Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 381
Filtrar
1.
Nature ; 628(8009): 826-834, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38538787

RESUMO

Empirical evidence suggests that heat exposure reduces food intake. However, the neurocircuit architecture and the signalling mechanisms that form an associative interface between sensory and metabolic modalities remain unknown, despite primary thermoceptive neurons in the pontine parabrachial nucleus becoming well characterized1. Tanycytes are a specialized cell type along the wall of the third ventricle2 that bidirectionally transport hormones and signalling molecules between the brain's parenchyma and ventricular system3-8. Here we show that tanycytes are activated upon acute thermal challenge and are necessary to reduce food intake afterwards. Virus-mediated gene manipulation and circuit mapping showed that thermosensing glutamatergic neurons of the parabrachial nucleus innervate tanycytes either directly or through second-order hypothalamic neurons. Heat-dependent Fos expression in tanycytes suggested their ability to produce signalling molecules, including vascular endothelial growth factor A (VEGFA). Instead of discharging VEGFA into the cerebrospinal fluid for a systemic effect, VEGFA was released along the parenchymal processes of tanycytes in the arcuate nucleus. VEGFA then increased the spike threshold of Flt1-expressing dopamine and agouti-related peptide (Agrp)-containing neurons, thus priming net anorexigenic output. Indeed, both acute heat and the chemogenetic activation of glutamatergic parabrachial neurons at thermoneutrality reduced food intake for hours, in a manner that is sensitive to both Vegfa loss-of-function and blockage of vesicle-associated membrane protein 2 (VAMP2)-dependent exocytosis from tanycytes. Overall, we define a multimodal neurocircuit in which tanycytes link parabrachial sensory relay to the long-term enforcement of a metabolic code.


Assuntos
Tronco Encefálico , Células Ependimogliais , Comportamento Alimentar , Temperatura Alta , Hipotálamo , Vias Neurais , Neurônios , Animais , Feminino , Masculino , Camundongos , Proteína Relacionada com Agouti/metabolismo , Núcleo Arqueado do Hipotálamo/metabolismo , Núcleo Arqueado do Hipotálamo/citologia , Tronco Encefálico/citologia , Tronco Encefálico/fisiologia , Dopamina/metabolismo , Ingestão de Alimentos/fisiologia , Células Ependimogliais/citologia , Células Ependimogliais/fisiologia , Comportamento Alimentar/fisiologia , Ácido Glutâmico/metabolismo , Hipotálamo/citologia , Hipotálamo/fisiologia , Vias Neurais/metabolismo , Neurônios/metabolismo , Núcleos Parabraquiais/citologia , Núcleos Parabraquiais/metabolismo , Núcleos Parabraquiais/fisiologia , Sensação Térmica/fisiologia , Fatores de Tempo , Fator A de Crescimento do Endotélio Vascular/líquido cefalorraquidiano , Fator A de Crescimento do Endotélio Vascular/metabolismo
2.
J Physiol ; 601(4): 801-829, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36696965

RESUMO

Prolonged high-fat diet (HFD) exposure is associated with hyperphagia, excess caloric intake and weight gain. After initial exposure to a HFD, a brief (24-48 h) period of hyperphagia is followed by the regulation of caloric intake and restoration of energy balance within an acute (3-5 day) period. Previous studies have demonstrated this occurs via a vagally mediated signalling cascade that increases glutamatergic transmission via activation of NMDA receptors located on gastric-projecting neurons of the dorsal motor nucleus of the vagus (DMV). The present study used electrophysiological recordings from thin brainstem slice preparations, in vivo recordings of gastric motility and tone, measurement of gastric emptying rates, and food intake studies to investigate the hypothesis that activation of brainstem astrocytes in response to acute HFD exposure is responsible for the increased glutamatergic drive to DMV neurons and the restoration of caloric balance. Pharmacological and chemogenetic inhibition of brainstem astrocytes reduced glutamatergic signalling and DMV excitability, dysregulated gastric tone and motility, attenuated the homeostatic delay in gastric emptying, and prevented the decrease in food intake that is observed during the period of energy regulation following initial exposure to HFD. Understanding the mechanisms involved in caloric regulation may provide critical insights into energy balance as well as into the hyperphagia that develops as these mechanisms are overcome. KEY POINTS: Initial exposure to a high fat diet is associated with a brief period of hyperphagia before caloric intake and energy balance is restored. This period of homeostatic regulation is associated with a vagally mediated signalling cascade that increases glutamatergic transmission to dorsal motor nucleus of the vagus (DMV) neurons via activation of synaptic NMDA receptors. The present study demonstrates that pharmacological and chemogenetic inhibition of brainstem astrocytes reduced glutamatergic signalling and DMV neuronal excitability, dysregulated gastric motility and tone and emptying, and prevented the regulation of food intake following high-fat diet exposure. Astrocyte regulation of glutamatergic transmission to DMV neurons appears to involve release of the gliotransmitters glutamate and ATP. Understanding the mechanisms involved in caloric regulation may provide critical insights into energy balance as well as into the hyperphagia that develops as these mechanisms are overcome.


Assuntos
Astrócitos , Ingestão de Energia , Hiperfagia , Animais , Ratos , Astrócitos/fisiologia , Tronco Encefálico/citologia , Ratos Sprague-Dawley , Receptores de N-Metil-D-Aspartato , Nervo Vago/fisiologia , Dieta Hiperlipídica
3.
Nature ; 609(7928): 761-771, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-36071158

RESUMO

Infections induce a set of pleiotropic responses in animals, including anorexia, adipsia, lethargy and changes in temperature, collectively termed sickness behaviours1. Although these responses have been shown to be adaptive, the underlying neural mechanisms have not been elucidated2-4. Here we use of a set of unbiased methodologies to show that a specific subpopulation of neurons in the brainstem can control the diverse responses to a bacterial endotoxin (lipopolysaccharide (LPS)) that potently induces sickness behaviour. Whole-brain activity mapping revealed that subsets of neurons in the nucleus of the solitary tract (NTS) and the area postrema (AP) acutely express FOS after LPS treatment, and we found that subsequent reactivation of these specific neurons in FOS2A-iCreERT2 (also known as TRAP2) mice replicates the behavioural and thermal component of sickness. In addition, inhibition of LPS-activated neurons diminished all of the behavioural responses to LPS. Single-nucleus RNA sequencing of the NTS-AP was used to identify LPS-activated neural populations, and we found that activation of ADCYAP1+ neurons in the NTS-AP fully recapitulates the responses elicited by LPS. Furthermore, inhibition of these neurons significantly diminished the anorexia, adipsia and locomotor cessation seen after LPS injection. Together these studies map the pleiotropic effects of LPS to a neural population that is both necessary and sufficient for canonical elements of the sickness response, thus establishing a critical link between the brain and the response to infection.


Assuntos
Tronco Encefálico , Comportamento de Doença , Neurônios , Animais , Anorexia/complicações , Área Postrema/citologia , Área Postrema/metabolismo , Tronco Encefálico/citologia , Tronco Encefálico/efeitos dos fármacos , Tronco Encefálico/fisiologia , Comportamento de Doença/efeitos dos fármacos , Letargia/complicações , Lipopolissacarídeos/farmacologia , Camundongos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/metabolismo , Proteínas Proto-Oncogênicas c-fos/metabolismo , Núcleo Solitário/citologia , Núcleo Solitário/metabolismo
4.
Nat Commun ; 12(1): 5809, 2021 10 04.
Artigo em Inglês | MEDLINE | ID: mdl-34608167

RESUMO

SARS-CoV-2 has caused a global pandemic of COVID-19 since its emergence in December 2019. The infection causes a severe acute respiratory syndrome and may also spread to central nervous system leading to neurological sequelae. We have developed and characterized two new organotypic cultures from hamster brainstem and lung tissues that offer a unique opportunity to study the early steps of viral infection and screening antivirals. These models are not dedicated to investigate how the virus reaches the brain. However, they allow validating the early tropism of the virus in the lungs and demonstrating that SARS-CoV-2 could infect the brainstem and the cerebellum, mainly by targeting granular neurons. Viral infection induces specific interferon and innate immune responses with patterns specific to each organ, along with cell death by apoptosis, necroptosis, and pyroptosis. Overall, our data illustrate the potential of rapid modeling of complex tissue-level interactions during infection by a newly emerged virus.


Assuntos
Tronco Encefálico/virologia , Pulmão/virologia , Modelos Biológicos , SARS-CoV-2/patogenicidade , Monofosfato de Adenosina/análogos & derivados , Monofosfato de Adenosina/farmacologia , Alanina/análogos & derivados , Alanina/farmacologia , Células Epiteliais Alveolares/virologia , Animais , Antivirais/farmacologia , Tronco Encefálico/citologia , Tronco Encefálico/imunologia , Tronco Encefálico/patologia , Cricetinae , Imunidade Inata , Inflamação , Pulmão/citologia , Pulmão/imunologia , Pulmão/patologia , Neurônios/virologia , Técnicas de Cultura de Órgãos , Morte Celular Regulada , SARS-CoV-2/efeitos dos fármacos , Tropismo Viral
5.
Nature ; 589(7842): 426-430, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33268898

RESUMO

Among numerous challenges encountered at the beginning of extrauterine life, the most celebrated is the first breath that initiates a life-sustaining motor activity1. The neural systems that regulate breathing are fragile early in development, and it is not clear how they adjust to support breathing at birth. Here we identify a neuropeptide system that becomes activated immediately after birth and supports breathing. Mice that lack PACAP selectively in neurons of the retrotrapezoid nucleus (RTN) displayed increased apnoeas and blunted CO2-stimulated breathing; re-expression of PACAP in RTN neurons corrected these breathing deficits. Deletion of the PACAP receptor PAC1 from the pre-Bötzinger complex-an RTN target region responsible for generating the respiratory rhythm-phenocopied the breathing deficits observed after RTN deletion of PACAP, and suppressed PACAP-evoked respiratory stimulation in the pre-Bötzinger complex. Notably, a postnatal burst of PACAP expression occurred in RTN neurons precisely at the time of birth, coinciding with exposure to the external environment. Neonatal mice with deletion of PACAP in RTN neurons displayed increased apnoeas that were further exacerbated by changes in ambient temperature. Our findings demonstrate that well-timed PACAP expression by RTN neurons provides an important supplementary respiratory drive immediately after birth and reveal key molecular components of a peptidergic neural circuit that supports breathing at a particularly vulnerable period in life.


Assuntos
Tronco Encefálico/fisiologia , Parto/fisiologia , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/metabolismo , Respiração , Animais , Apneia/metabolismo , Tronco Encefálico/citologia , Dióxido de Carbono/metabolismo , Feminino , Masculino , Camundongos , Neurônios/metabolismo , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/deficiência , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/genética , Receptores de Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/deficiência , Receptores de Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/genética , Receptores de Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/metabolismo
6.
J Neurosci ; 40(49): 9364-9371, 2020 12 02.
Artigo em Inglês | MEDLINE | ID: mdl-33122390

RESUMO

Mechanosensitivity is a well-known feature of astrocytes, however, its underlying mechanisms and functional significance remain unclear. There is evidence that astrocytes are acutely sensitive to decreases in cerebral perfusion pressure and may function as intracranial baroreceptors, tuned to monitor brain blood flow. This study investigated the mechanosensory signaling in brainstem astrocytes, as these cells reside alongside the cardiovascular control circuits and mediate increases in blood pressure and heart rate induced by falls in brain perfusion. It was found that mechanical stimulation-evoked Ca2+ responses in astrocytes of the rat brainstem were blocked by (1) antagonists of connexin channels, connexin 43 (Cx43) blocking peptide Gap26, or Cx43 gene knock-down; (2) antagonists of TRPV4 channels; (3) antagonist of P2Y1 receptors for ATP; and (4) inhibitors of phospholipase C or IP3 receptors. Proximity ligation assay demonstrated interaction between TRPV4 and Cx43 channels in astrocytes. Dye loading experiments showed that mechanical stimulation increased open probability of carboxyfluorescein-permeable membrane channels. These data suggest that mechanosensory Ca2+ responses in astrocytes are mediated by interaction between TRPV4 and Cx43 channels, leading to Cx43-mediated release of ATP which propagates/amplifies Ca2+ signals via P2Y1 receptors and Ca2+ recruitment from the intracellular stores. In astrocyte-specific Cx43 knock-out mice the magnitude of heart rate responses to acute increases in intracranial pressure was not affected by Cx43 deficiency. However, these animals displayed lower heart rates at different levels of cerebral perfusion, supporting the hypothesis of connexin hemichannel-mediated release of signaling molecules by astrocytes having an excitatory action on the CNS sympathetic control circuits.SIGNIFICANCE STATEMENT There is evidence suggesting that astrocytes may function as intracranial baroreceptors that play an important role in the control of systemic and cerebral circulation. To function as intracranial baroreceptors, astrocytes must possess a specialized membrane mechanism that makes them exquisitely sensitive to mechanical stimuli. This study shows that opening of connexin 43 (Cx43) hemichannels leading to the release of ATP is the key central event underlying mechanosensory Ca2+ responses in astrocytes. This astroglial mechanism plays an important role in the autonomic control of heart rate. These data add to the growing body of evidence suggesting that astrocytes function as versatile surveyors of the CNS metabolic milieu, tuned to detect conditions of potential metabolic threat, such as hypoxia, hypercapnia, and reduced perfusion.


Assuntos
Astrócitos/fisiologia , Mecanotransdução Celular/fisiologia , Trifosfato de Adenosina/metabolismo , Animais , Pressão Sanguínea/efeitos dos fármacos , Tronco Encefálico/citologia , Tronco Encefálico/efeitos dos fármacos , Tronco Encefálico/fisiologia , Sinalização do Cálcio/efeitos dos fármacos , Sinalização do Cálcio/fisiologia , Circulação Cerebrovascular/fisiologia , Conexina 43/antagonistas & inibidores , Conexina 43/genética , Feminino , Frequência Cardíaca/fisiologia , Masculino , Mecanotransdução Celular/efeitos dos fármacos , Camundongos , Camundongos Knockout , Peptídeos/antagonistas & inibidores , Peptídeos/genética , Estimulação Física , Ratos , Receptores Purinérgicos P2Y1/efeitos dos fármacos , Canais de Cátion TRPV/antagonistas & inibidores , Canais de Cátion TRPV/genética
7.
Transl Psychiatry ; 10(1): 357, 2020 10 21.
Artigo em Inglês | MEDLINE | ID: mdl-33087695

RESUMO

The neural circuits regulating motivation and movement include midbrain dopaminergic neurons and associated inhibitory GABAergic and excitatory glutamatergic neurons in the anterior brainstem. Differentiation of specific subtypes of GABAergic and glutamatergic neurons in the mouse embryonic brainstem is controlled by a transcription factor Tal1. This study characterizes the behavioral and neurochemical changes caused by the absence of Tal1 function. The Tal1cko mutant mice are hyperactive, impulsive, hypersensitive to reward, have learning deficits and a habituation defect in a novel environment. Only minor changes in their dopaminergic system were detected. Amphetamine induced striatal dopamine release and amphetamine induced place preference were normal in Tal1cko mice. Increased dopamine signaling failed to stimulate the locomotor activity of the Tal1cko mice, but instead alleviated their hyperactivity. Altogether, the Tal1cko mice recapitulate many features of the attention and hyperactivity disorders, suggesting a role for Tal1 regulated developmental pathways and neural structures in the control of motivation and movement.


Assuntos
Transtorno do Deficit de Atenção com Hiperatividade , Tronco Encefálico/citologia , Neurônios Dopaminérgicos , Proteína 1 de Leucemia Linfocítica Aguda de Células T/genética , Anfetamina/farmacologia , Animais , Transtorno do Deficit de Atenção com Hiperatividade/genética , Mesencéfalo , Camundongos
8.
Life Sci ; 234: 116784, 2019 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-31445026

RESUMO

Tobacco smoking is recognized as a life-threatening risk factor worldwide. Initiation of smoking primarily occurs during adolescence which is a critical developmental phase characterized by specific neurobehavioral alterations. The effect of adolescent nicotine exposure on vulnerability to opioid addiction has not been previously addressed. Furthermore, lateral paragigantocellularis (LPGi) is a key modulator of opiate effects. In this study we investigated the effect of adolescent nicotine treatment on development of morphine tolerance and dependence as well as LPGi neuronal responses to morphine during adulthood. Male Wistar rats received subcutaneous injections of either nicotine or saline during adolescence and then development of morphine tolerance and dependence was assessed during adulthood by tail-flick and withdrawal tests, respectively. In vivo single-unit recording was performed to examine the LPGi neuronal activities. Results indicated that adolescent nicotine exposure significantly facilitates the development of tolerance to analgesic effect of morphine and increases the expression of morphine withdrawal signs in adulthood. Also, it was observed that following adolescent nicotine treatment, the extent of morphine-induced excitation is attenuated in LPGi neurons of adult rats. Moreover, the onset of morphine-induced inhibition was increased in these animals. Neither the baseline, nor the regularity of firing was affected in our observations. It could be concluded that nicotine challenge during adolescence may enhance the future vulnerability to opioid addiction through induction of persistent neuroadaptations in LPGi neurons.


Assuntos
Tronco Encefálico/efeitos dos fármacos , Dependência de Morfina/etiologia , Neurônios/efeitos dos fármacos , Nicotina/efeitos adversos , Envelhecimento , Animais , Tronco Encefálico/citologia , Tronco Encefálico/fisiopatologia , Masculino , Dependência de Morfina/fisiopatologia , Neurônios/patologia , Transtornos Relacionados ao Uso de Opioides/etiologia , Transtornos Relacionados ao Uso de Opioides/fisiopatologia , Ratos Wistar , Síndrome de Abstinência a Substâncias/etiologia , Síndrome de Abstinência a Substâncias/fisiopatologia
9.
Sci Rep ; 9(1): 5280, 2019 03 27.
Artigo em Inglês | MEDLINE | ID: mdl-30918315

RESUMO

Convincing evidence demonstrated impairment of the blood-spinal cord barrier (BSCB) in Amyotrophic Lateral Sclerosis (ALS), mainly by endothelial cell (EC) alterations. Replacing damaged ECs by cell transplantation is a potential barrier repair strategy. Recently, we showed that intravenous (iv) administration of human bone marrow CD34+ (hBM34+) cells into symptomatic ALS mice benefits BSCB restoration and postpones disease progression. However, delayed effect on motor function and some severely damaged capillaries were noted. We hypothesized that hematopoietic cells from a restricted lineage would be more effective. This study aimed to establish the effects of human bone marrow-derived endothelial progenitor cells (hBMEPCs) systemically transplanted into G93A mice at symptomatic disease stage. Results showed that transplanted hBMEPCs significantly improved behavioral disease outcomes, engrafted widely into capillaries of the gray/white matter spinal cord and brain motor cortex/brainstem, substantially restored capillary ultrastructure, significantly decreased EB extravasation into spinal cord parenchyma, meaningfully re-established perivascular astrocyte end-feet, and enhanced spinal cord motor neuron survival. These results provide novel evidence that transplantation of hBMEPCs effectively repairs the BSCB, potentially preventing entry of detrimental peripheral factors, including immune/inflammatory cells, which contribute to motor neuron dysfunction. Transplanting EC progenitor cells may be a promising strategy for barrier repair therapy in this disease.


Assuntos
Esclerose Lateral Amiotrófica/metabolismo , Esclerose Lateral Amiotrófica/fisiopatologia , Células Progenitoras Endoteliais/citologia , Células Progenitoras Endoteliais/metabolismo , Neurônios Motores/fisiologia , Esclerose Lateral Amiotrófica/patologia , Animais , Tronco Encefálico/citologia , Tronco Encefálico/metabolismo , Modelos Animais de Doenças , Progressão da Doença , Humanos , Imuno-Histoquímica , Masculino , Camundongos , Microscopia Eletrônica , Córtex Motor/citologia , Córtex Motor/metabolismo , Medula Espinal/citologia , Medula Espinal/metabolismo , Células-Tronco/citologia , Células-Tronco/metabolismo
10.
Exp Neurol ; 314: 46-57, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30653967

RESUMO

Neural stem cells (NSCs) can differentiate into both neurons and glia after transplantation into spinal cord injury (SCI) sites. The neuronal component of stem cell grafts has the potential to form functional synaptic relays across the lesion site. The glial component may reform a blood-spinal cord barrier, support neuronal function, and contribute to remyelination. We performed a long-term, 1.5-year time course study focused on astrocyte migration, differentiation, integration, and safety following human NSC transplantation into C5 hemisection sites in immunodeficient rats. NSCs that adopted a neuronal fate did not migrate from the lesion site. In contrast, transplanted cells that adopted astrocyte fates exhibited long distance migration from the lesion site through host white matter in rostrocaudal directions. These cells migrated slowly at a mean rate of 2-3 mm/month, divided as they migrated, and gradually differentiated into astrocytes. After 1.5 years, human astrocytes migrated nine spinal cord segments, caudally to the mid-thoracic level, and rostrally into the brainstem. The migrated human astrocytes joined the endogenous population of astrocytes in the host spinal cord, extended perivascular endfeet towards host pericytes and endothelium, formed interspecies and intraspecies perivascular astrocytic networks connected by gap junctions, and expressed glutamate transporter proteins in perisynaptic processes, suggesting structural and functional integration. No adverse consequences of this extended glial migration were detected. Adjacent to the lesion site, chronic host astrocytic upregulation was significantly attenuated by NSC grafts. Thus, human astrocytes can migrate long distances from sites of SCI and safely integrate into the host central nervous system. SIGNIFICANCE STATEMENT: Neural stem cell (NSC) grafts are a candidate therapy for spinal cord injury (SCI). Here we report an 18-month study of astrocyte survival and migration from sites of SCI in immunodeficient rats. NSC grafts significantly attenuate host astrocyte reactivity at the lesion/host interface. Intra-graft astrocytes integrate into the host blood-spinal cord barrier (BSCB) and widely express glutamate transporter proteins characteristic of neurotransmitter regulation. Notably, astrocytic components of NSC grafts exhibit gradual yet extensive migration after implantation into the mid-cervical injury site; neurons do not migrate at all. This extensive astrocyte migration is not detectably associated with adverse outcomes anatomically or behaviorally.


Assuntos
Astrócitos , Movimento Celular/fisiologia , Células-Tronco Neurais , Traumatismos da Medula Espinal/terapia , Animais , Tronco Encefálico/citologia , Sobrevivência Celular , Feminino , Junções Comunicantes , Xenoenxertos , Humanos , Hospedeiro Imunocomprometido , Rede Nervosa/citologia , Células-Tronco Neurais/metabolismo , Neurogênese , Ratos , Ratos Nus , Medula Espinal/citologia , Traumatismos da Medula Espinal/patologia , Traumatismos da Medula Espinal/psicologia , Transplante de Células-Tronco , Proteínas Vesiculares de Transporte de Glutamato/metabolismo
11.
Cytokine ; 113: 185-194, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30539780

RESUMO

INTRODUCTION: A growing body of evidence indicates that brain cytokines are involved in the control of the cardiovascular system. Tumour necrosis factor (TNF) is an archetypal cytokine, which exerts its proinflammatory actions via type 1 receptor (TNFR1). Interleukin 10 (IL-10) plays a critical anti-inflammatory role by binding to its receptor (IL-10Ra). The orchestrated inflammatory response is largely dependent on an intricate balance between proinflammatory and anti-inflammatory cytokines and expression of their receptors. AIM: In the study we evaluated the expression of the cytokines and their receptors in the brains of spontaneously hypertensive (SH) and normotensive Wistar-Kyoto (WKY) rats, and how the cytokines affect arterial blood pressure. METHODS: In SH and WKY rats we recorded systolic blood pressure with tail cuff method and measured concentration of TNF, IL-10, TNFR1, and IL-10Ra in the serum, the brainstem, and the hypothalamus; we also measured serum concentrations of copeptin, a surrogate of vasopressin release, angiotensin II and norepinephrine. We immunostained brainstem sections for TNFR1, IL-10Ra, neurons, astrocytes and microglia for confocal imaging. In urethane anaesthetized SH and WKY rats, we invasively recorded blood pressure response to intracerebroventricular (IVC) infusion of TNF or IL-10. We also pharmacologically evaluated baroreflex with phenylephrine and chemoreflex with cyanide in SH and WKY rats. RESULTS: Compared to WKY rats, SH rats had: (1) higher blood pressure; (2) blunted baroreflex and augmented peripheral chemoreflex; (3) greater pressor response to ICV infused TNF and greater hypotensive response to ICV infused IL-10; (4) higher concentration of TNF in the ventral and dorsal aspects of the medulla oblongata; (5) higher expression of TNFR1 in the dorsal medulla; (6) higher concentration of IL-10 in both aspects of the medulla; (7) lower expression of IL-10Ra in the dorsal medulla. Confocal imaging showed co-localization of TNFR1 and IL-10Ra with neurons, astrocytes and microglia in both SH and WKY rats. The concentration of the cytokines and their receptors were significantly higher in the brain than in the serum. There were no significant differences in the concentration of the cytokines and their receptors in the hypothalamic region and in the serum between SH and WKY rats. Serum concentrations of norepinephrine, angiotensin II and copeptin were similar between SH and WKY rats. CONCLUSIONS: Taken together, these findings suggest the presence of a potent milieu for effective TNF signalling in the brainstem, which is associated with the hypertensive phenotype and enhanced hemodynamic response to intrabrain administration of the cytokines. In addition, we hypothesize that the increased IL-10 concentration in the brainstem is a compensatory mechanism for the upregulated TNF system.


Assuntos
Pressão Sanguínea , Tronco Encefálico/metabolismo , Hipotálamo/metabolismo , Interleucina-10/sangue , Receptores de Interleucina-10/sangue , Receptores Tipo I de Fatores de Necrose Tumoral/sangue , Fator de Necrose Tumoral alfa/sangue , Animais , Astrócitos/citologia , Astrócitos/metabolismo , Tronco Encefálico/citologia , Hipotálamo/citologia , Microglia/citologia , Microglia/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Ratos , Ratos Endogâmicos SHR , Ratos Endogâmicos WKY , Transdução de Sinais
12.
Nat Neurosci ; 21(9): 1229-1238, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30104734

RESUMO

Voluntary urination ensures that waste is eliminated when safe and socially appropriate, even without a pressing urge. Uncontrolled urination, or incontinence, is a common problem with few treatment options. Normal urine release requires a small region in the brainstem known as Barrington's nucleus (Bar), but specific neurons that relax the urethral sphincter and enable urine flow are unknown. Here we identify a small subset of Bar neurons that control the urethral sphincter in mice. These excitatory neurons express estrogen receptor 1 (BarESR1), project to sphincter-relaxing interneurons in the spinal cord and are active during natural urination. Optogenetic stimulation of BarESR1 neurons rapidly initiates sphincter bursting and efficient voiding in anesthetized and behaving animals. Conversely, optogenetic and chemogenetic inhibition reveals their necessity in motivated urination behavior. The identification of these cells provides an expanded model for the control of urination and its dysfunction.


Assuntos
Tronco Encefálico/fisiologia , Neurônios/fisiologia , Uretra/inervação , Uretra/fisiologia , Micção/fisiologia , Animais , Tronco Encefálico/citologia , Eletromiografia , Receptor alfa de Estrogênio/genética , Receptor alfa de Estrogênio/fisiologia , Masculino , Camundongos , Vias Neurais/fisiologia , Odorantes , Optogenética , Transtornos Urinários/genética , Transtornos Urinários/fisiopatologia
13.
Sci Rep ; 8(1): 8536, 2018 06 04.
Artigo em Inglês | MEDLINE | ID: mdl-29867218

RESUMO

The Alanine-Serine-Cysteine-1 transporter (SLC7A10, Asc-1) has been shown to play a role in synaptic availability of glycine although the exact mechanism remains unclear. We used electrophysiological recordings and biochemical experiments to investigate the role of Asc-1 transporter in glycinergic transmission in the brainstem respiratory network. Using both the Asc-1 substrate and transportable inhibitor D-isoleucine (D-Ile), and the non-transportable Asc-1 blocker Lu AE00527 (Lu), we found that D-Ile reduces glycinergic transmission and increases glycine release via hetero-exchange, whereas Lu has no acute effect on glycinergic synaptic transmission. Furthermore, D-Ile increases the frequency and reduces amplitude of the phrenic nerve activity in the arterially-perfused working heart brainstem preparation. These results suggest a role of Asc-1 in modulating presynaptic glycine levels that can impact on the respiratory network.


Assuntos
Sistema y+ de Transporte de Aminoácidos/metabolismo , Tronco Encefálico/metabolismo , Glicina/metabolismo , Neurônios/metabolismo , Respiração , Transmissão Sináptica , Sistema y+ de Transporte de Aminoácidos/antagonistas & inibidores , Animais , Tronco Encefálico/citologia , Camundongos , Camundongos Transgênicos , Neurônios/citologia
14.
J Neurosci ; 38(31): 6841-6853, 2018 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-29934352

RESUMO

The internal environment of an organism must remain stable to ensure optimal performance and ultimately survival. The generation of motivated behaviors is an adaptive mechanism for defending homeostasis. Although physiological state modulates motivated behaviors, the influence of physiological state on phasic dopamine signaling, an underlying neurobiological substrate of reward-driven behavior, is underexplored. Here, we use sodium depletion and water restriction, manipulations of body fluid homeostasis, to determine the flexibility and specificity of dopamine responses. Changes in dopamine concentration were measured using fast-scan cyclic voltammetry in the nucleus accumbens shell of male rats in response to intraoral infusions of fluids that either satisfied or did not satisfy homeostatic need. Increases in dopamine concentration during intraoral infusions were observed only under conditions of physiological deficit. Furthermore, dopamine increases were selective and limited to those that satisfied the need state of the animal. Thus, dopamine neurons track fluid balance and respond to salt and water stimuli in a state- and taste-dependent manner. Using Fluoro-Gold tracing and immunohistochemistry for c-Fos and Foxp2, a marker of sodium-deprivation responsive neurons, we revealed brainstem populations of neurons that are activated by sodium depletion and project directly to the ventral tegmental area. The identified projections may modulate dopamine neuron excitability and consequently the state-specific dopamine release observed in our experiments. This work illustrates the impact of physiological state on mesolimbic dopamine signaling and a potential circuit by which homeostatic disruptions are communicated to mesolimbic circuitry to drive the selective reinforcement of biologically-required stimuli under conditions of physiological need.SIGNIFICANCE STATEMENT Motivated behaviors arise during physiological need and are highly selective for homeostasis-restoring stimuli. Although phasic dopamine signaling has been shown to contribute to the generation of motivated behaviors, the state and stimulus specificity of phasic dopamine signaling is less clear. These studies use thirst and sodium appetite to show that dopamine neurons dynamically track body fluid homeostasis and respond to water and salt stimuli in a state- and taste-dependent manner. We also identify hindbrain sodium deprivation-responsive neurons that project directly to the ventral tegmental area, where dopamine neuron cell bodies reside. This work demonstrates command of homeostasis over dopamine signaling and proposes a circuit by which physiological need drives motivated behavior by state- and taste-selective recruitment of phasic dopamine signaling.


Assuntos
Dopamina/fisiologia , Comportamento de Ingestão de Líquido/fisiologia , Núcleo Accumbens/fisiologia , Paladar/fisiologia , Privação de Água/fisiologia , Equilíbrio Hidroeletrolítico/fisiologia , Vias Aferentes/fisiologia , Animais , Apetite/fisiologia , Tronco Encefálico/citologia , Dieta Hipossódica , Eletrodos Implantados , Furosemida/farmacologia , Homeostase , Masculino , Motivação , Natriurese/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-fos/análise , Ratos , Ratos Sprague-Dawley , Recompensa , Área Tegmentar Ventral/fisiologia
15.
Neurosci Bull ; 34(3): 485-496, 2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-29557546

RESUMO

The GABAergic neurons in the parafacial zone (PZ) play an important role in sleep-wake regulation and have been identified as part of a sleep-promoting center in the brainstem, but the long-range connections mediating this function remain poorly characterized. Here, we performed whole-brain mapping of both the inputs and outputs of the GABAergic neurons in the PZ of the mouse brain. We used the modified rabies virus EnvA-ΔG-DsRed combined with a Cre/loxP gene-expression strategy to map the direct monosynaptic inputs to the GABAergic neurons in the PZ, and found that they receive inputs mainly from the hypothalamic area, zona incerta, and parasubthalamic nucleus in the hypothalamus; the substantia nigra, pars reticulata and deep mesencephalic nucleus in the midbrain; and the intermediate reticular nucleus and medial vestibular nucleus (parvocellular part) in the pons and medulla. We also mapped the axonal projections of the PZ GABAergic neurons with adeno-associated virus, and defined the reciprocal connections of the PZ GABAergic neurons with their input and output nuclei. The newly-found inputs and outputs of the PZ were also listed compared with the literature. This cell-type-specific neuronal whole-brain mapping of the PZ GABAergic neurons may reveal the circuits underlying various functions such as sleep-wake regulation.


Assuntos
Mapeamento Encefálico , Tronco Encefálico/citologia , Encéfalo/anatomia & histologia , Neurônios GABAérgicos/fisiologia , Vias Neurais/fisiologia , Proteínas Vesiculares de Transporte de Aminoácidos Inibidores/metabolismo , Animais , Axônios/fisiologia , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fator 1 de Elongação de Peptídeos/genética , Fator 1 de Elongação de Peptídeos/metabolismo , Vírus da Raiva/genética , Vírus da Raiva/metabolismo , Transdução Genética , Proteínas Vesiculares de Transporte de Aminoácidos Inibidores/genética
16.
Nat Commun ; 9(1): 370, 2018 01 25.
Artigo em Inglês | MEDLINE | ID: mdl-29371650

RESUMO

Astrocytes are implicated in modulation of neuronal excitability and synaptic function, but it remains unknown if these glial cells can directly control activities of motor circuits to influence complex behaviors in vivo. This study focused on the vital respiratory rhythm-generating circuits of the preBötzinger complex (preBötC) and determined how compromised function of local astrocytes affects breathing in conscious experimental animals (rats). Vesicular release mechanisms in astrocytes were disrupted by virally driven expression of either the dominant-negative SNARE protein or light chain of tetanus toxin. We show that blockade of vesicular release in preBötC astrocytes reduces the resting breathing rate and frequency of periodic sighs, decreases rhythm variability, impairs respiratory responses to hypoxia and hypercapnia, and dramatically reduces the exercise capacity. These findings indicate that astrocytes modulate the activity of CNS circuits generating the respiratory rhythm, critically contribute to adaptive respiratory responses in conditions of increased metabolic demand and determine the exercise capacity.


Assuntos
Astrócitos/fisiologia , Tronco Encefálico/fisiologia , Periodicidade , Condicionamento Físico Animal/fisiologia , Respiração , Potenciais de Ação/fisiologia , Adenoviridae/genética , Adenoviridae/metabolismo , Animais , Animais Recém-Nascidos , Astrócitos/citologia , Tronco Encefálico/citologia , Cálcio/metabolismo , Feminino , Regulação da Expressão Gênica , Vetores Genéticos/química , Vetores Genéticos/metabolismo , Hipercapnia/metabolismo , Hipercapnia/fisiopatologia , Hipóxia/metabolismo , Hipóxia/fisiopatologia , Masculino , Bulbo/citologia , Bulbo/fisiologia , Cultura Primária de Células , Ratos , Ratos Sprague-Dawley , Proteínas SNARE/antagonistas & inibidores , Proteínas SNARE/genética , Proteínas SNARE/metabolismo
17.
Glia ; 66(3): 592-605, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29178321

RESUMO

Glucose is a key modulator of feeding behavior. By acting in peripheral tissues and in the central nervous system, it directly controls the secretion of hormones and neuropeptides and modulates the activity of the autonomic nervous system. GLUT2 is required for several glucoregulatory responses in the brain, including feeding behavior, and is localized in the hypothalamus and brainstem, which are the main centers that control this behavior. In the hypothalamus, GLUT2 has been detected in glial cells, known as tanycytes, which line the basal walls of the third ventricle (3V). This study aimed to clarify the role of GLUT2 expression in tanycytes in feeding behavior using 3V injections of an adenovirus encoding a shRNA against GLUT2 and the reporter EGFP (Ad-shGLUT2). Efficient in vivo GLUT2 knockdown in rat hypothalamic tissue was demonstrated by qPCR and Western blot analyses. Specificity of cell transduction in the hypothalamus and brainstem was evaluated by EGFP-fluorescence and immunohistochemistry, which showed EGFP expression specifically in ependymal cells, including tanycytes. The altered mRNA levels of both orexigenic and anorexigenic neuropeptides suggested a loss of response to increased glucose in the 3V. Feeding behavior analysis in the fasting-feeding transition revealed that GLUT2-knockdown rats had increased food intake and body weight, suggesting an inhibitory effect on satiety. Taken together, suppression of GLUT2 expression in tanycytes disrupted the hypothalamic glucosensing mechanism, which altered the feeding behavior.


Assuntos
Comportamento Alimentar/fisiologia , Transportador de Glucose Tipo 2/metabolismo , Hipotálamo/metabolismo , Neuroglia/metabolismo , Saciação/fisiologia , Animais , Peso Corporal , Tronco Encefálico/citologia , Tronco Encefálico/metabolismo , Células Cultivadas , Jejum/metabolismo , Técnicas de Silenciamento de Genes , Transportador de Glucose Tipo 2/genética , Hipotálamo/citologia , Masculino , Neuroglia/citologia , Neuropeptídeos/metabolismo , RNA Mensageiro/metabolismo , Ratos Sprague-Dawley
18.
J Comp Neurol ; 525(18): 3787-3808, 2017 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-28758209

RESUMO

Detailed anatomical tracing and mapping of the viscerotopic organization of the vagal motor nuclei has provided insight into autonomic function in health and disease. To further define specific cellular identities, we paired information based on visceral connectivity with a cell-type specific marker of a subpopulation of neurons in the dorsal motor nucleus of the vagus (DMV) and nucleus ambiguus (nAmb) that express the autism-associated MET receptor tyrosine kinase. As gastrointestinal disturbances are common in children with autism spectrum disorder (ASD), we sought to define the relationship between MET-expressing (MET+) neurons in the DMV and nAmb, and the gastrointestinal tract. Using wholemount tissue staining and clearing, or retrograde tracing in a METEGFP transgenic mouse, we identify three novel subpopulations of EGFP+ vagal brainstem neurons: (a) EGFP+ neurons in the nAmb projecting to the esophagus or laryngeal muscles, (b) EGFP+ neurons in the medial DMV projecting to the stomach, and (b) EGFP+ neurons in the lateral DMV projecting to the cecum and/or proximal colon. Expression of the MET ligand, hepatocyte growth factor (HGF), by tissues innervated by vagal motor neurons during fetal development reveal potential sites of HGF-MET interaction. Furthermore, similar cellular expression patterns of MET in the brainstem of both the mouse and nonhuman primate suggests that MET expression at these sites is evolutionarily conserved. Together, the data suggest that MET+ neurons in the brainstem vagal motor nuclei are anatomically positioned to regulate distinct portions of the gastrointestinal tract, with implications for the pathophysiology of gastrointestinal comorbidities of ASD.


Assuntos
Tronco Encefálico/citologia , Neurônios Motores/metabolismo , Proteínas Proto-Oncogênicas c-met/metabolismo , Nervo Vago/fisiologia , Animais , Animais Recém-Nascidos , Tronco Encefálico/embriologia , Tronco Encefálico/crescimento & desenvolvimento , Toxina da Cólera/metabolismo , Colina O-Acetiltransferase/metabolismo , Embrião de Mamíferos , Feminino , Trato Gastrointestinal/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/genética , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Fator de Crescimento de Hepatócito/genética , Fator de Crescimento de Hepatócito/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neurônios Motores/classificação , Proteínas do Tecido Nervoso/metabolismo , Neurotransmissores/metabolismo , Proteínas Proto-Oncogênicas c-met/genética , Tirosina 3-Mono-Oxigenase/metabolismo
19.
PLoS One ; 12(6): e0178616, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28570669

RESUMO

Recent studies show a relationship between the deficit of vascular endothelial growth factor (VEGF) and motoneuronal degeneration, such as that occurring in amyotrophic lateral sclerosis (ALS). VEGF delivery protects motoneurons from cell death and delayed neurodegeneration in animal models of ALS. Strikingly, extraocular motoneurons show lesser vulnerability to neurodegeneration in ALS compared to other cranial or spinal motoneurons. Therefore, the present study investigates possible differences in VEGF and its main receptor VEGFR-2 or Flk-1 between extraocular and non-extraocular brainstem motoneurons. We performed immunohistochemistry and Western blot to determine the presence of VEGF and Flk-1 in rat motoneurons located in the three extraocular motor nuclei (abducens, trochlear and oculomotor) and to compare it to that observed in two other brainstem nuclei (hypoglossal and facial) that are vulnerable to degeneration. Extraocular motoneurons presented higher amounts of VEGF and its receptor Flk-1 than other brainstem motoneurons, and thus these molecules could be participating in their higher resistance to neurodegeneration. In conclusion, we hypothesize that differences in VEGF availability and signaling could be a contributing factor to the different susceptibility of extraocular motoneurons, when compared with other motoneurons, in neurodegenerative diseases.


Assuntos
Tronco Encefálico/metabolismo , Neurônios Motores/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Animais , Western Blotting , Tronco Encefálico/citologia , Ratos , Ratos Wistar
20.
Brain Stimul ; 10(4): 735-743, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28551320

RESUMO

BACKGROUND: Abdominal bilateral vagal stimulation reduces food intake in animals. However, the classical square wave, mA range current generator is poorly effective to evoke action potentials on A∂ and C neurons that represent the majority of vagal neurons at the abdominal level. OBJECTIVE/HYPOTHESIS: METHODS: The current thresholds for pulsons (S2 & S3) and millisecond pulses (S1) required to trigger action potentials were calculated in 5 anaesthetized pigs using single fibre recording. Similar stimulation protocols were compared chronically to sham stimulation in 24 pigs. After two weeks of chronic stimulation, food intake and brain metabolism were investigated. The electrical characteristics and histology of the vagus nerve were also studied. RESULTS: S3 stimulation required a lower amount of charges to trigger an action potential. Chronically applied S2 & S3 activated the dorsal vagal complex and increased the metabolism of its afferent cortical structures. They also reduced energy intake together with a reduced ingestion of high fat and high sugar diets. All these effects were not observed for the S1 group. The vagal histology for the S1, S2 and S3 groups was not different from that of the sham. CONCLUSIONS: These findings demonstrate that pulsons applied bilaterally on the abdominal vagus reduced food intake as a consequence of the activation of the brainstem and higher-order brain areas.


Assuntos
Tronco Encefálico/fisiologia , Ingestão de Alimentos , Neurônios/fisiologia , Nervo Vago/fisiologia , Animais , Tronco Encefálico/citologia , Tronco Encefálico/diagnóstico por imagem , Tronco Encefálico/metabolismo , Potenciais Evocados , Feminino , Masculino , Neurônios/metabolismo , Tomografia por Emissão de Pósitrons , Suínos , Estimulação do Nervo Vago
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA