Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Metabolism ; 114: 154414, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33129839

RESUMO

AIMS/HYPOTHESIS: Type 2 diabetes (T2D) is characterized by a progressive loss of beta-cell function, and the "disappearance" of beta-cells in T2D may also be caused by the process of beta -cell dedifferentiation. Since noradrenergic innervation inhibits insulin secretion and density of noradrenergic fibers is increased in type 2 diabetes mouse models, we aimed to study the relation between islet innervation, dedifferentiation and beta-cell function in humans. METHODS: Using immunohistochemistry and electron microscopy, we analyzed pancreata from organ donors and from patients undergoing pancreatic surgery. In the latter, a pre-surgical detailed metabolic characterization by oral glucose tolerance test (OGTT) and hyperglycemic clamp was performed before surgery, thus obtaining in vivo functional parameters of beta-cell function and insulin secretion. RESULTS: The islets of diabetic subjects were 3 times more innervated than controls (0.91 ±â€¯0.21 vs 0.32 ±â€¯0.10, n.fibers/islet; p = 0.01), and directly correlated with the dedifferentiation score (r = 0.39; p = 0.03). In vivo functional parameters of insulin secretion, assessed by hyperglycemic clamp, negatively correlated with the increase in fibers [beta-cell Glucose Sensitivity (r = -0.84; p = 0.01), incremental second-phase insulin secretion (r = -0.84, p = 0.03) and arginine-stimulated insulin secretion (r = -0.76, p = 0.04)]. Moreover, we observed a progressive increase in fibers, paralleling worsening glucose tolerance (from NGT through IGT to T2D). CONCLUSIONS/INTERPRETATION: Noradrenergic fibers are significantly increased in the islets of diabetic subjects and this positively correlates with beta-cell dedifferentiation score. The correlation between in vivo insulin secretion parameters and the density of pancreatic noradrenergic fibers suggests a significant involvement of these fibers in the pathogenesis of the disease, and indirectly, in the islet dedifferentiation process.


Assuntos
Neurônios Adrenérgicos/fisiologia , Desdiferenciação Celular/fisiologia , Diabetes Mellitus Tipo 2/metabolismo , Glibureto/metabolismo , Secreção de Insulina/fisiologia , Células Secretoras de Insulina/metabolismo , Fibras Nervosas/fisiologia , Idoso , Glicemia/metabolismo , Feminino , Intolerância à Glucose/metabolismo , Humanos , Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , Masculino , Pessoa de Meia-Idade
2.
Front Immunol ; 11: 956, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32508835

RESUMO

As for many other adult stem cells, the behavior of hematopoietic stem and progenitor cells (HSPCs) is subjected to circadian regulatory patterns. Multiple HSPC functions, such as proliferation, differentiation or trafficking exhibit time-dependent patterns that require a tight coordination to ensure daily blood cell production. The autonomic nervous system, together with circulating hormones, relay circadian signals from the central clock-the suprachiasmatic nucleus in the brain-to synchronize HSC niche physiology according to light/darkness cycles. Research over the last 20 years has revealed how specific neural signals modulate certain aspects of circadian HSC biology. However, only recently some studies have started to decipher the cellular and molecular mechanisms that orchestrate this complex regulation in a time-dependent fashion. Here we firstly review some of the recent key findings illustrating how different neural signals (catecholaminergic or cholinergic) regulate circadian HSC egress, homing, maintenance, proliferation, and differentiation. In particular, we highlight the critical role of different neurotransmitter receptors in the bone marrow microenvironment to channel these neural signals and regulate antagonistic processes according to circadian cues and organismal demands. Then, we discuss the potential biological meaning of HSC circadian regulation and its possible utility for clinical purposes. Finally, we offer our perspective on emerging concepts in HSC chronobiology.


Assuntos
Sistema Nervoso Autônomo/fisiologia , Medula Óssea/inervação , Ritmo Circadiano , Células-Tronco Hematopoéticas/fisiologia , Periodicidade , Núcleo Supraquiasmático/fisiologia , Neurônios Adrenérgicos/fisiologia , Animais , Diferenciação Celular , Proliferação de Células , Neurônios Colinérgicos/fisiologia , Regulação da Expressão Gênica , Células-Tronco Hematopoéticas/metabolismo , Humanos , Transdução de Sinais , Nicho de Células-Tronco , Fatores de Tempo
5.
Anesth Analg ; 125(4): 1281-1288, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28787345

RESUMO

BACKGROUND: The tricyclic antidepressant amitriptyline, the serotonin and noradrenaline reuptake inhibitor duloxetine, and gabapentinoids are first-line drugs for treatment of neuropathic pain. The analgesic effect of these drugs relates to brainstem-spinal descending noradrenergic systems. However, amitriptyline utilizes a variety of mechanisms for analgesia in neuropathic pain, and it is unclear which mechanism is most important. In the present study, we investigated the role of descending noradrenergic systems in the analgesic effect of these drugs for treatment of neuropathic pain. We also examined whether amitriptyline modifies the descending noradrenergic systems. METHODS: Seven days after L5 spinal nerve ligation (SNL), rats received N-(2-chloroethyl)-N-ethyl-2-bromobenzylamine (DSP-4, 50 mg/kg) to degenerate noradrenergic fibers. The rats then received 5 daily intraperitoneal injections of amitriptyline (10 mg/kg), duloxetine (10 mg/kg), pregabalin (10 mg/kg), or gabapentin (50 mg/kg) from 21 days after SNL surgery. Paw withdrawal thresholds were determined to assess the effect of the drugs on hyperalgesia after SNL. To determine whether 5 daily injections of amitriptyline activated noradrenergic neurons in the locus coeruleus (LC) and spinal cord with or without DSP-4 treatment, we performed immunohistochemistry using antibodies for c-Fos and dopamine beta-hydroxylase (DßH). RESULTS: Five daily injections of amitriptyline, duloxetine, pregabalin, and gabapentin exerted antihyperalgesic effects in SNL rats (P < .001; estimated treatment effect of amitriptyline [99% confidence interval]: 59.9 [35.1-84.7] g). The antihyperalgesic effects of duloxetine, pregabalin, and gabapentin were reversed by pretreatment with DSP-4 (P < .001, respectively). However, antihyperalgesia was still observed after treatment of amitriptyline in SNL rats with DSP-4 pretreatment (P < .001, 59.7 [30.0-89.3] g), and this analgesic effect was not reversed by the α2-adrenoceptor antagonist idazoxan (30 µg). Additionally, 5 daily injections of amitriptyline increased the ratio of c-Fos-immunoreactive (IR) cells in noradrenergic LC neurons in SNL rats with or without DSP-4 pretreatment (P < .001, respectively). Five daily injections of amitriptyline increased DßH-IR in the LC and the spinal dorsal horn of SNL rats (P < .001, respectively). With DSP-4 pretreatment, DßH-IR was dramatically decreased with or without 5 daily injections of amitriptyline (P < .001). CONCLUSIONS: Five daily injections of amitriptyline produced antihyperalgesic effects against neuropathic pain despite suppression of noradrenergic descending inhibitory systems. Amitriptyline activated LC neurons and increased noradrenergic fibers density in SNL rats. These results suggest that amitriptyline could still produce analgesia under pathological dysfunction of the descending noradrenergic system. Amitriptyline may enhance the analgesic effect of drugs for neuropathic pain that require normal descending noradrenergic inhibition to produce analgesia, such as serotonin and noradrenaline reuptake inhibitors and gabapentinoids.


Assuntos
Neurônios Adrenérgicos/efeitos dos fármacos , Amitriptilina/administração & dosagem , Analgésicos não Narcóticos/administração & dosagem , Inibição Neural/efeitos dos fármacos , Neuralgia/tratamento farmacológico , Tratos Piramidais/efeitos dos fármacos , Neurônios Adrenérgicos/fisiologia , Animais , Masculino , Inibição Neural/fisiologia , Neuralgia/fisiopatologia , Medição da Dor/efeitos dos fármacos , Medição da Dor/métodos , Limiar da Dor/efeitos dos fármacos , Limiar da Dor/fisiologia , Tratos Piramidais/fisiopatologia , Ratos , Ratos Sprague-Dawley
6.
Brain Res ; 1663: 1-8, 2017 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-28284896

RESUMO

With the decline of ovarian steroids levels at menopause, many women experience an increase in anxiety and stress sensitivity. The locus coeruleus (LC), a central source of noradrenaline (NE), is activated by stress and is inhibited by ß-endorphin. Moreover, increased NE has been implicated in pathological anxiety syndromes. Hormone replacement therapy (HRT) in menopause appears to decrease anxiety and vulnerability to stress. Therefore, we questioned the effect of HRT on the inhibitory ß-endorphin innervation of the LC. In addition, we found that progesterone protects serotoninergic neurons in monkeys, leading us to question whether ovarian steroids are also neuroprotective in LC neurons in monkeys. Adult Rhesus monkeys (Macaca mulatta) were ovariectomized, and either treated with Silastic capsules that contained estradiol, estradiol+progesterone, progesterone alone or that were empty (ovariectomized; control). After 1month, the LC was obtained and processed for immunohistochemistry for ß-endorphin and terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end-labeling (TUNEL). The density of ß-endorphin axons was determined with image analysis using ImageJ. The TUNEL-positive neurons were counted in the entire LC. Progesterone-alone significantly increased the density of the ß-endorphin axons in the LC (p<0.01). No significant differences between groups in the number of TUNEL-positive cells in the LC were found. In conclusion, we found that HRT increases the inhibitory influence of ß-endorphin in the LC, which could, in turn, contribute to reduce anxiety and increase stress resilience. In addition, we did not find compelling evidence of neurodegeneration or neuroprotection by HRT in the LC of Rhesus monkeys.


Assuntos
Locus Cerúleo/efeitos dos fármacos , beta-Endorfina/efeitos dos fármacos , Neurônios Adrenérgicos/efeitos dos fármacos , Neurônios Adrenérgicos/fisiologia , Animais , Estradiol/farmacologia , Estrogênios/farmacologia , Feminino , Haplorrinos , Terapia de Reposição Hormonal , Marcação In Situ das Extremidades Cortadas , Locus Cerúleo/fisiologia , Macaca mulatta/fisiologia , Menopausa/efeitos dos fármacos , Modelos Animais , Neurônios/efeitos dos fármacos , Norepinefrina/metabolismo , Norepinefrina/farmacologia , Ovariectomia , Ovário , Progesterona/metabolismo , Progesterona/farmacologia , Esteroides , beta-Endorfina/metabolismo
7.
Brain Struct Funct ; 222(1): 317-339, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27056728

RESUMO

The vermis or "spinocerebellum" receives input from the spinal cord and motor cortex for controlling balance and locomotion, while the longitudinal hemisphere region or "cerebro-cerebellum" is interconnected with non-motor cortical regions, including the prefrontal cortex that underlies decision-making. Noradrenaline release in the cerebellum is known to be important for motor plasticity but less is known about plasticity of the cerebellar noradrenergic (NA) system, itself. We characterized plasticity of dopamine ß-hydroxylase-immunoreactive NA fibers in the cerebellum of adolescent female rats that are evoked by voluntary wheel running, food restriction (FR) or by both, in combination. When 8 days of wheel access was combined with FR during the last 4 days, some responded with excessive exercise, choosing to run even during the hours of food access: this exacerbated weight loss beyond that due to FR alone. In the vermis, exercise, with or without FR, shortened the inter-varicosity intervals and increased varicosity density along NA fibers, while excessive exercise, due to FR, also shortened NA fibers. In contrast, the hemisphere required the FR-evoked excessive exercise to evoke shortened inter-varicosity intervals along NA fibers and this change was exhibited more strongly by rats that suppressed the FR-evoked excessive exercise, a behavior that minimized weight loss. Presuming that shortened inter-varicosity intervals translate to enhanced NA release and synthesis of norepinephrine, this enhancement in the cerebellar hemisphere may contribute towards protection of individuals from the life-threatening activity-based anorexia via relays with higher-order cortical areas that mediate the animal's decision to suppress the innate FR-evoked hyperactivity.


Assuntos
Neurônios Adrenérgicos/citologia , Neurônios Adrenérgicos/fisiologia , Anorexia/patologia , Anorexia/fisiopatologia , Cerebelo/citologia , Cerebelo/fisiologia , Atividade Motora , Plasticidade Neuronal , Animais , Vermis Cerebelar/citologia , Vermis Cerebelar/fisiologia , Modelos Animais de Doenças , Dopamina/fisiologia , Dopamina beta-Hidroxilase/metabolismo , Ingestão de Alimentos , Feminino , Ratos , Ratos Sprague-Dawley
8.
Neuropharmacology ; 114: 114-122, 2017 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-27908769

RESUMO

Selective serotonin reuptake inhibitors (SSRIs) regulate brain noradrenergic neurotransmission both at somatodendritic and nerve terminal areas. Previous studies have demonstrated that noradrenaline (NA) reuptake inhibitors are able to desensitize α2-adrenoceptor-mediated responses. The present study was undertaken to elucidate the effects of repeated treatment with the SSRI citalopram on the α2-adrenoceptor sensitivity in locus coeruleus (LC) and prefrontal cortex (PFC), by using in vivo microdialysis and electrophysiological techniques, and in vitro stimulation of [35S]GTPγS binding autoradiography. Repeated, but not acute, treatment with citalopram (5 mg/kg, i.p., 14 days) increased extracellular NA concentration selectively in PFC. The α2-adrenoceptor agonist clonidine (0.3 mg/kg, i.p.), administered to saline-treated animals (1 ml/kg i.p., 14 days) induced NA decrease in LC (Emax = -44 ± 4%; p < 0.001) and in PFC (Emax = -61 ± 5%, p < 0.001). In citalopram chronically-treated rats, clonidine administration exerted a lower decrease of NA (Emax = -25 ± 7%; p < 0.001) in PFC whereas the effect in LC was not different to controls (Emax = -36 ± 4%). Clonidine administration (0.625-20 µg/kg, i.v.) evoked a dose-dependent decrease of the firing activity of LC noradrenergic neurons in both citalopram- (ED50 = 3.2 ± 0.4 µg/kg) and saline-treated groups (ED50 = 2.6 ± 0.5 µg/kg). No significant differences between groups were found in ED50 values. The α2-adrenoceptor agonist UK14304 stimulated specific [35S]GTPγS binding in brain sections containing LC (144 ± 14%) and PFC (194 ± 32%) of saline-treated animals. In citalopram-treated animals, this increase did not differ from controls in LC (146 ± 22%) but was lower in PFC (141 ± 8%; p < 0.05). Taken together, long-term citalopram treatment induces a desensitization of α2-adrenoceptors acting as axon terminal autoreceptors in PFC without changes in somatodendritic α2-adrenoceptor sensitivity.


Assuntos
Citalopram/administração & dosagem , Locus Cerúleo/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Córtex Pré-Frontal/efeitos dos fármacos , Receptores Adrenérgicos alfa 2/fisiologia , Inibidores Seletivos de Recaptação de Serotonina/administração & dosagem , Neurônios Adrenérgicos/efeitos dos fármacos , Neurônios Adrenérgicos/fisiologia , Agonistas de Receptores Adrenérgicos alfa 2/administração & dosagem , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Tartarato de Brimonidina/administração & dosagem , Citalopram/farmacocinética , Locus Cerúleo/metabolismo , Locus Cerúleo/fisiologia , Masculino , Neurônios/fisiologia , Norepinefrina/metabolismo , Córtex Pré-Frontal/metabolismo , Córtex Pré-Frontal/fisiologia , Ratos , Ratos Sprague-Dawley , Inibidores Seletivos de Recaptação de Serotonina/farmacocinética
9.
Physiol Behav ; 165: 339-49, 2016 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-27553574

RESUMO

The Wistar-Kyoto (WKY) rat is a widely used animal model of depression, which is characterized by dysregulation of noradrenergic signaling. We previously demonstrated that WKY rats show a unique behavioral profile on the forced swim test (FST), characterized by high levels of immobility upon initial exposure and a greater learning-like response by further increasing immobility upon re-exposure than the genetically related Wistar rats. In the current study we aimed to determine whether altered activation of brainstem noradrenergic cell groups contributes to this behavioral profile. We exposed WKY and Wistar rats, to either 5min of forced swim or to the standard two-day FST (i.e. 15min forced swim on Day 1, followed by 5min on Day 2). We then stained their brains for FOS/tyrosine hydroxylase double-immunocytochemistry to determine potential differences in the activation of the brainstem noradrenergic cell groups. We detected a relative hyperactivation in the locus coeruleus of WKY rats when compared to Wistars in response to both one- and two-day forced swim. In contrast, within the A2 noradrenergic cell group, WKY rats exhibited diminished levels of FOS across both days of the FST, suggesting their lesser activation. We followed up these observations by selectively lesioning the A2 neurons, using anti-dopamine-ß-hydroxylase-conjugated saporin, in Wistar rats, which resulted in increased FST immobility on both days of the test. Together these data indicate that the A2 noradrenergic cell group regulates FST behavior, and that its hypoactivation may contribute to the unique behavioral phenotype of WKY rats.


Assuntos
Neurônios Adrenérgicos/fisiologia , Transtorno Depressivo/patologia , Resposta de Imobilidade Tônica/fisiologia , Natação/psicologia , Neurônios Adrenérgicos/efeitos dos fármacos , Animais , Modelos Animais de Doenças , Dopamina beta-Hidroxilase/farmacologia , Imunotoxinas/uso terapêutico , Masculino , Microinjeções , Proteínas Oncogênicas v-fos/metabolismo , Ratos , Ratos Endogâmicos WKY , Ratos Sprague-Dawley , Ratos Wistar , Proteínas Inativadoras de Ribossomos Tipo 1/farmacologia , Saporinas , Núcleo Solitário/efeitos dos fármacos , Núcleo Solitário/patologia , Especificidade da Espécie , Fatores de Tempo , Tirosina 3-Mono-Oxigenase/metabolismo
10.
Psychoneuroendocrinology ; 70: 1-9, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27131036

RESUMO

Increased activity of the noradrenergic system in the amygdala has been suggested to contribute to the hyperarousal symptoms associated with post-traumatic stress disorder (PTSD). However, only two studies have examined the content of noradrenaline or its metabolites in the amygdala of rats previously exposed to traumatic stress showing inconsistent results. The aim of this study was to investigate the effects of an inescapable foot shock (IFS) procedure (1) on reactivity to novelty in an open-field (as an index of hyperarousal), and (2) on noradrenaline release in the amygdala during an acute stress. To test the role of noradrenaline in amygdala, we also investigated the effects of microinjections of propranolol, a ß-adrenoreceptor antagonist, and clenbuterol, a ß-adrenoreceptor agonist, into the amygdala of IFS and control animals. Finally, we evaluated the expression of mRNA levels of ß-adrenoreceptors (ß1 and ß2) in the amygdala, the hippocampus and the prefrontal cortex. Male Wistar rats (3 months) were stereotaxically implanted with bilateral guide cannulae. After recovering from surgery, animals were exposed to IFS (10 shocks, 0.86mA, and 6s per shock) and seven days later either microdialysis or microinjections were performed in amygdala. Animals exposed to IFS showed a reduced locomotion compared to non-shocked animals during the first 5min in the open-field. In the amygdala, IFS animals showed an enhanced increase of noradrenaline induced by stress compared to control animals. Bilateral microinjections of propranolol (0.5µg) into the amygdala one hour before testing in the open-field normalized the decreased locomotion observed in IFS animals. On the other hand, bilateral microinjections of clenbuterol (30ng) into the amygdala of control animals did not change the exploratory activity induced by novelty in the open field. IFS modified the mRNA expression of ß1 and ß2 adrenoreceptors in the prefrontal cortex and the hippocampus. These results suggest that an increased noradrenergic activity in the amygdala contributes to the expression of hyperarousal in an animal model of PTSD.


Assuntos
Neurônios Adrenérgicos/fisiologia , Tonsila do Cerebelo/fisiopatologia , Transtornos de Estresse Pós-Traumáticos/fisiopatologia , Neurônios Adrenérgicos/efeitos dos fármacos , Neurônios Adrenérgicos/metabolismo , Tonsila do Cerebelo/efeitos dos fármacos , Tonsila do Cerebelo/metabolismo , Animais , Clembuterol/farmacologia , Modelos Animais de Doenças , Eletrochoque , Hipocampo/metabolismo , Masculino , Norepinefrina/metabolismo , Córtex Pré-Frontal/metabolismo , Propranolol/farmacologia , Ratos , Ratos Wistar , Transtornos de Estresse Pós-Traumáticos/etiologia , Transtornos de Estresse Pós-Traumáticos/metabolismo
11.
J Basic Clin Physiol Pharmacol ; 27(6): 557-561, 2016 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-27226099

RESUMO

BACKGROUND: Pain is the most common cause of patients seeking medical advice as a result of its association with different pathologies. This study evaluated the antinociceptive property of Haematostaphis barteri as well as the possible mechanism(s) associated with its antinociceptive property. METHODS: Mice were administered H. barteri (30-300 mg kg-1; p.o.), followed by intraplantar injection of 10 µL of 5% formalin into the hind paws. The pain score was determined for 1 h in the formalin test. The possible nociceptive pathways involved in the antinociceptive action of H. barteri were determined by pre-treating mice with theophylline (5 mg kg-1, a non-selective adenosine receptor antagonist), naloxone (2 mg kg-1, a non-selective opioid receptor antagonist), glibenclamide (8 mg kg-1; an ATP-sensitive K+ channel inhibitor), and atropine (3 mg kg-1; non-selective muscarinic antagonist). RESULTS: H. barteri (30-300 mg kg-1) significantly and dose dependently precluded both first and second phases of nociception. Pre-treatment with naloxone had no effect on the analgesic activities of H. barteri in the first phase. Again, pre-treatment with atropine and glibenclamide did not significantly reverse the neurogenic antinociception of the extract in phase 1. However, theophylline reversed the analgesic effect of the extract in the first phase. In phase 2, theophylline had no effect on the analgesic activities of the extract. Naloxone, atropine, and glibenclamide significantly blocked the antinociception of H. barteri in the inflammatory phase of the formalin test. CONCLUSIONS: H. barteri possesses antinociceptive property mediated via the opioidergic, adrenergic, muscarinic, ATP-sensitive K+ channels, and adenosinergic nociceptive pathways.


Assuntos
Anacardiaceae , Analgésicos/farmacologia , Canais KATP/antagonistas & inibidores , Medição da Dor/efeitos dos fármacos , Extratos Vegetais/farmacologia , Folhas de Planta , Neurônios Adrenérgicos/efeitos dos fármacos , Neurônios Adrenérgicos/fisiologia , Analgésicos/isolamento & purificação , Animais , Feminino , Canais KATP/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos ICR , Medição da Dor/métodos , Extratos Vegetais/isolamento & purificação , Receptores Muscarínicos/fisiologia , Receptores Opioides/fisiologia , Receptores Purinérgicos P1/fisiologia , Neurônios Serotoninérgicos/efeitos dos fármacos , Neurônios Serotoninérgicos/fisiologia
12.
BJOG ; 123(7): 1076-85, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26301457

RESUMO

OBJECTIVE: To evaluate the impact of prolapse meshes on vaginal smooth muscle structure (VaSM) and function, and to evaluate these outcomes in the context of the mechanical and textile properties of the mesh. DESIGN: Three months following the implantation of three polypropylene prolapse meshes with distinct textile and mechanical properties, mesh tissue explants were evaluated for smooth muscle contraction, innervation, receptor function, and innervation density. SETTING: Magee-Womens Research Institute at the University of Pittsburgh. POPULATION: Thirty-four parous rhesus macaques of similar age, parity, and pelvic organ prolapse quantification (POP-Q) scores. METHODS: Macaques were implanted with mesh via sacrocolpopexy. The impact of Gynemesh(™)  PS (Ethicon; n = 7), Restorelle(®) (Coloplast; n = 7), UltraPro(™) parallel and UltraPro(™) perpendicular (Ethicon; n = 6 and 7, respectively) were compared with sham-operated controls (n = 7). Outcomes were analysed by Kruskal-Wallis ANOVA, Mann-Whitney U-tests and multiple regression analysis (P < 0.05). MEAN OUTCOME MEASURES: Vaginal tissue explants were evaluated for the maximum contractile force generated following muscle, nerve, and receptor stimulation, and for peripheral nerve density. RESULTS: Muscle myofibre, nerve, and receptor-mediated contractions were negatively affected by mesh only in the grafted region (P < 0.001, P = 0.002, and P = 0.008, respectively), whereas cholinergic and adrenergic nerve densities were affected in the grafted (P = 0.090 and P = 0.008, respectively) and non-grafted (P = 0.009 and P = 0.005, respectively) regions. The impact varied by mesh property, as mesh stiffness was a significant predictor of the negative affect on muscle function and nerve density (P < 0.001 and P = 0.013, respectively), whereas mesh and weight was a predictor of receptor function (P < 0.001). CONCLUSIONS: Mesh has an overall negative impact on VaSM, and the effects are a function of mesh properties, most notably, mesh stiffness. TWEETABLE ABSTRACT: Prolapse mesh affects vaginal smooth muscle.


Assuntos
Músculo Liso/patologia , Prolapso de Órgão Pélvico/cirurgia , Polipropilenos/efeitos adversos , Telas Cirúrgicas/efeitos adversos , Vagina/patologia , Neurônios Adrenérgicos/patologia , Neurônios Adrenérgicos/fisiologia , Animais , Fenômenos Biomecânicos , Biópsia/métodos , Neurônios Colinérgicos/patologia , Neurônios Colinérgicos/fisiologia , Desenho de Equipamento , Feminino , Macaca mulatta , Agonistas Muscarínicos/farmacologia , Contração Muscular/fisiologia , Fibras Musculares Esqueléticas/patologia , Fibras Musculares Esqueléticas/fisiologia , Músculo Liso/inervação , Músculo Liso/fisiopatologia , Prolapso de Órgão Pélvico/patologia , Prolapso de Órgão Pélvico/fisiopatologia , Nervos Periféricos/patologia , Nervos Periféricos/fisiopatologia , Polipropilenos/administração & dosagem , Receptores Muscarínicos/efeitos dos fármacos , Receptores Muscarínicos/fisiologia , Vagina/inervação , Vagina/fisiopatologia
13.
J Physiol ; 593(23): 5057-73, 2015 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-26420487

RESUMO

Sympathetic drive to the heart is a key modulator of cardiac function and interactions between heart tissue and innervating sympathetic fibres are established early in development. Significant innervation takes place during postnatal heart development, a period when cardiomyocytes undergo a rapid transition from proliferative to hypertrophic growth. The question of whether these innervating sympathetic fibres play a role in regulating the modes of cardiomyocyte growth was investigated using 6-hydroxydopamine (6-OHDA) to abolish early sympathetic innervation of the heart. Postnatal chemical sympathectomy resulted in rats with smaller hearts, indicating that heart growth is regulated by innervating sympathetic fibres during the postnatal period. In vitro experiments showed that sympathetic interactions resulted in delays in markers of cardiomyocyte maturation, suggesting that changes in the timing of the transition from hyperplastic to hypertrophic growth of cardiomyocytes could underlie changes in heart size in the sympathectomized animals. There was also an increase in the expression of Meis1, which has been linked to cardiomyocyte cell cycle withdrawal, suggesting that sympathetic signalling suppresses cell cycle withdrawal. This signalling involves ß-adrenergic activation, which was necessary for sympathetic regulation of cardiomyocyte proliferation and hypertrophy. The effect of ß-adrenergic signalling on cardiomyocyte hypertrophy underwent a developmental transition. While young postnatal cardiomyocytes responded to isoproterenol (isoprenaline) with a decrease in cell size, mature cardiomyocytes showed an increase in cell size in response to the drug. Together, these results suggest that early sympathetic effects on proliferation modulate a key transition between proliferative and hypertrophic growth of the heart and contribute to the sympathetic regulation of adult heart size.


Assuntos
Neurônios Adrenérgicos/fisiologia , Ciclo Celular , Coração/crescimento & desenvolvimento , Miócitos Cardíacos/citologia , Sistema Nervoso Simpático/fisiologia , Animais , Células Cultivadas , Feminino , Coração/inervação , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Masculino , Proteína Meis1 , Miocárdio/patologia , Miócitos Cardíacos/fisiologia , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Tamanho do Órgão , Ratos , Ratos Sprague-Dawley , Sistema Nervoso Simpático/citologia
14.
J Pharmacol Sci ; 124(4): 502-10, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24681698

RESUMO

Bortezomib, an inhibitor of proteasome holoenzyme, is used to treat relapsed and refractory multiple myeloma. Peripheral neuropathy is a treatment-limiting adverse effect of bortezomib and is very difficult to control. In this study, we examined the efficacy of gabapentin in inhibiting bortezomib-induced peripheral neuropathy. Single intravenous injections of bortezomib (0.03 - 0.3 mg/kg) dose-dependently induced mechanical allodynia with a peak effect 12 days after injection. Bortezomib (0.3 mg/kg) also caused mechanical hyperalgesia, but neither affected thermal nociception nor induced cold allodynia. Bortezomib increased the response of the saphenous nerve to weak punctate stimulation but not response to cool stimulation of the skin. When administered 12 days after bortezomib injection, oral and intracisternal gabapentin markedly inhibited mechanical allodynia. Intrathecal, but not intraplantar, gabapentin had a tendency to reduce mechanical allodynia. The antiallodynic activity of orally administered gabapentin was suppressed by noradrenaline, but not serotonin, depletion in the spinal cord. Bortezomib did not affect the expression levels of the calcium channel α2δ-1 subunit, a high-affinity binding site of gabapentin, in the plantar skin, spinal cord, medulla oblongata, and pons. These results suggest that gabapentin inhibits bortezomib-induced mechanical allodynia, most likely through the activation of the descending noradrenergic system.


Assuntos
Aminas/farmacologia , Antineoplásicos/efeitos adversos , Antineoplásicos/antagonistas & inibidores , Ácidos Borônicos/efeitos adversos , Ácidos Borônicos/antagonistas & inibidores , Ácidos Cicloexanocarboxílicos/farmacologia , Hiperalgesia/induzido quimicamente , Hiperalgesia/tratamento farmacológico , Pirazinas/efeitos adversos , Pirazinas/antagonistas & inibidores , Ácido gama-Aminobutírico/farmacologia , Neurônios Adrenérgicos/fisiologia , Aminas/administração & dosagem , Animais , Antineoplásicos/administração & dosagem , Ácidos Borônicos/administração & dosagem , Bortezomib , Ácidos Cicloexanocarboxílicos/administração & dosagem , Gabapentina , Camundongos , Camundongos Endogâmicos C57BL , Norepinefrina/fisiologia , Pirazinas/administração & dosagem , Medula Espinal/efeitos dos fármacos , Ácido gama-Aminobutírico/administração & dosagem
15.
Neuroscience ; 269: 199-214, 2014 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-24631866

RESUMO

Neuro-glucostasis is required for normal expression of the steroid positive-feedback-induced preovulatory pituitary luteinizing hormone (LH) surge, a critical element of female reproduction. Glucoprivic signals from the caudal hindbrain restrain this surge, but the cellular source of this stimulus is unclear. Norepinephrine (NE) exerts well-defined stimulatory effects on the reproductive neuroendocrine axis. Our studies show that medullary A2 noradrenergic neurons are both estrogen- and glucoprivic-sensitive. Here, we investigated the premise that the LH surge is inhibited by A2 cell reactivity to hindbrain glucopenia and diminished preoptic NE neurotransmission. Estradiol- and progesterone-primed ovariectomized (OVX) female rats were injected into the caudal fourth ventricle (CV4) with the glucose anti-metabolite, 5-thioglucose (5TG) or saline (SAL) prior to onset of the LH surge. Pretreatment by intra-CV4 delivery of the selective catecholamine neurotoxin, 6-OHDA, attenuated LH output, but prevented inhibition by 5TG. 5TG modified patterns of steroid feedback-associated Fos staining of A2, but not other medullary catecholamine cell groups. Intra-preoptic administration of the alpha1-adrenergic receptor agonist, methoxamine, elicited site-specific reversal of hindbrain glucoprivic suppression of gonadotropin-releasing hormone (GnRH) neuron Fos labeling and LH release. Western blotting of laser-microdissected A2 neurons revealed glucoprivic stimulation of Fos, but inhibition of the catecholamine synthetic enzyme, dopamine-ß-hydroxylase; 5TG also diminished A2 estrogen receptor (ER)-α and progesterone receptor profiles, but augmented ER-ß protein. Intriguingly, A2 AMPK activity was decreased in 5TG-treated rats, despite down-regulation of GLUT3 and no change in MCT2 protein expression. Rostral preoptic GnRH neurons also exhibited decreased AMPK activation simultaneous with apparent reduction of neuropeptide signaling to the pituitary. The present studies demonstrate that hindbrain glucoprivation inhibits the LH surge, in part, by reducing preoptic noradrenergic input, and furthermore implicate A2 neurons as a source of this altered signal. Results also suggest that AMPK sensor deactivation does not supersede the impact of pharmacological inhibition of glucose catabolism on A2 cell function nor afferent signaling of hindbrain glucopenia on GnRH neurons. Further studies are needed to determine if decreased AMPK activation in these cell populations reflect compensatory gain in positive energy balance and/or direct effects of estrogen on AMPK.


Assuntos
Neurônios Adrenérgicos/fisiologia , Hormônio Luteinizante/metabolismo , Receptores A2 de Adenosina/metabolismo , Rombencéfalo/fisiologia , Proteínas Quinases Ativadas por AMP/metabolismo , Adrenérgicos/farmacologia , Neurônios Adrenérgicos/efeitos dos fármacos , Agonistas de Receptores Adrenérgicos alfa 1/farmacologia , Animais , Biomarcadores/metabolismo , Fármacos do Sistema Nervoso Central/farmacologia , Estradiol/farmacologia , Feminino , Glucose/análogos & derivados , Glucose/farmacologia , Hormônio Liberador de Gonadotropina/metabolismo , Hormônios/farmacologia , Metoxamina/farmacologia , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Ovariectomia , Oxidopamina/farmacologia , Progesterona/farmacologia , Ratos Sprague-Dawley , Rombencéfalo/efeitos dos fármacos
16.
J Neurosci ; 33(43): 17138-49, 2013 Oct 23.
Artigo em Inglês | MEDLINE | ID: mdl-24155317

RESUMO

High-level spinal cord injury can lead to cardiovascular dysfunction, including disordered hemodynamics at rest and autonomic dysreflexia during noxious stimulation. To restore supraspinal control of sympathetic preganglionic neurons (SPNs), we grafted embryonic brainstem-derived neural stem cells (BS-NSCs) or spinal cord-derived neural stem cells (SC-NSCs) expressing green fluorescent protein into the T4 complete transection site of adult rats. Animals with injury alone served as controls. Implanting of BS-NSCs but not SC-NSCs resulted in recovery of basal cardiovascular parameters, whereas both cell grafts alleviated autonomic dysreflexia. Subsequent spinal cord retransection above the graft abolished the recovery of basal hemodynamics and reflexic response. BS-NSC graft-derived catecholaminergic and serotonergic neurons showed remarkable long-distance axon growth and topographical innervation of caudal SPNs. Anterograde tracing indicated growth of medullar axons into stem cell grafts and formation of synapses. Thus, grafted embryonic brainstem-derived neurons can act as functional relays to restore supraspinal regulation of denervated SPNs, thereby contributing to cardiovascular functional improvement.


Assuntos
Células-Tronco Embrionárias/transplante , Coração/inervação , Hemodinâmica , Regeneração Nervosa , Células-Tronco Neurais/transplante , Traumatismos da Medula Espinal/cirurgia , Fibras Adrenérgicas/fisiologia , Neurônios Adrenérgicos/fisiologia , Animais , Disreflexia Autonômica/cirurgia , Fibras Autônomas Pré-Ganglionares/fisiologia , Axônios/fisiologia , Tronco Encefálico/citologia , Processos de Crescimento Celular , Feminino , Coração/fisiopatologia , Ratos , Ratos Endogâmicos F344 , Reflexo , Neurônios Serotoninérgicos/fisiologia , Medula Espinal/citologia , Medula Espinal/fisiopatologia , Transplante de Células-Tronco , Sinapses/fisiologia
17.
J Neurophysiol ; 110(11): 2580-91, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24027106

RESUMO

The pathogenesis of psychiatric and neurodegenerative diseases is often associated with a deregulation of noradrenergic transmission. Considering the potential involvement of purinergic signaling in the modulation of noradrenergic transmission in the brain cortex, this study aimed to identify the P2Y receptor subtypes involved in the modulation of neuronal release and neuronal/glial uptake of norepinephrine. Electrical stimulation (100 pulses at 5 Hz) of rat cortical slices induced norepinephrine release that was inhibited by ATP and ADP (0.01-1 mM), adenosine 5'-O-(2-thiodiphosphate) (ADPßS, 0.03-0.3 mM), and UDP (0.1-1 mM). The effect of ADPßS was mediated by P2Y1 receptors and possibly by A1/P2Y1 heterodimers since it was attenuated by the A1 receptor antagonist DPCPX and by the P2Y1 receptor antagonist MRS 2500 but was resistant to the effect of adenosine deaminase (ADA). UDP inhibited norepinephrine release through activation of P2Y6 receptors, an effect that was abolished by the P2Y6 receptor antagonist MRS 2578 and by DPCPX, indicating that it depends on the formation and/or release of adenosine and activation of A1 receptors. Supporting this hypothesis, the inhibitory effect of UDP was also prevented by inhibition of ectonucleotidases, by ADA and was attenuated by the inhibitor of nucleoside transporter 6-[(4-nitrobenzyl)thio]-9-ß-d-ribofuranosylpurine (NBTI). Additionally, the inhibitory effect of UDP was attenuated when norepinephrine uptake 1 or 2 was inhibited. In astroglial cultures, ADPßS and UDP increased norepinephrine uptake mainly by activation of P2Y1 and P2Y6 receptors, respectively. The results indicate that neuronal and glial P2Y1 and P2Y6 receptors may represent new targets of intervention to regulate noradrenergic transmission in CNS diseases.


Assuntos
Astrócitos/metabolismo , Córtex Cerebral/metabolismo , Exocitose , Norepinefrina/metabolismo , Receptores Purinérgicos P2Y/metabolismo , Potenciais de Ação , Neurônios Adrenérgicos/metabolismo , Neurônios Adrenérgicos/fisiologia , Animais , Astrócitos/fisiologia , Córtex Cerebral/citologia , Córtex Cerebral/fisiologia , Estimulação Elétrica , Masculino , Agonistas do Receptor Purinérgico P2Y/farmacologia , Antagonistas do Receptor Purinérgico P2Y/farmacologia , Ratos , Ratos Wistar
18.
Am J Physiol Endocrinol Metab ; 305(10): E1266-73, 2013 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-24064338

RESUMO

Oleoylethanolamide (OEA) is a gut-derived endogenous lipid that stimulates vagal fibers to induce satiety. Our previous work has shown that peripherally administered OEA activates c-fos transcription in the nucleus of the solitary tract (NST) and in the paraventricular nucleus (PVN), where it enhances oxytocin (OXY) expression. The anorexigenic action of OEA is prevented by the intracerebroventricular administration of a selective OXY receptor antagonist, suggesting a necessary role of OXYergic mediation of OEA's effect. The NST is the source of direct noradrenergic afferent input to hypothalamic OXY neurons, and therefore, we hypothesized that the activation of this pathway might mediate OEA effects on PVN neurons. To test this hypothesis, we subjected rats to intra-PVN administration of the toxin saporin (DSAP) conjugated to an antibody against dopamine-ß-hydroxylase (DBH) to destroy hindbrain noradrenergic neurons. In these rats we evaluated the effects of OEA (10 mg/kg, ip) on feeding behavior, on c-Fos and OXY immunoreactivity in the PVN, and on OXY immunoreactivity in the posterior pituitary gland. We found that the DSAP lesion completely prevented OEA's effects on food intake, on Fos and OXY expression in the PVN, and on OXY immunoreactivity of the posterior pituitary gland; all effects were maintained in sham-operated rats. These results support the hypothesis that noradrenergic NST-PVN projections are involved in the activation of the hypothalamic OXY system, which mediates OEA's prosatiety action.


Assuntos
Neurônios Adrenérgicos/efeitos dos fármacos , Ácidos Oleicos/farmacologia , Ocitocina/metabolismo , Núcleo Hipotalâmico Paraventricular/efeitos dos fármacos , Rombencéfalo/efeitos dos fármacos , Saciação/efeitos dos fármacos , Neurônios Adrenérgicos/fisiologia , Animais , Dopamina beta-Hidroxilase/imunologia , Endocanabinoides , Imunotoxinas/farmacologia , Masculino , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Núcleo Hipotalâmico Paraventricular/fisiologia , Ratos , Ratos Wistar , Rombencéfalo/fisiologia , Proteínas Inativadoras de Ribossomos Tipo 1/farmacologia , Saporinas
19.
Hypertension ; 62(2): 263-73, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23753413

RESUMO

Catecholaminergic C1 cells of the rostral ventrolateral medulla (RVLM) are key determinants of the sympathoexcitatory response to peripheral chemoreceptor activation. Overactivation of this reflex is thought to contribute to increased sympathetic activity and hypertension; however, molecular mechanisms linking peripheral chemoreceptor drive to hypertension remain poorly understood. We have recently determined that activation of P2Y1 receptors in the RVLM mimicked effects of peripheral chemoreceptor activation. Therefore, we hypothesize that P2Y1 receptors regulate peripheral chemoreceptor drive in this region. Here, we determine whether P2Y1 receptors are expressed by C1 neurons in the RVLM and contribute to peripheral chemoreceptor control of breathing, sympathetic activity, and blood pressure. We found that injection of a specific P2Y1 receptor agonist (MRS2365) into the RVLM of anesthetized adult rats increased phrenic nerve activity (≈55%), sympathetic nerve activity (38 ± 6%), and blood pressure (23 ± 1 mm Hg), whereas application of a specific P2Y1 receptor antagonist (MRS2179) decreased peripheral chemoreceptor-mediated activation of phrenic nerve activity, sympathetic nerve activity, and blood pressure. To establish that P2Y1 receptors are expressed by C1 cells, we determine in the brain slice preparation using cell-attached recording techniques that cells responsive to MRS2365 are immunoreactive for tyrosine hydroxylase (a marker of C1 cells), and we determine in vivo that C1-lesioned animals do not respond to RVLM injection of MRS2365. These data identify P2Y1 receptors as key determinants of peripheral chemoreceptor regulation of breathing, sympathetic nerve activity, and blood pressure.


Assuntos
Neurônios Adrenérgicos/fisiologia , Pressão Sanguínea/fisiologia , Células Quimiorreceptoras/fisiologia , Bulbo/fisiologia , Receptores Purinérgicos P2Y1/fisiologia , Sistema Nervoso Simpático/fisiologia , Difosfato de Adenosina/análogos & derivados , Difosfato de Adenosina/farmacologia , Animais , Nervo Frênico/fisiologia , Ratos , Receptores Purinérgicos P2Y1/análise , Respiração
20.
Brain Struct Funct ; 218(2): 455-75, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22460939

RESUMO

The rostral ventrolateral medulla (RVLM) primarily regulates respiration and the autonomic nervous system. Its medial portion (mRVLM) contains many choline acetyltransferase (ChAT)-immunoreactive (ir) neurons of unknown function. We sought to clarify the role of these cholinergic cells by tracing their axonal projections. We first established that these neurons are neither parasympathetic preganglionic neurons nor motor neurons because they did not accumulate intraperitoneally administered Fluorogold. We traced their axonal projections by injecting a Cre-dependent vector (floxed-AAV2) expressing either GFP or mCherrry into the mRVLM of ChAT-Cre mice. Transduced neurons expressing GFP or mCherry were confined to the injection site and were exclusively ChAT-ir. Their axonal projections included the dorsal column nuclei, medullary trigeminal complex, cochlear nuclei, superior olivary complex and spinal cord lamina III. For control experiments, the floxed-AAV2 (mCherry) was injected into the RVLM of dopamine beta-hydroxylase-Cre mice. In these mice, mCherry was exclusively expressed by RVLM catecholaminergic neurons. Consistent with data from rats, these catecholaminergic neurons targeted brain regions involved in autonomic and endocrine regulation. These regions were almost totally different from those innervated by the intermingled mRVLM-ChAT neurons. This study emphasizes the advantages of using Cre-driver mouse strains in combination with floxed-AAV2 to trace the axonal projections of chemically defined neuronal groups. Using this technique, we revealed previously unknown projections of mRVLM-ChAT neurons and showed that despite their close proximity to the cardiorespiratory region of the RVLM, these cholinergic neurons regulate sensory afferent information selectively and presumably have little to do with respiration or circulatory control.


Assuntos
Fibras Colinérgicas/fisiologia , Bulbo/fisiologia , Sensação , Células Receptoras Sensoriais/fisiologia , Neurônios Adrenérgicos/metabolismo , Neurônios Adrenérgicos/fisiologia , Vias Aferentes/fisiologia , Animais , Biomarcadores/metabolismo , Catecolaminas/metabolismo , Colina O-Acetiltransferase/genética , Colina O-Acetiltransferase/metabolismo , Fibras Colinérgicas/metabolismo , Dependovirus/genética , Dopamina beta-Hidroxilase/genética , Feminino , Corantes Fluorescentes/administração & dosagem , Vetores Genéticos , Proteínas de Fluorescência Verde/biossíntese , Proteínas de Fluorescência Verde/genética , Imuno-Histoquímica , Injeções Intraperitoneais , Integrases/genética , Proteínas Luminescentes/biossíntese , Proteínas Luminescentes/genética , Masculino , Bulbo/citologia , Bulbo/metabolismo , Camundongos , Camundongos Transgênicos , Técnicas de Rastreamento Neuroanatômico , Marcadores do Trato Nervoso/administração & dosagem , Regiões Promotoras Genéticas , Células Receptoras Sensoriais/metabolismo , Estilbamidinas/administração & dosagem , Transdução Genética , Tirosina 3-Mono-Oxigenase/metabolismo , Proteína Vermelha Fluorescente
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA