Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
J Biol Chem ; 298(11): 102536, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36174675

RESUMEN

The cellular response to hypoxia is regulated through enzymatic oxygen sensors, including the prolyl hydroxylases, which control degradation of the well-known hypoxia inducible factors (HIFs). Other enzymatic oxygen sensors have been recently identified, including members of the KDM histone demethylase family. Little is known about how different oxygen-sensing pathways interact and if this varies depending on the form of hypoxia, such as chronic or intermittent. In this study, we investigated how two proposed cellular oxygen-sensing systems, HIF-1 and KDM4A, KDM4B, and KDM4C, respond in cells exposed to rapid forms of intermittent hypoxia (minutes) and compared to chronic hypoxia (hours). We found that intermittent hypoxia increases HIF-1α protein through a pathway distinct from chronic hypoxia, involving the KDM4A, KDM4B, and KDM4C histone lysine demethylases. Intermittent hypoxia increases the quantity and activity of KDM4A, KDM4B, and KDM4C, resulting in a decrease in histone 3 lysine 9 (H3K9) trimethylation near the HIF1A locus. We demonstrate that this contrasts with chronic hypoxia, which decreases KDM4A, KDM4B, and KDM4C activity, leading to hypertrimethylation of H3K9 globally and at the HIF1A locus. Altogether, we found that demethylation of histones bound to the HIF1A gene in intermittent hypoxia increases HIF1A mRNA expression, which has the downstream effect of increasing overall HIF-1 activity and expression of HIF target genes. This study highlights how multiple oxygen-sensing pathways can interact to regulate and fine tune the cellular hypoxic response depending on the period and length of hypoxia.


Asunto(s)
Histonas , Subunidad alfa del Factor 1 Inducible por Hipoxia , Procesamiento Proteico-Postraduccional , Humanos , Desmetilación , Histona Demetilasas/metabolismo , Histonas/genética , Histonas/metabolismo , Hipoxia , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Histona Demetilasas con Dominio de Jumonji/genética , Histona Demetilasas con Dominio de Jumonji/metabolismo , Oxígeno/metabolismo
2.
J Physiol ; 596(15): 3149-3169, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29159869

RESUMEN

KEY POINTS: Activity-dependent plasticity can be induced in carotid body (CB) chemosensory afferents without chronic intermittent hypoxia (CIH) preconditioning by acute intermittent hypoxia coincident with bouts of hypercapnia (AIH-Hc). Several properties of this acute plasticity are shared with CIH-dependent sensory long-term facilitation (LTF) in that induction is dependent on 5-HT, angiotensin II, protein kinase C and reactive oxygen species. Several properties differ from CIH-dependent sensory LTF; H2 O2 appears to play no part in induction, whereas maintenance requires purinergic P2X2/3 receptor activation and is dependent on transient receptor potential vanilloid type 1 (TRPV1) receptor sensitization. Because P2X2/3 and TRPV1 receptors are located in carotid sinus nerve (CSN) terminals but not presynaptic glomus cells, a primary site of the acute AIH-Hc induced sensory LTF appears to be postsynaptic. Our results obtained in vivo suggest a role for TRPV1-dependent CB activity in acute sympathetic LTF. We propose that P2X-TRPV1-receptor-dependent sensory LTF may constitute an important early mechanism linking sleep apnoea with hypertension and/or cardiovascular disease. ABSTRACT: Apnoeas constitute an acute existential threat to neonates and adults. In large part, this threat is detected by the carotid bodies, which are the primary peripheral chemoreceptors, and is combatted by arousal and acute cardiorespiratory responses, including increased sympathetic output. Similar responses occur with repeated apnoeas but they continue beyond the last apnoea and can persist for hours [i.e. ventilatory and sympathetic long-term facilitation (LTF)]. These long-term effects may be adaptive during acute episodic apnoea, although they may prolong hypertension causing chronic cardiovascular impairment. We report a novel mechanism of acute carotid body (CB) plasticity (sensory LTF) induced by repeated apnoea-like stimuli [i.e. acute intermittent hypoxia coincident with bouts of hypercapnia (AIH-Hc)]. This plasticity did not require chronic intermittent hypoxia preconditioning, was dependent on P2X receptors and protein kinase C, and involved heat-sensitive transient receptor potential vanilloid type 1 (TRPV1) receptors. Reactive oxygen species (O2 ·¯) were involved in initiating plasticity only; no evidence was found for H2 O2 involvement. Angiotensin II and 5-HT receptor antagonists, losartan and ketanserin, severely reduced CB responses to individual hypoxic-hypercapnic challenges and prevented the induction of sensory LTF but, if applied after AIH-Hc, failed to reduce plasticity-associated activity. Conversely, TRPV1 receptor antagonism had no effect on responses to individual hypoxic-hypercapnic challenges but reduced plasticity-associated activity by ∼50%. Further, TRPV1 receptor antagonism in vivo reduced sympathetic LTF caused by AIH-Hc, although only if the CBs were functional. These data demonstrate a new mechanism of CB plasticity and suggest P2X-TRPV1-dependent sensory LTF as a novel target for pharmacological intervention in some forms of neurogenic hypertension associated with recurrent apnoeas.


Asunto(s)
Cuerpo Carotídeo/fisiología , Hipercapnia/fisiopatología , Hipoxia/fisiopatología , Receptores Purinérgicos P2X/fisiología , Canales Catiónicos TRPV/fisiología , Animales , Masculino , Ratas Sprague-Dawley
3.
Am J Physiol Heart Circ Physiol ; 314(3): H563-H572, 2018 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-29212793

RESUMEN

Intermittent hypoxia causes a persistent increase in sympathetic activity that progresses to hypertension in chronic conditions such as obstructive sleep apnea. Pituitary adenylate cyclase-activating polypeptide (PACAP) is an excitatory neurotransmitter that causes long-lasting sympathetic excitation. We aimed to determine if intermittent activation of the rostral ventrolateral medulla (RVLM) causes PACAP-mediated elevation of sympathetic nerve activity, termed sympathetic long-term facilitation (sLTF). The role of PACAP in mediating sLTF in response to intermittent activation of the RVLM was investigated in urethane-anaesthetized and artificially ventilated rats ( n = 65, Sprague-Dawley). Bilateral RVLM microinjections of the PACAP type 1 receptor/vasoactive intestinal polypeptide receptor type 2 receptor antagonist PACAP-(6-38) [ n = 6, change (Δ): -16.4 ± 6.5%) or an ionotropic glutamate antagonist, kynurenate ( n = 6, Δ:-7.2 ± 2.3%), blocked the development of acute intermittent hypoxia-induced sLTF ( n = 6, Δ: 49.2 ± 14.2%). Intermittent RVLM microinjections of glutamate caused sLTF ( n = 5, Δ: 56.9 ± 14.7%) that was abolished by PACAP-(6-38) pretreatment ( n = 5, Δ:-1.2 ± 4.7%). Conversely, intermittent microinjections of PACAP in the RVLM did not elicit sLTF. Intermittent bilateral disinhibition of the RVLM by microinjection of γ-aminobutyric acid in the caudal ventrolateral medulla did not elicit sLTF. Direct activation of RVLM neurons is crucial for the development of sLTF. PACAP and glutamate act synergistically in the RVLM, with both being necessary for the sLTF response. We found that activation of glutamate but not PACAP receptors is necessary and sufficient to generate sLTF, even in the absence of intermittent hypoxia. Our results demonstrate that PACAP within the RVLM may contribute to the development of obstructive sleep apnea -induced hypertension. NEW & NOTEWORTHY Pharmacological blockade of either pituitary adenylate cyclase-activating polypeptide (PACAP) or ionotropic glutamate receptors in the rostral ventrolateral medulla prevents development of sympathetic long-term facilitation. PACAP receptor inhibition prevents the occurrence of hypoxia-induced peripheral chemoreflex sensitization. Thus, PACAP receptors may be a potential therapeutic target serving to reduce heightened sympathetic tone and hypersensitized cardiovascular reflexes.


Asunto(s)
Hipoxia/fisiopatología , Ácido Quinurénico/administración & dosificación , Potenciación a Largo Plazo/efectos de los fármacos , Bulbo Raquídeo/efectos de los fármacos , Fragmentos de Péptidos/administración & dosificación , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/administración & dosificación , Apnea Obstructiva del Sueño/fisiopatología , Sistema Nervioso Simpático/efectos de los fármacos , Enfermedad Aguda , Animales , Modelos Animales de Enfermedad , Ácido Glutámico/administración & dosificación , Hipertensión/etiología , Hipertensión/fisiopatología , Hipoxia/complicaciones , Hipoxia/metabolismo , Masculino , Bulbo Raquídeo/metabolismo , Bulbo Raquídeo/fisiopatología , Microinyecciones , Ratas Sprague-Dawley , Receptores de Glutamato/efectos de los fármacos , Receptores de Glutamato/metabolismo , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/efectos de los fármacos , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/metabolismo , Apnea Obstructiva del Sueño/complicaciones , Apnea Obstructiva del Sueño/metabolismo , Sistema Nervioso Simpático/metabolismo , Sistema Nervioso Simpático/fisiopatología , Transmisión Sináptica/efectos de los fármacos , Factores de Tiempo , Ácido gamma-Aminobutírico/administración & dosificación
4.
Am J Physiol Regul Integr Comp Physiol ; 315(6): R1115-R1122, 2018 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-30281326

RESUMEN

Activation of neurons in the rostral ventrolateral medulla (RVLM) following glucoprivation initiates sympathoadrenal activation, adrenaline release, and increased glucose production. Here, we aimed to determine the role of RVLM µ-opioid receptors in the counterregulatory response to systemic glucoprivation. Experiments were performed in pentobarbital sodium anesthetized male Sprague-Dawley rats ( n = 30). Bilateral activation of RVLM µ-opioid receptors with [d-Ala2, N-Me-Phe4, Gly5-ol]-enkephalin (DAMGO) (8 mM, 50 nl) depressed adrenal sympathetic nerve activity for ~60 min ( n = 6; Δ49.9 ± 5.8%, P < 0.05). The counterregulatory response to glucoprivation (measured by adrenal sympathetic efferent nerve activity) induced by 2-deoxyglucose (2-DG) ( n = 6; Δ63.6 ± 16.5%, P < 0.05) was completely blocked 60 min after DAMGO microinjections ( n = 6; Δ10.2 ± 3.5%, P < 0.05). Furthermore, DAMGO pretreatment attenuated the increase in blood glucose levels after 2-DG infusion ( n = 6; 6.1 ± 0.7mmol/l vs. baseline 5.2 ± 0.3mmol/l, P > 0.05) compared with 2-DG alone ( n = 6; 7.6 ± 0.4mmol/l vs. baseline 6.0 ± 0.4mmol/l, P < 0.05). Thus, activation of RVLM µ-opioid receptors attenuated the neural efferent response to glucoprivation and reduced glucose production.


