Network


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

Hotspot


Dive into the research topics where Marco Sisignano is active.

Publication


Featured researches published by Marco Sisignano.


The Journal of Neuroscience | 2012

5,6-EET Is Released upon Neuronal Activity and Induces Mechanical Pain Hypersensitivity via TRPA1 on Central Afferent Terminals

Marco Sisignano; Chul-Kyu Park; Carlo Angioni; Dong Dong Zhang; Christian von Hehn; Enrique J. Cobos; Nader Ghasemlou; Zhen-Zhong Xu; Vigneswara Kumaran; Ruirui Lu; Andrew D. Grant; Michael J. M. Fischer; Achim Schmidtko; Peter W. Reeh; Ru-Rong Ji; Clifford J. Woolf; Gerd Geisslinger; Klaus Scholich; Christian Brenneis

Epoxyeicosatrienoic acids (EETs) are cytochrome P450-epoxygenase-derived metabolites of arachidonic acid that act as endogenous signaling molecules in multiple biological systems. Here we have investigated the specific contribution of 5,6-EET to transient receptor potential (TRP) channel activation in nociceptor neurons and its consequence for nociceptive processing. We found that, during capsaicin-induced nociception, 5,6-EET levels increased in dorsal root ganglia (DRGs) and the dorsal spinal cord, and 5,6-EET is released from activated sensory neurons in vitro. 5,6-EET potently induced a calcium flux (100 nm) in cultured DRG neurons that was completely abolished when TRPA1 was deleted or inhibited. In spinal cord slices, 5,6-EET dose dependently enhanced the frequency, but not the amplitude, of spontaneous EPSCs (sEPSCs) in lamina II neurons that also responded to mustard oil (allyl isothiocyanate), indicating a presynaptic action. Furthermore, 5,6-EET-induced enhancement of sEPSC frequency was abolished in TRPA1-null mice, suggesting that 5,6-EET presynaptically facilitated spinal cord synaptic transmission by TRPA1. Finally, in vivo intrathecal injection of 5,6-EET caused mechanical allodynia in wild-type but not TRPA1-null mice. We conclude that 5,6-EET is synthesized on the acute activation of nociceptors and can produce mechanical hypersensitivity via TRPA1 at central afferent terminals in the spinal cord.


The Journal of Neuroscience | 2013

Phenotyping the Function of TRPV1-Expressing Sensory Neurons by Targeted Axonal Silencing

Christian Brenneis; Katrin Kistner; Michelino Puopolo; David Segal; David W. Roberson; Marco Sisignano; Sandra Labocha; Nerea Ferreirós; Amanda Strominger; Enrique J. Cobos; Nader Ghasemlou; Gerd Geisslinger; Peter W. Reeh; Bruce P. Bean; Clifford J. Woolf

Specific somatosensations may be processed by different subsets of primary afferents. C-fibers expressing heat-sensitive TRPV1 channels are proposed, for example, to be heat but not mechanical pain detectors. To phenotype in rats the sensory function of TRPV1+ afferents, we rapidly and selectively silenced only their activity, by introducing the membrane-impermeant sodium channel blocker QX-314 into these axons via the TRPV1 channel pore. Using tandem mass spectrometry we show that upon activation with capsaicin, QX-314 selectively accumulates in the cytosol only of TRPV1-expressing cells, and not in control cells. Exposure to QX-314 and capsaicin induces in small DRG neurons a robust sodium current block within 30 s. In sciatic nerves, application of extracellular QX-314 with capsaicin persistently reduces C-fiber but not A-fiber compound action potentials and this effect does not occur in TRPV1−/− mice. Behavioral phenotyping after selectively silencing TRPV1+ sciatic nerve axons by perineural injections of QX-314 and capsaicin reveals deficits in heat and mechanical pressure but not pinprick or light touch perception. The response to intraplantar capsaicin is substantially reduced, as expected. During inflammation, silencing TRPV1+ axons abolishes heat, mechanical, and cold hyperalgesia but tactile and cold allodynia remain following peripheral nerve injury. These results indicate that TRPV1-expressing sensory neurons process particular thermal and mechanical somatosensations, and that the sensory channels activated by mechanical and cold stimuli to produce pain in naive/inflamed rats differ from those in animals after peripheral nerve injury.


Nature Reviews Neurology | 2014

Mechanism-based treatment for chemotherapy-induced peripheral neuropathic pain

Marco Sisignano; Ralf Baron; Klaus Scholich; Gerd Geisslinger

Chemotherapy-induced peripheral neuropathic pain (CIPNP)—a severe adverse effect observed in up to 80% of patients during treatment with antineoplastic drugs—limits the tolerable dose of cytostatics, and can lead to discontinuation of chemotherapy. Many drugs that are approved for the treatment of other neuropathic pain states have shown little or no analgesic effect on CIPNP in large randomized, placebo-controlled clinical trials. Here, we review the known mechanisms of CIPNP induced by the three most commonly used cytostatics: paclitaxel, oxaliplatin and vincristine. These substances have distinct neurotoxic and neuroinflammatory properties, but they also have overlapping contributions to pathogenesis of CIPNP that could potentially be targeted for prevention or treatment of CIPNP. We discuss the failure of previously tested antioxidants, neuroprotective agents, anticonvulsants and antidepressants as therapeutic or preventative strategies, and suggest individualized, mechanism-based therapeutic options for CIPNP associated with each of the three main drug groups. We point out the necessity to assess drug efficacy in CIPNP independently of other neuropathic pain states, and emphasize the need for delineation of subpopulations of patients with CIPNP for more-efficient treatment. Finally, we discuss novel therapeutic strategies and recent progress in treatment of CIPNP, and evaluate the potential benefits of these recent proceedings for future therapies.


Progress in Lipid Research | 2014

TRP-channels as key integrators of lipid pathways in nociceptive neurons.

Marco Sisignano; David L. H. Bennett; Gerd Geisslinger; Klaus Scholich

TRP-channels are the most prominent family of ligand-gated ion channels for pain perception. In sensory neurons, TRPV1-V4, TRPA1 and TRPM8 are expressed and are responsible for the conversion of external stimuli to painful sensations. Under pathophysiological conditions, excessive activity of TRP-channels leads to mechanical allodynia and thermal hyperalgesia. Among the endogenous TRP-channel sensitizers, activators and inhibitors, more than 50 arachidonic acid- and linoleic acid-metabolites from the COX-, LOX- and CYP-pathways, as well as lysophospholipids and isoprenoids can be found. As a consequence, these lipids represent the vast majority of endogenous TRP-channel modulators in sensory neurons. Although the precise mechanisms of TRP-channel modulation by most lipids are still unknown, it became clear that lipids can either bind directly to the target TRP-channel or modulate TRP-channels indirectly by activating G-protein coupled receptors. Thus, TRP-channels seem to be key sensors for lipids, integrating and interpreting incoming signals from the different metabolic lipid pathways. Here, we discuss the specific properties of the currently known endogenous lipid-derived TRP-channel modulators concerning their ability to activate or inhibit TRP-channels, the molecular mechanisms of lipid/TRP-channel interactions and specific TRP-regulatory characteristics of the individual lipid families.


Molecular Pain | 2011

Soluble epoxide hydrolase limits mechanical hyperalgesia during inflammation

Christian Brenneis; Marco Sisignano; Ovidiu Coste; Kai Altenrath; Michael J. M. Fischer; Carlo Angioni; Ingrid Fleming; Ralf P. Brandes; Peter W. Reeh; Clifford J. Woolf; Gerd Geisslinger; Klaus Scholich

BackgroundCytochrome-P450 (CYP450) epoxygenases metabolise arachidonic acid (AA) into four different biologically active epoxyeicosatrienoic acid (EET) regioisomers. Three of the EETs (i.e., 8,9-, 11,12- and 14,15-EET) are rapidly hydrolysed by the enzyme soluble epoxide hydrolase (sEH). Here, we investigated the role of sEH in nociceptive processing during peripheral inflammation.ResultsIn dorsal root ganglia (DRG), we found that sEH is expressed in medium and large diameter neurofilament 200-positive neurons. Isolated DRG-neurons from sEH-/- mice showed higher EET and lower DHET levels. Upon AA stimulation, the largest changes in EET levels occurred in culture media, indicating both that cell associated EET concentrations quickly reach saturation and EET-hydrolyzing activity mostly effects extracellular EET signaling. In vivo, DRGs from sEH-deficient mice exhibited elevated 8,9-, 11,12- and 14,15-EET-levels. Interestingly, EET levels did not increase at the site of zymosan-induced inflammation. Cellular imaging experiments revealed direct calcium flux responses to 8,9-EET in a subpopulation of nociceptors. In addition, 8,9-EET sensitized AITC-induced calcium increases in DRG neurons and AITC-induced calcitonin gene related peptide (CGRP) release from sciatic nerve axons, indicating that 8,9-EET sensitizes TRPA1-expressing neurons, which are known to contribute to mechanical hyperalgesia. Supporting this, sEH-/- mice showed increased nociceptive responses to mechanical stimulation during zymosan-induced inflammation and 8,9-EET injection reduced mechanical thresholds in naive mice.ConclusionOur results show that the sEH can regulate mechanical hyperalgesia during inflammation by inactivating 8,9-EET, which sensitizes TRPA1-expressing nociceptors. Therefore we suggest that influencing the CYP450 pathway, which is actually highly considered to treat cardiovascular diseases, may cause pain side effects.


PLOS ONE | 2013

Synthesis of lipid mediators during UVB-induced inflammatory hyperalgesia in rats and mice.

Marco Sisignano; Carlo Angioni; Nerea Ferreirós; Claus-Dieter Schuh; Jing Suo; Yannick Schreiber; John M. Dawes; Ana Antunes-Martins; David L. H. Bennett; Stephen B. McMahon; Gerd Geisslinger; Klaus Scholich

Peripheral sensitization during inflammatory pain is mediated by a variety of endogenous proalgesic mediators including a number of oxidized lipids, some of which serve endogenous modulators of sensory TRP-channels. These lipids are eicosanoids of the arachidonic acid and linoleic acid pathway, as well as lysophophatidic acids (LPAs). However, their regulation pattern during inflammatory pain and their contribution to peripheral sensitization is still unclear. Here, we used the UVB-model for inflammatory pain to investigate alterations of lipid concentrations at the site of inflammation, the dorsal root ganglia (DRGs) as well as the spinal dorsal horn and quantified 21 lipid species from five different lipid families at the peak of inflammation 48 hours post irradiation. We found that known proinflammatory lipids as well as lipids with unknown roles in inflammatory pain to be strongly increased in the skin, whereas surprisingly little changes of lipid levels were seen in DRGs or the dorsal horn. Importantly, although there are profound differences between the number of cytochrome (CYP) genes between mice and rats, CYP-derived lipids were regulated similarly in both species. Since TRPV1 agonists such as LPA 18∶1, 9- and 13-HODE, 5- and 12-HETE were elevated in the skin, they may contribute to thermal hyperalgesia and mechanical allodynia during UVB-induced inflammatory pain. These results may explain why some studies show relatively weak analgesic effects of cyclooxygenase inhibitors in UVB-induced skin inflammation, as they do not inhibit synthesis of other proalgesic lipids such as LPA 18∶1, 9-and 13-HODE and HETEs.


British Journal of Pharmacology | 2014

Bupivacaine-induced cellular entry of QX-314 and its contribution to differential nerve block

C Brenneis; Katrin Kistner; Michelino Puopolo; Sooyeon Jo; Dp Roberson; Marco Sisignano; David Segal; Enrique J. Cobos; Brian J. Wainger; Sandra Labocha; Nerea Ferreirós; C von Hehn; J Tran; Gerd Geisslinger; Peter W. Reeh; Bruce P. Bean; Clifford J. Woolf

Selective nociceptor fibre block is achieved by introducing the cell membrane impermeant sodium channel blocker lidocaine N‐ethyl bromide (QX‐314) through transient receptor potential V1 (TRPV1) channels into nociceptors. We screened local anaesthetics for their capacity to activate TRP channels, and characterized the nerve block obtained by combination with QX‐314.


PLOS ONE | 2014

Genome-wide transcriptional profiling of skin and dorsal root ganglia after ultraviolet-B-induced inflammation.

John M. Dawes; Ana Antunes-Martins; James R. Perkins; Kathryn J. Paterson; Marco Sisignano; Ramona Schmid; Werner Rust; Tobias Hildebrandt; Gerd Geisslinger; Christine A. Orengo; David L. H. Bennett; Stephen B. McMahon

Ultraviolet-B (UVB)-induced inflammation produces a dose-dependent mechanical and thermal hyperalgesia in both humans and rats, most likely via inflammatory mediators acting at the site of injury. Previous work has shown that the gene expression of cytokines and chemokines is positively correlated between species and that these factors can contribute to UVB-induced pain. In order to investigate other potential pain mediators in this model we used RNA-seq to perform genome-wide transcriptional profiling in both human and rat skin at the peak of hyperalgesia. In addition we have also measured transcriptional changes in the L4 and L5 DRG of the rat model. Our data show that UVB irradiation produces a large number of transcriptional changes in the skin: 2186 and 3888 genes are significantly dysregulated in human and rat skin, respectively. The most highly up-regulated genes in human skin feature those encoding cytokines (IL6 and IL24), chemokines (CCL3, CCL20, CXCL1, CXCL2, CXCL3 and CXCL5), the prostanoid synthesising enzyme COX-2 and members of the keratin gene family. Overall there was a strong positive and significant correlation in gene expression between the human and rat (R = 0.8022). In contrast to the skin, only 39 genes were significantly dysregulated in the rat L4 and L5 DRGs, the majority of which had small fold change values. Amongst the most up-regulated genes in DRG were REG3B, CCL2 and VGF. Overall, our data shows that numerous genes were up-regulated in UVB irradiated skin at the peak of hyperalgesia in both human and rats. Many of the top up-regulated genes were cytokines and chemokines, highlighting again their potential as pain mediators. However many other genes were also up-regulated and might play a role in UVB-induced hyperalgesia. In addition, the strong gene expression correlation between species re-emphasises the value of the UVB model as translational tool to study inflammatory pain.


Pain | 2014

BKCa channels expressed in sensory neurons modulate inflammatory pain in mice

Ruirui Lu; Robert Lukowski; Matthias Sausbier; Dong Dong Zhang; Marco Sisignano; Claus-Dieter Schuh; Rohini Kuner; Peter Ruth; Gerd Geisslinger; Achim Schmidtko

Summary The in vivo function of large conductance calcium‐activated potassium channels in sensory neurons includes control of inflammatory pain, but not of acute nociceptive or nerve injury‐induced neuropathic pain. ABSTRACT Large conductance calcium‐activated potassium (BKCa) channels are important regulators of neuronal excitability. Although there is electrophysiological evidence for BKCa channel expression in sensory neurons, their in vivo functions in pain processing have not been fully defined. Using a specific antibody, we demonstrate here that BKCa channels are expressed in subpopulations of peptidergic and nonpeptidergic nociceptors. To test a functional association of BKCa channel activity in sensory neurons with particular pain modalities, we generated mice in which BKCa channels are ablated specifically from sensory neurons and analyzed their behavior in various models of pain. Mutant mice showed increased nociceptive behavior in models of persistent inflammatory pain. However, their behavior in models of neuropathic or acute nociceptive pain was normal. Moreover, systemic administration of the BKCa channel opener, NS1619, inhibited persistent inflammatory pain. Our investigations provide in vivo evidence that BKCa channels expressed in sensory neurons exert inhibitory control on sensory input in inflammatory pain states.


Trends in Pharmacological Sciences | 2016

Drug Repurposing for the Development of Novel Analgesics.

Marco Sisignano; Michael J. Parnham; Gerd Geisslinger

Drug development consumes huge amounts of time and money and the search for novel analgesics, which are urgently required, is particularly difficult, having resulted in many setbacks in the past. Drug repurposing - the identification of new uses for existing drugs - is an alternative approach, which bypasses most of the time- and cost-consuming components of drug development. Recent, unexpected findings suggest a role for several existing drugs, such as minocycline, ceftriaxone, sivelestat, and pioglitazone, as novel analgesics in chronic and neuropathic pain states. Here, we discuss these findings as well as their proposed antihyperalgesic mechanisms and outline the merits of pathway-based repurposing screens, in combination with bioinformatics and novel cellular reprogramming techniques, for the identification of novel analgesics.

Collaboration


Dive into the Marco Sisignano's collaboration.

Top Co-Authors

Avatar

Gerd Geisslinger

Goethe University Frankfurt

View shared research outputs
Top Co-Authors

Avatar

Klaus Scholich

Goethe University Frankfurt

View shared research outputs
Top Co-Authors

Avatar

Carlo Angioni

Goethe University Frankfurt

View shared research outputs
Top Co-Authors

Avatar

Clifford J. Woolf

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar

Nerea Ferreirós

Goethe University Frankfurt

View shared research outputs
Top Co-Authors

Avatar

Christian Brenneis

Goethe University Frankfurt

View shared research outputs
Top Co-Authors

Avatar

Dong Dong Zhang

Goethe University Frankfurt

View shared research outputs
Top Co-Authors

Avatar

Ruirui Lu

Goethe University Frankfurt

View shared research outputs
Top Co-Authors

Avatar

Achim Schmidtko

Goethe University Frankfurt

View shared research outputs
Top Co-Authors

Avatar

Jing Suo

Goethe University Frankfurt

View shared research outputs
Researchain Logo
Decentralizing Knowledge