Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Respir Physiol Neurobiol ; 323: 104227, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38295924

RESUMO

Breathing is the only vital function that can be volitionally controlled. However, a detailed understanding how volitional (cortical) motor commands can transform vital breathing activity into adaptive breathing patterns that accommodate orofacial behaviors such as swallowing, vocalization or sniffing remains to be developed. Recent neuroanatomical tract tracing studies have identified patterns and origins of descending forebrain projections that target brain nuclei involved in laryngeal adductor function which is critically involved in orofacial behavior. These nuclei include the midbrain periaqueductal gray and nuclei of the respiratory rhythm and pattern generating network in the brainstem, specifically including the pontine Kölliker-Fuse nucleus and the pre-Bötzinger complex in the medulla oblongata. This review discusses the functional implications of the forebrain-brainstem anatomical connectivity that could underlie the volitional control and coordination of orofacial behaviors with breathing.


Assuntos
Tronco Encefálico , Núcleo de Kölliker-Fuse , Bulbo , Respiração , Ponte , Vias Neurais
2.
J Physiol ; 602(1): 93-112, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38063489

RESUMO

The Kölliker-Fuse nucleus (KF), which is part of the parabrachial complex, participates in the generation of eupnoea under resting conditions and the control of active abdominal expiration when increased ventilation is required. Moreover, dysfunctions in KF neuronal activity are believed to play a role in the emergence of respiratory abnormalities seen in Rett syndrome (RTT), a progressive neurodevelopmental disorder associated with an irregular breathing pattern and frequent apnoeas. Relatively little is known, however, about the intrinsic dynamics of neurons within the KF and how their synaptic connections affect breathing pattern control and contribute to breathing irregularities. In this study, we use a reduced computational model to consider several dynamical regimes of KF activity paired with different input sources to determine which combinations are compatible with known experimental observations. We further build on these findings to identify possible interactions between the KF and other components of the respiratory neural circuitry. Specifically, we present two models that both simulate eupnoeic as well as RTT-like breathing phenotypes. Using nullcline analysis, we identify the types of inhibitory inputs to the KF leading to RTT-like respiratory patterns and suggest possible KF local circuit organizations. When the identified properties are present, the two models also exhibit quantal acceleration of late-expiratory activity, a hallmark of active expiration featuring forced exhalation, with increasing inhibition to KF, as reported experimentally. Hence, these models instantiate plausible hypotheses about possible KF dynamics and forms of local network interactions, thus providing a general framework as well as specific predictions for future experimental testing. KEY POINTS: The Kölliker-Fuse nucleus (KF), a part of the parabrachial complex, is involved in regulating normal breathing and controlling active abdominal expiration during increased ventilation. Dysfunction in KF neuronal activity is thought to contribute to respiratory abnormalities seen in Rett syndrome (RTT). This study utilizes computational modelling to explore different dynamical regimes of KF activity and their compatibility with experimental observations. By analysing different model configurations, the study identifies inhibitory inputs to the KF that lead to RTT-like respiratory patterns and proposes potential KF local circuit organizations. Two models are presented that simulate both normal breathing and RTT-like breathing patterns. These models provide testable hypotheses and specific predictions for future experimental investigations, offering a general framework for understanding KF dynamics and potential network interactions.


Assuntos
Núcleo de Kölliker-Fuse , Síndrome de Rett , Humanos , Núcleo de Kölliker-Fuse/fisiologia , Respiração , Neurônios , Simulação por Computador
3.
J Comp Neurol ; 530(10): 1658-1699, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35134251

RESUMO

Diverse neurons in the parabrachial nucleus (PB) communicate with widespread brain regions. Despite evidence linking them to a variety of homeostatic functions, it remains difficult to determine which PB neurons influence which functions because their subpopulations intermingle extensively. An improved framework for identifying these intermingled subpopulations would help advance our understanding of neural circuit functions linked to this region. Here, we present the foundation of a developmental-genetic ontology that classifies PB neurons based on their intrinsic, molecular features. By combining transcription factor labeling with Cre fate-mapping, we find that the PB is a blend of two, developmentally distinct macropopulations of glutamatergic neurons. Neurons in the first macropopulation express Lmx1b (and, to a lesser extent, Lmx1a) and are mutually exclusive with those in a second macropopulation, which derive from precursors expressing Atoh1. This second, Atoh1-derived macropopulation includes many Foxp2-expressing neurons, but Foxp2 also identifies a subset of Lmx1b-expressing neurons in the Kölliker-Fuse nucleus (KF) and a population of GABAergic neurons ventrolateral to the PB ("caudal KF"). Immediately ventral to the PB, Phox2b-expressing glutamatergic neurons (some coexpressing Lmx1b) occupy the KF, supratrigeminal nucleus, and reticular formation. We show that this molecular framework organizes subsidiary patterns of adult gene expression (including Satb2, Calca, Grp, and Pdyn) and predicts output projections to the amygdala (Lmx1b), hypothalamus (Atoh1), and hindbrain (Phox2b/Lmx1b). Using this molecular ontology to organize, interpret, and communicate PB-related information could accelerate the translation of experimental findings from animal models to human patients.


Assuntos
Núcleo de Kölliker-Fuse , Núcleos Parabraquiais , Animais , Encéfalo/metabolismo , Neurônios GABAérgicos/metabolismo , Humanos , Hipotálamo/metabolismo , Ponte/fisiologia , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
4.
Anesthesiology ; 135(4): 649-672, 2021 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-34352068

RESUMO

BACKGROUND: Recent studies showed partial reversal of opioid-induced respiratory depression in the pre-Bötzinger complex and the parabrachial nucleus/Kölliker-Fuse complex. The hypothesis for this study was that opioid antagonism in the parabrachial nucleus/Kölliker-Fuse complex plus pre-Bötzinger complex completely reverses respiratory depression from clinically relevant opioid concentrations. METHODS: Experiments were performed in 48 adult, artificially ventilated, decerebrate rabbits. The authors decreased baseline respiratory rate ~50% with intravenous, "analgesic" remifentanil infusion or produced apnea with remifentanil boluses and investigated the reversal with naloxone microinjections (1 mM, 700 nl) into the Kölliker-Fuse nucleus, parabrachial nucleus, and pre-Bötzinger complex. In another group of animals, naloxone was injected only into the pre-Bötzinger complex to determine whether prior parabrachial nucleus/Kölliker-Fuse complex injection impacted the naloxone effect. Last, the µ-opioid receptor agonist [d-Ala,2N-MePhe,4Gly-ol]-enkephalin (100 µM, 700 nl) was injected into the parabrachial nucleus/Kölliker-Fuse complex. The data are presented as medians (25 to 75%). RESULTS: Remifentanil infusion reduced the respiratory rate from 36 (31 to 40) to 16 (15 to 21) breaths/min. Naloxone microinjections into the bilateral Kölliker-Fuse nucleus, parabrachial nucleus, and pre-Bötzinger complex increased the rate to 17 (16 to 22, n = 19, P = 0.005), 23 (19 to 29, n = 19, P < 0.001), and 25 (22 to 28) breaths/min (n = 11, P < 0.001), respectively. Naloxone injection into the parabrachial nucleus/Kölliker-Fuse complex prevented apnea in 12 of 17 animals, increasing the respiratory rate to 10 (0 to 12) breaths/min (P < 0.001); subsequent pre-Bötzinger complex injection prevented apnea in all animals (13 [10 to 19] breaths/min, n = 12, P = 0.002). Naloxone injection into the pre-Bötzinger complex alone increased the respiratory rate to 21 (15 to 26) breaths/min during analgesic concentrations (n = 10, P = 0.008) but not during apnea (0 [0 to 0] breaths/min, n = 9, P = 0.500). [d-Ala,2N-MePhe,4Gly-ol]-enkephalin injection into the parabrachial nucleus/Kölliker-Fuse complex decreased respiratory rate to 3 (2 to 6) breaths/min. CONCLUSIONS: Opioid reversal in the parabrachial nucleus/Kölliker-Fuse complex plus pre-Bötzinger complex only partially reversed respiratory depression from analgesic and even less from "apneic" opioid doses. The lack of recovery pointed to opioid-induced depression of respiratory drive that determines the activity of these areas.


Assuntos
Analgésicos Opioides/efeitos adversos , Núcleo de Kölliker-Fuse/efeitos dos fármacos , Núcleos Parabraquiais/efeitos dos fármacos , Remifentanil/efeitos adversos , Insuficiência Respiratória/induzido quimicamente , Analgésicos Opioides/administração & dosagem , Animais , Relação Dose-Resposta a Droga , Feminino , Infusões Intravenosas , Núcleo de Kölliker-Fuse/fisiologia , Masculino , Núcleos Parabraquiais/fisiologia , Coelhos , Remifentanil/administração & dosagem , Insuficiência Respiratória/fisiopatologia
5.
Respir Physiol Neurobiol ; 284: 103563, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33053424

RESUMO

The pontine Kölliker-Fuse nucleus (KFn) is a core nucleus of respiratory network that mediates the inspiratory-expiratory phase transition and gates eupneic motor discharges in the vagal and hypoglossal nerves. In the present study, we investigated whether the same KFn circuit may also gate motor activities that control the resistance of the nasal airway, which is of particular importance in rodents. To do so, we simultaneously recorded phrenic, facial, vagal and hypoglossal cranial nerve activity in an in situ perfused brainstem preparation before and after bilateral injection of the GABA-receptor agonist isoguvacine (50-70 nl, 10 mM) into the KFn (n = 11). Our results show that bilateral inhibition of the KFn triggers apneusis (prolonged inspiration) and abolished pre-inspiratory discharge of facial, vagal and hypoglossal nerves as well as post-inspiratory discharge in the vagus. We conclude that the KFn plays a critical role for the eupneic regulation of naso-pharyngeal airway patency and the potential functions of the KFn in regulating airway patency and orofacial behavior is discussed.


Assuntos
Nervo Facial/fisiologia , Nervo Hipoglosso/fisiologia , Núcleo de Kölliker-Fuse/fisiologia , Atividade Motora/fisiologia , Rede Nervosa/fisiologia , Nervo Frênico/fisiologia , Respiração , Nervo Vago/fisiologia , Animais , Nervo Facial/efeitos dos fármacos , Feminino , Agonistas GABAérgicos/farmacologia , Nervo Hipoglosso/efeitos dos fármacos , Ácidos Isonicotínicos/farmacologia , Núcleo de Kölliker-Fuse/efeitos dos fármacos , Masculino , Atividade Motora/efeitos dos fármacos , Rede Nervosa/efeitos dos fármacos , Nervo Frênico/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Respiração/efeitos dos fármacos , Centro Respiratório , Taxa Respiratória/efeitos dos fármacos , Taxa Respiratória/fisiologia , Nervo Vago/efeitos dos fármacos
6.
J Neurochem ; 156(1): 16-37, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-32396650

RESUMO

The Kölliker-Fuse nucleus (KF) is a functionally distinct component of the parabrachial complex, located in the dorsolateral pons of mammals. The KF has a major role in respiration and upper airway control. A comprehensive understanding of the KF and its contributions to respiratory function and dysfunction requires an appreciation for its neurochemical characteristics. The goal of this review is to summarize the diverse neurochemical composition of the KF, focusing on the neurotransmitters, neuromodulators, and neuropeptides present. We also include a description of the receptors expressed on KF neurons and transporters involved in each system, as well as their putative roles in respiratory physiology. Finally, we provide a short section reviewing the literature regarding neurochemical changes in the KF in the context of respiratory dysfunction observed in SIDS and Rett syndrome. By over-viewing the current literature on the neurochemical composition of the KF, this review will serve to aid a wide range of topics in the future research into the neural control of respiration in health and disease.


Assuntos
Núcleo de Kölliker-Fuse/química , Núcleo de Kölliker-Fuse/fisiologia , Respiração , Animais , Humanos
7.
Pflugers Arch ; 472(8): 1051-1063, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32617654

RESUMO

The Kölliker-Fuse (KF) nucleus is a part of the parabrachial complex, located in the dorsolateral pons. It is involved in the chemoreflex-evoked cardiovascular and respiratory changes, but the role of GABA and glutamate in cardiovascular chemoreflex has not been shown yet. This study was performed to determine the role of GABA, glutamate, and their interaction in the KF, in cardiovascular chemoreflex in anesthetized rat. The antagonists were microinjected into the KF, and arterial pressure, heart rate, and single-unit responses were recorded simultaneously. The chemoreflex was evoked by i.v. injection of KCN, consisted of a short pressor followed by long bradycardia responses. Both responses were significantly attenuated by injection of a synaptic blocker (CoCl2) into the KF, confirming involvement of the KF in generating the reflex. Microinjection of AP5, an NMDA receptor antagonist, into the KF significantly attenuated the pressor and bradycardia responses, while blocking the AMPA receptors by CNQX had no significant effect. Blockade of GABAA receptors by bicuculline methiodide (BMI) potentiated both responses. Co-injection of BMI and CNQX potentiated the responses too. Co-injection of BMI and AP5 had no significant effect on the pressor response but significantly attenuated the bradycardia response. In conclusion, the KF plays a role in generating cardiovascular chemoreflex via its glutamate NMDA but not AMPA receptors. GABA inhibits both components of this reflex through GABAA receptors. There is an interaction between GABAA and NMDA receptors in regulating the bradycardia response of the reflex. Single-unit results were also presented which were correlated with and supported the homodynamic findings.


Assuntos
Sistema Cardiovascular/metabolismo , Células Quimiorreceptoras/metabolismo , Ácido Glutâmico/metabolismo , Núcleo de Kölliker-Fuse/metabolismo , Reflexo/fisiologia , Ácido gama-Aminobutírico/metabolismo , Animais , Bicuculina/análogos & derivados , Bicuculina/farmacologia , Pressão Sanguínea/efeitos dos fármacos , Pressão Sanguínea/fisiologia , Células Quimiorreceptoras/efeitos dos fármacos , Frequência Cardíaca/efeitos dos fármacos , Frequência Cardíaca/fisiologia , Núcleo de Kölliker-Fuse/efeitos dos fármacos , Masculino , Ponte/efeitos dos fármacos , Ponte/fisiologia , Ratos , Ratos Sprague-Dawley , Receptores de AMPA/metabolismo , Receptores de GABA-A/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Reflexo/efeitos dos fármacos , Respiração/efeitos dos fármacos
8.
Respir Physiol Neurobiol ; 277: 103401, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32036030

RESUMO

The Kölliker-Fuse Nucleus (KF) has been widely investigated for its contribution to "inspiratory off-switch" while more recent studies showed that activation of the Parabrachial Nucleus (PBN) shortened expiratory duration. This study used an adult, in vivo, decerebrate rabbit model to delineate the contribution of each site to inspiratory and expiratory duration through sequential block of glutamatergic excitation with the receptor antagonists 2,3-dihydroxy-6-nitro-7-sulfamoyl-benzo[f]quinoxaline-2,3-dione (NBQX) and d(-)-2-amino-5-phosphonopentanoic acid (AP5). Glutamatergic disfacilitation caused large increases in inspiratory and expiratory duration and minor decrease in peak phrenic activity (PPA). Hypoxia only partially reversed respiratory rate depression but PPA was increased to >200 % of control. The contribution of PBN activity to inspiratory and expiratory duration was equal while block of the KF affected inspiratory duration more than expiratory. We conclude that in the in vivo preparation respiratory rate greatly depends on PBN/KF activity, which contributes to the "inspiratory on- "and "off-switch", but is of minor importance for the magnitude of phrenic motor output.


Assuntos
Ácido Glutâmico/fisiologia , Núcleo de Kölliker-Fuse/fisiologia , Núcleos Parabraquiais/fisiologia , Centro Respiratório/fisiologia , Taxa Respiratória/fisiologia , Animais , Agonistas de Aminoácidos Excitatórios/administração & dosagem , Antagonistas de Aminoácidos Excitatórios/administração & dosagem , Feminino , Núcleo de Kölliker-Fuse/efeitos dos fármacos , Masculino , Microinjeções/métodos , Núcleos Parabraquiais/efeitos dos fármacos , Quinoxalinas/administração & dosagem , Coelhos , Centro Respiratório/efeitos dos fármacos , Taxa Respiratória/efeitos dos fármacos , Ácido alfa-Amino-3-hidroxi-5-metil-4-isoxazol Propiônico/administração & dosagem
9.
Respir Physiol Neurobiol ; 275: 103388, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31953234

RESUMO

Overdoses caused by the opioid agonist fentanyl have increased exponentially in recent years. Identifying mechanisms to counter progression to fatal respiratory apnea during opioid overdose is desirable, but difficult to study in vivo. The pontine Kölliker-Fuse/Parabrachial complex (KF/PB) provides respiratory drive and contains opioid-sensitive neurons. The contribution of the KF/PB complex to fentanyl-induced apnea was investigated using the in situ arterially perfused preparation of rat. Systemic application of fentanyl resulted in concentration-dependent respiratory disturbances. At low concentrations, respiratory rate slowed and subsequently transitioned to an apneustic-like, 2-phase pattern. Higher concentrations caused prolonged apnea, interrupted by occasional apneustic-like bursts. Application of CTAP, a selective mu opioid receptor antagonist, directly into the KF/PB complex reversed and prevented fentanyl-induced apnea by increasing the frequency of apneustic-like bursting. These results demonstrate that countering opioid effects in the KF/PB complex is sufficient to restore phasic respiratory output at a rate similar to pre-fentanyl conditions, which could be beneficial in opioid overdose.


Assuntos
Analgésicos Opioides/farmacologia , Apneia/induzido quimicamente , Apneia/prevenção & controle , Fentanila/farmacologia , Núcleo de Kölliker-Fuse/efeitos dos fármacos , Antagonistas de Entorpecentes/farmacologia , Núcleos Parabraquiais/efeitos dos fármacos , Receptores Opioides mu/efeitos dos fármacos , Taxa Respiratória/efeitos dos fármacos , Animais , Feminino , Masculino , Ratos , Ratos Sprague-Dawley
10.
Respir Physiol Neurobiol ; 266: 95-102, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31055189

RESUMO

Spinal phrenic nerve activity (PNA) drives the diaphragm but cranial hypoglossal nerve activity (HNA) also expresses synchronous activity during inspiration. Here, we investigated the effects of local disinhibition (bilateral microinjections of bicuculline) of the nucleus tractus solitarius (NTS), the pre-Bötzinger complex and Bötzinger complex core circuit (pre-BötC/BötC) and the Kölliker-Fuse nuclei (KFn) on the synchronization of PNA and HNA in arterially-perfused brainstem preparations of rats. To quantitatively analyze the bicuculline effects on a putatively distributed inspiratory central pattern generator (i-CPG), we quantified the phase synchronization properties between PNA and HNA. The analysis revealed that bicuculline-evoked local disinhibition significantly reduced the strength of phase synchronization between PNA and HNA at any target site. However, the emergence of desynchronized HNA following disinhibition was more prevalent after NTS or pre-BötC/BötC microinjections compared to the KFn. We conclude that the primary i-CPG is located in a distributed medullary circuit whereas pontine contributions are restricted to synaptic gating of synchronous HNA and PNA.


Assuntos
Geradores de Padrão Central/fisiologia , Núcleo de Kölliker-Fuse/fisiologia , Bulbo/fisiologia , Rede Nervosa/fisiologia , Fenômenos Fisiológicos do Sistema Nervoso , Nervo Frênico/fisiologia , Respiração , Núcleo Solitário/fisiologia , Animais , Bicuculina/farmacologia , Geradores de Padrão Central/efeitos dos fármacos , Feminino , Antagonistas de Receptores de GABA-A/farmacologia , Núcleo de Kölliker-Fuse/efeitos dos fármacos , Masculino , Bulbo/efeitos dos fármacos , Rede Nervosa/efeitos dos fármacos , Fenômenos Fisiológicos do Sistema Nervoso/efeitos dos fármacos , Nervo Frênico/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Núcleo Solitário/efeitos dos fármacos
11.
Behav Brain Res ; 369: 111916, 2019 09 02.
Artigo em Inglês | MEDLINE | ID: mdl-31004684

RESUMO

Upper airway and vocalization control areas such as the periaqueductal gray (PAG), kölliker-fuse nucleus (KF) and nucleus retroambiguus (NRA) are prone to developing tauopathy in mice expressing the mutant human tau P301L protein. Consequently, impaired ultrasonic vocalization (USV) previously identified in tau-P301L mice at the terminal disease stage of 8-9 months of age, was attributed to the presence of tauopathy in these regions. Our aim was to establish whether the onset of USV disorders manifest prior to the terminal stage, and if USV disorders are predictive of the presence of tauopathy in the PAG, KF and NRA. USVs produced by tau-P301L and wildtype mice aged 3-4, 5-6 or 8-9 months were recorded during male-female interaction. Immunohistochemistry was then performed to assess the presence or degree of tauopathy in the PAG, KF and NRA of mice displaying normal or abnormal USV patterns. Comparing various USV measurements, including the number, duration and frequency of calls, revealed no differences between tau-P301L and wildtype mice across all age groups, and linear discriminant analysis also failed to identify separate USV populations. Finally, the presence of tauopathy in the PAG, KF and NRA in individual tau-P301L mice did not reliably associate with USV disorders. Our findings that tauopathy in designated mammalian vocalization centres, such as the PAG, KF and NRA, did not associate with USV disturbances in tau-P301L mice questions whether USV phenotypes in this transgenic mouse are valid for studying tauopathy-related human voice and speech disorders.


Assuntos
Tauopatias/metabolismo , Vocalização Animal/fisiologia , Proteínas tau/genética , Animais , Feminino , Núcleo de Kölliker-Fuse/fisiologia , Masculino , Camundongos , Camundongos Transgênicos , Substância Cinzenta Periaquedutal/fisiologia , Ondas Ultrassônicas
12.
J Physiol ; 597(10): 2651-2672, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30908648

RESUMO

KEY POINTS: Reduced computational models are used to test effects of loss of inhibition to the Kölliker-Fuse nucleus (KFn). Three reduced computational models that simulate eupnoeic and vagotomized respiratory rhythms are considered. All models exhibit the emergence of respiratory perturbations associated with Rett syndrome as inhibition to the KFn is diminished. Simulations suggest that application of 5-HT1A agonists can mitigate the respiratory pathology. The three models can be distinguished and tested based on their predictions about connections and dynamics within the respiratory circuit and about effects of perturbations on certain respiratory neuron populations. ABSTRACT: Rett syndrome (RTT) is a developmental disorder that can lead to respiratory disturbances featuring prolonged apnoeas of variable durations. Determining the mechanisms underlying these effects at the level of respiratory neural circuits would have significant implications for treatment efforts and would also enhance our understanding of respiratory rhythm generation and control. While experimental studies have suggested possible factors contributing to the respiratory patterns of RTT, we take a novel computational approach to the investigation of RTT, which allows for direct manipulation of selected system parameters and testing of specific hypotheses. Specifically, we present three reduced computational models, developed using an established framework, all of which successfully simulate respiratory outputs across eupnoeic and vagotomized conditions. All three models show that loss of inhibition to the Kölliker-Fuse nucleus reproduces the key respiratory alterations associated with RTT and, as suggested experimentally, that effects of 5-HT1A agonists on the respiratory neural circuit suffice to alleviate this respiratory pathology. Each of the models makes distinct predictions regarding the neuronal populations and interactions underlying these effects, suggesting natural directions for future experimental testing.


Assuntos
Simulação por Computador , Núcleo de Kölliker-Fuse/fisiologia , Modelos Biológicos , Síndrome de Rett/fisiopatologia , Fenômenos Fisiológicos Respiratórios , Nervo Vago
13.
Am J Perinatol ; 36(5): 460-471, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30170332

RESUMO

OBJECTIVE: To find a possible pathogenetic mechanism of the early sudden infant death occurring in newborns during the skin-to-skin care (SSC), through the examination of neuronal centers regulating the vital activities. STUDY DESIGN: This is an in-depth examination of the brain stem in 22 healthy term newborns, suddenly died in the first hour of life without the identification of a cause at autopsy (early sudden infant death syndrome [eSIDS]), 12 of them concomitantly with SSC, and 10 with age-matched controls died of known pathology. RESULTS: Developmental alterations of neuronal structures of the brain stem were highlighted in 19 of the 22 eSIDS, but not in control. The hypoplasia of the pontine Kölliker-Fuse nucleus (KFN), an important respiratory center, was diagnosed at the histological examination, validated by morphometric quantifications, in 11 of the 12 eSIDS while they were placed on the mother's chest and in 2 of the 10 SSC unrelated neonatal deaths. CONCLUSION: The delayed development of the KFN could represent a specific finding of eSIDS occurring during SSC. Therefore, it is necessary to point out that the SSC represents a further risk factor that must be added to others already known for sudden infant death syndrome. Then this practice needs appropriate monitoring strategies of the infant's conditions.


Assuntos
Tronco Encefálico/patologia , Método Canguru , Núcleo de Kölliker-Fuse/anormalidades , Morte Súbita do Lactente/patologia , Adulto , Autopsia , Feminino , Humanos , Recém-Nascido , Núcleo de Kölliker-Fuse/patologia , Masculino , Neuropatologia , Decúbito Ventral/fisiologia , Respiração , Fatores de Risco , Adulto Jovem
14.
J Neurophysiol ; 119(2): 401-412, 2018 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-29070631

RESUMO

Coordination of respiratory pump and valve muscle activity is essential for normal breathing. A hallmark respiratory response to hypercapnia and hypoxia is the emergence of active exhalation, characterized by abdominal muscle pumping during the late one-third of expiration (late-E phase). Late-E abdominal activity during hypercapnia has been attributed to the activation of expiratory neurons located within the parafacial respiratory group (pFRG). However, the mechanisms that control emergence of active exhalation, and its silencing in restful breathing, are not completely understood. We hypothesized that inputs from the Kölliker-Fuse nucleus (KF) control the emergence of late-E activity during hypercapnia. Previously, we reported that reversible inhibition of the KF reduced postinspiratory (post-I) motor output to laryngeal adductor muscles and brought forward the onset of hypercapnia-induced late-E abdominal activity. Here we explored the contribution of the KF for late-E abdominal recruitment during hypercapnia by pharmacologically disinhibiting the KF in in situ decerebrate arterially perfused rat preparations. These data were combined with previous results and incorporated into a computational model of the respiratory central pattern generator. Disinhibition of the KF through local parenchymal microinjections of gabazine (GABAA receptor antagonist) prolonged vagal post-I activity and inhibited late-E abdominal output during hypercapnia. In silico, we reproduced this behavior and predicted a mechanism in which the KF provides excitatory drive to post-I inhibitory neurons, which in turn inhibit late-E neurons of the pFRG. Although the exact mechanism proposed by the model requires testing, our data confirm that the KF modulates the formation of late-E abdominal activity during hypercapnia. NEW & NOTEWORTHY The pons is essential for the formation of the three-phase respiratory pattern, controlling the inspiratory-expiratory phase transition. We provide functional evidence of a novel role for the Kölliker-Fuse nucleus (KF) controlling the emergence of abdominal expiratory bursts during active expiration. A computational model of the respiratory central pattern generator predicts a possible mechanism by which the KF interacts indirectly with the parafacial respiratory group and exerts an inhibitory effect on the expiratory conditional oscillator.


Assuntos
Hipercapnia/fisiopatologia , Núcleo de Kölliker-Fuse/fisiologia , Nervos Periféricos/fisiologia , Respiração , Animais , Geradores de Padrão Central/fisiologia , Potencial Evocado Motor , Núcleo de Kölliker-Fuse/fisiopatologia , Masculino , Modelos Neurológicos , Nervos Periféricos/fisiopatologia , Ratos , Ratos Wistar , Músculos Respiratórios/inervação
15.
Neuroscience ; 348: 63-72, 2017 04 21.
Artigo em Inglês | MEDLINE | ID: mdl-28188852

RESUMO

While the transition from the inspiratory to the post-inspiratory (post-I) phase is dependent on the pons, little attention has been paid to understanding the role of the pontine respiratory nuclei, specifically the Kölliker-Fuse nucleus (KF), in transitioning from post-I to the late expiratory (late-E) activity seen with elevated respiratory drive. To elucidate this, we used the in situ working heart-brainstem preparation of juvenile male Holtzman rats and recorded from the vagus (cVN), phrenic (PN) and abdominal nerves (AbN) during baseline conditions and during chemoreflex activation [with potassium cyanide (KCN; n=13) or hypercapnia (8% CO2; n=10)] to recruit active expiration. Chemoreflex activation with KCN increased PN frequency and cVN post-I and AbN activities. The inhibition of KF with isoguvacine microinjections (10mM) attenuated the typical increase in PN frequency and cVN post-I activity, and amplified the AbN response. During hypercapnia, AbN late-E activity emerged in association with a significant reduction in expiratory time. KF inhibition during hypercapnia significantly decreased PN frequency and reduced the duration and amplitude of post-I cVN activity, while the onset of the AbN late-E bursts occurred significantly earlier. Our data reveal a negative relationship between KF-induced post-I and AbN late-E activities, suggesting that the KF coordinates the transition between post-I to late-E activity during conditions of elevated respiratory drive.


Assuntos
Abdome/inervação , Expiração/fisiologia , Hipercapnia/fisiopatologia , Inalação/fisiologia , Nervo Frênico/fisiologia , Nervo Vago/fisiologia , Animais , Expiração/efeitos dos fármacos , Inalação/efeitos dos fármacos , Núcleo de Kölliker-Fuse/fisiologia , Masculino , Nervo Frênico/efeitos dos fármacos , Cianeto de Potássio/farmacologia , Ratos , Ratos Sprague-Dawley , Respiração/efeitos dos fármacos , Nervo Vago/efeitos dos fármacos
16.
J Comp Neurol ; 525(8): 1844-1860, 2017 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-28032634

RESUMO

The Kölliker-Fuse nucleus (KF) is known primarily for its respiratory function as the "pneumotaxic center" or "pontine respiratory group." Considered part of the parabrachial (PB) complex, KF contains glutamatergic neurons that project to respiratory-related targets in the medulla and spinal cord (Yokota, Oka, Tsumori, Nakamura, & Yasui, 2007). Here we describe an unexpected population of neurons in the caudal KF and adjacent lateral crescent subnucleus (PBlc), which are γ-aminobutyric acid (GABA)ergic and have an entirely different pattern of projections than glutamatergic KF neurons. First, immunofluorescence, in situ hybridization, and Cre-reporter labeling revealed that many of these GABAergic neurons express FoxP2 in both rats and mice. Next, using Cre-dependent axonal tracing in Vgat-IRES-Cre and Vglut2-IRES-Cre mice, we identified different projection patterns from GABAergic and glutamatergic neurons in this region. GABAergic neurons in KF and PBlc project heavily and almost exclusively to trigeminal sensory nuclei, with minimal projections to cardiorespiratory nuclei in the brainstem, and none to the spinal cord. In contrast, glutamatergic KF neurons project heavily to the autonomic, respiratory, and motor regions of the medulla and spinal cord previously identified as efferent targets mediating KF cardiorespiratory effects. These findings identify a novel, GABAergic subpopulation of KF/PB neurons with a distinct efferent projection pattern targeting the brainstem trigeminal sensory system. Rather than regulating breathing, we propose that these neurons influence vibrissal sensorimotor function.


Assuntos
Neurônios GABAérgicos/citologia , Núcleo de Kölliker-Fuse/citologia , Vias Neurais/citologia , Neurônios/citologia , Animais , Feminino , Imunofluorescência , Ácido Glutâmico , Processamento de Imagem Assistida por Computador , Imuno-Histoquímica , Hibridização In Situ , Masculino , Camundongos , Camundongos Transgênicos , Ratos , Ratos Wistar
17.
Am J Physiol Regul Integr Comp Physiol ; 312(2): R172-R188, 2017 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-27974314

RESUMO

Respiration varies from breath to breath. On the millisecond timescale of spiking, neuronal circuits exhibit variability due to the stochastic properties of ion channels and synapses. Does this fast, microscopic source of variability contribute to the slower, macroscopic variability of the respiratory period? To address this question, we modeled a stochastic oscillator with forcing; then, we tested its predictions experimentally for the respiratory rhythm generated by the in situ perfused preparation during vagal nerve stimulation (VNS). Our simulations identified a relationship among the gain of the input, entrainment strength, and rhythm variability. Specifically, at high gain, the periodic input entrained the oscillator and reduced variability, whereas at low gain, the noise interacted with the input, causing events known as "phase slips", which increased variability on a slow timescale. Experimentally, the in situ preparation behaved like the low-gain model: VNS entrained respiration but exhibited phase slips that increased rhythm variability. Next, we used bilateral muscimol microinjections in discrete respiratory compartments to identify areas involved in VNS gain control. Suppression of activity in the nucleus tractus solitarii occluded both entrainment and amplification of rhythm variability by VNS, confirming that these effects were due to the activation of the Hering-Breuer reflex. Suppressing activity of the Kölliker-Fuse nuclei (KFn) enhanced entrainment and reduced rhythm variability during VNS, consistent with the predictions of the high-gain model. Together, the model and experiments suggest that the KFn regulates respiratory rhythm variability via a gain control mechanism.


Assuntos
Relógios Biológicos/fisiologia , Retroalimentação Fisiológica/fisiologia , Núcleo de Kölliker-Fuse/fisiologia , Modelos Biológicos , Taxa Respiratória/fisiologia , Animais , Geradores de Padrão Central , Simulação por Computador , Humanos , Reflexo/fisiologia , Reprodutibilidade dos Testes , Mecânica Respiratória/fisiologia , Sensibilidade e Especificidade , Processos Estocásticos
18.
Brain Res ; 1648(Pt A): 512-523, 2016 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-27544422

RESUMO

The neural pathways underlying the respiratory variation dependent on vigilance states remain unsettled. In the present study, we examined the orexinergic innervation of Kölliker-Fuse nucleus (KFN) neurons sending their axons to the rostral ventral respiratory group (rVRG) and phrenic nucleus (PhN) as well as to the hypoglossal nucleus (HGN) by using a combined retrograde tracing and immunohistochemistry. After injection of cholera toxin B subunit (CTb) into the KFN, CTb-labeled neurons that are also immunoreactive for orexin (ORX) were found prominently in the perifornical and medial regions and additionally in the lateral region of the hypothalamic ORX field. After injection of fluorogold (FG) into the rVRG, PhN or HGN, we found an overlapping distribution of ORX-immunoreactive axon terminals and FG-labeled neurons in the KFN. Within the neuropil of the KFN, asymmetrical synaptic contacts were made between these terminals and neurons. We further demonstrated that many neurons labeled with FG injected into the rVRG, PhN, or HGN are immunoreactive for ORX receptor 2. Present data suggest that rVRG-, PhN- and HGN-projecting KFN neurons may be under the excitatory influence of the ORXergic neurons for the state-dependent regulation of respiration.


Assuntos
Medula Cervical/citologia , Núcleo de Kölliker-Fuse/citologia , Bulbo/citologia , Neurônios/citologia , Orexinas/metabolismo , Respiração , Medula Espinal/citologia , Animais , Axônios/metabolismo , Medula Cervical/metabolismo , Hipotálamo/citologia , Imuno-Histoquímica , Núcleo de Kölliker-Fuse/ultraestrutura , Masculino , Bulbo/metabolismo , Vias Neurais/citologia , Vias Neurais/metabolismo , Técnicas de Rastreamento Neuroanatômico , Neurônios/metabolismo , Receptores de Orexina/metabolismo , Ratos , Ratos Wistar , Medula Espinal/metabolismo
19.
Neurol Res ; 38(8): 706-16, 2016 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-27353953

RESUMO

OBJECTIVES: As well known, the sudden infant death syndrome (SIDS) is characterized by the sudden death of a seemingly healthy infant during sleep, frequently resulted from a deficit in arousal phase. Awakening from sleep requires a fully developed and functioning neuronal respiratory network to modulate the ventilation as needed. The pontine Kölliker-Fuse nucleus (KFN) plays a pivotal role in breathing control, thanks to its interconnections with the widespread serotonin and noradrenaline neurons in the brainstem. Numerous studies to date have focused on the implication of orexin, a neuropeptide synthesized by neurons of the lateral hypothalamus, with major projections to the brainstem raphé nuclei and locus coeruleus, in arousal, a neurobiological process closely linked to breathing modifications. The aim of our research has been to demonstrate that also the KFN is a fundamental component of the orexin system, actively involved in arousal. METHODS: We have evaluated the expression and distribution of the orexin receptors (orexin-1 and orexin-2 receptors) particularly in the rostral pons, where the KFN is located, of 25 SIDS cases and 18 controls. RESULTS: An intense orexin-1 innervation around the KF neurons has been detected in almost all the controls and only in 20% of SIDS cases. DISCUSSION: On the basis of these results, we believe that: (1) the KFN plays a leading role not only in providing a regular breathing rhythm but also in the coordination of the sleep-to-wake transition; (2) a defective orexin expression in the KFN could prevent arousal, thus assuming a crucial importance in causing SIDS.


Assuntos
Núcleo de Kölliker-Fuse/metabolismo , Núcleo de Kölliker-Fuse/fisiopatologia , Receptores de Orexina/metabolismo , Morte Súbita do Lactente/patologia , Feminino , Proteína Glial Fibrilar Ácida/metabolismo , Humanos , Lactente , Núcleo de Kölliker-Fuse/patologia , Masculino , Neurônios/metabolismo , Fosfopiruvato Hidratase/metabolismo
20.
Respir Physiol Neurobiol ; 226: 110-4, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26840837

RESUMO

The neurotransmitter serotonin (5HT) acting via 5HT1a receptors (5HT1aR) is a potent determinant of respiratory rhythm variability. Here, we address the 5HT1aR-dependent control of respiratory rhythm variability in C57BL6/J mice. Using the in situ perfused preparation, we compared the effects of systemic versus focal blockade of 5HT1aRs. Blocking 5HT1aRs in the Kölliker-Fuse nucleus (KFn) increased the occurrence of spontaneous apneas and accounted for the systemic effects of 5HT1aR antagonists. Further, 5HT1aRs of the KFn stabilized the respiratory rhythm's response to arterial chemoreflex perturbations; reducing the recovering time, e.g., the latency to return to the baseline pattern. Together, these results suggest that the KFn regulates both intrinsic and sensory determinants of respiratory rhythm variability.


Assuntos
Cicloexanos/farmacologia , Núcleo de Kölliker-Fuse/efeitos dos fármacos , Núcleo de Kölliker-Fuse/metabolismo , Piperazinas/farmacologia , Receptor 5-HT1A de Serotonina/metabolismo , Respiração/efeitos dos fármacos , Antagonistas do Receptor 5-HT1 de Serotonina/farmacologia , Animais , Apneia/induzido quimicamente , Apneia/metabolismo , Células Quimiorreceptoras/efeitos dos fármacos , Células Quimiorreceptoras/fisiologia , Feminino , Potenciais da Membrana/efeitos dos fármacos , Camundongos da Linhagem 129 , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Fármacos do Sistema Nervoso Periférico/farmacologia , Nervo Frênico/efeitos dos fármacos , Nervo Frênico/fisiologia , Reflexo/efeitos dos fármacos , Reflexo/fisiologia , Cianeto de Sódio/farmacologia , Técnicas de Cultura de Tecidos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...