Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 541
Filtrar
1.
Pharmacol Rev ; 76(3): 358-387, 2024 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-38697858

RESUMO

G-protein coupled receptors (GPCRs) transduce a wide range of extracellular signals. They are key players in the majority of biologic functions including vision, olfaction, chemotaxis, and immunity. However, as essential as most of them are to body function and homeostasis, overactivation of GPCRs has been implicated in many pathologic diseases such as cancer, asthma, and heart failure (HF). Therefore, an important feature of G protein signaling systems is the ability to control GPCR responsiveness, and one key process to control overstimulation involves initiating receptor desensitization. A number of steps are appreciated in the desensitization process, including cell surface receptor phosphorylation, internalization, and downregulation. Rapid or short-term desensitization occurs within minutes and involves receptor phosphorylation via the action of intracellular protein kinases, the binding of ß-arrestins, and the consequent uncoupling of GPCRs from their cognate heterotrimeric G proteins. On the other hand, long-term desensitization occurs over hours to days and involves receptor downregulation or a decrease in cell surface receptor protein level. Of the proteins involved in this biologic phenomenon, ß-arrestins play a particularly significant role in both short- and long-term desensitization mechanisms. In addition, ß-arrestins are involved in the phenomenon of biased agonism, where the biased ligand preferentially activates one of several downstream signaling pathways, leading to altered cellular responses. In this context, this review discusses the different patterns of desensitization of the α 1-, α 2- and the ß adrenoceptors and highlights the role of ß-arrestins in regulating physiologic responsiveness through desensitization and biased agonism. SIGNIFICANCE STATEMENT: A sophisticated network of proteins orchestrates the molecular regulation of GPCR activity. Adrenoceptors are GPCRs that play vast roles in many physiological processes. Without tightly controlled desensitization of these receptors, homeostatic imbalance may ensue, thus precipitating various diseases. Here, we critically appraise the mechanisms implicated in adrenoceptor desensitization. A better understanding of these mechanisms helps identify new druggable targets within the GPCR desensitization machinery and opens exciting therapeutic fronts in the treatment of several pathologies.


Assuntos
Transdução de Sinais , Humanos , Animais , Receptores Adrenérgicos/metabolismo , Receptores Adrenérgicos/fisiologia , Receptores Acoplados a Proteínas G/metabolismo , Receptores Acoplados a Proteínas G/fisiologia , beta-Arrestinas/metabolismo
2.
J Craniofac Surg ; 32(2): 783-786, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33705036

RESUMO

ABSTRACT: This study aimed to investigate the effects of systemic propranol on the osseointegration of titanium implants. After the surgical insertion of titanium implants into the metaphyseal part of the tibial bone, the rats were randomly divided into 2 equal groups: the control (CNT) (n = 10) and propranol group (P) (n = 10); CNT: Rats received no further treatment during the 4 week experimental period after surgery. Rats received 10 mg/kg propranol in every day during the 4 week experimental period in PRP group after the surgical insertion of the implants. After the experimental period, the rats were euthanized, blood serum were collected to biochemical analysis and the implants and surrounding bone tissues were used for the histopathologic analysis. To analysis of the data between tests and controls student T test was used. There were no significant differences in the biochemical parameters (alcaline phosphatase, calcium, phosphor) of the groups (P > 0.05). Bone implant connection (BIC) ratios was detected higher in test animals compared with the controls (P < 0.05). Systemic propranolol may increases titanium implant osseointegration.


Assuntos
Osseointegração , Propranolol , Próteses e Implantes , Receptores Adrenérgicos/fisiologia , Titânio , Animais , Propranolol/farmacologia , Ratos , Tíbia/cirurgia
3.
Ann Endocrinol (Paris) ; 82(3-4): 193-197, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-32473788

RESUMO

Activation of the sympathetic nervous system is responsible for the body's "fight or flight" reaction. The physiological responses to the activation of the sympathetic nervous system and adrenal medulla are mediated through the action of the endogenous catecholamines norepinephrine (or noradrenaline) and epinephrine (or adrenaline) on adrenergic receptors. Adrenergic receptors belong to the superfamily of G protein-coupled receptors (GPCR). Adrenoceptors are divided into alpha1, alpha2, beta1, beta2 and beta3 receptors. Norepinephrine stimulates both subtypes of α receptors and ß1 receptors. Epinephrine stimulates all subtypes ofα and ß adrenoreceptors. α1 adrenergic receptors, coupled to stimulatory Gq proteins, activate the enzyme phospholipase C and are mainly found in the smooth muscle cells of blood vessels and urinary tract, where they induce constriction. α2 receptors are coupled to inhibitory Gi proteins, that inactivate adenylyl cyclase, decreasing cyclic adenosine monophosphate (AMP) production. They are mainly found in the central nervous system, where their activation results in a decreased arterial blood pressure. ß1 adrenoreceptors predominate in the heart, activate the Gs-adenylyl cyclase -cAMP-protein kinase A signaling cascade, and induce positive inotropic and chronotropic effects. ß2 adrenoreceptors are distributed extensively throughout the body, but are expressed predominantly in bronchial smooth muscle cells. ß2 adrenergic receptors activate adenylyl cyclase, dilate blood vessels and bronchioles, relax the muscles of the uterus, bladder and gastrointestinal duct, and also decrease platelet aggregation and glycogenolysis. ß3 receptors can couple interchangeably to both stimulating and inhibiting G proteins. They are abundantly expressed in white and brown adipose tissue, and increase fat oxidation, energy expenditure and insulin-mediated glucose uptake. This review details the regulation of cardiac and vascular function by adrenergic receptors.


Assuntos
Sistema Cardiovascular/efeitos dos fármacos , Catecolaminas/farmacologia , Receptores Adrenérgicos/fisiologia , Animais , Fenômenos Fisiológicos Cardiovasculares/efeitos dos fármacos , Sistema Cardiovascular/fisiopatologia , Feminino , Coração/efeitos dos fármacos , Coração/fisiologia , Humanos , Masculino
4.
Sheng Li Xue Bao ; 72(2): 227-234, 2020 Apr 25.
Artigo em Chinês | MEDLINE | ID: mdl-32328616

RESUMO

Adrenergic receptor (AR), one of the key receptors for nervous system, plays an important role in the immune microenvironment and the progression of many diseases. In recent years, the regulation of ARs and its signal on macrophages has become a research hotspot. Researchers found that ARs could exert different regulatory functions on macrophages in different microenvironments, which in turn affects occurrence and development of diseases such as tumor, heart failure, obesity, acute injury, infection and pregnancy-related diseases. This review summarizes the expression and functional regulation of ARs on macrophages, and the role of ARs in microenvironment of related diseases, which might provide new ideas for the treatments.


Assuntos
Doença , Macrófagos/fisiologia , Receptores Adrenérgicos/fisiologia , Transdução de Sinais , Humanos
5.
Sheng Li Xue Bao ; 72(2): 235-242, 2020 Apr 25.
Artigo em Chinês | MEDLINE | ID: mdl-32328617

RESUMO

Gliomas are malignant tumors with strong invasiveness. The current treatment strategy is surgical treatment assisted by a variety of radiotherapies, chemotherapies and immunotherapies. However, the curative efficacy is limited. Adrenergic receptor (AR) is an important stress hormone receptor, which is highly involved in the regulation of the tumorigenesis and progression of various tumors by activating different downstream signal transduction pathways. Recent studies have shown that AR is dysregulated in glioma cells and tissues, and plays an important role in a series of biological behaviors such as tumorigenesis, invasion and metastasis of glioma. This article reviews the research progress of AR in the field of glioma in recent years, which provides a theoretical basis for the prevention and treatment of glioma targeting the AR.


Assuntos
Neoplasias Encefálicas/patologia , Glioma/patologia , Receptores Adrenérgicos/fisiologia , Transdução de Sinais , Carcinogênese , Linhagem Celular Tumoral , Proliferação de Células , Regulação Neoplásica da Expressão Gênica , Humanos , Invasividade Neoplásica , Metástase Neoplásica
6.
Mol Metab ; 37: 100986, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32247064

RESUMO

OBJECTIVE: 3',5'-Cyclic adenosine monophosphate (cAMP) is a central second messenger governing brown adipocyte differentiation and function. ß-adrenergic receptors (ß-ARs) stimulate adenylate cyclases which produce cAMP. Moreover, cyclic nucleotide levels are tightly controlled by phosphodiesterases (PDEs), which can generate subcellular microdomains of cAMP. Since the spatio-temporal organisation of the cAMP signalling pathway in adipocytes is still unclear, we sought to monitor real-time cAMP dynamics by live cell imaging in pre-mature and mature brown adipocytes. METHODS: We measured the real-time dynamics of cAMP in murine pre-mature and mature brown adipocytes during stimulation of individual ß-AR subtypes, as well as its regulation by PDEs using a Förster Resonance Energy Transfer based biosensor and pharmacological tools. We also correlated these data with ß-AR stimulated lipolysis and analysed the expression of ß-ARs and PDEs in brown adipocytes using qPCR and immunoblotting. Furthermore, subcellular distribution of PDEs was studied using cell fractionation and immunoblots. RESULTS: Using pre-mature and mature brown adipocytes isolated from transgenic mice expressing a highly sensitive cytosolic biosensor Epac1-camps, we established real-time measurements of cAMP responses. PDE4 turned out to be the major PDE regulating cytosolic cAMP in brown preadipocytes. Upon maturation, PDE3 gets upregulated and contributes with PDE4 to control ß1-AR-induced cAMP. Unexpectedly, ß3-AR initiated cAMP is resistant to increased PDE3 protein levels and simultaneously, the control of this microdomain by PDE4 is reduced upon brown adipocyte maturation. Therefore we postulate the existence of distinct cAMP pools in brown adipocytes. One cAMP pool is formed by ß1-AR associated with PDE3 and PDE4, while another pool is centred around ß3-AR and is much less controlled by these PDEs. Functionally, lower control of ß3-AR initiated cAMP by PDE3 and PDE4 facilitates brown adipocyte lipolysis, while lipolysis activated by ß1-AR and is under tight control of PDE3 and PDE4. CONCLUSIONS: We have established a real-time live cell imaging approach to analyse brown adipocyte cAMP dynamics in real-time using a cAMP biosensor. We showed that during the differentiation from pre-mature to mature murine brown adipocytes, there was a change in PDE-dependent compartmentation of ß1-and ß3-AR-initiated cAMP responses by PDE3 and PDE4 regulating lipolysis.


Assuntos
Adipócitos Marrons/metabolismo , AMP Cíclico/metabolismo , Receptores Adrenérgicos/fisiologia , Animais , Diferenciação Celular/fisiologia , AMP Cíclico/análise , Feminino , Humanos , Lipólise/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Receptores Adrenérgicos/metabolismo , Receptores Adrenérgicos beta 1/metabolismo , Receptores Adrenérgicos beta 1/fisiologia , Receptores Adrenérgicos beta 3/metabolismo , Receptores Adrenérgicos beta 3/fisiologia , Sistemas do Segundo Mensageiro , Transdução de Sinais/fisiologia
7.
Nat Commun ; 11(1): 471, 2020 01 24.
Artigo em Inglês | MEDLINE | ID: mdl-31980655

RESUMO

Astrocytes may function as mediators of the impact of noradrenaline on neuronal function. Activation of glial α1-adrenergic receptors triggers rapid astrocytic Ca2+ elevation and facilitates synaptic plasticity, while activation of ß-adrenergic receptors elevates cAMP levels and modulates memory consolidation. However, the dynamics of these processes in behaving mice remain unexplored, as do the interactions between the distinct second messenger pathways. Here we simultaneously monitored astrocytic Ca2+ and cAMP and demonstrate that astrocytic second messengers are regulated in a temporally distinct manner. In behaving mice, we found that while an abrupt facial air puff triggered transient increases in noradrenaline release and large cytosolic astrocytic Ca2+ elevations, cAMP changes were not detectable. By contrast, repeated aversive stimuli that lead to prolonged periods of vigilance were accompanied by robust noradrenergic axonal activity and gradual sustained cAMP increases. Our findings suggest distinct astrocytic signaling pathways can integrate noradrenergic activity during vigilance states to mediate distinct functions supporting memory.


Assuntos
Nível de Alerta/fisiologia , Astrócitos/fisiologia , Norepinefrina/fisiologia , Sistemas do Segundo Mensageiro/fisiologia , Animais , Sinalização do Cálcio/fisiologia , Condicionamento Clássico/fisiologia , AMP Cíclico/metabolismo , Medo/fisiologia , Corantes Fluorescentes , Locus Cerúleo/citologia , Locus Cerúleo/fisiologia , Memória/fisiologia , Camundongos , Plasticidade Neuronal/fisiologia , Lobo Parietal/citologia , Lobo Parietal/fisiologia , Receptores Adrenérgicos/fisiologia
8.
Arch Oral Biol ; 110: 104590, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-31743801

RESUMO

OBJECTIVE: To investigate the influence of chronic stress and adrenergic blockade in a rat model of apical periodontitis. METHODS: Thirty-two Wistar rats were submitted to an animal model of periapical lesion and randomly divided into 4 groups (n = 8): no stress (NS); stress + saline solution (SS); stress + ß-adrenergic blocker (Sß); stress + α-adrenergic blocker (Sα). The SS, Sß and Sα groups were submitted to an animal model of chronic stress for 28 days and received daily injections of saline solution, propranolol (ß adrenergic blocker) and phentolamine (α adrenergic blocker), respectively. After 28 days the animals were euthanized and the following analyses were carried out: a) serum corticosterone levels through Radioimmunoassay; b) measurement of serum levels of IL-1B, IL-6, IL-10 and IL-17 by enzyme-linked immunosorbent assay (ELISA); c) volume of periapical bone resorption by micro-computed tomography; d) histomorphometric analysis by staining with hematoxylin and eosin; e) expression of ß-AR, α-AR, receptor activator of nuclear factor kappa-B ligand (RANKL) and osteoprotegerin (OPG) by immunohistochemistry; f) tartrate-resistant acid phosphatase (TRAP) staining; g) ex-vivo cytokine release followed by the stimulation with LPS in superfusion system, by ELISA. RESULTS: SS group displayed significantly higher corticosterone levels than NS group (non-stressed). Higher IL-1ß serum level was observed in the NS group (p < .05); compared to all stressed groups. Other cytokines were present in similar amounts in the serum of all groups. All groups presented similar periapical lesions. All groups presented moderate inflammatory infiltrate, without statistically significant differences between them. No differences were observed regarding ß-AR, α-AR, Rank-L and OPG expression. The number of TRAP-positive cells was significantly decreased in the groups that received daily injections of adrenergic blockers. The IL-1ß release followed LPS stimulation was significantly suppressed when the superfusion media contained propranolol (p < .05). Perfusion containing phentolamine induced a greater release of IL-10. TGF-ß was significantly suppressed by phentolamine perfusion in the NS group (p < .05). CONCLUSIONS: Chronic stress can significantly change the inflammatory cytokines release. Rank-L/OPG system and periapical lesion volume were not affected following the current method applied. The administration of adrenergic blockers was not able to modulate the inflammatory response but presented effectivity in reducing the number of osteoclasts in the periapical region.


Assuntos
Reabsorção Óssea , Inflamação , Periodontite Periapical , Receptores Adrenérgicos , Estresse Fisiológico , Adrenérgicos , Animais , Osteoprotegerina , Estresse Oxidativo , Periodontite Periapical/fisiopatologia , Ligante RANK , Ratos , Ratos Wistar , Receptores Adrenérgicos/fisiologia , Microtomografia por Raio-X
9.
Artigo em Inglês, Espanhol | MEDLINE | ID: mdl-31031167

RESUMO

Imaging of cardiac sympathetic innervation is only possible by nuclear cardiology techniques and its assessment is key in the evaluation of and decision-making for patients with cardiac sympathetic impairment. This review includes the basis of cardiac sympathetic scintigraphy with 123I-meta-iodobenzylguanidine (123I-MIBG), recommended protocols, patient preparation, image acquisition and quantification, reproducibility, dosimetry, etc., and also the clinical indications for cardiac patients, mainly with regard to heart failure, arrhythmia, coronary artery disease, cardiotoxicity, including its contribution to establishing the indication for and monitoring the response to implantable cardiac devices, pharmacological treatment, heart transplantation and other.


Assuntos
3-Iodobenzilguanidina , Sistema de Condução Cardíaco/diagnóstico por imagem , Cardiopatias/diagnóstico por imagem , Coração/inervação , Radioisótopos do Iodo , Compostos Radiofarmacêuticos , Sistema Nervoso Simpático/diagnóstico por imagem , 3-Iodobenzilguanidina/administração & dosagem , 3-Iodobenzilguanidina/farmacocinética , Dispositivos de Terapia de Ressincronização Cardíaca , Pré-Escolar , Tomada de Decisão Clínica , Desfibriladores Implantáveis , Coração/diagnóstico por imagem , Sistema de Condução Cardíaco/fisiopatologia , Cardiopatias/induzido quimicamente , Cardiopatias/fisiopatologia , Transplante de Coração , Humanos , Processamento de Imagem Assistida por Computador , Infusões Intravenosas , Radioisótopos do Iodo/administração & dosagem , Radioisótopos do Iodo/farmacocinética , Norepinefrina/fisiologia , Percloratos/administração & dosagem , Compostos de Potássio/administração & dosagem , Prognóstico , Radiometria , Compostos Radiofarmacêuticos/administração & dosagem , Compostos Radiofarmacêuticos/farmacocinética , Receptores Adrenérgicos/fisiologia , Reprodutibilidade dos Testes , Sistema Nervoso Simpático/fisiopatologia , Glândula Tireoide/efeitos dos fármacos , Resultado do Tratamento
10.
J Trauma Acute Care Surg ; 80(6): 972-6, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26958793

RESUMO

BACKGROUND: Hemorrhagic shock results in ileal mucosa damage and intestinal bacterial translocation. Additionally, during hemorrhagic shock, norepinephrine levels increase. Past research has shown that the QseC sensor kinase of Escherichia coli modulates the quorum-sensing response to epinephrine and norepinephrine. Therefore, the aim of our study was to examine whether the absence of the ability of E. coli to sense epinephrine/norepinephrine would attenuate the bacterial translocation to extraintestinal organs in a rat model of hemorrhagic shock. METHODS: An E. coli MC1000 qseC mutant was constructed, and E. coli MC1000 and MC1000ΔqseC with streptomycin resistance were used to track bacterial translocation after gavage in rats. A rat model of nonlethal hemorrhagic shock was used. The rats were divided into six groups: controls (SS), rats that received a sham shock and MC1000 (M-SS), rats that received a sham shock and MC1000ΔqseC (Δ-SS), rats that received a hemorrhagic shock alone (HS), rats that received a hemorrhagic shock and MC1000 (M-HS), and rats that received a hemorrhagic shock and MC1000ΔqseC (Δ-HS). RESULTS: We found the incidence of bacterial translocation in the M-HS rats was higher than in the Δ-HS rats. The observed effects seem to be largely dependent on the inability to sense epinephrine/norepinephrine and the decreased motility of E. coli MC1000ΔqseC. CONCLUSION: Therefore, a role for E. coli sensing epinephrine/norepinephrine in the pathophysiology of bacterial translocation following hemorrhagic shock is proposed. The demonstration of such an effect would suggest a new mechanism for the development of shock-induced sepsis.


Assuntos
Translocação Bacteriana , Proteínas de Escherichia coli/fisiologia , Receptores Adrenérgicos/fisiologia , Choque Hemorrágico/fisiopatologia , Animais , Modelos Animais de Doenças , Masculino , Plasmídeos , Reação em Cadeia da Polimerase , Ratos , Ratos Sprague-Dawley
11.
Mol Neurobiol ; 53(1): 500-517, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25482049

RESUMO

Calcium-independent phospholipase A2 (iPLA2) is an 85-kDa enzyme that releases docosahexaenoic acid (DHA) from glycerophospholipids. DHA can be metabolized to resolvins and neuroprotectins that have anti-inflammatory properties and effects on neural plasticity. Recent studies show an important role of prefrontal cortical iPLA2 in hippocampo-prefrontal cortical LTP and antidepressant-like effect of the norepinephrine reuptake inhibitor (NRI) antidepressant, maprotiline. In this study, we elucidated the cellular mechanisms through which stimulation of adrenergic receptors could lead to increased iPLA2 expression. Treatment of SH-SY5Y neuroblastoma cells with maprotiline, another tricyclic antidepressant with noradrenaline reuptake inhibiting properties, nortriptyline, and the adrenergic receptor agonist, phenylephrine, resulted in increased iPLA2ß mRNA expression. This increase was blocked by inhibitors to alpha-1 adrenergic receptor, mitogen-activated protein (MAP) kinase or extracellular signal-regulated kinase (ERK) 1/2, and sterol regulatory element-binding protein (SREBP). Maprotiline and phenylephrine induced binding of SREBP-2 to sterol regulatory element (SRE) region on the iPLA2 promoter, as determined by electrophoretic mobility shift assay (EMSA). Together, results indicate that stimulation of adrenoreceptors causes increased iPLA2 expression via MAP kinase/ERK 1/2 and SREBP, and suggest a possible mechanism for effect of CNS noradrenaline on neural plasticity and crosstalk between sterol and glycerophospholipid mediators, that may play a role in physiological or pathophysiological processes in the brain and other organs.


Assuntos
Glicerofosfolipídeos/fisiologia , Fosfolipases A2 do Grupo VI/biossíntese , Receptor Cross-Talk/fisiologia , Receptores Adrenérgicos/fisiologia , Proteína de Ligação a Elemento Regulador de Esterol 2/metabolismo , Agonistas Adrenérgicos/farmacologia , Antagonistas de Receptores Adrenérgicos alfa 1/farmacologia , Antidepressivos/farmacologia , Linhagem Celular Tumoral , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/fisiologia , Maprotilina/farmacologia , Receptor Cross-Talk/efeitos dos fármacos , Esteróis/metabolismo
12.
Biochem Biophys Res Commun ; 460(4): 1053-8, 2015 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-25843795

RESUMO

Adrenaline reacts with three types of adrenergic receptors, α1, α2 and ß-adrenergic receptors (ARs), inducing many physiological events including exocytosis. Although adrenaline has been shown to induce glucagon-like peptide-1 (GLP-1) secretion from intestinal L cells, the precise molecular mechanism by which adrenaline regulates GLP-1 secretion remains unknown. Here we show by live cell imaging that all types of adrenergic receptors are stimulated by adrenaline in enteroendocrine L cell line GLUTag cells and are involved in GLP-1 exocytosis. We performed RT-PCR analysis and found that α1B-, α2A-, α2B-, and ß1-ARs were expressed in GLUTag cells. Application of adrenaline induced a significant increase of intracellular Ca(2+) and cAMP concentration ([Ca(2+)]i and [cAMP]i, respectively), and GLP-1 exocytosis in GLUTag cells. Blockade of α1-AR inhibited adrenaline-induced [Ca(2+)]i increase and exocytosis but not [cAMP]i increase, while blockade of ß1-AR inhibited adrenaline-induced [cAMP]i increase and exocytosis but not [Ca(2+)]i increase. Furthermore, overexpression of α2A-AR suppressed the adrenaline-induced [cAMP]i increase and exocytosis. These results suggest that the fine-turning of GLP-1 secretion from enteroendocrine L cells is established by the balance between α1-, α2-, and ß-ARs activation.


Assuntos
Células Enteroendócrinas/metabolismo , Epinefrina/metabolismo , Exocitose , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Receptores Adrenérgicos/fisiologia , Animais , Sequência de Bases , Cálcio/metabolismo , Linhagem Celular , AMP Cíclico/metabolismo , Primers do DNA , Células Enteroendócrinas/citologia , Camundongos , Microscopia de Fluorescência , Reação em Cadeia da Polimerase , Receptores Adrenérgicos/metabolismo
13.
Nephron Clin Pract ; 127(1-4): 153-7, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25343841

RESUMO

Acute kidney injury (AKI) is an increasing medical burden and is independently associated with mortality. AKI is a common comorbidity in the intensive care unit (ICU), with sepsis-associated AKI seen in almost a quarter of all ICU patients. Due to the high mortality seen in these patients, improved therapeutic options are needed. Data from experimental studies in animals support observations in humans that the host immune response to sepsis and trauma contributes to multiorgan failure and the high morbidity and mortality seen in critically ill patients. The spleen, a major component of the reticuloendothelial system, appears to be a key player in the 'cytokine storm' that develops after infection and trauma, and the resultant systemic inflammation is regulated by the autonomic nervous system. Over the past decade, evidence has suggested that controlling the splenic cytokine response improves tissue function and mortality in sepsis and other inflammatory-mediated diseases. One pathway that controls the response of the spleen to sepsis and trauma is the cholinergic anti-inflammatory pathway, and it may provide a key target for therapeutic intervention. Here, we review this concept and highlight the potential use of ultrasound to stimulate the cholinergic anti-inflammatory pathway and reduce systemic inflammation and disease severity.


Assuntos
Injúria Renal Aguda/imunologia , Estado Terminal , Baço/imunologia , Injúria Renal Aguda/etiologia , Injúria Renal Aguda/fisiopatologia , Injúria Renal Aguda/terapia , Transferência Adotiva , Animais , Sistema Nervoso Autônomo/imunologia , Sistema Nervoso Autônomo/fisiopatologia , Linfócitos T CD4-Positivos/imunologia , Fibras Colinérgicas/diagnóstico por imagem , Fibras Colinérgicas/fisiologia , Cuidados Críticos , Citocinas/fisiologia , Humanos , Inflamação , Sistema Fagocitário Mononuclear/imunologia , Sistema Fagocitário Mononuclear/fisiopatologia , Neuroimunomodulação/fisiologia , Norepinefrina/metabolismo , Fagócitos/imunologia , Receptores Adrenérgicos/fisiologia , Sepse/complicações , Sepse/imunologia , Baço/fisiopatologia , Esplenectomia , Síndrome de Resposta Inflamatória Sistêmica/imunologia , Síndrome de Resposta Inflamatória Sistêmica/fisiopatologia , Fator de Necrose Tumoral alfa/fisiologia , Terapia por Ultrassom , Ultrassonografia
14.
Zhonghua Nan Ke Xue ; 20(4): 372-6, 2014 Apr.
Artigo em Chinês | MEDLINE | ID: mdl-24873168

RESUMO

Adrenergic receptors are members of the G-protein coupled receptor superfamily. Recent studies revealed that these adrenergic receptors are playing an important role in the growth and metastasis of prostate cancer cells. The expression of adrenergic receptors rises significantly in prostate cancer cells and tissues. Agonists of these receptors promote the growth and mobility of prostate cancer cells, while antagonists may suppress their proliferation, trigger their apoptosis, and inhibit their metastasis. Clinically, receptor antagonists can significantly reduce the risk of prostate cancer and improve its prognosis after androgen depravation therapy. This article presents an overview on the roles of adrenergic receptors in prostate cancer.


Assuntos
Neoplasias da Próstata/metabolismo , Receptores Adrenérgicos/fisiologia , Agonistas Adrenérgicos/farmacologia , Antagonistas Adrenérgicos/farmacologia , Apoptose , Humanos , Masculino , Neoplasias da Próstata/patologia , Receptores Adrenérgicos/efeitos dos fármacos
15.
Pharmacol Biochem Behav ; 121: 146-57, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24291648

RESUMO

In recent years, research on penile erection has increasingly been centered on the molecular mechanisms involved. Major progress has been made in the field and at present a whole number of neurotransmitters, chemical effectors, growth factors, second-messenger molecules, ions, intercellular proteins, and hormones have been characterized as components of the complex process of erection. This knowledge has led to the discovery of several new therapeutic targets and multiple medical approaches for the treatment of erectile dysfunction (ED). This review focuses on the progress made in this field within the last few years.


Assuntos
Disfunção Erétil/fisiopatologia , Disfunção Erétil/terapia , Terapias Complementares , AMP Cíclico/fisiologia , GMP Cíclico/fisiologia , Humanos , Sulfeto de Hidrogênio/metabolismo , Masculino , Óxido Nítrico/fisiologia , Receptores Adrenérgicos/fisiologia , Receptores de Angiotensina/fisiologia , Receptores de Endotelina/fisiologia , Medicina Regenerativa , Transdução de Sinais , Transplante de Células-Tronco , Engenharia Tecidual , Urotensinas/fisiologia , Vasoconstrição/fisiologia , Vasodilatação/fisiologia , Proteína rhoA de Ligação ao GTP/fisiologia
16.
Naunyn Schmiedebergs Arch Pharmacol ; 386(4): 311-8, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23338711

RESUMO

Melanocortin is a potent anti-inflammatory molecule. However, little is known about the effect of melanocortin on acute inflammatory processes such as neutrophil migration. In the present study, we investigated the ability of [Nle4, D-Phe7]-melanocyte-stimulating hormone (NDP-MSH), a semisynthetic melanocortin compound, in the inhibition of neutrophil migration in carrageenin-induced peritonitis model. Herein, subcutaneous pretreatment with NDP-MSH decreased neutrophil trafficking in the peritoneal cavity in a dose-dependent manner. NDP-MSH inhibited vascular leakage, leukocyte rolling, and adhesion and reduced peritoneal macrophage inflammatory protein 2, but not TNF-alpha, IL-1beta, IL-10, and keratinocyte-derived chemokine production. In addition, the effect on neutrophil migration was reverted by the pretreatment with both propranolol (a nonselective beta-adrenergic antagonist) and mecamylamine (a nonselective nicotinic antagonist) but not by splenectomy surgery. Moreover, NDP-MSH intracerebroventricular administration inhibited neutrophil migration, indicating participation of the central nervous system. Our results propose that the NDP-MSH effect may be due to a spleen-independent neuro-immune pathway that efficiently regulates excessive neutrophil recruitment to tissues.


Assuntos
Neutrófilos/efeitos dos fármacos , Peritonite/imunologia , alfa-MSH/análogos & derivados , Animais , Carragenina , Adesão Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Quimiocina CXCL2/imunologia , Citocinas/imunologia , Migração e Rolagem de Leucócitos/efeitos dos fármacos , Masculino , Camundongos , Neutrófilos/fisiologia , Peritonite/induzido quimicamente , Receptores Adrenérgicos/fisiologia , Receptores Nicotínicos/fisiologia , alfa-MSH/farmacologia
17.
Gen Comp Endocrinol ; 175(3): 500-13, 2012 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-22202599

RESUMO

Ovarian pieces containing postvitellogenic follicles were incubated in vitro with different concentrations of the catecholestrogen 2-hydroxyestradiol-17ß (2-OHE(2)) to evaluate its effects on steroid production and germinal vesicle breakdown (GVBD) in the catfish Heteropneustes fossilis. The incubation with 2-OHE(2) induced a shift in steroidogenic pattern: the C(19) and C(18) steroids testosterone (T) and estradiol-17ß (E(2)), respectively were significantly decreased with a concomitant significant increase in the C(21) steroids progesterone (P(4)), 17-hydroxyprogesterone (17-OHP), 17,20ß-dihydroxy-4-pregnen-3-one (17,20ß-DP), 17,20α-dihydroxy-4-pregnen-3-one (17,20α-DP) and cortisol (F). Concomitantly, the catecholestrogen induced dose-dependently GVBD response, the first sign of meiosis resumption. The co- and pre-incubations of the ovarian pieces with 2-OHE(2), and adrenergic (phentolamine, α-blocker and propranolol, ß-blocker) or estrogen (tamoxifen) receptor blockers resulted in inhibition of the stimulatory effect of the catecholestrogen on C(21) steroids and reversed the inhibition of testosterone and E(2). The α-blocker was more effective than the ß-blocker. Our results suggest that 2-OHE(2) appears to employ both adrenergic (α-type) and estrogen receptor mechanisms in mediating the effects. The co- or pre-incubation of ovarian pieces with IBMX (a cAMP elevating drug), H89 (a protein kinase A inhibitor), and PD098059 (a MAP kinase kinase inhibitor) significantly inhibited the stimulatory effect of 2-OHE(2) on the C(21) steroids. The effect of chelerythrine (a protein kinase C inhibitor), on the other hand, varied with the incubation condition. In the co-incubation, the steroids showed varied effects: 17,20ß-DP, testosterone and E(2) were elevated, and P(4) and 17-OHP were decreased. In the pre-incubation set up, all the steroids were inhibited except E(2). The inhibition by the blockers was higher in the pre-incubation groups. Taken together, the data suggest the involvement cAMP-protein kinase A, protein kinase C and MAP kinase pathways in the modulation of the steroidogenic activity.


Assuntos
Peixes-Gato/metabolismo , Estradiol/análogos & derivados , Hormônios Esteroides Gonadais/metabolismo , Folículo Ovariano/efeitos dos fármacos , Folículo Ovariano/metabolismo , Receptores Adrenérgicos/fisiologia , Receptores de Estrogênio/fisiologia , Transdução de Sinais/fisiologia , 17-alfa-Hidroxiprogesterona/metabolismo , Animais , Estradiol/metabolismo , Estradiol/farmacologia , Feminino , Hidrocortisona/metabolismo , Hidroxiprogesteronas/metabolismo , Técnicas In Vitro , Meiose/efeitos dos fármacos , Progesterona/metabolismo , Testosterona/metabolismo
18.
Sports Med ; 41(8): 621-39, 2011 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-21780849

RESUMO

This article reviews the interaction between the neuroendocrine and immune systems in response to exercise stress, considering gender differences. The body's response to exercise stress is a system-wide effort coordinated by the integration between the immune and the neuroendocrine systems. Although considered distinct systems, increasing evidence supports the close communication between them. Like any stressor, the body's response to exercise triggers a systematic series of neuroendocrine and immune events directed at bringing the system back to a state of homeostasis. Physical exercise presents a unique physiological stress where the neuroendocrine and immune systems contribute to accommodating the increase in physiological demands. These systems of the body also adapt to chronic overload, or exercise training. Such adaptations alleviate the magnitude of subsequent stress or minimize the exercise challenge to within homeostatic limits. This adaptive capacity of collaborating systems resembles the acquired, or adaptive, branch of the immune system, characterized by the memory capacity of the cells involved. Specific to the adaptive immune response, once a specific antigen is encountered, memory cells, or lymphocytes, mount a response that reduces the magnitude of the immune response to subsequent encounters of the same stress. In each case, the endocrine response to physical exercise and the adaptive branch of the immune system share the ability to adapt to a stressful encounter. Moreover, each of these systemic responses to stress is influenced by gender. In both the neuroendocrine responses to exercise and the adaptive (B lymphocyte) immune response, gender differences have been attributed to the 'protective' effects of estrogens. Thus, this review will create a paradigm to explain the neuroendocrine communication with leukocytes during exercise by reviewing (i) endocrine and immune interactions; (ii) endocrine and immune systems response to physiological stress; and (iii) gender differences (and the role of estrogen) in both endocrine response to physiological stress and adaptive immune response.


Assuntos
Exercício Físico/fisiologia , Sistema Imunitário/fisiologia , Neuroimunomodulação/imunologia , Neuroimunomodulação/fisiologia , Sistemas Neurossecretores/imunologia , Sistemas Neurossecretores/fisiologia , Animais , Catecolaminas/imunologia , Catecolaminas/fisiologia , Estradiol/imunologia , Estradiol/fisiologia , Feminino , Humanos , Hidrocortisona/imunologia , Hidrocortisona/fisiologia , Masculino , Camundongos , Ratos , Receptores Adrenérgicos/imunologia , Receptores Adrenérgicos/fisiologia , Receptores de Glucocorticoides/imunologia , Receptores de Glucocorticoides/fisiologia , Testosterona/imunologia , Testosterona/fisiologia
19.
Endocr Regul ; 45(2): 65-90, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21615192

RESUMO

The WHO classification of endocrine tumors defines pheochromocytoma as a tumor arising from chromaffin cells in the adrenal medulla - an intra-adrenal paraganglioma. Closely related tumors of extra-adrenal sympathetic and parasympathetic paraganglia are classified as extra-adrenal paragangliomas. Almost all pheochromocytomas and paragangliomas produce catecholamines. The concentrations of catecholamines in pheochromocytoma tissues are enormous, potentially creating a volcano that can erupt at any time. Significant eruptions result in catecholamine storms called "attacks" or "spells". Acute catecholamine crisis can strike unexpectedly, leaving traumatic memories of acute medical disaster that champions any intensive care unit. A very well-defined genotype-biochemical phenotype relationship exists, guiding proper and cost-effective genetic testing of patients with these tumors. Currently, the production of norepinephrine and epinephrine is optimally assessed by the measurement of their O-methylated metabolites, normetanephrine or metanephrine, respectively. Dopamine is a minor component, but some paragangliomas produce only this catecholamine or this together with norepinephrine. Methoxytyramine, the O-methylated metabolite of dopamine, is the best biochemical marker of these tumors. In those patients with equivocal biochemical results, a modified clonidine suppression test coupled with the measurement of plasma normetanephrine has recently been introduced. In addition to differences in catecholamine enzyme expression, the presence of either constitutive or regulated secretory pathways contributes further to the very unique mutation-dependent catecholamine production and release, resulting in various clinical presentations. Oxidative stress results from a significant imbalance between levels of prooxidants, generated during oxidative phosphorylation, and antioxidants. The gradual accumulation of prooxidants due to metabolic oxidative stress results in proto-oncogene activation, tumor suppressor gene inactivation, DNA damage, and genomic instability. Since the mitochondria serves as the main source of prooxidants, any mitochondrial impairment leads to severe oxidative stress, a major outcome of which is tumor development. In terms of cancer pathogenesis, pheochromocytomas and paragangliomas represent tumors where the oxidative phosphorylation defect due to the mutation of succinate dehydrogenase is the cause, not a consequence, of tumor development. Any succinate dehydrogenase pathogenic mutation results in the shift from oxidative phosphorylation to aerobic glycolysis in the cytoplasm (also called anaerobic glycolysis if hypoxia is the main cause of such a shift). This phenomenon, also called the Warburg effect, is well demonstrated by a positive [18F]-fluorodeoxyglycose positron emission tomography scan. Microarray studies, genome-wide association studies, proteomics and protein arrays, metabolomics, transcriptomics, and bioinformatics approaches will remain powerful tools to further uncover the pathogenesis of these tumors and their unique markers, with the ultimate goal to introduce new therapeutic options for those with metastatic or malignant pheochromocytoma and paraganglioma. Soon oxidative stress will be tightly linked to a multistep cancer process in which the mutation of various genes (perhaps in a logistic way) ultimately results in uncontrolled growth, proliferation, and metastatic potential of practically any cell. Targeting the mTORC, IGF-1, HIF and other pathways, topoisomerases, protein degradation by proteosomes, balancing the activity of protein kinases and phosphatases or even synchronizing the cell cycle before any exposure to any kind of therapy will soon become a reality. Facing such a reality today will favor our chances to "beat" this disease tomorrow.


Assuntos
Neoplasias das Glândulas Suprarrenais , Catecolaminas/metabolismo , Estresse Oxidativo/fisiologia , Feocromocitoma , Succinato Desidrogenase/genética , Trifosfato de Adenosina/biossíntese , Catecolaminas/efeitos adversos , Catecolaminas/fisiologia , Humanos , Metanefrina/sangue , Neoplasia Endócrina Múltipla Tipo 2a/fisiopatologia , Mutação , Neurofibromatose 1/fisiopatologia , Normetanefrina/sangue , Paraganglioma/genética , Paraganglioma Extrassuprarrenal/genética , Feocromocitoma/genética , Feocromocitoma/fisiopatologia , Proto-Oncogene Mas , Receptores Adrenérgicos/efeitos dos fármacos , Receptores Adrenérgicos/fisiologia , Doença de von Hippel-Lindau/fisiopatologia
20.
J Cardiol ; 57(3): 338-44, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21367584

RESUMO

AIMS: MicroRNAs (miRNAs) are small non-coding RNAs discovered as potential new gene regulators. Their roles in the development of chronic heart failure (CHF), however, are largely unknown. Reduced catecholamine sensitivity is an early step of CHF. We examined whether altered expression of miRNAs was related to reduced catecholamine sensitivity in patients with CHF. METHODS AND RESULTS: Maximum first derivative of left ventricular pressure (LV dP/dt(max)) at baseline and during infusion of dobutamine (10 µg kg(-1)min(-1)) were determined in 14 asymptomatic or mildly symptomatic (New York Heart Association class I or II) patients with idiopathic dilated cardiomyopathy (DCM). We performed microarray analysis for a total of 485 miRNAs using endomyocardial biopsy specimens from these 14 patients. Patients were classified into 2 groups based on a percent increase in LV dP/dt(max) by dobutamine infusion (ΔLV dP/dt(max)). These are Group I (n=7) with ΔLV dP/dt(max)>90%, and Group II (n=7) with ΔLV dP/dt(max)<90%. Out of 485 miRNAs, 32 were differentially expressed in the myocardium with reduced catecholamine sensitivity. Among those, four miRNAs were decreased and one miRNA was increased in the Group II compared to the Group I (p<0.01). LVEF measured by left ventriculography at baseline did not differ between the 2 groups. Also there were no differences in plasma norepinephrine levels between the 2 groups. CONCLUSIONS: Altered expression of several miRNAs was related to the reduced catecholamine sensitivity in mildly symptomatic patients with DCM. These findings shed light on the potential of miRNAs to provide possible etiologic insights as well as therapeutic targets for CHF.


Assuntos
Catecolaminas/fisiologia , Insuficiência Cardíaca/genética , MicroRNAs/análise , Cateterismo Cardíaco , Cardiomiopatia Dilatada/complicações , Doença Crônica , Dobutamina , Feminino , Regulação da Expressão Gênica/fisiologia , Humanos , Masculino , Análise em Microsséries , Pessoa de Meia-Idade , Contração Miocárdica/fisiologia , Miocárdio/química , Norepinefrina/sangue , Receptores Adrenérgicos/fisiologia , Função Ventricular/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA