Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Mol Metab ; 80: 101886, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38246589

RESUMO

OBJECTIVE: The central melanocortin system is essential for the regulation of food intake and body weight. Agouti-related protein (AgRP) is the sole orexigenic component of the central melanocortin system and is conserved across mammalian species. AgRP is currently known to be expressed exclusively in the mediobasal hypothalamus, and hypothalamic AgRP-expressing neurons are essential for feeding. Here we characterized a previously unknown population of AgRP cells in the mouse hindbrain. METHODS: Expression of AgRP in the hindbrain was investigated using gene expression analysis, single-cell RNA sequencing, immunofluorescent analysis and multiple transgenic mice with reporter expressions. Activation of AgRP neurons was achieved by Designer Receptors Exclusively Activated by Designer Drugs (DREADD) and by transcranial focal photo-stimulation using a step-function opsin with ultra-high light sensitivity (SOUL). RESULTS: AgRP expressing cells were present in the area postrema (AP) and the adjacent subpostrema area (SubP) and commissural nucleus of the solitary tract (cNTS) of the mouse hindbrain (termed AgRPHind herein). AgRPHind cells consisted of locally projecting neurons as well as tanycyte-like cells. Food deprivation stimulated hindbrain Agrp expression as well as neuronal activity of subsets of AgRPHind cells. In adult mice that lacked hypothalamic AgRP neurons, chemogenetic activation of AgRP neurons resulted in hyperphagia and weight gain. In addition, transcranial focal photo-stimulation of hindbrain AgRP cells increased food intake in adult mice with or without hypothalamic AgRP neurons. CONCLUSIONS: Our study indicates that the central melanocortin system in the hindbrain possesses an orexigenic component, and that AgRPHind neurons stimulate feeding independently of hypothalamic AgRP neurons.


Assuntos
Hipotálamo , Melanocortinas , Camundongos , Animais , Proteína Relacionada com Agouti/genética , Proteína Relacionada com Agouti/metabolismo , Hipotálamo/metabolismo , Camundongos Transgênicos , Melanocortinas/metabolismo , Rombencéfalo/metabolismo , Mamíferos/metabolismo
2.
Cell Biosci ; 13(1): 20, 2023 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-36732847

RESUMO

BACKGROUND: Foraging for food precedes food consumption and is an important component of the overall metabolic programming that regulates feeding. Foraging is governed by central nervous system neuronal circuits but how it is influenced by diet and hormonal signals is still not well understood. RESULTS: In this study, we show that dietary cholesterol exerted suppressive effects on locomotor activity and that these effects were partially mediated by the neuropeptide Agouti-related protein (AgRP). High dietary cholesterol stimulated intestinal expression of fibroblast growth factor 15 (Fgf15), an ortholog of the human fibroblast growth factor 19 (FGF19). Intracerebroventricular infusion of FGF19 peptide reduced exploratory activity in the open field test paradigm. On the other hand, the lack of dietary cholesterol enhanced exploratory activity in the open field test, but this effect was abolished by central administration of FGF19. CONCLUSIONS: Experiments in this study show that dietary cholesterol suppresses locomotor activity and foraging-like behaviors, and this regulation is in part mediated by AgRP neurons. Dietary cholesterol or the central action of FGF19 suppresses exploratory behaviors, and the anxiogenic effects of dietary cholesterol may be mediated by the effect of FGF19 in the mouse brain. This study suggests that dietary cholesterol and intestinal hormone FGF15/19 signal a satiating state to the brain, thereby suppressing foraging-like behaviors.

3.
Sci Signal ; 15(733): eabj8204, 2022 05 10.
Artigo em Inglês | MEDLINE | ID: mdl-35536884

RESUMO

Variants in the gene encoding ankyrin repeat and SOCS box-containing 4 (ASB4) are linked to human obesity. Here, we characterized the pathways underlying the metabolic functions of ASB4. Hypothalamic Asb4 expression was suppressed by fasting in wild-type mice but not in mice deficient in AgRP, which encodes Agouti-related protein (AgRP), an appetite-stimulating hormone, suggesting that ASB4 is a negative target of AgRP. Many ASB4 neurons in the brain were adjacent to AgRP terminals, and feeding induced by AgRP neuronal activation was disrupted in Asb4-deficient mice. Acute knockdown of Asb4 in the brain caused marked hyperphagia due to increased meal size, and Asb4 deficiency led to increased meal size and food intake at the onset of refeeding, when very large meals were consumed. Asb4-deficient mice were resistant to the meal-terminating effects of exogenously administered calcitonin and showed decreased neuronal expression of Calcr, which encodes the calcitonin receptor. Pro-opiomelanocortin (POMC) neurons in the arcuate nucleus in mice are involved in glucose homeostasis, and Asb4 deficiency specifically in POMC neurons resulted in glucose intolerance that was independent of obesity. Furthermore, individuals with type 2 diabetes showed reduced ASB4 abundance in the infundibular nuclei, the human equivalent of the arcuate nucleus. Together, our results indicate that ASB4 acts in the brain to improve glucose homeostasis and to induce satiety after substantial meals, particularly those after food deprivation.


Assuntos
Diabetes Mellitus Tipo 2 , Neuropeptídeos , Proteína Relacionada com Agouti/genética , Proteína Relacionada com Agouti/metabolismo , Proteína Relacionada com Agouti/farmacologia , Animais , Calcitonina/metabolismo , Calcitonina/farmacologia , Diabetes Mellitus Tipo 2/metabolismo , Glucose/metabolismo , Homeostase , Hipotálamo/metabolismo , Camundongos , Neurônios/metabolismo , Neuropeptídeos/metabolismo , Obesidade/genética , Obesidade/metabolismo , Pró-Opiomelanocortina/genética , Pró-Opiomelanocortina/metabolismo , Pró-Opiomelanocortina/farmacologia
4.
Mol Metab ; 58: 101442, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35051651

RESUMO

OBJECTIVE: Preference for dietary fat vs. carbohydrate varies markedly across free-living individuals. It is recognized that food choice is under genetic and physiological regulation, and that the central melanocortin system is involved. However, how genetic and dietary factors interact to regulate relative macronutrient intake is not well understood. METHODS: We investigated how the choice for food rich in carbohydrate vs. fat is influenced by dietary cholesterol availability and agouti-related protein (AGRP), the orexigenic component of the central melanocortin system. We assessed how macronutrient intake and different metabolic parameters correlate with plasma AGRP in a cohort of obese humans. We also examined how both dietary cholesterol levels and inhibiting de novo cholesterol synthesis affect carbohydrate and fat intake in mice, and how dietary cholesterol deficiency during the postnatal period impacts macronutrient intake patterns in adulthood. RESULTS: In obese human subjects, plasma levels of AGRP correlated inversely with consumption of carbohydrates over fats. Moreover, AgRP-deficient mice preferred to consume more calories from carbohydrates than fats, more so when each diet lacked cholesterol. Intriguingly, inhibiting cholesterol biosynthesis (simvastatin) promoted carbohydrate intake at the expense of fat without altering total caloric consumption, an effect that was remarkably absent in AgRP-deficient mice. Finally, feeding lactating C57BL/6 dams and pups a cholesterol-free diet prior to weaning led the offspring to prefer fats over carbohydrates as adults, indicating that altered cholesterol metabolism early in life programs adaptive changes to macronutrient intake. CONCLUSIONS: Together, our study illustrates a specific gene-diet interaction in modulating food choice.


Assuntos
Colesterol na Dieta , Carboidratos da Dieta , Adulto , Proteína Relacionada com Agouti , Animais , Dieta , Feminino , Humanos , Lactação , Melanocortinas , Camundongos , Camundongos Endogâmicos C57BL , Obesidade
5.
Transl Psychiatry ; 9(1): 141, 2019 05 10.
Artigo em Inglês | MEDLINE | ID: mdl-31076569

RESUMO

Obesity is associated with an increased risk of depression. The aim of the present study was to investigate whether obesity is a causative factor for the development of depression and what is the molecular pathway(s) that link these two disorders. Using lipidomic and transcriptomic methods, we identified a mechanism that links exposure to a high-fat diet (HFD) in mice with alterations in hypothalamic function that lead to depression. Consumption of an HFD selectively induced accumulation of palmitic acid in the hypothalamus, suppressed the 3', 5'-cyclic AMP (cAMP)/protein kinase A (PKA) signaling pathway, and increased the concentration of free fatty acid receptor 1 (FFAR1). Deficiency of phosphodiesterase 4A (PDE4A), an enzyme that degrades cAMP and modulates stimulatory regulative G protein (Gs)-coupled G protein-coupled receptor signaling, protected animals either from genetic- or dietary-induced depression phenotype. These findings suggest that dietary intake of saturated fats disrupts hypothalamic functions by suppressing cAMP/PKA signaling through activation of PDE4A. FFAR1 inhibition and/or an increase of cAMP signaling in the hypothalamus could offer potential therapeutic targets to counteract the effects of dietary or genetically induced obesity on depression.


Assuntos
AMP Cíclico/metabolismo , Nucleotídeo Cíclico Fosfodiesterase do Tipo 4/metabolismo , Depressão/fisiopatologia , Dieta Hiperlipídica/efeitos adversos , Hipotálamo/fisiopatologia , Obesidade/fisiopatologia , Animais , Comportamento Animal , Depressão/etiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Obesidade/etiologia , Transdução de Sinais
6.
Endocrinology ; 159(6): 2408-2420, 2018 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-29750244

RESUMO

Proper regulation of energy metabolism requires neurons in the central nervous system to respond dynamically to signals that reflect the body's energy reserve, and one such signal is leptin. Agouti-related protein (AgRP) is a hypothalamic neuropeptide that is markedly upregulated in leptin deficiency, a condition that is associated with severe obesity, diabetes, and hepatic steatosis. Because deleting AgRP in mice does not alter energy balance, we sought to determine whether AgRP plays an indispensable role in regulating energy and hepatic lipid metabolism in the sensitized background of leptin deficiency. We generated male mice that are deficient for both leptin and AgRP [double-knockout (DKO)]. DKO mice and ob/ob littermates had similar body weights, food intake, energy expenditure, and plasma insulin levels, although DKO mice surprisingly developed heightened hyperglycemia with advancing age. Overall hepatic lipid content was reduced in young prediabetic DKO mice, but not in the older diabetic counterparts. Intriguingly, however, both young and older DKO mice had an altered zonal distribution of hepatic lipids with reduced periportal lipid deposition. Moreover, leptin stimulated, whereas AgRP inhibited, hepatic sympathetic activity. Ablating sympathetic nerves to the liver, which primarily innervate the portal regions, produced periportal lipid accumulation in wild-type mice. Collectively, our results highlight AgRP as a regulator of hepatic sympathetic activity and metabolic zonation.


Assuntos
Proteína Relacionada com Agouti/fisiologia , Metabolismo dos Lipídeos/genética , Fígado/metabolismo , Proteína Relacionada com Agouti/genética , Animais , Fígado Gorduroso/genética , Fígado Gorduroso/metabolismo , Fígado Gorduroso/patologia , Leptina/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Obesos , Obesidade Mórbida/genética , Obesidade Mórbida/metabolismo , Obesidade Mórbida/patologia , Distribuição Tecidual/genética
7.
Cell Rep ; 19(11): 2257-2271, 2017 06 13.
Artigo em Inglês | MEDLINE | ID: mdl-28614713

RESUMO

Neurons expressing agouti-related protein (AgRP) are essential for feeding. The majority of these neurons are located outside the blood-brain barrier (BBB), allowing them to directly sense circulating metabolic factors. Here, we show that, in adult mice, AgRP neurons outside the BBB (AgRPOBBB) were rapidly ablated by peripheral administration of monosodium glutamate (MSG), whereas AgRP neurons inside the BBB and most proopiomelanocortin (POMC) neurons were spared. MSG treatment induced proliferation of tanycytes, the putative hypothalamic neural progenitor cells, but the newly proliferated tanycytes did not become neurons. Intriguingly, AgRPOBBB neuronal number increased within a week after MSG treatment, and newly emerging AgRP neurons were derived from post-mitotic cells, including some from the Pomc-expressing cell lineage. Our study reveals that the lack of protection by the BBB renders AgRPOBBB vulnerable to lesioning by circulating toxins but that the rapid re-emergence of AgRPOBBB is part of a reparative process to maintain energy balance.


Assuntos
Barreira Hematoencefálica/citologia , Hipotálamo/citologia , Neurônios/citologia , Pró-Opiomelanocortina/metabolismo , Animais , Transporte Biológico , Barreira Hematoencefálica/metabolismo , Hipotálamo/metabolismo , Camundongos , Neurônios/metabolismo
8.
Cell Rep ; 14(2): 255-68, 2016 Jan 12.
Artigo em Inglês | MEDLINE | ID: mdl-26748707

RESUMO

Obesity and metabolic syndrome reflect the dysregulation of molecular pathways that control energy homeostasis. Here, we show that the p75 neurotrophin receptor (p75(NTR)) controls energy expenditure in obese mice on a high-fat diet (HFD). Despite no changes in food intake, p75(NTR)-null mice were protected from HFD-induced obesity and remained lean as a result of increased energy expenditure without developing insulin resistance or liver steatosis. p75(NTR) directly interacts with the catalytic subunit of protein kinase A (PKA) and regulates cAMP signaling in adipocytes, leading to decreased lipolysis and thermogenesis. Adipocyte-specific depletion of p75(NTR) or transplantation of p75(NTR)-null white adipose tissue (WAT) into wild-type mice fed a HFD protected against weight gain and insulin resistance. Our results reveal that signaling from p75(NTR) to cAMP/PKA regulates energy balance and suggest that non-CNS neurotrophin receptor signaling could be a target for treating obesity and the metabolic syndrome.


Assuntos
Metabolismo dos Lipídeos/fisiologia , Obesidade/metabolismo , Receptor de Fator de Crescimento Neural/metabolismo , Animais , Camundongos , Camundongos Knockout , Transdução de Sinais
9.
Nat Commun ; 6: 8164, 2015 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-26353940

RESUMO

Autoimmunity and macrophage recruitment into the central nervous system (CNS) are critical determinants of neuroinflammatory diseases. However, the mechanisms that drive immunological responses targeted to the CNS remain largely unknown. Here we show that fibrinogen, a central blood coagulation protein deposited in the CNS after blood-brain barrier disruption, induces encephalitogenic adaptive immune responses and peripheral macrophage recruitment into the CNS leading to demyelination. Fibrinogen stimulates a unique transcriptional signature in CD11b(+) antigen-presenting cells inducing the recruitment and local CNS activation of myelin antigen-specific Th1 cells. Fibrinogen depletion reduces Th1 cells in the multiple sclerosis model, experimental autoimmune encephalomyelitis. Major histocompatibility complex (MHC) II-dependent antigen presentation, CXCL10- and CCL2-mediated recruitment of T cells and macrophages, respectively, are required for fibrinogen-induced encephalomyelitis. Inhibition of the fibrinogen receptor CD11b/CD18 protects from all immune and neuropathologic effects. Our results show that the final product of the coagulation cascade is a key determinant of CNS autoimmunity.


Assuntos
Autoimunidade/imunologia , Encéfalo/imunologia , Doenças Desmielinizantes/imunologia , Encefalomielite Autoimune Experimental/imunologia , Fibrinogênio/imunologia , Genes MHC da Classe II/imunologia , Macrófagos/imunologia , Medula Espinal/imunologia , Células Th1/imunologia , Imunidade Adaptativa/efeitos dos fármacos , Imunidade Adaptativa/genética , Imunidade Adaptativa/imunologia , Animais , Apresentação de Antígeno/efeitos dos fármacos , Apresentação de Antígeno/genética , Apresentação de Antígeno/imunologia , Autoimunidade/efeitos dos fármacos , Autoimunidade/genética , Barreira Hematoencefálica , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/patologia , Antígeno CD11b/genética , Antígeno CD11b/imunologia , Receptor 1 de Quimiocina CX3C , Proliferação de Células , Quimiocina CCL2/imunologia , Quimiocina CXCL10/genética , Quimiocina CXCL10/imunologia , Quimiocinas , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/imunologia , Doenças Desmielinizantes/genética , Fibrina , Fibrinogênio/farmacologia , Citometria de Fluxo , Perfilação da Expressão Gênica , Genes MHC da Classe II/genética , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/imunologia , Imuno-Histoquímica , Camundongos , Camundongos Knockout , Microglia , Glicoproteína Mielina-Oligodendrócito/imunologia , Ratos , Receptores de Antígenos de Linfócitos T/imunologia , Receptores de Quimiocinas/genética , Receptores de Quimiocinas/imunologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Medula Espinal/efeitos dos fármacos , Medula Espinal/metabolismo , Medula Espinal/patologia
10.
Nat Neurosci ; 18(8): 1077-80, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26120963

RESUMO

Astrocytes modulate neuronal activity and inhibit regeneration. We show that cleaved p75 neurotrophin receptor (p75(NTR)) is a component of the nuclear pore complex (NPC) required for glial scar formation and reduced gamma oscillations in mice via regulation of transforming growth factor (TGF)-ß signaling. Cleaved p75(NTR) interacts with nucleoporins to promote Smad2 nucleocytoplasmic shuttling. Thus, NPC remodeling by regulated intramembrane cleavage of p75(NTR) controls astrocyte-neuronal communication in response to profibrotic factors.


Assuntos
Astrócitos/metabolismo , Ritmo Gama/fisiologia , Atividade Motora/fisiologia , Poro Nuclear/metabolismo , Receptor de Fator de Crescimento Neural/metabolismo , Transdução de Sinais , Fator de Crescimento Transformador beta/metabolismo , Animais , Comportamento Animal/fisiologia , Eletroencefalografia , Gliose/metabolismo , Células HEK293 , Humanos , Hidrocefalia/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Células NIH 3T3 , Complexo de Proteínas Formadoras de Poros Nucleares/metabolismo , Receptor de Fator de Crescimento Neural/deficiência , Proteína Smad2/metabolismo
11.
J Neurosci ; 33(25): 10221-34, 2013 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-23785138

RESUMO

The p75 neurotrophin receptor (p75(NTR)) is a member of the tumor necrosis factor receptor superfamily with a widespread pattern of expression in tissues such as the brain, liver, lung, and muscle. The mechanisms that regulate p75(NTR) transcription in the nervous system and its expression in other tissues remain largely unknown. Here we show that p75(NTR) is an oscillating gene regulated by the helix-loop-helix transcription factors CLOCK and BMAL1. The p75(NTR) promoter contains evolutionarily conserved noncanonical E-box enhancers. Deletion mutagenesis of the p75(NTR)-luciferase reporter identified the -1039 conserved E-box necessary for the regulation of p75(NTR) by CLOCK and BMAL1. Accordingly, gel-shift assays confirmed the binding of CLOCK and BMAL1 to the p75(NTR-)1039 E-box. Studies in mice revealed that p75(NTR) transcription oscillates during dark and light cycles not only in the suprachiasmatic nucleus (SCN), but also in peripheral tissues including the liver. Oscillation of p75(NTR) is disrupted in Clock-deficient and mutant mice, is E-box dependent, and is in phase with clock genes, such as Per1 and Per2. Intriguingly, p75(NTR) is required for circadian clock oscillation, since loss of p75(NTR) alters the circadian oscillation of clock genes in the SCN, liver, and fibroblasts. Consistent with this, Per2::Luc/p75(NTR-/-) liver explants showed reduced circadian oscillation amplitude compared with those of Per2::Luc/p75(NTR+/+). Moreover, deletion of p75(NTR) also alters the circadian oscillation of glucose and lipid homeostasis genes. Overall, our findings reveal that the transcriptional activation of p75(NTR) is under circadian regulation in the nervous system and peripheral tissues, and plays an important role in the maintenance of clock and metabolic gene oscillation.


Assuntos
Proteínas CLOCK/fisiologia , Ritmo Circadiano/fisiologia , Metabolismo/fisiologia , Receptor de Fator de Crescimento Neural/fisiologia , Fatores de Transcrição ARNTL/biossíntese , Fatores de Transcrição ARNTL/genética , Animais , Glicemia/metabolismo , Proteínas CLOCK/genética , Ritmo Circadiano/genética , DNA/genética , Ensaio de Desvio de Mobilidade Eletroforética , Células HEK293 , Homeostase/genética , Humanos , Fígado/metabolismo , Luciferases/genética , Metabolismo/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Atividade Motora/fisiologia , Reação em Cadeia da Polimerase em Tempo Real , Receptor de Fator de Crescimento Neural/genética , Choque Séptico/fisiopatologia , Núcleo Supraquiasmático/fisiologia , Transfecção
13.
Proc Natl Acad Sci U S A ; 109(15): 5838-43, 2012 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-22460790

RESUMO

Insulin resistance is a key factor in the etiology of type 2 diabetes. Insulin-stimulated glucose uptake is mediated by the glucose transporter 4 (GLUT4), which is expressed mainly in skeletal muscle and adipose tissue. Insulin-stimulated translocation of GLUT4 from its intracellular compartment to the plasma membrane is regulated by small guanosine triphosphate hydrolases (GTPases) and is essential for the maintenance of normal glucose homeostasis. Here we show that the p75 neurotrophin receptor (p75(NTR)) is a regulator of glucose uptake and insulin resistance. p75(NTR) knockout mice show increased insulin sensitivity on normal chow diet, independent of changes in body weight. Euglycemic-hyperinsulinemic clamp studies demonstrate that deletion of the p75(NTR) gene increases the insulin-stimulated glucose disposal rate and suppression of hepatic glucose production. Genetic depletion or shRNA knockdown of p75(NTR) in adipocytes or myoblasts increases insulin-stimulated glucose uptake and GLUT4 translocation. Conversely, overexpression of p75(NTR) in adipocytes decreases insulin-stimulated glucose transport. In adipocytes, p75(NTR) forms a complex with the Rab5 family GTPases Rab5 and Rab31 that regulate GLUT4 trafficking. Rab5 and Rab31 directly interact with p75(NTR) primarily via helix 4 of the p75(NTR) death domain. Adipocytes from p75(NTR) knockout mice show increased Rab5 and decreased Rab31 activities, and dominant negative Rab5 rescues the increase in glucose uptake seen in p75(NTR) knockout adipocytes. Our results identify p75(NTR) as a unique player in glucose metabolism and suggest that signaling from p75(NTR) to Rab5 family GTPases may represent a unique therapeutic target for insulin resistance and diabetes.


Assuntos
Glucose/metabolismo , Homeostase , Resistência à Insulina , Receptor de Fator de Crescimento Neural/metabolismo , Adipócitos/metabolismo , Sequência de Aminoácidos , Animais , Peso Corporal , Transportador de Glucose Tipo 4/metabolismo , Células HEK293 , Humanos , Camundongos , Dados de Sequência Molecular , Células Musculares/metabolismo , Músculo Esquelético/citologia , Ligação Proteica , Estrutura Terciária de Proteína , Transporte Proteico , Receptor de Fator de Crescimento Neural/química , Receptor de Fator de Crescimento Neural/deficiência , Proteínas rab de Ligação ao GTP/metabolismo , Proteínas rab5 de Ligação ao GTP/metabolismo
14.
J Neurosci ; 30(17): 5843-54, 2010 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-20427645

RESUMO

Scar formation in the nervous system begins within hours after traumatic injury and is characterized primarily by reactive astrocytes depositing proteoglycans that inhibit regeneration. A fundamental question in CNS repair has been the identity of the initial molecular mediator that triggers glial scar formation. Here we show that the blood protein fibrinogen, which leaks into the CNS immediately after blood-brain barrier (BBB) disruption or vascular damage, serves as an early signal for the induction of glial scar formation via the TGF-beta/Smad signaling pathway. Our studies revealed that fibrinogen is a carrier of latent TGF-beta and induces phosphorylation of Smad2 in astrocytes that leads to inhibition of neurite outgrowth. Consistent with these findings, genetic or pharmacologic depletion of fibrinogen in mice reduces active TGF-beta, Smad2 phosphorylation, glial cell activation, and neurocan deposition after cortical injury. Furthermore, stereotactic injection of fibrinogen into the mouse cortex is sufficient to induce astrogliosis. Inhibition of the TGF-beta receptor pathway abolishes the fibrinogen-induced effects on glial scar formation in vivo and in vitro. These results identify fibrinogen as a primary astrocyte activation signal, provide evidence that deposition of inhibitory proteoglycans is induced by a blood protein that leaks in the CNS after vasculature rupture, and point to TGF-beta as a molecular link between vascular permeability and scar formation.


Assuntos
Astrócitos/fisiologia , Córtex Cerebral/lesões , Córtex Cerebral/fisiopatologia , Cicatriz/fisiopatologia , Fibrinogênio/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Animais , Células Cultivadas , Córtex Cerebral/irrigação sanguínea , Fibrinogênio/genética , Gliose/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas do Tecido Nervoso/metabolismo , Neuritos/fisiologia , Neurocam , Fosforilação , Proteoglicanas/metabolismo , Transdução de Sinais , Proteína Smad2/metabolismo
15.
Curr Drug Metab ; 9(7): 628-60, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18781915

RESUMO

Arylamine N-acetyltransferases (NATs) are xenobiotic metabolizing enzymes found in prokaryotes and eukaryotes. NATs have been characterized in bacteria (Bacilli, Mycobacteria, Salmonella etc.), laboratory animals (chicken, rabbit, rodents etc.) and humans, where the NAT loci occupy 230 kilobases on chromosome 8p22. Our previous comprehensive search for NAT genes involved 416 genomes (340 prokaryotic, 76 eukaryotic) and identified NAT homologues in several taxa, while also reporting on taxa that appeared to lack NAT genes [Boukouvala, S. and Fakis, G. (2005) Drug Metab. Rev. 37(3), 511-564]. Here, we present an update of this genomic search, covering 2138 genomes (1674 prokaryotic, 464 eukaryotic), of which 1167 (986 prokaryotic, 181 eukaryotic) were accessible using the advanced search algorithm tBLASTn. We have reconstructed the full-length open reading frames for putative proteins with sequence homology and features characteristic of NAT from 274 bacterial genomes (31 actinobacteria, 6 bacteroidetes/chlorobi, 2 cyanobacteria, 65 firmicutes and 170 proteobacteria) and 27 animals (1 sea-urchin, 5 fishes, 1 lizard, 1 bird and 19 mammals). Partial NAT sequences were recovered from several other organisms, including fungi, where NAT genes were found in 30 ascomycetes and 2 basidiomycetes. No NATs were found in arhaea, plants and lower invertebrates (insects and worms), while it is also uncertain whether NAT genes exist in protista. We present comparative genomic and phylogenetic analyses of the identified NAT homologues and announce a new database that will maintain information on non-human NATs and will provide recommendations for a standardized nomenclature, along the lines of the NAT Gene Nomenclature Committee.


Assuntos
Archaea/enzimologia , Arilamina N-Acetiltransferase/genética , Bactérias/enzimologia , Fungos/enzimologia , Genoma , Animais , Archaea/genética , Arilamina N-Acetiltransferase/classificação , Arilamina N-Acetiltransferase/metabolismo , Bactérias/genética , Bases de Dados como Assunto , Fungos/genética , Humanos , Filogenia , Terminologia como Assunto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...