Network


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

Hotspot


Dive into the research topics where Xiao-Ying Hua is active.

Publication


Featured researches published by Xiao-Ying Hua.


European Journal of Neuroscience | 2005

Intrathecal minocycline attenuates peripheral inflammation-induced hyperalgesia by inhibiting p38 MAPK in spinal microglia.

Xiao-Ying Hua; Camilla I. Svensson; Tomohiro Matsui; Bethany Fitzsimmons; Tony L. Yaksh; Michael Webb

Activation of p38 mitogen‐activated protein kinase (p38) in spinal microglia is implicated in spinal nociceptive processing. Minocycline, a tetracycline derivative, displays selective inhibition of microglial activation, a function that is distinct from its antibiotic activity. In the present study we examined antinociceptive effects of intrathecal (IT) administration of minocycline in experimental models of inflammation‐evoked hyperalgesia in addition to the effect of minocycline on stimulation‐induced activation of p38 in spinal microglia. Intrathecal minocycline produced a dose‐dependent reduction of formalin‐evoked second‐phase flinching behaviour in rats, and prevented thermal hyperalgesia induced by carrageenan injection into the paw. In contrast, systemic delivery (intraperitoneally) of minocycline inhibited the first but not the second phase of formalin‐induced flinching, and it had no effect on carrageenan‐induced hyperalgesia. Centrally mediated hyperalgesia induced by IT delivery of N‐methyl‐d‐aspartate was completely blocked by IT minocycline. An increase in phosphorylation (activation) of p38 (P‐p38) was observed in the dorsal spinal cord after carrageenan paw injection, assessed by both Western blotting and immunohistochemistry. The increased P‐p38 immunoreactivity was seen primarily in microglia but also in a small population of neurons. Minocycline, at the IT dose that blocked carrageenan‐induced hyperalgesia, also attenuated the increased P‐p38 in microglia. In addition, minocycline suppressed lipopolysaccharide‐evoked P‐p38 in cultured spinal microglial cells. Taken together, these findings show that minocycline given IT produces a potent and consistent antinociception in models of tissue injury and inflammation‐evoked pain, and they provide strong support for the idea that this effect is mediated by direct inhibition of spinal microglia and subsequent activation of p38 in these cells.


Journal of Neurochemistry | 2005

Spinal p38β isoform mediates tissue injury‐induced hyperalgesia and spinal sensitization

Camilla I. Svensson; Bethany Fitzsimmons; Sara Azizi; Henry C. Powell; Xiao-Ying Hua; Tony L. Yaksh

Antagonist studies show that spinal p38 mitogen‐activated protein kinase plays a crucial role in spinal sensitization. However, there are two p38 isoforms found in spinal cord and the relative contribution of these two to hyperalgesia is not known. Here we demonstrate that the isoforms are distinctly expressed in spinal dorsal horn: p38α in neurons and p38β in microglia. In lieu of isoform selective inhibitors, we examined the functional role of these two individual isoforms in nociception by using intrathecal isoform‐specific antisense oligonucleotides to selectively block the expression of the respective isoform. In these rats, down‐regulation of spinal p38β, but not p38α, prevented nocifensive flinching evoked by intraplantar injection of formalin and hyperalgesia induced by activation of spinal neurokinin‐1 receptors through intrathecal injection of substance P. Both intraplantar formalin and intrathecal substance P produced an increase in spinal p38 phosphorylation and this phosphorylation (activation) was prevented when spinal p38β, but not p38α, was down‐regulated. Thus, spinal p38β, probably in microglia, plays a significant role in spinal nociceptive processing and represents a potential target for pain therapy.


Pain | 2000

Spinal PKC activity and expression: role in tolerance produced by continuous spinal morphine infusion.

Vinicio Granados-Soto; Iveta Kalcheva; Xiao-Ying Hua; Alexandra C. Newton; Tony L. Yaksh

&NA; It has been hypothesized that spinal morphine tolerance results from protein kinase C (PKC) mediated phosphorylation. Chronic lumbar intrathecal (i.t.) infusion of morphine (20 nmol/&mgr;l/h) was shown to produce antinociception on day 1 (d1) that disappeared by d5 (tolerance). On d6, a bolus i.t. probe dose of morphine (60 nmol) produced a more profound antinociception in saline‐infused rats than in morphine‐infused rats. Coinfusion of morphine with a PKC inhibitor, chelerythrine, prevented tolerance to the probe morphine dose. Bolus i.t. chelerythrine or GF109203X (GF), another PKC inhibitor, on d5, but not the inactive homologue of GF Bisindolymaleimide V, also blocked development of tolerance after 24 h. I.t. morphine infusion, but not saline, produced a 2‐fold increase in dorsal horn PKC phosphorylating activity and in the expression of PKC&agr;/&ggr;. Bolus chelerythrine on d5 after spinal morphine infusion blocked upon an increase in PKC activity, confirming that at the behaviorally active dose the drug had the intended biochemical effect upon spinal PKC activity. PKC activity and protein expression did not change when assessed 1 h after bolus i.t. morphine in naive rats. Thus, tolerance produced by morphine infusion is dependent upon an increase in local phosphorylating activity by PKC. Blocking the PKC activity prevents expression of the morphine tolerance.


The Journal of Neuroscience | 2005

Inhibition by Spinal μ- and δ-Opioid Agonists of Afferent-Evoked Substance P Release

Ichiro Kondo; Juan Carlos G. Marvizón; Bingbing Song; Frances Salgado; Simone Codeluppi; Xiao-Ying Hua; Tony L. Yaksh

Opioid μ- and δ-receptors are present on the central terminals of primary afferents, where they are thought to inhibit neurotransmitter release. This mechanism may mediate analgesia produced by spinal opiates; however, when they used neurokinin 1 receptor (NK1R) internalization as an indicator of substance P release, Trafton et al. (1999) noted that this evoked internalization was altered only modestly by morphine delivered intrathecally at spinal cord segment S1-S2. We reexamined this issue by studying the effect of opiates on NK1R internalization in spinal cord slices and in vivo. In slices, NK1R internalization evoked by dorsal root stimulation at C-fiber intensity was abolished by the μ agonist [d-Ala2, N-Me-Phe4, Gly-ol5]-enkephalin (DAMGO) (1 μm) and decreased by the δ agonist [d-Phe2,5]-enkephalin (DPDPE) (1 μm). In vivo, hindpaw compression induced NK1R internalization in ipsilateral laminas I-II. This evoked internalization was significantly reduced by morphine (60 nmol), DAMGO (1 nmol), and DPDPE (100 nmol), but not by the κ agonist trans-(1S,2S)-3,4-dichloro-N-mathyl-N-[2-(1-pyrrolidinyl)cyclohexyl]-benzeneacetamide hydrochloride (200 nmol), delivered at spinal cord segment L2 using intrathecal catheters. These doses of the μ and δ agonists were equi-analgesic as measured by a thermal escape test. Lower doses neither produced analgesia nor inhibited NK1R internalization. In contrast, morphine delivered by percutaneous injections at S1-S2 had only a modest effect on thermal escape, even at higher doses. Morphine decreased NK1R internalization after systemic delivery, but at a dose greater than that necessary to produce equivalent analgesia. All effects were reversed by naloxone. These results indicate that lumbar opiates inhibit noxious stimuli-induced neurotransmitter release from primary afferents at doses that are confirmed behaviorally as analgesic.


The Journal of Neuroscience | 2011

Spinal phosphinositide 3-kinase-Akt-mammalian target of rapamycin signaling cascades in inflammation-induced hyperalgesia.

Qinghao Xu; Bethany Fitzsimmons; Joanne J. Steinauer; Audrey O’Neill; Alexandra C. Newton; Xiao-Ying Hua; Tony L. Yaksh

Phosphinositide 3-kinase (PI3K), Akt, and their downstream kinase, mammalian target of rapamycin (mTOR), are implicated in neural plasticity. The functional linkages of this signaling cascade in spinal dorsal horn and their role in inflammatory hyperalgesia have not been elucidated. In the present work, we identified the following characteristics of this cascade. (1) Local inflammation led to increase in rat dorsal horn phosphorylation (activation) of Akt (pAkt) and mTOR (pmTOR), as assessed by Western blotting and immunocytochemistry. (2) Increased pAkt and pmTOR were prominent in neurons in laminae I, III, and IV, whereas pmTOR and its downstream targets (pS6, p4EBP) were also observed in glial cells. (3) Intrathecal treatment with inhibitors to PI3K or Akt attenuated Formalin-induced second-phase flinching behavior, as well as carrageenan-induced thermal hyperalgesia and tactile allodynia. (4) Intrathecal rapamycin (an mTORC1 inhibitor) displayed anti-hyperalgesic effect in both inflammatory pain models. Importantly, intrathecal wortmannin at anti-hyperalgesic doses reversed the evoked increase not only in Akt but also in mTORC1 signaling (pS6/p4EBP). (5) pAkt and pmTOR are expressed in neurokinin 1 receptor-positive neurons in laminae I–III after peripheral inflammation. Intrathecal injection of Substance P activated this cascade (increased phosphorylation) and resulted in hyperalgesia, both of which effects were blocked by intrathecal wortmannin and rapamycin. Together, these findings reveal that afferent inputs trigged by peripheral inflammation initiate spinal activation of PI3K–Akt–mTOR signaling pathway, a component of which participates in neuronal circuits of facilitated pain processing.


FEBS Letters | 2006

Descending serotonergic facilitation of spinal ERK activation and pain behavior

Camilla I. Svensson; Thao K. Tran; Bethany Fitzsimmons; Tony L. Yaksh; Xiao-Ying Hua

Serotonin (5‐HT) derived from bulbo‐spinal projection is released by nociceptive input into the spinal dorsal horn. Here we report that formalin injection in the paw produced pain behavior (flinching) and phosphorylation of spinal ERK1/2 (P‐ERK1/2, indicating activation) in rats. Depletion of spinal 5‐HT by intrathecal (IT) 5,7‐DHT, a serotonergic neurotoxin, profoundly reduced formalin evoked flinching and the increase in P‐ERK1/2. Ondansetron (a 5‐HT3 receptor antagonist) at IT doses that inhibited flinching also attenuated spinal ERK activation. These findings reveal that primary afferent‐evoked activation of spinal ERK requires the input from an excitatory 5‐HT descending pathway.


Neuroscience Letters | 1999

Inhibition of spinal protein kinase C reduces nerve injury-induced tactile allodynia in neuropathic rats

Xiao-Ying Hua; Ping Chen; Tony L. Yaksh

We investigated the effects of inhibiting spinal protein kinases including PKC, PKA and PKG on tactile allodynia in rats with a unilateral tight ligation on L5/L6 spinal nerves (Chung model). The intrathecal (IT) delivery of GF109203X, a PKC inhibitor, produced a potent and long lasting anti-allodynic effect. The effect was dose-dependent and stereospecific. Bisindolymaleimide V, an inactive homologue of GF, had no effect. Additionally, two other PKC inhibitors, PKC19-31 and chelerythrine, displayed significant anti-allodynic action. Spinal PKA, but not PKG, is likely involved in Chung tactile allodynia, since H89 (a PKA inhibitor) showed anti-allodynic activity, while KT5823 (a PKG inhibitor) had only a minor effect. These data emphasize that spinal PKC plays an important role in nerve injury-induced tactile allodynia. Other protein kinases such as PKA may also contribute to this phenomenon.


Neuroscience | 1999

Intrathecal substance P-induced thermal hyperalgesia and spinal release of prostaglandin E2 and amino acids.

Xiao-Ying Hua; Ping Chen; Martin Marsala; Tony L. Yaksh

Substance P is an important neuromediator in spinal synaptic transmission, particularly in processing nociceptive afferent information. The effects of substance P are mediated by activation of the neurokinin 1 receptor. Evidence has suggested that excitatory amino acids such as glutamate, and prostaglandins including prostaglandin E2 are involved in the enhanced spinal excitability and hyperalgesia produced by spinal substance P. In the present study, we have demonstrated that intrathecal injection of substance P (20 nmol) in rats chronically implanted with intrathecal dialysis catheters induced a decrease in thermal paw withdrawal latency (before: 10.4+/-0.3 s; after 7.6+/-0.6 s), which was accompanied by an increase in prostaglandin E2 (362+/-37% of baseline), glutamate (267+/-84%) and taurine (279+/-57%), but not glycine, glutamine, serine or asparagine. Intrathecal injection of artificial cerebrospinal fluid had no effect upon the behavior or release. Substance P-induced thermal hyperalgesia and prostaglandin E2 release were significantly attenuated by a selective neurokinin 1 receptor antagonist RP67580, but not by an enantiomer RP68651. However, substance P-induced release of glutamate and taurine was not reduced by treatment with RP67580. SR140333, another neurokinin 1 receptor antagonist, displayed the same effects as RP67580 (i.e. block of thermal hyperalgesia and prostaglandin E2 release, but not release of amino acids). These results provide direct evidence suggesting that the spinal substance P-induced thermal hyperalgesia is mediated by an increase in spinal prostaglandin E2 via activation of the neurokinin 1 receptor. These findings define an important linkage between small afferents, sensory neurotransmitter release and spinal prostanoids in the cascade of spinally-mediated hyperalgesia. The evoked release of glutamate is apparently not a result of activation of neurokinin 1 receptors. Accordingly, consistent with other pharmacological data, acute spinal glutamate release does not contribute to the hyperalgesia induced by activation of spinal neurokinin 1 receptors.


British Journal of Pharmacology | 2010

Spinal glial TLR4-mediated nociception and production of prostaglandin E2 and TNF

Saito O; Camilla I. Svensson; Matthew W. Buczynski; Wegner K; Xiao-Ying Hua; Codeluppi S; Schaloske Rh; Deems Ra; Edward A. Dennis; Tony L. Yaksh

Background and purpose:  Toll‐like receptor 4 (TLR4) expressed on spinal microglia and astrocytes has been suggested to play an important role in the regulation of pain signalling. The purpose of the present work was to examine the links between TLR4, glial activation and spinal release of prostaglandin E2 (PGE2) and tumour necrosis factor (TNF), and the role these factors play in TLR4‐induced tactile allodynia.


British Journal of Pharmacology | 2005

Spinal phospholipase A2 in inflammatory hyperalgesia: role of group IVA cPLA2.

Karin Killermann Lucas; Camilla I. Svensson; Xiao-Ying Hua; Tony L. Yaksh; Edward A. Dennis

1 Current work has shown the importance of spinal cyclooxygenase (COX) products in facilitatory processes leading to tissue injury induced hyperalgesia. This cascade must originate with free arachidonic acid (AA) released by the activity of spinal phospholipase A2s (PLA2). In the present work, we studied the role of PLA2s in spinal sensitization. 2 We first demonstrate the presence of constitutive mRNA in the spinal cord for PLA2 Groups IB, IIA, IIC, IVA, V and VI by reverse transcription–polymerase chain reaction (RT–PCR) and sequencing. Using quantitative‐PCR, we found that Group IVA cPLA2 and Group VI iPLA2 are the predominant PLA2 messages in the spinal cord. Western blotting and activity assays specific for Group IVA cPLA2 and Group VI iPLA2 verified the presence of these enzymes. PLA2 activity in spinal cord homogenates was suppressed by methyl arachidonyl fluorophosphonate (MAFP) and arachidonyl trifluoromethylketone (AACOCF3), mixed inhibitors of Group IVA cPLA2 and Group VI iPLA2 as well as by bromoenol lactone (BEL), a Group VI iPLA2 inhibitor. The spinal expression of PLA2 mRNA or protein was not altered in the face of peripheral inflammation. Secondly, we showed that intrathecal (i.t.) administration of MAFP and AACOCF3, but not BEL, dose‐dependently prevented thermal hyperalgesia induced by intraplantar carrageenan as well as formalin‐induced flinching. Finally, i.t. injection of AACOCF3, at antihyperalgesic doses, decreased the release of prostaglandin E2 (PGE2) into spinal dialysate evoked by i.t. NMDA, while i.t. injection of BEL had no effect. 3 Taken together, this work points to a role for constitutive Group IVA cPLA2 in spinal nociceptive processing.

Collaboration


Dive into the Xiao-Ying Hua's collaboration.

Top Co-Authors

Avatar

Tony L. Yaksh

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Shelle Malkmus

University of California

View shared research outputs
Top Co-Authors

Avatar

Tamas Bartfai

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar

Martin Marsala

University of California

View shared research outputs
Top Co-Authors

Avatar

Ping Chen

University of California

View shared research outputs
Top Co-Authors

Avatar

B. K Crawley

University of California

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge