Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Neuropeptides ; 87: 102136, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-33721592

RESUMEN

Anxiety induced by excess mental or physical stress is deeply involved in the onset of human psychiatric diseases such as depression, bipolar disorder, and panic disorder. Recently, Kampo medicines have received focus as antidepressant drugs for clinical use because of their synergistic and additive effects. Thus, we evaluated the anxiolytic activity of Ninjinyoeito (NYT) using neuropeptide Y-knockout (NPY-KO) zebrafish that exhibit severe anxiety responses to acute stress. Adult NPY-KO zebrafish were fed either a 3% NYT-supplemented or normal diet (i.e., the control diet) for four days and were then examined via behavioral tests. After short-term cold stress (10 °C, 2 s) was applied, control-fed NPY-KO zebrafish exhibited anxiety behaviors such as freezing, erratic movement, and increased swimming time along the tank wall. On the other hand, NYT-fed NPY-KO zebrafish significantly suppressed these anxiety behaviors, accompanied by a downregulation of tyrosine hydroxylase levels and phosphorylation of extracellular signal-regulated kinases in the brain. To understand the responsible component(s) in NYT, twelve kinds of herbal medicines that composed NYT were tested in behavioral trials with the zebrafish. Among them, nine significantly reduced freezing behavior in NPY-KO zebrafish. In particular, Schisandra fruit induced the most potent effect on abnormal zebrafish behavior, even in the lower amount (0.3% equivalent to NYT), followed by Atractylodes rhizome and Cinnamon bark. Subsequently, four lignans uniquely found in Schisandra fruit (i.e., gomisin A, gomisin N, schizandrin, and schizandrin B) were investigated for their anxiolytic activity in NPY-KO zebrafish. As a result, schizandrin was identified as a responsible compound in the anxiolytic effect of NYT. These results suggest that NYT has a positive effect on mental stress-induced anxiety and may be a promising therapeutic for psychiatric diseases.


Asunto(s)
Ansiolíticos/uso terapéutico , Ansiedad/tratamiento farmacológico , Medicamentos Herbarios Chinos/uso terapéutico , Neuropéptido Y/deficiencia , Fitoterapia , Animales , Ansiolíticos/química , Ansiedad/genética , Encéfalo/enzimología , Respuesta al Choque por Frío , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Medicamentos Herbarios Chinos/química , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Reacción Cataléptica de Congelación , Frutas , Regulación de la Expresión Génica/efectos de los fármacos , Técnicas de Inactivación de Genes , Lignanos/aislamiento & purificación , Lignanos/farmacología , Lignanos/uso terapéutico , Medicina Kampo , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Schisandra , Natación , Tirosina 3-Monooxigenasa/metabolismo , Pez Cebra , Proteínas de Pez Cebra
2.
J Cell Physiol ; 236(3): 1903-1912, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-32710469

RESUMEN

Macrophage migration is thought to participate in obesity-related cardiovascular diseases. Matrix metalloproteinase-8 (MMP-8) possesses proteolytic activity on the extracellular matrix (ECM), which promotes macrophage migration to the site of vascular injury. Neuropeptide Y (NPY) is a bioactive peptide involved in MMP expression. However, it is uncertain whether NPY can regulate the expression of matrix metalloproteinase-8 (MMP-8) in macrophages. In this study, wild-type C57BL/6 and NPY-/- mice were fed a high-fat diet and subjected to subcutaneous carotid artery injury with ferric chloride, to observe the role of NPY and macrophages in neointima formation. In addition, Raw264.7 cells were treated with NPY and its antagonists to observe MMP-8 expression and macrophage migration. We found that NPY-/- mice exhibited significantly reduced neointima formation after carotid artery injury. The content of macrophages and MMP-8 in the neointima and media were also significantly reduced in NPY-/- mice compared with C57BL/6 mice. Moreover, the expression of MMP-8 in macrophages was also decreased in NPY-/- mice. NPY increased MMP-8 messenger RNA and protein expression in Raw264.7 cells in vitro, and this effect was abrogated by the Y1R antagonist. In addition, NPY increased the phosphorylation of ERK1/2, which was significantly attenuated by co-treatment with the Y1R antagonist. Moreover, NPY-induced MMP-8 expression could be decreased by the ERK1/2 inhibitor PD98059. Furthermore, NPY promoted macrophage migration across type I collagen in vitro. In conclusion, NPY promotes macrophage migration by upregulating MMP-8 expression, which we believe to be an underappreciated mechanism of the increased progression of neointima formation.


Asunto(s)
Movimiento Celular , Macrófagos/citología , Macrófagos/enzimología , Metaloproteinasa 8 de la Matriz/metabolismo , Neuropéptido Y/metabolismo , Animales , Activación Enzimática , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neointima/metabolismo , Neointima/patología , Neuropéptido Y/deficiencia , Placa Aterosclerótica/patología , Células RAW 264.7 , Receptores de Neuropéptido/antagonistas & inhibidores , Receptores de Neuropéptido/metabolismo
3.
Sci Rep ; 10(1): 5913, 2020 04 03.
Artículo en Inglés | MEDLINE | ID: mdl-32246073

RESUMEN

Neuropeptide Y (NPY) controls energy homeostasis including orexigenic actions in mammalians and non-mammalians. Recently, NPY has attracted attention as a mediator of emotional behaviour and psychosomatic diseases. However, its functions are not fully understood. We established npy gene-deficient (NPY-KO) zebrafish (Danio rerio) to assess the relationship between NPY and emotional behaviours. The NPY-KO zebrafish exhibited similar growth, but pomc and avp mRNA levels in the brain were higher as compared to wild-type fish. NPY-KO zebrafish exhibited several anxiety-like behaviours, such as a decrease in social interaction in mirror test and decreased locomotion in black-white test. The acute cold stress-treated NPY-KO zebrafish exhibited anxiety-like behaviours such as remaining stationary and swimming along the side of the tank in the mirror test. Moreover, expression levels of anxiety-associated genes (orx and cck) and catecholamine production (gr, mr, th1 and th2) were significantly higher in NPY-KO zebrafish than in wild-type fish. We demonstrated that NPY-KO zebrafish have an anxiety phenotype and a stress-vulnerability like NPY-KO mice, whereby orx and/or catecholamine signalling may be involved in the mechanism actions.


Asunto(s)
Ansiedad/genética , Metabolismo Energético/fisiología , Neuropéptido Y/deficiencia , Estrés Psicológico/fisiopatología , Animales , Animales Modificados Genéticamente , Ansiedad/fisiopatología , Ansiedad/psicología , Conducta Animal/fisiología , Catecolaminas/metabolismo , Modelos Animales de Enfermedad , Femenino , Técnicas de Inactivación de Genes , Humanos , Masculino , Neuropéptido Y/genética , Orexinas/metabolismo , Receptores de Neuropéptido Y/metabolismo , Transducción de Señal/fisiología , Pez Cebra , Proteínas de Pez Cebra/metabolismo
4.
J Gerontol A Biol Sci Med Sci ; 75(6): 1042-1049, 2020 05 22.
Artículo en Inglés | MEDLINE | ID: mdl-30561530

RESUMEN

The orexigenic hormone neuropeptide Y (NPY) plays a pivotal role in the peripheral regulation of fat metabolism. However, the mechanisms underlying the effects of sex on NPY function have not been extensively analyzed. In this study, we examined the effects of NPY deficiency on fat metabolism in male and female mice. Body weight was slightly decreased, whereas white adipose tissue (WAT) mass was significantly decreased as the thermogenic program was upregulated in NPY-/- female mice compared with that in wild-type mice; these factors were not altered in response to NPY deficiency in male mice. Moreover, lack of NPY resulted in an increase in luteinizing hormone (LH) expression in the pituitary gland, with concomitant activation of the estradiol-mediated thermogenic program in inguinal WAT, and alleviated age-related modification of adiposity in female mice. Taken together, these data revealed a novel intracellular mechanism of NPY in the regulation of fat metabolism and highlighted the sexual dimorphism of NPY as a promising target for drug development to reduce postmenopausal adiposity.


Asunto(s)
Tejido Adiposo Blanco/metabolismo , Estradiol/farmacología , Neuropéptido Y/deficiencia , Posmenopausia , Animales , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Femenino , Hormona Luteinizante/metabolismo , Masculino , Ratones , Obesidad/metabolismo , Fenotipo , Hipófisis/metabolismo , Caracteres Sexuales
5.
Neuropeptides ; 80: 101994, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-31740068

RESUMEN

Neuropeptide Y (NPY) producing neurons in the arcuate nucleus (Arc) of the hypothalamus are essential to the regulation of food intake and energy homeostasis. Whilst they have classically been thought to co-express agouti-related peptide (AgRP), it is now clear that there is a sub-population of NPY neurons in the Arc that do not. Here, we show that a subset of AgRP-negative, NPY-positive neurons in the Arc also express neurotensin (NTS) and we use an NTS-Cre line to investigate the function of this sub-population of NPY neurons. The lack of NPY in NTS-positive neurons led to a marked reduction in fat mass and bodyweight as well as a significant reduction in food intake in male NPYlox/lox; NTScre/+ mice compared to controls. Despite the reduction in food intake, overall energy expenditure was similar between genotypes due to concomitant reduction in activity in NPYlox/lox; NTScre/+ mice. Furthermore, cortical bone mass was significantly reduced in NPYlox/lox;NTScre/+ mice with no evident alterations in the cancellous bone compartment, likely due to reduced leptin levels as a result of their reduced adiposity. Taken together, these data suggest that the sub-population of Arc NPY neurons expressing NTS are critical for regulating food intake, activity and fat mass but are not directly involved in the control of bone mass.


Asunto(s)
Peso Corporal/fisiología , Metabolismo Energético/fisiología , Neuronas/metabolismo , Neuropéptido Y/deficiencia , Neurotensina/metabolismo , Proteína Relacionada con Agouti/genética , Proteína Relacionada con Agouti/metabolismo , Animales , Núcleo Arqueado del Hipotálamo/metabolismo , Homeostasis/fisiología , Hipotálamo/metabolismo , Leptina/metabolismo , Ratones Transgénicos , Neuropéptido Y/metabolismo , Fenotipo
6.
Nat Commun ; 10(1): 1318, 2019 03 21.
Artículo en Inglés | MEDLINE | ID: mdl-30899013

RESUMEN

Macroautophagy is an evolutionarily conserved cellular maintenance program, meant to protect the brain from premature aging and neurodegeneration. How neuronal autophagy, usually loosing efficacy with age, intersects with neuronal processes mediating brain maintenance remains to be explored. Here, we show that impairing autophagy in the Drosophila learning center (mushroom body, MB) but not in other brain regions triggered changes normally restricted to aged brains: impaired associative olfactory memory as well as a brain-wide ultrastructural increase of presynaptic active zones (metaplasticity), a state non-compatible with memory formation. Mechanistically, decreasing autophagy within the MBs reduced expression of an NPY-family neuropeptide, and interfering with autocrine NPY signaling of the MBs provoked similar brain-wide metaplastic changes. Our results in an exemplary fashion show that autophagy-regulated signaling emanating from a higher brain integration center can execute high-level control over other brain regions to steer life-strategy decisions such as whether or not to form memories.


Asunto(s)
Envejecimiento/metabolismo , Autofagia/genética , Drosophila melanogaster/metabolismo , Memoria/fisiología , Cuerpos Pedunculados/metabolismo , Neuropéptido Y/genética , Envejecimiento/genética , Animales , Comunicación Autocrina/genética , Proteínas Relacionadas con la Autofagia/antagonistas & inhibidores , Proteínas Relacionadas con la Autofagia/genética , Proteínas Relacionadas con la Autofagia/metabolismo , Encéfalo/citología , Encéfalo/metabolismo , Proteínas de Drosophila/antagonistas & inhibidores , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/crecimiento & desarrollo , Regulación de la Expresión Génica , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Cuerpos Pedunculados/citología , Neuronas/citología , Neuronas/metabolismo , Neuropéptido Y/deficiencia , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Sinapsis/metabolismo , Transmisión Sináptica
7.
J Clin Invest ; 128(9): 3866-3871, 2018 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-30080182

RESUMEN

Hypoglycemia activates the counterregulatory response (CRR), a neural-endocrine reflex that restores euglycemia. Although effective if occasionally activated, repeated induction of the CRR leads to a decline in responsiveness and prolonged exposure to hypoglycemia. The mechanism underlying this impairment is not known. We found that the reduction in epinephrine release that characterizes a suppressed CRR involves a long-lasting form of sympatho-adrenal synaptic plasticity. Using optogenetically evoked catecholamine release, we show that recurrent hypoglycemia reduced the secretory capacity of mouse adrenal chromaffin cells. Single activation of the CRR increased the adrenal levels of tyrosine hydroxylase (TH), the rate-limiting enzyme for catecholamine synthesis, but this was prevented by repeated activation. In contrast, the level of neuropeptide Y (NPY), an adrenal cotransmitter, remained elevated after recurrent hypoglycemia. Inhibition of NPY or Y1 signaling, either transgenically or pharmacologically, prevented the attenuation of both TH expression and epinephrine release. These results indicate that impairment of the CRR involves suppressed activity at the adrenal level. Interfering with the peripheral NPY-dependent negative feedback loop may provide a way to avoid the pathophysiological consequences of recurrent hypoglycemia which are common in the diabetic state.


Asunto(s)
Glándulas Suprarrenales/metabolismo , Hipoglucemia/metabolismo , Animales , Catecolaminas/biosíntesis , Células Cromafines/metabolismo , Epinefrina/metabolismo , Retroalimentación Fisiológica , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Plasticidad Neuronal , Neuropéptido Y/deficiencia , Neuropéptido Y/genética , Neuropéptido Y/metabolismo , Recurrencia , Tirosina 3-Monooxigenasa/metabolismo
8.
Int J Obes (Lond) ; 42(11): 1925-1938, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-29523877

RESUMEN

OBJECTIVE: To determine whether age and neuropeptide Y (NPY) were involved in the skeletal response to extended periods of diet-induced obesity. METHODS: Male wild-type (WT) and NPY null (NPYKO) mice were fed a mild (23% fat) high-fat diet for 10 weeks from 6 or 16 weeks of age. Metabolism and bone density were assessed during feeding. Skeletal changes were assessed by microCT and histomorphometry. RESULTS: High-fat feeding in 6-week-old WT mice led to significantly increased body weight, adiposity and serum leptin levels, accompanied with markedly suppressed cortical bone accrual. NPYKO mice were less susceptible to fat accrual but, importantly, displayed a complete lack of suppression of bone accrual or cortical bone loss. In contrast, when skeletally mature (16 week old) mice underwent 10 weeks of fat feeding, the metabolic response to HFD was similar to younger mice, however bone mass was not affected in either WT or NPYKO. Thus, growing mice are particularly susceptible to the detrimental effects of HFD on bone mass, through suppression of bone accrual involving NPY signalling. CONCLUSION: This study provides new insights into the relationship between the opposing processes of a positive weight/bone relationship and the negative 'metabolic' effect of obesity on bone mass. This negative effect is particularly active in growing skeletons, which have heightened sensitivity to changes in obesity. In addition, NPY is identified as a fundamental driver of this negative 'metabolic' pathway to bone.


Asunto(s)
Remodelación Ósea/fisiología , Hueso Cortical/patología , Neuropéptido Y/deficiencia , Obesidad/patología , Aumento de Peso/fisiología , Animales , Densidad Ósea , Dieta Alta en Grasa/efectos adversos , Modelos Animales de Enfermedad , Ratones , Ratones Endogámicos , Neuropéptido Y/fisiología , Obesidad/metabolismo
9.
Am J Physiol Regul Integr Comp Physiol ; 314(4): R533-R539, 2018 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-29351428

RESUMEN

Although central melanocortin 4 receptor (MC4R) blockade abolishes the central nervous system (CNS)-mediated anorexogenic, antidiabetic, and cardiovascular actions of leptin, chronic MC4R stimulation fails to completely mimic the effects of leptin. Because neuropeptide Y (NPY) and MC4R exert opposite effects on cardiovascular and metabolic functions, we tested the role of NPY in offsetting the long-term actions of MC4R activation. Wild-type (WT) and NPY-deficient (NPY-/-) mice were implanted with telemetry probes for measuring mean arterial pressure (MAP) and heart rate (HR) 24 h/day. After the mice recovered from surgery and stable baseline measurements, the MC3/4R agonist melanotan II (MTII, 120 µg·kg-1·day-1 iv) was infused for 7 days followed by a recovery period. No major differences between groups were observed at baseline except for slightly higher food intake and HR in NPY-/- mice (4.3 ± 0.2 vs. 3.4 ± 0.2 g/day and 567 ± 14 vs. 522 ± 13 beats/min). Chronic MTII infusion reduced food intake in both groups while causing transient increases in MAP and HR only in WT mice (peaks of 11 ± 3 mmHg and 126 ± 13 beats/min). To examine whether NPY deficiency would amplify the antidiabetic effects of MC4R activation, diabetes was induced with streptozotocin (STZ) 1 wk before baseline measurements were taken, and the same experimental protocol was followed. In WT and NPY-/- mice, STZ-induced diabetes led to similar hyperphagia, hyperglycemia, and weight loss, which were not reversed by chronic MTII treatment. Our results demonstrate that chronic MC4R activation, even in NPY-deficient mice, does not mimic chronic antidiabetic, cardiovascular, or metabolic actions of leptin, and that NPY is not essential for hyperphagia or cardiovascular changes associated with diabetes.


Asunto(s)
Depresores del Apetito/administración & dosificación , Regulación del Apetito/efectos de los fármacos , Presión Arterial/efectos de los fármacos , Conducta Animal/efectos de los fármacos , Glucemia/efectos de los fármacos , Diabetes Mellitus Experimental/tratamiento farmacológico , Hipoglucemiantes/administración & dosificación , Neuropéptido Y/deficiencia , Péptidos Cíclicos/administración & dosificación , Receptor de Melanocortina Tipo 4/agonistas , alfa-MSH/análogos & derivados , Animales , Biomarcadores/sangre , Glucemia/metabolismo , Diabetes Mellitus Experimental/sangre , Diabetes Mellitus Experimental/fisiopatología , Diabetes Mellitus Experimental/psicología , Frecuencia Cardíaca/efectos de los fármacos , Infusiones Intravenosas , Insulina/sangre , Leptina/sangre , Masculino , Ratones Noqueados , Neuropéptido Y/genética , Receptor de Melanocortina Tipo 4/metabolismo , Transducción de Señal/efectos de los fármacos , alfa-MSH/administración & dosificación
10.
Aging Cell ; 16(2): 339-348, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28101970

RESUMEN

Neuropeptide Y (NPY) is an orexigenic peptide that plays an essential role in caloric restriction (CR)-mediated lifespan extension. However, the mechanisms underlying the NPY-mediated effects in CR are poorly defined. Here, we report that NPY deficiency in male mice during CR increases mortality in association with lipodystrophy. NPY-/- mice displayed a rapid decrease in body weight and fat mass, as well as increased lipolysis during CR. These alterations in fat regulation were inhibited by the lipolysis inhibitor, acipimox, a treatment associated with reduced mortality. The lipolytic/thermogenic signaling, ß3-adrenergic receptor/hormone sensitive lipase, was markedly activated in white adipose tissue of NPY-/- mice compared with that of NPY+/+ mice, and thermogenesis was controlled by NPY under negative energy balance. These results demonstrate the critical role of NPY in the regulation of lipid metabolic homeostasis and survival via control of lipolysis and thermogenesis in a state of negative energy balance.


Asunto(s)
Adiposidad , Restricción Calórica , Esperanza de Vida , Lipólisis , Neuropéptido Y/metabolismo , Adipocitos/efectos de los fármacos , Adipocitos/metabolismo , Adipocitos/patología , Adipogénesis/efectos de los fármacos , Tejido Adiposo Blanco/efectos de los fármacos , Tejido Adiposo Blanco/metabolismo , Adiposidad/efectos de los fármacos , Animales , Peso Corporal/efectos de los fármacos , Tamaño de la Célula/efectos de los fármacos , Metabolismo Energético/efectos de los fármacos , Lipólisis/efectos de los fármacos , Masculino , Ratones , Modelos Biológicos , Mortalidad , Neuropéptido Y/deficiencia , Pirazinas/farmacología , Receptores Adrenérgicos beta 3/metabolismo , Transducción de Señal/efectos de los fármacos , Esterol Esterasa/metabolismo , Termogénesis/efectos de los fármacos
11.
J Genet Genomics ; 43(7): 421-30, 2016 07 20.
Artículo en Inglés | MEDLINE | ID: mdl-27461754

RESUMEN

Neuropeptide Y (NPY) is widely expressed in the central nervous system and influences many physiological processes. It is located within the rat quantitative trait locus (QTL) for alcohol preference on chromosome 4. Alcohol-nonpreferring (NP) rats consume very little alcohol, but have significantly higher NPY expression in the brain than alcohol-preferring (P) rats. We capitalized on this phenotypic difference by creating an Npy knockout (KO) rat using the inbred NP background to evaluate NPY effects on alcohol consumption. Zinc finger nuclease (ZNF) technology was applied, resulting in a 26-bp deletion in the Npy gene. RT-PCR, Western blotting and immunohistochemistry confirmed the absence of Npy mRNA and protein in KO rats. Alcohol consumption was increased in Npy(+/-) but not Npy(-/-) rats, while Npy(-/-) rats displayed significantly lower body weight when compared to Npy(+/+) rats. In whole brain tissue, expression levels of Npy-related and other alcohol-associated genes, Npy1r, Npy2r, Npy5r, Agrp, Mc3r, Mc4r, Crh and Crh1r, were significantly greater in Npy(-/-) rats, whereas Pomc and Crhr2 expressions were highest in Npy(+/-) rats. These findings suggest that the NPY-system works in close coordination with the melanocortin (MC) and corticotropin-releasing hormone (CRH) systems to modulate alcohol intake and body weight.


Asunto(s)
Consumo de Bebidas Alcohólicas/genética , Peso Corporal/genética , Técnicas de Inactivación de Genes , Neuropéptido Y/deficiencia , Neuropéptido Y/genética , Animales , Conducta Animal , Línea Celular , Ingestión de Alimentos/genética , Ratas
12.
Stem Cells ; 34(8): 2145-56, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27090492

RESUMEN

Hematopoietic stem/progenitor cell (HSPC) mobilization is an essential homeostatic process regulated by the interaction of cellular and molecular components in bone marrow niches. It has been shown by others that neurotransmitters released from the sympathetic nervous system regulate HSPC egress from bone marrow to peripheral blood. In this study, we investigate the functional role of neuropeptide Y (NPY) on this process. NPY deficient mice had significantly impaired HSPC mobilization due to increased expression of HSPC maintenance factors by reduction of matrix metalloproteinase-9 (MMP-9) activity in bone marrow. Pharmacological or endogenous elevation of NPY led to decrease of HSPC maintenance factors expression by activating MMP-9 in osteoblasts, resulting in HSPC mobilization. Mice in which the Y1 receptor was deleted in osteoblasts did not exhibit HSPC mobilization by NPY. Furthermore, NPY treatment in ovariectomized mice caused reduction of bone loss due to HSPC mobilization. These results suggest a new role of NPY on HSPC mobilization, as well as the potential therapeutic application of this neuropeptide for stem cell-based therapy. Stem Cells 2016;34:2145-2156.


Asunto(s)
Movilización de Célula Madre Hematopoyética , Metaloproteinasa 9 de la Matriz/metabolismo , Neuropéptido Y/metabolismo , Osteoblastos/metabolismo , Receptores de Neuropéptido Y/metabolismo , Animales , Huesos/metabolismo , Quimiotaxis , Femenino , Homeostasis , Ratones Endogámicos C57BL , Neuropéptido Y/deficiencia , Osteoblastos/citología , Osteoblastos/enzimología , Receptores CXCR4/metabolismo
13.
Am J Physiol Regul Integr Comp Physiol ; 310(2): R134-42, 2016 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-26561644

RESUMEN

Neuropeptide Y (NPY) in the dorsomedial hypothalamus (DMH) plays an important role in the regulation of energy balance. While DMH NPY overexpression causes hyperphagia and obesity in rats, knockdown of NPY in the DMH via adeno-associated virus (AAV)-mediated RNAi (AAVshNPY) ameliorates these alterations. Whether this knockdown has a therapeutic effect on obesity and glycemic disorder has yet to be determined. The present study sought to test this potential using a rat model of high-fat diet (HFD)-induced obesity and insulin resistance, mimicking human obesity with impaired glucose homeostasis. Rats had ad libitum access to rodent regular chow (RC) or HFD. Six weeks later, an oral glucose tolerance test (OGTT) was performed for verifying HFD-induced glucose intolerance. After verification, obese rats received bilateral DMH injections of AAVshNPY or the control vector AAVshCTL, and OGTT and insulin tolerance test (ITT) were performed at 16 and 18 wk after viral injection (23 and 25 wk on HFD), respectively. Rats were killed at 26 wk on HFD. We found that AAVshCTL rats on HFD remained hyperphagic, obese, glucose intolerant, and insulin resistant relative to lean control RC-fed rats receiving DMH injection of AAVshCTL, whereas these alterations were reversed in NPY knockdown rats fed a HFD. NPY knockdown rats exhibited normal food intake, body weight, glucose tolerance, and insulin sensitivity, as seen in lean control rats. Together, these results demonstrate a therapeutic action of DMH NPY knockdown against obesity and impaired glucose homeostasis in rats, providing a potential target for the treatment of obesity and diabetes.


Asunto(s)
Dieta Alta en Grasa , Técnicas de Silenciamiento del Gen , Intolerancia a la Glucosa/terapia , Hipotálamo Medio/metabolismo , Neuropéptido Y/deficiencia , Obesidad/terapia , Tratamiento con ARN de Interferencia/métodos , Adiposidad , Animales , Glucemia/metabolismo , Peso Corporal , Dependovirus/genética , Modelos Animales de Enfermedad , Regulación hacia Abajo , Ingestión de Alimentos , Metabolismo Energético , Vectores Genéticos , Intolerancia a la Glucosa/sangre , Intolerancia a la Glucosa/genética , Intolerancia a la Glucosa/fisiopatología , Hipotálamo Medio/fisiopatología , Insulina/sangre , Resistencia a la Insulina , Masculino , Neuropéptido Y/genética , Obesidad/sangre , Obesidad/genética , Obesidad/fisiopatología , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Ratas Sprague-Dawley , Factores de Tiempo
14.
BMB Rep ; 48(12): 645-6, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26538251

RESUMEN

The sympathetic nervous system (SNS) or neurotransmitters in the bone marrow microenvironment has been known to regulate hematopoietic stem cell (HSC) functions such as self-renewal, proliferation and differentiation. However, the specific role of neuropeptide Y (NPY) in this process remains relatively unexplored. In this study, we demonstrated that NPY deficient mice have significantly reduced HSC numbers and impaired bone marrow regeneration due to apoptotic destruction of SNS fibers and/or endothelial cells. Moreover, NPY treatment prevented bone marrow impairments in a mouse model of chemotherapy-induced SNS injury, while conditional knockout mice lacking the Y1 receptor in macrophages did not restore bone marrow dysfunction in spite of NPY injection. Transforming growth factor-beta (TGF-ß) secreted by NPY-mediated Y1 receptor stimulation in macrophages plays a key role in neuroprotection and HSC survival in the bone marrow. Therefore, this study reveals a new role of NPY in bone marrow HSC microenvironment, and provides an insight into the therapeutic application of this neuropeptide.


Asunto(s)
Médula Ósea/metabolismo , Células Madre Hematopoyéticas/metabolismo , Neuropéptido Y/metabolismo , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Diferenciación Celular/fisiología , Células Endoteliales/citología , Células Endoteliales/metabolismo , Células Madre Hematopoyéticas/citología , Ratones , Ratones Noqueados , Neuropéptido Y/deficiencia , Receptores de Neuropéptido Y/metabolismo , Nicho de Células Madre
15.
EMBO J ; 34(12): 1648-60, 2015 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-25916827

RESUMEN

Many reports have revealed the importance of the sympathetic nervous system (SNS) in the control of the bone marrow environment. However, the specific role of neuropeptide Y (NPY) in this process has not been systematically studied. Here we show that NPY-deficient mice have significantly reduced hematopoietic stem cell (HSC) numbers and impaired regeneration in bone marrow due to apoptotic destruction of SNS fibers and/or endothelial cells. Furthermore, pharmacological elevation of NPY prevented bone marrow impairments in a mouse model of chemotherapy-induced SNS injury, while NPY injection into conditional knockout mice lacking the Y1 receptor in macrophages did not relieve bone marrow dysfunction. These results indicate that NPY promotes neuroprotection and restores bone marrow dysfunction from chemotherapy-induced SNS injury through the Y1 receptor in macrophages. They also reveal a new role of NPY as a regulator of the bone marrow microenvironment and highlight the potential therapeutic value of this neuropeptide.


Asunto(s)
Células de la Médula Ósea/metabolismo , Médula Ósea/fisiología , Microambiente Celular/fisiología , Células Madre Hematopoyéticas/fisiología , Fibras Nerviosas Mielínicas/metabolismo , Neuropéptido Y/metabolismo , Análisis de Varianza , Animales , Apoptosis/fisiología , Células Endoteliales/fisiología , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Células Madre Hematopoyéticas/metabolismo , Ratones , Ratones Noqueados , Neuropéptido Y/deficiencia , Sistema Nervioso Simpático/citología
16.
Cell Metab ; 18(4): 588-95, 2013 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-24093681

RESUMEN

Agouti-related peptide (AgRP) neurons of the hypothalamus release a fast transmitter (GABA) in addition to neuropeptides (neuropeptide Y [NPY] and Agouti-related peptide [AgRP]). This raises questions as to their respective functions. The acute activation of AgRP neurons robustly promotes food intake, while central injections of AgRP, NPY, or GABA agonist results in the marked escalation of food consumption with temporal variance. Given the orexigenic capability of all three of these neuroactive substances in conjunction with their coexpression in AgRP neurons, we looked to unravel their relative temporal role in driving food intake. After the acute stimulation of AgRP neurons with DREADD technology, we found that either GABA or NPY is required for the rapid stimulation of feeding, and the neuropeptide AgRP, through action on MC4 receptors, is sufficient to induce feeding over a delayed yet prolonged period. These studies help to elucidate the neurochemical mechanisms of AgRP neurons in controlling temporally distinct phases of eating.


Asunto(s)
Proteína Relacionada con Agouti/metabolismo , Neuronas/metabolismo , Neuropéptido Y/metabolismo , Ácido gamma-Aminobutírico/metabolismo , Proteína Relacionada con Agouti/genética , Animales , Clozapina/análogos & derivados , Clozapina/farmacología , Ingestión de Alimentos/efectos de los fármacos , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Neuronas/efectos de los fármacos , Neuropéptido Y/deficiencia , Neuropéptido Y/genética , Receptor de Melanocortina Tipo 4/deficiencia , Receptor de Melanocortina Tipo 4/genética , Receptor de Melanocortina Tipo 4/metabolismo , Transducción de Señal , Proteínas del Transporte Vesicular de Aminoácidos Inhibidores/deficiencia , Proteínas del Transporte Vesicular de Aminoácidos Inhibidores/genética , Proteínas del Transporte Vesicular de Aminoácidos Inhibidores/metabolismo
17.
Br J Pharmacol ; 170(5): 1014-26, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23992467

RESUMEN

BACKGROUND AND PURPOSE: Immune challenge of mice with Bacille Calmette-Guérin (BCG) has been reported to cause transient weight loss and a behavioural sickness response. Although BCG-induced depression involves the kynurenine pathway, weight loss occurs independently of this factor. Because neuropeptide Y (NPY) and peptide YY (PYY) are involved in the regulation of food intake, we hypothesized that they play a role in the BCG-induced weight loss. EXPERIMENTAL APPROACH: Male wild-type, PYY knockout (PYY-/-), NPY knockout (NPY-/-) and NPY-/-;PYY-/- double knockout mice were injected with vehicle or BCG (approximately 10(8) colony-forming units per mouse), and their weight, locomotion, exploration and ingestion were recorded for 2 weeks post-treatment. KEY RESULTS: Deletion of PYY and NPY aggravated the BCG-induced loss of body weight, which was most pronounced in NPY-/-;PYY-/- mice (maximum loss: 15%). The weight loss in NPY-/-;PYY-/- mice did not normalize during the 2 week observation period. BCG suppressed the circadian pattern of locomotion, exploration and food intake. However, these changes took a different time course than the prolonged weight loss caused by BCG in NPY-/-;PYY-/- mice. The effect of BCG to increase circulating IL-6 (measured 16 days post-treatment) remained unaltered by knockout of PYY, NPY or NPY plus PYY. CONCLUSIONS AND IMPLICATIONS: These data show that NPY and PYY are both required to protect from the action of BCG-evoked immune challenge to cause prolonged weight loss and disturb energy balance. The findings attest to an important role of NPY and PYY in orchestrating homeostatic reactions to infection and immune stimulation.


Asunto(s)
Vacuna BCG/toxicidad , Neuropéptido Y/metabolismo , Péptido YY/metabolismo , Pérdida de Peso/efectos de los fármacos , Animales , Vacuna BCG/inmunología , Conducta Animal/efectos de los fármacos , Ritmo Circadiano/efectos de los fármacos , Corticosterona/sangre , Ingestión de Alimentos/efectos de los fármacos , Metabolismo Energético/efectos de los fármacos , Conducta Exploratoria/efectos de los fármacos , Femenino , Interleucina-6/sangre , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Actividad Motora/efectos de los fármacos , Neuropéptido Y/deficiencia , Neuropéptido Y/genética , Péptido YY/deficiencia , Péptido YY/genética , Factores de Tiempo
18.
J Neurosci ; 33(31): 12705-17, 2013 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-23904607

RESUMEN

Acute stress evokes the fight-or-flight reflex, which via release of the catecholamine hormones affects the function of every major organ. Although the reflex is transient, it has lasting consequences that produce an exaggerated response when stress is reexperienced. How this change is encoded is not known. We investigated whether the reflex affects the adrenal component of the sympathetic nervous system, a major branch of the stress response. Mice were briefly exposed to the cold-water forced swim test (FST) which evoked an increase in circulating catecholamines. Although this hormonal response was transient, the FST led to a long-lasting increase in the catecholamine secretory capacity measured amperometrically from chromaffin cells and in the expression of tyrosine hydroxylase. A variety of approaches indicate that these changes are regulated postsynaptically by neuropeptide Y (NPY), an adrenal cotransmitter. Using immunohistochemistry, RT-PCR, and NPY(GFP) BAC mice, we find that NPY is synthesized by all chromaffin cells. Stress failed to increase secretory capacity in NPY knock-out mice. Genetic or pharmacological interference with NPY and Y1 (but not Y2 or Y5) receptor signaling attenuated the stress-induced change in tyrosine hydroxylase expression. These results indicate that, under basal conditions, adrenal signaling is tonically inhibited by NPY, but stress overrides this autocrine negative feedback loop. Because acute stress leads to a lasting increase in secretory capacity in vivo but does not alter sympathetic tone, these postsynaptic changes appear to be an adaptive response. We conclude that the sympathetic limb of the stress response exhibits an activity-dependent form of long-lasting plasticity.


Asunto(s)
Plasticidad Neuronal/fisiología , Neuropéptido Y/metabolismo , Estrés Psicológico/metabolismo , Estrés Psicológico/patología , Sistema Nervioso Simpático/metabolismo , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/genética , Glándulas Suprarrenales/patología , Animales , Animales Recién Nacidos , Células Cromafines/efectos de los fármacos , Células Cromafines/metabolismo , Modelos Animales de Enfermedad , Conducta Exploratoria/efectos de los fármacos , Conducta Exploratoria/fisiología , Estimulantes Ganglionares/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Plasticidad Neuronal/efectos de los fármacos , Plasticidad Neuronal/genética , Neuropéptido Y/deficiencia , Feniletanolamina N-Metiltransferasa/metabolismo , Receptores de Neuropéptido Y/agonistas , Receptores de Neuropéptido Y/antagonistas & inhibidores , Receptores de Neuropéptido Y/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Natación/psicología , Sistema Nervioso Simpático/efectos de los fármacos , Factores de Tiempo
19.
FASEB J ; 27(6): 2244-55, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23457218

RESUMEN

We previously reported that the sympathetic neurotransmitter neuropeptide Y (NPY) is potently angiogenic, primarily through its Y2 receptor, and that endogenous NPY is crucial for capillary angiogenesis in rodent hindlimb ischemia. Here we sought to identify the source of NPY responsible for revascularization and its mechanisms of action. At d 3, NPY(-/-) mice demonstrated delayed recovery of blood flow and limb function, consistent with impaired collateral conductance, while ischemic capillary angiogenesis was reduced (~70%) at d 14. This biphasic temporal response was confirmed by 2 peaks of NPY activation in rats: a transient early increase in neuronally derived plasma NPY and increase in platelet NPY during late-phase recovery. Compared to NPY-null platelets, collagen-activated NPY-rich platelets were more mitogenic (~2-fold vs. ~1.6-fold increase) for human microvascular endothelial cells, and Y2/Y5 receptor antagonists ablated this difference in proliferation. In NPY(+/+) mice, ischemic angiogenesis was prevented by platelet depletion and then restored by transfusion of platelets from NPY(+/+) mice, but not NPY(-/-) mice. In thrombocytopenic NPY(-/-) mice, transfusion of wild-type platelets fully restored ischemia-induced angiogenesis. These findings suggest that neuronally derived NPY accelerates the early response to femoral artery ligation by promoting collateral conductance, while platelet-derived NPY is critical for sustained capillary angiogenesis.


Asunto(s)
Plaquetas/metabolismo , Isquemia/sangre , Neovascularización Fisiológica , Neuropéptido Y/fisiología , Animales , Proliferación Celular , Células Cultivadas , Modelos Animales de Enfermedad , Células Endoteliales/patología , Miembro Posterior , Humanos , Isquemia/genética , Isquemia/fisiopatología , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Noqueados , Neovascularización Fisiológica/genética , Neuropéptido Y/deficiencia , Neuropéptido Y/genética , Ratas , Ratas Wistar
20.
J Bone Miner Res ; 28(4): 886-98, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23044938

RESUMEN

Leptin signaling is required for normal bone homeostasis; however, loss of leptin results in differing effects on cortical and cancellous bone, as well as altered responses between the axial and appendicular regions. Local ß-adrenergic actions are responsible for the greater cancellous bone volume in leptin-deficient (ob/ob) mice; however, the mechanism responsible for the opposing reduction in cortical bone in ob/ob mice is not known. Here we show that blocking the leptin-deficient increase in neuropeptide Y (NPY) expression reverses the cortical bone loss in ob/ob mice. Mice null for both NPY and leptin (NPY(-/-) ob/ob), display greater cortical bone mass in both long-bones and vertebra, with NPY(-/-) ob/ob mice exhibiting thicker and denser cortical bone, associated with greater endocortical and periosteal mineral apposition rate (MAR), compared to ob/ob animals. Importantly, these cortical changes occurred without significant increases in body weight, with NPY(-/-) ob/ob mice showing significantly reduced adiposity compared to ob/ob controls, most likely due to the reduced respiratory exchange ratio seen in these animals. Interestingly, cancellous bone volume was not different between NPY(-/-) ob/ob and ob/ob, suggesting that NPY is not influencing the adrenergic axis. Taken together, this work demonstrates the critical role of NPY signaling in the regulation of bone and energy homeostasis, and more importantly, suggests that reduced leptin levels or leptin resistance, which occurs in obesity, could potentially inhibit cortical bone formation via increased central NPY signaling.


Asunto(s)
Huesos/metabolismo , Leptina/metabolismo , Neuropéptido Y/metabolismo , Absorciometría de Fotón , Adiposidad , Animales , Densidad Ósea , Huesos/anatomía & histología , Huesos/diagnóstico por imagen , Corticosterona/sangre , Ayuno/sangre , Conducta Alimentaria , Fémur/diagnóstico por imagen , Fémur/metabolismo , Fertilidad , Eliminación de Gen , Masculino , Ratones Endogámicos C57BL , Ratones Obesos , Minerales/metabolismo , Neuropéptido Y/deficiencia , Tamaño de los Órganos , Fenotipo , Columna Vertebral/anatomía & histología , Columna Vertebral/diagnóstico por imagen , Columna Vertebral/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...