Network


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

Hotspot


Dive into the research topics where Michael Costigan is active.

Publication


Featured researches published by Michael Costigan.


Annual Review of Neuroscience | 2009

Neuropathic Pain: A Maladaptive Response of the Nervous System to Damage

Michael Costigan; Joachim Scholz; Clifford J. Woolf

Neuropathic pain is triggered by lesions to the somatosensory nervous system that alter its structure and function so that pain occurs spontaneously and responses to noxious and innocuous stimuli are pathologically amplified. The pain is an expression of maladaptive plasticity within the nociceptive system, a series of changes that constitute a neural disease state. Multiple alterations distributed widely across the nervous system contribute to complex pain phenotypes. These alterations include ectopic generation of action potentials, facilitation and disinhibition of synaptic transmission, loss of synaptic connectivity and formation of new synaptic circuits, and neuroimmune interactions. Although neural lesions are necessary, they are not sufficient to generate neuropathic pain; genetic polymorphisms, gender, and age all influence the risk of developing persistent pain. Treatment needs to move from merely suppressing symptoms to a disease-modifying strategy aimed at both preventing maladaptive plasticity and reducing intrinsic risk.


BMC Neuroscience | 2002

Replicate high-density rat genome oligonucleotide microarrays reveal hundreds of regulated genes in the dorsal root ganglion after peripheral nerve injury.

Michael Costigan; Katia Befort; Laurie A. Karchewski; Robert S. Griffin; Donatella D'urso; Andrew Allchorne; Joanne Sitarski; James W Mannion; Richard E. Pratt; Clifford J. Woolf

BackgroundRat oligonucleotide microarrays were used to detect changes in gene expression in the dorsal root ganglion (DRG) 3 days following sciatic nerve transection (axotomy). Two comparisons were made using two sets of triplicate microarrays, naïve versus naïve and naïve versus axotomy.ResultsMicroarray variability was assessed using the naïve versus naïve comparison. These results support use of a P < 0.05 significance threshold for detecting regulated genes, despite the large number of hypothesis tests required. For the naïve versus axotomy comparison, a 2-fold cut off alone led to an estimated error rate of 16%; combining a >1.5-fold expression change and P < 0.05 significance reduced the estimated error to 5%. The 2-fold cut off identified 178 genes while the combined >1.5-fold and P < 0.05 criteria generated 240 putatively regulated genes, which we have listed. Many of these have not been described as regulated in the DRG by axotomy. Northern blot, quantitative slot blots and in situ hybridization verified the expression of 24 transcripts. These data showed an 83% concordance rate with the arrays; most mismatches represent genes with low expression levels reflecting limits of array sensitivity. A significant correlation was found between actual mRNA differences and relative changes between microarrays (r2 = 0.8567). Temporal patterns of individual genes regulation varied.ConclusionsWe identify parameters for microarray analysis which reduce error while identifying many putatively regulated genes. Functional classification of these genes suggest reorganization of cell structural components, activation of genes expressed by immune and inflammatory cells and down-regulation of genes involved in neurotransmission.


Nature Medicine | 2006

GTP cyclohydrolase and tetrahydrobiopterin regulate pain sensitivity and persistence.

Irmgard Tegeder; Michael Costigan; Robert S. Griffin; Andrea Abele; Inna Belfer; Helmut Schmidt; Corina Ehnert; Jemiel Nejim; Claudiu Marian; Joachim Scholz; Tianxia Wu; Andrew Allchorne; Luda Diatchenko; Alexander M. Binshtok; David Goldman; Jan Adolph; Swetha Sama; Steven J. Atlas; William A. Carlezon; Aram Parsegian; Jörn Lötsch; Roger B. Fillingim; William Maixner; Gerd Geisslinger; Mitchell B. Max; Clifford J. Woolf

We report that GTP cyclohydrolase (GCH1), the rate-limiting enzyme for tetrahydrobiopterin (BH4) synthesis, is a key modulator of peripheral neuropathic and inflammatory pain. BH4 is an essential cofactor for catecholamine, serotonin and nitric oxide production. After axonal injury, concentrations of BH4 rose in primary sensory neurons, owing to upregulation of GCH1. After peripheral inflammation, BH4 also increased in dorsal root ganglia (DRGs), owing to enhanced GCH1 enzyme activity. Inhibiting this de novo BH4 synthesis in rats attenuated neuropathic and inflammatory pain and prevented nerve injury–evoked excess nitric oxide production in the DRG, whereas administering BH4 intrathecally exacerbated pain. In humans, a haplotype of the GCH1 gene (population frequency 15.4%) was significantly associated with less pain following diskectomy for persistent radicular low back pain. Healthy individuals homozygous for this haplotype exhibited reduced experimental pain sensitivity, and forskolin-stimulated immortalized leukocytes from haplotype carriers upregulated GCH1 less than did controls. BH4 is therefore an intrinsic regulator of pain sensitivity and chronicity, and the GTP cyclohydrolase haplotype is a marker for these traits.


Nature Neuroscience | 1998

Two sodium channels contribute to the TTX-R sodium current in primary sensory neurons

Simon Tate; Susanna C. Benn; Caroline Hick; Derek J. Trezise; Victoria H. John; Richard J. Mannion; Michael Costigan; Chris Plumpton; David Grose; Zoe Marie Gladwell; Giles S. Kendall; Katie Dale; C. Bountra; Clifford J. Woolf

We have cloned a second tetrodotoxin-resistant (TTX-R) sodium channel α subunit, SNS2, with the same amino-acid sequence as a putative sodium channel α subunit NaN (ref. 1). SNS2 expression in HEK293T cells produced a TTX-R voltage-gated sodium current with faster kinetics and a lower TTX IC50 than the previously cloned sensory-neuron-specific sodium channel, SNS/PN3 (Refs 2, 3). SNS2 was co-expressed with SNS/PN3 in small DRG neurons.


The Journal of Neuroscience | 2006

The Voltage-Gated Sodium Channel Nav1.9 Is an Effector of Peripheral Inflammatory Pain Hypersensitivity

Fumimasa Amaya; Haibin Wang; Michael Costigan; Andrew Allchorne; Jon P. Hatcher; Julie Egerton; Tania O. Stean; Valerie Morisset; David Thomas Grose; Martin J. Gunthorpe; Iain P. Chessell; Simon Tate; Paula J. Green; Clifford J. Woolf

We used a mouse with deletion of exons 4, 5, and 6 of the SCN11A (sodium channel, voltage-gated, type XI, α) gene that encodes the voltage-gated sodium channel Nav1.9 to assess its contribution to pain. Nav1.9 is present in nociceptor sensory neurons that express TRPV1, bradykinin B2, and purinergic P2X3 receptors. In Nav1.9−/− mice, the non-inactivating persistent tetrodotoxin-resistant sodium TTXr-Per current is absent, whereas TTXr-Slow is unchanged. TTXs currents are unaffected by the mutation of Nav1.9. Pain hypersensitivity elicited by intraplantar administration of prostaglandin E2, bradykinin, interleukin-1β, capsaicin, and P2X3 and P2Y receptor agonists, but not NGF, is either reduced or absent in Nav1.9−/− mice, whereas basal thermal and mechanical pain sensitivity is unchanged. Thermal, but not mechanical, hypersensitivity produced by peripheral inflammation (intraplanatar complete Freunds adjuvant) is substantially diminished in the null allele mutant mice, whereas hypersensitivity in two neuropathic pain models is unchanged in the Nav1.9−/− mice. Nav1.9 is, we conclude, an effector of the hypersensitivity produced by multiple inflammatory mediators on nociceptor peripheral terminals and therefore plays a key role in mediating peripheral sensitization.


Molecular and Cellular Neuroscience | 2000

Diversity of Expression of the Sensory Neuron-Specific TTX-Resistant Voltage-Gated Sodium Ion Channels SNS and SNS2

Fumimasa Amaya; Isabelle Decosterd; Tarek A. Samad; Christopher Plumpton; Simon Tate; Richard J. Mannion; Michael Costigan; Clifford J. Woolf

The differential distribution of two tetrodotoxin resistant (TTXr) voltage-gated sodium channels SNS (PN3) and SNS2 (NaN) in rat primary sensory neurons has been investigated. Both channels are sensory neuron specific with SNS2 restricted entirely to those small dorsal root ganglion (DRG) cells with unmyelinated axons (C-fibers). SNS, in contrast, is expressed both in small C-fiber DRG cells and in 10% of cells with myelinated axons (A-fibers). All SNS expressing A-fiber cells are Trk-A positive and many express the vanilloid-like receptor VRL1. About half of C-fiber DRG neurons express either SNS or SNS2, and in most, the channels are colocalized. SNS and SNS2 are found both in NGF-responsive and GDNF-responsive C-fibers and many of these cells also express the capsaicin receptor VR1. A very small proportion of small DRG cells express either only SNS or only SNS2. At least four different classes of A- and C-fiber DRG neurons exist, therefore, with respect to expression of these sodium channels.


The Journal of Neuroscience | 2009

T-Cell Infiltration and Signaling in the Adult Dorsal Spinal Cord Is a Major Contributor to Neuropathic Pain-Like Hypersensitivity

Michael Costigan; Andrew Moss; Alban Latremoliere; Caroline Johnston; Monica Verma-Gandhu; Teri A. Herbert; Lee B. Barrett; Gary J. Brenner; Daniel Vardeh; Clifford J. Woolf; Maria Fitzgerald

Partial peripheral nerve injury in adult rats results in neuropathic pain-like hypersensitivity, while that in neonatal rats does not, a phenomenon also observed in humans. We therefore compared gene expression profiles in the dorsal horn of adult and neonatal rats in response to the spared nerve injury (SNI) model of peripheral neuropathic pain. The 148 differentially regulated genes in adult, but not young, rat spinal cords indicate a greater microglial and T-cell response in adult than in young animals. T-cells show a large infiltration in the adult dorsal horn but not in the neonate after SNI. T-cell-deficient Rag1-null adult mice develop less neuropathic mechanical allodynia than controls, and central expression of cytokines involved in T-cell signaling exhibits large relative differences between young and adult animals after SNI. One such cytokine, interferon-γ (IFNγ), is upregulated in the dorsal horn after nerve injury in the adult but not neonate, and we show that IFNγ signaling is required for full expression of adult neuropathic hypersensitivity. These data reveal that T-cell infiltration and activation in the dorsal horn of the spinal cord following peripheral nerve injury contribute to the evolution of neuropathic pain-like hypersensitivity. The neuroimmune interaction following peripheral nerve injury has therefore a substantial adaptive immune component, which is absent or suppressed in the young CNS.


Nature Medicine | 2012

Genetically determined P2X7 receptor pore formation regulates variability in chronic pain sensitivity

Tuan Trang; Ruslan Dorfman; Shad B. Smith; Simon Beggs; Jennifer Ritchie; Jean Sebastien Austin; Dmitri V. Zaykin; Heather Vander Meulen; Michael Costigan; Teri A. Herbert; Merav Yarkoni-Abitbul; David Tichauer; Jessica Livneh; Edith Gershon; Ming Zheng; Keith Tan; Sally John; Gary D. Slade; Joanne M. Jordan; Clifford J. Woolf; Gary Peltz; William Maixner; Luda Diatchenko; Ze'ev Seltzer; Michael W. Salter; Jeffrey S. Mogil

Chronic pain is highly variable between individuals, as is the response to analgesics. Although much of the variability in chronic pain and analgesic response is heritable, an understanding of the genetic determinants underlying this variability is rudimentary. Here we show that variation within the coding sequence of the gene encoding the P2X7 receptor (P2X7R) affects chronic pain sensitivity in both mice and humans. P2X7Rs, which are members of the family of ionotropic ATP-gated receptors, have two distinct modes of function: they can function through their intrinsic cationic channel or by forming nonselective pores that are permeable to molecules with a mass of up to 900 Da. Using genome-wide linkage analyses, we discovered an association between nerve-injury–induced pain behavior (mechanical allodynia) and the P451L mutation of the mouse P2rx7 gene, such that mice in which P2X7Rs have impaired pore formation as a result of this mutation showed less allodynia than mice with the pore-forming P2rx7 allele. Administration of a peptide corresponding to the P2X7R C-terminal domain, which blocked pore formation but not cation channel activity, selectively reduced nerve injury and inflammatory allodynia only in mice with the pore-forming P2rx7 allele. Moreover, in two independent human chronic pain cohorts, a cohort with pain after mastectomy and a cohort with osteoarthritis, we observed a genetic association between lower pain intensity and the hypofunctional His270 (rs7958311) allele of P2RX7. Our findings suggest that selectively targeting P2X7R pore formation may be a new strategy for individualizing the treatment of chronic pain.


The Journal of Neuroscience | 2010

TRPA1 Contributes to Cold Hypersensitivity

Donato del Camino; Sarah Murphy; Melissa Heiry; Lee B. Barrett; Taryn J. Earley; Colby A. Cook; Matt J. Petrus; Michael Zhao; Marc D'Amours; Nate Deering; Gary J. Brenner; Michael Costigan; Neil Hayward; Jayhong A. Chong; Christopher Fanger; Clifford J. Woolf; Ardem Patapoutian; Magdalene M. Moran

TRPA1 is a nonselective cation channel expressed by nociceptors. Although it is widely accepted that TRPA1 serves as a broad irritancy receptor for a variety of reactive chemicals, its role in cold sensation remains controversial. Here, we demonstrate that mild cooling markedly increases agonist-evoked rat TRPA1 currents. In the absence of an agonist, even noxious cold only increases current amplitude slightly. These results suggest that TRPA1 is a key mediator of cold hypersensitivity in pathological conditions in which reactive oxygen species and proinflammatory activators of the channel are present, but likely plays a comparatively minor role in acute cold sensation. Supporting this, cold hypersensitivity can be induced in wild-type but not Trpa1−/− mice by subcutaneous administration of a TRPA1 agonist. Furthermore, the selective TRPA1 antagonist HC-030031 [2-(1,3-dimethyl-2,6-dioxo-1,2,3,6-tetrahydro-7H-purin-7-yl)-N-(4-isopropylphenyl)acetamide] reduces cold hypersensitivity in rodent models of inflammatory and neuropathic pain.


Cell | 2010

A Genome-wide Drosophila Screen for Heat Nociception Identifies α2δ3 as an Evolutionarily Conserved Pain Gene

G. Gregory Neely; Andreas Hess; Michael Costigan; Alex C. Keene; Spyros Goulas; Michiel Langeslag; Robert S. Griffin; Inna Belfer; Feng Dai; Shad B. Smith; Luda Diatchenko; Vaijayanti Gupta; Cui ping Xia; Sabina Amann; Silke Kreitz; Cornelia Heindl-Erdmann; Susanne Wolz; Cindy V. Ly; Suchir Arora; Rinku Sarangi; Debasis Dan; Maria Novatchkova; Mark R. Rosenzweig; Dustin G. Gibson; Darwin Truong; Daniel Schramek; Tamara Zoranovic; Shane J. Cronin; Belinda Angjeli; Kay Brune

Worldwide, acute, and chronic pain affects 20% of the adult population and represents an enormous financial and emotional burden. Using genome-wide neuronal-specific RNAi knockdown in Drosophila, we report a global screen for an innate behavior and identify hundreds of genes implicated in heat nociception, including the α2δ family calcium channel subunit straightjacket (stj). Mice mutant for the stj ortholog CACNA2D3 (α2δ3) also exhibit impaired behavioral heat pain sensitivity. In addition, in humans, α2δ3 SNP variants associate with reduced sensitivity to acute noxious heat and chronic back pain. Functional imaging in α2δ3 mutant mice revealed impaired transmission of thermal pain-evoked signals from the thalamus to higher-order pain centers. Intriguingly, in α2δ3 mutant mice, thermal pain and tactile stimulation triggered strong cross-activation, or synesthesia, of brain regions involved in vision, olfaction, and hearing.

Collaboration


Dive into the Michael Costigan's collaboration.

Top Co-Authors

Avatar

Clifford J. Woolf

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Lee B. Barrett

Boston Children's Hospital

View shared research outputs
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

Inna Belfer

University of Pittsburgh

View shared research outputs
Researchain Logo
Decentralizing Knowledge