Asunto(s)
Desoxiglucosa/farmacología , Bulbo Raquídeo/efectos de los fármacos , Neuronas/fisiología , Receptores Opioides/efectos de los fármacos , Animales , Presión Sanguínea/efectos de los fármacos , Presión Sanguínea/fisiología , Desoxiglucosa/metabolismo , Masculino , Bulbo Raquídeo/metabolismo , Microinyecciones/métodos , Antagonistas de Narcóticos/farmacología , Neuronas/efectos de los fármacos , Ratas Sprague-Dawley , Receptores Opioides/metabolismo , Sistema Nervioso Simpático/efectos de los fármacos , Sistema Nervioso Simpático/fisiología
5.
Am J Physiol Regul Integr Comp Physiol ; 315(6): R1115-R1122, 2018 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-30499309

RESUMEN

Activation of neurons in the rostral ventrolateral medulla (RVLM) following glucoprivation initiates sympathoadrenal activation, adrenaline release, and increased glucose production. Here, we aimed to determine the role of RVLM µ-opioid receptors in the counterregulatory response to systemic glucoprivation. Experiments were performed in pentobarbital sodium anesthetized male Sprague-Dawley rats ( n = 30). Bilateral activation of RVLM µ-opioid receptors with [d-Ala2, N-Me-Phe4, Gly5-ol]-enkephalin (DAMGO) (8 mM, 50 nl) depressed adrenal sympathetic nerve activity for ~60 min ( n = 6; Δ49.9 ± 5.8%, P < 0.05). The counterregulatory response to glucoprivation (measured by adrenal sympathetic efferent nerve activity) induced by 2-deoxyglucose (2-DG) ( n = 6; Δ63.6 ± 16.5%, P < 0.05) was completely blocked 60 min after DAMGO microinjections ( n = 6; Δ10.2 ± 3.5%, P < 0.05). Furthermore, DAMGO pretreatment attenuated the increase in blood glucose levels after 2-DG infusion ( n = 6; 6.1 ± 0.7mmol/l vs. baseline 5.2 ± 0.3mmol/l, P > 0.05) compared with 2-DG alone ( n = 6; 7.6 ± 0.4mmol/l vs. baseline 6.0 ± 0.4mmol/l, P < 0.05). Thus, activation of RVLM µ-opioid receptors attenuated the neural efferent response to glucoprivation and reduced glucose production.


Asunto(s)
Desoxiglucosa/farmacología , Bulbo Raquídeo/efectos de los fármacos , Neuronas/efectos de los fármacos , Receptores Opioides/efectos de los fármacos , Animales , Presión Sanguínea/efectos de los fármacos , Presión Sanguínea/fisiología , Epinefrina/metabolismo , Masculino , Bulbo Raquídeo/fisiología , Antagonistas de Narcóticos/farmacología , Neuronas/fisiología , Ratas Sprague-Dawley , Sistema Nervioso Simpático/efectos de los fármacos , Sistema Nervioso Simpático/fisiología
6.
J Neurosci ; 36(2): 506-17, 2016 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-26758841

RESUMEN

Cardiovascular autonomic dysfunction in seizure is a major cause of sudden unexpected death in epilepsy. The catecholaminergic neurons in the rostral ventrolateral medulla (RVLM) maintain sympathetic vasomotor tone and blood pressure through their direct excitatory projections to the intermediolateral (IML) cell column. Glutamate, the principal excitatory neurotransmitter in brain, is increased in seizures. Pituitary adenylate cyclase activating polypeptide (PACAP) is an excitatory neuropeptide with neuroprotective properties, whereas microglia are key players in inflammatory responses in CNS. We investigated the roles of glutamate, PACAP, and microglia on RVLM catecholaminergic neurons during the cardiovascular responses to 2 mg/kg kainic acid (KA)-induced seizures in urethane anesthetized, male Sprague Dawley rats. Microinjection of the glutamate antagonist, kynurenic acid (50 nl; 100 mM) into RVLM, blocked the seizure-induced 43.2 ± 12.6% sympathoexcitation (p ≤ 0.05), and abolished the pressor responses, tachycardia, and QT interval prolongation. PACAP or microglia antagonists (50 nl) (PACAP(6-38), 15 pmol; minocycline 10 mg/ml) microinjected bilaterally into RVLM had no effect on seizure-induced sympathoexcitation, pressor responses, or tachycardia but abolished the prolongation of QT interval. The actions of PACAP or microglia on RVLM neurons do not cause sympathoexcitation, but they do elicit proarrhythmogenic changes. An immunohistochemical analysis in 2 and 10 mg/kg KA-induced seizure rats revealed that microglia surrounding catecholaminergic neurons are in a "surveillance" state with no change in the number of M2 microglia (anti-inflammatory). In conclusion, seizure-induced sympathoexcitation is caused by activation of glutamatergic receptors in RVLM that also cause proarrhythmogenic changes mediated by PACAP and microglia. SIGNIFICANCE STATEMENT: Sudden unexpected death in epilepsy is a major cause of death in epilepsy. Generally, seizures are accompanied by changes in brain function leading to uncontrolled nerve activity causing high blood pressure, rapid heart rate, and abnormal heart rhythm. Nevertheless, the brain chemicals causing these cardiovascular changes are unknown. Chemicals, such as glutamate and pituitary adenylate cyclase activating polypeptide, whose expression is increased after seizures, act on specific cardiovascular nuclei in the brain and influence the activity of the heart, and blood vessels. Microglia, which manage excitation in the brain, are commonly activated after seizure and produce pro- and/or anti-inflammatory factors. Hence, we aimed to determine the effects of blocking glutamate, pituitary adenylate cyclase activating polypeptide, and microglia in the RVLM and their contribution to cardiovascular autonomic dysfunction in seizure.


Asunto(s)
Anomalías Cardiovasculares/etiología , Bulbo Raquídeo/efectos de los fármacos , Microglía/efectos de los fármacos , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/farmacología , Receptores de Glutamato/metabolismo , Convulsiones/complicaciones , Animales , Proteínas de Unión al Calcio/metabolismo , Modelos Animales de Enfermedad , Antagonistas de Aminoácidos Excitadores/farmacología , Ritmo Gamma/efectos de los fármacos , Ritmo Gamma/fisiología , Ácido Kaínico/toxicidad , Masculino , Bulbo Raquídeo/metabolismo , Proteínas de Microfilamentos/metabolismo , Minociclina/farmacología , Fragmentos de Péptidos/farmacología , Ratas , Ratas Sprague-Dawley , Convulsiones/inducido químicamente , Convulsiones/patología , Nervios Esplácnicos/efectos de los fármacos , Nervios Esplácnicos/fisiología , Tirosina 3-Monooxigenasa/metabolismo , Vagotomía
7.
J Neurosci ; 35(5): 2191-9, 2015 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-25653374

RESUMEN

Seizures are accompanied by cardiovascular changes that are a major cause of sudden unexpected death in epilepsy (SUDEP). Seizures activate inflammatory responses in the cardiovascular nuclei of the medulla oblongata and increase neuronal excitability. Pituitary adenylate cyclase-activating polypeptide (PACAP) is a neuropeptide with autocrine and paracrine neuroprotective properties. Microglia are key players in inflammatory responses in the CNS. We sought to determine whether PACAP and microglia mitigate the adverse effects of seizure on cardiovascular function in a rat model of temporal lobe epilepsy. Kainic acid (KA)-induced seizures increased splanchnic sympathetic nerve activity by 97%, accompanied by increase in heart rate (HR) but not blood pressure (BP). Intrathecal infusion of the PACAP antagonist PACAP(6-38) or the microglia antagonists minocycline and doxycycline augmented sympathetic responses to KA-induced seizures. PACAP(6-38) caused a 161% increase, whereas minocycline and doxycycline caused a 225% and 215% increase, respectively. In intrathecal PACAP-antagonist-treated rats, both BP and HR increased, whereas after treatment with microglial antagonists, only BP was significantly increased compared with control. Our findings support the idea that PACAP and its action on microglia at the level of the spinal cord elicit cardioprotective effects during seizure. However, intrathecal PACAP did not show additive effects, suggesting that the agonist effect was at maximum. The protective effect of microglia may occur by adoption of an M2 phenotype and expression of factors such as TGF-ß and IL-10 that promote neuronal quiescence. In summary, therapeutic interventions targeting PACAP and microglia could be a promising strategy for preventing SUDEP.


Asunto(s)
Arritmias Cardíacas/etiología , Corazón/efectos de los fármacos , Microglía/efectos de los fármacos , Fragmentos de Péptidos/toxicidad , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/toxicidad , Convulsiones/fisiopatología , Animales , Arritmias Cardíacas/fisiopatología , Presión Sanguínea , Doxiciclina/toxicidad , Epilepsia del Lóbulo Temporal/complicaciones , Epilepsia del Lóbulo Temporal/metabolismo , Epilepsia del Lóbulo Temporal/fisiopatología , Frecuencia Cardíaca , Masculino , Minociclina/toxicidad , Ratas , Ratas Sprague-Dawley , Convulsiones/complicaciones , Convulsiones/metabolismo , Médula Espinal/efectos de los fármacos , Médula Espinal/fisiopatología , Sistema Nervioso Simpático/efectos de los fármacos , Sistema Nervioso Simpático/fisiopatología
8.
J Pharmacol Exp Ther ; 358(3): 492-501, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27384072

RESUMEN

Intermittent hypoxia causes a persistent increase in sympathetic nerve activity (SNA), which progresses to hypertension in conditions such as obstructive sleep apnea. Orexins (A and B) are hypothalamic neurotransmitters with arousal-promoting and sympathoexcitatory effects. We investigated whether the sustained elevation of SNA, termed sympathetic long-term facilitation, after acute intermittent hypoxia (AIH) is caused by endogenous orexin acting on spinal sympathetic preganglionic neurons. The role of orexin in the increased SNA response to AIH was investigated in urethane-anesthetized, vagotomized, and artificially ventilated Sprague-Dawley rats (n = 58). A spinally infused subthreshold dose of orexin-A (intermittent; 0.1 nmol × 10) produced long-term enhancement in SNA (41.4% ± 6.9%) from baseline. This phenomenon was not produced by the same dose of orexin-A administered as a bolus intrathecal infusion (1 nmol; 7.3% ± 2.3%). The dual orexin receptor blocker, Almorexant, attenuated the effect of sympathetic long-term facilitation generated by intermittent orexin-A (20.7% ± 4.5% for Almorexant at 30 mg∙kg(-1) and 18.5% ± 1.2% for 75 mg∙kg(-1)), but not in AIH. The peripheral chemoreflex sympathoexcitatory response to hypoxia was greatly enhanced by intermittent orexin-A and AIH. In both cases, the sympathetic chemoreflex sensitization was reduced by Almorexant. Taken together, spinally acting orexin-A is mechanistically sufficient to evoke sympathetic long-term facilitation. However, AIH-induced sympathetic long-term facilitation appears to rely on mechanisms that are independent of orexin neurotransmission. Our findings further reveal that the activation of spinal orexin receptors is critical to enhance peripheral chemoreceptor responses to hypoxia after AIH.


Asunto(s)
Células Quimiorreceptoras/citología , Células Quimiorreceptoras/efectos de los fármacos , Potenciación a Largo Plazo/efectos de los fármacos , Orexinas/administración & dosificación , Orexinas/farmacología , Sistema Nervioso Simpático/efectos de los fármacos , Sistema Nervioso Simpático/fisiología , Animales , Hipoxia de la Célula/efectos de los fármacos , Inyecciones Espinales , Ratas , Ratas Sprague-Dawley , Factores de Tiempo
9.
Am J Physiol Regul Integr Comp Physiol ; 307(11): R1292-302, 2014 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-25274906

RESUMEN

Neuronal stimulation by light is a novel approach in the emerging field of optogenetics, where genetic engineering is used to introduce light-activated channels. However, light is also capable of stimulating neurons even in the absence of genetic modifications through a range of physical and biological mechanisms. As a result, rigorous design of optogenetic experiments needs to take note of alternative and parallel effects of light illumination of neuronal tissues. Thus all matters relating to light penetration are critical to the development of studies using light-activated proteins. This paper discusses ways to quantify light, light penetration in tissue, as well as light stimulation of neurons in physiological conditions. We also describe the direct effect of light on neurons investigated at different sites.


Asunto(s)
Neuronas/fisiología , Neurociencias , Optogenética , Estimulación Luminosa , Física , Animales , Humanos , Luz
10.
Am J Physiol Regul Integr Comp Physiol ; 303(7): R719-26, 2012 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-22874427

RESUMEN

This study focuses on presympathetic neurons of the rostral ventrolateral medulla (RVLM) that regulate sympathetic vasomotor tone. Many neurotransmitters are colocalized in RVLM neurons and are released under specific conditions to modulate efferent homeostatic responses. Of particular interest here are two peptides colocalized in catecholaminergic RVLM neurons: catestatin and pituitary adenylate cyclase-activating polypeptide (PACAP). Chromogranin A-derived catestatin is a potent endogenous noncompetitive nicotinic and adrenoreceptor antagonist. Catestatin impairs adenylate cyclase and phospholipase C action: mechanisms engaged by PACAP. Although PACAP and catestatin are likely coreleased, the possible effects of this are unknown. We aimed to determine whether catestatin affects the normal sympathoexcitatory but isotensive responses to intrathecal PACAP. Urethane-anesthetized, vagotomized, ventilated Sprague-Dawley rats (n = 22) were given an intrathecal injection of catestatin at different times prior to intrathecal administration of PACAP-38. Arterial pressure, splanchnic sympathetic nerve activity, heart rate, and reflex responses to baroreceptor and chemoreceptor activation were recorded. The key findings of this study are that pretreatment with catestatin time dependently enhances the PACAP-38 effect on mean arterial pressure and enhances sympathetic barosensitivity and chemosensitivity. The time-scale of the effect of catestatin on the response to PACAP-38 strongly suggests that catestatin is either causing changes in gene expression to exert its effects, or modifying intracellular mechanisms normally engaged by PAC(1) receptors. The ability of catestatin pretreatment to enhance barosensitivity and chemosensitivity after PACAP-38 injection supports the hypothesis that catestatin manipulates the intracellular environment within sympathetic neurons in a way that increases responses to PACAP.


Asunto(s)
Presión Sanguínea/efectos de los fármacos , Presión Sanguínea/fisiología , Cromogranina A/farmacología , Fragmentos de Péptidos/farmacología , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/farmacología , Médula Espinal/efectos de los fármacos , Animales , Sistema Cardiovascular/efectos de los fármacos , Cromogranina A/administración & dosificación , Frecuencia Cardíaca/efectos de los fármacos , Frecuencia Cardíaca/fisiología , Inyecciones Espinales , Masculino , Modelos Animales , Fragmentos de Péptidos/administración & dosificación , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/administración & dosificación , Ratas , Ratas Sprague-Dawley , Médula Espinal/fisiología , Sistema Nervioso Simpático/efectos de los fármacos , Sistema Nervioso Simpático/fisiología , Vasodilatadores/administración & dosificación , Vasodilatadores/farmacología
11.
Metabolites ; 13(1)2022 Dec 27.
Artículo en Inglés | MEDLINE | ID: mdl-36676967

RESUMEN

Hypoglycemia-associated autonomic failure (HAAF) is a serious, life-threatening complication of intensive insulin therapy, particularly in people with type 1 diabetes. The ketogenic diet is reported to beneficially affect glycemic control in people with type 1 diabetes, however its effects on the neurohormonal counterregulatory response to recurrent hypoglycemia and HAAF development are understudied. In this study we used Sprague Dawley rats to establish a HAAF model under non-diabetic and streptozotocin (STZ)-induced diabetic conditions and determined how nutritional ketosis affected the neurohormonal counterregulation and the activity of energy-sensing orexin (OX) neurons. We found that antecedent hypoglycemia diminished the sympathoexcitatory epinephrine response to subsequent hypoglycemia in chow-fed non-diabetic rats, but this did not occur in STZ-diabetic animals. In all cases a ketogenic diet preserved the epinephrine response. Contrary to expectations, STZ-diabetic keto-fed rats showed reduced OX activity in the recurrent hypoglycemia group, which did not occur in any other group. It is possible that the reduced activation of OX neurons is an adaptation aimed at energy conservation accompanied by diminished arousal and exploratory behaviour. Our data suggests that while a ketogenic diet has beneficial effects on glycemia, and epinephrine response, the reduced activation of OX neurons could be detrimental and warrants further investigation.

12.
Am J Physiol Heart Circ Physiol ; 300(1): H214-22, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20952662

RESUMEN

The rostral ventrolateral medulla contains presympathetic neurons that project monosynaptically to sympathetic preganglionic neurons (SPN) in the spinal cord and are essential for the tonic and reflex control of the cardiovascular system. SPN directly innervate the adrenal medulla and, via postganglionic axons, affect the heart, kidneys, and blood vessels to alter sympathetic outflow and hence blood pressure. Over 80% of bulbospinal, catecholaminergic (C1) neurons contain pituitary adenylate cyclase-activating polypeptide (PACAP) mRNA. Activation of PACAP receptors with intrathecal infusion of PACAP-38 causes a robust, prolonged elevation in sympathetic tone. Given that a common feature of most forms of hypertension is elevated sympathetic tone, this study aimed to determine in the spontaneously hypertensive rat (SHR) and the Wistar Kyoto rat (normotensive control) 1) the proportion of C1 neurons containing PACAP mRNA and 2) responsiveness to intrathecal PACAP-38. We further investigated whether intrathecal infusion of the PACAP antagonist, PACAP(6-38), reduces the hypertension in the SHR. The principal findings are that 1) the proportion of PACAP mRNA-containing C1 neurons is not different between normotensive and hypertensive rats, 2) intrathecal PACAP-38 causes a strain-dependent, sustained sympathoexcitation and tachycardia with variable effects on mean arterial pressure in normotensive and hypertensive rats, and 3) PACAP(6-38) effectively attenuated the effects of intrathecal PACAP-38, but had no effect alone, on any baseline variables. This finding indicates that PACAP-38 is not tonically released in the spinal cord of rats. A role for PACAP in hypertension in conscious rats remains to be determined.


Asunto(s)
Presión Sanguínea/efectos de los fármacos , Frecuencia Cardíaca/efectos de los fármacos , Hipertensión/fisiopatología , Bulbo Raquídeo/efectos de los fármacos , Neuronas/efectos de los fármacos , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/farmacología , Sistema Nervioso Simpático/efectos de los fármacos , Análisis de Varianza , Animales , Técnica del Anticuerpo Fluorescente , Hipertensión/metabolismo , Hibridación in Situ , Inyecciones Espinales , Bulbo Raquídeo/metabolismo , Bulbo Raquídeo/fisiopatología , Neuronas/metabolismo , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/metabolismo , ARN Mensajero/metabolismo , Ratas , Ratas Endogámicas SHR , Ratas Endogámicas WKY , Especificidad de la Especie , Sistema Nervioso Simpático/metabolismo , Sistema Nervioso Simpático/fisiopatología , Taquicardia/inducido químicamente , Tirosina 3-Monooxigenasa/metabolismo , Vasodilatadores/farmacología
13.
Am J Physiol Heart Circ Physiol ; 300(6): H2300-7, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21460201

RESUMEN

The rostral ventrolateral medulla differentially regulates sympathetic output to different vascular beds, possibly through the release of various neurotransmitters and peptides that may include pituitary adenylate cyclase-activating polypeptide (PACAP). An intrathecal administration of PACAP increases splanchnic sympathetic nerve activity and heart rate, but not mean arterial blood pressure. The mechanism behind this response is unknown but may be due to a differential control of sympathetic outflows. In this study we sought 1) to investigate whether intrathecal PACAP differentially affects sympathetic outflow, 2) to determine whether the intrathecal responses to PACAP are solely due to a spinally mediated mechanism, and 3) to determine whether intrathecal PACAP affects metabolic function. Experiments using urethane-anesthetized, vagotomized, ventilated, and paralyzed adult male Sprague-Dawley rats were conducted in this study. Intrathecal injections of PACAP-38 were given, and mean arterial pressure, heart rate, the activity of regional sympathetic nerves, end-tidal CO(2), and core temperature were recorded. The novel findings of this study are that 1) intrathecal PACAP-38 causes a prolonged widespread sympathoexcitation in multiple sympathetic beds, 2) this widespread sympathoexcitation is mediated within the spinal cord itself since spinal transection does not abrogate the response, and 3) that intrathecal PACAP-38 increases basal metabolic rate. Therefore, we conclude that intrathecal PACAP acts in the spinal cord to cause a prolonged widespread sympathoexcitation and that PACAP also causes an increase in basal metabolic rate that includes an increase in brown adipose tissue thermogenesis in our rat preparation.


Asunto(s)
Metabolismo Basal/efectos de los fármacos , Neurotransmisores/farmacología , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/farmacología , Médula Espinal/fisiología , Sistema Nervioso Simpático/efectos de los fármacos , Sistema Nervioso Simpático/fisiología , Inconsciencia/fisiopatología , Animales , Metabolismo Basal/fisiología , Glucemia/metabolismo , Presión Sanguínea/efectos de los fármacos , Presión Sanguínea/fisiología , Temperatura Corporal/efectos de los fármacos , Temperatura Corporal/fisiología , Frecuencia Cardíaca/efectos de los fármacos , Frecuencia Cardíaca/fisiología , Concentración de Iones de Hidrógeno , Inyecciones Espinales , Masculino , Modelos Animales , Neurotransmisores/administración & dosificación , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/administración & dosificación , Ratas , Ratas Sprague-Dawley
14.
Front Physiol ; 12: 645392, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33746780

RESUMEN

A key feature of sleep disordered breathing syndromes, such as obstructive sleep apnea is intermittent hypoxia. Intermittent hypoxia is well accepted to drive the sympathoexcitation that is frequently associated with hypertension and diabetes, with measurable effects after just 1 h. The aim of this study was to directly measure the glucose response to 1 h of acute intermittent hypoxia in pentobarbital anesthetized rats, compared to conscious rats. However, we found that while a glucose response is measurable in conscious rats exposed to intermittent hypoxia, it is suppressed in anesthetized rats. Intermittent hypoxia for 1, 2, or 8 h increased blood glucose by 0.7 ± 0.1 mmol/L in conscious rats but had no effect in anesthetized rats (-0.1 ± 0.2 mmol/L). These results were independent of the frequency of the hypoxia challenges, fasting state, vagotomy, or paralytic agents. A supraphysiological challenge of 3 min of hypoxia was able to induce a glycemic response indicating that the reflex response is not abolished under pentobarbital anesthesia. We conclude that pentobarbital anesthesia is unsuitable for investigations into glycemic response pathways in response to intermittent hypoxia in rats.

15.
Nutrients ; 13(8)2021 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-34444787

RESUMEN

The sympathoadrenal counterregulatory response to hypoglycemia is critical for individuals with type 1 diabetes due to impaired ability to produce glucagon. Ketogenic diets (KD) are an increasingly popular diabetes management tool; however, the effects of KD on the sympathoadrenal response are largely unknown. Here, we determined the effects of KD-induced ketosis on the sympathoadrenal response to a single insulin-induced hypoglycemic challenge. We investigated how a 3 week KD feeding regimen affected the main components of the sympathoadrenal counterregulatory response: adrenal sympathetic nerve activity (ASNA), adrenal gland activity, plasma epinephrine, and brainstem glucose-responsive C1 neuronal activation in anesthetized, nondiabetic male Sprague-Dawley rats. Rats on KD had similar blood glucose (BG) levels and elevated ketone body ß-hydroxybutyrate (BHB) levels compared to the control Chow diet group. All KD rats responded to hypoglycemia with a robust increase in ASNA, which was initiated at significantly lower BG levels compared to Chow-fed rats. The delay in hypoglycemia-induced ASNA increase was concurrent with rapid disappearance of BHB from cerebral and peripheral circulation. Adrenal gland activity paralleled epinephrine and ASNA response. Overall, KD-induced ketosis was associated with initiation of the sympathoadrenal response at lower blood glucose levels; however, the magnitude of the response was not diminished.


Asunto(s)
Dieta Cetogénica , Hipoglucemiantes/farmacología , Sistema Simpatoadrenal , Ácido 3-Hidroxibutírico/farmacología , Glándulas Suprarrenales/efectos de los fármacos , Animales , Glucemia , Modelos Animales de Enfermedad , Epinefrina/sangre , Glucagón , Glucosa/efectos adversos , Hipoglucemia/sangre , Hipoglucemia/terapia , Hipoglucemiantes/uso terapéutico , Insulina , Cetosis , Masculino , Ratas , Ratas Sprague-Dawley , Sistema Nervioso Simpático/efectos de los fármacos
16.
Respir Physiol Neurobiol ; 166(2): 80-6, 2009 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-19429523

RESUMEN

Controversy surrounds the respiratory responses to baroreceptor activation. Although many reflexes that effect respiration (e.g. chemoreflexes and nociceptive reflexes) frequently affect cardiovascular parameters, the effect of baroreflex stimulation within normal physiological limits is generally considered to affect only blood pressure and heart rate. Even though previous authors have reported that baroreceptor activation can affect respiratory activity, the effects on respiratory frequency and amplitude are highly variable, and changes in perfusion evoked by blood pressure manipulation could account for the observed effects. Here, we determined the respiratory effects of activating arterial baroreceptors by intravenous injection of phenylephrine or angiotensin II, or by electrical stimulation of the aortic depressor nerve (ADN). In urethane-anesthetized vagotomized rats, 1, 2 and 4s trains of tetanic ADN stimulation evoked 3.1+/-1.1%, 11.2+/-13.6% and 21.9+/-8.9% increases in inspiratory (TI) time and 26.5+/-18%, 23.4+/-15.7% and 34.6+/-20.9% increases in expiratory (TE) time, respectively (P<0.05 in both cases), but no effect on the amplitude of bursts recorded in the phrenic nerve. Similar effects were observed following pressor trials evoked by intravenous PE (TE: +26.1+/-9.1%, P<0.01), but not Ang II. Intermittent ADN stimulation (single pulse, 1 Hz) significantly increased the variability of TI during periods of low respiratory drive (P<0.05) without significantly affecting any other parameters. We propose that a specific baroreceptor-respiratory response exists that is independent of changes in blood flow. In contrast to the effects of baroreceptor stimulation on sympathetic nerve activity, the baro-respiratory response is subtle and highly dependent on respiratory drive.


Asunto(s)
Barorreflejo/fisiología , Presorreceptores/fisiología , Respiración , Análisis de Varianza , Angiotensina II/farmacología , Animales , Aorta/inervación , Desnervación Autonómica/métodos , Barorreflejo/efectos de los fármacos , Biofisica , Presión Sanguínea/efectos de los fármacos , Presión Sanguínea/fisiología , Fenómenos Fisiológicos Cardiovasculares , Estimulación Eléctrica/métodos , Frecuencia Cardíaca/efectos de los fármacos , Frecuencia Cardíaca/fisiología , Inyecciones Intravenosas/métodos , Masculino , Fenilefrina/farmacología , Nervio Frénico/efectos de los fármacos , Nervio Frénico/fisiología , Ratas , Ratas Sprague-Dawley , Vagotomía/métodos , Vasoconstrictores/farmacología
17.
Respir Physiol Neurobiol ; 265: 83-91, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30172780

RESUMEN

Autonomic reflex responses are critical in restoring changes to circulatory factors reduced beyond the domain of homeostasis. Intermittent hypoxia triggers repeated activation of chemoreflexes, resulting in baroreflex dysfunction and widespread changes in cellular and neuronal activity regulated by sensory/motor pathways. Hypoglycaemia initiates a rapid neurally-mediated counter-regulatory response. This counter-regulatory response to hypoglycaemia increases plasma adrenaline levels, liver glycogenolysis, and thus blood glucose levels. Context-dependent activation of rostral ventral medullary neurons initiates baroreceptor unloading, peripheral chemoreflex firing and the counter-regulatory response to hypoglycaemia. In this review, we briefly focus on the functional integration between peripheral and medullary pathways comprising the sympathetic baroreflex, chemoreflexes, and the counter-regulatory response to hypoglycaemia.


Asunto(s)
Sistema Nervioso Autónomo/fisiología , Cuerpo Carotídeo/fisiología , Hipoglucemia/fisiopatología , Hipoxia/fisiopatología , Reflejo/fisiología , Respiración , Rombencéfalo/fisiología , Animales , Sistema Nervioso Autónomo/fisiopatología , Cuerpo Carotídeo/fisiopatología , Humanos , Rombencéfalo/fisiopatología
18.
Auton Neurosci ; 216: 9-16, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30598122

RESUMEN

The RVLM of spontaneously hypertensive rats (SHR) contains over-active C1 neurons, which model the pathology of essential hypertension. Hypertension involves chronic low-grade neuroinflammation. Inflammation in the brain is produced and maintained primarily by microglia. We assessed microglial gene expression (P2Y12R and CX3CR1) and morphology in the RVLM of SHR compared to normotensive Wistar-Kyoto rats (WKY). The gene expression of the metabotropic purinergic receptor P2Y12 and the fractalkine receptor CX3CR1 was downregulated in the RVLM of SHR compared to WKY (by 37.3% and 30.9% respectively). P2Y12R and CX3CR1 are required for normal microglial function, and reduced P2Y12R expression is associated with changes in microglial activity. Histological analysis showed a 22.9% reduction in microglial cell density, along with 18.7% shorter microglial processes, a phenotypic indicator of activation, in the RVLM of SHR compared to WKY. These results indicate a subtle loss of function, or a mild state of inflammation, in the RVLM microglia of SHR.


Asunto(s)
Receptor 1 de Quimiocinas CX3C/biosíntesis , Bulbo Raquídeo/citología , Microglía/citología , Microglía/metabolismo , Receptores Purinérgicos P2Y12/biosíntesis , Animales , Recuento de Células , Regulación hacia Abajo , Expresión Génica/fisiología , Masculino , Bulbo Raquídeo/metabolismo , Ratas , Ratas Endogámicas SHR , Ratas Endogámicas WKY , Especificidad de la Especie
19.
Front Neurosci ; 13: 881, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31496933

RESUMEN

Repetitive hypoxia is a key feature of obstructive sleep apnoea (OSA), a condition characterized by intermittent airways obstruction. Patients with OSA present with persistent increases in sympathetic activity and commonly develop hypertension. The objectives of this study were to determine if the persistent increases in sympathetic nerve activity, known to be induced by acute intermittent hypoxia (AIH), are mediated through activation of the pituitary adenylate cyclase activating polypeptide (PACAP) signaling system. Here, we show that the excitatory neuropeptide PACAP, acting in the spinal cord, is important for generating the sympathetic response seen following AIH. Using PACAP receptor knockout mice, and pharmacological agents in Sprague Dawley rats, we measured blood pressure, heart rate, pH, PaCO2, and splanchnic sympathetic nerve activity, under anaesthesia, to demonstrate that the sympathetic response to AIH is mediated via the PAC1 receptor, in a cAMP-dependent manner. We also report that both intermittent microinjection of glutamate into the rostroventrolateral medulla (RVLM) and intermittent infusion of a sub-threshold dose of PACAP into the subarachnoid space can mimic the sympathetic response to AIH. All the sympathetic responses are independent of blood pressure, pH or PaCO2 changes. Our results show that in AIH, PACAP signaling in the spinal cord helps drive persistent increases in sympathetic nerve activity. This mechanism may be a precursor to the development of hypertension in conditions of chronic intermittent hypoxia, such as OSA.

20.
Clin Exp Pharmacol Physiol ; 35(4): 508-11, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18307751

RESUMEN

1. Cardiovascular sympathetic nerve activity at rest is grouped into waves, or bursts, that are generally, although not exclusively, related to the heart rate and to respiration. In addition, activity is also generated in response to central commands and to environmental stimuli. 2. Responsibility for the integration of all these different elements of sympathetic activity rests with pre-motoneurons in the rostral ventrolateral medulla oblongata. These pre-motoneurons are glutamatergic and spinally projecting where they form synapses with sympathetic preganglionic neurons. 3. Pre-motoneurons also contain and presumably release, neurotransmitters other than glutamate, including amines and neuropeptides that act on metabotropic receptors with long-term effects on cell function. 4. Similarly, in the rostral ventrolateral medulla oblongata the pre-motoneurons are mainly regulated by excitatory influences from glutamate and inhibitory influences from gamma-aminobutyric acid (GABA). Major focuses of recent studies are the interactions between non-glutamatergic and GABAergic systems and reflexes that regulate the activity of the sympathetic nervous system. 5. The results indicate that neurotransmitters acting at metabotropic receptors selectively affect different reflexes in the rostral ventrolateral medulla. It is suggested that this differential activation or attenuation of reflexes by different neurotransmitters is a mechanism by which the organism can fine-tune its responses to different homeostatic requirements.


Asunto(s)
Fibras Adrenérgicas/fisiología , Bulbo Raquídeo/citología , Bulbo Raquídeo/fisiología , Animales , Electrofisiología , Ratas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA