Network


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

Hotspot


Dive into the research topics where Theodore J. Price is active.

Publication


Featured researches published by Theodore J. Price.


British Journal of Pharmacology | 2004

Modulation of trigeminal sensory neuron activity by the dual cannabinoid–vanilloid agonists anandamide, N‐arachidonoyl‐dopamine and arachidonyl‐2‐chloroethylamide

Theodore J. Price; Amol M. Patwardhan; Armen N. Akopian; Kenneth M. Hargreaves; Christopher M. Flores

Peripheral cannabinoids have been shown to suppress nociceptive neurotransmission in a number of behavioral and neurophysiological studies. It is not known, however, whether cannabinoids exert this action through direct interactions with nociceptors in the periphery and/or if other processes are involved. To gain a better understanding of the direct actions of cannabinoid‐vanilloid agonists on sensory neurons, we examined the effects of these compounds on trigeminal ganglion (TG) neurons in vitro. AEA (EC50=11.0 μM), NADA (EC50=857 nM) and arachidonyl‐2‐chloroethylamide ACEA (EC50=14.0 μM) each evoked calcitonin gene‐related peptide (CGRP) release from TG neurons. The TRPV1 antagonists iodo‐resiniferatoxin (I‐RTX) and capsazepine (CPZ) each obtunded AEA‐, NADA‐, ACEA‐ and capsaicin (CAP)‐evoked CGRP release with individually equivalent IC50s for each of the compounds (I‐RTX IC50 range=2.6–4.0 nM; CPZ IC50 range=523–1140 μM). The pro‐inflammatory mediator prostaglandin E2 significantly increased the maximal effect of AEA‐evoked CGRP release without altering the EC50. AEA, ACEA and CAP stimulated cAMP accumulation in TG neurons in a calcium‐ and TRPV1‐dependent fashion. Moreover, the protein kinase inhibitor staurosporine significantly inhibited AEA‐ and CAP‐evoked CGRP release. The pungency of AEA, NADA, ACEA and CAP in the rat eye‐wipe assay was also assessed. Interestingly, when applied intraocularly, NADA or CAP each produced nocifensive responses, while AEA or ACEA did not. Finally, the potential inhibitory effects of these cannabinoids on TG nociceptors were evaluated. Neither AEA nor ACEA decreased CAP‐evoked CGRP release. Furthermore, neither of the cannabinoid receptor type 1 antagonists SR141716A nor AM251 had any impact on either basal or CAP‐evoked CGRP release. AEA also did not inhibit 50 mM K+‐evoked CGRP release and did not influence bradykinin‐stimulated inositol phosphate accumulation. We conclude that the major action of AEA, NADA and ACEA on TG neurons is excitatory, while, of these, only NADA is pungent. These findings are discussed in relation to our current understanding of interactions between the cannabinoid and vanilloid systems and nociceptive processing in the periphery.


Journal of Biological Chemistry | 2006

Cannabinoid WIN 55,212-2 Regulates TRPV1 Phosphorylation in Sensory Neurons

Nathaniel Aaron Jeske; Amol M. Patwardhan; Nikita Gamper; Theodore J. Price; Armen N. Akopian; Kenneth M. Hargreaves

Cannabinoids are known to have multiple sites of action in the nociceptive system, leading to reduced pain sensation. However, the peripheral mechanism(s) by which this phenomenon occurs remains an issue that has yet to be resolved. Because phosphorylation of TRPV1 (transient receptor potential subtype V1) plays a key role in the induction of thermal hyperalgesia in inflammatory pain models, we evaluated whether the cannabinoid agonist WIN 55,212-2 (WIN) regulates the phosphorylation state of TRPV1. Here, we show that treatment of primary rat trigeminal ganglion cultures with WIN led to dephosphorylation of TRPV1, specifically at threonine residues. Utilizing Chinese hamster ovary cell lines, we demonstrate that Thr144 and Thr370 were dephosphorylated, leading to desensitization of the TRPV1 receptor. This post-translational modification occurred through activation of the phosphatase calcineurin (protein phosphatase 2B) following WIN treatment. Furthermore, knockdown of TRPA1 (transient receptor potential subtype A1) expression in sensory neurons by specific small interfering RNA abolished the WIN effect on TRPV1 dephosphorylation, suggesting that WIN acts through TRPA1. We also confirm the importance of TRPA1 in WIN-induced dephosphorylation of TRPV1 in Chinese hamster ovary cells through targeted expression of one or both receptor channels. These results imply that the cannabinoid WIN modulates the sensitivity of sensory neurons to TRPV1 activation by altering receptor phosphorylation. In addition, our data could serve as a useful strategy in determining the potential use of certain cannabinoids as peripheral analgesics.


Neuroscience | 2003

The neuronal distribution of cannabinoid receptor type 1 in the trigeminal ganglion of the rat

Theodore J. Price; G Helesic; D Parghi; Kenneth M. Hargreaves; Christopher M. Flores

Cannabinoid compounds have been shown to produce antinociception and antihyperalgesia by acting upon cannabinoid receptors located in both the CNS and the periphery. A potential mechanism by which cannabinoids could inhibit nociception in the periphery is the activation of cannabinoid receptors located on one or more classes of primary nociceptive neurons. To address this hypothesis, we evaluated the neuronal distribution of cannabinoid receptor type 1 (CB1) in the trigeminal ganglion (TG) of the adult rat through combined in situ hybridization (ISH) and immunohistochemistry (IHC). CB1 receptor mRNA was localized mainly to medium and large diameter neurons of the maxillary and mandibular branches of the TG. Consistent with this distribution, in a de facto nociceptive sensory neuron population that exhibited vanilloid receptor type 1 immunoreactivity, colocalization with CB1 mRNA was also sparse (<5%). Furthermore, very few neurons (approximately 5%) in the peptidergic (defined as calcitonin gene-related peptide- or substance P-immunoreactive) or the isolectin B4-binding sensory neuron populations contained CB1 mRNA. In contrast, and consistent with the neuron-size distribution for CB1, nearly 75% of CB1-positive neurons exhibited N52-immunoreactivity, a marker of myelinated axons. These results indicate that in the rat TG, CB1 receptors are expressed predominantly in neurons that are not thought to subserve nociceptive neurotransmission in the noninjured animal. Taken together with the absence of an above background in situ signal for CB2 mRNA in TG neurons, these findings suggest that the peripherally mediated antinociceptive effects of cannabinoids may involve either as yet unidentified receptors or interaction with afferent neuron populations that normally subserve non-nociceptive functions.


BMC Neuroscience | 2005

Treatment of trigeminal ganglion neurons in vitro with NGF, GDNF or BDNF: effects on neuronal survival, neurochemical properties and TRPV1-mediated neuropeptide secretion

Theodore J. Price; Michael D Louria; Damaries Candelario-Soto; Gregory O. Dussor; Nathanial A. Jeske; Amol M. Patwardhan; Anibal Diogenes; Amanda A Trott; Kenneth M. Hargreaves; Christopher M. Flores

BackgroundNerve growth factor (NGF), glial cell line-derived neurotrophic factor (GDNF) and brain-derived neurotrophic factor (BDNF) all play important roles in the development of the peripheral sensory nervous system. Additionally, these growth factors are proposed to modulate the properties of the sensory system in the adult under pathological conditions brought about by nerve injury or inflammation. We have examined the effects of NGF, GDNF and BDNF on adult rat trigeminal ganglion (TG) neurons in culture to gain a better understanding of how these growth factors alter the cytochemical and functional phenotype of these neurons, with special attention to properties associated with nociception.ResultsCompared with no growth factor controls, GDNF, at 1 and 100 ng/ml, significantly increased by nearly 100% the number of neurons in culture at 5 days post-plating. A significant, positive, linear trend of increasing neuron number as a function of BDNF concentration was observed, also peaking at nearly 100%. NGF treatment was without effect. Chronic treatment with NGF and GDNF significantly and concentration-dependently increased 100 nM capsaicin (CAP)-evoked calcitonin gene-related peptide (CGRP) release, reaching approximately 300% at the highest concentration tested (100 ng/ml). Also, NGF and GDNF each augmented anandamide (AEA)- and arachidonyl-2-chloroethylamide (ACEA)-evoked CGRP release, while BDNF was without effect. Utilizing immunohistochemistry to account for the proportions of TRPV1- or CGRP-positive neurons under each growth factor treatment condition and then standardizing evoked CGRP release to these proportions, we observed that NGF was much more effective in enhancing CAP- and 50 mM K+-evoked CGRP release than was GDNF. Furthermore, NGF and GDNF each altered the concentration-response function for CAP- and AEA-evoked CGRP release, increasing the Emax without altering the EC50 for either compound.ConclusionsTaken together, our results illustrate that NGF, GDNF and BDNF differentially alter TG sensory neuron survival, neurochemical properties and TRPV1-mediated neuropeptide release in culture. In particular, our findings suggest that GDNF and NGF differentially modulate TRPV1-mediated neuropeptide secretion sensitivity, with NGF having a much greater effect on a per neuron basis than GDNF. These findings are discussed in relation to possible therapeutic roles for growth factors or their modulators in pathological pain states, especially as these relate to the trigeminal system.


Neuroscience | 2006

The RNA binding and transport proteins staufen and fragile X mental retardation protein are expressed by rat primary afferent neurons and localize to peripheral and central axons.

Theodore J. Price; Christopher M. Flores; Fernando Cervero; Kenneth M. Hargreaves

Neuronal proteins have been traditionally viewed as being derived solely from the soma; however, accumulating evidence indicates that dendritic and axonal sites are capable of a more autonomous role in terms of new protein synthesis. Such extra-somal translation allows for more rapid, on-demand regulation of neuronal structure and function than would otherwise be possible. While mechanisms of dendritic RNA transport have been elucidated, it remains unclear how RNA is trafficked into the axon for this purpose. Primary afferent neurons of the dorsal root (DRG) and trigeminal (TG) ganglia have among the longest axons in the neuraxis and such axonal protein synthesis would be advantageous, given the greater time involved for protein trafficking to occur via axonal transport. Therefore, we hypothesized that these primary sensory neurons might express proteins involved in RNA transport. Rat DRG and TG neurons expressed staufen (stau) 1 and 2 (detected at the mRNA level) and stau2 and fragile x mental retardation protein (FMRP; detected at the protein level). Stau2 mRNA was also detected in human TG neurons. Stau2 and FMRP protein were localized to the sciatic nerve and dorsal roots by immunohistochemistry and to dorsal roots by Western blot. Stau2 and FMRP immunoreactivities colocalized with transient receptor potential channel type 1 immunoreactivity in sensory axons of the sciatic nerve and dorsal root, suggesting that these proteins are being transported into the peripheral and central terminals of nociceptive sensory axons. Based on these findings, we propose that stau2 and FMRP proteins are attractive candidates to subserve RNA transport in sensory neurons, linking somal transcriptional events to axonal translation.


Brain Research | 2006

Protein expression and mRNA cellular distribution of the NKCC1 cotransporter in the dorsal root and trigeminal ganglia of the rat

Theodore J. Price; Kenneth M. Hargreaves; Fernando Cervero

Primary afferent neurons maintain depolarizing responses to GABA into adulthood. The molecular basis for this GABAergic response appears to be the Na+K+2Cl- cotransporter NKCC1 that contributes to the maintenance of a high intracellular chloride concentration. Recently, a role for NKCC1 has been proposed in nociceptive processing which makes it timely to gain a better understanding of the distribution of NKCC1 in sensory ganglia. Here, we describe that, in the rat, NKCC1 mRNA is predominately expressed by small and medium diameter dorsal root (DRG) and trigeminal (TG) ganglion neurons. The colocalization of NKCC1 mRNA with sensory neuron population markers was assessed. In the DRG, many NKCC1 mRNA-expressing neurons colocalized peripherin (57.0+/-2.5%), calcitonin-gene-related peptide (CGRP, 39.2+/-4.4%) or TRPV1 immunoreactivity (50.0+/-1.9%) whereas only 8.7+/-1.2% were co-labeled with a marker for large diameter afferents (N52). Similarly, in the TG, NKCC1 mRNA-expressing neurons frequently colocalized peripherin (50.0+/-3.0%), CGRP (35.4+/-2.6%) or TRPV1 immunoreactivity (44.7+/-1.2%) while 14.8+/-1.3% were co-labeled with the N52 antibody. NKCC1 mRNA was also detected in satellite glial (SGCs) in both the DRG and TG. Colocalization of NKCC1 protein with the SGC marker NG2 confirmed the phenotype of these NKCC1-expressing glial cells. In contrast to in situ hybridization experiments, we did not observe NKCC1 immunoreactivity in primary afferent somata. These findings suggest that NKCC1 is expressed in anatomically appropriate cells in order to modulate GABAergic responses in nociceptive neurons. Moreover, these results suggest the possibility of a functional role of NKCC1 in the glial cells closely apposed to primary sensory afferents.


Molecular Pain | 2007

Spinal NKCC1 blockade inhibits TRPV1-dependent referred allodynia

Mark H. Pitcher; Theodore J. Price; José Manuel Entrena; Fernando Cervero

BackgroundThe Na+, K+, 2Cl- type I cotransporter (NKCC1) and TRPV1 receptors, at the level of the dorsal horn, have been implicated in mediating allodynia in response to an inflammatory insult. The NKCC1 cotransporter regulates intracellular [Cl-] and thus the magnitude and polarity of GABAA receptor responses in neurons. TRPV1 receptors transduce diverse chemical and natural stimuli in nociceptors and are critical for inflammatory hyperalgesia.ResultsHere we have tested the role of spinal NKCC1 cotransporters and TRPV1 receptors in referred allodynia in a model of visceral hyperalgesia in mice. Intrathecal (IT) injection of the NKCC1 inhibitor bumetanide (BUM, 1 nmol) inhibited referred, abdominal allodynia evoked by an intracolonic capsaicin injection. BUM was effective when injected IT either before or up to 4 hrs after the establishment of referred allodynia. The TRPV1 antagonist AMG 9810 (1 nmol) also inhibited referred allodynia in this model suggesting the involvement of an endogenous TRPV1 agonist in the dorsal horn in referred allodynia. In support of this suggestion, the endovanilloid TRPV1 agonist, narachidonoyl- dopamine (NADA, 1 or 10 nmol, IT) evoked stroking allodynia in the hindpaw that was blocked by co-treatment with AMG 9810 (1 nmol). The TRPV1-dependent stroking allodynia caused by NADA appeared to be functionally linked to NKCC1 because BUM (1 nmol) also inhibited NADA-evoked stroking allodynia.ConclusionOur findings indicate that spinal NKCC1 and TRPV1 are critical for referred allodynia mediated by a painful visceral stimulus. Moreover, they suggest that endogenous TRPV1 agonists, released in the CNS in painful conditions, might stimulate TRPV1 receptors on primary afferents that, in turn, play a role in increasing NKCC1 activity leading to allodynia.


Neuroscience Letters | 2005

Pharmacological interactions between calcium/calmodulin-dependent kinase II α and TRPV1 receptors in rat trigeminal sensory neurons

Theodore J. Price; Nathanial A. Jeske; Christopher M. Flores; Kenneth M. Hargreaves

Multiple lines of evidence suggest that calcium/calmodulin-dependent kinase II alpha (CaMKIIalpha) plays an important role in the spinal dorsal horn in nociceptive models of chemical, inflammatory and nerve injury. Moreover, CaMKIIalpha phosphorylates the vanilloid receptor type 1 (TRPV1), thereby regulating vanilloid agonist binding to the receptor. Herein, we have explored a possible interaction of CaMKIIalpha activity with the TRPV1 receptor in rat trigeminal ganglion (TG) neurons in vitro. Inhibition of CaMKIIalpha with KN-93 (5 microM) inhibited capsaicin (CAP)- and n-arachidonoyl-dopamine (NADA)-evoked calcitonin gene-related peptide (CGRP) release effectively decreasing the Emax for both compounds. This effect was not mimicked by the inactive compound KN-92 (5 microM), indicating that the effect was mediated by CaMKIIalpha inhibition. CAP also stimulated a significant approximately 50% increase in autophosphorylation of CaMKIIalpha at Thr286/287. Immunocytochemistry for phospho-CaMKIIalpha indicated that this effect specifically occurred in TRPV1-positive TG neurons. These findings indicate that phopho-CaMKIIalpha is likely to play a role in presynaptic primary afferents in animal models of nociceptive hypersensitivity and provide support for CaMKIIalpha modulation of TRPV1 activity in sensory neurons.


European Journal of Neuroscience | 2003

Potentiation of evoked calcitonin gene‐related peptide release from oral mucosa: a potential basis for the pro‐inflammatory effects of nicotine

Gregory O. Dussor; Anthony S. Leong; Nicholas B. Gracia; Sonja Kilo; Theodore J. Price; Kenneth M. Hargreaves; Christopher M. Flores

Inflammation of the buccal mucosa, gingiva and periodontal tissues is a significant problem in users of nicotine‐containing tobacco products; however, the potential role of nicotine in the development of this inflammation is unclear. In many tissues, nicotine, acting through nicotinic acetylcholine receptors (nAChRs), has been shown to increase the release of the pro‐inflammatory mediator calcitonin gene‐related peptide (CGRP) thereby potentially contributing to neurogenic inflammation. The purpose of the present studies was to determine the effects of nicotine and other nAChR agonists on capsaicin‐evoked immunoreactive CGRP (iCGRP) release from rat buccal mucosa and to identify a potential cellular basis for these effects. Using a previously validated model of in vitro superfusion, we show that the nAChR agonists nicotine (EC50 557u2003µm), epibatidine (EC50 317u2003pm) and cytisine (EC50 4.83u2003nm) potentiated capsaicin‐evoked iCGRP release in a concentration‐dependent manner by 123, 70 and 76%, respectively. The expression and distribution patterns of the mRNA transcripts encoding the α3, α4 and α6 nAChR subunits and their colocalization with CGRP and the capsaicin receptor VR1 were examined in rat trigeminal ganglion using combined in situ hybridization and immunohistofluorescence. Of all trigeminal neurons counted, mRNA encoding the α3, α4 and α6 subunits was found, respectively, in 14.45, 9.2 and 19.21% of neurons. The cell body diameter of most neurons containing any nAChR subunit was in the 30–40u2003µm range with slightly fewer in the 20–30u2003µm range. Co‐localization of these α subunit transcripts with either CGRP or VR1 immunoreactivity ranged from approximately 5 to 7% for α4 and over 8% for α3 to 18% for α6. These data support the hypothesis that nicotinic agents, acting at nAChRs contained on primary sensory neurons, are capable of directly modulating the stimulated release of iCGRP. In the case of users of nicotine‐containing tobacco products, this modulation could contribute to inflammatory processes within the oral cavity.


British Journal of Pharmacology | 2004

Cannabinoid receptor‐independent actions of the aminoalkylindole WIN 55,212‐2 on trigeminal sensory neurons

Theodore J. Price; Amol M. Patwardhan; Armen N. Akopian; Kenneth M. Hargreaves; Christopher M. Flores

The prototypical aminoalkylindole cannabinoid WIN 55,212‐2 (WIN‐2) has been shown to produce antihyperalgesia through a peripheral mechanism of action. However, it is not known whether WIN‐2 exerts this action directly via cannabinoid receptors located on primary afferents or if other, perhaps indirect or noncannabinoid, mechanisms are involved. To address this question, we have examined the specific actions of WIN‐2 on trigeminal ganglion (TG) neurons in vitro by quantifying its ability to modulate the evoked secretion of the proinflammatory neuropeptide CGRP as well as the inflammatory mediator‐induced generation of cAMP. WIN‐2 evoked CGRP release from TG neurons in vitro (EC50=26 μM) in a concentration‐ and calcium‐dependent manner, which was mimicked by the cannabinoid receptor‐inactive enantiomer WIN 55,212‐3 (WIN‐3). Moreover, WIN‐2‐evoked CGRP release was attenuated by the nonselective cation channel blocker ruthenium red but not by the vanilloid receptor type 1 (TRPV1) antagonist capsazepine, suggesting that, unlike certain endogenous and synthetic cannabinoids, WIN‐2 is not a TRPV1 agonist but rather acts at an as yet unidentified cation channel. The inhibitory effects of WIN‐2 on TG neurons were also examined. WIN‐2 neither inhibited capsaicin‐evoked CGRP release nor did it inhibit forskolin‐, isoproteranol‐ or prostaglandin E2‐stimulated cAMP accumulation. On the other hand, WIN‐2 significantly inhibited (EC50=1.7 μM) 50 mM K+‐evoked CGRP release by approximately 70%. WIN‐2 inhibition of 50 mM K+‐evoked CGRP release was not reversed by antagonists of cannabinoid type 1 (CB1) receptor, but was mimicked in magnitude and potency (EC50=2.7 μM) by its cannabinoid‐inactive enantiomer WIN‐3. These findings indicate that WIN‐2 exerts both excitatory and inhibitory effects on TG neurons, neither of which appear to be mediated by CB1, CB2 or TRPV1 receptors, but by a novel calcium‐dependent mechanism. The ramifications of these results are discussed in relation to our current understanding of cannabinoid/vanilloid interactions with primary sensory neurons.

Collaboration


Dive into the Theodore J. Price's collaboration.

Top Co-Authors

Avatar

Kenneth M. Hargreaves

University of Texas Health Science Center at San Antonio

View shared research outputs
Top Co-Authors

Avatar

Christopher M. Flores

University of Texas Health Science Center at San Antonio

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Gregory O. Dussor

University of Texas Health Science Center at San Antonio

View shared research outputs
Top Co-Authors

Avatar

Armen N. Akopian

University of Texas Health Science Center at San Antonio

View shared research outputs
Top Co-Authors

Avatar

Nathanial A. Jeske

University of Texas Health Science Center at San Antonio

View shared research outputs
Top Co-Authors

Avatar

Nathaniel Aaron Jeske

University of Texas Health Science Center at San Antonio

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge