Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
1.
J Neurosci ; 43(5): 846-862, 2023 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-36564184

RESUMEN

Stress disorders impair sleep and quality of life; however, their pathomechanisms are unknown. Prolactin-releasing peptide (PrRP) is a stress mediator; we therefore hypothesized that PrRP may be involved in the development of stress disorders. PrRP is produced by the medullary A1/A2 noradrenaline (NA) cells, which transmit stress signals to forebrain centers, and by non-NA cells in the hypothalamic dorsomedial nucleus. We found in male rats that both PrRP and PrRP-NA cells innervate melanin-concentrating hormone (MCH) producing neurons in the dorsolateral hypothalamus (DLH). These cells serve as a key hub for regulating sleep and affective states. Ex vivo, PrRP hyperpolarized MCH neurons and further increased the hyperpolarization caused by NA. Following sleep deprivation, intracerebroventricular PrRP injection reduced the number of REM sleep-active MCH cells. PrRP expression in the dorsomedial nucleus was upregulated by sleep deprivation, while downregulated by REM sleep rebound. Both in learned helplessness paradigm and after peripheral inflammation, impaired coping with sustained stress was associated with (1) overactivation of PrRP cells, (2) PrRP protein and receptor depletion in the DLH, and (3) dysregulation of MCH expression. Exposure to stress in the PrRP-insensitive period led to increased passive coping with stress. Normal PrRP signaling, therefore, seems to protect animals against stress-related disorders. PrRP signaling in the DLH is an important component of the PrRP's action, which may be mediated by MCH neurons. Moreover, PrRP receptors were downregulated in the DLH of human suicidal victims. As stress-related mental disorders are the leading cause of suicide, our findings may have particular translational relevance.SIGNIFICANCE STATEMENT Treatment resistance to monoaminergic antidepressants is a major problem. Neuropeptides that modulate the central monoaminergic signaling are promising targets for developing alternative therapeutic strategies. We found that stress-responsive prolactin-releasing peptide (PrRP) cells innervated melanin-concentrating hormone (MCH) neurons that are crucial in the regulation of sleep and mood. PrRP inhibited MCH cell activity and enhanced the inhibitory effect evoked by noradrenaline, a classic monoamine, on MCH neurons. We observed that impaired PrRP signaling led to failure in coping with chronic/repeated stress and was associated with altered MCH expression. We found alterations of the PrRP system also in suicidal human subjects. PrRP dysfunction may underlie stress disorders, and fine-tuning MCH activity by PrRP may be an important part of the mechanism.


Asunto(s)
Hormonas Hipotalámicas , Privación de Sueño , Ratas , Masculino , Humanos , Animales , Hormona Liberadora de Prolactina/farmacología , Hormona Liberadora de Prolactina/metabolismo , Privación de Sueño/metabolismo , Trastornos del Humor/etiología , Calidad de Vida , Ratas Wistar , Hormonas Hipotalámicas/metabolismo , Sueño/fisiología , Neuronas/fisiología , Norepinefrina/metabolismo
2.
Int J Mol Sci ; 25(8)2024 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-38673899

RESUMEN

According to previous studies, the median raphe region (MRR) is known to contribute significantly to social behavior. Besides serotonin, there have also been reports of a small population of dopaminergic neurons in this region. Dopamine is linked to reward and locomotion, but very little is known about its role in the MRR. To address that, we first confirmed the presence of dopaminergic cells in the MRR of mice (immunohistochemistry, RT-PCR), and then also in humans (RT-PCR) using healthy donor samples to prove translational relevance. Next, we used chemogenetic technology in mice containing the Cre enzyme under the promoter of the dopamine transporter. With the help of an adeno-associated virus, designer receptors exclusively activated by designer drugs (DREADDs) were expressed in the dopaminergic cells of the MRR to manipulate their activity. Four weeks later, we performed an extensive behavioral characterization 30 min after the injection of the artificial ligand (Clozapine-N-Oxide). Stimulation of the dopaminergic cells in the MRR decreased social interest without influencing aggression and with an increase in social discrimination. Additionally, inhibition of the same cells increased the friendly social behavior during social interaction test. No behavioral changes were detected in anxiety, memory or locomotion. All in all, dopaminergic cells were present in both the mouse and human samples from the MRR, and the manipulation of the dopaminergic neurons in the MRR elicited a specific social response.


Asunto(s)
Clozapina/análogos & derivados , Neuronas Dopaminérgicas , Conducta Social , Animales , Neuronas Dopaminérgicas/metabolismo , Masculino , Ratones , Humanos , Clozapina/farmacología , Núcleos del Rafe/metabolismo , Conducta Animal , Dopamina/metabolismo , Ratones Endogámicos C57BL
3.
Int J Mol Sci ; 23(3)2022 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-35163282

RESUMEN

The relevance of vasopressin (AVP) of magnocellular origin to the regulation of the endocrine stress axis and related behaviour is still under discussion. We aimed to obtain deeper insight into this process. To rescue magnocellular AVP synthesis, a vasopressin-containing adeno-associated virus vector (AVP-AAV) was injected into the supraoptic nucleus (SON) of AVP-deficient Brattleboro rats (di/di). We compared +/+, di/di, and AVP-AAV treated di/di male rats. The AVP-AAV treatment rescued the AVP synthesis in the SON both morphologically and functionally. It also rescued the peak of adrenocorticotropin release triggered by immune and metabolic challenges without affecting corticosterone levels. The elevated corticotropin-releasing hormone receptor 1 mRNA levels in the anterior pituitary of di/di-rats were diminished by the AVP-AAV-treatment. The altered c-Fos synthesis in di/di-rats in response to a metabolic stressor was normalised by AVP-AAV in both the SON and medial amygdala (MeA), but not in the central and basolateral amygdala or lateral hypothalamus. In vitro electrophysiological recordings showed an AVP-induced inhibition of MeA neurons that was prevented by picrotoxin administration, supporting the possible regulatory role of AVP originating in the SON. A memory deficit in the novel object recognition test seen in di/di animals remained unaffected by AVP-AAV treatment. Interestingly, although di/di rats show intact social investigation and aggression, the SON AVP-AAV treatment resulted in an alteration of these social behaviours. AVP released from the magnocellular SON neurons may stimulate adrenocorticotropin secretion in response to defined stressors and might participate in the fine-tuning of social behaviour with a possible contribution from the MeA.


Asunto(s)
Hormona Adrenocorticotrópica/metabolismo , Núcleo Supraóptico/metabolismo , Vasopresinas/metabolismo , Hormona Adrenocorticotrópica/genética , Animales , Núcleo Basal de Meynert/metabolismo , Encéfalo/metabolismo , Corticosterona/metabolismo , Hormona Liberadora de Corticotropina/metabolismo , Sistema Hipotálamo-Hipofisario/metabolismo , Masculino , Neuronas/metabolismo , Núcleo Hipotalámico Paraventricular/metabolismo , ARN Mensajero/metabolismo , Ratas , Ratas Brattleboro , Conducta Social , Vasopresinas/fisiología
4.
Proc Natl Acad Sci U S A ; 114(10): E2006-E2015, 2017 03 07.
Artículo en Inglés | MEDLINE | ID: mdl-28223495

RESUMEN

The rostral migratory stream (RMS) is viewed as a glia-enriched conduit of forward-migrating neuroblasts in which chemorepulsive signals control the pace of forward migration. Here we demonstrate the existence of a scaffold of neurons that receive synaptic inputs within the rat, mouse, and human fetal RMS equivalents. These neurons express secretagogin, a Ca2+-sensor protein, to execute an annexin V-dependent externalization of matrix metalloprotease-2 (MMP-2) for reconfiguring the extracellular matrix locally. Mouse genetics combined with pharmacological probing in vivo and in vitro demonstrate that MMP-2 externalization occurs on demand and that its loss slows neuroblast migration. Loss of function is particularly remarkable upon injury to the olfactory bulb. Cumulatively, we identify a signaling cascade that provokes structural remodeling of the RMS through recruitment of MMP-2 by a previously unrecognized neuronal constituent. Given the life-long presence of secretagogin-containing neurons in human, this mechanism might be exploited for therapeutic benefit in rescue strategies.


Asunto(s)
Calcio/metabolismo , Metaloproteinasa 2 de la Matriz/genética , Neuroglía/metabolismo , Neuronas/metabolismo , Bulbo Olfatorio/metabolismo , Secretagoginas/genética , Animales , Anexina A5/genética , Anexina A5/metabolismo , Movimiento Celular , Feto , Regulación de la Expresión Génica , Humanos , Masculino , Metaloproteinasa 2 de la Matriz/metabolismo , Ratones , Microtomía , Neuroglía/ultraestructura , Neuronas/ultraestructura , Bulbo Olfatorio/citología , Cultivo Primario de Células , Ratas , Ratas Wistar , Secretagoginas/metabolismo , Sinapsis/metabolismo , Sinapsis/ultraestructura , Técnicas de Cultivo de Tejidos
5.
Int J Obes (Lond) ; 43(4): 917-927, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-29907842

RESUMEN

BACKGROUND/OBJECTIVES: Dysfunction in reward-related aspects of feeding, and consequent overeating in humans, is a major contributor to obesity. Intrauterine undernutrition and overnutrition are among the predisposing factors, but the exact mechanism of how overeating develops is still unclear. Consummatory behavior is regulated by the medial shell (mSh) of the accumbens nucleus (Nac) through direct connections with the rostral part of the lateral hypothalamic area (LHA). Our aim was to investigate whether an altered Nac-LHA circuit may underlie hyperphagic behavior. SUBJECTS/METHODS: Intrauterine protein-restricted (PR) male Wistar rats were used as models for hyperphagia. The experiments were performed using young adult control (normally nourished) and PR animals. Sweet condensed milk (SCM) served as a reward to test consumption and subsequent activation (Fos+) of Nac and LHA neurons. Expression levels of type 1 and 2 dopamine receptors (D1R, D2R) in the Nac, as well as tyrosine hydroxylase (TH) levels in the ventral tegmental area, were determined. The D1R agonist SKF82958 was injected into the mSh-Nac of control rats to test the effect of D1R signaling on SCM intake and neuronal cell activation in the LHA. RESULTS: A group of food reward-representing D1R+ neurons was identified in the mSh-Nac. Activation (Fos+) of these neurons was highly proportional to the consumed palatable food. D1R agonist treatment attenuated SCM intake and diminished the number of SCM-activated cells in the LHA. Hyperphagic PR rats showed increased intake of SCM, reduced D1R expression, and an impaired response to SCM-evoked neuronal activation in the mSh-Nac, accompanied by an elevated number of Fos+ neurons in the LHA compared to controls. CONCLUSIONS: Sensitivity of food reward-representing neurons in the mSh-Nac determines the level of satisfaction that governs cessation of consumption, probably through connections with the LHA. D1R signaling is a key element in this function, and is impaired in obesity-prone rats.


Asunto(s)
Conducta Alimentaria/fisiología , Vías Nerviosas/fisiología , Neuronas/metabolismo , Núcleo Accumbens/fisiopatología , Animales , Modelos Animales de Enfermedad , Femenino , Masculino , Embarazo , Ratas , Ratas Wistar , Recompensa
7.
Cells ; 13(13)2024 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-38994950

RESUMEN

The RFamide peptide family is a group of proteins that share a common C-terminal arginine-phenylalanine-amide motif. To date, the family comprises five groups in mammals: neuropeptide FF, LPXRFamides/RFamide-related peptides, prolactin releasing peptide, QRFP, and kisspeptins. Different RFamide peptides have their own cognate receptors and are produced by different cell populations, although they all can also bind to neuropeptide FF receptors with different affinities. RFamide peptides function in the brain as neuropeptides regulating key aspects of homeostasis such as energy balance, reproduction, and cardiovascular function. Furthermore, they are involved in the organization of the stress response including modulation of pain. Considering the interaction between stress and various parameters of homeostasis, the role of RFamide peptides may be critical in the development of stress-related neuropathologies. This review will therefore focus on the role of RFamide peptides as possible key hubs in stress and stress-related psychopathologies. The neurotransmitter coexpression profile of RFamide-producing cells is also discussed, highlighting its potential functional significance. The development of novel pharmaceutical agents for the treatment of stress-related disorders is an ongoing need. Thus, the importance of RFamide research is underlined by the emergence of peptidergic and G-protein coupled receptor-based therapeutic targets in the pharmaceutical industry.


Asunto(s)
Encéfalo , Neuropéptidos , Estrés Psicológico , Humanos , Neuropéptidos/metabolismo , Animales , Encéfalo/metabolismo , Encéfalo/patología , Estrés Psicológico/metabolismo
8.
J Biol Chem ; 287(37): 31540-50, 2012 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-22787147

RESUMEN

In the vascular system angiotensin II (Ang II) causes vasoconstriction via the activation of type 1 angiotensin receptors. Earlier reports have shown that in cellular expression systems diacylglycerol produced during type 1 angiotensin receptor signaling can be converted to 2-arachidonoylglycerol, an important endocannabinoid. Because activation of CB(1) cannabinoid receptors (CB(1)R) induces vasodilation and reduces blood pressure, we have tested the hypothesis that Ang II-induced 2-arachidonoylglycerol release can modulate its vasoconstrictor action in vascular tissue. Rat and mouse skeletal muscle arterioles and mouse saphenous arteries were isolated, pressurized, and subjected to microangiometry. Vascular expression of CB(1)R was demonstrated using Western blot and RT-PCR. In accordance with the functional relevance of these receptors WIN55212, a CB(1)R agonist, caused vasodilation, which was absent in CB(1)R knock-out mice. Inhibition of CB(1)Rs using O2050, a neutral antagonist, enhanced the vasoconstrictor effect of Ang II in wild type but not in CB(1)R knock-out mice. Inverse agonists of CB(1)R (SR141716 and AM251) and inhibition of diacylglycerol lipase using tetrahydrolipstatin also augmented the Ang II-induced vasoconstriction, suggesting that endocannabinoid release modulates this process via CB(1)R activation. This effect was independent of nitric-oxide synthase activity and endothelial function. These data demonstrate that Ang II stimulates vascular endocannabinoid formation, which attenuates its vasoconstrictor effect, suggesting that endocannabinoid release from the vascular wall and CB(1)R activation reduces the vasoconstrictor and hypertensive effects of Ang II.


Asunto(s)
Angiotensina II/metabolismo , Arterias/metabolismo , Endocannabinoides/metabolismo , Endotelio Vascular/metabolismo , Músculo Esquelético/metabolismo , Receptor Cannabinoide CB1/metabolismo , Vasoconstricción/fisiología , Analgésicos/farmacología , Angiotensina II/genética , Animales , Benzoxazinas/farmacología , Endocannabinoides/antagonistas & inhibidores , Endocannabinoides/genética , Hipertensión/genética , Hipertensión/metabolismo , Masculino , Ratones , Ratones Noqueados , Morfolinas/farmacología , Músculo Esquelético/irrigación sanguínea , Naftalenos/farmacología , Piperidinas/farmacología , Pirazoles/farmacología , Ratas , Ratas Wistar , Receptor Cannabinoide CB1/genética , Rimonabant , Vasoconstricción/efectos de los fármacos
9.
Front Neurosci ; 16: 828571, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35386592

RESUMEN

Intrauterine growth retardation (IUGR) poses a high risk for developing late-onset, non-obese type 2 diabetes (T2DM). The exact mechanism underlying this phenomenon is unknown, although the contribution of the central nervous system is recognized. The main hypothalamic nuclei involved in the homeostatic regulation express nesfatin-1, an anorexigenic neuropeptide and identified regulator of blood glucose level. Using intrauterine protein restricted rat model (PR) of IUGR, we investigated, whether IUGR alters the function of nesfatin-1. We show that PR rats develop fat preference and impaired glucose homeostasis by adulthood, while the body composition and caloric intake of normal nourished (NN) and PR rats are similar. Plasma nesfatin-1 levels are unaffected by IUGR in both neonates and adults, but pro-nesfatin-1 mRNA expression is upregulated in the hypothalamus of adult PR animals. We find that centrally injected nesfatin-1 inhibits the fasting induced neuronal activation in the hypothalamic arcuate nucleus in adult NN rats. This effect of nesfatin-1 is not seen in PR rats. The anorexigenic effect of centrally injected nesfatin-1 is also reduced in adult PR rats. Moreover, chronic central nesfatin-1 administration improves the glucose tolerance and insulin sensitivity in NN rats but not in PR animals. Birth dating of nesfatin-1 cells by bromodeoxyuridine (BrDU) reveals that formation of nesfatin-1 cells in the hypothalamus of PR rats is disturbed. Our results suggest that adult PR rats acquire hypothalamic nesfatin-1-resistance, probably due to the altered development of the hypothalamic nesfatin-1 cells. Hypothalamic nesfatin-1-resistance, in turn, may contribute to the development of non-obese type T2DM.

10.
Neurosci Lett ; 765: 136274, 2021 11 20.
Artículo en Inglés | MEDLINE | ID: mdl-34592370

RESUMEN

Ample evidence indicates that in several mammalian species the pineal body contains neurons. In adult white albino rats neurons are not present in the pineal body; however, in perinatal rats many neurons were described. It was demonstrated that in adult mammalian species the pineal neurons contained some neuropeptides and neurotransmitters such as leu-enkephalin, met-enkephalin, substance-P, somatostatin and γ-aminobutiric acid. Oxytocin, vasopressin mRNAs and peptides were also demonstrated. No data are available on the chemical nature of the neurons in perinatal rats. In the present experiment we used immunohistochemistry to clarify this issue. After paraformaldehyde fixation frozen sections were prepared and stained for immunoreactivities of several neuropeptides and neurotransmitters. Dopamine ß-hydroxylase, neuropeptide-Y, vesicular acetylcholine transporter, vesicular glutamate transporter and calcitonin gene-related peptide antibodies were able to stain fibers. According to previous data these fibers may be sympathetic, parasympathetic or sensory. Vesicular glutamate transporter antibody may stain pinealocytes as well. Some cells were immunoreactive for substance-P, oxytocin, vasopressin, leu-enkefalin and glutamic acid decarboxylase. These immnoreactivities showed colocalization with neuron-specific nuclear protein immunoreactivity indicating that these cells were neurons. Calbindin was observed in oval and elongated cells resembling pinealocytes. Based on the results obtained in adult mammals, the pineal neurons may be analogue to retinal ganglion cells, or they may function as interneurons in the retino-pinealo-retinal neuronal circuit or peptidergic neurons may influence pinealocytes in a paracrine manner.


Asunto(s)
Neuronas/citología , Neuropéptidos/análisis , Neurotransmisores/análisis , Glándula Pineal/química , Glándula Pineal/citología , Animales , Animales Recién Nacidos , Femenino , Masculino , Neuronas/metabolismo , Glándula Pineal/metabolismo , Ratas
11.
Cells ; 10(4)2021 03 24.
Artículo en Inglés | MEDLINE | ID: mdl-33805075

RESUMEN

Angiotensin II (Ang II) has various cardiac effects and causes vasoconstriction. Ang II activates the type-1 angiotensin receptor-Gq/11 signaling pathway resulting in the release of 2-arachidonoylglycerol (2-AG). We aimed to investigate whether cardiac Ang II effects are modulated by 2-AG-release and to identify the role of type-1 cannabinoid receptors (CB1R) in these effects. Expression of CB1R in rat cardiac tissue was confirmed by immunohistochemistry. To characterize short-term Ang II effects, increasing concentrations of Ang II (10-9-10-7 M); whereas to assess tachyphylaxis, repeated infusions of Ang II (10-7 M) were administered to isolated Langendorff-perfused rat hearts. Ang II infusions caused a decrease in coronary flow and ventricular inotropy, which was more pronounced during the first administration. CB agonist 2-AG and WIN55,212-2 administration to the perfusate enhanced coronary flow. The flow-reducing effect of Ang II was moderated in the presence of CB1R blocker O2050 and diacylglycerol-lipase inhibitor Orlistat. Our findings indicate that Ang II-induced cardiac effects are modulated by simultaneous CB1R-activation, most likely due to 2-AG-release during Ang II signalling. In this combined effect, the response to 2-AG via cardiac CB1R may counteract the positive inotropic effect of Ang II, which may decrease metabolic demand and augment Ang II-induced coronary vasoconstriction.


Asunto(s)
Angiotensina II/farmacología , Endocannabinoides/metabolismo , Corazón/efectos de los fármacos , Receptor Cannabinoide CB1/metabolismo , Animales , Ácidos Araquidónicos/farmacología , Circulación Coronaria/efectos de los fármacos , Endocannabinoides/farmacología , Glicéridos/farmacología , Lipoproteína Lipasa/antagonistas & inhibidores , Lipoproteína Lipasa/metabolismo , Masculino , Contracción Miocárdica/efectos de los fármacos , Orlistat/farmacología , Ratas Sprague-Dawley , Receptor Cannabinoide CB1/agonistas , Receptor Cannabinoide CB1/antagonistas & inhibidores
12.
Eur J Neurosci ; 31(2): 302-14, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20074225

RESUMEN

We report that satiation evokes neuronal activity in the ventral subdivision of the hypothalamic dorsomedial nucleus (DMH) as indicated by increased c-fos expression in response to refeeding in fasted rats. The absence of significant Fos activation following food presentation without consumption suggests that satiation but not craving for food elicits the activation of ventral DMH neurons. The distribution pattern of the prolactin-releasing peptide (PrRP)-immunoreactive (ir) network showed remarkable correlations with the distribution of activated neurons within the DMH. The PrRP-ir fibers and terminals were immunolabeled with tyrosine hydroxylase, suggesting their origin in lower brainstem instead of local, hypothalamic PrRP cells. PrRP-ir fibers arising from neurons of the nucleus of the solitary tract could be followed to the hypothalamus. Unilateral transections of these fibers at pontine and caudal hypothalamic levels resulted in a disappearance of the dense PrRP-ir network in the ventral DMH while PrRP immunoreactivity was increased in transected fibers caudal to the knife cuts as well as in perikarya of the nucleus of the solitary tract ipsilateral to the transections. In accord with these changes, the number of Fos-expressing neurons following refeeding declined in the ipsilateral but remained high in the contralateral DMH. However, the Fos response in the ventral DMH was not attenuated following chemical lesion (neonatal monosodium glutamate treatment) of the hypothalamic arcuate nucleus, another possible source of DMH inputs. These findings suggest that PrRP projections from the nucleus of the solitary tract contribute to the activation of ventral DMH neurons during refeeding, possibly by transferring information on cholecystokinin-mediated satiation.


Asunto(s)
Núcleo Hipotalámico Dorsomedial/citología , Núcleo Hipotalámico Dorsomedial/metabolismo , Ingestión de Alimentos , Ayuno , Vías Nerviosas , Neuronas/metabolismo , Núcleo Solitario , Animales , Conducta Alimentaria/fisiología , Aditivos Alimentarios/farmacología , Masculino , Vías Nerviosas/anatomía & histología , Vías Nerviosas/fisiología , Neuronas/citología , Neuronas/efectos de los fármacos , Proteínas Oncogénicas v-fos/genética , Proteínas Oncogénicas v-fos/metabolismo , Hormona Liberadora de Prolactina/metabolismo , Ratas , Ratas Wistar , Glutamato de Sodio/farmacología , Núcleo Solitario/anatomía & histología , Núcleo Solitario/fisiología
13.
Blood ; 111(12): 5544-52, 2008 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-18268092

RESUMEN

Granulocyte colony-stimulating factor (G-CSF) induces proliferation of bone marrow-derived cells. G-CSF is neuroprotective after experimental brain injury, but the mechanisms involved remain unclear. Stem cell factor (SCF) is a cytokine important for the survival and differentiation of hematopoietic stem cells. Its receptor (c-kit or CD117) is present in some endothelial cells. We aimed to determine whether the combination of G-CSF/SCF induces angiogenesis in the central nervous system by promoting entry of endothelial precursors into the injured brain and causing them to proliferate there. We induced permanent middle cerebral artery occlusion in female mice that previously underwent sex-mismatched bone marrow transplantation from enhanced green fluorescent protein (EGFP)-expressing mice. G-CSF/SCF treatment reduced infarct volumes by more than 50% and resulted in a 1.5-fold increase in vessel formation in mice with stroke, a large percentage of which contain endothelial cells of bone marrow origin. Most cells entering the brain maintained their bone marrow identity and did not transdifferentiate into neural cells. G-CSF/SCF treatment also led to a 2-fold increase in the number of newborn cells in the ischemic hemisphere. These findings suggest that G-CSF/SCF treatment might help recovery through induction of bone marrow-derived angiogenesis, thus improving neuronal survival and functional outcome.


Asunto(s)
Trasplante de Médula Ósea , Isquemia Encefálica/tratamiento farmacológico , Células Endoteliales/citología , Factor Estimulante de Colonias de Granulocitos/farmacología , Factor de Células Madre/farmacología , Animales , Isquemia Encefálica/patología , División Celular/efectos de los fármacos , Quimioterapia Combinada , Células Endoteliales/efectos de los fármacos , Femenino , Proteínas Fluorescentes Verdes , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Infarto de la Arteria Cerebral Media/patología , Ratones , Ratones Endogámicos C57BL , Neovascularización Fisiológica/efectos de los fármacos , Recuperación de la Función/efectos de los fármacos
14.
Brain Struct Funct ; 225(3): 969-984, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-32200401

RESUMEN

Chronic hypernatremia activates the central osmoregulatory mechanisms and inhibits the function of the hypothalamic-pituitary-adrenal (HPA) axis. Noradrenaline (NE) release into the periventricular anteroventral third ventricle region (AV3V), the supraoptic (SON) and hypothalamic paraventricular nuclei (PVN) from efferents of the caudal ventrolateral (cVLM) and dorsomedial (cDMM) medulla has been shown to be essential for the hypernatremia-evoked responses and for the HPA response to acute restraint. Notably, the medullary NE cell groups highly coexpress prolactin-releasing peptide (PrRP) and nesfatin-1/NUCB2 (nesfatin), therefore, we assumed they contributed to the reactions to chronic hypernatremia. To investigate this, we compared two models: homozygous Brattleboro rats with hereditary diabetes insipidus (DI) and Wistar rats subjected to chronic high salt solution (HS) intake. HS rats had higher plasma osmolality than DI rats. PrRP and nesfatin mRNA levels were higher in both models, in both medullary regions compared to controls. Elevated basal tyrosine hydroxylase (TH) expression and impaired restraint-induced TH, PrRP and nesfatin expression elevations in the cVLM were, however, detected only in HS, but not in DI rats. Simultaneously, only HS rats exhibited classical signs of chronic stress and severely blunted hormonal reactions to acute restraint. Data suggest that HPA axis responsiveness to restraint depends on the type of hypernatremia, and on NE capacity in the cVLM. Additionally, NE and PrRP signalization primarily of medullary origin is increased in the SON, PVN and AV3V in HS rats. This suggests a cooperative action in the adaptation responses and designates the AV3V as a new site for PrRP's action in hypernatremia.


Asunto(s)
Adaptación Fisiológica , Hipernatremia/fisiopatología , Hipotálamo/fisiopatología , Bulbo Raquídeo/fisiopatología , Nucleobindinas/fisiología , Hormona Liberadora de Prolactina/fisiología , Animales , Masculino , Nucleobindinas/análisis , Hormona Liberadora de Prolactina/análisis , Ratas Brattleboro , Ratas Wistar , Estrés Psicológico/metabolismo , Tirosina 3-Monooxigenasa/análisis
15.
J Neurosci ; 27(1): 141-51, 2007 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-17202481

RESUMEN

Vasopressin (VP) transcription in the rat suprachiasmatic nucleus (SCN) in organotypic culture was studied by in situ hybridization histochemistry using an intron-specific VP heteronuclear RNA probe. The circadian peak of VP gene transcription in the SCN in vitro is completely blocked by a 2 h exposure to tetrodotoxin (TTX) in the culture medium, and this TTX inhibition of VP gene transcription is reversed by exposure of the SCN to either forskolin or potassium depolarization. This suggests that an intrinsic, spontaneously active neuronal mechanism in the SCN is responsible for the cAMP- and depolarization-dependent pathways involved in maintaining peak VP gene transcription. In this paper, we evaluate a variety of neurotransmitter candidates, membrane receptors, and signal-transduction cascades that might constitute the mechanisms responsible for the peak of VP gene transcription. We find that vasoactive intestinal peptide (VIP) and a VPAC2 (VIP receptor subtype 2) receptor-specific agonist, Ro-25-1553, are the most effective ligands tested in evoking a cAMP-mitogen-activated protein kinase signal transduction cascade leading to an increase in VP gene transcription in the SCN. In addition, a second independent pathway involving depolarization activating L-type voltage-gated calcium channels and a Ca-dependent kinase pathway [inhibited by KN62 (1-[N,O-bis(5-isoquinolinesulphonyl)-N-methyl-L-tyrosyl]-4-phenylpiperazine)] rescues VP gene transcription in the presence of TTX. In the absence of TTX, these independent pathways appear to act in a cooperative manner to generate the circadian peak of VP gene transcription in the SCN.


Asunto(s)
Regulación de la Expresión Génica/fisiología , Potenciales de la Membrana/fisiología , Neurotransmisores/metabolismo , Receptores de Vasopresinas/metabolismo , Núcleo Supraquiasmático/fisiología , Transmisión Sináptica/fisiología , Vasopresinas/metabolismo , Animales , Células Cultivadas , Ratas , Ratas Sprague-Dawley
16.
J Neurochem ; 104(3): 653-66, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18199117

RESUMEN

In this study, we investigated the effect of chronic repeated restraint (RR) on prolactin-releasing peptide (PrRP) expression. In the brainstem, where PrRP colocalize with norepinephrine in neurons of the A1 and A2 catecholaminergic cell groups, the expression of tyrosine hydroxylase (TH) has also been examined. In the brainstem, but not in the hypothalamus, the basal PrRP expression in female rats was higher than that in the males that was abolished by ovariectomy. RR evoked an elevation of PrRP expression in all areas investigated, with smaller reaction in the brainstems of females. There was no gender-related difference in the RR-evoked TH expression. Elevation of PrRP was relatively higher than elevation of TH, causing a shift in PrRP/TH ratio in the brainstem after RR. Estrogen alpha receptors were found in the PrRP neurons of the A1 and A2 cell groups, but not in the hypothalamus. Bilateral lesions of the hypothalamic paraventricular nucleus did not prevent RR-evoked changes. Elevated PrRP production parallel with increased PrRP/TH ratio in A1/A2 neurons indicate that: (i) there is a clear difference in the regulation of TH and PrRP expression after RR, and (ii) among other factors this may also contribute to the changed sensitivity of the hypothalamo-pituitary-adrenal axis during chronic stress.


Asunto(s)
Encéfalo/metabolismo , Hormonas Hipotalámicas/metabolismo , Neuropéptidos/metabolismo , Restricción Física/efectos adversos , Caracteres Sexuales , Estrés Psicológico/patología , Tirosina 3-Monooxigenasa/metabolismo , Análisis de Varianza , Animales , Corticosterona/sangre , Femenino , Regulación de la Expresión Génica/fisiología , Masculino , Ovariectomía/métodos , Núcleo Hipotalámico Paraventricular/lesiones , Núcleo Hipotalámico Paraventricular/metabolismo , Prolactina/sangre , Hormona Liberadora de Prolactina , Ratas , Ratas Wistar , Estrés Psicológico/sangre
17.
Stem Cells ; 25(11): 2820-6, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17656643

RESUMEN

The uterine endometrium is composed of epithelial and stromal cells, which undergo extensive degeneration and regeneration in every estrous cycle, and dramatic changes occur during pregnancy. The high turnover of cells requires a correspondingly high level of cell division by progenitor cells in the uterus, but the character and source of these cells remain obscure. In the present study, using a novel transgenic mouse, we showed that CD45-positive hematopoietic progenitor cells colonize the uterine epithelium and that in pregnancy more than 80% of the epithelium can derive from these cells. Since we also found green fluorescent protein (GFP)-positive uterine endothelial cells in long-term GFP bone marrow-transplanted mice, we conclude that circulating CD45+ cells play an important role in regenerating the uterine epithelium.


Asunto(s)
Diferenciación Celular , Células Epiteliales/citología , Células Madre Hematopoyéticas/metabolismo , Antígenos Comunes de Leucocito/biosíntesis , Antígenos Comunes de Leucocito/sangre , Útero/citología , Envejecimiento/genética , Animales , Diferenciación Celular/genética , Células Epiteliales/fisiología , Femenino , Células Madre Hematopoyéticas/fisiología , Antígenos Comunes de Leucocito/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Embarazo , Útero/fisiología
18.
Free Radic Biol Med ; 116: 41-49, 2018 02 20.
Artículo en Inglés | MEDLINE | ID: mdl-29278739

RESUMEN

The p22phox protein is an essential component of the phagocytic- and inner ear NADPH oxidases but its relationship to other Nox proteins is less clear. We have studied the role of p22phox in the TGF-ß1-stimulated H2O2 production of primary human and murine fibroblasts. TGF-ß1 induced H2O2 release of the examined cells, and the response was dependent on the expression of both Nox4 and p22phox. Interestingly, the p22phox protein was present in the absence of any detectable Nox/Duox expression, and the p22phox level was unaffected by TGF-ß1. On the other hand, Nox4 expression was dependent on the presence of p22phox, establishing an asymmetrical relationship between the two proteins. Nox4 and p22phox proteins localized to the endoplasmic reticulum and their distribution was unaffected by TGF-ß1. We used a chemically induced protein dimerization method to study the orientation of p22phox and Nox4 in the endoplasmic reticulum membrane. This technique is based on the rapamycin-mediated heterodimerization of the mammalian FRB domain with the FK506 binding protein. The results of these experiments suggest that the enzyme complex produces H2O2 into the lumen of the endoplasmic reticulum, indicating that Nox4 contributes to the development of the oxidative milieu within this organelle.


Asunto(s)
Grupo Citocromo b/metabolismo , Retículo Endoplásmico/metabolismo , Fibroblastos/fisiología , Complejos Multiproteicos/metabolismo , NADPH Oxidasa 4/metabolismo , NADPH Oxidasas/metabolismo , Animales , Grupo Citocromo b/genética , Dimerización , Células HeLa , Humanos , Peróxido de Hidrógeno/metabolismo , Ratones , Ratones Mutantes , NADPH Oxidasa 4/genética , NADPH Oxidasas/genética , Oxidación-Reducción , Unión Proteica , Especies Reactivas de Oxígeno/metabolismo , Sirolimus/metabolismo , Factor de Crecimiento Transformador beta1/inmunología
19.
Sci Rep ; 8(1): 3490, 2018 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-29472575

RESUMEN

Altered pain sensations such as hyperalgesia and allodynia are characteristic features of various pain states, and remain difficult to treat. We have shown previously that spinal application of dipeptidyl peptidase 4 (DPP4) inhibitors induces strong antihyperalgesic effect during inflammatory pain. In this study we observed low level of DPP4 mRNA in the rat spinal dorsal horn in physiological conditions, which did not change significantly either in carrageenan-induced inflammatory or partial nerve ligation-generated neuropathic states. In naïve animals, microglia and astrocytes expressed DPP4 protein with one and two orders of magnitude higher than neurons, respectively. DPP4 significantly increased in astrocytes during inflammation and in microglia in neuropathy. Intrathecal application of two DPP4 inhibitors tripeptide isoleucin-prolin-isoleucin (IPI) and the antidiabetic drug vildagliptin resulted in robust opioid-dependent antihyperalgesic effect during inflammation, and milder but significant opioid-independent antihyperalgesic action in the neuropathic model. The opioid-mediated antihyperalgesic effect of IPI was exclusively related to mu-opioid receptors, while vildagliptin affected mainly delta-receptor activity, although mu- and kappa-receptors were also involved. None of the inhibitors influenced allodynia. Our results suggest pathology and glia-type specific changes of DPP4 activity in the spinal cord, which contribute to the development and maintenance of hyperalgesia and interact with endogenous opioid systems.


Asunto(s)
Dipeptidil Peptidasa 4/genética , Hiperalgesia/tratamiento farmacológico , Inflamación/tratamiento farmacológico , Neuralgia/tratamiento farmacológico , Analgésicos Opioides/administración & dosificación , Animales , Astrocitos/efectos de los fármacos , Linaje de la Célula/genética , Inhibidores de la Dipeptidil-Peptidasa IV/administración & dosificación , Hiperalgesia/genética , Hiperalgesia/patología , Inflamación/genética , Inflamación/patología , Masculino , Neuralgia/genética , Neuralgia/patología , Neuroglía/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Ratas Wistar , Receptores Opioides kappa/genética , Receptores Opioides mu , Médula Espinal/efectos de los fármacos , Médula Espinal/patología
20.
J Comp Neurol ; 505(4): 443-58, 2007 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-17912746

RESUMEN

Hibernation results in dramatic changes in body temperature and metabolism; however, the central nervous system remains active during deep torpor. By cloning c-fos cDNA from the 13-lined ground squirrel (Spermophilus tridecemlineatus) and using squirrel c-fos mRNA probe for in situ hybridization histochemistry, we systematically analyzed and identified specific brain regions that were activated during six different phases of the hibernation bout. During entrance into torpor, we detected activation of the ventrolateral subdivision of the medial preoptic area ('thermoregulatory center'), and the reticular thalamic nucleus, which is known to inhibit the somatomotor cortex. During torpor, c-fos expression in the cortex was suppressed while the reticular thalamic nucleus remained uniformly active. Throughout torpor the suprachiasmatic nucleus ('biological clock') showed increasing activity, likely participating in phase-change regulation of the hibernation bout. Interestingly, during torpor very strong c-fos activation was seen in the epithelial cells of the choroid plexus and in tanycytes at the third ventricle, both peaking near the beginning of arousal. In arousal, activity of the suprachiasmatic and reticular thalamic nuclei and choroid epithelial cells diminished, while ependymal cells in the lateral and fourth ventricles showed stronger activity. Increasing body temperature during arousal was driven by the activation of neurons in the medial part of the preoptic area. In interbout awake animals, we demonstrated the activation of hypothalamic neurons located in the arcuate nucleus and the dorsolateral hypothalamus, areas involved in food intake. Our observations indicate that the hibernation bout is closely regulated and orchestrated by specific regions of the central nervous system. J. Comp. Neurol. 505:443-458, 2007. (c) 2007 Wiley-Liss, Inc.


Asunto(s)
Encéfalo/fisiología , Hibernación/fisiología , Proteínas Proto-Oncogénicas c-fos/metabolismo , Sciuridae/fisiología , Animales , Nivel de Alerta/fisiología , Ritmo Circadiano/fisiología , Hibridación in Situ , Proteínas Proto-Oncogénicas c-fos/genética , ARN Mensajero/análisis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA