Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Semin Cell Dev Biol ; 155(Pt B): 66-73, 2024 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-37391348

RESUMO

Thrombospondin-4 (TSP-4) belongs to the extracellular matrix glycoprotein family of thrombospondins (TSPs). The multidomain, pentameric structure of TSP-4 allows its interactions with numerous extracellular matrix components, proteins and signaling molecules that enable its modulation to various physiological and pathological processes. Characterization of TSP-4 expression under development and pathogenesis of disorders has yielded important insights into mechanisms underlying the unique role of TSP-4 in mediating various processes including cell-cell, cell-extracellular matrix interactions, cell migration, proliferation, tissue remodeling, angiogenesis, and synaptogenesis. Maladaptation of these processes in response to pathological insults and stress can accelerate the development of disorders including skeletal dysplasia, osteoporosis, degenerative joint disease, cardiovascular diseases, tumor progression/metastasis and neurological disorders. Overall, the diverse functions of TSP-4 suggest that it may be a potential marker or therapeutic target for prognosis, diagnosis, and treatment of various pathological conditions upon further investigations. This review article highlights recent findings on the role of TSP-4 in both physiological and pathological conditions with a focus on what sets it apart from other TSPs.


Assuntos
Doenças Cardiovasculares , Trombospondinas , Humanos , Trombospondinas/genética , Trombospondinas/química , Trombospondinas/metabolismo , Matriz Extracelular/metabolismo , Movimento Celular , Morfogênese , Doenças Cardiovasculares/metabolismo
2.
eNeuro ; 6(5)2019.
Artigo em Inglês | MEDLINE | ID: mdl-31533959

RESUMO

The spinal dorsal horn is the first relay structure coding for pain transmission and modulation. Previous anatomical and electrophysiological studies have examined spinal dorsal horn circuit connections and network activity. Further work is required to understand spinal cord sensory information processing that underlies pathological neuropathic pain states. Our previous studies suggest that peripheral nerve injury enhances presynaptic excitatory input onto spinal superficial dorsal horn neurons, which in turn contributes to pathologic nociception. The potential changes in local postsynaptic circuits in the dorsal horn that lead to pathologically heightened behavioral responses to pain remain largely unexplored. We combined whole-cell electrophysiological recordings with laser-scanning photostimulation to test whether peripheral nerve injury in the spinal nerve ligation (SNL) mouse model of neuropathic pain leads to alterations in the functional connectivity of spinal cord circuits including lamina II excitatory interneurons. Here we show that SNL enhances excitation and decreases inhibition to lamina II excitatory interneurons along with their increased glutamate-evoked excitability. The enhanced excitatory postsynaptic input and connectivity evoked by SNL eventually return to normal levels concurrently with the resolution of the neuropathic pain states. The physiological pattern highly correlates with mouse pain behaviors following SNL, supporting a neurophysiological mechanism of central sensitization and neuropathic pain that is functionally localized to the spinal dorsal horn. Together, these data support that SNL induces functional changes in synaptic input and connectivity to lamina II excitatory interneurons that code for pain perception, and thus provide new insights into the mechanism and locus of pain hypersensitivity.


Assuntos
Potenciais Pós-Sinápticos Excitadores/fisiologia , Potenciais Pós-Sinápticos Inibidores/fisiologia , Rede Nervosa/fisiopatologia , Neuralgia/fisiopatologia , Plasticidade Neuronal/fisiologia , Corno Dorsal da Medula Espinal/fisiopatologia , Animais , Masculino , Camundongos , Camundongos Transgênicos , Técnicas de Cultura de Órgãos , Medição da Dor/métodos
3.
J Biol Chem ; 293(42): 16453-16463, 2018 10 19.
Artigo em Inglês | MEDLINE | ID: mdl-30194282

RESUMO

Up-regulation of thrombospondin-4 (TSP4) or voltage-gated calcium channel subunit α2δ1 (Cavα2δ1) proteins in the spinal cord contributes to neuropathic pain development through an unidentified mechanism. We have previously shown that TSP4 interacts with Cavα2δ1 to promote excitatory synaptogenesis and the development of chronic pain states. However, the TSP4 determinants responsible for these changes are not known. Here, we tested the hypothesis that the Cavα2δ1-binding domains of TSP4 are synaptogenic and pronociceptive. We mapped the major Cavα2δ1-binding domains of TSP4 within the coiled-coil and epidermal growth factor (EGF)-like domains in vitro Intrathecal injection of TSP4 fragment proteins containing the EGF-like domain (EGF-LIKE) into naïve rodents was sufficient for inducing behavioral hypersensitivity similar to that produced by an equal molar dose of full-length TSP4. Gabapentin, a drug that binds to Cavα2δ1, blocked EGF-LIKE-induced behavioral hypersensitivity in a dose-dependent manner, supporting the notion that EGF-LIKE interacts with Cavα2δ1 and thereby mediates behavioral hypersensitivity. This notion was further supported by our findings that a peptide within EGF-LIKE (EGFD355-369) could block TSP4- or Cavα2δ1-induced behavioral hypersensitivity after intrathecal injections. Furthermore, only TSP4 proteins that contained EGF-LIKE could promote excitatory synaptogenesis between sensory and spinal cord neurons, which could be blocked by peptide EGFD355-369. Together, these findings indicate that EGF-LIKE is the molecular determinant that mediates aberrant excitatory synaptogenesis and chronic pain development. Blocking interactions between EGF-LIKE and Cavα2δ1 could be an alternative approach in designing target-specific pain medications.


Assuntos
Fator de Crescimento Epidérmico/química , Neuralgia/etiologia , Trombospondinas/química , Animais , Canais de Cálcio/metabolismo , Medição da Dor , Domínios Proteicos , Ratos , Células Receptoras Sensoriais/metabolismo , Medula Espinal/metabolismo , Sinapses
4.
Br J Pharmacol ; 175(12): 2348-2361, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29338087

RESUMO

BACKGROUND AND PURPOSE: Nerve injury induces concurrent up-regulation of the voltage-gated calcium channel subunit Cav α2 δ1 and the extracellular matrix protein thrombospondin-4 (TSP4) in dorsal root ganglia and dorsal spinal cord, leading to the development of a neuropathic pain state. Interactions of these proteins promote aberrant excitatory synaptogenesis that contributes to neuropathic pain state development through unknown mechanisms. We investigated the contributions of Cav α2 δ1 subunits and TSP4 to synaptogenesis, and the pathways involved in vitro, and whether treatment with gabapentin could block this process and pain development in vivo. EXPERIMENTAL APPROACH: A co-culture system of sensory and spinal cord neurons was used to study the contribution from each protein to synaptogenesis and the pathway(s) involved. Anti-synaptogenic actions of gabapentin were studied in TSP4-injected mice. KEY RESULTS: Only presynaptic, but not postsynaptic, Cav α2 δ1 subunits interacted with TSP4 to initiate excitatory synaptogenesis through a pathway modulated by T-type calcium channels. Cav α2 δ1 /TSP4 interactions were not required for maintenance of already formed synapses. In vivo, early, but not delayed, treatment with low-dose gabapentin blocked this pathway and the development of the pain state. CONCLUSIONS AND IMPLICATIONS: Cav α2 δ1 /TSP4 interactions were critical for the initiation, but not for the maintenance, of abnormal synapse formation between sensory and spinal cord neurons. This process was blocked by early, but was not reversed by delayed, treatment with gabapentin. Early intervention with gabapentin may prevent the development of injury-induced chronic pain, resulting from Cav α2 δ1 /TSP4-initiated abnormal synapse formation. LINKED ARTICLES: This article is part of a themed section on Recent Advances in Targeting Ion Channels to Treat Chronic Pain. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v175.12/issuetoc.


Assuntos
Canais de Cálcio/metabolismo , Gabapentina/farmacologia , Neurônios/efeitos dos fármacos , Medula Espinal/citologia , Medula Espinal/efeitos dos fármacos , Sinapses/efeitos dos fármacos , Trombospondinas/metabolismo , Animais , Células Cultivadas , Feminino , Masculino , Neurônios/metabolismo , Medula Espinal/metabolismo , Sinapses/metabolismo
5.
Br J Pharmacol ; 175(12): 2231-2243, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-28646556

RESUMO

Voltage-gated calcium channels (VGCCs) play important roles in physiological functions including the modulation of neurotransmitter release, neuronal network activities, intracellular signalling pathways and gene expression. Some pathological conditions, including nerve injuries, can cause the dysregulation of VGCCs and their subunits. This in turn can lead to a functional maladaptation of VGCCs and their subunits, which can contribute to the development of disorders such as pain sensations. This review has summarized recent findings related to maladaptive changes in the dysregulated VGCC α2 δ1 subunit (Cav α2 δ1 ) with a focus on exploring the mechanisms underlying the contribution of Cav α2 δ1 to pain signal transduction. At least under neuropathic pain conditions, the dysregulated Cav α2 δ1 can modulate VGCC functions as well as other plasticity changes. The latter includes abnormal excitatory synaptogenesis resulting from its interactions with injury-induced extracellular matrix glycoprotein molecule thrombospondins, which is independent of the VGCC functions. Blocking Cav α2 δ1 with gabapentinoids can reverse neuropathic pain significantly with relatively mild side effects, but only in a small population of neuropathic pain patients due to reasons yet to be explored. There are emerging data suggesting that early preventive treatment with gabapentinoids can prevent aberrant excitatory synapse formation and the development of chronic pain. If these findings are confirmed clinically, this could be an attractive approach for neuropathic pain management. LINKED ARTICLES: This article is part of a themed section on Recent Advances in Targeting Ion Channels to Treat Chronic Pain. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v175.12/issuetoc.


Assuntos
Canais de Cálcio/metabolismo , Neuralgia/metabolismo , Animais , Canais de Cálcio/deficiência , Canais de Cálcio/genética , Humanos , Camundongos
6.
Neuropharmacology ; 117: 292-304, 2017 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-28232180

RESUMO

Painful nerve injury disrupts Ca2+ signaling in primary sensory neurons by elevating plasma membrane Ca2+-ATPase (PMCA) function and depressing sarco-endoplasmic reticulum Ca2+-ATPase (SERCA) function, which decreases endoplasmic reticulum (ER) Ca2+ stores and stimulates store-operated Ca2+ entry (SOCE). The extracellular matrix glycoprotein thrombospondin-4 (TSP4), which is increased after painful nerve injury, decreases Ca2+ current (ICa) through high-voltage-activated Ca2+ channels and increases ICa through low-voltage-activated Ca2+ channels in dorsal root ganglion neurons, which are events similar to the effect of nerve injury. We therefore examined whether TSP4 plays a critical role in injury-induced disruption of intracellular Ca2+ signaling. We found that TSP4 increases PMCA activity, inhibits SERCA, depletes ER Ca2+ stores, and enhances store-operated Ca2+ influx. Injury-induced changes of SERCA and PMCA function are attenuated in TSP4 knock-out mice. Effects of TSP4 on intracellular Ca2+ signaling are attenuated in voltage-gated Ca2+ channel α2δ1 subunit (Cavα2δ1) conditional knock-out mice and are also Protein Kinase C (PKC) signaling dependent. These findings suggest that TSP4 elevation may contribute to the pathogenesis of chronic pain following nerve injury by disrupting intracellular Ca2+ signaling via interacting with the Cavα2δ1 and the subsequent PKC signaling pathway. Controlling TSP4 mediated intracellular Ca2+ signaling in peripheral sensory neurons may be a target for analgesic drug development for neuropathic pain.


Assuntos
Sinalização do Cálcio/fisiologia , Células Receptoras Sensoriais/metabolismo , Nervos Espinhais/lesões , Trombospondinas/metabolismo , Animais , Cálcio/metabolismo , Canais de Cálcio/metabolismo , Citoplasma/metabolismo , Modelos Animais de Doenças , Retículo Endoplasmático/metabolismo , Feminino , Gânglios Espinais/metabolismo , Masculino , Potenciais da Membrana/fisiologia , Camundongos da Linhagem 129 , Camundongos Knockout , Neuralgia/metabolismo , ATPases Transportadoras de Cálcio da Membrana Plasmática/metabolismo , Proteína Quinase C/metabolismo , Ratos Sprague-Dawley , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/metabolismo , Trombospondinas/genética
7.
Neurobiol Dis ; 102: 70-80, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28193459

RESUMO

The alpha2delta-1 subunit (α2δ-1) of voltage-gated calcium channels is a receptor for astrocyte-secreted thrombospondins that promote developmental synaptogenesis. Alpha2delta-1 receptors are upregulated in models of injury-induced peripheral pain and epileptogenic neocortical trauma associated with an enhancement of excitatory synaptic connectivity. These results lead to the hypothesis that overexpression of α2δ-1 alone in neocortex of uninjured transgenic (TG) mice might result in increased excitatory connectivity and consequent cortical hyperexcitability and epileptiform activity. Whole cell recordings from layer V pyramidal neurons in somatosensory cortical slices of TG mice showed increased frequency and amplitude of miniature and spontaneous EPSCs and prolonged bursts of polysynaptic EPSCs. Epileptiform field potentials were evoked in layers II/III and V of brain slices from TG mice, but not controls. Dual immunoreactivity for Vglut-2 and PSD95 showed increased density of close appositions in TG mice compared to controls, suggesting an increased number of excitatory synapses. Video-EEG monitoring showed that 13/13 implanted TG mice aged >P21, but not controls, had frequent abnormal spontaneous epileptiform events, consisting of variable duration, high amplitude bi-hemispheric irregular bursts of delta activity, spikes and sharp waves lasting many seconds, with a variable peak frequency of ~1-3Hz, associated with behavioral arrest. The epileptiform EEG abnormalities and behavioral arrests were reversibly eliminated by treatment with i.p. ethosuximide. Behavioral seizures, consisting of ~15-30s duration episodes of rigid arched tail and head and body extension, followed by loss of balance and falling, frequently occurred in adult TG mice during recovery from isoflurane-induced anesthesia, but were rare in WT mice. Results show that over-expression of α2δ-1 subunits increases cortical excitatory connectivity and leads to neocortical hyperexcitability and epileptiform activity associated with behavioral arrests in adult TG mice. Similar increases in expression of α2δ-1 in models of cortical injury may play an important role in epileptogenesis. SIGNIFICANCE: Binding of astrocytic-secreted thrombospondins to their α2δ-1 receptor facilitates excitatory synapse formation and excitatory transmission during cortical development and after injury. Upregulation of α2δ-1 is present in models of injury-induced pain and epileptogenic cortical trauma, along with many other molecular alterations. Here we show that overexpression of α2δ-1 alone in TG mice can enhance excitatory connectivity in neocortex and lead to neural circuit hyperexcitability and episodes of electrographic epileptiform activity, associated with behavioral arrests in transgenic mice. α2δ-1 is the high-affinity receptor for gabapentinoids and a potential target for prophylactic treatment of posttraumatic epilepsy and other disorders in which excessive aberrant excitatory connectivity is a pathophysiological feature.


Assuntos
Canais de Cálcio/metabolismo , Epilepsia/metabolismo , Córtex Somatossensorial/metabolismo , Animais , Anticonvulsivantes/farmacologia , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Astrócitos/patologia , Canais de Cálcio/genética , Epilepsia/tratamento farmacológico , Epilepsia/patologia , Etossuximida/farmacologia , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Hipocampo/patologia , Isoflurano/toxicidade , Masculino , Camundongos Transgênicos , Células Piramidais/efeitos dos fármacos , Células Piramidais/metabolismo , Células Piramidais/patologia , Córtex Somatossensorial/efeitos dos fármacos , Córtex Somatossensorial/patologia , Sinapses/efeitos dos fármacos , Sinapses/metabolismo , Sinapses/patologia , Transmissão Sináptica/efeitos dos fármacos , Transmissão Sináptica/fisiologia , Técnicas de Cultura de Tecidos
8.
Neuron ; 92(1): 160-173, 2016 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-27641496

RESUMO

Experience alters cortical networks through neural plasticity mechanisms. During a developmental critical period, the most dramatic consequence of occluding vision through one eye (monocular deprivation) is a rapid loss of excitatory synaptic inputs to parvalbumin-expressing (PV) inhibitory neurons in visual cortex. Subsequent cortical disinhibition by reduced PV cell activity allows for excitatory ocular dominance plasticity. However, the molecular mechanisms underlying critical period synaptic plasticity are unclear. Here we show that brief monocular deprivation during the critical period downregulates neuregulin-1(NRG1)/ErbB4 signaling in PV neurons, causing retraction of excitatory inputs to PV neurons. Exogenous NRG1 rapidly restores excitatory inputs onto deprived PV cells through downstream PKC-dependent activation and AMPA receptor exocytosis, thus enhancing PV neuronal inhibition to excitatory neurons. NRG1 treatment prevents the loss of deprived eye visual cortical responsiveness in vivo. Our findings reveal molecular, cellular, and circuit mechanisms of NRG1/ErbB4 in regulating the initiation of critical period visual cortical plasticity.


Assuntos
Dominância Ocular/fisiologia , Neuregulina-1/fisiologia , Plasticidade Neuronal/fisiologia , Neurônios/fisiologia , Receptor ErbB-4/fisiologia , Córtex Visual/fisiologia , Animais , Período Crítico Psicológico , Regulação para Baixo/fisiologia , Feminino , Masculino , Camundongos , Inibição Neural/efeitos dos fármacos , Inibição Neural/fisiologia , Neuregulina-1/farmacologia , Plasticidade Neuronal/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Parvalbuminas/metabolismo , Privação Sensorial/fisiologia , Córtex Visual/crescimento & desenvolvimento
9.
Pain ; 157(9): 2068-2080, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27168360

RESUMO

Loss of high-voltage-activated (HVA) calcium current (ICa) and gain of low-voltage-activated (LVA) ICa after painful peripheral nerve injury cause elevated excitability in sensory neurons. Nerve injury is also accompanied by increased expression of the extracellular matrix glycoprotein thrombospondin-4 (TSP4), and interruption of TSP4 function can reverse or prevent behavioral hypersensitivity after injury. We therefore investigated TSP4 regulation of ICa in dorsal root ganglion (DRG) neurons. During depolarization adequate to activate HVA ICa, TSP4 decreases both N- and L-type ICa and the associated intracellular calcium transient. In contrast, TSP4 increases ICa and the intracellular calcium signal after low-voltage depolarization, which we confirmed is due to ICa through T-type channels. These effects are blocked by gabapentin, which ameliorates neuropathic pain by targeting the α2δ1 calcium subunit. Injury-induced changes of HVA and LVA ICa are attenuated in TSP4 knockout mice. In the neuropathic pain model of spinal nerve ligation, TSP4 application did not further regulate ICa of injured DRG neurons. Taken together, these findings suggest that elevated TSP4 after peripheral nerve injury may contribute to hypersensitivity of peripheral sensory systems by decreasing HVA and increasing LVA in DRG neurons by targeting the α2δ1 calcium subunit. Controlling TSP4 overexpression in peripheral sensory neurons may be a target for analgesic drug development for neuropathic pain.


Assuntos
Canais de Cálcio/metabolismo , Regulação da Expressão Gênica/genética , Traumatismos dos Nervos Periféricos/genética , Traumatismos dos Nervos Periféricos/patologia , Células Receptoras Sensoriais/metabolismo , Trombospondinas/deficiência , Análise de Variância , Animais , Cálcio/metabolismo , Bloqueadores dos Canais de Cálcio/farmacologia , Bloqueadores dos Canais de Cálcio/uso terapêutico , Canais de Cálcio/genética , Toxina da Cólera/metabolismo , Modelos Animais de Doenças , Potenciais Evocados/efeitos dos fármacos , Potenciais Evocados/genética , Gânglios Espinais/patologia , Camundongos , Camundongos Knockout , Células Receptoras Sensoriais/efeitos dos fármacos , Trombospondinas/genética , Trombospondinas/farmacologia
10.
J Biol Chem ; 291(25): 13335-48, 2016 Jun 17.
Artigo em Inglês | MEDLINE | ID: mdl-27129212

RESUMO

Peripheral nerve injury induces increased expression of thrombospondin-4 (TSP4) in spinal cord and dorsal root ganglia that contributes to neuropathic pain states through unknown mechanisms. Here, we test the hypothesis that TSP4 activates its receptor, the voltage-gated calcium channel Cavα2δ1 subunit (Cavα2δ1), on sensory afferent terminals in dorsal spinal cord to promote excitatory synaptogenesis and central sensitization that contribute to neuropathic pain states. We show that there is a direct molecular interaction between TSP4 and Cavα2δ1 in the spinal cord in vivo and that TSP4/Cavα2δ1-dependent processes lead to increased behavioral sensitivities to stimuli. In dorsal spinal cord, TSP4/Cavα2δ1-dependent processes lead to increased frequency of miniature and amplitude of evoked excitatory post-synaptic currents in second-order neurons as well as increased VGlut2- and PSD95-positive puncta, indicative of increased excitatory synapses. Blockade of TSP4/Cavα2δ1-dependent processes with Cavα2δ1 ligand gabapentin or genetic Cavα2δ1 knockdown blocks TSP4 induced nociception and its pathological correlates. Conversely, TSP4 antibodies or genetic ablation blocks nociception and changes in synaptic transmission in mice overexpressing Cavα2δ1 Importantly, TSP4/Cavα2δ1-dependent processes also lead to similar behavioral and pathological changes in a neuropathic pain model of peripheral nerve injury. Thus, a TSP4/Cavα2δ1-dependent pathway activated by TSP4 or peripheral nerve injury promotes exaggerated presynaptic excitatory input and evoked sensory neuron hyperexcitability and excitatory synaptogenesis, which together lead to central sensitization and pain state development.


Assuntos
Canais de Cálcio/metabolismo , Neuralgia/metabolismo , Trombospondinas/fisiologia , Animais , Células HEK293 , Humanos , Masculino , Camundongos Transgênicos , Células do Corno Posterior/fisiologia , Sinapses/fisiologia , Potenciais Sinápticos
11.
J Comp Neurol ; 524(2): 309-22, 2016 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-26132987

RESUMO

Trigeminal nerves collecting sensory information from the orofacial area synapse on second-order neurons in the dorsal horn of subnucleus caudalis and cervical C1/C2 spinal cord (Vc/C2, or trigeminocervical complex), which is critical for sensory information processing. Injury to the trigeminal nerves may cause maladaptive changes in synaptic connectivity that plays an important role in chronic pain development. Here we examined whether injury to the infraorbital nerve, a branch of the trigeminal nerves, led to synaptic ultrastructural changes when the injured animals have developed neuropathic pain states. Transmission electron microscopy was used to examine synaptic profiles in Vc/C2 at 3 weeks postinjury, corresponding to the time of peak behavioral hypersensitivity following chronic constriction injury to the infraorbital nerve (CCI-ION). Using established criteria, synaptic profiles were classified as associated with excitatory (R-), inhibitory (F-), and primary afferent (C-) terminals. Each type was counted within the superficial dorsal horn of the Vc/C2 and the means from each rat were compared between sham and injured animals; synaptic contact length was also measured. The overall analysis indicates that rats with orofacial pain states had increased numbers and decreased mean synaptic length of R-profiles within the Vc/C2 superficial dorsal horn (lamina I) 3 weeks post-CCI-ION. Increases in the number of excitatory synapses in the superficial dorsal horn of Vc/C2 could lead to enhanced activation of nociceptive pathways, contributing to the development of orofacial pain states.


Assuntos
Dor Facial/etiologia , Sinapses/patologia , Sinapses/ultraestrutura , Núcleo Inferior Caudal do Nervo Trigêmeo/citologia , Traumatismos do Nervo Trigêmeo/complicações , Análise de Variância , Animais , Modelos Animais de Doenças , Dor Facial/patologia , Lateralidade Funcional , Masculino , Microscopia Eletrônica , Medição da Dor , Ratos , Ratos Sprague-Dawley , Fatores de Tempo , Traumatismos do Nervo Trigêmeo/patologia
12.
J Neurosci Res ; 93(3): 443-53, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25327416

RESUMO

Thrombospondin-4 (TSP4) belongs to a family of large, oligomeric extracellular matrix glycoproteins that mediate interactions between cells and interactions of cells with underlying matrix components. Recent evidence shows that TSP4 might contribute to the generation of neuropathic pain. However, there has been no systematic examination of TSP4 expression in the dorsal root ganglia (DRG) after injury. This study, therefore, investigates whether TSP4 protein level is changed in DRG after injury following spinal nerve ligation (SNL) and spared nerve injury in rats by performing Western blotting, immunohistochemistry, and immunocytochemistry. After nerve ligation, TSP4 protein level is upregulated in the axotomized somata of the fifth lumbar (L5) DRG. There is substantial additional TSP4 in the nonneuronal compartment of the L5 DRG that does not costain for markers of satellite glia, microglia, or Schwann cells and appears to be in the interstitial space. Evidence of intracellular overexpression of TSP4 persists in neurons dissociated from the L5 DRG after SNL. These findings indicate that, following peripheral nerve injury, TSP4 protein expression is elevated in the cytoplasm of axotomized sensory neurons and in the surrounding interstitial space.


Assuntos
Gânglios Espinais/metabolismo , Neuralgia/metabolismo , Neuroglia/metabolismo , Neurônios/metabolismo , Traumatismos dos Nervos Periféricos/metabolismo , Trombospondinas/metabolismo , Animais , Axotomia , Masculino , Neuralgia/etiologia , Traumatismos dos Nervos Periféricos/complicações , Ratos , Ratos Sprague-Dawley , Nervos Espinhais/lesões , Regulação para Cima
13.
Exp Neurol ; 264: 111-20, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25483397

RESUMO

Facet joint injury induces persistent pain that may be maintained by structural plasticity in the spinal cord. Astrocyte-derived thrombospondins, especially thrombospondin-4 (TSP4), have been implicated in synaptogenesis and spinal sensitization in neuropathic pain, but the TSP4 response and its relationship to synaptic changes in the spinal cord have not been investigated for painful joint injury. This study investigates the role of TSP4 in the development and maintenance of persistent pain following injurious facet joint distraction in rats and tests the hypothesis that excitatory synaptogenesis contributes to such pain. Painful facet joint loading induces dorsal horn excitatory synaptogenesis along with decreased TSP4 in the DRG and increased astrocytic release of TSP4 in the spinal cord, all of which parallel the time course of sustained tactile allodynia. Blocking injury-induced spinal TSP4 expression with antisense oligonucleotides or reducing TSP4 activity at its neuronal receptor in the spinal cord with gabapentin treatment both attenuate the allodynia and dorsal horn synaptogenesis that develop after painful facet joint loading. Increased spinal TSP4 also facilitates the development of allodynia and spinal hyperexcitability, even after non-painful physiological loading of the facet joint. These results suggest that spinal TSP4 plays an important role in the development and maintenance of persistent joint-mediated pain by inducing excitatory synaptogenesis and facilitating the transduction of mechanical loading of the facet joint that leads to spinal hyperexcitability.


Assuntos
Artralgia/patologia , Medula Espinal/metabolismo , Sinapses/patologia , Trombospondinas/metabolismo , Articulação Zigapofisária , Potenciais de Ação/efeitos dos fármacos , Aminas/uso terapêutico , Analgésicos/uso terapêutico , Animais , Artralgia/tratamento farmacológico , Ácidos Cicloexanocarboxílicos/uso terapêutico , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Gabapentina , Gânglios Espinais/metabolismo , Gânglios Espinais/patologia , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Proteína Glial Fibrilar Ácida/metabolismo , Hiperalgesia/tratamento farmacológico , Cápsula Articular/patologia , Masculino , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Medição da Dor , Ratos , Medula Espinal/patologia , Sinapses/efeitos dos fármacos , Sinapses/metabolismo , Trombospondinas/genética , Fatores de Tempo , Tubulina (Proteína)/metabolismo , Articulação Zigapofisária/efeitos dos fármacos , Ácido gama-Aminobutírico/uso terapêutico
14.
J Biol Chem ; 289(10): 7025-7037, 2014 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-24459143

RESUMO

To investigate a potential mechanism underlying trigeminal nerve injury-induced orofacial hypersensitivity, we used a rat model of chronic constriction injury to the infraorbital nerve (CCI-ION) to study whether CCI-ION caused calcium channel α2δ1 (Cavα2δ1) protein dysregulation in trigeminal ganglia and associated spinal subnucleus caudalis and C1/C2 cervical dorsal spinal cord (Vc/C2). Furthermore, we studied whether this neuroplasticity contributed to spinal neuron sensitization and neuropathic pain states. CCI-ION caused orofacial hypersensitivity that correlated with Cavα2δ1 up-regulation in trigeminal ganglion neurons and Vc/C2. Blocking Cavα2δ1 with gabapentin, a ligand for the Cavα2δ1 proteins, or Cavα2δ1 antisense oligodeoxynucleotides led to a reversal of orofacial hypersensitivity, supporting an important role of Cavα2δ1 in orofacial pain processing. Importantly, increased Cavα2δ1 in Vc/C2 superficial dorsal horn was associated with increased excitatory synaptogenesis and increased frequency, but not the amplitude, of miniature excitatory postsynaptic currents in dorsal horn neurons that could be blocked by gabapentin. Thus, CCI-ION-induced Cavα2δ1 up-regulation may contribute to orofacial neuropathic pain states through abnormal excitatory synapse formation and enhanced presynaptic excitatory neurotransmitter release in Vc/C2.


Assuntos
Canais de Cálcio/metabolismo , Dor Facial/metabolismo , Neuralgia/metabolismo , Gânglio Trigeminal/metabolismo , Traumatismos do Nervo Trigêmeo/complicações , Animais , Canais de Cálcio/genética , Canais de Cálcio Tipo L , Modelos Animais de Doenças , Dor Facial/etiologia , Dor Facial/genética , Masculino , Neuralgia/etiologia , Neuralgia/genética , Ratos , Ratos Sprague-Dawley , Núcleo Inferior Caudal do Nervo Trigêmeo/metabolismo
15.
Curr Biol ; 24(2): 117-123, 2014 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-24388848

RESUMO

BACKGROUND: Current pain management is limited, in particular, with regard to chronic pain. In an attempt to discover novel analgesics, we combined the approach developed to characterize traditional Chinese medicine (TCM), as part of the "herbalome" project, with the reverse pharmacology approach aimed at discovering new endogenous transmitters and hormones. RESULTS: In a plant used for centuries for its analgesic properties, we identify a compound, dehydrocorybulbine (DHCB), that is effective at alleviating thermally induced acute pain. We synthesize DHCB and show that it displays moderate dopamine receptor antagonist activities. By using selective pharmacological compounds and dopamine receptor knockout (KO) mice, we show that DHCB antinociceptive effect is primarily due to its interaction with D2 receptors, at least at low doses. We further show that DHCB is effective against inflammatory pain and injury-induced neuropathic pain and furthermore causes no antinociceptive tolerance. CONCLUSIONS: Our study casts DHCB as a different type of analgesic compound and as a promising lead in pain management.


Assuntos
Alcaloides/farmacologia , Analgésicos/farmacologia , Corydalis/química , Isoquinolinas/farmacologia , Medicina Tradicional Chinesa , Dor/tratamento farmacológico , Alcaloides/química , Alcaloides/isolamento & purificação , Analgésicos/química , Analgésicos/isolamento & purificação , Animais , Células HEK293 , Humanos , Inflamação/complicações , Isoquinolinas/química , Isoquinolinas/isolamento & purificação , Masculino , Camundongos , Dor/etiologia
16.
J Neurosci ; 32(26): 8977-87, 2012 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-22745497

RESUMO

Neuropathic pain is a common cause of pain after nerve injury, but its molecular basis is poorly understood. In a post-gene chip microarray effort to identify new target genes contributing to neuropathic pain development, we report here the characterization of a novel neuropathic pain contributor, thrombospondin-4 (TSP4), using a neuropathic pain model of spinal nerve ligation injury. TSP4 is mainly expressed in astrocytes and significantly upregulated in the injury side of dorsal spinal cord that correlates with the development of neuropathic pain states. TSP4 blockade by intrathecal antibodies, antisense oligodeoxynucleotides, or inactivation of the TSP4 gene reverses or prevents behavioral hypersensitivities. Intrathecal injection of TSP4 protein into naive rats is sufficient to enhance the frequency of EPSCs in spinal dorsal horn neurons, suggesting an increased excitatory presynaptic input, and to cause similar behavioral hypersensitivities. Together, these findings support that injury-induced spinal TSP4 may contribute to spinal presynaptic hypersensitivity and neuropathic pain states. Development of TSP4 antagonists has the therapeutic potential for target-specific neuropathic pain management.


Assuntos
Neuralgia/metabolismo , Limiar da Dor/fisiologia , Medula Espinal/metabolismo , Trombospondinas/metabolismo , Regulação para Cima/fisiologia , 6-Ciano-7-nitroquinoxalina-2,3-diona , Análise de Variância , Animais , Anticorpos/uso terapêutico , Modelos Animais de Doenças , Antagonistas de Aminoácidos Excitatórios/farmacologia , Proteínas de Fluorescência Verde/genética , Humanos , Hiperalgesia/metabolismo , Hiperalgesia/patologia , Técnicas In Vitro , Potenciais Pós-Sinápticos Inibidores/efeitos dos fármacos , Potenciais Pós-Sinápticos Inibidores/genética , Injeções Espinhais , Masculino , Camundongos , Camundongos Transgênicos , Atividade Motora/efeitos dos fármacos , Neuralgia/tratamento farmacológico , Neuralgia/etiologia , Oligodesoxirribonucleotídeos Antissenso/administração & dosagem , Medição da Dor , Limiar da Dor/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Bloqueadores dos Canais de Sódio/farmacologia , Medula Espinal/efeitos dos fármacos , Medula Espinal/patologia , Medula Espinal/fisiopatologia , Nervos Espinhais/lesões , Tetrodotoxina/farmacologia , Trombospondinas/deficiência , Trombospondinas/genética , Regulação para Cima/efeitos dos fármacos , Valina/análogos & derivados , Valina/farmacologia
17.
Methods Mol Biol ; 851: 1-8, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22351079

RESUMO

After the publication of the First Edition of this book in the series of Methods in Molecular Medicine (volume 99 in the series) in 2004, pain research continues its rapid acceleration until 2009, during which it experienced a plateau of growth that likely resulted from the economic downturn started in 2008 (Fig. 1.1). This rapid growth in pain research could be the driving force for an impressive 66% increase in new randomized, double-blind, placebo-control trials for neuropathic pain medications in the past 5 years compared with the last four decades. Unfortunately, little improvement in pain medications has been obtained [1] due to primarily our limited understanding of mechanisms mediating different pain states, especially that for chronic pain. It is highly possible that the growth in pain research will continue for decades to come due to three main reasons. First, there is an urgent need for more efficacious and safer pain medications that are necessary for better and individualized pain management. The increase in life expectancy of the general population and patients due to improvements in quality of health care and medicine is likely to increase the demand for better pain medications for improving the quality of daily life of those living with pain. It is estimated that the continuous increase in percentage of patients suffering from chronic pain (pain conditions lasting more than 6 months) arranges from 11 to 47% between 40 and 75 years of age [2], which will inevitably and continually increase the demand for better pain medications. Second, the cost of pain conditions to our society is high, estimated $55 billion per year in loss of productivity from full-time workers alone [3], so better pain management can significantly help economic growth and stability. Third, the swift advancement in technologies and our better understanding of sensory circuitries and pain pathways serves as a driving force for timely drug discovery research and development at an unprecedented pace to meet the demand for better pain medications.


Assuntos
Dor Crônica/metabolismo , Manejo da Dor/tendências , Dor Crônica/tratamento farmacológico , Descoberta de Drogas , Humanos , Manejo da Dor/métodos , Projetos de Pesquisa
18.
Methods Mol Biol ; 851: 195-203, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22351092

RESUMO

Spinal cord injury-induced pain is a common clinical problem affecting adversely the quality of daily lives of spinal cord injured patients. Management with current pain medications can only lead to partial pain relief in some spinal cord injured patients, which is usually associated with unfavorable side effects. Development of specific medications for spinal cord injury-induced pain states relies on identification of new targets and/or pathways that contribute to chronic pain development post injury. We describe here the generation of a spinal cord contusion injury model that mimics the etiology and phenotypes of chronic pain states in spinal cord injured patients. Therefore, this model can be a useful tool for studying spinal cord injury mechanisms, functional recovery, research, and development of new medications for better functional and symptomatic improvements, including pain management.


Assuntos
Dor Crônica/etiologia , Modelos Animais de Doenças , Traumatismos da Medula Espinal/complicações , Medula Espinal/fisiopatologia , Animais , Contusões/complicações , Contusões/fisiopatologia , Camundongos , Manejo da Dor/métodos , Ratos , Traumatismos da Medula Espinal/fisiopatologia
19.
Methods Mol Biol ; 851: 275-84, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22351099

RESUMO

The spinal nerve ligation model of neuropathic pain in rats, as originally described by Kim and Chung (Pain 50:355-363, 1992), provides an excellent venue to study the antinociception and modulation effects of pulsed radiofrequency (PRF) current in pain processing. We describe the procedure of application of PRF current near the exposed L5 dorsal root ganglion (DRG) in rats with L5 spinal nerve ligation injury-induced behavioral hypersensitivity. This method employs the direct visualization of the L5 DRG, allowing for confirmation of the location of the PRF probe adjacent to the DRG.


Assuntos
Gânglios Espinais/efeitos da radiação , Neuralgia/etiologia , Traumatismos dos Nervos Periféricos/complicações , Animais , Modelos Animais de Doenças , Gânglios Espinais/patologia , Gânglios Espinais/fisiopatologia , Neuralgia/patologia , Medição da Dor , Traumatismos dos Nervos Periféricos/patologia , Ondas de Rádio , Ratos , Ratos Sprague-Dawley
20.
Anesth Analg ; 113(3): 610-6, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21596869

RESUMO

BACKGROUND: Application of pulsed radiofrequency (PRF) currents to the dorsal root ganglia (DRG) has been reported to produce relief from certain pain states without causing thermal ablation. In this study, we examined the direct correlation between PRF application to DRG associated with spinal nerve injury and reversal of injury-induced behavioral hypersensitivity in a rat neuropathic pain model. METHODS: Neuropathic lesioning was performed via left L5 spinal nerve ligation on male adult Sprague-Dawley rats. Once the injured rats had developed tactile allodynia, one group was then assigned to PRF treatment of the L5 DRG and another group was assigned to the sham treatment to the DRG. Behavioral testing was performed on both the control and treated paws using the von Frey filament test before the surgery and at indicated days. The resulting data were analyzed using a linear mixed model to assess the overall difference between the treatment groups and the overall difference among the study days. Cohen's d statistic was computed from paired difference-from-baseline scores for each of the 14 study days after treatment and these measures of effect size were then used to descriptively compare the recovery patterns over time for each study group. RESULTS: Spinal nerve injury resulted in the development of behavioral hypersensitivity to von Frey filament stimulation (allodynia) in the hindpaw of the left (injury) side. Mixed linear modeling showed a significant difference between the treatment groups (P = 0.0079) and a significant change of paw withdrawal threshold means over time (P = 0.0006) for all 12 animals. Evaluation of Cohen's d (effect size) revealed that the PRF-treated animals exhibited better recovery and recorded larger effect sizes than the sham-treated animals on 10 of the 14 post-PRF treatment days and exhibited moderate-to-strong effects posttreatment at days 8 to 10 and at and beyond day 32. CONCLUSIONS: Findings from this study support that PRF of the DRG causes reversal of nerve injury (spinal nerve ligation)-induced tactile allodynia in rats. This allodynia reversal indicates that nonablative PRF acting via modulation of the DRG can speed recovery in nerve injury-induced pain.


Assuntos
Terapia por Estimulação Elétrica , Gânglios Espinais/fisiopatologia , Hiperalgesia/terapia , Neuralgia/terapia , Nervos Espinhais/fisiopatologia , Análise de Variância , Animais , Comportamento Animal , Modelos Animais de Doenças , Hiperalgesia/etiologia , Hiperalgesia/fisiopatologia , Hiperalgesia/psicologia , Ligadura , Modelos Lineares , Masculino , Neuralgia/etiologia , Neuralgia/fisiopatologia , Neuralgia/psicologia , Medição da Dor , Limiar da Dor , Ratos , Ratos Sprague-Dawley , Nervos Espinhais/cirurgia , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...