Network


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

Hotspot


Dive into the research topics where Peter Lundbäck is active.

Publication


Featured researches published by Peter Lundbäck.


Nature | 2012

Novel role of PKR in inflammasome activation and HMGB1 release

Ben Lu; Takahisa Nakamura; Karen Inouye; Jianhua Li; Yiting Tang; Peter Lundbäck; Sergio Valdes-Ferrer; Peder S. Olofsson; Thomas Kalb; Jesse Roth; Yong-Rui Zou; Helena Erlandsson-Harris; Huan Yang; Jenny P.-Y. Ting; Haichao Wang; Ulf Andersson; Daniel J. Antoine; Sangeeta Chavan; Gökhan S. Hotamisligil; Kevin J. Tracey

The inflammasome regulates the release of caspase activation-dependent cytokines, including interleukin (IL)-1β, IL-18 and high-mobility group box 1 (HMGB1). By studying HMGB1 release mechanisms, here we identify a role for double-stranded RNA-dependent protein kinase (PKR, also known as EIF2AK2) in inflammasome activation. Exposure of macrophages to inflammasome agonists induced PKR autophosphorylation. PKR inactivation by genetic deletion or pharmacological inhibition severely impaired inflammasome activation in response to double-stranded RNA, ATP, monosodium urate, adjuvant aluminium, rotenone, live Escherichia coli, anthrax lethal toxin, DNA transfection and Salmonella typhimurium infection. PKR deficiency significantly inhibited the secretion of IL-1β, IL-18 and HMGB1 in E. coli-induced peritonitis. PKR physically interacts with several inflammasome components, including NOD-like receptor (NLR) family pyrin domain-containing 3 (NLRP3), NLRP1, NLR family CARD domain-containing protein 4 (NLRC4), absent in melanoma 2 (AIM2), and broadly regulates inflammasome activation. PKR autophosphorylation in a cell-free system with recombinant NLRP3, apoptosis-associated speck-like protein containing a CARD (ASC, also known as PYCARD) and pro-caspase-1 reconstitutes inflammasome activity. These results show a crucial role for PKR in inflammasome activation, and indicate that it should be possible to pharmacologically target this molecule to treat inflammation.


Molecular Medicine | 2012

Redox modification of cysteine residues regulates the cytokine activity of high mobility group box-1 (HMGB1).

Huan Yang; Peter Lundbäck; Lars Ottosson; Helena Erlandsson-Harris; Emilie Venereau; Marco Bianchi; Yousef Al-Abed; Ulf Andersson; Kevin J. Tracey; Daniel J. Antoine

High mobility group box 1 (HMGB1) is a nuclear protein with extracellular inflammatory cytokine activity. It is released passively during cell injury and necrosis, and secreted actively by immune cells. HMGB1 contains three conserved redox-sensitive cysteine residues: C23 and C45 can form an intramolecular disulfide bond, whereas C106 is unpaired and is essential for the interaction with Toll-Like Receptor (TLR) 4. However, a comprehensive characterization of the dynamic redox states of each cysteine residue and of their impacts on innate immune responses is lacking. Using tandem mass spectrometric analysis, we now have established that the C106 thiol and the C23-C45 disulfide bond are required for HMGB1 to induce nuclear NF-κB translocation and tumor necrosis factor (TNF) production in macrophages. Both irreversible oxidation to sulphonates and complete reduction to thiols of these cysteines inhibited TNF production markedly. In a proof of concept murine model of hepatic necrosis induced by acetaminophen, during inflammation, the predominant form of serum HMGB1 is the active one, containing a C106 thiol group and a disulfide bond between C23 and C45, whereas the inactive form of HMGB1, containing terminally oxidized cysteines, accumulates during inflammation resolution and hepatic regeneration. These results reveal critical posttranslational redox mechanisms that control the proinflammatory activity of HMGB1 and its inactivation during pathogenesis.


Proceedings of the National Academy of Sciences of the United States of America | 2014

JAK/STAT1 signaling promotes HMGB1 hyperacetylation and nuclear translocation

Ben Lu; Daniel J. Antoine; Kevin Kwan; Peter Lundbäck; H Wähämaa; Hanna Schierbeck; Melissa Robinson; Marieke A. D. van Zoelen; Huan Yang; Jianhua Li; Helena Erlandsson-Harris; Sangeeta Chavan; Haichao Wang; Ulf Andersson; Kevin J. Tracey

Significance High-mobility group box (HMGB)1 is a nuclear protein that we have identified as a proinflammatory mediator during infection or sterile tissue injury, which importantly orchestrates the innate immune responses. The mechanisms of HMGB1 release require translocation of HMGB1 from nucleus to cytoplasm and release into the extracellular space. We recently reported that the inflammasome and PKR mediates HMGB1 release from the cytoplasm, but the mechanism of HMGB1 translocation from nucleus to cytoplasm was previously unknown. Here, we describe our discovery that JAK/STAT1 is required for LPS- or interferon-induced HMGB1 nuclear translocation. These findings have significant implications for the field, and for designing therapeutics for potential use in inflammatory diseases. Extracellular high-mobility group box (HMGB)1 mediates inflammation during sterile and infectious injury and contributes importantly to disease pathogenesis. The first critical step in the release of HMGB1 from activated immune cells is mobilization from the nucleus to the cytoplasm, a process dependent upon hyperacetylation within two HMGB1 nuclear localization sequence (NLS) sites. The inflammasomes mediate the release of cytoplasmic HMGB1 in activated immune cells, but the mechanism of HMGB1 translocation from nucleus to cytoplasm was previously unknown. Here, we show that pharmacological inhibition of JAK/STAT1 inhibits LPS-induced HMGB1 nuclear translocation. Conversely, activation of JAK/STAT1 by type 1 interferon (IFN) stimulation induces HMGB1 translocation from nucleus to cytoplasm. Mass spectrometric analysis unequivocally revealed that pharmacological inhibition of the JAK/STAT1 pathway or genetic deletion of STAT1 abrogated LPS- or type 1 IFN-induced HMGB1 acetylation within the NLS sites. Together, these results identify a critical role of the JAK/STAT1 pathway in mediating HMGB1 cytoplasmic accumulation for subsequent release, suggesting that the JAK/STAT1 pathway is a potential drug target for inhibiting HMGB1 release.


Journal of Experimental Medicine | 2015

MD-2 is required for disulfide HMGB1–dependent TLR4 signaling

Huan Yang; Haichao Wang; Zhongliang Ju; Ahmed A. Ragab; Peter Lundbäck; Wei Long; Sergio Valdes-Ferrer; Mingzhu He; John P. Pribis; Jianhua Li; Ben Lu; Domokos Gero; Csaba Szabó; Daniel J. Antoine; Helena Erlandsson Harris; Doug T. Golenbock; Jianmin Meng; Jesse Roth; Sangeeta Chavan; Ulf Andersson; Timothy R. Billiar; Kevin J. Tracey; Yousef Al-Abed

Yang et al. show that a disulfide isoform of HMGB1, with a role in TLR4 signaling, physically interacts with and binds MD-2. MD-2 deficiency in macrophage cell lines or in primary mouse macrophages stimulated with HMGB1 implicates MD-2 in TLR4 signaling. They also identify an HGMB1 peptide inhibitor, P5779, which when administered in vivo can protect mice from acetaminophen-induced hepatoxicity, ischemia/reperfusion injury, and sepsis.


Molecular Medicine | 2011

Monoclonal anti-HMGB1 (high mobility group box chromosomal protein 1) antibody protection in two experimental arthritis models.

Hanna Schierbeck; Peter Lundbäck; Karin Palmblad; Lena Klevenvall; Helena Erlandsson-Harris; Ulf Andersson; Lars Ottosson

High mobility group box chromosomal protein 1 (HMGB1) is a DNA-binding nuclear protein that can be released from dying cells and activated myeloid cells. Extracellularly, HMGB1 promotes inflammation. Experimental studies demonstrate HMGB1 to be a pathogenic factor in many inflammatory conditions including arthritis. HMGB1-blocking therapies in arthritis models alleviate disease and confer significant protection against cartilage and bone destruction. So far, the most successful HMGB1-targeted therapies have been demonstrated with HMGB1-specific polyclonal antibodies and with recombinant A box protein, a fragment of HMGB1. The present study is the first to evaluate the potential of a monoclonal anti-HMGB1 antibody (2G7, mouse IgG2b) to ameliorate arthritis. Effects of repeated injections of this antibody have now been studied in two conceptually different models of arthritis: collagen type II-induced arthritis (CIA) in DBA/1 mice and in a spontaneous arthritis disease in mice with combined deficiencies for genes encoding for the enzyme DNase type II and interferon type I receptors. These mice are unable to degrade phagocytozed DNA in macrophages and develop chronic, destructive polyarthritis. Therapeutic intervention in CIA and prophylactic administration of anti-HMGB1 monoclonal antibody (mAb) in the spontaneous arthritis model significantly ameliorated the clinical courses. Anti-HMGB1 mAb therapy also partially prevented joint destruction, as demonstrated by histological examination. The beneficial antiarthritic effects by the anti-HMGB1 mAb in two diverse models of arthritis represent additional proof-of-concept, indicating that HMGB1 may be a valid target molecule to consider for development of future clinical therapy.


Pain | 2014

Spinal HMGB1 induces TLR4-mediated long-lasting hypersensitivity and glial activation and regulates pain-like behavior in experimental arthritis.

Nilesh M. Agalave; Max Larsson; Sally Abdelmoaty; Jie Su; Azar Baharpoor; Peter Lundbäck; Karin Palmblad; Ulf Andersson; Helena Erlandsson Harris; Camilla I. Svensson

Summary Spinal injection of disulfide extracellular high mobility group box‐1 protein (HMGB1) induces mechanical hypersensitivity and spinal glial activation, and inhibition of spinal HMGB1 resolves mechanical hypersensitivity induced by collagen antibody‐induced arthritis. ABSTRACT Extracellular high mobility group box‐1 protein (HMGB1) plays important roles in the pathogenesis of nerve injury‐ and cancer‐induced pain. However, the involvement of spinal HMGB1 in arthritis‐induced pain has not been examined previously and is the focus of this study. Immunohistochemistry showed that HMGB1 is expressed in neurons and glial cells in the spinal cord. Subsequent to induction of collagen antibody‐induced arthritis (CAIA), Hmgb1 mRNA and extranuclear protein levels were significantly increased in the lumbar spinal cord. Intrathecal (i.t.) injection of a neutralizing anti‐HMGB1 monoclonal antibody or recombinant HMGB1 box A peptide (Abox), which each prevent extracellular HMGB1 activities, reversed CAIA‐induced mechanical hypersensitivity. This occurred during ongoing joint inflammation as well as during the postinflammatory phase, indicating that spinal HMGB1 has an important function in nociception persisting beyond episodes of joint inflammation. Importantly, only HMGB1 in its partially oxidized isoform (disulfide HMGB1), which activates toll‐like receptor 4 (TLR4), but not in its fully reduced or fully oxidized isoforms, evoked mechanical hypersensitivity upon i.t. injection. Interestingly, although both male and female mice developed mechanical hypersensitivity in response to i.t. HMGB1, female mice recovered faster. Furthermore, the pro‐nociceptive effect of i.t. injection of HMGB1 persisted in Tlr2‐ and Rage‐, but was absent in Tlr4‐deficient mice. The same pattern was observed for HMGB1‐induced spinal microglia and astrocyte activation and cytokine induction. These results demonstrate that spinal HMGB1 contributes to nociceptive signal transmission via activation of TLR4 and point to disulfide HMGB1 inhibition as a potential therapeutic strategy in treatment of chronic inflammatory pain.


The EMBO Journal | 2012

TLR activation regulates damage‐associated molecular pattern isoforms released during pyroptosis

Sanna Nyström; Daniel J. Antoine; Peter Lundbäck; John G. Lock; Andreia Florina Nita; Kari Högstrand; Alf Grandien; Helena Erlandsson-Harris; Ulf Andersson; Steven E. Applequist

Infection of macrophages by bacterial pathogens can trigger Toll‐like receptor (TLR) activation as well as Nod‐like receptors (NLRs) leading to inflammasome formation and cell death dependent on caspase‐1 (pyroptosis). Complicating the study of inflammasome activation is priming. Here, we develop a priming‐free NLRC4 inflammasome activation system to address the necessity and role of priming in pyroptotic cell death and damage‐associated molecular pattern (DAMP) release. We find pyroptosis is not dependent on priming and when priming is re‐introduced pyroptosis is unaffected. Cells undergoing unprimed pyroptosis appear to be independent of mitochondrial involvement and do not produce inflammatory cytokines, nitrous oxide (NO), or reactive oxygen species (ROS). Nevertheless, they undergo an explosive cell death releasing a chemotactic isoform of the DAMP high mobility group protein box 1 (HMGB1). Importantly, priming through surface TLRs but not endosomal TLRs during pyroptosis leads to the release of a new TLR4‐agonist cysteine redox isoform of HMGB1. These results show that pyroptosis is dominant to priming signals and indicates that metabolic changes triggered by priming can affect how cell death is perceived by the immune system.


Hepatology | 2016

A novel high mobility group box 1 neutralizing chimeric antibody attenuates drug-induced liver injury and postinjury inflammation in mice

Peter Lundbäck; Jonathan D. Lea; Agnieszka Sowinska; Lars Ottosson; Camilla Melin Fürst; Johanna Steen; C. Aulin; Joanna I. Clarke; Anja Kipar; Lena Klevenvall; Huan Yang; Karin Palmblad; B. Kevin Park; Kevin J. Tracey; Anna M. Blom; Ulf Andersson; Daniel J. Antoine; Helena Erlandsson Harris

Acetaminophen (APAP) overdoses are of major clinical concern. Growing evidence underlines a pathogenic contribution of sterile postinjury inflammation in APAP‐induced acute liver injury (APAP‐ALI) and justifies development of anti‐inflammatory therapies with therapeutic efficacy beyond the therapeutic window of the only current treatment option, N‐acetylcysteine (NAC). The inflammatory mediator, high mobility group box 1 (HMGB1), is a key regulator of a range of liver injury conditions and is elevated in clinical and preclinical APAP‐ALI. The anti‐HMGB1 antibody (m2G7) is therapeutically beneficial in multiple inflammatory conditions, and anti‐HMGB1 polyclonal antibody treatment improves survival in a model of APAP‐ALI. Herein, we developed and investigated the therapeutic efficacy of a partly humanized anti‐HMGB1 monoclonal antibody (mAb; h2G7) and identified its mechanism of action in preclinical APAP‐ALI. The mouse anti‐HMGB1 mAb (m2G7) was partly humanized (h2G7) by merging variable domains of m2G7 with human antibody‐Fc backbones. Effector function‐deficient variants of h2G7 were assessed in comparison with h2G7 in vitro and in preclinical APAP‐ALI. h2G7 retained identical antigen specificity and comparable affinity as m2G7. 2G7 treatments significantly attenuated APAP‐induced serum elevations of alanine aminotransferase and microRNA‐122 and completely abrogated markers of APAP‐induced inflammation (tumor necrosis factor, monocyte chemoattractant protein 1, and chemokine [C‐X‐C motif] ligand 1) with prolonged therapeutic efficacy as compared to NAC. Removal of complement and/or Fc receptor binding did not affect h2G7 efficacy. Conclusion: This is the first report describing the generation of a partly humanized HMGB1‐neutralizing antibody with validated therapeutic efficacy and with a prolonged therapeutic window, as compared to NAC, in APAP‐ALI. The therapeutic effect was mediated by HMGB1 neutralization and attenuation of postinjury inflammation. These results represent important progress toward clinical implementation of HMGB1‐specific therapy as a means to treat APAP‐ALI and other inflammatory conditions. (Hepatology 2016;64:1699‐1710).


Annals of the Rheumatic Diseases | 2012

The pro-inflammatory effect of HMGB1, a mediator of inflammation in arthritis, is dependent on the redox status of the protein

Lars Ottosson; Peter Lundbäck; Hulda Hreggvidsdottir; Huan Yang; Kevin J. Tracey; Daniel J. Antoine; Helena Erlandsson-Harris; Ulf Andersson

Background and objectives High mobility group box protein 1 (HMGB1) is a nuclear protein involved in chromatin architecture and is present in all cells. HMGB1 also functions as a prototype alarmin and is released passively during necrotic cell death, as well as actively secreted from certain immunocompetent cells. Levels of HMGB1 are highly elevated in synovial tissue of rheumatoid arthritis patients, and the protein is an important mediator of different inflammatory processes. Blocking the inflammatory effect of HMGB1 with antibodies or specific inhibitors ameliorates disease in different experimental arthritis models. In this study, the authors wanted to investigate if the redox status of HMGB1 is important for the pro-inflammatory effect of the protein. We also investigated if the presence of the redox sensitive cysteine is necessary for binding of HMGB1 to its receptor TLR4/MD2. Methods To determine the redox status of HMGB1, meaning the three cysteines present in the molecule, the authors used tryptic digestion followed by liquid chromatography electrospray tandem mass spectrometry. To investigate the pro-inflammatory effect of HMGB1 and HMGB1-mutants, primary human and murine macrophages and macrophage-like RAW 264.7-cells were stimulated with HMGB1 and cytokine production was measured by ELISA. Binding studies of HMGB1 to its receptor TLR4/MD2, and investigation of the importance of redox status, was performed by Surface-Plasmon Resonance analysis. Results The authors could determine that, in order for HMGB1 to have a pro-inflammatory effect, the redox sensitive cysteine at position 106 (C106) needs to be in a reduced thiol state and the cysteines at position 23 and 45 have to form a disulfide bridge. Oxidation of C106 or lack of a disulfide bridge between C23 and C45 then causes HMGB1 to lose its pro-inflammatory effect. In Surface-Plasmon Resonance experiments the authors could demonstrate that a substitution of C106 for an alanine or a serine inhibited the binding of HMGB1 to its receptor TLR4/MD2. Conclusion The pro-inflammatory effect of HMGB1 is dependent on the redox status of all three present cysteines. The reduced C106 is necessary for the binding of HMGB1 to its receptor TLR4/MD2 explaining the lack of cytokine inducing effect of HMGB1 where C106 has been substituted for another amino acid or if the cysteine has been oxidised. The knowledge of the importance of the redox status of HMGB1 can promote the development of therapies targeted at only blocking HMGB1 when it is in its pro-inflammatory form.


Scandinavian Journal of Pain | 2015

Spinal disulfide HMGB1, but not all-thiol HMGB1, induces mechanical hypersensitivity in a TLR4-dependent manner

Nilesh M. Agalave; Sally Abdelmoaty; Peter Lundbäck; Ulf Andersson; Helena Erlandsson Harris; Camilla I. Svensson

Abstract Aims Increasing evidence indicates that extracellular high mobility group box-1 protein (HMGB1) is involved in the pathogenesis of inflammatory and autoimmune disease. Data from our laboratory demonstrates that HMGB1 contributes to nociceptive behavior in a model of rheumatoid arthritis-induced pain. HMGB1 binds to multiple receptors, including toll like receptor (TLR) 2, TLR4 and receptor for advanced glycation end products (RAGE). When the cysteine in position C106 is in the reduced thiol form and C23 and C45 are engaged in a disulfide bridge (disulfide HMGB1), the molecule functions as a cytokine-inducing TLR4 ligand. In contrast, when these three cysteines are all reduced (all-thiol HMGB1), HMGB1 exclusively potentiates chemotactic activity via CXCR4. It is currently not well understood which receptor and which redox form of HMGB1 that mediates pain hypersensitivity and is therefore the aim of this study. Methods All animal work was carried out in accordance with protocol approved by the local ethics committee for animal experiments in Sweden. Balb/c, C57B/l6 (WT), Tlr2–/–, Tlr4–/– and Rage–/– male mice were used for this study. Disulfide (ds) and all thiol (at) form of HMGB1 were injected intrathecally (1 μg) and mechanical hypersensitivity assessed by von Frey filaments. Lumbar spinal cords were collected after i.t. injection of atHMGB1 and ds HMGB1 and mRNA levels for cytokine and glia markers assessed by quantitative PCR. Results In Balb/c and C57Bl/6 WT mice, i.t injection of dsHMGB1, but not atHMGB1, led to a significant reduction in mechanical thresholds. dsHMGB1 induced mechanical hypersensitivity 6 h after i.t. injection, which lasted for 5 days, compared to i.t. injection of saline. When dsHMGB1 was injected i.t. to Tlr4 deficient mice it did not induce mechanical hypersensitivity. In contrast Tlr2 and Rage deficient mice were still susceptible to dsHMGB1-induced mechanical hypersensitivity. Analysis of mRNA for cytokines and glial cell-associated factors in lumbar spinal cords revealed increased levels of Tnf, Ccl2, Cxcl1, Cxcl2, Gfap and Cd11b in mice injected with dsHMGB1, but not atHMGB1, with exception for Il1β and Cxcr3 that was induced also by atHMGB1. Intrathecal injection of dsHMGB1 to Tlr4–/– deficient mice, did not increase mRNA levels for Tnf, Il1β, Ccl2, Gfap and Cd11b. Conclusions We found the i.t. injection of the disulfide, but not the all-thiol, form of HMGB1 to induce pronouncedand long-lasting mechanical hypersensitivity, glial reactivity and cytokine induction in a TLR4-, but not TLR2- or RAGE-dependent manner. Thus our data indicates that, the redox state of HMGB1 is key for determining its nociceptive property and receptor usage and thus also the functional consequences of HMGB1 release. Agents interfering with extracellular HMGB1 may be considered in the development of new pain relieving therapeutics.

Collaboration


Dive into the Peter Lundbäck's collaboration.

Top Co-Authors

Avatar

Ulf Andersson

Karolinska University Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Huan Yang

The Feinstein Institute for Medical Research

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kevin J. Tracey

The Feinstein Institute for Medical Research

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge