhenden und aktivierten NK-Zellen exprimiert wird, bei FcεRIγ-defizienten NK-Zellen nicht exprimiert wird. Interessanterweise führte FcεRIγ durch die Hemmung der proteasomalen Degradation von NCR1 zur Stabilisierung der NCR1-Expression. Durch die Übertragung von monoklonalen LCMV-spezifischen CD8+-T-Zellen und die Depletion der NK-Zellen konnten wir die wichtige Rolle des NCR1-intrinsischen FcεRIγ bei der Eliminierung der Immunantwort der LCMV-spezifischen CD8+T-Zellen zeigen. Aufgrund unserer Ergebnisse lässt sich zusammenfassend sagen, dass der Mangel an FcεRIγ zu einem Defekt in der NKp46-Expression in den NK-Zellen führt und somit deren Aktivität beeinträchtigt. Die Immunantwort der CD8+-T-Zellen wird im Verlauf einer LCMV-Infektion durch die Expression des intrinsischen FcεRIγ der NK-Zellen vermindert, wodurch die akute Infektion in eine chronische Infektion mit Erschöpfung der T Zell Antwort und viraler Persistenz umgewandelt wird., Chronic viral infection is a health condition that afflicts a huge sector of the global population. An effective CD8+ T cell regulation is critical to eradicate the viral infection. Furthermore, NK cells are regarded as innate sentinels and widely defined to regulate the antiviral CD8+ T cells negatively. IFN-I signalling in CD8+ T cells is one of the regulating mediators that renders CD8+ T cells protective against NK cell–mediated killing, however, factors modulating the regulatory functions of NK cells are mainly unknown. Albeit, lymphocytic choriomeningitis virus (LCMV) is NK cells-resistant, it is a prototypical model to investigate the NK-CD8+ T cells crosstalk and it is well-studied model for acute and chronic infections. Herein, we exploited mice that are devoid Fc receptor common gamma chain (FcRγ or FcεRIγ) (Fcer1g–/– mice) to address the role of NK cell-intrinsic Fc receptor on shaping the T cell responses in the chronic LCMV settings. We report here that, FcεRIγ deficiency led to potent CD8+ T cell response and efficient control of LCMV, despite the unaffected NK cells quality in FcεRIγ –deficient animals. In addition, we noticed that FcεRIγ is highly expressed intracellularly by NK cells. More specifically, we found that, FcεRIγ-deficient NK cells are not expressing NCR1/NKp46, a unique activating receptor expressed by both resting and activated NK cells. Intriguingly, FcεRIγ was found to stabilize the NCR1 expression via preventing its proteasomal degradation. With the aid of monoclonal LCMV-specific CD8+ T cells transfer and NK cell depletion experiments, we highlight the direct role of NCR1-intrinsic FcεRIγ in eliminating the LCMV-specific CD8+ T cells response. In summary, our study unravels that lack of FcεRIγ abrogates NKp46 expression on NK cells, and hence compromising their activity on target cells. Thus, NK cell-intrinsic FcεRIγ curtails the CD8+ T cells response in the course of viral infection, converting the acute signature of the disease, whereby the robust CD8+ T cells response and efficient viral control, into a chronic one where the T cells exhaustion, immunopathology and virus persistence.
- Published
- 2020
29. Role of viral infection in transplantation medicine
- Author
-
Gassa, Asmae, Lang, Karl S. (Akademische Betreuung), and Lang, Karl Sebastian
- Subjects
Transplantation ,Medizinische Fakultät » Universitätsklinikum Essen » Institut für Immunologie ,Medizin ,ddc:61 ,ddc:610 ,transgenes Mausmodell ,LCMV ,CD8 T Zellen - Abstract
Dissertation, Universität Duisburg-Essen, 2016 Graft versus host disease (GvHD) tritt in 40% der Fälle bei Patienten nach MHC I abgeglichener Knochenmarktransplantation (KMT) auf. Mechanismen, die diese Krankheit verursachen, sind noch zu untersuchen. In dieser Arbeit verwendeten wir eine transgene CD8+ T Zell Mauslinie (P14/CD 45.1+) und eine transgene DEE Mauslinie, die ubiquitär mit dem Fremdantigen (LCMV-Glykoprotein) versehen ist, um Toleranzmechanismen aus Spender abgeleiteten host-spezifischen CD8+ T Zellen nach KMT zu untersuchen. Wir stellten fest, dass host-reaktive CD8+ T Zellen im Thymus nicht negativ selektiert wurden und sich ähnlich in den Empfängermäusen entwickelten wie die nicht host-spezifischen CD8+ T Zellkontrollen. Host-spezifische CD8+ T Zellen ignorierten das ubiquitär exprimierte Antigen auf den Empfänger-Zellen, konnten jedoch ex vivo durch eine LCMV (Lymphozytäre Choriomeningitis Virus)-Infektion aktiviert werden. Lipopolysaccharide (LPS) induzierten einen transienten Zellschaden in den transgenen DEE-Empfängermäusen mit host-spezifischen CD8+ T Zellen, was vermuten ließ, dass die Einführung einer inflammatorischen Immunantwort die Ignoranz unterbrach. Schlussfolgernd konnten wir feststellen, dass host-spezifische CD8+ T Zellen das Antigen im Empfänger nach KMT ignorierten und dass sie nur eliminiert wurden, wenn das Host-Antigen im hämatopoetischen System vorhanden war. Darüber hinaus trug eine LPS-induzierte Immunaktivierung u.a. dazu bei, eine Alloreaktivität von host-spezifischen CD8+ T Zellen nach KMT zu induzieren. Unerwartete Übertragungen von viralen Keimen nach solider Organtransplantation (SOT) kann beim Empfänger eine schwere, lebensgefährliche Erkrankung auslösen. Eine Immunaktivierung trägt dann zum Krankheitsausbruch bei; Mechanismen, die jedoch eine Immunantwort gegen transplantatvermittelte übertragbare Virus Infektionen beeinflussen, sind noch unbekannt. In diesem Modell haben wir herausgefunden, dass bei Verwendung des LCMV eine Transplantation von LCMV-infizierten Herzen zu einer zellulären Erschöpfung von virus-spezifischen CD8+ T Zellen, Virus Persistenz in den Empfängerorganen sowie Überleben des Transplantorgans und –Empfängers führten. Die genetische Depletion des Interleukin-10 (IL-10) resultierte in einer starken Immunaktivierung, Transplantatdysfunktion und Tod der Empfängermäuse, was vermuten ließ, dass IL-10 ein wichtiger Regler für CD8+ T Zell Erschöpfung während SOT darstellte. In Anwesenheit von Memory-CD8+ T Zellen konnte das Virus kontrolliert werden; es kam jedoch aufgrund der vorhandenen antiviralen Immunantwort zu einer Abstoßung des transplantierten Herzens. Schlussfolgernd stellten wir fest, dass durch SOT übertragbare Virusinfektionen nicht durch naive Empfängermäuse aufgrund der IL-10 vermittelten CD8+ T Zell Erschöpfung kontrolliert werden konnten, wodurch Immunpathologie und Transplantatversagen vermieden werden konnte, während infizierte Memorymausempfänger im Stande waren, dass transplantatvermittelte Virus zu kontrollieren und dadurch eine Transplantatabstoßung zu induzieren. Graft versus host disease (GvHD) occurs in 40% of cases with patients having a MHC I matched bone marrow transplantation (BMT). Mechanisms causing this disease remain to be studied. Here we used a CD8+ T cell transgenic mouse strain (P14/CD45.1+) and DEE mice bearing the foreign antigen (LCMV-GP33-41) to study mechanisms of tolerance in donor derived host specific CD8+ T cells after BMT. We found that host reactive CD8+ T cells were not negatively selected in the thymus and developed comparably to host non-specific CD8+ T cells. Host specific CD8+ T cells ignored the antigen expressed ubiquitously by host cells but they could be activated ex vivo via LCMV (lymphocytic choriomeningitis virus)-infection. Lipopolysaccharides (LPS) induced transient cell damage in DEE mice bearing host specific CD8+ T cells, suggesting that induction of host inflammatory response could break this ignorance. In conclusion, we found that after BMT host specific CD8+ T cells ignore antigen in recipients and that they are only deleted when host antigen is present in the hematopoietic system. Moreover, LPS-induced immune activation contributes to induction of alloreactivity of host specific CD8+ T cells after BMT. Unexpected transmissions of viral pathogens during solid organ transplantation (SOT) can result in severe, life-threatening diseases in transplant recipients. Immune activation contributes to disease onset; however mechanisms balancing the immune response against transmitted virus infection through organ transplantation remain unknown. Here, we found, using LCMV, that transplantation of LCMV infected hearts led to exhaustion of virus specific CD8+ T cells, viral persistence in organs and survival of graft and recipient. Genetic depletion of IL-10 resulted in a strong immune activation, graft dysfunction and death of mice, suggesting that IL-10 was a major regulator of CD8+ T cell exhaustion during SOT. In the presence of memory CD8+ T cells, virus could be controlled; however sufficient antiviral immune response resulted in rejection of transplanted heart. In conclusion, we found that virus transmitted by SOT cannot be controlled by naive recipients due to IL-10 mediated CD8+ T cell exhaustion which thereby prevented immunopathology and graft failure whereas memory mice recipients were able to control the virus and induced graft failure.
- Published
- 2017
30. B-Myb is a crucial proviral host-factor and is involved in arenavirus mediated tumor suppression
- Author
-
Sharma, Piyush, Lang, Karl S. (Akademische Betreuung), and Lang, Karl Sebastian
- Subjects
Medizinische Fakultät » Universitätsklinikum Essen » Institut für Immunologie ,ddc:570 ,viruses ,ddc:576 ,Biologie - Abstract
Viren besitzen eine minimale Überlebensausstattung, die ihren Fortbestand in widrigen Umständen sichert, aber sie nicht zur unabhängigen Reproduktion befähigt. Letzteres zu kompensieren, berauben sie die Resourcen ihres Wirts und beuten die Wirts-Maschinerie zu ihrem Vorteil aus. Diese Faktoren ermöglichen in erster Linie die Virus-Replikation und führen daher zur Infektion des Wirts. Sie sind aber auch - vielleicht eher kontraintuitiv – nötig zur Sicherung der Virusreplikation in antigen-präsentierenden Zellen (antigen presenting cells; APCs), wodurch es zur Antigen-Prozessierung und -Präsentation kommt, welche widerum die Immunaktivierung auslöst, damit die Infektion beseitigt werden kann. Das angeborene und das adaptive Immunsystem arbeiten nebeneinander, um Infektionen effektiv zu bekämpfen. Wir haben gezeigt, dass eine verstärkte Virusreplikation in den CD169+ Makrophagen der Milz wichtig für eine Immunaktivierung ist. In dieser Arbeit haben wir eine neue Funktion eines Transkriptions-Faktors identifiziert, der bereits für seine bedeutende Rolle in Zellreplikation und -proliferation bekannt ist. Die vorliegende Arbeit erarbeitet Rolle von B-Myb in der Virus-Replikation und untersucht den therapeutischen Effekt einer LCMV-Infektion bei Tumoren. B-Myb ist ein wichtiger Transkriptionsfaktor für Zellproliferation, Mitglied des DREAM-Komplexes und bekannt dafür, die Expression vielzähliger Gene zu induzieren. Hier wird gezeigt, dass B-Myb auch ein bedeutender Faktor ist, der während der Virus-Replikation aktiviert wird und dem Virus hilft, indem er die Expression verschiedener Wirts-Faktoren erhält, die für die Virion-Produktion und -Freisetzung benötigt werden. Wir haben beobachtet, dass die Aktivierung von B-Myb bei der Virus-Replikation nicht zell-spezifisch ist. Wir zeigen, dass B-Myb-abhängige frühe Virus-Replikation in der Milz wichtig für die Aktivierung des adaptiven Immunsystems und somit der anti-viralen Antwort ist und das sowohl bei RNA- als auch DNA-Viren. Zudem wird gezeigt, dass die B-Myb-abhängige Replikation eine Bedingung für die humanen Viren HSV und HIV ist. Im zweiten Teil der Arbeit wird der Effekt von B-Myb abhängiger LCMV-Replikation bei Tumoren untersucht. Interessanterweise, decken wir das Potential von LCMV als Anti-Tumor-Therapeutikum auf, dass verglichen mit onkolytischen Viren vergleichbar oder sogar effektiver ist. Wir demonstrieren, dass eine LCMV-Behandlung ein Eindringen von Immunzellen in Tumore bewirkt. Darüberhinaus wird gezeigt, dass eine LCMV-Behandlung zur Tumor-Regression aufgrund von IFN-I produzierenden Ly6C+ Monozyten führt, welches anti-angiogenetisch wirkt. Zusammenfassend haben wir eine neue Funktion von B-Myb als Wirtsfaktor identifiziert, der die Replikation mehrerer Viren vereinfacht. Wir zeigten die Wichtigkeit B-Myb’s für die frühe Virus-Replikation, Immun-Aktivierung und Infektionskontrolle auf. Zudem etablierten wir LCMV als eine IFN-I-abhängige Anti-Tumor-Therapie. Viruses have minimal survival requirements thus ensuring their survival in harsh conditions but are unable to replicate independently. To overcome this, they hijack host resources and exploit host machinery for their benefit. These factors enable virus replication in the first place thus infecting the host but perhaps rather counterintuitively they are also necessary to ensure virus replication in antigen presenting cells (APCs) to ensure virus antigen processing and presentation ensuring immune activation such that the infection can be cleared. Innate and adaptive immune system works side-by-side in order to effectively combat infections. We have shown that enforced virus replication in splenic CD169+ macrophages is crucial for immune activation. In this study, we have identified a novel function of a transcription factor known for its crucial role in cell replication and proliferation. The present study thus deals with elaborating the role of B-Myb in virus replication and exploring the therapeutic effect of LCMV replication in tumors. B-Myb is an important transcription factor for cell proliferation, member of DREAM complex and is also known to induce expression of multiple genes. Here, we show that B-Myb is also a major transcription factor activated during virus replication and helps the virus by maintaining the expression of various host-factors required for virion production and release. We observed that activation of B-Myb on viral replication is not cell-specific. We show that B-Myb dependent early virus replication in spleen is crucial for adaptive immune activation and thus anti-viral response in both RNA and DNA viruses. We also show here that B-Myb dependent replication is also a requirement for human viruses HSV and HIV. In the second portion of the thesis we explored the effect of B-Myb dependent LCMV replication in tumors. Interestingly, we show the potential of LCMV as anti-tumor virotheray which when compared to oncolytic viruses, is comparable or more effective. We demonstrated that LCMV treatment induces evasive immune infiltration in tumors. We show that LCMV treatment leads to tumor regression via IFN-I producing Ly6C+ monocytes which acts as anti-angiogenesis. In conclusion, we have identified a new function of B-Myb as host factor facilitating the replication of multiple viruses. We demonstrated the importance of B-Myb in early virus replication for immune activation and infection control. We also established LCMV as an IFN-I dependent anti-tumor therapy. Dissertation, Universität Duisburg-Essen, 2017
- Published
- 2017
31. Crosstalk between Splenic CD169+ Macrophages and Adaptive Immune System
- Author
-
Duhan, Vikas, Lang, Karl S. (Akademische Betreuung), and Lang, Karl Sebastian
- Subjects
Medizinische Fakultät » Universitätsklinikum Essen » Institut für Immunologie ,ddc:570 ,viruses ,ddc:572 ,Biologie - Abstract
Krankheiten, die durch Virusinfektionen ausgelöst werden, stellen eine große Besorgnis für die Menschheit dar. Viren wie das humane Immundefizienz-Virus (HIV), Hepatitis B Virus (HBV) und Hepatitis C Virus (HCV), die chronische Infektionen verursachen, sind immer lebensbedrohlich. Die Mechanismen zu verstehen, wie das Immunsystem Virusinfektionen bekämpft, kann helfen bessere Impfstoffe oder Therapien zu entwickeln, um tödlichen Virusinfektionen vorzubeugen. In dieser Studie untersuchten wir die Rolle von CD169+ Makrophagen, die in der Marginalzone der Milz lokalisiert sind, für der adaptiven Immunantwort. Die anfängliche Virus-Replikation in den CD169+ Makrophagen der Milz bei einer systemischen Infektion mit dem vesikulären Stomatitis-Virus (VSV), welche das angeborene und adaptive Immunsystem aktiviert, wurde bereits beschrieben. Indem wir hier das lymphozytäre Choriomeningitis-Virus (LCMV) im Maus-Modell untersuchten, fanden wir, dass die CD169+ Makrophagen in Milz und Lymphknoten Typ-I-Interferon (IFN-I) produzieren. IFN-I agierte antiviral, inhibierte somit die Virus-Replikation in verschiedenen peripheren Organen und stimulierte die Expression von program death ligand 1 (PD-L1) in der Leber. PD-L1 ist ein Ligand für PD1, das auf aktivierten CD8+ T-Zellen exprimiert wird. Die Signale über die PD-L1/PD1-Achse induzieren die Erschöpfung von CD8+ T-Zellen. In Abwesenheit von CD169+ Makrophagen war die IFN-I-Produktion und PD-L1-Expression stark reduziert. Die führte zu einer überwältigenden Virus-Replikation bei fehlender CD8+ T-Zell-Erschöpfung, was in schwerer Immunpathologie und dem Tod der Mäuse endete. Diese Arbeit zeigt klar die Rolle von CD169+ Makrophagen bei der funktionalen Regulierung der CD8+ T-Zellen auf. In einer zweiten Studie beschrieben wir den Einfluss Virus-spezifischer Antikörper oder Virus-spezifischer CD8+ T-Zellen auf die CD169+ Makrophagen der Milz während einer systemischen Zweit-Infektion mit LCMV. Wir berichteten, dass die Anwesenheit von LCMV-spezifischen Antikörpern die intrazelluläre Virus-Replikation in den CD169+ Makrophagen der Marginalzone der Milz zulässt, aber die Virus-Replikation in peripheren Organen unterdrückt. Während einer persistenten Infektion mit LCMV-Docile war die Replikation in den CD169+ Makrophagen der Milz bei Anwesenheit Virus-spezifischer Antikörper essentiell für die Vorbereitung der CD8+ T-Zellen, was zur Beseitung des Virus führte. Parallel zu spezifischen Antikörpern inhibierten CD8+ T-Gedächtnis-Zellen die Virus-Replikation in CD169+ Makrophagen und die systemische IFN-I-Produktion und schafften es nicht eine effective CD8+ T-Zell-Antwort aufzubauen, was in der Persistenz des Virus resultierte. Diese Untersuchung erklärt die Mechanismen der Immun-Aktivierung und des Immun-Schutzes bei Anwesenheit von Virus-spezifischen Antikörpern bei viralen Zweit-Infektionen. Insgesamt kann man aus diesen beiden Studien schlussfolgern, dass CD169+ Makrophagen signifikante Rollen für die Induzierung einer starken antiviralen angeborenen und adaptiven Immunantwort bei viralen Erst- und Zweit-Infektionen einnehmen. Diseases caused by viral infections are major concerns for human being. Viruses like human immunodeficiency virus (HIV), hepatitis B virus (HBV) and hepatitis C virus (HCV) that induce chronic infection, is always a risk for human life. Understanding the mechanisms how immune system combat against viral infection can help to develop better vaccines or treatment therapies to prevent from lethal viral infections. Here in this study, we analyzed the roles of CD169+ macrophages present in splenic marginal zone on adaptive immunity. Initial virus replication in splenic CD169+ macrophages during systemic infection of vesicular stomatitis virus (VSV) was described earlier which activate innate and adaptive immune system. Here using Lymphocytic choriomeningitis virus (LCMV) in mouse model, we found that CD169+ macrophages in spleen and lymph node produce type I interferon (IFN-I). IFN-I acts as antiviral and inhibited virus replication in different peripheral organs, and stimulated program death ligand 1 (PD-L1) expression in liver. PD-L1 is a ligand for PD-1 expressed on activated CD8+ T cells, and PD-L1/PD-1 axis signaling induces CD8+ T cell exhaustion. In absence of CD169+ macrophages, IFN-I production was impaired and PD-L1 expression was highly reduced. This led to overwhelming virus replication in absence of CD8+ T cells exhaustion that resulted in severe immunopathology and death of mice. This study clearly shows the role of CD169+ macrophages in regulating the functions of CD8+ T cells. In another study, we described the influence of virus-specific antibodies or virus-specific memory CD8+ T cells on splenic CD169+ macrophages during systemic recall infection of LCMV. We reported that the presence of LCMV-specific antibodies permit intracellular viral replication in CD169+ macrophages of splenic marginal zone but suppressed viral replication in peripheral organs. During persistent infection with the LCMV-Docile, viral replication in CD169+ macrophages of spleen in presence of virus specific antibodies was found to be essential for priming of CD8+ T cells that led to viral clearance. In parallel to specific antibodies, memory CD8+ T cells inhibited viral replication in CD169+ macrophages and systemic IFN-I production, and failed to mount effective CD8+ T cell response that resulted in viral persistence. This investigation explains the mechanism of immune activation and protection in presence of virus specific antibodies during secondary viral infection. Over all from these two studies we can conclude that CD169+ macrophages play significant roles for inducing strong antiviral innate and adaptive immunity during primary and secondary viral infection. Dissertation, Universität Duisburg-Essen, 2017
- Published
- 2017
32. Immune suppression of vaccine-induced CD8 + T-cell responses by gamma retrovirus envelope is mediated by interleukin-10-producing CD4 + T cells.
- Author
-
Podschwadt P, Malyshkina A, Windmann S, Papadamakis A, Kerkmann L, Lapuente D, Tenbusch M, Lu M, Schindler M, Lang KS, Hansen W, and Bayer W
- Subjects
- Animals, Mice, CD4-Positive T-Lymphocytes, CD8-Positive T-Lymphocytes, Friend murine leukemia virus, Gene Products, env, Immunosuppression Therapy, Interleukin-10, Retroviridae, Retroviridae Proteins, Humans, Retroviridae Infections, Viral Vaccines
- Abstract
Retroviral envelope (Env) proteins have long been recognized to exhibit immunosuppressive properties, which affect the CD8
+ T-cell response to an infection but also to immunization. Interestingly, we previously showed in the Friend murine leukemia virus (F-MuLV) model that the surface Env protein gp70 also plays a role in immunosuppression, in addition to the immunosuppressive function attributed to the transmembrane Env protein. We now demonstrate that immunization with F-MuLV Env leads to a significant increase in interleukin-10 (IL-10)-producing CD4+ T cells and that the induction of CD8+ T-cell responses in the presence of Env is rescued if the capacity of CD4+ T cells to produce IL-10 is abrogated, indicating a mechanistic role of IL-10-producing CD4+ T cells in mediating the Env-induced suppression of CD8+ T-cell responses in Env co-immunization. We found that CD8+ T-cell responses against different immunogens are not all equally affected. On the other hand, suppression of immunity was observed not only in co-immunization experiments but also for immune control of subcutaneous tumor growth after an Env immunization. Finally, we show that suppression of CD8+ T cells by the surface Env protein is observed not only for Friend MuLV Env but also for the Env proteins of other gamma retroviruses. Taken together, our results show that IL-10-producing CD4+ T cells mechanistically underlie the Env-mediated suppression of CD8+ T-cell responses and suggest the presence of an immunosuppressive motif in the surface Env protein of gamma retroviruses., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2022 Podschwadt, Malyshkina, Windmann, Papadamakis, Kerkmann, Lapuente, Tenbusch, Lu, Schindler, Lang, Hansen and Bayer.)- Published
- 2022
- Full Text
- View/download PDF
33. CD107a + (LAMP-1) Cytotoxic CD8 + T-Cells in Lupus Nephritis Patients.
- Author
-
Wiechmann A, Wilde B, Tyczynski B, Amann K, Abdulahad WH, Kribben A, Lang KS, Witzke O, and Dolff S
- Abstract
Cytotoxic CD8
+ T-cells play a pivotal role in the pathogenesis of systemic lupus erythematosus (SLE). The aim of this study was to investigate the role of CD107a (LAMP-1) on cytotoxic CD8+ T-cells in SLE-patients in particular with lupus nephritis. Peripheral blood of SLE-patients ( n = 31) and healthy controls ( n = 21) was analyzed for the expression of CD314 and CD107a by flow cytometry. Kidney biopsies of lupus nephritis patients were investigated for the presence of CD8+ and C107a+ cells by immunohistochemistry and immunofluorescence staining. The percentages of CD107a+ on CD8+ T-cells were significantly decreased in SLE-patients as compared to healthy controls (40.2 ± 18.5% vs. 47.9 ± 15.0%, p = 0.02). This was even more significant in SLE-patients with inactive disease. There was a significant correlation between the percentages of CD107a+ CD8+ T-cells and SLEDAI. The evaluation of lupus nephritis biopsies showed a significant number of CD107a+ CD8+ T-cells mainly located in the peritubular infiltrates. The intrarenal expression of CD107a+ was significantly correlated with proteinuria. These results demonstrate that CD8+ T-cells of patients with systemic lupus erythematosus have an altered expression of CD107a which seems to be associated with disease activity. The proof of intrarenal CD107a+ CD8+ suggests a role in the pathogenesis of lupus nephritis., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2021 Wiechmann, Wilde, Tyczynski, Amann, Abdulahad, Kribben, Lang, Witzke and Dolff.)- Published
- 2021
- Full Text
- View/download PDF
34. Usp18 Expression in CD169 + Macrophages is Important for Strong Immune Response after Vaccination with VSV-EBOV.
- Author
-
Friedrich SK, Schmitz R, Bergerhausen M, Lang J, Cham LB, Duhan V, Häussinger D, Hardt C, Addo M, Prinz M, Asano K, Lang PA, and Lang KS
- Abstract
Ebola virus epidemics can be effectively limited by the VSV-EBOV vaccine (Ervebo) due to its rapid protection abilities; however, side effects prevent the broad use of VSV-EBOV as vaccine. Mechanisms explaining the efficient immune activation after single injection with the VSV-EBOV vaccine remain mainly unknown. Here, using the clinically available VSV-EBOV vaccine (Ervebo), we show that the cell-intrinsic expression of the interferon-inhibitor Usp18 in CD169
+ macrophages is one important factor modulating the anti-Ebola virus immune response. The absence of Usp18 in CD169+ macrophages led to the reduced local replication of VSV-EBOV followed by a diminished innate as well as adaptive immune response. In line, CD169 -Cre+/ki x Usp18fl/fl mice showed reduced innate and adaptive immune responses against the VSV wildtype strain and died quickly after infection, suggesting that a lack of Usp18 makes mice more susceptible to the side effects of the VSV vector. In conclusion, our study shows that Usp18 expression in CD169+ macrophages is one important surrogate marker for effective vaccination against VSV-EBOV, and probably other VSV-based vaccines also.- Published
- 2020
- Full Text
- View/download PDF
35. Diverse Immunomodulatory Effects of Individual IFNα Subtypes on Virus-Specific CD8 + T Cell Responses.
- Author
-
Dickow J, Francois S, Kaiserling RL, Malyshkina A, Drexler I, Westendorf AM, Lang KS, Santiago ML, Dittmer U, and Sutter K
- Subjects
- Animals, Antiviral Agents immunology, Antiviral Agents pharmacology, CD8-Positive T-Lymphocytes immunology, CD8-Positive T-Lymphocytes virology, Cell Survival drug effects, Cell Survival immunology, Cytokines immunology, Cytokines metabolism, Dendritic Cells drug effects, Dendritic Cells immunology, Dendritic Cells virology, HEK293 Cells, Humans, Immunologic Factors immunology, Immunologic Factors pharmacology, Interferon-alpha classification, Interferon-alpha immunology, Mice, Inbred BALB C, Mice, Inbred C57BL, Mice, Knockout, Protein Isoforms immunology, Protein Isoforms pharmacology, Virus Replication immunology, CD8-Positive T-Lymphocytes drug effects, Cell Proliferation drug effects, Interferon-alpha pharmacology, Virus Replication drug effects
- Abstract
Clinical administration of Interferon α (IFNα) resulted in limited therapeutic success against some viral infections. Immune modulation of CD8
+ T cell responses during IFNα therapy is believed to play a pivotal role in promoting viral clearance. However, these clinical studies primarily focused on IFNα subtype 2. To date, the immunomodulatory roles of the remaining 10-13 IFNα subtypes remains poorly understood, thereby precluding assessments of their potential for more effective treatments. Here, we report that virus-specific CD8+ T cell responses were influenced to various extents by individual IFNα subtypes. IFNα4, 6, and 9 had the strongest effects on CD8+ T cells, including antiproliferative effects, improved cytokine production and cytotoxicity. Interestingly, augmented cytokine responses were dependent on IFNα subtype stimulation of dendritic cells (DCs), while antiproliferative effects and cytotoxicity were mediated by IFNAR signaling in either CD8+ T cells or DCs. Thus, precise modulation of virus-specific CD8+ T cell responses may be feasible for specific antiviral immunotherapies through careful selection and administration of individual IFNα subtypes., (Copyright © 2019 Dickow, Francois, Kaiserling, Malyshkina, Drexler, Westendorf, Lang, Santiago, Dittmer and Sutter.)- Published
- 2019
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.