Network


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

Hotspot


Dive into the research topics where Christian König is active.

Publication


Featured researches published by Christian König.


Arthritis & Rheumatism | 2012

Interleukin-17 sensitizes joint nociceptors to mechanical stimuli and contributes to arthritic pain through neuronal interleukin-17 receptors in rodents

Frank Richter; Gabriel Natura; Matthias Ebbinghaus; Gisela Segond von Banchet; Susanne Hensellek; Christian König; Rolf Bräuer; Hans-Georg Schaible

OBJECTIVE Interleukin-17 (IL-17) is considered a proinflammatory cytokine, but whether neuronal IL-17 receptors contribute to the generation of arthritic pain is unknown. This study was undertaken to explore whether IL-17A acts on neurons, whether it sensitizes joint nociceptors, and whether neutralization of IL-17 is antinociceptive. METHODS We recorded action potentials from rat joint nociceptors after intraarticular injection of IL-17A. We studied the expression of the IL-17A receptor in the rat dorsal root ganglia (DRG), explored the effect of IL-17A on signaling pathways in cultured rat DRG neurons, and using patch clamp recordings, monitored changes of excitability by IL-17A. We tested whether an antibody to IL-17 influences pain behaviors in mice with antigen-induced arthritis (AIA). RESULTS A single injection of IL-17A into the rat knee joint elicited a slowly developing and long-lasting sensitization of nociceptive C fibers of the joint to mechanical stimuli, which was not attenuated by neutralizing tumor necrosis factor α or IL-6. The IL-17A receptor was visualized in most rat DRG neurons, the cell bodies of primary sensory neurons. In isolated and cultured rat DRG neurons, IL-17A caused rapid phosphorylation of protein kinase B and ERK, and it rapidly enhanced excitability. In mice with unilateral AIA in the knee, an antibody against IL-17 improved the guarding score and reduced secondary mechanical hyperalgesia at the ipsilateral paw. CONCLUSION Our findings indicate that IL-17A has the potential to act as a pain mediator by targeting IL-17 receptors in nociceptive neurons, and these receptors are particularly involved in inflammation-evoked mechanical hyperalgesia.


Molecular and Cellular Neuroscience | 2013

Neuronal IL-17 receptor upregulates TRPV4 but not TRPV1 receptors in DRG neurons and mediates mechanical but not thermal hyperalgesia.

Gisela Segond von Banchet; Michael Karl Boettger; Christian König; Yoichiro Iwakura; Rolf Bräuer; Hans-Georg Schaible

In addition to the proinflammatory cytokines tumor necrosis factor-α, interleukin-6 and interleukin-1ß, the cytokine interleukin-17 (IL-17) is considered an important mediator of autoimmune diseases such as rheumatoid arthritis. Because tumor necrosis factor-α and interleukin-1ß have the potential to influence the expression of transduction molecules such as transient receptor potential vanilloid 1 (TRPV1) in dorsal root ganglion (DRG) neurons and thus to contribute to pain we explored in the present study whether IL-17A activates DRG neurons and influences the expression of TRPV1. The IL-17A receptor was visualized in most neurons in dorsal root ganglion (DRG) sections as well as in cultured DRG neurons. Upon long-term exposure to IL-17A, isolated and cultured rat DRG neurons showed a significant upregulation of extracellular-regulated kinase (ERK) and nuclear factor κB (NFκB). Long-term exposure of neurons to IL-17A did not upregulate the expression of TRPV1. However, we found a pronounced upregulation of transient receptor potential vanilloid 4 (TRPV4) which is considered a candidate transduction molecule for mechanical hyperalgesia. Upon the injection of zymosan into the paw, IL-17A-deficient mice showed less mechanical hyperalgesia than wild type mice but thermal hyperalgesia was not attenuated in IL-17A-deficient mice. These data show, therefore, a particular role of IL-17 in mechanical hyperalgesia, and they suggest that this effect is linked to an activation and upregulation of TRPV4.


Arthritis & Rheumatism | 2012

Spinal interleukin-6 is an amplifier of arthritic pain in the rat

Enrique Vazquez; Jan Kahlenbach; Gisela Segond von Banchet; Christian König; Hans-Georg Schaible; Andrea Ebersberger

OBJECTIVE Significant joint pain is usually widespread beyond the affected joint, which results from the sensitization of nociceptive neurons in the central nervous system (central sensitization). This study was undertaken to explore whether the proinflammatory cytokine interleukin-6 (IL-6) in the joint induces central sensitization, whether joint inflammation causes the release of IL-6 from the spinal cord, and whether spinal IL-6 contributes to central sensitization. METHODS In anesthetized rats, electrophysiologic recordings of spinal cord neurons with sensory input from the knee joint were made. Neuronal responses to mechanical stimulation of the rat knee and leg were monitored. IL-6 and soluble IL-6 receptor (sIL-6R) were applied to the knee joint or the spinal cord. Spinal release of IL-6 was measured by enzyme-linked immunosorbent assay. Soluble gp130, which neutralizes IL-6/sIL-6R, was spinally applied during the development of joint inflammation or during established inflammation. RESULTS A single injection of IL-6/sIL-6R into the rat knee joint as well as application of IL-6/sIL-6R to the rat spinal cord significantly increased the responses of spinal neurons to mechanical stimulation of the knee and ankle joint, i.e., induced central sensitization. Application of soluble gp130 to the rat spinal cord attenuated this effect of IL-6. The development of knee inflammation in the rat caused spinal release of IL-6. Spinal application of soluble gp130 attenuated the development of inflammation-evoked central sensitization but did not reverse it. CONCLUSION Our findings indicate that the generation of joint pain in the rat involves not only IL-6 in the joint but also IL-6 released from the spinal cord. Spinal IL-6 contributes to central sensitization and thus promotes the widespread hyperalgesia observed in the course of joint disease.


Arthritis & Rheumatism | 2014

Involvement of Peripheral and Spinal Tumor Necrosis Factor α in Spinal Cord Hyperexcitability During Knee Joint Inflammation in Rats

Christian König; Maxim Zharsky; Christian Möller; Hans-Georg Schaible; Andrea Ebersberger

Tumor necrosis factor α (TNFα) is produced not only in peripheral tissues, but also in the spinal cord. The purpose of this study was to address the potential of peripheral and spinal TNFα to induce and maintain spinal hyperexcitability, which is a hallmark of pain states in the joints during rheumatoid arthritis and osteoarthritis.


Neuroscience | 2010

Modulation of μ-opioid receptor desensitization in peripheral sensory neurons by phosphoinositide 3-kinase γ

Christian König; O. Gavrilova-Ruch; G Segond von Banchet; R. Bauer; M. Grün; Emilio Hirsch; I. Rubio; S. Schulz; Sh Heinemann; H.-G Schaible; R. Wetzker

G protein-coupled opioid receptors undergo desensitization after prolonged agonist exposure. Recent in vitro studies of mu-opioid receptor (MOR) signaling revealed an involvement of phosphoinositide 3-kinases (PI3K) in agonist-induced MOR desensitization. Here we document a specific role of the G protein-coupled class IB isoform PI3Kgamma in MOR desensitization in mice and isolated sensory neurons. The tail-withdrawal nociception assay evidenced a compromised morphine-induced tolerance of PI3Kgamma-deficient mice compared to wild-type animals. Consistent with a role of PI3Kgamma in MOR signaling, PI3Kgamma was expressed in a subgroup of small-diameter dorsal root ganglia (DRG) along with MOR and the transient receptor potential vanilloid type 1 (TRPV1) receptor. In isolated DRG acute stimulation of MOR blocked voltage-gated calcium currents (VGCC) in both wild-type and PI3Kgamma-deficient DRG neurons. By contrast, following long-term opioid administration the attenuating effect of MOR was strongly compromised in wild-type DRG but not in PI3Kgamma-deficient DRG. Our results uncover PI3Kgamma as an essential modulator of long-term MOR desensitization and tolerance development induced by chronic opioid treatment in sensory neurons.


BMC Musculoskeletal Disorders | 2010

Loss of phosphoinositide 3-kinase γ decreases migration and activation of phagocytes but not T cell activation in antigen-induced arthritis

Michael Gruen; Christina Rose; Christian König; Mieczyslaw Gajda; Reinhard Wetzker; Rolf Bräuer

BackgroundPhosphoinositide 3-kinase γ (PI3Kγ) has been depicted as a major regulator of inflammatory processes, including leukocyte activation and migration towards several chemokines. This study aims to explore the role of PI3Kγ in the murine model of antigen-induced arthritis (AIA).MethodsDevelopment of AIA was investigated in wildtype and PI3Kγ-deficient mice as well as in mice treated with a specific inhibitor of PI3Kγ (AS-605240) in comparison to untreated animals. Inflammatory reactions of leukocytes, including macrophage and T cell activation, and macrophage migration, were studied in vivo and in vitro.ResultsGenetic deletion or pharmacological inhibition of PI3Kγ induced a marked decrease of clinical symptoms in early AIA, together with a considerably diminished macrophage migration and activation (lower production of NO, IL-1β, IL-6). Also, macrophage and neutrophil infiltration into the knee joint were impaired in vivo. However, T cell functions, measured by cytokine production (TNFα, IFNγ, IL-2, IL-4, IL-5, IL-17) in vitro and DTH reaction in vivo were not altered, and accordingly, disease developed normally at later timepointsConclusionPI3Kγ specifically affects phagocyte function in the AIA model but has no impact on T cell activation.


Journal of Cancer Research and Clinical Oncology | 2010

Targeting PI3K in neuroblastoma

Volker Spitzenberg; Christian König; Susanne Ulm; Romina Marone; Luise Röpke; Jörg P. Müller; Michael Grün; Reinhard Bauer; Ignacio Rubio; Matthias P. Wymann; Astrid Voigt; Reinhard Wetzker

PurposeThis work employs pharmacological targeting of phosphoinositide 3-kinases (PI3K) in selected neuroblastoma (NB) tumors with the inhibitor AS605240, which has been shown to express low toxicity and relative specificity for the PI3K species γ.MethodsThe expression pattern of PI3K isoforms in 7 NB cell lines and 14 tumor patient samples was determined by Western blotting and immunocytochemistry. The effect of AS605240 on the growth of four selected tumor cell lines was assessed. Two cell lines exhibiting (SK-N-LO) or lacking (SK-N-AS) PI3Kγ expression were chosen for further in vitro analysis, which involved propidium iodide (PI)-based cell cycle staining, terminal deoxynucleotidyl transferase–mediated dUTP-biotin nick end labeling (TUNEL-staining) of apoptotic cells and analysis of PI3K/Akt-related signaling pathways via Western blotting and translocation experiments. The action of AS605240 in vivo was addressed by xenograft experiments in severe combined immunodeficiency (SCID) mice, thereby comparing SK-N-LO and SK-N-AS derived tumors. Apoptosis induced in SK-N-LO tumors was shown by immunohistochemical TUNEL-staining.ResultsSignificant expression of PI3Kγ in neuroblastoma patient biopsies and tumor cell lines was detected. AS605240 induced apoptosis in NB cell lines proportional to this expression and suppressed growth of PI3Kγ positive, but not negative, tumors in a xenograft mouse model. No adverse effects of the inhibitor treatment were observed.ConclusionsOur observations hint to an oncogenic function of PI3Kγ in distinct neuroblastoma entities and reveal PI3K targeting by AS605240 as a promising molecular therapy of these tumors.


The Journal of Neuroscience | 2016

Involvement of Spinal IL-6 Trans-Signaling in the Induction of Hyperexcitability of Deep Dorsal Horn Neurons by Spinal Tumor Necrosis Factor-Alpha

Christian König; Eric Morch; Annett Eitner; Christian Möller; Brian Turnquist; Hans-Georg Schaible; Andrea Ebersberger

During peripheral inflammation, both spinal TNF-α and IL-6 are released within the spinal cord and support the generation of inflammation-evoked spinal hyperexcitability. However, whether spinal TNF-α and IL-6 act independently in parallel or in a functionally dependent manner has not been investigated. In extracellular recordings from mechanonociceptive deep dorsal horn neurons of normal rats in vivo, we found that spinal application of TNF-α increased spinal neuronal responses to mechanical stimulation of knee and ankle joints. This effect was significantly attenuated by either sgp130, which blocks IL-6 trans-signaling mediated by IL-6 and its soluble receptor IL-6R (sIL-6R); by an antibody to the IL-6 receptor; or by minocycline, which inhibits the microglia. IL-6 was localized in neurons of the spinal cord and, upon peripheral noxious stimulation in the presence of spinal TNF-α, IL-6 was released spinally. Furthermore, TNF-α recruited microglial cells to provide sIL-6R, which can form complexes with IL-6. Spinal application of IL-6 plus sIL-6R, but not of IL-6 alone, enhanced spinal hyperexcitability similar to TNF-α and the inhibition of TNF-α-induced hyperexcitability by minocycline was overcome by coadministration of sIL-6R, showing that sIL-6R is required. Neither minocycline nor the TNF-α-neutralizing compound etanercept inhibited the induction of hyperexcitability by IL-6 plus sIL-6R. Together, these data show that the induction of hyperexcitability of nociceptive deep dorsal horn neurons by TNF-α largely depends on the formation of IL-6/sIL-6R complexes that are downstream of TNF-α and requires the interactions of neurons and microglia orchestrated by TNF-α. SIGNIFICANCE STATEMENT Both spinal TNF-α and IL-6 induce a state of spinal hyperexcitability. We present the novel finding that the full effect of TNF-α on the development of spinal hyperexcitability depends on IL-6 trans-signaling acting downstream of TNF-α. IL-6 trans-signaling requires the formation of complexes of IL-6 and soluble IL-6 receptor. Spinal TNF-α furthers the release of IL-6 from neurons in the spinal cord during peripheral noxious stimulation and recruits microglial cells to provide soluble IL-6 receptor, which can form complexes with IL-6. Therefore, a specific interaction between neurons and microglia is required for the full development of TNF-α-induced hyperexcitability of nociceptive deep horsal horn neurons.


Arthritis & Rheumatism | 2016

Long-Lasting Activation of the Transcription Factor CREB in Sensory Neurons by Interleukin-1β During Antigen-Induced Arthritis in Rats: A Mechanism of Persistent Arthritis Pain?

Gisela Segond von Banchet; Christian König; Jessica Patzer; Annett Eitner; Johannes Leuchtweis; Matthias Ebbinghaus; Michael Karl Boettger; Hans-Georg Schaible

In spite of successful treatment of immune‐mediated arthritis, many patients still experience pain. We undertook this study to investigate whether antigen‐induced arthritis (AIA) in rats triggers neuronal changes in sensory neurons that outlast the inflammatory process.


British Journal of Pharmacology | 2014

PI3Kγ integrates cAMP and Akt signalling of the μ-opioid receptor.

Sreedhar Madishetti; Nadine Schneble; Christian König; Emilio Hirsch; Stefan Schulz; Jörg P. Müller; Reinhard Wetzker

The μ‐opioid receptor has been characterized as the main mediator of opioid signalling in neuronal cells. Opioid‐induced pain suppression was originally proposed to be mediated by μ‐opioid receptor‐induced inhibitory effects on cAMP, which is known to mediate inflammatory hypernociception. Recent investigations revealed PI3Kγ and Akt (PKB) as additional elements of μ‐opioid receptor signalling. Hence, we investigated the interaction between pronociceptive cAMP and antinociceptive PI3K/Akt signalling pathways.

Collaboration


Dive into the Christian König's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge