Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Monika Braun is active.

Publication


Featured researches published by Monika Braun.


Journal of Experimental Medicine | 2011

Rapid expansion and long-term persistence of elevated NK cell numbers in humans infected with hantavirus

Niklas K. Björkström; Therese Lindgren; Malin Stoltz; Cyril Fauriat; Monika Braun; Magnus Evander; Jakob Michaëlsson; Karl-Johan Malmberg; Jonas Klingström; Clas Ahlm; Hans-Gustaf Ljunggren

Acute hantavirus infection in humans triggers a rapid expansion and long-term persistence of NK cells.


Clinical Cancer Research | 2011

Natural Killer Cells are Scarce in Colorectal Carcinoma Tissue Despite High Levels of Chemokines and Cytokines

Niels Halama; Monika Braun; Christoph Kahlert; Anna Spille; Christian Quack; Nuh N. Rahbari; Moritz Koch; Jürgen Weitz; Matthias Kloor; Inka Zoernig; Peter Schirmacher; Karsten Brand; Niels Grabe; Christine S. Falk

Purpose: Tumor infiltrating T lymphocytes in colorectal cancer (CRC) have prognostic impact, but the role of natural killer (NK) cells in CRC tissue is unclear. The contribution of intratumoral cytokines and chemokines in shaping the composition of the inflammatory lymphocytic infiltrate is also unclear. Experimental Design: In this study, localization and densities of NK and T cells within primary CRC, CRC liver metastases, adenomas, and normal tissues were analyzed on whole tissue sections from 112 patients. In a subset of these patients, the most important 50 cytokines and chemokines were quantified in paired serum, primary CRC and adjacent mucosa samples and in CRC liver metastases and correlated with NK and T-cell infiltration, respectively. Results: The various compartments displayed characteristic differences like significantly higher chemokine concentrations in CRC tissue. Most importantly, despite high local chemokine levels, NK cells were generally scarce within CRC tumor tissues, independent of human leukocyte antigen (HLA) class I expression. Adjacent normal mucosa contained normal levels of NK cells. In contrast, corresponding T-cell numbers varied substantially and were positively correlated with higher chemokine levels. Conclusions: Our findings indicate a distinct regulation of NK cells versus T cells in the CRC tumor microenvironment. NK-cell migration into CRC tumor tissue is obviously impaired early during tumor development by mechanisms that do not affect T-cell infiltration. Clin Cancer Res; 17(4); 678–89. ©2011 AACR.


International Journal of Cancer | 2009

Down‐regulation of HLA Class I and NKG2D ligands through a concerted action of MAPK and DNA methyltransferases in colorectal cancer cells

Christine Sers; Ruprecht Kuner; Christine S. Falk; Per Lund; Holger Sueltmann; Monika Braun; Andreas Buness; Markus Ruschhaupt; Janine Conrad; Shila Mang-Fatehi; Iwona Stelniec; Ulf Krapfenbauer; Annemarie Poustka; Reinhold Schäfer

Most malignant features of cancer cells are triggered by activated oncogenes and the loss of tumor suppressors due to mutation or epigenetic inactivation. It is still unclear, to what extend the escape of emerging cancer cells from recognition and elimination by the immune system is determined by similar mechanisms. We compared the transcriptomes of HCT116 colorectal cancer cells deficient in DNA methyltransferases (DNMTs) and of cells, in which the RAS pathway as the major growth‐promoting signaling system is blocked by inhibition of MAPK. We identified the MHC Class I genes HLA‐A1/A2 and the ULBP2 gene encoding 1 of the 8 known ligands of the activating NK receptor NKG2D among a cluster of immune genes up‐regulated under the conditions of both DNMT‐deficiency and MEK‐inhibition. Bisulphite sequencing analyses of HCT116 with DNMT deficiency or after MEK‐inhibition showed that de‐methylation of the ULPB2 promoter correlated with its enhanced surface expression. The HLA‐A promoters were not methylated indicating that components of the HLA assembly machinery were also suppressed in DNMT‐deficient and MEK‐inhibited cells. Increased HLA‐A2 surface expression was correlated with enhanced recognition and lysis by A2‐specific CTL. On the contrary, elevated ULBP2 expression was not reflected by enhanced recognition and lysis by NK cells. Cosuppression of HLA Class I and NKG2D ligands and genes encoding peptide transporters or proteasomal genes mediates a strong functional link between RAS activation, DNMT activity and disruption of the antigen presenting system controlling immune recognition in colorectal cancer cells.


Journal of Immunology | 2013

Temporal Dynamics of the Primary Human T Cell Response to Yellow Fever Virus 17D As It Matures from an Effector- to a Memory-Type Response

Kim Blom; Monika Braun; Martin A. Ivarsson; Veronica D. Gonzalez; Karolin Falconer; Markus Moll; Hans-Gustaf Ljunggren; Jakob Michaëlsson; Johan K. Sandberg

The live attenuated yellow fever virus (YFV) 17D vaccine provides a good model to study immune responses to an acute viral infection in humans. We studied the temporal dynamics, composition, and character of the primary human T cell response to YFV. The acute YFV-specific effector CD8 T cell response was broad and complex; it was composed of dominant responses that persisted into the memory population, as well as of transient subdominant responses that were not detected at the memory stage. Furthermore, HLA-A2– and HLA-B7–restricted YFV epitope–specific effector cells predominantly displayed a CD45RA−CCR7−PD-1+CD27high phenotype, which transitioned into a CD45RA+CCR7−PD-1−CD27low memory population phenotype. The functional profile of the YFV-specific CD8 T cell response changed in composition as it matured from an effector- to a memory-type response, and it tended to become less polyfunctional during the course of this transition. Interestingly, activation of CD4 T cells, as well as FOXP3+ T regulatory cells, in response to YFV vaccination preceded the kinetics of the CD8 T cell response. The present results contribute to our understanding of how immunodominance patterns develop, as well as the phenotypic and functional characteristics of the primary human T cell response to a viral infection as it evolves and matures into memory.


PLOS Pathogens | 2013

Hantavirus-infection Confers Resistance to Cytotoxic Lymphocyte-Mediated Apoptosis

Shawon Gupta; Monika Braun; Nicole D. Tischler; Malin Stoltz; Karin Sundström; Niklas K. Björkström; Hans-Gustaf Ljunggren; Jonas Klingström

Hantaviruses cause hemorrhagic fever with renal syndrome (HFRS) and hantavirus cardio-pulmonary syndrome (HCPS; also called hantavirus pulmonary syndrome (HPS)), both human diseases with high case-fatality rates. Endothelial cells are the main targets for hantaviruses. An intriguing observation in patients with HFRS and HCPS is that on one hand the virus infection leads to strong activation of CD8 T cells and NK cells, on the other hand no obvious destruction of infected endothelial cells is observed. Here, we provide an explanation for this dichotomy by showing that hantavirus-infected endothelial cells are protected from cytotoxic lymphocyte-mediated induction of apoptosis. When dissecting potential mechanisms behind this phenomenon, we discovered that the hantavirus nucleocapsid protein inhibits the enzymatic activity of both granzyme B and caspase 3. This provides a tentative explanation for the hantavirus-mediated block of cytotoxic granule-mediated apoptosis-induction, and hence the protection of infected cells from cytotoxic lymphocytes. These findings may explain why infected endothelial cells in hantavirus-infected patients are not destroyed by the strong cytotoxic lymphocyte response.


Journal of Innate Immunity | 2011

The CD6 Scavenger Receptor Is Differentially Expressed on a CD56dim Natural Killer Cell Subpopulation and Contributes to Natural Killer-Derived Cytokine and Chemokine Secretion

Monika Braun; Bernadette Müller; Dominik ter Meer; Silke Raffegerst; Barbara Simm; Susanne Wilde; Stefani Spranger; Joachim Ellwart; Barbara Mosetter; Ludmila Umansky; Tina Lerchl; Dolores J. Schendel; Christine S. Falk

The CD6 scavenger receptor is known to be expressed on virtually all T cells and is supposed to be involved in costimulation, synapse formation, thymic selection and leukocyte migration. Here, we demonstrate that CD6 is differentially expressed by a subpopulation of peripheral CD56dim natu- ral killer (NK) cells and absent on CD56bright NK cells. CD56dimCD16+ cells represent the major NK subset in the periphery, and most cells within this group are positive for CD6. Most killer immunoglobulin-like receptor- and immunoglobulin-like transcript-positive cells also belong to the CD6+ subpopulation, as expected from their restricted expression on CD56dim NK cells. In addition, CD6+ NK cells are similar to the newly identified CD94lowCD56dim NK subpopulation and most distant from the recently defined CD27+ NK subpopulation based on the reverse correlation of expression between CD6 and CD27, a marker associated primarily with CD56bright NK cells. With respect to CD6 function on NK cells, direct CD6 triggering did not result in degranulation but induced secretion of cytokines (interferon-γ and tumor necrosis factor-α) and chemokines [CXCL10 (IP-10), CXCL1 (GRO-α)]. Thus, CD6 expression on peripheral NK cells marks a novel CD56dim subpopulation associated with distinct patterns of cytokine and chemokine secretion.


European Journal of Immunology | 2006

TCR-independent cytokine stimulation induces non-MHC-restricted T cell activity and is negatively regulated by HLA class I.

Marion von Geldern; Barbara Simm; Monika Braun; Elisabeth H. Weiß; Dolores J. Schendel; Christine S. Falk

Recent evidence suggests that the functional status of T cells activated independently from their TCR differs substantially from classical MHC‐restricted T cells. Here, we show that TCR‐independent, short‐term stimulation via the common γ‐chain of the IL‐2/IL‐15 receptor induces non‐MHC‐restricted cytotoxicity and sustained cytokine secretion in purified CD4+ or CD8+ T cells. NK‐like cytotoxicity is directed against MHC class I‐negative targets and can be inhibited by classical and non‐classical HLA class I molecules. Known inhibitory receptors, such as CD85j (ILT2) and leukocyte‐associated Ig‐like receptor‐1, are not responsible for this HLA‐mediated inhibition. NK‐like cytotoxicity can be costimulated by NKG2D (CD314) triggering, but 2B4 (CD244) and DNAM‐1 (CD226) are not involved. NK‐like T cells display an activated phenotype and secrete various cytokines, including IFN‐γ, TNF‐α, IL‐5, IL‐13 and MIP‐1β. Under normal conditions, HLA class I‐mediated inhibition may function as a safety mechanism to prevent unbalanced cytokine production and effector killing mechanisms by T cells that were activated independently from their TCR. Non‐MHC‐restricted activity represents a functional status rather than a property of distinct T cell subpopulations. Thus, cytokine‐induced, non‐MHC‐restricted T cells may be relevant in immune responses against tumors showing aberrant MHC expression through their capacities of cytokine production and direct tumor cell eradication.


PLOS Pathogens | 2014

NK Cell Activation in Human Hantavirus Infection Explained by Virus-Induced IL-15/IL15Rα Expression

Monika Braun; Niklas K. Björkström; Shawon Gupta; Karin Sundström; Clas Ahlm; Jonas Klingström; Hans-Gustaf Ljunggren

Clinical infection with hantaviruses cause two severe acute diseases, hemorrhagic fever with renal syndrome (HFRS) and hantavirus pulmonary syndrome (HPS). These diseases are characterized by strong immune activation, increased vascular permeability, and up to 50% case-fatality rates. One prominent feature observed in clinical hantavirus infection is rapid expansion of natural killer (NK) cells in peripheral blood of affected individuals. We here describe an unusually high state of activation of such expanding NK cells in the acute phase of clinical Puumala hantavirus infection. Expanding NK cells expressed markedly increased levels of activating NK cell receptors and cytotoxic effector molecules. In search for possible mechanisms behind this NK cell activation, we observed virus-induced IL-15 and IL-15Rα on infected endothelial and epithelial cells. Hantavirus-infected cells were shown to strongly activate NK cells in a cell-cell contact-dependent way, and this response was blocked with anti-IL-15 antibodies. Surprisingly, the strength of the IL-15-dependent NK cell response was such that it led to killing of uninfected endothelial cells despite expression of normal levels of HLA class I. In contrast, hantavirus-infected cells were resistant to NK cell lysis, due to a combination of virus-induced increase in HLA class I expression levels and hantavirus-mediated inhibition of apoptosis induction. In summary, we here describe a possible mechanism explaining the massive NK cell activation and proliferation observed in HFRS patients caused by Puumala hantavirus infection. The results add further insights into mechanisms behind the immunopathogenesis of hantavirus infections in humans and identify new possible targets for intervention.


PLOS Pathogens | 2015

Specificity and Dynamics of Effector and Memory CD8 T Cell Responses in Human Tick-Borne Encephalitis Virus Infection

Kim Blom; Monika Braun; Jolita Pakalniene; Laura Dailidyte; Vivien Béziat; Margit H. Lampen; Jonas Klingström; Nina Lagerqvist; Torbjörn Kjerstadius; Jakob Michaëlsson; Lars Lindquist; Hans-Gustaf Ljunggren; Johan K. Sandberg; Aukse Mickiene; Sara Gredmark-Russ

Tick-borne encephalitis virus (TBEV) is transferred to humans by ticks. The virus causes tick-borne encephalitis (TBE) with symptoms such as meningitis and meningoencephalitis. About one third of the patients suffer from long-lasting sequelae after clearance of the infection. Studies of the immune response during TBEV-infection are essential to the understanding of host responses to TBEV-infection and for the development of therapeutics. Here, we studied in detail the primary CD8 T cell response to TBEV in patients with acute TBE. Peripheral blood CD8 T cells mounted a considerable response to TBEV-infection as assessed by Ki67 and CD38 co-expression. These activated cells showed a CD45RA-CCR7-CD127- phenotype at day 7 after hospitalization, phenotypically defining them as effector cells. An immunodominant HLA-A2-restricted TBEV epitope was identified and utilized to study the characteristics and temporal dynamics of the antigen-specific response. The functional profile of TBEV-specific CD8 T cells was dominated by variants of mono-functional cells as the effector response matured. Antigen-specific CD8 T cells predominantly displayed a distinct Eomes+Ki67+T-bet+ effector phenotype at the peak of the response, which transitioned to an Eomes-Ki67-T-bet+ phenotype as the infection resolved and memory was established. These transcription factors thus characterize and discriminate stages of the antigen-specific T cell response during acute TBEV-infection. Altogether, CD8 T cells responded strongly to acute TBEV infection and passed through an effector phase, prior to gradual differentiation into memory cells with distinct transcription factor expression-patterns throughout the different phases.


Journal of Immunology | 2015

The Human NK Cell Response to Yellow Fever Virus 17D Is Primarily Governed by NK Cell Differentiation Independently of NK Cell Education.

Nicole Marquardt; Martin A. Ivarsson; Kim Blom; Veronica D. Gonzalez; Monika Braun; Karolin Falconer; Rasmus Gustafsson; Anna Fogdell-Hahn; Johan K. Sandberg; Jakob Michaëlsson

NK cells play an important role in the defense against viral infections. However, little is known about the regulation of NK cell responses during the first days of acute viral infections in humans. In this study, we used the live attenuated yellow fever virus (YFV) vaccine 17D as a human in vivo model to study the temporal dynamics and regulation of NK cell responses in an acute viral infection. YFV induced a robust NK cell response in vivo, with an early activation and peak in NK cell function at day 6, followed by a delayed peak in Ki67 expression, which was indicative of proliferation, at day 10. The in vivo NK cell response correlated positively with plasma type I/III IFN levels at day 6, as well as with the viral load. YFV induced an increased functional responsiveness to IL-12 and IL-18, as well as to K562 cells, indicating that the NK cells were primed in vivo. The NK cell responses were associated primarily with the stage of differentiation, because the magnitude of induced Ki67 and CD69 expression was distinctly higher in CD57− NK cells. In contrast, NK cells expressing self- and nonself-HLA class I–binding inhibitory killer cell Ig-like receptors contributed, to a similar degree, to the response. Taken together, our results indicate that NK cells are primed by type I/III IFN in vivo early after YFV infection and that their response is governed primarily by the differentiation stage, independently of killer cell Ig-like receptor/HLA class I–mediated inhibition or education.

Collaboration


Dive into the Monika Braun's collaboration.

Top Co-Authors

Avatar

Hans-Gustaf Ljunggren

Karolinska University Hospital

View shared research outputs
Top Co-Authors

Avatar

Jakob Michaëlsson

Karolinska University Hospital

View shared research outputs
Top Co-Authors

Avatar

Jonas Klingström

Karolinska University Hospital

View shared research outputs
Top Co-Authors

Avatar

Kim Blom

Karolinska University Hospital

View shared research outputs
Top Co-Authors

Avatar

Johan K. Sandberg

Karolinska University Hospital

View shared research outputs
Top Co-Authors

Avatar

Karolin Falconer

Karolinska University Hospital

View shared research outputs
Top Co-Authors

Avatar

Martin A. Ivarsson

Karolinska University Hospital

View shared research outputs
Top Co-Authors

Avatar

Niklas K. Björkström

Karolinska University Hospital

View shared research outputs
Top Co-Authors

Avatar

Aukse Mickiene

Karolinska University Hospital

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge