Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
BMC Anesthesiol ; 24(1): 100, 2024 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-38475719

RESUMO

BACKGROUND: Mediastinal tumors pose a challenging respiratory and circulatory management during anesthesia procedures, there is a risk of circulatory collapse or complete airway obstruction, which in severe cases can lead to cardiac arrest. We reported a case of anesthetic management using a bronchial blocker placed outside the tracheal tube. In this case report, the patient's trachea was so severely compressed that the airway was extremely narrow, only 4 mm at its narrowest point. By reporting the anesthetic management of this patient, we intend to provide an unusual approach for airway management. CASE PRESENTATION: A 52-year-old male patient was admitted to the hospital due to cough and expectoration for one year. Additionally, the patient experienced chest tightness and asthma after physical activity. The enhanced computed tomography revealed there existed an irregular soft tissue mass in the right upper mediastinum, which significantly compressed the trachea and esophagus. The results of the mediastinal puncture pathology showed the presence of mesenchymal tumors. According to the results above, the patient was diagnosed with a mediastinal tumor and scheduled to undergo tumor resection under general anesthesia. We used a bronchial occluder outside the tracheal tube for general anesthesia. After surgery, the patient received thorough treatment and was subsequently discharged from the hospital. CONCLUSION: In patients with severe airway compression from a mediastinal tumor airway compression, positioning a bronchial occluder externally to the tracheal tube is an effective method of airway management. However, we still need more clinical practice to help the process become more standardized.


Assuntos
Anestésicos , Neoplasias do Mediastino , Masculino , Humanos , Pessoa de Meia-Idade , Neoplasias do Mediastino/cirurgia , Brônquios , Traqueia , Anestesia Geral/métodos
2.
Int J Surg Case Rep ; 117: 109427, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38452638

RESUMO

INTRODUCTION AND IMPORTANCE: Intratracheal tumors account for approximately 0.2 % of respiratory tumors, including primary and secondary tumors. Secondary tumors of the upper trachea are most commonly derived from advanced thyroid cancer. Surgical resection is currently the general curative modality for thyroid cancer with tracheal invasion. Patients with tracheal tumors invading and protruding into the lumen may have reduced oxygen reserve capacity, leading to a shorter safe window for general anesthesia induction. Establishing an appropriate artificial airway is essential to ensure intraoperative safety for these patients. Here, we report a case of tracheal tumor caused by differentiated papillary thyroid carcinoma invading the upper segment of the trachea and the non-conventional approach used for intraoperative airway management without traditional endotracheal intubation. PRESENTATION OF CASE: A 59-year-old female presented with bilateral neck masses and hemoptysis. The CT scan revealed suspicious malignant thyroid nodules, and tracheoscopy showed an approximately 50 % obstruction of the tracheal lumen. The patient's physical examination and biochemical examination showed no significant abnormalities. Based on imaging studies and pre-anesthetic assessment, a multidisciplinary team decided against performing endotracheal intubation in the patient due to the risk of tumor bleeding during the procedure. Instead, they opted for a modified endotracheal tube and the insertion of a laryngeal mask airway (LMA). The anesthesia induction and maintenance proceeded smoothly, with stable intraoperative hemodynamics. The tumor was successfully resected and tracheal anastomosis was performed without any complications. CLINICAL DISCUSSION: The strategy adroitly evades the risk of bleeding and dislodgement due to tumor contact during the intubation process. In this case report, the anesthetic highlight is the employment of a reverse insertion technique for endotracheal intubation, facilitated by a sterile suction catheter and complemented by an innovative modification to the tracheal tube. CONCLUSION: For patients with thyroid cancer invading the upper segment of the trachea, and in whom rapid induction anesthesia is anticipated not to cause tumor collapse, the use of laryngeal mask airway combined with modified tracheal tube mechanical ventilation is both safe and feasible.

3.
Nat Biomed Eng ; 6(6): 683-705, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35361935

RESUMO

Peripheral neurons that sense glucose relay signals of glucose availability to integrative clusters of neurons in the brain. However, the roles of such signalling pathways in the maintenance of glucose homoeostasis and their contribution to disease are unknown. Here we show that the selective activation of the nerve plexus of the hepatic portal system via peripheral focused ultrasound stimulation (pFUS) improves glucose homoeostasis in mice and rats with insulin-resistant diabetes and in swine subject to hyperinsulinemic-euglycaemic clamps. pFUS modulated the activity of sensory projections to the hypothalamus, altered the concentrations of metabolism-regulating neurotransmitters, and enhanced glucose tolerance and utilization in the three species, whereas physical transection or chemical blocking of the liver-brain nerve pathway abolished the effect of pFUS on glucose tolerance. Longitudinal multi-omic profiling of metabolic tissues from the treated animals confirmed pFUS-induced modifications of key metabolic functions in liver, pancreas, muscle, adipose, kidney and intestinal tissues. Non-invasive ultrasound activation of afferent autonomic nerves may represent a non-pharmacologic therapy for the restoration of glucose homoeostasis in type-2 diabetes and other metabolic diseases.


Assuntos
Diabetes Mellitus Experimental , Glucose , Animais , Diabetes Mellitus Experimental/terapia , Glucose/metabolismo , Homeostase , Hipotálamo/metabolismo , Fígado/metabolismo , Camundongos , Ratos , Suínos
4.
Diabetes ; 68(1): 163-171, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30327383

RESUMO

The impact of glycemic variability on brain glucose transport kinetics among individuals with type 1 diabetes mellitus (T1DM) remains unclear. Fourteen individuals with T1DM (age 35 ± 4 years; BMI 26.0 ± 1.4 kg/m2; HbA1c 7.6 ± 0.3) and nine healthy control participants (age 32 ± 4; BMI 23.1 ± 0.8; HbA1c 5.0 ± 0.1) wore a continuous glucose monitor (Dexcom) to measure hypoglycemia, hyperglycemia, and glycemic variability for 5 days followed by 1H MRS scanning in the occipital lobe to measure the change in intracerebral glucose levels during a 2-h glucose clamp (target glucose concentration 220 mg/dL). Hyperglycemic clamps were also performed in a rat model of T1DM to assess regional differences in brain glucose transport and metabolism. Despite a similar change in plasma glucose levels during the hyperglycemic clamp, individuals with T1DM had significantly smaller increments in intracerebral glucose levels (P = 0.0002). Moreover, among individuals with T1DM, the change in brain glucose correlated positively with the lability index (r = 0.67, P = 0.006). Consistent with findings in humans, streptozotocin-treated rats had lower brain glucose levels in the cortex, hippocampus, and striatum compared with control rats. These findings that glycemic variability is associated with brain glucose levels highlight the need for future studies to investigate the impact of glycemic variability on brain glucose kinetics.


Assuntos
Encéfalo/metabolismo , Diabetes Mellitus Tipo 1/sangue , Diabetes Mellitus Tipo 1/metabolismo , Glucose/metabolismo , Adulto , Animais , Glicemia/metabolismo , Diabetes Mellitus Tipo 1/tratamento farmacológico , Feminino , Hemoglobinas Glicadas , Humanos , Hiperglicemia/sangue , Hiperglicemia/tratamento farmacológico , Hiperglicemia/metabolismo , Hipoglicemiantes/uso terapêutico , Masculino , Modelos Teóricos , Ratos , Ratos Sprague-Dawley
5.
Diabetes ; 67(7): 1401-1413, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29669745

RESUMO

Insulin-induced hypoglycemia in diabetes is associated with impaired glucagon secretion. In this study, we tested whether stimulation of GPR119, a G-protein-coupled receptor expressed in pancreatic islet as well as enteroendocrine cells and previously shown to stimulate insulin and incretin secretion, might enhance glucagon secretion during hypoglycemia. In the study, GPR119 agonists were applied to isolated islets or perfused pancreata to assess insulin and glucagon secretion during hypoglycemic or hyperglycemic conditions. Insulin infusion hypoglycemic clamps were performed with or without GPR119 agonist pretreatment to assess glucagon counterregulation in healthy and streptozotocin (STZ)-induced diabetic rats, including those exposed to recurrent bouts of insulin-induced hypoglycemia that leads to suppression of hypoglycemia-induced glucagon release. Hypoglycemic clamp studies were also conducted in GPR119 knockout (KO) mice to evaluate whether the pharmacological stimulatory actions of GPR119 agonists on glucagon secretion during hypoglycemia were an on-target effect. The results revealed that GPR119 agonist-treated pancreata or cultured islets had increased glucagon secretion during low glucose perfusion. In vivo, GPR119 agonists also significantly increased glucagon secretion during hypoglycemia in healthy and STZ-diabetic rats, a response that was absent in GPR119 KO mice. In addition, impaired glucagon counterregulatory responses were restored by a GPR119 agonist in STZ-diabetic rats that were exposed to antecedent bouts of hypoglycemia. Thus, GPR119 agonists have the ability to pharmacologically augment glucagon secretion, specifically in response to hypoglycemia in diabetic rodents. Whether this effect might serve to diminish the occurrence and severity of iatrogenic hypoglycemia during intensive insulin therapy in patients with diabetes remains to be established.


Assuntos
Glucagon/metabolismo , Hipoglicemia/induzido quimicamente , Hipoglicemia/metabolismo , Insulina/efeitos adversos , Receptores Acoplados a Proteínas G/agonistas , Adulto , Animais , Células Cultivadas , Diabetes Mellitus Experimental/tratamento farmacológico , Diabetes Mellitus Experimental/metabolismo , Teste de Tolerância a Glucose , Humanos , Hipoglicemiantes/efeitos adversos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pessoa de Meia-Idade , Ratos , Ratos Wistar , Receptores Acoplados a Proteínas G/genética , Estreptozocina , Adulto Jovem
6.
Am J Physiol Endocrinol Metab ; 309(12): E960-7, 2015 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-26506851

RESUMO

Leptin has been shown to diminish hyperglycemia via reduced glucagon secretion, although it can also enhance sympathoadrenal responses. However, whether leptin can also inhibit glucagon secretion during insulin-induced hypoglycemia or increase epinephrine during acute or recurrent hypoglycemia has not been examined. To test whether leptin acts in the brain to influence counterregulation, hyperinsulinemic hypoglycemic (∼45 mg/dl) clamps were performed on rats exposed to or not exposed to recurrent hypoglycemia (3 days, ∼40 mg/dl). Intracerebroventricular artificial cerebral spinal fluid or leptin was infused during the clamp. During acute hypoglycemia, leptin decreased glucagon responses by 51% but increased epinephrine and norepinephrine by 24 and 48%, respectively. After recurrent hypoglycemia, basal plasma leptin levels were undetectable. Subsequent brain leptin infusion during hypoglycemia paradoxically increased glucagon by 45% as well as epinephrine by 19%. In conclusion, leptin acts within the brain to diminish glucagon secretion during acute hypoglycemia but increases epinephrine, potentially limiting its detrimental effects during hypoglycemia. Exposure to recurrent hypoglycemia markedly suppresses plasma leptin, whereas exogenous brain leptin delivery enhances both glucagon and epinephrine release to subsequent hypoglycemia. These data suggest that recurrent hypoglycemia may diminish counterregulatory responses in part by reducing brain leptin action.


Assuntos
Encéfalo/metabolismo , Glucagon/metabolismo , Hipoglicemia/metabolismo , Leptina/metabolismo , Animais , Retroalimentação Fisiológica , Masculino , Ratos , Ratos Sprague-Dawley , Recidiva
7.
Diabetes ; 64(10): 3564-72, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26130763

RESUMO

The objective of this study was to determine whether the sodium-glucose transporter SGLT1 in the ventromedial hypothalamus (VMH) plays a role in glucose sensing and in regulating the counterregulatory response to hypoglycemia, and if so, whether knockdown of in the VMH can improve counterregulatory responses to hypoglycemia in diabetic rats or rats exposed to recurrent bouts of hypoglycemia (RH). Normal Sprague-Dawley rats as well as RH or streptozotocin (STZ)-diabetic rats received bilateral VMH microinjections of an adenoassociated viral vector containing either the SGLT1 short hairpin RNA (shRNA) or a scrambled RNA sequence. Subsequently, these rats underwent a hypoglycemic clamp to assess hormone responses. In a subgroup of rats, glucose kinetics was determined using tritiated glucose. The shRNA reduced VMH SGLT1 expression by 53% in nondiabetic rats, and this augmented glucagon and epinephrine responses and hepatic glucose production during hypoglycemia. Similarly, SGLT1 knockdown improved the glucagon and epinephrine responses in RH rats and restored the impaired epinephrine response to hypoglycemia in STZ-diabetic animals. These findings suggest that SGLT1 in the VMH plays a significant role in the detection and activation of counterregulatory responses to hypoglycemia. Inhibition of SGLT1 may offer a potential therapeutic target to diminish the risk of hypoglycemia in diabetes.


Assuntos
Diabetes Mellitus Experimental/metabolismo , Regulação da Expressão Gênica/fisiologia , Hipoglicemia/metabolismo , RNA Mensageiro/metabolismo , Transportador 1 de Glucose-Sódio/metabolismo , Núcleo Hipotalâmico Ventromedial/metabolismo , Animais , Glicemia , Masculino , Interferência de RNA , RNA Mensageiro/genética , RNA Interferente Pequeno , Ratos , Ratos Sprague-Dawley , Transportador 1 de Glucose-Sódio/genética
8.
Diabetes ; 60(5): 1582-9, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21411513

RESUMO

OBJECTIVE: Impaired glucose counterregulation during hypoglycemia is well documented in patients with type 1 diabetes; however, the molecular mechanisms underlying this defect remain uncertain. We reported that the inhibitory neurotransmitter γ-aminobutyric acid (GABA), in a crucial glucose-sensing region within the brain, the ventromedial hypothalamus (VMH), plays an important role in modulating the magnitude of the glucagon and epinephrine responses to hypoglycemia and investigated whether VMH GABAergic tone is altered in diabetes and therefore might contribute to defective counterregulatory responses. RESEARCH DESIGN AND METHODS: We used immunoblots to measure GAD(65) protein (a rate-limiting enzyme in GABA synthesis) and microdialysis to measure extracellular GABA levels in the VMH of two diabetic rat models, the diabetic BB rat and the streptozotocin (STZ)-induced diabetic rat, and compared them with nondiabetic controls. RESULTS: Both diabetic rat models exhibited an ~50% increase in GAD(65) protein as well as a twofold increase in VMH GABA levels compared with controls under baseline conditions. Moreover, during hypoglycemia, VMH GABA levels did not change in the diabetic animals, whereas they significantly declined in nondiabetic animals. As expected, glucagon responses were absent and epinephrine responses were attenuated in diabetic rats compared with their nondiabetic control counterparts. The defective counterregulatory response in STZ-diabetic animals was restored to normal with either local blockade of GABA(A) receptors or knockdown of GAD(65) in the VMH. CONCLUSIONS: These data suggest that increased VMH GABAergic inhibition is an important contributor to the absent glucagon response to hypoglycemia and the development of counterregulatory failure in type 1 diabetes.


Assuntos
Diabetes Mellitus Experimental/metabolismo , Hipoglicemia/metabolismo , Núcleo Hipotalâmico Ventromedial/metabolismo , Ácido gama-Aminobutírico/metabolismo , Animais , Técnica Clamp de Glucose , Glutamato Descarboxilase/genética , Glutamato Descarboxilase/metabolismo , Immunoblotting , Masculino , Microdiálise , Ratos , Ratos Sprague-Dawley
9.
Diabetes ; 59(10): 2646-52, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20627933

RESUMO

OBJECTIVE: To determine whether the medial amygdalar nucleus (MAN) represents a novel brain glucose-sensing region involved in the detection of hypoglycemia and generation of a counterregulatory hormone response. RESEARCH DESIGN AND METHODS: Fura-2 calcium imaging was used to assess glucose responsivity in neurons isolated from the MAN and single-cell real-time reverse transcription PCR used to examine gene expression within glucose-responsive neurons. In vivo studies with local MAN perfusion of the glucoprivic agent, 2-deoxyglucose (2-DG), under normal and hypoglycemic conditions and also after MAN lesioning with ibotenic acid, were used to examine the functional role of MAN glucose sensors. In addition, retrograde neuronal tracer studies were used to examine reciprocal pathways between the MAN and the ventromedial hypothalamus (VMH). RESULTS: The MAN contains a population of glucose-sensing neurons (13.5%), which express glucokinase, and the selective urocortin 3 (UCN3) receptor CRH-R2, but not UCN3 itself. Lesioning the MAN suppressed, whereas 2-DG infusion amplified, the counterregulatory response to hyperinsulinemic hypoglycemia in vivo. However, 2-DG infusion to the MAN or VMH under normoglycemic conditions had no systemic effect. The VMH is innervated by UCN3 neurons that arise mainly from the MAN, and ∼1/3 of MAN UCN3 neurons are active during mild hypoglycemia. CONCLUSIONS: The MAN represents a novel limbic glucose-sensing region that contains characteristic glucokinase-expressing glucose-sensing neurons that respond directly to manipulations of glucose availability both in vitro and in vivo. Moreover, UCN3 neurons may provide feedback inhibitory regulation of the counterregulatory response through actions within the VMH and the MAN.


Assuntos
Tonsila do Cerebelo/fisiologia , Homeostase/fisiologia , Hipoglicemia/fisiopatologia , Tonsila do Cerebelo/anatomia & histologia , Tonsila do Cerebelo/fisiopatologia , Animais , Conscientização , Cálcio/metabolismo , Artéria Carótida Interna/cirurgia , Diabetes Mellitus Tipo 1/fisiopatologia , Diabetes Mellitus Tipo 2/fisiopatologia , Agonistas de Aminoácidos Excitatórios/farmacologia , Regulação da Expressão Gênica , Glucoquinase/metabolismo , Glucose/farmacologia , Humanos , Hiperinsulinismo/fisiopatologia , Hipoglicemia/psicologia , Ácido Ibotênico/farmacologia , Veias Jugulares/cirurgia , Masculino , Neurônios/fisiologia , Reação em Cadeia da Polimerase , Ratos , Ratos Sprague-Dawley
10.
Diabetes ; 57(12): 3327-34, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18776135

RESUMO

OBJECTIVE: In glucose-sensing neurons, ATP-sensitive K(+) channels (K(ATP) channels) are thought to translate metabolic signals into an alteration in neuronal firing rates. Because these neurons express the Kir6.2/SUR-1 isoform of the K(ATP) channel, we sought to examine the therapeutic potential of the SUR-1-selective potassium channel opener (KCO), NN414, to amplify counterregulatory response to hypoglycemia. RESEARCH DESIGN AND METHODS: In vivo dose-response studies with NN414 delivered intravenously to normal Sprague-Dawley rats before the induction of controlled hypoglycemia were performed. Based on these studies, the potential for NN414 to restore counterregulatory responses in chronically cannulated nondiabetic and diabetic BB rats was explored using the in vivo hyperinsulinemic-hypoglycemic clamp technique. RESULTS: NN414 delivered systemically amplified epinephrine responses during acute hypoglycemia and showed a persisting effect to amplify the epinephrine response when given 24 h before the hypoglycemic study. Local delivery of a potassium-channel blocker to the ventromedial hypothalamus reversed the effects of systemic NN414. In addition, NN414 amplified the epinephrine response to hypoglycemia in both nondiabetic and diabetic BB rats with defective hormonal counterregulation. CONCLUSIONS: These studies demonstrate in a variety of rodent models that systemic delivery of Kir6.2/SUR-1-selective KCOs enhance the glucose counterregulatory response to insulin-induced hypoglycemia. Future studies in human subjects are now required to determine their potential as a therapy for hypoglycemia-associated autonomic failure in type 1 diabetes.


Assuntos
Transportadores de Cassetes de Ligação de ATP/fisiologia , Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Óxidos S-Cíclicos/farmacologia , Hipoglicemia/fisiopatologia , Canais de Potássio Corretores do Fluxo de Internalização/fisiologia , Receptores de Droga/fisiologia , Transportadores de Cassetes de Ligação de ATP/efeitos dos fármacos , Animais , Glicemia/efeitos dos fármacos , Glicemia/metabolismo , Diabetes Mellitus Tipo 1/complicações , Epinefrina/farmacologia , Homeostase/efeitos dos fármacos , Homeostase/fisiologia , Hipoglicemia/etiologia , Hipoglicemia/prevenção & controle , Canais de Potássio Corretores do Fluxo de Internalização/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Receptores de Droga/efeitos dos fármacos , Receptores de Sulfonilureias
11.
Diabetes ; 57(2): 444-50, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17977955

RESUMO

OBJECTIVE: To examine in vivo in a rodent model the potential role of AMP-activated protein kinase (AMPK) within the ventromedial hypothalamus (VMH) in glucose sensing during hypoglycemia. RESEARCH DESIGN AND METHODS: Using gene silencing technology to selectively downregulate AMPK in the VMH, a key hypothalamic glucose-sensing region, we demonstrate a key role for AMPK in the detection of hypoglycemia. In vivo hyperinsulinemic-hypoglycemic (50 mg dl(-1)) clamp studies were performed in awake, chronically catheterized Sprague-Dawley rats that had been microinjected bilaterally to the VMH with an adeno-associated viral (AAV) vector expressing a short hairpin RNA for AMPKalpha. RESULTS: In comparison with control studies, VMH AMPK downregulation resulted in suppressed glucagon ( approximately 60%) and epinephrine (approximately 40%) responses to acute hypoglycemia. Rats with VMH AMPK downregulation also required more exogenous glucose to maintain the hypoglycemia plateau and showed significant reductions in endogenous glucose production and whole-body glucose uptake. CONCLUSIONS: We conclude that AMPK in the VMH plays a key role in the detection of acute hypoglycemia and initiation of the glucose counterregulatory response.


Assuntos
Hipoglicemia/fisiopatologia , Complexos Multienzimáticos/genética , Proteínas Serina-Treonina Quinases/genética , Núcleo Hipotalâmico Ventromedial/enzimologia , Proteínas Quinases Ativadas por AMP , Animais , Sequência de Bases , Primers do DNA , Inativação Gênica , Vetores Genéticos , Técnica Clamp de Glucose , Homeostase , Hipoglicemia/enzimologia , Hipoglicemia/genética , Masculino , Microinjeções , Dados de Sequência Molecular , Complexos Multienzimáticos/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , RNA Mensageiro/genética , Ratos , Ratos Sprague-Dawley , Moldes Genéticos
12.
Diabetes ; 55(6): 1755-60, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16731839

RESUMO

Defective counterregulatory responses (CRRs) to hypoglycemia are associated with a marked increase in the risk of severe hypoglycemia. The mechanisms leading to the development of defective CRRs remain largely unknown, although they are associated with antecedent hypoglycemia. Activation of AMP-activated protein kinase (AMPK) in the ventromedial hypothalamus (VMH) amplifies the counterregulatory increase in glucose production during acute hypoglycemia. To examine whether activation of AMPK in the VMH restores defective CRR, controlled hypoglycemia ( approximately 2.8 mmol/l) was induced in a group of 24 Sprague-Dawley rats, all of which had undergone a 3-day model of recurrent hypoglycemia before the clamp study. Before the acute study, rats were microinjected to the VMH with either 5-aminoimidazole-4-carboxamide (AICAR; n=12), to activate AMPK, or saline (n=12). In a subset of rats, an infusion of H(3)-glucose was additionally started to calculate glucose turnover. Stimulation of AMPK within the VMH was found to amplify hormonal CRR and increase endogenous glucose production. In addition, analysis of tissue from both whole hypothalamus and VMH showed that recurrent hypoglycemia induces an increase in the gene expression of AMPK alpha(1) and alpha(2). These findings suggest that the development of novel drugs designed to selectively activate AMPK in the VMH offer a future therapeutic potential for individuals with type 1 diabetes who have defective CRRs to hypoglycemia.


Assuntos
Hormônios/fisiologia , Hipotálamo Médio/enzimologia , Complexos Multienzimáticos/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Quinases Ativadas por AMP , Aminoimidazol Carboxamida/administração & dosagem , Aminoimidazol Carboxamida/análogos & derivados , Aminoimidazol Carboxamida/farmacologia , Animais , Glicemia/metabolismo , Diabetes Mellitus Tipo 1/tratamento farmacológico , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/patologia , Ativação Enzimática/efeitos dos fármacos , Glucose/administração & dosagem , Glucose/metabolismo , Glucose/farmacologia , Hormônios/metabolismo , Hipoglicemia/tratamento farmacológico , Hipoglicemia/metabolismo , Hipoglicemia/patologia , Hipotálamo Médio/efeitos dos fármacos , Hipotálamo Médio/metabolismo , Masculino , Complexos Multienzimáticos/genética , Proteínas Serina-Treonina Quinases/genética , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Ribonucleosídeos/administração & dosagem , Ribonucleosídeos/farmacologia , Cloreto de Sódio/administração & dosagem , Cloreto de Sódio/farmacologia
13.
Diabetes ; 55(4): 1080-7, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16567532

RESUMO

Hypoglycemia provokes a multifaceted counterregulatory response involving the sympathoadrenal system, stimulation of glucagon secretion, and the hypothalamo-pituitary-adrenal axis that is commonly impaired in diabetes. We examined whether modulation of inhibitory input from gamma-aminobutyric acid (GABA) in the ventromedial hypothalamus (VMH), a major glucose-sensing region within the brain, plays a role in affecting counterregulatory responses to hypoglycemia. Normal Sprague-Dawley rats had carotid artery and jugular vein catheters chronically implanted, as well as bilateral steel microinjection guide cannulas inserted down to the level of the VMH. Seven to 10 days following surgery, the rats were microinjected with artificial extracellular fluid, the GABA(A) receptor agonist muscimol (1 nmol/side), or the GABA(A) receptor antagonist bicuculline methiodide (12.5 pmol/side) before being subjected to a hyperinsulinemic-hypoglycemic (2.5 mmol/l) glucose clamp for 90 min. Following VMH administration of bicuculline methiodide, glucose infusion rates were significantly suppressed, whereas muscimol raised glucose infusion rates significantly compared with controls. Glucagon and epinephrine responses were elevated with the antagonist and suppressed with the agonist compared with controls. Corticosterone responses, however, were unaffected by either administration of the agonist or antagonist into the VMH. These data demonstrate that modulation of the GABAergic system in the VMH alters both glucagon and sympathoadrenal, but not corticosterone, responses to hypoglycemia. Our findings are consistent with the hypothesis that GABAergic inhibitory tone within the VMH can modulate glucose counterregulatory responses.


Assuntos
Antagonistas GABAérgicos/farmacologia , Glucagon/metabolismo , Hipoglicemia/fisiopatologia , Sistema Hipotálamo-Hipofisário/fisiologia , Sistema Hipófise-Suprarrenal/fisiologia , Receptores de GABA-A/fisiologia , Núcleo Hipotalâmico Ventromedial/fisiologia , Animais , Bicuculina/farmacologia , Glicemia/efeitos dos fármacos , Glicemia/metabolismo , Corticosterona/sangue , Epinefrina/sangue , Técnica Clamp de Glucose , Sistema Hipotálamo-Hipofisário/efeitos dos fármacos , Cinética , Masculino , Muscimol/farmacologia , Norepinefrina/sangue , Sistema Hipófise-Suprarrenal/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Núcleo Hipotalâmico Ventromedial/efeitos dos fármacos
14.
Diabetes ; 54(11): 3169-74, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16249441

RESUMO

The mechanism(s) by which glucosensing neurons detect fluctuations in glucose remains largely unknown. In the pancreatic beta-cell, ATP-sensitive K+ channels (K ATP channels) play a key role in glucosensing by providing a link between neuronal metabolism and membrane potential. The present study was designed to determine in vivo whether the pharmacological opening of ventromedial hypothalamic K ATP channels during systemic hypoglycemia would amplify hormonal counterregulatory responses in normal rats and those with defective counterregulation arising from prior recurrent hypoglycemia. Controlled hypoglycemia (approximately 2.8 mmol/l) was induced in vivo using a hyperinsulinemic (20 mU x kg(-1) x min(-1)) glucose clamp technique in unrestrained, overnight-fasted, chronically catheterized Sprague-Dawley rats. Immediately before the induction of hypoglycemia, the rats received bilateral ventromedial hypothalamic microinjections of either the potassium channel openers (KCOs) diazoxide and NN414 or their respective controls. In normal rats, both KCOs amplified epinephrine and glucagon counterregulatory responses to hypoglycemia. Moreover, diazoxide also amplified the counterregulatory responses in a rat model of defective hormonal counterregulation. Taken together, our data suggest that the K ATP channel plays a key role in vivo within glucosensing neurons in the ventromedial hypothalamus in the detection of incipient hypoglycemia and the initiation of protective counterregulatory responses. We also conclude that KCOs may offer a future potential therapeutic option for individuals with insulin-treated diabetes who develop defective counterregulation.


Assuntos
Hipoglicemia/fisiopatologia , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , Núcleo Hipotalâmico Ventromedial/efeitos dos fármacos , Núcleo Hipotalâmico Ventromedial/metabolismo , Animais , Glicemia , Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Carbonatos/farmacologia , Óxidos S-Cíclicos/farmacologia , Diazóxido/farmacologia , Epinefrina/metabolismo , Glucagon/metabolismo , Glucose/administração & dosagem , Masculino , Potássio/farmacologia , Ratos , Ratos Sprague-Dawley
15.
Diabetes ; 53(8): 1953-8, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15277372

RESUMO

The mechanisms by which specialized glucose-sensing neurons within the hypothalamus are able to detect a falling blood glucose remain largely unknown but may be linked to some gauge of neuronal energy status. We sought to test the hypothesis that AMP-activated protein kinase (AMPK), an intracellular kinase purported to act as a fuel sensor, plays a role in hypoglycemia sensing in the ventromedial hypothalamus (VMH) of the Sprague-Dawley rat by chemically activating AMPK in vivo through bilateral microinjection, before performing hyperinsulinemic-hypoglycemic or hyperinsulinemic-euglycemic clamp studies. In a subgroup of rats, H3-glucose was infused to determine glucose kinetics. The additional chemical activation by AICAR of AMPK in the VMH during hypoglycemia markedly reduced the amount of exogenous glucose required to maintain plasma glucose during hypoglycemia, an effect that was almost completely accounted for by a three- to fourfold increase in hepatic glucose production in comparison to controls. In contrast, no differences were seen between groups in hypoglycemia-induced rises in the principal counterregulatory hormones. In conclusion, activation of AMPK within the VMH may play an important role in hypoglycemia sensing. The combination of hypoglycemia- and AICAR-induced AMPK activity appears to result in a marked stimulus to hepatic glucose counterregulation.


Assuntos
Aminoimidazol Carboxamida/análogos & derivados , Glicemia/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/fisiologia , Hipoglicemia/fisiopatologia , Neurônios/fisiologia , Núcleo Hipotalâmico Ventromedial/fisiologia , Aminoimidazol Carboxamida/administração & dosagem , Aminoimidazol Carboxamida/farmacologia , Animais , Glicemia/efeitos dos fármacos , Epinefrina/sangue , Glucagon/sangue , Técnica Clamp de Glucose , Hiperinsulinismo , Infusões Intravenosas , Insulina/administração & dosagem , Insulina/farmacologia , Masculino , Microinjeções , Neurônios/efeitos dos fármacos , Norepinefrina/sangue , Ratos , Ratos Sprague-Dawley , Ribonucleotídeos/administração & dosagem , Ribonucleotídeos/farmacologia , Núcleo Hipotalâmico Ventromedial/efeitos dos fármacos
16.
Nucleic Acids Res ; 30(20): 4481-8, 2002 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-12384595

RESUMO

Previous studies have shown that human dihydrofolate reductase (DHFR) acts as an RNA-binding protein, in which it binds to its own mRNA and, in so doing, results in translational repression. In this study, we used RNA gel mobility shift and nitrocellulose filter-binding assays to further investigate the specificity of the interaction between human DHFR protein and human DHFR mRNA. Site-directed mutagenesis was used to identify the critical amino acid residues on DHFR protein required for RNA recognition. Human His-Tag DHFR protein specifically binds to human DHFR mRNA, while unrelated proteins including thymidylate synthase, p53 and glutathione-S-transferase were unable to form a ribonucleoprotein complex with DHFR mRNA. The Cys6 residue is essential for RNA recognition, as mutation at this amino acid with either an alanine (C6A) or serine (C6S) residue almost completely abrogated RNA-binding activity. Neither one of the cysteine mutant proteins was able to repress the in vitro translation of human DHFR mRNA. Mutations at amino acids Ile7, Arg28 and Phe34, significantly reduced RNA-binding activity. An RNA footprinting analysis identified three different RNA sequences, bound to DHFR protein, ranging in size from 16 to 45 nt, while a UV cross-linking analysis isolated an approximately 16 nt RNA sequence bound to DHFR. These studies begin to identify the critical amino acid residues on human DHFR that mediate RNA binding either through forming direct contact points with RNA or through maintaining the protein in an optimal structure that allows for the critical RNA-binding domain to be accessible.


Assuntos
Aminoácidos/análise , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/química , Proteínas de Ligação a RNA/metabolismo , Tetra-Hidrofolato Desidrogenase/química , Tetra-Hidrofolato Desidrogenase/metabolismo , Sítios de Ligação , Humanos , Substâncias Macromoleculares , Peso Molecular , Biossíntese de Proteínas , Proteínas de Ligação a RNA/genética , Ribonucleoproteínas/metabolismo , Tetra-Hidrofolato Desidrogenase/genética , Raios Ultravioleta
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...