Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
FASEB J ; 35(10): e21869, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34469026

RESUMO

The leucine-rich repeat-containing family 8 member A (LRRC8A) is an essential subunit of the volume-regulated anion channel (VRAC). VRAC is critical for cell volume control, but its broader physiological functions remain under investigation. Recent studies in the field indicate that Lrrc8a disruption in the brain astrocytes reduces neuronal excitability, impairs synaptic plasticity and memory, and protects against cerebral ischemia. In the present work, we generated brain-wide conditional LRRC8A knockout mice (LRRC8A bKO) using NestinCre -driven Lrrc8aflox/flox excision in neurons, astrocytes, and oligodendroglia. LRRC8A bKO animals were born close to the expected Mendelian ratio and developed without overt histological abnormalities, but, surprisingly, all died between 5 and 9 weeks of age with a seizure phenotype, which was confirmed by video and EEG recordings. Brain slice electrophysiology detected changes in the excitability of pyramidal cells and modified GABAergic inputs in the hippocampal CA1 region of LRRC8A bKO. LRRC8A-null hippocampi showed increased immunoreactivity of the astrocytic marker GFAP, indicating reactive astrogliosis. We also found decreased whole-brain protein levels of the GABA transporter GAT-1, the glutamate transporter GLT-1, and the astrocytic enzyme glutamine synthetase. Complementary HPLC assays identified reduction in the tissue levels of the glutamate and GABA precursor glutamine. Together, these findings suggest that VRAC provides vital control of brain excitability in mouse adolescence. VRAC deletion leads to a lethal phenotype involving progressive astrogliosis and dysregulation of astrocytic uptake and supply of amino acid neurotransmitters and their precursors.


Assuntos
Astrócitos/patologia , Gliose/mortalidade , Ácido Glutâmico/metabolismo , Proteínas de Membrana/fisiologia , Convulsões/mortalidade , Animais , Astrócitos/metabolismo , Região CA1 Hipocampal/metabolismo , Região CA1 Hipocampal/patologia , Feminino , Gliose/etiologia , Gliose/patologia , Transporte de Íons , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Convulsões/etiologia , Convulsões/patologia
2.
Int J Hyperthermia ; 39(1): 1283-1293, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36162814

RESUMO

BACKGROUND: In stereotactic radiosurgery, isodose lines must be considered to determine how surrounding tissue is affected. In thermal ablative therapy, such as laser interstitial thermal therapy (LITT), transcranial MR-guided focused ultrasound (tcMRgFUS), and needle-based therapeutic ultrasound (NBTU), how the surrounding area is affected has not been well studied. OBJECTIVE: We aimed to quantify the transition zone surrounding the ablation core created by magnetic resonance-guided robotically-assisted (MRgRA) delivery of NBTU using multi-slice volumetric 2-D magnetic resonance thermal imaging (MRTI) and subsequent characterization of the resultant tissue damage using histopathologic analysis. METHODS: Four swine underwent MRgRA NBTU using varying duration and wattage for treatment delivery. Serial MRI images were obtained, and the most representative were overlaid with isodose lines and compared to brain tissue acquired postmortem which underwent histopathologic analysis. These results were also compared to predicted volumes using a finite element analysis model. Contralateral brain tissue was used for control data. RESULTS: Intraoperative MRTI thermal isodose contours were characterized and comprehensively mapped to post-operative MRI images and qualitatively compared with histological tissue sections postmortem. NBTU 360° ablations induced smaller lesion volumes (33.19 mm3; 120 s, 3 W; 30.05 mm3, 180 s, 4 W) versus 180° ablations (77.20 mm3, 120 s, 3 W; 109.29 mm3; 180 s; 4 W). MRTI/MRI overlay demonstrated the lesion within the proximal isodose lines. The ablation-zone was characterized by dense macrophage infiltration and glial/neuronal loss as demonstrated by glial fibrillary acidic protein (GFAP) and neurofilament (NF) absence and avid CD163 staining. The transition-zone between lesion and normal brain demonstrated decreased macrophage infiltration and measured ∼345 microns (n - 3). We did not detect overt hemorrhages or signs of edema in the adjacent spared tissue. CONCLUSION: We successfully performed MRgRA NBTU ablation in swine and demonstrated minimal histologic changes extended past the ablation-zone. The lesion was characterized by macrophage infiltration and glial/neuronal loss which decreased through the transition-zone.


Assuntos
Encéfalo , Terapia por Ultrassom , Animais , Encéfalo/diagnóstico por imagem , Encéfalo/cirurgia , Proteína Glial Fibrilar Ácida , Fígado , Imageamento por Ressonância Magnética/métodos , Suínos
3.
Int J Hyperthermia ; 38(1): 907-915, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34148489

RESUMO

BACKGROUND: High-intensity focused ultrasound (HIFU) serves as a noninvasive stereotactic system for the ablation of brain metastases; however, treatments are limited to simple geometries and energy delivery is limited by the high acoustic attenuation of the calvarium. Minimally-invasive magnetic resonance-guided robotically-assisted (MRgRA) needle-based therapeutic ultrasound (NBTU) using multislice volumetric 2-D magnetic resonance thermal imaging (MRTI) overcomes these limitations and has potential to produce less collateral tissue damage than current methods. OBJECTIVE: To correlate multislice volumetric 2-D MRTI volumes with histologically confirmed regions of tissue damage in MRgRA NBTU. METHODS: Seven swine underwent a total of 8 frontal MRgRA NBTU lesions. MRTI ablation volumes were compared to histologic tissue damage on brain sections stained with 2,3,5-triphenyltetrazolium chloride (TTC). Bland-Altman analyses and correlation trends were used to compare MRTI and TTC ablation volumes. RESULTS: Data from the initial and third swine's ablations were excluded due to sub-optimal tissue staining. For the remaining ablations (n = 6), the limits of agreement between the MRTI and histologic volumes ranged from -0.149 cm3 to 0.252 cm3 with a mean difference of 0.052 ± 0.042 cm3 (11.1%). There was a high correlation between the MRTI and histology volumes (r2 = 0.831) with a strong linear relationship (r = 0.868). CONCLUSION: We used a volumetric MRTI technique to accurately track thermal changes during MRgRA NBTU in preparation for human trials. Improved volumetric coverage with MRTI enhanced our delivery of therapy and has far-reaching implications for focused ultrasound in the broader clinical setting.


Assuntos
Neoplasias Encefálicas , Ablação por Ultrassom Focalizado de Alta Intensidade , Terapia por Ultrassom , Animais , Encéfalo/diagnóstico por imagem , Encéfalo/cirurgia , Neoplasias Encefálicas/diagnóstico por imagem , Neoplasias Encefálicas/cirurgia , Imageamento por Ressonância Magnética , Espectroscopia de Ressonância Magnética , Suínos
4.
J Neurosci ; 36(28): 7485-96, 2016 07 13.
Artigo em Inglês | MEDLINE | ID: mdl-27413158

RESUMO

UNLABELLED: The occurrence of recurrent, unprovoked seizures is the hallmark of human epilepsy. Currently, only two-thirds of this patient population has adequate seizure control. New epilepsy models provide the potential for not only understanding the development of spontaneous seizures, but also for testing new strategies to treat this disorder. Here, we characterize a primary generalized seizure model of epilepsy following repeated exposure to the GABAA receptor antagonist, flurothyl, in which mice develop spontaneous seizures that remit within 1 month. In this model, we expose C57BL/6J mice to flurothyl until they experience a generalized seizure. Each of these generalized seizures typically lasts <30 s. We induce one seizure per day for 8 d followed by 24 h video-electroencephalographic recordings. Within 1 d following the last of eight flurothyl-induced seizures, ∼50% of mice have spontaneous seizures. Ninety-five percent of mice tested have seizures within the first week of the recording period. Of the spontaneous seizures recorded, the majority are generalized clonic seizures, with the remaining 7-12% comprising generalized clonic seizures that transition into brainstem seizures. Over the course of an 8 week recording period, spontaneous seizure episodes remit after ∼4 weeks. Overall, the repeated flurothyl paradigm is a model of epileptogenesis with spontaneous seizures that remit. This model provides an additional tool in our armamentarium for understanding the mechanisms underlying epileptogenesis and may provide insights into why spontaneous seizures remit without anticonvulsant treatment. Elucidating these processes could lead to the development of new epilepsy therapeutics. SIGNIFICANCE STATEMENT: Epilepsy is a chronic disorder characterized by the occurrence of recurrent, unprovoked seizures in which the individual seizure-ictal events are self-limiting. Remission of recurrent, unprovoked seizures can be achieved in two-thirds of cases by treatment with anticonvulsant medication, surgical resection, and/or nerve/brain electrode stimulation. However, there are examples in humans of epilepsy with recurrent, unprovoked seizures remitting without any intervention. While elucidating how recurrent, unprovoked seizures develop is critical for understanding epileptogenesis, an understanding of how and why recurrent, unprovoked seizures remit may further our understanding and treatment of epilepsy. Here, we describe a new model of recurrent, unprovoked spontaneous seizures in which the occurrence of spontaneous seizures naturally remits over time without any therapeutic intervention.


Assuntos
Convulsivantes/toxicidade , Flurotila/toxicidade , Convulsões/induzido quimicamente , Análise de Variância , Animais , Anticonvulsivantes/uso terapêutico , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Esquema de Medicação , Eletroencefalografia , Fluoresceínas/metabolismo , Hipocampo/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Convulsões/tratamento farmacológico , Convulsões/patologia , Fatores de Tempo , Gravação em Vídeo
5.
Drug Metab Dispos ; 43(9): 1326-30, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26109562

RESUMO

Cytochrome P450 monooxygenases (P450s), which are well-known drug-metabolizing enzymes, are thought to play a signal transduction role in µ opioid analgesia and may serve as high-affinity (3)H-cimetidine ((3)HCIM) binding sites in the brain. (3)HCIM binding sites may also be related to opioid or nonopioid analgesia. However, of the more than 100 murine P450 enzymes, the specific isoform(s) responsible for either function have not been identified. Presently, three lines of constitutive P450 gene cluster knockout (KO) mice with full-length deletions of 14 Cyp2c, 9 Cyp2d, and 7 Cyp3a genes were studied for deficiencies in (3)HCIM binding and for opioid analgesia. Liver and brain homogenates from all three genotypes showed normal (3)HCIM binding values, indicating that gene products of Cyp2d, Cyp3a, and Cyp2c are not (3)HCIM-binding proteins. Cyp2d KO and Cyp3a KO mice showed normal antinociceptive responses to a moderate systemic dose of morphine (20 mg/kg, s.c.), thereby excluding 16 P450 isoforms as mediators of opioid analgesia. In contrast, Cyp2c KO mice showed a 41% reduction in analgesic responses following systemically (s.c.) administered morphine. However, the significance of brain Cyp2c gene products in opioid analgesia is uncertain because little or no analgesic deficits were noted in Cyp2c KO mice following intracerebroventricular or intrathecalmorphine administration, respectively. These results show that the gene products of Cyp2d and Cyp3a do not contribute to µ opioid analgesia in the central nervous system. A possible role for Cyp2c gene products in opioid analgesia requires further consideration.


Assuntos
Analgésicos Opioides/administração & dosagem , Sistema Enzimático do Citocromo P-450/metabolismo , Isoenzimas/metabolismo , Analgésicos Opioides/metabolismo , Animais , Sistema Enzimático do Citocromo P-450/genética , Isoenzimas/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
6.
ArXiv ; 2024 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-39279835

RESUMO

Intracorporeal needle-based therapeutic ultrasound (NBTU) is a minimally invasive option for intervening in malignant brain tumors, commonly used in thermal ablation procedures. This technique is suitable for both primary and metastatic cancers, utilizing a high-frequency alternating electric field (up to 10 MHz) to excite a piezoelectric transducer. The resulting rapid deformation of the transducer produces an acoustic wave that propagates through tissue, leading to localized high-temperature heating at the target tumor site and inducing rapid cell death. To optimize the design of NBTU transducers for thermal dose delivery during treatment, numerical modeling of the acoustic pressure field generated by the deforming piezoelectric transducer is frequently employed. The bioheat transfer process generated by the input pressure field is used to track the thermal propagation of the applicator over time. Magnetic resonance thermal imaging (MRTI) can be used to experimentally validate these models. Validation results using MRTI demonstrated the feasibility of this model, showing a consistent thermal propagation pattern. However, a thermal damage isodose map is more advantageous for evaluating therapeutic efficacy. To achieve a more accurate simulation based on the actual brain tissue environment, a new finite element method (FEM) simulation with enhanced damage evaluation capabilities was conducted. The results showed that the highest temperature and ablated volume differed between experimental and simulation results by 2.1884°C (3.71%) and 0.0631 cm3 (5.74%), respectively. The lowest Pearson correlation coefficient (PCC) for peak temperature was 0.7117, and the lowest Dice coefficient for the ablated area was 0.7021, indicating a good agreement in accuracy between simulation and experiment.

7.
iScience ; 26(5): 106669, 2023 May 19.
Artigo em Inglês | MEDLINE | ID: mdl-37182109

RESUMO

The ubiquitous volume-regulated anion channels (VRACs) facilitate cell volume control and contribute to many other physiological processes. Treatment with non-specific VRAC blockers or brain-specific deletion of the essential VRAC subunit LRRC8A is highly protective in rodent models of stroke. Here, we tested the widely accepted idea that the harmful effects of VRACs are mediated by release of the excitatory neurotransmitter glutamate. We produced conditional LRRC8A knockout either exclusively in astrocytes or in the majority of brain cells. Genetically modified mice were subjected to an experimental stroke (middle cerebral artery occlusion). The astrocytic LRRC8A knockout yielded no protection. Conversely, the brain-wide LRRC8A deletion strongly reduced cerebral infarction in both heterozygous (Het) and full KO mice. Yet, despite identical protection, Het mice had full swelling-activated glutamate release, whereas KO animals showed its virtual absence. These findings suggest that LRRC8A contributes to ischemic brain injury via a mechanism other than VRAC-mediated glutamate release.

8.
J Neurophysiol ; 108(9): 2393-404, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22956800

RESUMO

Respiratory depression is a therapy-limiting side effect of opioid analgesics, yet our understanding of the brain circuits mediating this potentially lethal outcome remains incomplete. Here we studied the contribution of the rostral ventromedial medulla (RVM), a region long implicated in pain modulation and homeostatic regulation, to opioid-induced respiratory depression. Microinjection of the µ-opioid agonist DAMGO in the RVM of lightly anesthetized rats produced both analgesia and respiratory depression, showing that neurons in this region can modulate breathing. Blocking opioid action in the RVM by microinjecting the opioid antagonist naltrexone reversed the analgesic and respiratory effects of systemically administered morphine, showing that this region plays a role in both the analgesic and respiratory-depressant properties of systemically administered morphine. The distribution of neurons directly inhibited by RVM opioid microinjection was determined with a fluorescent opioid peptide, dermorphin-Alexa 594, and found to be concentrated in and around the RVM. The non-opioid analgesic improgan, like DAMGO, produced antinociception but, unlike DAMGO, stimulated breathing when microinjected into the RVM. Concurrent recording of RVM neurons during improgan microinjection showed that this agent activated RVM ON-cells, OFF-cells, and NEUTRAL-cells. Since opioids are known to activate OFF-cells but suppress ON-cell firing, the differential respiratory response to these two analgesic drugs is best explained by their opposing effects on the activity of RVM ON-cells. These findings show that pain relief can be separated pharmacologically from respiratory depression and identify RVM OFF-cells as important central targets for continued development of potent analgesics with fewer side effects.


Assuntos
Analgésicos Opioides/toxicidade , Bulbo/efeitos dos fármacos , Neurônios/fisiologia , Dor Nociceptiva/fisiopatologia , Insuficiência Respiratória/induzido quimicamente , Analgésicos Opioides/antagonistas & inibidores , Animais , Ala(2)-MePhe(4)-Gly(5)-Encefalina/antagonistas & inibidores , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , Potenciais Evocados/efeitos dos fármacos , Potenciais Evocados/fisiologia , Masculino , Bulbo/citologia , Bulbo/fisiologia , Morfina/antagonistas & inibidores , Morfina/farmacologia , Naltrexona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Neurônios/efeitos dos fármacos , Nociceptividade/efeitos dos fármacos , Nociceptividade/fisiologia , Ratos , Ratos Sprague-Dawley , Insuficiência Respiratória/fisiopatologia
9.
Neurosci Lett ; 757: 135977, 2021 07 13.
Artigo em Inglês | MEDLINE | ID: mdl-34023413

RESUMO

BACKGROUND: Changes in inflammatory cytokine levels contribute to the induction and maintenance of neuropathic pain. We have shown that external low intensity focused ultrasound (liFUS) reduces allodynia in a common peroneal nerve injury (CPNI). Here, we investigate an underlying mechanism of action for this treatment and measure the effect of liFUS on inflammatory markers. METHODS: Male rats were divided into four groups: CPNI/liFUS, CPNI/shamliFUS, shamCPNI/liFUS, and shamCPNI/shamliFUS. Mechanical nociceptive thresholds were measured using Von Frey filaments (VFF) to confirm the absence/presence of allodynia at baseline, after CPNI, and after liFUS. Commercial microarray and ELISA assays were used to assess cytokine expression in the treated L5 dorsal root ganglion (DRG) and dorsal horn (DH) tissue 24 and 72 h after liFUS. RESULTS: VFF thresholds were significantly reduced following CPNI in both groups that received the injury (p < 0.001). After liFUS, only the CPNI/liFUS cohort showed a significant increase in mechanical thresholds (p < 0.001). CPNI significantly increased TNFa, IL6, CNTF, IL1b (p < 0.05 for all) levels in the DRG and DH, compared to baseline, consistent with previous work in sciatic nerve injury. LiFUS in CPNI rats resulted in a decrease in these cytokines in DRG 72 h post-therapy (TNFa, IL6, CNTF and IL1b, p < 0.001). In the DH, IL1b, CNTF, and TNFa (p < 0.05 for all) decreased 72 h after liFUS. CONCLUSION: We have demonstrated that liFUS modifies inflammatory cytokines in both DRG and DH in CPNI rats. These data provide evidence that liFUS, reverses the allodynic phenotype, in part, by altering inflammatory cytokine pathways.


Assuntos
Hiperalgesia/terapia , Neuralgia/terapia , Traumatismos dos Nervos Periféricos/complicações , Terapia por Ultrassom/métodos , Animais , Citocinas/metabolismo , Modelos Animais de Doenças , Gânglios Espinais/imunologia , Gânglios Espinais/metabolismo , Humanos , Hiperalgesia/diagnóstico , Hiperalgesia/imunologia , Masculino , Neuralgia/diagnóstico , Neuralgia/imunologia , Traumatismos dos Nervos Periféricos/imunologia , Traumatismos dos Nervos Periféricos/terapia , Nervo Fibular/lesões , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/imunologia , Transdução de Sinais/efeitos da radiação , Corno Dorsal da Medula Espinal/imunologia , Corno Dorsal da Medula Espinal/metabolismo , Ondas Ultrassônicas
10.
J Neurosurg ; : 1-8, 2021 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-33862597

RESUMO

OBJECTIVE: The authors' laboratory has previously demonstrated beneficial effects of noninvasive low intensity focused ultrasound (liFUS), targeted at the dorsal root ganglion (DRG), for reducing allodynia in rodent neuropathic pain models. However, in rats the DRG is 5 mm below the skin when approached laterally, while in humans the DRG is typically 5-8 cm deep. Here, using a modified liFUS probe, the authors demonstrated the feasibility of using external liFUS for modulation of antinociceptive responses in neuropathic swine. METHODS: Two cohorts of swine underwent a common peroneal nerve injury (CPNI) to induce neuropathic pain. In the first cohort, pigs (14 kg) were iteratively tested to determine treatment parameters. liFUS penetration to the L5 DRG was verified by using a thermocouple to monitor tissue temperature changes and by measuring nerve conduction velocity (NCV) at the corresponding common peroneal nerve (CPN). Pain behaviors were monitored before and after treatment. DRG was evaluated for tissue damage postmortem. Based on data from the first cohort, a treatment algorithm was developed, parameter predictions were verified, and neuropathic pain was significantly modified in a second cohort of larger swine (20 kg). RESULTS: The authors performed a dose-response curve analysis in 14-kg CPNI swine. Specifically, after confirming that the liFUS probe could reach 5 cm in ex vivo tissue experiments, the authors tested liFUS in 14-kg CPNI swine. The mean ± SEM DRG depth was 3.79 ± 0.09 cm in this initial cohort. The parameters were determined and then extrapolated to larger animals (20 kg), and predictions were verified. Tissue temperature elevations at the treatment site did not exceed 2°C, and the expected increases in the CPN NCV were observed. liFUS treatment eliminated pain guarding in all animals for the duration of follow-up (up to 1 month) and improved allodynia for 5 days postprocedure. No evidence of histological damage was seen using Fluoro-Jade and H&E staining. CONCLUSIONS: The results demonstrate that a 5-cm depth can be reached with external liFUS and alters pain behavior and allodynia in a large-animal model of neuropathic pain.

11.
J Neurosurg ; : 1-8, 2021 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-33862596

RESUMO

OBJECTIVE: To date, muscular and bone pain have been studied in domestic swine models, but the only neuropathic pain model described in swine is a mixed neuritis model. Common peroneal nerve injury (CPNI) neuropathic pain models have been utilized in both mice and rats. METHODS: The authors developed a swine surgical CPNI model of neuropathic pain. Behavioral outcomes were validated with von Frey filament testing, thermal sensitivity assessments, and social and motor scoring. Demyelination of the nerve was confirmed through standard histological assessment. The contralateral nerve served as the control. RESULTS: CPNI induced mechanical and thermal allodynia (p < 0.001 [n = 10] and p < 0.05 [n = 4], respectively) and increased pain behavior, i.e., guarding of the painful leg (n = 12). Myelin protein zero (P0) staining revealed demyelination of the ligated nerve upstream of the ligation site. CONCLUSIONS: In a neuropathic pain model in domestic swine, the authors demonstrated that CPNI induces demyelination of the common peroneal nerve, which the authors hypothesize is responsible for the resulting allodynic pain behavior. As the anatomical features of domestic swine resemble those of humans more closely than previously used rat and mouse models, utilizing this swine model, which is to the authors' knowledge the first of its kind, will aid in the translation of experimental treatments to clinical trials.

12.
J Neurophysiol ; 104(6): 3222-30, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20926616

RESUMO

Many analgesic drugs, including µ-opioids, cannabinoids, and the novel nonopioid analgesic improgan, produce antinociception by actions in the rostral ventromedial medulla (RVM). There they activate pain-inhibiting neurons, termed "OFF-cells," defined by a nociceptive reflex-related pause in activity. Based on recent functional evidence that neuronal P450 epoxygenases are important for the central antinociceptive actions of morphine and improgan, we explored the convergence of opioid and nonopioid analgesic drug actions in RVM by studying the effects of the P450 epoxygenase inhibitor CC12 on the analgesic drug-induced activation of these OFF-cells and on behavioral antinociception. In rats lightly anesthetized with isoflurane, we recorded the effects of intraventricular morphine and improgan, with and without CC12 pretreatment, on tail flick latency and activity of identified RVM neurons: OFF-cells, ON-cells (pronociceptive neurons), and neutral cells (unresponsive to analgesic drugs). CC12 pretreatment preserved reflex-related changes in OFF-cell firing and blocked the analgesic actions of both drugs, without interfering with the increase in spontaneous firing induced by improgan or morphine. CC12 blocked suppression of evoked ON-cell firing by improgan, but not morphine. CC12 pretreatment had no effect by itself on RVM neurons or behavior. These data show that the epoxygenase inhibitor CC12 works downstream from receptors for both µ-opioid and improgan, at the inhibitory input mediating the OFF-cell pause. This circuit-level analysis thus provides a cellular basis for the convergence of opioid and nonopioid analgesic actions in the RVM. A presynaptic P450 epoxygenase may therefore be an important target for development of clinically useful nonopioid analgesic drugs.


Assuntos
Analgésicos/antagonistas & inibidores , Cimetidina/análogos & derivados , Imidazóis/farmacologia , Bulbo/efeitos dos fármacos , Morfina/antagonistas & inibidores , Percepção da Dor/efeitos dos fármacos , Receptores Opioides mu/efeitos dos fármacos , Sulfetos/farmacologia , Potenciais de Ação/efeitos dos fármacos , Animais , Cimetidina/antagonistas & inibidores , Citocromo P-450 CYP2J2 , Inibidores das Enzimas do Citocromo P-450 , Sistema Enzimático do Citocromo P-450 , Masculino , Bulbo/citologia , Bulbo/fisiologia , Modelos Neurológicos , Percepção da Dor/fisiologia , Ratos , Ratos Sprague-Dawley , Tempo de Reação/efeitos dos fármacos , Tempo de Reação/fisiologia , Receptor CB1 de Canabinoide/fisiologia , Receptores Opioides mu/fisiologia , Receptores Pré-Sinápticos/efeitos dos fármacos , Receptores Pré-Sinápticos/fisiologia , Transdução de Sinais/efeitos dos fármacos , Ácido gama-Aminobutírico/fisiologia
13.
Neuroscience ; 430: 82-93, 2020 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-32032575

RESUMO

Previously, we showed internal low intensity focused ultrasound (liFUS) improves nociceptive thresholds in rats with vincristine-induced neuropathy (VIN) for 48-h post-treatment. Here, we perform more rigorous behavioral testing with the internal device and introduce external liFUS treatment. Behavioral testing confirmed VIN (Von Frey fibers, VFF; hot plate, HPT; locomotion, OFT). This was followed by internal or external liFUS treatment (2.5 W or 8 W, for 3 min, respectively) to the left L5 dorsal root ganglia (DRG). A thermocouple placed at the DRG documented temperature changes during treatment, to confirm the modulatory nature of our treatment. Behavioral testing was performed pre-liFUS, and for five consecutive days post-liFUS. Groups included: (1) VIN/liFUS, (2) saline/liFUS, (3) VIN/sham liFUS, and (4) saline/sham liFUS. Significant improvements in mechanical (VFF) and thermal (HPT) nociceptive thresholds were seen in the VIN/liFUS group following both internal and external treatment. Hematoxylin and Eosin, and Fluorojade staining showed no histological damage to the DRG. Internal liFUS treatment produced a mean temperature rise of 3.21 ±â€¯0.30 °C, whereas external liFUS resulted in a mean temperature rise of 1.78 °C ±â€¯0.21 °C. We demonstrate that, in a VIN rat model, external liFUS treatment of the L5 DRG significantly reduces nociceptive sensitivity thresholds without causing tissue damage.


Assuntos
Hiperalgesia , Neuralgia , Animais , Gânglios Espinais , Hiperalgesia/induzido quimicamente , Neuralgia/induzido quimicamente , Neuralgia/tratamento farmacológico , Ratos , Ratos Sprague-Dawley , Vincristina
14.
Neuroscience ; 429: 264-272, 2020 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-32001366

RESUMO

Non-invasive treatment methods for neuropathic pain are lacking. We assess how modulatory low intensity focused ultrasound (liFUS) at the L5 dorsal root ganglion (DRG) affects behavioral responses and sensory nerve action potentials (SNAPs) in a common peroneal nerve injury (CPNI) model. Rats were assessed for mechanical and thermal responses using Von Frey filaments (VFF) and the hot plate test (HPT) following CPNI surgery. Testing was repeated 24 h after liFUS treatment. Significant increases in mechanical and thermal sensory thresholds were seen post-liFUS treatment, indicating a reduction in sensitivity to pain (p < 0.0001, p = 0.02, respectively). Animals who received CPNI surgery had significant increases in SNAP latencies compared to sham CPNI surgery animals (p = 0.0003) before liFUS treatment. LiFUS induced significant reductions in SNAP latency in both CPNI liFUS and sham CPNI liFUS cohorts, for up to 35 min post treatment. No changes were seen in SNAP amplitude and there was no evidence of neuronal degeneration 24 h after liFUS treatment, showing that liFUS did not damage the tissue being modulated. This is the first in vivo study of the impact of liFUS on peripheral nerve electrophysiology in a model of chronic pain. This study demonstrates the effects of liFUS on peripheral nerve electrophysiology in vivo. We found that external liFUS treatment results in transient decreased latency in common peroneal nerve (CPN) sensory nerve action potentials (SNAPs) with no change in signal amplitude.


Assuntos
Traumatismos dos Nervos Periféricos , Nervo Fibular , Animais , Gânglios Espinais , Hiperalgesia , Ratos , Ratos Sprague-Dawley , Roedores
15.
Drug Metab Dispos ; 36(3): 614-21, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18094038

RESUMO

[(3)H]Cimetidine (3HCIM) specifically binds to an unidentified site in the rat brain. Because recently described ligands for this site have pharmacological activity, 3HCIM binding was characterized. 3HCIM binding was saturable, heat-labile, and distinct from the histamine H(2) receptor. To test the hypothesis that 3HCIM binds to a cytochrome P450 (P450), the effects of nonselective and isoform-selective P450 inhibitors were studied. The heme inhibitor KCN and the nonselective P450 inhibitor metyrapone both produced complete, concentration-dependent inhibition of 3HCIM binding (K(i) = 1.3 mM and 11.9 muM, respectively). Binding was largely unaffected by inhibitors of CYP1A2, 2B6, 2C8, 2C9, 2D6, 2E1, and 19A1 but was eliminated by inhibitors of CYP2C19 (tranylcypromine) and CYP3A4 (ketoconazole). Synthesis and testing of CC11 [4(5)-(benzylthiomethyl)-1H-imidazole] and CC12 [4(5)-((4-iodobenzyl)-thiomethyl)-1H-imidazole] confirmed both drugs to be high-affinity inhibitors of 3HCIM binding. On recombinant human P450s, CC12 was a potent inhibitor of CYP2B6 (IC(50) = 11.7 nM), CYP2C19 (51.4 nM), and CYP19A1 (140.7 nM) and had a range of activities (100-494 nM) on nine other isoforms. Although the 3HCIM binding site pharmacologically resembles some P450s, eight recombinant human P450s and three recombinant rat P450s did not exhibit 3HCIM binding. Inhibition by KCN and metyrapone suggests that 3HCIM binds to a heme-containing brain protein (possibly a P450). However, results with selective P450 inhibitors, recombinant P450 isoforms, and a P450 antibody did not identify a 3HCIM-binding P450 isoform. Finally, CC12 is a new, potent inhibitor of CYP2B6 and CYP2C19 that may be a valuable tool for P450 research.


Assuntos
Encéfalo/metabolismo , Cimetidina/metabolismo , Sistema Enzimático do Citocromo P-450/metabolismo , Animais , Sítios de Ligação , Ligação Competitiva , Cimetidina/farmacologia , Inibidores das Enzimas do Citocromo P-450 , Inibidores Enzimáticos/metabolismo , Inibidores Enzimáticos/farmacologia , Antagonistas dos Receptores H2 da Histamina/metabolismo , Antagonistas dos Receptores H2 da Histamina/farmacologia , Isoenzimas , Cinética , Ligantes , Ligação Proteica , Ratos , Ratos Sprague-Dawley , Trítio
16.
J Neurosci Methods ; 294: 1-6, 2018 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-29061345

RESUMO

BACKGROUND: Morphometric analyses of biological features have become increasingly common in recent years with such analyses being subject to a large degree of observer bias, variability, and time consumption. While commercial software packages exist to perform these analyses, they are expensive, require extensive user training, and are usually dependent on the observer tracing the morphology. NEW METHOD: To address these issues, we have developed a broadly applicable, no-cost ImageJ plugin we call 'BranchAnalysis2D/3D', to perform morphometric analyses of structures with branching morphologies, such as neuronal dendritic spines, vascular morphology, and primary cilia. RESULTS: Our BranchAnalysis2D/3D algorithm allows for rapid quantification of the length and thickness of branching morphologies, independent of user tracing, in both 2D and 3D data sets. COMPARISON WITH EXISTING METHODS: We validated the performance of BranchAnalysis2D/3D against pre-existing software packages using trained human observers and images from brain and retina. We found that the BranchAnalysis2D/3D algorithm outputs results similar to available software (i.e., Metamorph, AngioTool, Neurolucida), while allowing faster analysis times and unbiased quantification. CONCLUSIONS: BranchAnalysis2D/3D allows inexperienced observers to output results like a trained observer but more efficiently, thereby increasing the consistency, speed, and reliability of morphometric analyses.


Assuntos
Encéfalo/citologia , Imageamento Tridimensional/métodos , Microscopia Confocal/métodos , Neurônios/citologia , Software , Algoritmos , Animais , Camundongos , Variações Dependentes do Observador , Reprodutibilidade dos Testes , Retina/anatomia & histologia
17.
Neuropharmacology ; 52(5): 1244-55, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17336343

RESUMO

Improgan, a chemical congener of cimetidine, is a highly effective non-opioid analgesic when injected into the CNS. Despite extensive characterization, neither the improgan receptor, nor a pharmacological antagonist of improgan has been previously described. Presently, the specific binding of [(3)H]cimetidine (3HCIM) in brain fractions was used to discover 4(5)-((4-iodobenzyl)thiomethyl)-1H-imidazole, which behaved in vivo as the first improgan antagonist. The synthesis and pharmacological properties of this drug (named CC12) are described herein. In rats, CC12 (50-500nmol, i.c.v.) produced dose-dependent inhibition of improgan (200-400nmol) antinociception on the tail flick and hot plate tests. When given alone to rats, CC12 had no effects on nociceptive latencies, or on other observable behavioral or motor functions. Maximal inhibitory effects of CC12 (500nmol) were fully surmounted with a large i.c.v. dose of improgan (800nmol), demonstrating competitive antagonism. In mice, CC12 (200-400nmol, i.c.v.) behaved as a partial agonist, producing incomplete improgan antagonism, but also limited antinociception when given alone. Radioligand binding, receptor autoradiography, and electrophysiology experiments showed that CC12's antagonist properties are not explained by activity at 25 sites relevant to analgesia, including known receptors for cannabinoids, opioids or histamine. The use of CC12 as an improgan antagonist will facilitate the characterization of improgan analgesia. Furthermore, because CC12 was also found presently to inhibit opioid and cannabinoid antinociception, it is suggested that this drug modifies a biochemical mechanism shared by several classes of analgesics. Elucidation of this mechanism will enhance understanding of the biochemistry of pain relief.


Assuntos
Cimetidina/análogos & derivados , Antagonistas dos Receptores H2 da Histamina/metabolismo , Imidazóis/farmacologia , Receptores Histamínicos H2/efeitos dos fármacos , Sulfetos/farmacologia , Analgésicos Opioides/farmacologia , Animais , Autorradiografia , Benzoxazinas/farmacologia , Sítios de Ligação/efeitos dos fármacos , Cimetidina/antagonistas & inibidores , Cimetidina/metabolismo , Cimetidina/farmacologia , Relação Dose-Resposta a Droga , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , Guanosina 5'-O-(3-Tiotrifosfato)/farmacologia , Histamina/farmacologia , Imidazóis/síntese química , Indicadores e Reagentes , Injeções Intraventriculares , Ligantes , Masculino , Membranas/efeitos dos fármacos , Membranas/metabolismo , Camundongos , Morfolinas/farmacologia , Naloxona/farmacologia , Naftalenos/farmacologia , Antagonistas de Entorpecentes/farmacologia , Medição da Dor/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Sulfetos/síntese química
18.
Brain Res ; 1152: 42-8, 2007 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-17433267

RESUMO

Improgan, a congener of the H(2) antagonist cimetidine, produces non-opioid antinociception which is blocked by the CB(1) antagonist rimonabant, implying a cannabinoid mechanism of action. Since cannabinoids produce hypothermia as well as antinociception in rodents, the present study investigated the pharmacological activity of improgan on core body temperature and nociceptive (tail flick) responses. Improgan (60, 100 and 140 microg, intraventricular [ivt]) elicited significant decreases in core temperature 3-30 min following injection with a maximal hypothermic effect of -1.3 degrees C. Pretreatment with rimonabant (50 microg, ivt) produced a statistically significant but incomplete (29-42%) antagonism of improgan hypothermia. In control experiments, the CB(1) agonist CP-55,940 (37.9 microg, ivt) induced significant decreases in core temperature (-1.8 degrees C) 3-30 min following injection. However, unlike the case with improgan, pretreatment with rimonabant completely blocked CP-55,940 hypothermia. Furthermore, CP-55,940 and improgan elicited maximal antinociception over the same time course and dose ranges, and both effects were attenuated by rimonabant. These results show that, like cannabinoid agonists in the rat, improgan produces antinociception and hypothermia which is blocked by a CB(1) antagonist. Unlike cannabinoid agonists, however, improgan does not produce locomotor inhibition at antinociceptive doses. Additional experiments were performed to determine the effect of CC12, a recently discovered improgan antagonist which lacks affinity at CB(1) receptors. Pretreatment with CC12 (183 microg, ivt) produced complete inhibition of both the antinociception and the hypothermia produced by improgan, suggesting the possible role of an unknown improgan receptor in both of these effects.


Assuntos
Analgésicos/farmacologia , Temperatura Corporal/efeitos dos fármacos , Cimetidina/análogos & derivados , Limiar da Dor/efeitos dos fármacos , Receptor CB1 de Canabinoide/fisiologia , Animais , Cimetidina/antagonistas & inibidores , Cimetidina/farmacologia , Imidazóis/farmacologia , Masculino , Piperidinas/farmacologia , Pirazóis/farmacologia , Ratos , Ratos Sprague-Dawley , Receptor CB1 de Canabinoide/antagonistas & inibidores , Rimonabanto , Sulfetos/farmacologia
19.
J Pain ; 8(11): 850-60, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17644043

RESUMO

UNLABELLED: Improgan is a congener of the H(2) antagonist cimetidine, which produces potent antinociception. Because a) the mechanism of action of improgan remains unknown and b) this drug may indirectly activate cannabinoid CB(1) receptors, the effects of the CB(1) antagonist/inverse agonist rimonabant (SR141716A) and 3 congeners with varying CB(1) potencies were studied on improgan antinociception after intracerebroventricular (icv) dosing in rats. Consistent with blockade of brain CB(1) receptors, rimonabant (K(d) = 0.23 nM), and O-1691 (K(d) = 0.22 nM) inhibited improgan antinociception by 48% and 70% after icv doses of 43 nmol and 25 nmol, respectively. However, 2 other derivatives with much lower CB(1) affinity (O-1876, K(d) = 139 nM and O-848, K(d) = 352 nM) unexpectedly blocked improgan antinociception by 65% and 50% after icv doses of 300 nmol and 30 nmol, respectively. These derivatives have 600-fold to 1500-fold lower CB(1) potencies than that of rimonabant, yet they retained improgan antagonist activity in vivo. In vitro dose-response curves with (35)S-GTPgammaS on CB(1) receptor-containing membranes confirmed the approximate relative potency of the derivatives at the CB(1) receptor. Although antagonism of improgan antinociception by rimonabant has previously implicated a mechanistic role for the CB(1) receptor, current findings with rimonabant congeners suggest that receptors other than, or in addition to CB(1) may participate in the pain-relieving mechanisms activated by this drug. The use of congeners such as O-848, which lack relevant CB(1)-blocking properties, will help to identify these cannabinoid-like, non-CB(1) mechanisms. PERSPECTIVE: This article describes new pharmacological characteristics of improgan, a pain-relieving drug that acts by an unknown mechanism. Improgan may use a marijuana-like (cannabinoid) pain-relieving mechanism, but it is shown presently that the principal cannabinoid receptor in the brain (CB(1)) is not solely responsible for improgan analgesia.


Assuntos
Analgésicos/administração & dosagem , Cimetidina/análogos & derivados , Limiar da Dor/efeitos dos fármacos , Dor/tratamento farmacológico , Receptor CB1 de Canabinoide/fisiologia , Análise de Variância , Animais , Cimetidina/administração & dosagem , Cimetidina/química , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Injeções Intraventriculares/métodos , Masculino , Medição da Dor/métodos , Piperidinas/administração & dosagem , Piperidinas/química , Pirazóis/administração & dosagem , Pirazóis/química , Ratos , Ratos Sprague-Dawley , Tempo de Reação/efeitos dos fármacos , Receptor CB1 de Canabinoide/agonistas , Receptor CB1 de Canabinoide/antagonistas & inibidores , Rimonabanto , Fatores de Tempo
20.
Neuropharmacology ; 51(3): 447-56, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16806300

RESUMO

Improgan is a chemical congener of the H2 antagonist cimetidine which shows the profile of a highly effective analgesic when administered directly into the CNS. Although the improgan receptor is unknown, improgan activates analgesic pathways which are independent of opioids, but may utilize cannabinoid mechanisms. To discover selective, potent, improgan-like drugs, seven compounds chemically related to improgan were synthesized and tested for antinociceptive activity in rats after intracerebroventricular (icv) administration. Among a series of improgan congeners in which the alkyl chain length of improgan ((-CH2)3-) was varied, five compounds showed full agonist antinociceptive activity with potencies greater than that of improgan. VUF5420 (containing (-CH2)4-, EC50 = 86.1 nmol) produced maximal antinociceptive activity after doses which showed no motor impairment or other obvious toxicity, and was 2.3-fold more potent than improgan (EC50 = 199.5 nmol). As found previously with improgan, VUF5420-induced antinociception was unaffected by administration of the opioid antagonist naltrexone, but was inhibited by the CB1 antagonist SR141716A, suggesting a non-opioid, cannabinoid-related analgesic action. However, VUF5420 showed very low affinity (Kd approximately 10 microM) on CB1-receptor activation of 35S-GTPgammaS binding, indicating that this drug does not directly interact with the CB1 receptor in vivo. The present results show that VUF5420 is a high potency, improgan-like, non-opioid analgesic which may indirectly activate cannabinoid pain-relieving mechanisms.


Assuntos
Analgésicos não Narcóticos/química , Analgésicos não Narcóticos/farmacologia , Cimetidina/análogos & derivados , Medição da Dor/efeitos dos fármacos , Analgésicos/síntese química , Analgésicos/química , Analgésicos/farmacologia , Analgésicos não Narcóticos/síntese química , Animais , Comportamento Animal/efeitos dos fármacos , Antagonistas de Receptores de Canabinoides , Linhagem Celular , Cimetidina/síntese química , Cimetidina/química , Cimetidina/farmacologia , Relação Dose-Resposta a Droga , Guanosina 5'-O-(3-Tiotrifosfato)/farmacocinética , Humanos , Injeções Intraventriculares/métodos , Masculino , Limiar da Dor/efeitos dos fármacos , Piperidinas/farmacologia , Ligação Proteica/efeitos dos fármacos , Pirazóis/farmacologia , Ensaio Radioligante , Ratos , Ratos Sprague-Dawley , Tempo de Reação/efeitos dos fármacos , Receptores de Canabinoides/efeitos dos fármacos , Receptores de Canabinoides/fisiologia , Rimonabanto , Isótopos de Enxofre/farmacocinética
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa