Network


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

Hotspot


Dive into the research topics where Shad B. Smith is active.

Publication


Featured researches published by Shad B. Smith.


Science | 2006

Social Modulation of Pain as Evidence for Empathy in Mice

Dale J. Langford; Sara E. Crager; Zarrar Shehzad; Shad B. Smith; Susana G. Sotocinal; Jeremy S. Levenstadt; Mona Lisa Chanda; Daniel J. Levitin; Jeffrey S. Mogil

Empathy is thought to be unique to higher primates, possibly to humans alone. We report the modulation of pain sensitivity in mice produced solely by exposure to their cagemates, but not to strangers, in pain. Mice tested in dyads and given an identical noxious stimulus displayed increased pain behaviors with statistically greater co-occurrence, effects dependent on visual observation. When familiar mice were given noxious stimuli of different intensities, their pain behavior was influenced by their neighbors status bidirectionally. Finally, observation of a cagemate in pain altered pain sensitivity of an entirely different modality, suggesting that nociceptive mechanisms in general are sensitized.


Proceedings of the National Academy of Sciences of the United States of America | 2010

Pain perception is altered by a nucleotide polymorphism in SCN9A

Frank Reimann; James J. Cox; Inna Belfer; Luda Diatchenko; Dmitri V. Zaykin; Duncan P. McHale; Joost P. H. Drenth; Feng Dai; Jerry Wheeler; Frances A. Sanders; Linda S. Wood; Tianxia Wu; Jaro Karppinen; Lone Nikolajsen; Minna Männikkö; Mitchell B. Max; Carly Kiselycznyk; Minakshi Poddar; Rene H. M. te Morsche; Shad B. Smith; Dustin G. Gibson; Anthi Kelempisioti; William Maixner; Fiona M. Gribble; C. Geoffrey Woods

The gene SCN9A is responsible for three human pain disorders. Nonsense mutations cause a complete absence of pain, whereas activating mutations cause severe episodic pain in paroxysmal extreme pain disorder and primary erythermalgia. This led us to investigate whether single nucleotide polymorphisms (SNPs) in SCN9A were associated with differing pain perception in the general population. We first genotyped 27 SCN9A SNPs in 578 individuals with a radiographic diagnosis of osteoarthritis and a pain score assessment. A significant association was found between pain score and SNP rs6746030; the rarer A allele was associated with increased pain scores compared to the commoner G allele (P = 0.016). This SNP was then further genotyped in 195 pain-assessed people with sciatica, 100 amputees with phantom pain, 179 individuals after lumbar discectomy, and 205 individuals with pancreatitis. The combined P value for increased A allele pain was 0.0001 in the five cohorts tested (1277 people in total). The two alleles of the SNP rs6746030 alter the coding sequence of the sodium channel Nav1.7. Each was separately transfected into HEK293 cells and electrophysiologically assessed by patch-clamping. The two alleles showed a difference in the voltage-dependent slow inactivation (P = 0.042) where the A allele would be predicted to increase Nav1.7 activity. Finally, we genotyped 186 healthy females characterized by their responses to a diverse set of noxious stimuli. The A allele of rs6746030 was associated with an altered pain threshold and the effect mediated through C-fiber activation. We conclude that individuals experience differing amounts of pain, per nociceptive stimulus, on the basis of their SCN9A rs6746030 genotype.


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.


Journal of Medical Genetics | 2005

Melanocortin-1 receptor gene variants affect pain and μ-opioid analgesia in mice and humans

Jeffrey S. Mogil; Jennifer Ritchie; Shad B. Smith; Kate Strasburg; Lee M. Kaplan; Margaret R. Wallace; R. R. Romberg; H. Bijl; Elise Sarton; Roger B. Fillingim; Albert Dahan

Background: A recent genetic study in mice and humans revealed the modulatory effect of MC1R (melanocortin-1 receptor) gene variants on κ-opioid receptor mediated analgesia. It is unclear whether this gene affects basal pain sensitivity or the efficacy of analgesics acting at the more clinically relevant μ-opioid receptor. Objective: To characterise sensitivity to pain and μ-opioid analgesia in mice and humans with non-functional melanocortin-1 receptors. Methods: Comparisons of spontaneous mutant C57BL/6-Mc1re/e mice to C57BL/6 wildtype mice, followed by a gene dosage study of pain and morphine-6-glucuronide (M6G) analgesia in humans with MC1R variants. Results: C57BL/6-Mc1re/e mutant mice and human redheads—both with non-functional MC1Rs—display reduced sensitivity to noxious stimuli and increased analgesic responsiveness to the μ-opioid selective morphine metabolite, M6G. In both species the differential analgesia is likely due to pharmacodynamic factors, as plasma levels of M6G are similar across genotype. Conclusions: Genotype at MC1R similarly affects pain sensitivity and M6G analgesia in mice and humans. These findings confirm the utility of cross species translational strategies in pharmacogenetics.


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.


The Journal of Pain | 2011

Potential Genetic Risk Factors for Chronic TMD: Genetic Associations from the OPPERA Case Control Study

Shad B. Smith; Dylan W. Maixner; Joel D. Greenspan; Ronald Dubner; Roger B. Fillingim; Richard Ohrbach; Charles Knott; Gary D. Slade; Eric Bair; Dustin G. Gibson; Dmitri V. Zaykin; Bruce S. Weir; William Maixner; Luda Diatchenko

UNLABELLED Genetic factors play a role in the etiology of persistent pain conditions, putatively by modulating underlying processes such as nociceptive sensitivity, psychological well-being, inflammation, and autonomic response. However, to date, only a few genes have been associated with temporomandibular disorders (TMD). This study evaluated 358 genes involved in pain processes, comparing allelic frequencies between 166 cases with chronic TMD and 1,442 controls enrolled in the OPPERA (Orofacial Pain: Prospective Evaluation and Risk Assessment) study cooperative agreement. To enhance statistical power, 182 TMD cases and 170 controls from a similar study were included in the analysis. Genotyping was performed using the Pain Research Panel, an Affymetrix gene chip representing 3,295 single nucleotide polymorphisms, including ancestry-informative markers that were used to adjust for population stratification. Adjusted associations between genetic markers and TMD case status were evaluated using logistic regression. The OPPERA findings provided evidence supporting previously reported associations between TMD and 2 genes: HTR2A and COMT. Other genes were revealed as potential new genetic risk factors for TMD, including NR3C1, CAMK4, CHRM2, IFRD1, and GRK5. While these findings need to be replicated in independent cohorts, the genes potentially represent important markers of risk for TMD, and they identify potential targets for therapeutic intervention. PERSPECTIVE Genetic risk factors for TMD pain were explored in the case-control component of the OPPERA cooperative agreement, a large population-based prospective cohort study. Over 350 candidate pain genes were assessed using a candidate gene panel, with several genes displaying preliminary evidence for association with TMD status.


Nature Neuroscience | 2011

Pain sensitivity and vasopressin analgesia are mediated by a gene-sex-environment interaction

Jeffrey S. Mogil; Michael L. LaCroix-Fralish; Shad B. Smith; Anny Fortin; Susana G. Sotocinal; Jennifer Ritchie; Jean Sebastien Austin; Ara Schorscher-Petcu; Kara Melmed; Jan T. Czerminski; Rosalie A. Bittong; J. Brad Mokris; John K. Neubert; C. Campbell; Robert R. Edwards; James N. Campbell; Jacqueline N. Crawley; William R. Lariviere; Margaret R. Wallace; Wendy F. Sternberg; Carey D. Balaban; Inna Belfer; Roger B. Fillingim

Quantitative trait locus mapping of chemical/inflammatory pain in the mouse identified the Avpr1a gene, which encodes the vasopressin-1A receptor (V1AR), as being responsible for strain-dependent pain sensitivity to formalin and capsaicin. A genetic association study in humans revealed the influence of a single nucleotide polymorphism (rs10877969) in AVPR1A on capsaicin pain levels, but only in male subjects reporting stress at the time of testing. The analgesic efficacy of the vasopressin analog desmopressin revealed a similar interaction between the drug and acute stress, as desmopressin inhibition of capsaicin pain was only observed in nonstressed subjects. Additional experiments in mice confirmed the male-specific interaction of V1AR and stress, leading to the conclusion that vasopressin activates endogenous analgesia mechanisms unless they have already been activated by stress. These findings represent, to the best of our knowledge, the first explicit demonstration of analgesic efficacy depending on the emotional state of the recipient, and illustrate the heuristic power of a bench-to-bedside-to-bench translational strategy.


Pain | 2006

Screening for pain phenotypes: Analysis of three congenic mouse strains on a battery of nine nociceptive assays

Jeffrey S. Mogil; Jennifer Ritchie; Susana G. Sotocinal; Shad B. Smith; Sylvie Croteau; Daniel J. Levitin; Anna K. Naumova

&NA; In an attempt to identify new genes responsible for variability in pain sensitivity, we tested three congenic mouse strains – in which a small portion of the genome of the MOLF/Ei strain has been placed on a C57BL/6 genetic background – on a battery of nine nociceptive assays, chosen to reflect those assays in most common use in the pain literature. Mice of both sexes were evaluated by two different testers at different points in time, allowing us to examine the relative importance of genotype, sex, tester and cohort effects on data from these assays. We find strong evidence for the existence of two quantitative trait loci (i.e., genomic regions containing variability‐causing genes), one for thermal nociception on mouse chromosome (Chr) 17 (Chr 17; Tpnr3) and one for formalin test nociception on mouse Chr 12 (Nociq3). We note, however, that the nociceptive assays in this battery feature strong main effects and interactions of sex, tester, and cohort, which if not controlled or covaried can seriously confound interpretation of genetic experiments, including the comparison of transgenic knockout mice to their wild‐type controls.


Pain | 2011

Cytokine biomarkers and chronic pain: association of genes, transcription, and circulating proteins with temporomandibular disorders and widespread palpation tenderness.

Gary D. Slade; Mathew S. Conrad; Luda Diatchenko; Naim Rashid; Sheng Zhong; Shad B. Smith; Jesse Rhodes; Alex Medvedev; Sergei S. Makarov; William Maixner; Andrea G. Nackley

Summary Women with widespread vs localized chronic pain exhibit distinctive alterations in molecular‐genetic profiles of the inflammatory mediators MCP‐1, IL‐1ra, IL‐8, and TGFβ1. ABSTRACT For reasons unknown, temporomandibular disorder (TMD) can manifest as localized pain or in conjunction with widespread pain. We evaluated relationships between cytokines and TMD without or with widespread palpation tenderness (TMD−WPT or TMD+WPT, respectively) at protein, transcription factory activity, and gene levels. Additionally, we evaluated the relationship between cytokines and intermediate phenotypes characteristic of TMD and WPT. In a case‐control study of 344 females, blood samples were analyzed for levels of 22 cytokines and activity of 48 transcription factors. Intermediate phenotypes were measured by quantitative sensory testing and questionnaires asking about pain, health, and psychological status. Single nucleotide polymorphisms (SNPs) coding cytokines and transcription factors were genotyped. TMD−WPT cases had elevated protein levels of proinflammatory cytokine monocyte chemotactic protein (MCP‐1) and antiinflammatory cytokine interleukin (IL)‐1ra, whereas TMD+WPT cases had elevated levels of proinflammatory cytokine IL‐8. MCP‐1, IL‐1ra, and IL‐8 were differentially associated with experimental pain, self‐rated pain, self‐rated health, and psychological phenotypes. TMD−WPT and TMD+WPT cases had inhibited transcription activity of the antiinflammatory cytokine transforming growth factor β1 (TGFβ1). Interactions were observed between TGFβ1 and IL‐8 SNPs: an additional copy of the TGFβ1 rs2241719 minor T allele was associated with twice the odds of TMD+WPT among individuals homozygous for the IL‐8 rs4073 major A allele, and half the odds of TMD+WPT among individuals heterozygous for rs4073. These results demonstrate how pro‐ and antiinflammatory cytokines contribute to the pathophysiology of TMD and WPT in genetically susceptible people. Furthermore, they identify MCP‐1, IL‐1ra, IL‐8, and TGFβ1 as potential diagnostic markers and therapeutic targets for pain in patients with TMD.


Pain | 2013

Pain modality- and sex-specific effects of COMT genetic functional variants

Inna Belfer; Samantha K. Segall; William R. Lariviere; Shad B. Smith; Feng Dai; Gary D. Slade; Naim Rashid; Jeffrey S. Mogil; C. Campbell; Robert R. Edwards; Qian Liu; Eric Bair; William Maixner; Luda Diatchenko

&NA; The effects of COMT genetic polymorphism on pain and analgesia are modality – selective and gender – dependent, in both mouse and humanPlease approve Summary as edited. &NA; The enzyme catechol‐O‐methyltransferase (COMT) metabolizes catecholamine neurotransmitters involved in a number of physiological functions, including pain perception. Both human and mouse COMT genes possess functional polymorphisms contributing to interindividual variability in pain phenotypes such as sensitivity to noxious stimuli, severity of clinical pain, and response to pain treatment. In this study, we found that the effects of Comt functional variation in mice are modality specific. Spontaneous inflammatory nociception and thermal nociception behaviors were correlated the most with the presence of the B2 SINE transposon insertion residing in the 3′UTR mRNA region. Similarly, in humans, COMT functional haplotypes were associated with thermal pain perception and with capsaicin‐induced pain. Furthermore, COMT genetic variations contributed to pain behaviors in mice and pain ratings in humans in a sex‐specific manner. The ancestral Comt variant, without a B2 SINE insertion, was more strongly associated with sensitivity to capsaicin in female vs male mice. In humans, the haplotype coding for low COMT activity increased capsaicin‐induced pain perception in women, but not men. These findings reemphasize the fundamental contribution of COMT to pain processes, and provide a fine‐grained resolution of this contribution at the genetic level that can be used to guide future studies in the area of pain genetics.

Collaboration


Dive into the Shad B. Smith's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Gary D. Slade

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Eric Bair

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Dmitri V. Zaykin

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Dustin G. Gibson

University of North Carolina at Chapel Hill

View shared research outputs
Researchain Logo
Decentralizing Knowledge